US20180250424A1 - Compositions and methods for promoting homology directed repair - Google Patents

Compositions and methods for promoting homology directed repair Download PDF

Info

Publication number
US20180250424A1
US20180250424A1 US15/518,105 US201515518105A US2018250424A1 US 20180250424 A1 US20180250424 A1 US 20180250424A1 US 201515518105 A US201515518105 A US 201515518105A US 2018250424 A1 US2018250424 A1 US 2018250424A1
Authority
US
United States
Prior art keywords
domain
nucleotides
cas9
molecule
nucleic acid
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US15/518,105
Inventor
Cecilia Cotta-Ramusino
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Editas Medicine Inc
Original Assignee
Editas Medicine Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Editas Medicine Inc filed Critical Editas Medicine Inc
Priority to US15/518,105 priority Critical patent/US20180250424A1/en
Assigned to EDITAS MEDICINE, INC. reassignment EDITAS MEDICINE, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: COTTA-RAMUSINO, Cecilia
Publication of US20180250424A1 publication Critical patent/US20180250424A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • A61K48/0066Manipulation of the nucleic acid to modify its expression pattern, e.g. enhance its duration of expression, achieved by the presence of particular introns in the delivered nucleic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/0083Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the administration regime
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/0091Purification or manufacturing processes for gene therapy compositions
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/111General methods applicable to biologically active non-coding nucleic acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/62DNA sequences coding for fusion proteins
    • C12N15/625DNA sequences coding for fusion proteins containing a sequence coding for a signal sequence
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/64General methods for preparing the vector, for introducing it into the cell or for selecting the vector-containing host
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/16Hydrolases (3) acting on ester bonds (3.1)
    • C12N9/22Ribonucleases RNAses, DNAses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/20Fusion polypeptide containing a tag with affinity for a non-protein ligand
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/80Fusion polypeptide containing a DNA binding domain, e.g. Lacl or Tet-repressor
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/85Fusion polypeptide containing an RNA binding domain
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/20Type of nucleic acid involving clustered regularly interspaced short palindromic repeats [CRISPRs]

Definitions

  • the CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats)/Cas (CRISPR-associated) system evolved in bacteria and archaea as an adaptive immune system to defend against viral attack.
  • CRISPR locus Upon exposure to a virus, short segments of viral DNA are integrated into the CRISPR locus.
  • RNA is transcribed from a portion of the CRISPR locus that includes the viral sequence. That RNA, which contains sequence complimentary to the viral genome, mediates targeting of a Cas9 protein to the sequence in the viral genome.
  • the Cas9 protein cleaves and thereby silences the viral target.
  • DSBs site-specific double strand breaks
  • HDR homology-directed repair
  • a Cas9 fusion molecule or Cas9 system may be used to mediate a break or nick near a target position, e.g., a chromosome position, that one desires to edit.
  • the cell then utilizes one of several repair pathways, e.g., HDR, to repair the break or nick.
  • a Cas9 repair system comprising a Cas9 fusion molecule, a nucleic acid template system, and optionally, at least one gRNA molecule.
  • the proximity of the template nucleic acid used by the cell to repair a Cas9-mediated cleavage event can be increased, and the frequency of use of a particular DNA repair pathway, e.g., HDR, can be modulated.
  • a particular DNA repair pathway e.g., HDR
  • a Cas9 fusion molecule comprising a Cas9 molecule linked to a template binding domain.
  • the template binding domain comprises a specific affinity for a template binding domain partner. In one embodiment, the template binding domain does not comprise substantial affinity for a nucleic acid from one or more of a human, a non-human animal, a mammal, a eukaryote, a plant, or a pathogen.
  • the template binding domain is not a nuclease, a transcription factor, an antibody or other complementarity determining region (CDR)-based molecule, a protein that alters chromatin, a protein that binds chromatin, a protein that modifies DNA, a DNA methylase, a protein that cleaves DNA, a protein that unwinds DNA, or any combination thereof.
  • the nuclease is an endonuclease or an exonuclease.
  • the chromatin comprises a histone.
  • the template binding domain does not comprise substantial affinity for a nucleic acid from a preselected species.
  • the preselected species is a human, a non-human animal, a mammal, a eukaryote, a plant, a pathogen, or any combination thereof. In one embodiment, the preselected species is human.
  • the Cas9 molecule is covalently linked to the template binding domain. In another embodiment, the Cas9 molecule is non-covalently linked to the template binding domain.
  • the template binding domain is linked to the N-terminus of the Cas9 molecule. In one embodiment, the template binding domain is covalently linked to the N-terminus of the Cas9 molecule. In another embodiment, the template binding domain is non-covalently linked to the N-terminus of the Cas9 molecule.
  • the template binding domain is linked to the C-terminus of the Cas9 molecule. In one embodiment, the template binding domain is covalently linked to the C-terminus of the Cas9 molecule. In another embodiment, the template binding domain is non-covalently linked to the C-terminus of the Cas9 molecule.
  • the Cas9 fusion molecule comprises at least two template binding domains.
  • the template binding domain comprises a protein, a nucleic acid, or a small molecule.
  • the nucleic acid is a DNA or an RNA.
  • the template binding domain comprises a protein, wherein the protein comprises a DNA binding domain.
  • the protein comprises a repressor protein, or a fragment of a repressor protein.
  • the repressor protein, or the fragment of the repressor protein is a TetR repressor, or a fragment of the TetR repressor; a LacI repressor, or a fragment of the LacI repressor; a Gal4 repressor, or a fragment of the Gal4 repressor; or a repressor protein C1, or a fragment of the repressor protein C1.
  • the Cas9 fusion protein comprises a linker between the Cas9 molecule and the template binding domain.
  • the linker is sufficiently long to allow the Cas9 molecule to bind to a target nucleic acid and the template binding domain to bind to a template binding domain partner without steric interference.
  • the linker is at least 6, but no longer than 60 amino acids in length.
  • the linker is at least 18, but no longer than 180 nucleotides in length.
  • the linker is at least 10, 50, 100, 200, 500, 1000, 2000, 5000, or 10000 Angstroms in length.
  • the linker is no more than 10, 50, 100, 200, 500, 1000, 2000, 5000, or 10000 Angstroms in length.
  • the linker comprises a polypeptide.
  • the linker comprises serine, glycine, or glycine and serine.
  • the linker comprises a sequence of Table 2 or Table 3.
  • the Cas9 molecule is a Cas9 molecule selected from Table 100. In one embodiment, the Cas9 molecule is a Cas9 molecule selected from Table 600. In one embodiment, the Cas9 molecule is not a wild-type Cas9 molecule.
  • the Cas9 molecule is an eaCas9 molecule. In one embodiment, the eaCas9 molecule forms a double stranded break in a target nucleic acid. In one embodiment, the eaCas9 molecule comprises N-terminal RuvC-like domain cleavage activity and HNH-like domain cleavage activity. In another embodiment, the eaCas9 molecule forms a single stranded break in a target nucleic acid. In one embodiment, the eaCas9 molecule comprises HNH-like domain cleavage activity but has no, or no significant, N-terminal RuvC-like domain cleavage activity.
  • the eaCas9 molecule is an HNH-like domain nickase. In one embodiment, the eaCas9 molecule comprises a mutation at an amino acid position corresponding to amino acid position D10 of Streptococcus pyogenes Cas9. In yet another embodiment, the eaCas9 molecule comprises N-terminal RuvC-like domain cleavage activity but has no, or no significant, HNH-like domain cleavage activity. In one embodiment, the eaCas9 molecule is an N-terminal RuvC-like domain nickase. In one embodiment, the eaCas9 molecule comprises a mutation at an amino acid position corresponding to amino acid position H840 or N863 of S. pyogenes Cas9.
  • the Cas9 molecule is an eiCas9 molecule.
  • the Cas9 molecule comprises a mutation, e.g., a point mutation, that causes the Cas9 molecule to be inactive, e.g., a mutation that eliminates the Cas9 molecule cleavage activity.
  • the Cas9 molecule comprises a REC2 deletion, REC1 CT deletion, or a REC1 SUB deletion, or any combination thereof.
  • the Cas9 molecule comprises an altered PI domain.
  • the Cas9 molecule is less than about 1300 amino acids in length. In one embodiment, the Cas9 molecule is less than about 1200, 1100, 1000, 900, or 800 amino acids in length. In one embodiment, the Cas9 molecule is between about 800 to 1300, 900 to 1200, 900 to 1100, or 900 to 1000 amino acids in length.
  • the Cas9 fusion molecule is substantially purified.
  • a Cas9 system comprising a Cas9 fusion molecule described herein, and a nucleic acid template system comprising a template binding domain partner and a template nucleic acid.
  • the template binding domain of the Cas9 fusion molecule is bound to the template binding domain partner. In one embodiment, the template binding domain of the Cas9 fusion molecule is covalently bound to the template binding domain partner. In another embodiment, the template binding domain of the Cas9 fusion molecule is non-covalently bound to the template binding domain partner.
  • the template binding domain partner is linked to the template nucleic acid. In one embodiment, the template binding domain partner is covalently linked to the template nucleic acid. In another embodiment, the template binding domain partner is non-covalently linked to the template nucleic acid.
  • the template binding domain partner comprises a protein, a nucleic acid, or a small molecule.
  • the nucleic acid is a DNA or an RNA.
  • the template binding domain comprises a protein and the template binding domain partner comprises a protein.
  • the template binding domain comprises a protein and the template binding domain partner comprises a nucleic acid.
  • the nucleic acid is a DNA.
  • the DNA is a double-stranded DNA or a single-stranded DNA.
  • the nucleic acid is an RNA.
  • the template binding domain comprises a protein and the template binding domain partner comprises a small molecule.
  • the template binding domain comprises a small molecule
  • the template binding domain partner comprises a protein
  • the template binding domain comprises a small molecule
  • the template binding domain partner comprises a small molecule
  • the template binding domain comprises a small molecule, and the template binding domain partner comprises a nucleic acid.
  • the template binding domain comprises a nucleic acid, and the template binding domain partner comprises a protein.
  • the template binding domain comprises a nucleic acid, and the template binding domain partner comprises a nucleic acid.
  • the template binding domain comprises a nucleic acid, and the template binding domain partner comprises a small molecule.
  • the nucleic acid is an RNA.
  • the nucleic acid is a DNA.
  • the DNA is a double-stranded DNA or a single-stranded DNA.
  • the template binding domain partner is a DNA sequence recognized by a DNA binding protein.
  • the DNA sequence recognized by the DNA binding protein is selected from a Tet-O sequence, a Lac operon O1 sequence, a UAS sequence, or an Operator L and R sequence.
  • the template binding domain partner comprises a repressor-binding DNA sequence from a bacterial operon.
  • the protein comprises a TetR repressor, or a fragment of the TetR repressor, and the DNA comprises at least one Tet-O sequence.
  • the DNA comprises at least two, three, four, five, or six Tet-O sequences.
  • the DNA comprises a spacer sequence between the Tet-O sequences.
  • the spacer sequence is at least 17 nucleotides in length. In another embodiment, the spacer sequence is at least 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 nucleotides in length. In one embodiment, the DNA does not comprise a spacer sequence between the Tet-O sequences.
  • the template binding domain partner comprises a protein that is not a nuclease, a transcription factor, an antibody or other CDR-based molecule, a protein that alters chromatin, a protein that binds chromatin, a protein that modifies DNA, a DNA methylase, a protein that cleaves DNA, a protein that unwinds DNA, or any combination thereof.
  • the nuclease is an endonuclease or an exonuclease.
  • the chromatin comprises a histone.
  • the Cas9 system comprises at least one additional template binding domain and at least one additional template binding domain partner.
  • the template binding domain partner comprises a linear nucleic acid. In another embodiment, the template binding domain partner comprises a nucleic acid sequence located on a circular nucleic acid.
  • the template binding domain partner comprises a double stranded nucleic acid sequence. In one embodiment, the template binding domain partner comprises a single stranded nucleic acid sequence.
  • the template nucleic acid comprises a double stranded nucleic acid sequence or a single stranded nucleic acid sequence.
  • the nucleic acid template system comprises a double stranded nucleic acid sequence or a single stranded nucleic acid sequence.
  • the template nucleic acid comprises a linear nucleic acid. In another embodiment, the template nucleic acid is a nucleic acid located on a circular nucleic acid.
  • the template binding domain partner comprises a double stranded nucleic acid; and the template nucleic acid comprises a double stranded nucleic acid.
  • the template binding domain partner comprises a double stranded nucleic acid; and the template nucleic acid comprises a single stranded nucleic acid.
  • the nucleic acid template system is a circular nucleic acid. In another embodiment, the nucleic acid template system is a linear nucleic acid.
  • the template binding domain partner comprises a single stranded nucleic acid; and the template nucleic acid comprises a single stranded nucleic acid.
  • the template binding domain partner comprises a single stranded nucleic acid; and the template nucleic acid comprises a double stranded nucleic acid.
  • the nucleic acid template system is a single stranded nucleic acid, and the template binding domain partner is 5′ of the template nucleic acid. In another embodiment, the nucleic acid template system is a single stranded nucleic acid, and the template binding domain partner is 3′ of the template nucleic acid.
  • the nucleic acid template system is a double stranded nucleic acid, and the template binding domain partner is 5′ of the template nucleic acid. In another embodiment, the nucleic acid template system is a double stranded nucleic acid, and the template binding domain partner is 3′ of the template nucleic acid.
  • the template nucleic acid comprises about 50-500 nucleotides of homology with a target nucleic acid. In one embodiment, the template nucleic acid comprises about 100-200 nucleotides of homology with a target nucleic acid. In one embodiment, the template nucleic acid comprises about 500-2000 nucleotides of homology with a target nucleic acid.
  • the template nucleic acid comprises a human nucleic acid sequence. In one embodiment, the template nucleic acid comprises a wild-type human nucleic acid sequence.
  • the Cas9 system further comprises at least one additional template binding domain partner.
  • the template nucleic acid lacks repeated elements.
  • the repeated element is an Alu element or a LINE element.
  • the Cas9 system further comprises a gRNA.
  • the gRNA comprises a targeting domain, first and second complementary domains, and a proximal domain.
  • the gRNA is a chimeric gRNA.
  • the Cas9 system further comprises at least one additional Cas9 molecule.
  • the Cas9 molecule is an eaCas9 molecule.
  • the Cas9 molecule is an eiCas9 molecule.
  • Cas9 system comprises a Cas9 fusion molecule, comprising an eiCas9 molecule linked to a template binding domain, and a Cas9 molecule.
  • Cas9 system comprises a Cas9 fusion molecule, comprising an eiCas9 molecule linked to a template binding domain, and an eaCas9 molecule.
  • described herein is a cell, or a population of cells, comprising a Cas9 system described herein.
  • described herein is a cell, or a population of cells, comprising a Cas9 fusion molecule described herein.
  • described herein is a nucleic acid encoding a Cas9 fusion molecule described herein.
  • a vector comprising said nucleic acid.
  • the vector is an AAV vector.
  • described herein is a cell, or a population of cells, comprising said nucleic acid.
  • described herein is a method of altering a nucleic acid at a target position in a cell, or a population of cells, the method comprising contacting the cell with the Cas9 system described herein, wherein the gRNA molecule and Cas9 fusion molecule interact with the nucleic acid, resulting in a cleavage event, wherein the cleavage event is repaired by at least one DNA repair pathway, and wherein the sequence of the nucleic acid after the cleavage event is different than the sequence of the nucleic acid prior to the cleavage event, thereby altering the nucleic acid at the target position in the cell, or in the population of cells.
  • the method further comprises contacting the cell, or the population of cells, with a second gRNA molecule, wherein the second gRNA molecule and the Cas9 fusion molecule interact with the nucleic acid, resulting in a second cleavage event.
  • the at least one DNA repair pathway is selected from the group consisting of resection, mismatch repair (MMR), nucleotide excision repair (NER), base excision repair (BER), canonical non-homologous end joining (canonical NHEJ), alternative non-homologous end joining (ALT-NHEJ), canonical homology directed-repair (canonical HDR), alternative homology directed repair (ALT-HDR), microhomology-mediated end joining (MMEJ), Blunt End Joining, Synthesis Dependent Microhomology Mediated End Joining, single strand annealing (SSA), Holliday junction model or double strand break repair (DSBR), synthesis-dependent strand annealing (SDSA), single strand break repair (SSBR), translesion synthesis repair (TLS), and interstrand crosslink repair (ICL), and DNA/RNA processing.
  • MMR mismatch repair
  • NER nucleotide excision repair
  • BER base excision repair
  • canonical non-homologous end joining can
  • the at least one DNA repair pathway is canonical homology directed-repair (canonical HDR) or alternative homology directed repair (ALT-HDR).
  • the cleavage event comprises one or more single strand breaks, one or more double strand breaks, or a combination of single strand breaks and double strand breaks.
  • the gRNA molecule positions one cleavage event on each strand of the nucleic acid.
  • the gRNA molecule positions the cleavage event on a strand of the nucleic acid that binds to the gRNA molecule.
  • the second gRNA molecule positions the second cleavage event on a strand of the nucleic acid that binds to the second gRNA molecule.
  • the Cas9 fusion molecule makes a single strand break in the nucleic acid. In another embodiment, the Cas9 fusion molecule makes a double strand break in the nucleic acid. In one embodiment, the single strand break is made in the strand of the nucleic acid to which a targeting domain of the gRNA molecule is complementary. In another embodiment, the single strand break is made in the strand of the nucleic acid other than the strand to which a targeting domain of the gRNA is complementary.
  • the target position is in an HBB gene.
  • the cell, or the population of cells is a eukaryotic cell, or a population of eukaryotic cells. In one embodiment, the cell, or the population of cells, is a plant cell, or a population of plant cells. In one embodiment, the plant cell, or the population of plant cells, is a monocot plant cell, a dicot plant cell, a population of monocot plant cells, or a population of dicot plant cells.
  • the cell, or the population of cells is a mammalian cell, or a population of mammalian cells. In one embodiment, the cell, or the population of cells, is a human cell, or a population of human cells.
  • the cell, or the population of cells is a vertebrate, mammalian, rodent, goat, pig, bird, chicken, turkey, cow, horse, sheep, fish, primate, or human cell or population of cells.
  • the cell, or the population of cells is a somatic cell, a germ cell, or a prenatal cell or population of cells.
  • the cell, or the population of cells is a zygotic cell, a blastocyst, an embryonic cell, a stem cell, a mitotically competent cell, a meiotically competent cell or population of cells.
  • the cell, or the population of cells is a T cell, a CD8+ T cell, a CD8+ na ⁇ ve T cell, a central memory T cell, an effector memory T cell, a CD4+ T cell, a stem cell memory T cell, a helper T cell, a regulatory T cell, a cytotoxic T cell, a natural killer T cell, a Hematopoietic Stem Cell, a long term hematopoietic stem cell, a short term hematopoietic stem cell, a multipotent progenitor cell, a lineage restricted progenitor cell, a lymphoid progenitor cell, a myeloid progenitor cell, a common myeloid progenitor cell, an erythroid progenitor cell, a megakaryocyte erythroid progenitor cell, a monocytic precursor cell, an endocrine precursor cell, an exocrine cell, a fibroblast, a
  • a progenitor B cell a Pre B cell, a Pro B cell, a memory B cell, a plasma B cell, a gastrointestinal epithelial cell, a biliary epithelial cell, a pancreatic ductal epithelial cell, an intestinal stem cell, a hepatocyte, a liver stellate cell, a Kupffer cell, an osteoblast, an osteoclast, an adipocyte, a preadipocyte, a pancreatic precursor cell, a pancreatic islet cell, a pancreatic beta cell, a pancreatic alpha cell, a pancreatic delta cell, a pancreatic exocrine cell, a Schwann cell, or an oligodendrocyte, or population of such cells.
  • the cell, or population of cells is from a subject suffering from a disease or disorder.
  • the disease is a blood disease, an immune disease, a neurological disease, a cancer, an infectious disease, a genetic disease, a disorder caused by aberrant mtDNA, a metabolic disease, a disorder caused by aberrant cell cycle, a disorder caused by aberrant angiogenesis, a disorder cause by aberrant DNA damage repair, or a pain disorder.
  • the cell, or population of cells is from a subject having at least one mutation at the target position.
  • the method further comprises isolating the cell, or population of cells, from the subject prior to contacting the cell, or population of cells, with the Cas9 system described herein.
  • the method further comprises introducing the cell, or the population of cells, into a subject after contacting the cell, or the population of cells, with the Cas9 system described herein.
  • the contacting the cell, or the population of cells, with the Cas9 system described herein is performed ex vivo. In another embodiment, the contacting the cell, or the population of cells, with the Cas9 system described herein is performed in vivo. In one embodiment, the contacting the cell, or the population of cells, with the Cas9 system described herein is performed in vitro.
  • the method further comprises sequencing the nucleic acid, or a portion of the nucleic acid, prior to contacting the cell, or the population of cells, with the Cas9 system described herein.
  • the method further comprises sequencing the nucleic acid, or a portion of the nucleic acid, after the cleavage event.
  • the cell, or the population of cells is contacted with the gRNA molecule and the Cas9 fusion molecule as a pre-formed complex.
  • described herein is a cell, or a population of cells, altered by the methods described herein.
  • described herein is a pharmaceutical composition comprising said cell, or said population of cells.
  • described herein is a pharmaceutical composition comprising a Cas9 system described herein.
  • described herein is a method of treating a subject comprising administering to the subject the cell, or the population of cells, altered by the methods described herein, or a pharmaceutical composition comprising said cell, or said population of cells.
  • described herein is a method of treating a subject suffering from a disease or disorder, the method comprising contacting a cell, or a population of cells, from the subject with the Cas9 system described herein, wherein the gRNA molecule and the Cas9 fusion molecule interact with a nucleic acid at a target position, resulting in a cleavage event, wherein the cleavage event is repaired by at least one DNA repair pathway, and wherein the sequence of the nucleic acid after the cleavage event is different than the sequence of the nucleic acid prior to the cleavage event, thereby treating the subject suffering from the disease or disorder.
  • the method further comprises contacting the cell from the subject with a second gRNA molecule, wherein the second gRNA molecule and the Cas9 fusion molecule interact with the nucleic acid, resulting in a second cleavage event.
  • the contacting the cell is performed ex vivo. In another embodiment, the contacting the cell is performed in vivo.
  • the present disclosure provides a cell comprising:
  • template binding domain comprises specific affinity for a template binding domain partner and lacks substantial affinity for:
  • the endogenous organellar nucleic acid e.g., mitochondrial, chloroplast, or both, of said cell.
  • the present disclosure also provides a cell comprising:
  • said template binding domain comprises specific affinity for a template binding domain partner and lacks substantial affinity for a nucleic acid sequence within 100, 500, 1000, 2000, 5,000, or 10,000 nucleotides of said target sequence.
  • the present disclosure also provides a kit comprising packaging and at least three of:
  • (a) and (b) form part of a single composition; (a) and (c) form part of a single composition; (a) and (d) form part of a single composition; (b) and (c) form part of a single composition; (b) and (d) form part of a single composition; (c) and (d) form part of a single composition; (a), (b), and (c) form part of a single composition; (a), (b), and (d) form part of a single composition; (a), (c), and (d) form part of a single composition; (b), (c), and (d) form part of a single composition; or (a), (b), (c), and (d) form part of a single composition.
  • the present disclosure also provides a reaction mixture comprising the cell and a solution.
  • the present disclosure also provides a reaction mixture wherein the solution is a cell growth medium.
  • the present disclosure provides a method of altering the structure of a cell, e.g., altering the sequence, of a target nucleic acid of the cell, comprising contacting the cell with the Cas9 system as described herein under conditions that allow for alteration of the structure of the cell, thereby altering the structure of a cell, e.g., altering the sequence of a target nucleic acid.
  • the method comprises bringing a template nucleic acid in proximity with a target nucleic acid in a cell, e.g., sufficient proximity that the efficiency of altering the cell, e.g., a target nucleic acid of the cell, is increased over the level seen with a non bound template nucleic acid.
  • the target nucleic acid is cleaved at a position that is:
  • the target nucleic acid is cleaved at two positions that are:
  • FIG. 1 illustrates embodiments where a Cas9 molecule is engineered to selectively associate with a template nucleic acid, and therefore increase the proximity of the template nucleic acid to a target nucleic acid.
  • the Cas9 molecule (which can be, e.g., wild-type or a nickase) is shown associated with a target nucleic acid, e.g., a mutant gene.
  • the Cas9 molecule is associated with a gRNA that targets the Cas9 molecule to the target nucleic acid.
  • the target nucleic acid comprises a target sequence which is recognized by a gRNA, as well as a PAM sequence that is recognized by the Cas9 molecule.
  • the Cas9 molecule is shown linked to a template binding domain (illustrated as a trapezoid) using a linker region (illustrated as a line connecting the Cas9 molecule and the template binding domain).
  • the template binding domain (which can be a DNA binding domain) has affinity for a template binding domain partner (which can be a DNA sequence specifically recognized by the DNA binding domain) which is shown as a shaded rectangle.
  • the template binding domain partner is part of a circular double stranded nucleic acid molecule.
  • the circular nucleic acid molecule also comprises a template nucleic acid which is capable of participating in homology-dependent repair of a break in the target nucleic acid.
  • the figure also shows alternative nucleic acid template systems, one which is a double stranded linear nucleic acid, and one which is a single stranded linear nucleic acid.
  • FIG. 2 depicts exemplary nucleic acid template systems.
  • the template binding domain partner e.g., a stretch of DNA that a DNA-binding domain can bind
  • the template nucleic acid is illustrated as a single line which represents single-stranded DNA, or a double line which represents double stranded DNA.
  • the figure illustrates that the template nucleic acid system can be a circular double stranded DNA molecule like a plasmid; a linear double stranded DNA molecule, or a linear DNA molecule that is single stranded in the template nucleic acid region and double stranded in the template binding domain partner region.
  • the template binding domain partner can be, e.g., on either end of the molecule.
  • FIG. 3 depicts exemplary arrangements of linear nucleic acid template systems.
  • the target nucleic acid (a long double stranded DNA segment like a gene or chromosome) is depicted as a double stranded shaded line with a break.
  • the regions of the target nucleic acid that are homologous to a template nucleic acid are shown as thick solid or dotted lines.
  • the Cas9 molecule is depicted as a shaded circle positioned over the break, and the template binding domain extends from the Cas9 molecule.
  • the nucleic acid template systems are shown below the target nucleic acids, with the template binding domain partner shown as a shaded box and the template nucleic acid as a dotted or solid line.
  • the nucleic acid template system can be double stranded, single stranded, or have a single stranded portion and a double stranded portion (e.g., where the template binding domain partner is double stranded and the template nucleic acid is single stranded).
  • the top row shows embodiments where the template nucleic acid has homology on either side of the cut, e.g., approximately 50 to 100 bp or nucleotides both on the left and on the right side of the cut.
  • the homology regions to the left are shown as solid lines and the homology regions to the right are shown as dotted lines.
  • the central row shows embodiments where the template nucleic acid has the majority of the homology on the right side of the cut, e.g., approximately 150-200 bp or nucleotides of homology.
  • the bottom row shows embodiments where the template nucleic acid has the majority of the homology on the left side of the cut, e.g., approximately 150-200 bp or nucleotides of homology.
  • the template binding domain partner can be placed at either end of the nucleic acid template system; in the left column the template binding domain partner is placed at the right of the nucleic acid template system, and in the right column the template binding domain partner is placed at the left of the nucleic acid template system.
  • FIG. 4 depicts exemplary arrangements of circular nucleic acid template systems.
  • the target nucleic acid (a long double stranded DNA segment like a gene or chromosome) is depicted as a double stranded shaded line with a break.
  • the regions of the target nucleic acid that are homologous to a template nucleic acid are shown as thick solid or dotted lines.
  • the Cas9 molecule is depicted as a shaded circle positioned over the break, and the template binding domain extends from the Cas9 molecule.
  • the nucleic acid template systems are shown below the target nucleic acids, with the template binding domain partner is shown as a shaded box and the template nucleic acid as a dotted or solid line.
  • the circular nucleic acid template systems are double stranded DNA, but it is understood that the disclosure also contemplates circular molecules that are partly or whole single stranded.
  • the top row shows embodiments where the template nucleic acid has a homology region to the right of the template binding domain partner.
  • the center row shows embodiments where the template nucleic acid has a homology region to the left of the template binding domain partner.
  • the bottom row shows embodiments where the template nucleic acid has a homology region on the opposite side of the circular DNA molecule from the template binding domain partner.
  • the homology region on the template nucleic acid can corresponds to a region on the target nucleic acid, where a solid line is homologous with a solid line and a dotted line is homologous with a dotted line.
  • the template nucleic acid has homology to the target nucleic acid on either side of the break.
  • the template nucleic acid has homology to the target nucleic acid on the left side of the break.
  • the template nucleic acid has homology to the target nucleic acid on the right side of the break.
  • the homology region can be, e.g., 500 to 2000 bp.
  • FIGS. 5A and 5B are schematic representations of the domain organization of S. pyogenes Cas9.
  • FIG. 5A shows the organization of the Cas9 domains, including amino acid positions, in reference to the two lobes of Cas9 (recognition (REC) and nuclease (NUC) lobes).
  • FIG. 5B shows the percent homology of each domain across 83 Cas9 orthologs.
  • FIG. 6 is a schematic drawing of a Cas9 fusion molecule, comprising a Cas9 molecule fused to a template binding domain, bound to a target nucleic acid, and complexed with a nucleic acid template system comprising a template binding domain partner and a template nucleic acid.
  • FIG. 7 is a schematic drawing of a Cas9 system comprising an eaCas9 molecule bound to a target nucleic acid, and a Cas9 fusion molecule, comprising an eiCas9 molecule fused to a template binding domain, bound to a target nucleic acid, and complexed with a nucleic acid template system comprising a template binding domain partner and a template nucleic acid.
  • the Cas9 fusion molecule positions the nucleic acid template system in close proximity to the eaCas9 molecule.
  • FIG. 8A illustrates four exemplary ssDNA nucleic acid template systems comprising a template binding domain partner, comprising three Tet Operator sequences, and a template nucleic acid.
  • V1.1 is an exemplary ssDNA nucleic acid template system comprising a template binding domain partner, comprising three Tet Operator sequences separated by two 17 nucleotide spacers, N terminal of a 109 nucleotide template nucleic acid.
  • V1.2 is an exemplary ssDNA nucleic acid template system comprising a template binding domain partner, comprising three Tet Operator sequences separated by two 17 nucleotide spacers, C terminal of a 109 nucleotide template nucleic acid.
  • V2 is an exemplary ssDNA nucleic acid template system comprising a template binding domain partner, comprising three Tet Operator sequences separated by two 8 nucleotide spacers, N terminal of a 109 nucleotide template nucleic acid.
  • V3 is an exemplary ssDNA nucleic acid template system comprising a template binding domain partner, comprising three Tet Operator sequences adjacent to one another without intervening spacers, N terminal of a 109 nucleotide template nucleic acid.
  • FIG. 8B shows Tet Repressor protein binding activity for four exemplary ssDNA nucleic acid template systems comprising a template binding domain partner, comprising three Tet Operator sequences, and a template nucleic acid (i.e., V1.1, V1.2, V2, and V3) and two control DNA templates (i.e., ssODN 179, and ssODN109.
  • An electrophoretic mobility shift assay using a 10% polyacrylamide gel was performed using 500 nM of recombinant Tet Repressor protein 50 nM of ssDNA nucleic acid template system or control DNA template.
  • FIG. 9 illustrates four exemplary ssDNA nucleic acid template systems comprising a template binding domain partner, comprising three or four Tet Operator sequences, and a template nucleic acid.
  • V4.1 is an exemplary ssDNA nucleic acid template system comprising a template binding domain partner, comprising three Tet Operator sequences separated by three 8 nucleotide spacers, N terminal of a 119 nucleotide template nucleic acid.
  • V4.2 is an exemplary ssDNA nucleic acid template system comprising a template binding domain partner, comprising three Tet Operator sequences separated by three 8 nucleotide spacers, C terminal of a 119 nucleotide template nucleic acid.
  • V5.1 is an exemplary ssDNA nucleic acid template system comprising a template binding domain partner, comprising four Tet Operator sequences separated by three 8 nucleotide spacers, N terminal of a 100 nucleotide template nucleic acid.
  • V5.2 is an exemplary ssDNA nucleic acid template system comprising a template binding domain partner, comprising four Tet Operator sequences separated by three 8 nucleotide spacers, C terminal of a 100 nucleotide template nucleic acid.
  • amino acids as used herein encompasses the canonical amino acids as well as analogs thereof.
  • amino acid residues that flank a deletion refers to the amino acid residue that immediately precedes the deletion and the amino acid residue that immediately follows the deletion.
  • the flanking amino acid residues are, CT 3 and CT 7.
  • Cas9 core domain refers to a polypeptide that does not include a functional PI domain, e.g., a polypeptide not having an endogenous PI domain, e.g., wherein the endogenous PI domain is deleted (deleted, as used in this context, refers merely to a sequence difference or the absence of amino acid residues and implies no process or origin limitation), or generally, a Cas9 molecule lacking a PI domain.
  • a Cas9 core domain comprises a REC1 domain, a REC2 domain, a BH domain, a RuvC domain, and an HNH domain.
  • a Cas9 core domain, together with an altered PI domain comprises a functional Cas9 molecule.
  • Cas9 fusion molecule refers to a chimeric protein comprising a Cas9 molecule, e.g., Cas9 protein or Cas9 polypeptide, or a fragment thereof, linked to a template binding domain.
  • the template binding domain is a protein or polypeptide.
  • the template binding domain is a nucleic acid, e.g., DNA or RNA.
  • the template binding domain is a small molecule.
  • the Cas9 fusion molecule comprises a Cas9 molecule covalently linked to the template binding domain.
  • the Cas9 fusion protein may be a chimeric protein comprising one or more Cas9 protein domains and a DNA binding domain from a protein disclosed herein.
  • the Cas9 fusion molecule comprises a Cas9 molecule noncovalently linked to the template binding domain.
  • a species X Cas9 core domain has at least 20, 30, 40, 50, 60, 70, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% homology with the corresponding sequence of a reference sequence, e.g., a naturally occurring species X Cas9 core domain, e.g., from a Cas9 core domain from Table 100.
  • each of a REC1 domain, a REC2 domain, a BH domain, a RuvC domain, and/or an HNH domain of a species X Cas9 core domain has, independently, at least 20, 30, 40, 50, 60, 70, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% homology with the corresponding sequence of a reference sequence, e.g., a naturally occurring species X Cas9 core domain, e.g., from a Cas9 core domain from Table 100.
  • a reference sequence e.g., a naturally occurring species X Cas9 core domain, e.g., from a Cas9 core domain from Table 100.
  • Cas9 molecule or “Cas9 polypeptide”, as that term is used herein, refers to a polypeptide that can bind (1) a PAM (a protospacer adjacent motif) in a nucleic acid, and (2) a guide RNA (gRNA) molecule.
  • gRNA guide RNA
  • a Cas9 molecule or Cas9 polypeptide in concert with the gRNA molecule, can localize to a site which comprises a target domain.
  • Cas9 may be a nuclease (an enzyme that cleaves both strands of a double-stranded nucleic acid), a nickase (an enzyme that cleaves one strand of a double-stranded nucleic acid), or an enzymatically inactive (or dead) molecule.
  • a Cas9 molecule having nuclease or nickase activity is referred to as an enzymatically active Cas9 molecule (an eaCas9 molecule).
  • a Cas9 molecule lacking the ability to cleave target nucleic acid is referred to as an enzymatically inactive Cas9 molecule (an eiCas9 molecule).
  • a Cas9 molecule can have the amino acid sequence of a naturally occurring Cas9 molecule or can be an altered, engineered or modified Cas9 molecule, which differs by at least one amino acid residue, from a reference sequence, e.g., the most similar naturally occurring Cas9 molecule, e.g., a Cas9 molecule from Table 100.
  • an altered, engineered or modified Cas9 molecule can comprise one or more point mutations which alter (e.g., increase, decrease and/or eliminate), one or more Cas9 molecule activities, e.g., a nuclease activity.
  • a Cas9 molecule meets one or both of the following criteria:
  • the Cas9 molecule may be a Cas9 deletion, e.g., the Cas9 may comprise a deletion in one or more of the following domains: a REC2, REC1 CT , or REC1 SUB domain. Except for any REC deletion, a Cas9 molecule meets one or both of the following criteria:
  • a reference sequence e.g., naturally occurring Cas9 molecule, e.g., a Cas9 molecule described in Table 100 herein.
  • Homology except for any REC deletion is determined as follows: a sequence having a deletion is altered by replacing the deleted sequence with the corresponding sequence from the reference sequence, and the altered sequence is compared with the reference sequence.
  • the Cas9 molecule may be a Cas9 variant, e.g., the Cas9 may comprise an altered PI domain, or other modified amino acid sequence, or the Cas9 may comprise a linker.
  • a Cas9 molecule meets one or both of the following criteria:
  • a sequence having an altered PI domain (or other modified amino acid sequence) is altered by restoring the altered PI domain (or other modified amino acid sequence) to the naturally occurring PI domain (or other naturally occurring sequence) from the reference sequence, and the thus altered sequence is compared with the reference sequence.
  • a Cas9 molecule meets one or both of the following criteria:
  • each domain of the Cas9 molecule (e.g., the domains named in FIG. 5 or elsewhere herein), including any remaining portion of a REC2, REC1 CT , or REC1 SUB domain having a deletion or an unaltered portion of a PI domain, will, independently have:
  • At least 1, 2, 3, 4, 5, of 6 domains will have, independently, at least 50, 60, 70, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% homology with a corresponding domain, while any remaining domains will be absent, or have less homology to their corresponding naturally occurring domains.
  • PI domain refers to the region of a Cas9 molecule that interacts with the PAM sequence of a target nucleic acid.
  • altered PI domain refers to a PI domain other than the native or endogenous PI domain associated with the naturally occurring Cas9 molecule.
  • a Cas9 molecule comprises an altered PI domain if its PI domain is other than the PI domain naturally associated with the Cas9 core domain of the Cas9 molecule, or if its PI domain is not a naturally occurring PI domain associated with any Cas9 molecule. (Derived, as used in this sense, is not limited to physical derivation or even derivation from a specific source, and does not require a process limitation, but in some embodiments, includes mere structural similarity).
  • An altered PI domain may have less than 99, 98, 97, 96, 95, 94, 93, 92, 91, 90, 89, 88, 87, 86, 85, 84, 83, 82, 81, 80, 70, 60, 50, 30, 40, 30, 20, or 10% homology with the native or endogenous PI domain of a subject naturally occurring Cas9 molecule from which the Cas9 core domain is derived.
  • An altered PI domain may have a different RKR motif (the PAM recognition sequence) than that of the native or endogenous PI domain of the Cas9 species that supplies the Cas9 core domain.
  • the RKR motif of an altered PI domain may differ from the RKR motif of the native or endogenous PI domain of the Cas9 core domain by 1, 2, or 3 residues.
  • the RKR motif of the altered PI differs at the first position, the second position, the third position, the first and second positions, the first and third positions, the second and third positions, or all three positions, from the RKR motif of the PI endogenous to or naturally associated with the Cas9 core domain.
  • an altered PI domain is one having greater homology with the PI domain of a reference or donor naturally occurring Cas9 molecule (a heterologous Cas9) that with the native PI domain of a subject Cas9.
  • identity refers to sequence identity between two amino acid sequences or two nucleic acid sequences, with identity being a more strict comparison.
  • percent identity or homology and “% identity or homology” refer to the percentage of sequence identity found in a comparison of two or more amino acid sequences or nucleic acid sequences. Two or more sequences can be anywhere from 0-100% identical, or any value there between. Identity can be determined by comparing a position in each sequence that can be aligned for purposes of comparison to a reference sequence. When a position in the compared sequence is occupied by the same nucleotide base or amino acid, then the molecules are identical at that position.
  • a degree of identity of amino acid sequences is a function of the number of identical amino acids at positions shared by the amino acid sequences.
  • a degree of identity between nucleic acid sequences is a function of the number of identical or matching nucleotides at positions shared by the nucleic acid sequences.
  • a degree of homology of amino acid sequences is a function of the number of amino acids at positions shared by the polypeptide sequences.
  • Calculations of homology or sequence identity between two sequences are performed as follows.
  • the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in one or both of a first and a second amino acid or nucleic acid sequence for optimal alignment and non-homologous sequences can be disregarded for comparison purposes).
  • the optimal alignment is determined as the best score using the GAP program in the GCG software package with a Blossum 62 scoring matrix with a gap penalty of 12, a gap extend penalty of 4, and a frame shift gap penalty of 5.
  • the amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared.
  • Linker refers to a molecular entity that may link a Cas9 molecule, or a fragment thereof, and a template binding domain, or may link a template binding domain partner to a template nucleic acid, or may link a Cas9 molecule fragment or domain to another Cas9 molecule fragment or domain.
  • the linker is a nucleic acid, e.g. an oligonucleotide.
  • the linker is a small molecule.
  • the linker is a polypeptide comprising at least one amino acid.
  • a polypeptide linker may be disposed between sequences or domains of a Cas9 molecule.
  • the linker is disposed between the amino acid residues that flank a deletion. In an embodiment, the linker is disposed between the amino acid residues of a Cas9 core domain and an altered PI domain.
  • the linker is located immediately C-terminal to the amino acid residue CT 3 and immediately N-terminal to the amino acid residue CT 7.
  • the linker is selected such that the Cas9 molecule exhibits a tertiary structure or folded conformation similar to that of the corresponding naturally occurring Cas9 molecule, such that some Cas9 activity is retained. Suitable linkers are described herein.
  • the linker comprises a combination of Gly and Ser residues, e.g., (GS) x or (GGS) x , where x is 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10.
  • the linker comprises a linker comprising the amino acid sequence SGSETPGTSESATPES (SEQ ID NO: _), referred to herein as XTEN linker or XTEN.
  • Alternative linkers include (GSAGSAAGSGEF) x , wherein x is 1, 2, 3 or 4 (SEQ ID NO: _) and (SIVAQLSRPDPA) x , wherein x is 1, 2, 3 or 4 (SEQ ID NO: _).
  • Linkers also include a combination of linkers described herein or known in the art.
  • REC deletion refers to a REC2 deletion, a REC1 CT deletion, or a REC1 SUB deletion.
  • REC2 deletion refers to a deletion of at least 10% of the amino acid residues of the REC2 domain.
  • REC2 domain refers to a region, in the N terminal half of a naturally occurring Cas9 molecule that is not needed for cleavage or gRNA-mediated targeting. Its length and boundaries differ between Cas9 molecules from various species.
  • the REC2 domain is about 41 amino acid residues in length and corresponds, approximately, to residues 126 to 166, of S. aureus Cas9.
  • the REC2 domain is about 139 amino acid residues in length and corresponds, approximately, to residues 176 to 314 of S. pyogenes Cas9.
  • C the case of C.
  • the REC2 domain is about 45 amino acid residues in length and corresponds, approximately, to residues 137 to 181 of C. jejuni Cas9. These, and the approximate sizes and boundaries of REC2 domains from other species, are provided in Table 100.
  • REC1 CT deletion refers to a deletion of at least 10% of the amino acid residues of the REC1 CT domain.
  • REC1 CT domain refers to a region, C terminal of the REC1 domain, of a naturally occurring Cas9 polypeptide that is not needed for cleavage or gRNA-mediated targeting. Its length and boundaries differ between Cas9 proteins from various species.
  • the REC1 CT domain is about 146 amino acid residues in length and corresponds, approximately, to residues 288 to 166, of S. aureus Cas9.
  • the REC1 CT domain is about 219 amino acid residues in length and corresponds, approximately, to residues 500 to 718 of S. pyogenes Cas9.
  • the REC1 CT domain is about 134 amino acid residues in length and corresponds, approximately, to residues 305 to 438 of C. jejuni Cas9. These, and the approximate sizes and boundaries of REC1 CT domains from other species, are provided in Table 100.
  • REC1 SUB deletion refers to a deletion of at least 10% of the amino acid residues of the REC1 SUB domain.
  • REC1 SUB domain refers to a region, located within the REC1 CT domain, of a naturally occurring Cas9 polypeptide that is not needed for cleavage or gRNA-mediated targeting. Its length and boundaries differ between Cas9 proteins from various species.
  • the REC1 SUB domain is about 57 amino acid residues in length and corresponds, approximately, to residues 296 to 352, of S. aureus Cas9.
  • the REC1 SUB domain is about 82 amino acid residues in length and corresponds, approximately, to residues 511 to 592 of S. pyogenes Cas9.
  • C the case of C.
  • the REC1 SUB domain is about 45 amino acid residues in length and corresponds, approximately, to residues 316 to 360 of C. jejuni Cas9. These, and the approximate sizes and boundaries of REC1 SUB domains from other species, are provided in Table 100.
  • n refers to the number of amino acid residues that are deleted in a REC2, REC1 CT , or REC1 SUB deletion, unless otherwise specified.
  • X as used herein in the context of an amino acid sequence of a linker sequence, refers to any number of repeating units unless otherwise specified.
  • a disorder “caused by” a mutation refers to a disorder that is made more likely or severe by the presence of the mutation, compared to a subject that does not have the mutation.
  • the mutation need not be the only cause of a disorder, i.e., the disorder can still be caused by the mutation even if other causes, such as environmental factors or lifestyle factors, contribute causally to the disorder.
  • the disorder is caused by the mutation if the mutation is a medically recognized risk factor for developing the disorder, and/or if a study has found that the mutation correlates with development of the disorder.
  • “Derived from”, as used herein, refers to the source or origin of a molecular entity, e.g., a nucleic acid or protein.
  • the source of a molecular entity may be naturally-occurring, recombinant, unpurified, or a purified molecular entity.
  • a polypeptide that is derived from a second polypeptide comprises an amino acid sequence that is identical or substantially similar, e.g., is more than 50% homologous to, the amino acid sequence of the second protein.
  • the derived molecular entity, e.g., a nucleic acid or protein can comprise one or more modifications, e.g., one or more amino acid or nucleotide changes.
  • Domain is used to describe segments of a protein or nucleic acid. Unless otherwise indicated, a domain is not required to have any specific functional property.
  • HDR homology-directed repair
  • a homologous nucleic acid e.g., a sister chromatid or an exogenous nucleic acid.
  • HDR typically involves a series of steps such as recognition of the break, stabilization of the break, resection, stabilization of single stranded DNA, formation of a DNA crossover intermediate, resolution of the crossover intermediate, and ligation.
  • Large molecule refers to a molecule having a molecular weight of at least 2, 3, 5, 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100 kDa. Large molecules include proteins, polypeptides, nucleic acids, biologics, and carbohydrates.
  • Polypeptide refers to a polymer of amino acids.
  • Reference molecule refers to a molecule to which a subject molecule, e.g., a subject Cas9 molecule or a subject gRNA molecule, e.g., a modified or candidate Cas9 molecule, is compared.
  • a Cas9 molecule can be characterized as having no more than 10% of the nuclease activity of a reference Cas9 molecule.
  • reference Cas9 molecules include naturally occurring unmodified Cas9 molecules, e.g., a naturally occurring Cas9 molecule such as a Cas9 molecule of S. aureus, S. pyogenes , or S.
  • the reference Cas9 molecule is the naturally occurring Cas9 molecule having the closest sequence identity or homology with the Cas9 molecule to which it is being compared.
  • the reference Cas9 molecule is a sequence, e.g., a naturally occurring or known sequence, which is the parental form on which a change, e.g., a mutation has been made.
  • Replacement or “replaced”, as used herein with reference to a modification of a molecule does not require a process limitation but merely indicates that the replacement entity is present.
  • “Small molecule”, as used herein, refers to a compound having a molecular weight less than about 2 kDa, e.g., less than about 2 kDa, less than about 1.5 kDa, less than about 1 kDa, or less than about 0.75 kDa.
  • Step interference or “steric hindrance”, as used herein, refers to the restriction or prevention of the binding or interaction of one molecular entity (e.g., a protein or a protein fragment) with another molecular entity (e.g., a nucleic acid or a protein).
  • one molecular entity e.g., a protein or a protein fragment
  • another molecular entity e.g., a nucleic acid or a protein
  • Subject may mean either a human or non-human animal.
  • the term includes, but is not limited to, mammals (e.g., humans, other primates, pigs, rodents (e.g., mice and rats or hamsters), rabbits, guinea pigs, cows, horses, cats, dogs, sheep, and goats.
  • the subject is a human. In other embodiments, the subject is poultry.
  • the linker is at least 6, but no longer than 60 amino acids in length.
  • the linker is at least 18, but no longer than 180 nucleotides in length.
  • the linker is at least 10, 50, 100, 200, 500, 1000, 2000, 5000, or 10000 Angstroms in length.
  • the linker is no more than 10, 50, 100, 200, 500, 1000, 2000, 5000, or 10000 Angstroms in length.
  • Treatment mean the treatment of a disease in a mammal, e.g., in a human, including (a) inhibiting the disease, i.e., arresting or preventing its development; (b) relieving the disease, i.e., causing regression of the disease state; and (c) curing the disease.
  • Prevent means the prevention of a disease in a subject, e.g., a mammal, e.g., in a human, including (a) avoiding or precluding the disease; (2) affecting the predisposition toward the disease, e.g., preventing at least one symptom of the disease or to delay onset of at least one symptom of the disease.
  • Specific affinity refers to a binding interaction between two or more molecular entities, e.g., a template binding domain and a template binding domain partner, such as, for example, a DNA-binding polypeptide and a DNA molecule, wherein one molecular entity preferentially binds to another molecular entity, but does not substantially bind to other molecular entities present in a molecular milieu, e.g., a heterologous molecular milieu, in a cell or in solution.
  • the term “specific affinity” refers to a DNA-binding protein or polypeptide that binds to a specific sequence of a nucleic acid molecule.
  • Exemplary protein-protein pairs that specifically bind to each other are provided in Table V.6.
  • Exemplary protein-small molecule pairs that specifically bind to each other are provided in Table V.7.
  • Exemplary protein-DNA sequence pairs that specifically bind to each other are provided in Table V.1.
  • “Substantial affinity”, as the term is used herein, refers to a binding interaction between two or more molecular entities, e.g., a template binding domain and a template binding domain partner, such as, for example, a DNA-binding polypeptide and a DNA molecule, wherein the binding event induces a significant event, change or alteration in the molecular entity being bound, or a significant phenotypic change in a cell in which the binding event occurs.
  • Target position refers to a site on a nucleic acid (e.g., a region of a chromosome) that is modified by a Cas9 molecule-dependent process.
  • the target position can be modified by a Cas9 molecule-mediated cleavage of the nucleic acid using a template nucleic acid.
  • a target position can be a site between two nucleotides, e.g., adjacent nucleotides, on the target nucleic acid into which one or more nucleotides is added.
  • the target position may comprise one or more nucleotides that are altered, e.g., corrected, by a template nucleic acid.
  • the target position is within a “target sequence” (e.g., the sequence to which the gRNA binds). In an embodiment, a target position is upstream or downstream of a target sequence (e.g., the sequence to which the gRNA binds).
  • the “targeting domain” of the gRNA is complementary to the “target domain” on the target nucleic acid.
  • template binding domain partner is a molecule with specific affinity for a template binding domain.
  • the template binding domain partner may be, e.g., DNA, protein, or a small molecule.
  • the target nucleic acid is modified to have some or all of the sequence of the template nucleic acid, typically at or near cleavage site(s).
  • the template nucleic acid is single stranded. In an alternate embodiment, the template nucleic acid is double stranded.
  • the template nucleic acid is DNA, e.g., double stranded DNA. In an alternate embodiment, the template nucleic acid is single stranded DNA. In an embodiment, the template nucleic acid is encoded on the same vector backbone, e.g. AAV genome, plasmid DNA, as the Cas9 and gRNA. In an embodiment, the template nucleic acid is excised from a vector backbone in vivo, e.g., it is flanked by gRNA recognition sequences.
  • Wild type refers to a gene or polypeptide which has the characteristics, e.g., the nucleotide or amino acid sequence, of a gene or polypeptide from a naturally-occurring source.
  • wild type typically includes the most frequent observation of a particular gene or polypeptide in a population of organisms found in nature.
  • X as used herein in the context of an amino acid sequence, refers to any amino acid (e.g., any of the twenty natural amino acids) unless otherwise specified.
  • template binding domain refers to an entity which, by virtue of its specific affinity for a template binding domain partner, mediates the association of a template nucleic acid with a Cas9 with which the template binding domain is associated.
  • the template binding domain associates with, e.g., by non-covalent or covalent interactions, with a template binding domain partner.
  • the template binding domain partner is associated with, e.g., covalently or non-covalently bound to, the template nucleic acid.
  • the template binding domain comprises a polypeptide and the template binding domain partner comprises a nucleic acid.
  • the template binding domain does not cleave nucleic acid.
  • a gRNA molecule refers to a nucleic acid that promotes the specific targeting or homing of a gRNA molecule/Cas9 molecule complex to a target nucleic acid.
  • the nucleic acid will incorporate the functions or structure of both crRNA and tracrRNA, e.g., the functions of processed or mature crRNA and of processed or mature tracrRNA.
  • gRNA molecules can be unimolecular (having a single nucleic acid molecule, e.g., which incorporates both crRNA function or structure and the tracrRNA function or structure), sometimes referred to herein as “chimeric” gRNAs, or modular (comprising more than one, and typically two, separate nucleic acid molecules, e.g., where one incorporates the crRNA function or structure and the other incorporates the tracrRNA function or structure).
  • a gRNA molecule comprises a number of domains. The gRNA molecule domains are described in more detail below. Additional details on gRNAs are provided in Section I entitled “gRNA molecules” of PCT Application WO 2015/048577, the entire contents of which are expressly incorporated herein by reference.
  • a unimolecular, or chimeric, gRNA comprises, preferably from 5′ to 3′: a targeting domain (which is complementary to a target nucleic acid, and which is sometimes referred to as a spacer); a first complementarity domain; a linking domain; a second complementarity domain (which is complementary to the first complementarity domain); a proximal domain; and optionally, a tail domain.
  • the targeting domain, and first complementarity domain correspond functionally or structurally to elements of a crRNA, e.g., a mature or processed crRNA.
  • the second complementarity domain, proximal domain, and tail domain correspond functionally or structurally to elements of a tracrRNA, e.g., a processed or mature tracrRNA.
  • a modular gRNA comprises: a first strand (which corresponds to a crRNA) comprising, preferably from 5′ to 3′; a targeting domain (which is complementary to a target nucleic acid); and a first complementarity domain; and a second strand (which corresponds to a tracrRNA), comprising, preferably from 5′ to 3′: optionally, a 5′ extension domain; a second complementarity domain; a proximal domain; and optionally, a tail domain.
  • the targeting domain (which can also be referred to as a “spacer”) comprises a nucleotide sequence that is complementary, e.g., at least 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, or 99% complementary, e.g., fully complementary, to the target sequence on the target nucleic acid.
  • the targeting domain is part of an RNA molecule and will therefore comprise the base uracil (U), while any DNA encoding the gRNA molecule will comprise the base thymine (T).
  • the complementarity of the targeting domain with the target sequence contributes to specificity of the interaction of the gRNA molecule/Cas9 molecule complex with a target nucleic acid. It is understood that in a targeting domain and target sequence pair, the uracil bases in the targeting domain will pair with the adenine bases in the target sequence.
  • the target domain itself comprises in the 5′ to 3′ direction, an optional secondary domain, and a core domain.
  • the core domain is fully complementary with the target sequence.
  • the targeting domain is 5 to 50 nucleotides in length, e.g., 10 to 30, e.g., 16, 17, 18, 19, 20, 21, 22, 23, 24, 25 or 26, nucleotides in length.
  • the strand of the target nucleic acid with which the targeting domain is complementary is referred to herein as the complementary strand.
  • Some or all of the nucleotides of the targeting domain can have a modification, e.g., a modification found in Section XI herein.
  • the targeting domain is 16 nucleotides in length.
  • the targeting domain is 17 nucleotides in length.
  • the targeting domain is 18 nucleotides in length.
  • the targeting domain is 19 nucleotides in length.
  • the targeting domain is 20 nucleotides in length.
  • the targeting domain is 21 nucleotides in length.
  • the targeting domain is 22 nucleotides in length.
  • the targeting domain is 23 nucleotides in length.
  • the targeting domain is 24 nucleotides in length.
  • the targeting domain is 25 nucleotides in length.
  • the targeting domain is 26 nucleotides in length.
  • the targeting domain comprises 16 nucleotides.
  • the targeting domain comprises 17 nucleotides.
  • the targeting domain comprises 18 nucleotides.
  • the targeting domain comprises 19 nucleotides.
  • the targeting domain comprises 20 nucleotides.
  • the targeting domain comprises 21 nucleotides.
  • the targeting domain comprises 22 nucleotides.
  • the targeting domain comprises 23 nucleotides.
  • the targeting domain comprises 24 nucleotides.
  • the targeting domain comprises 25 nucleotides.
  • the targeting domain comprises 26 nucleotides.
  • the first complementarity domain is complementary with the second complementarity domain, and in an embodiment, has sufficient complementarity to the second complementarity domain to form a duplexed region under at least some physiological conditions.
  • the first complementarity domain is 5 to 30 nucleotides in length. In an embodiment, the first complementarity domain is 5 to 25 nucleotides in length. In an embodiment, the first complementary domain is 7 to 25 nucleotides in length. In an embodiment, the first complementary domain is 7 to 22 nucleotides in length. In an embodiment, the first complementary domain is 7 to 18 nucleotides in length. In an embodiment, the first complementary domain is 7 to 15 nucleotides in length. In an embodiment, the first complementary domain is 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25 nucleotides in length.
  • the first complementarity domain comprises 3 subdomains, which, in the 5′ to 3′ direction are: a 5′ subdomain, a central subdomain, and a 3′ subdomain.
  • the 5′ subdomain is 4 to 9, e.g., 4, 5, 6, 7, 8 or 9 nucleotides in length.
  • the central subdomain is 1, 2, or 3, e.g., 1, nucleotide in length.
  • the 3′ subdomain is 3 to 25, e.g., 4 to 22, 4 to 18, or 4 to 10, or 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25 nucleotides in length.
  • the first complementarity domain can share homology with, or be derived from, a naturally occurring first complementarity domain. In an embodiment, it has at least 50% homology with a first complementarity domain disclosed herein, e.g., an S. pyogenes, S. aureus or S. thermophilus , first complementarity domain.
  • nucleotides of the domain can have a modification, e.g., a modification found in Section XI herein.
  • a linking domain serves to link the first complementarity domain with the second complementarity domain of a unimolecular gRNA.
  • the linking domain can link the first and second complementarity domains covalently or non-covalently.
  • the linkage is covalent.
  • the linking domain covalently couples the first and second complementarity domains.
  • the linking domain is, or comprises, a covalent bond interposed between the first complementarity domain and the second complementarity domain.
  • the linking domain comprises one or more, e.g., 2, 3, 4, 5, 6, 7, 8, 9, or 10 nucleotides.
  • linking domains are suitable for use in unimolecular gRNA molecules.
  • Linking domains can consist of a covalent bond, or be as short as one or a few nucleotides, e.g., 1, 2, 3, 4, or 5 nucleotides in length.
  • a linking domain is 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, or 25 or more nucleotides in length.
  • a linking domain is 2 to 50, 2 to 40, 2 to 30, 2 to 20, 2 to 10, or 2 to 5 nucleotides in length.
  • a linking domain shares homology with, or is derived from, a naturally occurring sequence, e.g., the sequence of a tracrRNA that is 5′ to the second complementarity domain.
  • the linking domain has at least 50% homology with a linking domain disclosed herein.
  • nucleotides of the domain can have a modification, e.g., a modification found in Section XI herein.
  • a modular gRNA can comprise additional sequence, 5′ to the second complementarity domain, referred to herein as the 5′ extension domain.
  • the 5′ extension domain is, 2 to 10, 2 to 9, 2 to 8, 2 to 7, 2 to 6, 2 to 5, or 2 to 4, nucleotides in length.
  • the 5′ extension domain is 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more nucleotides in length.
  • the second complementarity domain is complementary with the first complementarity domain, and in an embodiment, has sufficient complementarity to the second complementarity domain to form a duplexed region under at least some physiological conditions.
  • the second complementarity domain can include sequence that lacks complementarity with the first complementarity domain, e.g., sequence that loops out from the duplexed region.
  • the second complementarity domain is 5 to 27 nucleotides in length. In an embodiment, it is longer than the first complementarity region. In an embodiment the second complementary domain is 7 to 27 nucleotides in length. In an embodiment, the second complementary domain is 7 to 25 nucleotides in length. In an embodiment, the second complementary domain is 7 to 20 nucleotides in length. In an embodiment, the second complementary domain is 7 to 17 nucleotides in length. In an embodiment, the complementary domain is 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25 or 26 nucleotides in length.
  • the second complementarity domain comprises 3 subdomains, which, in the 5′ to 3′ direction are: a 5′ subdomain, a central subdomain, and a 3′ subdomain.
  • the 5′ subdomain is 3 to 25, e.g., 4 to 22, 4 to 18, or 4 to 10, or 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25 nucleotides in length.
  • the central subdomain is 1, 2, 3, 4 or 5, e.g., 3, nucleotides in length.
  • the 3′ subdomain is 4 to 9, e.g., 4, 5, 6, 7, 8 or 9 nucleotides in length.
  • the 5′ subdomain and the 3′ subdomain of the first complementarity domain are respectively, complementary, e.g., fully complementary, with the 3′ subdomain and the 5′ subdomain of the second complementarity domain.
  • the second complementarity domain can share homology with or be derived from a naturally occurring second complementarity domain. In an embodiment, it has at least 50% homology with a second complementarity domain disclosed herein, e.g., an S. pyogenes, S. aureus or S. thermophilus , first complementarity domain.
  • nucleotides of the domain can have a modification, e.g., a modification found in Section XI herein.
  • the proximal domain is 5 to 20 nucleotides in length.
  • the proximal domain can share homology with or be derived from a naturally occurring proximal domain. In an embodiment, it has at least 50% homology with a proximal domain disclosed herein, e.g., an S. pyogenes, S. aureus or S. thermophilus , proximal domain.
  • nucleotides of the domain can have a modification, e.g., a modification found in Section XI herein.
  • tail domains are suitable for use in gRNA molecules.
  • the tail domain is 0 (absent), 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 nucleotides in length.
  • the tail domain nucleotides are from or share homology with sequence from the 5′ end of a naturally occurring tail domain.
  • the tail domain includes sequences that are complementary to each other and which, under at least some physiological conditions, form a duplexed region.
  • the tail domain is absent or is 1 to 50 nucleotides in length.
  • the tail domain can share homology with or be derived from a naturally occurring proximal tail domain. In an embodiment, it has at least 50% homology with a tail domain disclosed herein, e.g., an S. pyogenes, S. aureus or S. thermophilus , tail domain.
  • the tail domain includes nucleotides at the 3′ end that are related to the method of in vitro or in vivo transcription.
  • these nucleotides may be any nucleotides present before the 3′ end of the DNA template.
  • these nucleotides may be the sequence UUUUUU.
  • alternate pol-III promoters are used, these nucleotides may be various numbers or uracil bases or may include alternate bases.
  • gRNA molecules The domains of gRNA molecules are described in more detail below.
  • the “targeting domain” of the gRNA is complementary to the “target domain” on the target nucleic acid.
  • the strfnd of the target nucleic acid comprising the nucleotide sequence complementary to the core domain of the gRNA is referred to herein as the “complementary strand” of the target nucleic acid.
  • Guidance on the selection of targeting domains can be found, e.g., in Fu Y et al. (2014) N AT . B IOTECHNOL . 32: 279-84 (doi: 10.1038/nbt.2808) and Sternberg S H et al. (2014) N ATURE 507: 62-7 (doi: 10.1038/nature13011).
  • the targeting domain is 16, 17, 18, 19, 20, 21, 22, 23, 24, 25 or 26 nucleotides in length.
  • the targeting domain is 16 nucleotides in length.
  • the targeting domain is 17 nucleotides in length.
  • the targeting domain is 18 nucleotides in length.
  • the targeting domain is 19 nucleotides in length.
  • the targeting domain is 20 nucleotides in length.
  • the targeting domain is 21 nucleotides in length.
  • the targeting domain is 22 nucleotides in length.
  • the targeting domain is 23 nucleotides in length.
  • the targeting domain is 24 nucleotides in length.
  • the targeting domain is 25 nucleotides in length.
  • the targeting domain is 26 nucleotides in length.
  • the targeting domain comprises 16 nucleotides.
  • the targeting domain comprises 17 nucleotides.
  • the targeting domain comprises 18 nucleotides.
  • the targeting domain comprises 19 nucleotides.
  • the targeting domain comprises 20 nucleotides.
  • the targeting domain comprises 21 nucleotides.
  • the targeting domain comprises 22 nucleotides.
  • the targeting domain comprises 23 nucleotides.
  • the targeting domain comprises 24 nucleotides.
  • the targeting domain comprises 25 nucleotides.
  • the targeting domain comprises 26 nucleotides.
  • the targeting domain is 10+/ ⁇ 5, 20+/ ⁇ 5, 30+/ ⁇ 5, 40+/ ⁇ 5, 50+/ ⁇ 5, 60+/ ⁇ 5, 70+/ ⁇ 5, 80+/ ⁇ 5, 90+/ ⁇ 5, or 100+/ ⁇ 5 nucleotides, in length.
  • the targeting domain is 20+/ ⁇ 5 nucleotides in length.
  • the targeting domain is 20+/ ⁇ 10, 30+/ ⁇ 10, 40+/ ⁇ 10, 50+/ ⁇ 10, 60+/ ⁇ 10, 70+/ ⁇ 10, 80+/ ⁇ 10, 90+/ ⁇ 10, or 100+/ ⁇ 10 nucleotides, in length.
  • the targeting domain is 30+/ ⁇ 10 nucleotides in length.
  • the targeting domain is 10 to 100, 10 to 90, 10 to 80, 10 to 70, 10 to 60, 10 to 50, 10 to 40, 10 to 30, 10 to 20 or 10 to 15 nucleotides in length.
  • the targeting domain is 20 to 100, 20 to 90, 20 to 80, 20 to 70, 20 to 60, 20 to 50, 20 to 40, 20 to 30, or 20 to 25 nucleotides in length.
  • the targeting domain has full complementarity with the target sequence.
  • the targeting domain has or includes 1, 2, 3, 4, 5, 6, 7 or 8 nucleotides that are not complementary with the corresponding nucleotide of the targeting domain.
  • the target domain includes 1, 2, 3, 4 or 5 nucleotides that are complementary with the corresponding nucleotide of the targeting domain within 5 nucleotides of its 5′ end. In an embodiment, the target domain includes 1, 2, 3, 4 or 5 nucleotides that are complementary with the corresponding nucleotide of the targeting domain within 5 nucleotides of its 3′ end.
  • the target domain includes 1, 2, 3, or 4 nucleotides that are not complementary with the corresponding nucleotide of the targeting domain within 5 nucleotides of its 5′ end. In an embodiment, the target domain includes 1, 2, 3, or 4 nucleotides that are not complementary with the corresponding nucleotide of the targeting domain within 5 nucleotides of its 3′ end.
  • the degree of complementarity, together with other properties of the gRNA, is sufficient to allow targeting of a Cas9 fusion molecule to the target nucleic acid.
  • the targeting domain comprises two consecutive nucleotides that are not complementary to the target domain (“non-complementary nucleotides”), e.g., two consecutive noncomplementary nucleotides that are within 5 nucleotides of the 5′ end of the targeting domain, within 5 nucleotides of the 3′ end of the targeting domain, or more than 5 nucleotides away from one or both ends of the targeting domain.
  • non-complementary nucleotides two consecutive noncomplementary nucleotides that are within 5 nucleotides of the 5′ end of the targeting domain, within 5 nucleotides of the 3′ end of the targeting domain, or more than 5 nucleotides away from one or both ends of the targeting domain.
  • no two consecutive nucleotides within 5 nucleotides of the 5′ end of the targeting domain, within 5 nucleotides of the 3′ end of the targeting domain, or within a region that is more than 5 nucleotides away from one or both ends of the targeting domain, are not complementary to the targeting domain.
  • the targeting domain nucleotides do not comprise modifications, e.g., modifications of the type provided in Section XI.
  • the targeting domain comprises one or more modifications, e.g., modifications that it render it less susceptible to degradation or more bio-compatible, e.g., less immunogenic.
  • the backbone of the targeting domain can be modified with a phosphorothioate, or other modification from Section XI.
  • a nucleotide of the targeting domain can comprise a 2′ modification (e.g., a modification at the 2′ position on ribose), e.g., a 2-acetylation, e.g., a 2′ methylation, or other modification(s) from Section XI.
  • a 2′ modification e.g., a modification at the 2′ position on ribose
  • 2-acetylation e.g., a 2′ methylation
  • the targeting domain includes 1, 2, 3, 4, 5, 6, 7 or 8 or more modifications. In an embodiment, the targeting domain includes 1, 2, 3, or 4 modifications within 5 nucleotides of its 5′ end. In an embodiment, the targeting domain comprises as many as 1, 2, 3, or 4 modifications within 5 nucleotides of its 3′ end.
  • the targeting domain comprises modifications at two consecutive nucleotides, e.g., two consecutive nucleotides that are within 5 nucleotides of the 5′ end of the targeting domain, within 5 nucleotides of the 3′ end of the targeting domain, or more than 5 nucleotides away from one or both ends of the targeting domain.
  • no two consecutive nucleotides are modified within 5 nucleotides of the 5′ end of the targeting domain, within 5 nucleotides of the 3′ end of the targeting domain, or within a region that is more than 5 nucleotides away from one or both ends of the targeting domain.
  • no nucleotide is modified within 5 nucleotides of the 5′ end of the targeting domain, within 5 nucleotides of the 3′ end of the targeting domain, or within a region that is more than 5 nucleotides away from one or both ends of the targeting domain.
  • Modifications in the targeting domain can be selected to not interfere with targeting efficacy, which can be evaluated by testing a candidate modification in the system described in Section VII.
  • gRNAs having a candidate targeting domain having a selected length, sequence, degree of complementarity, or degree of modification can be evaluated in a system in Section VII.
  • the candidate targeting domain can be placed, either alone, or with one or more other candidate changes in a gRNA molecule/Cas9 molecule system known to be functional with a selected target and evaluated.
  • all of the modified nucleotides are complementary to and capable of hybridizing to corresponding nucleotides present in the target domain. In another embodiment, 1, 2, 3, 4, 5, 6, 7 or 8 or more modified nucleotides are not complementary to or capable of hybridizing to corresponding nucleotides present in the target domain.
  • the targeting domain comprises, preferably in the 5′-3′ direction: a secondary domain and a core domain. These domains are discussed in more detail below.
  • the “core domain” of the targeting domain is complementary to the “core domain target” on the target nucleic acid.
  • the core domain comprises about 8 to about 13 nucleotides from the 3′ end of the targeting domain (e.g., the most 3′ 8 to 13 nucleotides of the targeting domain).
  • the core domain of the targeting domain and core domain target are independently, 6+/ ⁇ 2, 7+/ ⁇ 2, 8+/ ⁇ 2, 9+/ ⁇ 2, 10+/ ⁇ 2, 11+/ ⁇ 2, 12+/ ⁇ 2, 13+/ ⁇ 2, 14+/ ⁇ 2, 15+/ ⁇ 2, or 16+ ⁇ 2, nucleotides in length.
  • the core domain of the targeting domain and core domain target are independently, 10+/ ⁇ 2 nucleotides in length.
  • the core domain of the targeting domain and core domain target are independently, 10+/ ⁇ 4 nucleotides in length.
  • the core domain of the targeting domain and core domain target are independently 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18 nucleotides in length.
  • the core domain of the targeting domain and core domain target are independently 3 to 20, 4 to 20, 5 to 20, 6 to 20, 7 to 20, 8 to 20, 9 to 20 10 to 20 or 15 to 20 nucleotides in length.
  • the core domain of the targeting domain and core domain target are independently 3 to 15, e.g., 6 to 15, 7 to 14, 7 to 13, 6 to 12, 7 to 12, 7 to 11, 7 to 10, 8 to 14, 8 to 13, 8 to 12, 8 to 11, 8 to 10 or 8 to 9 nucleotides in length.
  • the core domain of the targeting domain is complementary with the core domain target.
  • the core domain has exact complementarity with the core domain target.
  • the core domain of the targeting domain can have 1, 2, 3, 4 or 5 nucleotides that are not complementary with the corresponding nucleotide of the core domain target.
  • the degree of complementarity, together with other properties of the gRNA molecule is sufficient to allow targeting of a Cas9 molecule to the target nucleic acid.
  • the “secondary domain” of the targeting domain of the gRNA is complementary to the “secondary domain target” of the target nucleic acid.
  • the secondary domain is positioned 5′ to the core domain.
  • the secondary domain is absent or optional.
  • the targeting domain is 26 nucleotides in length and the core domain (counted from the 3′ end of the targeting domain) is 8 to 13 nucleotides in length
  • the secondary domain is 13 to 18 nucleotides in length.
  • the targeting domain is 25 nucleotides in length and the core domain (counted from the 3′ end of the targeting domain) is 8 to 13 nucleotides in length
  • the secondary domain is 12 to 17 nucleotides in length.
  • the targeting domain is 24 nucleotides in length and the core domain (counted from the 3′ end of the targeting domain) is 8 to 13 nucleotides in length
  • the secondary domain is 11 to 16 nucleotides in length.
  • the targeting domain is 23 nucleotides in length and the core domain (counted from the 3′ end of the targeting domain) is 8 to 13 nucleotides in length
  • the secondary domain is 10 to 15 nucleotides in length.
  • the targeting domain is 22 nucleotides in length and the core domain (counted from the 3′ end of the targeting domain) is 8 to 13 nucleotides in length
  • the secondary domain is 9 to 14 nucleotides in length.
  • the targeting domain is 21 nucleotides in length and the core domain (counted from the 3′ end of the targeting domain) is 8 to 13 nucleotides in length
  • the secondary domain is 8 to 13 nucleotides in length.
  • the targeting domain is 20 nucleotides in length and the core domain (counted from the 3′ end of the targeting domain) is 8 to 13 nucleotides in length
  • the secondary domain is 7 to 12 nucleotides in length.
  • the targeting domain is 19 nucleotides in length and the core domain (counted from the 3′ end of the targeting domain) is 8 to 13 nucleotides in length
  • the secondary domain is 6 to 11 nucleotides in length.
  • the targeting domain is 18 nucleotides in length and the core domain (counted from the 3′ end of the targeting domain) is 8 to 13 nucleotides in length
  • the secondary domain is 5 to 10 nucleotides in length.
  • the targeting domain is 17 nucleotides in length and the core domain (counted from the 3′ end of the targeting domain) is 8 to 13 nucleotides in length
  • the secondary domain is 4 to 9 nucleotides in length.
  • the targeting domain is 16 nucleotides in length and the core domain (counted from the 3′ end of the targeting domain) is 8 to 13 nucleotides in length
  • the secondary domain is 3 to 8 nucleotides in length.
  • the secondary domain is 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18 nucleotides in length.
  • the secondary domain of the targeting domain is complementary with the secondary domain target.
  • the secondary domain of the targeting domain has exact complementarity with the secondary domain target.
  • the secondary domain of the targeting domain can have 1, 2, 3, 4 or 5 nucleotides that are not complementary with the corresponding nucleotide of the secondary domain target.
  • the degree of complementarity, together with other properties of the gRNA, is sufficient to allow targeting of a Cas9 fusion molecule to the target nucleic acid.
  • the core domain nucleotides do not comprise modifications, e.g., modifications of the type provided in Section XI.
  • the core domain comprises one or more modifications, e.g., modifications that it render it less susceptible to degradation or more bio-compatible, e.g., less immunogenic.
  • the backbone of the core domain can be modified with a phosphorothioate, or other modification(s) from Section XI.
  • a nucleotide of the core domain can comprise a 2′ modification (e.g., a modification at the 2′ position on ribose), e.g., a 2-acetylation, e.g., a 2′ methylation, or other modification(s) from Section XI.
  • a core domain will contain no more than 1, 2, or 3 modifications.
  • Modifications in the core domain can be selected to not interfere with targeting efficacy, which can be evaluated by testing a candidate modification in the system described in Section VII.
  • gRNAs having a candidate core domain having a selected length, sequence, degree of complementarity, or degree of modification can be evaluated in the system described at Section VII.
  • the candidate core domain can be placed, either alone, or with one or more other candidate changes in a gRNA molecule/Cas9 molecule system known to be functional with a selected target and evaluated.
  • the secondary domain nucleotides do not comprise modifications, e.g., modifications of the type provided in Section XI.
  • the secondary domain comprises one or more modifications, e.g., modifications that render it less susceptible to degradation or more bio-compatible, e.g., less immunogenic.
  • the backbone of the secondary domain can be modified with a phosphorothioate, or other modification(s) from Section XI.
  • a nucleotide of the secondary domain can comprise a 2′ modification (e.g., a modification at the 2′ position on ribose), e.g., a 2-acetylation, e.g., a 2′ methylation, or other modification from Section XI.
  • a secondary domain will contain no more than 1, 2, or 3 modifications.
  • Modifications in the secondary domain can be selected to not interfere with targeting efficacy, which can be evaluated by testing a candidate modification in the system described in Section VII.
  • gRNAs having a candidate secondary domain having a selected length, sequence, degree of complementarity, or degree of modification can be evaluated in the system described at Section VII.
  • the candidate secondary domain can be placed, either alone, or with one or more other candidate changes in a gRNA molecule/Cas9 molecule system known to be functional with a selected target and evaluated.
  • (1) the degree of complementarity between the core domain of the targeting domain and its target (i.e., the core domain target), and (2) the degree of complementarity between the secondary domain of the targeting domain and its target (i.e., the secondary domain target), may differ.
  • (1) may be greater than (2).
  • (1) may be less than (2).
  • (1) and (2) are the same, e.g., each may be completely complementary with its target.
  • (1) the number of modifications (e.g., modifications from Section XI) of the nucleotides of the core domain and (2) the number of modification (e.g., modifications from Section XI) of the nucleotides of the secondary domain may differ. In an embodiment, (1) may be less than (2). In an embodiment, (1) may be greater than (2). In an embodiment, (1) and (2) may be the same, e.g., each may be free of modifications.
  • the first complementarity domain is complementary with the second complementarity domain.
  • the first domain does not have exact complementarity with the second complementarity domain.
  • the first complementarity domain can have 1, 2, 3, 4 or 5 nucleotides that are not complementary with the corresponding nucleotide of the second complementarity domain.
  • 1, 2, 3, 4, 5 or 6, e.g., 3 nucleotides will not pair in the duplex, and, e.g., form a non-duplexed or looped-out region.
  • an unpaired, or loop-out, region e.g., a loop-out of 3 nucleotides, is present on the second complementarity domain.
  • the unpaired region begins 1, 2, 3, 4, 5, or 6, e.g., 4, nucleotides from the 5′ end of the second complementarity domain.
  • the degree of complementarity, together with other properties of the gRNA, is sufficient to allow targeting of a Cas9 molecule to the target nucleic acid.
  • the first and second complementarity domains are:
  • the second complementarity domain is longer than the first complementarity domain, e.g., 2, 3, 4, 5, or 6, e.g., 6, nucleotides longer.
  • the first and second complementary domains independently, do not comprise modifications, e.g., modifications of the type provided in Section XI.
  • the first and second complementary domains independently, comprise one or more modifications, e.g., modifications that the render the domain less susceptible to degradation or more bio-compatible, e.g., less immunogenic.
  • the backbone of the domain can be modified with a phosphorothioate, or other modification(s) from Section XI.
  • a nucleotide of the domain can comprise a 2′ modification (e.g., a modification at the 2′ position on ribose), e.g., a 2-acetylation, e.g., a 2′ methylation, or other modification(s) from Section XI.
  • the first and second complementary domains independently, include 1, 2, 3, 4, 5, 6, 7 or 8 or more modifications. In an embodiment, the first and second complementary domains, independently, include 1, 2, 3, or 4 modifications within 5 nucleotides of its 5′ end. In an embodiment, the first and second complementary domains, independently, include as many as 1, 2, 3, or 4 modifications within 5 nucleotides of its 3′ end.
  • the first and second complementary domains independently, include modifications at two consecutive nucleotides, e.g., two consecutive nucleotides that are within 5 nucleotides of the 5′ end of the domain, within 5 nucleotides of the 3′ end of the domain, or more than 5 nucleotides away from one or both ends of the domain.
  • the first and second complementary domains independently, include no two consecutive nucleotides that are modified, within 5 nucleotides of the 5′ end of the domain, within 5 nucleotides of the 3′ end of the domain, or within a region that is more than 5 nucleotides away from one or both ends of the domain.
  • the first and second complementary domains independently, include no nucleotide that is modified within 5 nucleotides of the 5′ end of the domain, within 5 nucleotides of the 3′ end of the domain, or within a region that is more than 5 nucleotides away from one or both ends of the domain.
  • Modifications in a complementarity domain can be selected to not interfere with targeting efficacy, which can be evaluated by testing a candidate modification in the system described in Section VII.
  • gRNAs having a candidate complementarity domain having a selected length, sequence, degree of complementarity, or degree of modification can be evaluated in the system described in SectionVII.
  • the candidate complementarity domain can be placed, either alone, or with one or more other candidate changes in a gRNA molecule/Cas9 molecule system known to be functional with a selected target and evaluated.
  • the first complementarity domain has at least 60, 70, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99% homology with, or differs by no more than 1, 2, 3, 4, 5, or 6 nucleotides from, a reference first complementarity domain, e.g., a naturally occurring, e.g., an S. pyogenes, S. aureus or S. thermophilus , first complementarity domain, or a first complementarity domain described herein.
  • a reference first complementarity domain e.g., a naturally occurring, e.g., an S. pyogenes, S. aureus or S. thermophilus
  • first complementarity domain e.g., a naturally occurring, e.g., an S. pyogenes, S. aureus or S. thermophilus
  • the second complementarity domain has at least 60, 70, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99% homology with, or differs by no more than 1, 2, 3, 4, 5, or 6 nucleotides from, a reference second complementarity domain, e.g., a naturally occurring, e.g., an S. pyogenes, S. aureus or S. thermophilus , second complementarity domain, or a second complementarity domain described herein.
  • a reference second complementarity domain e.g., a naturally occurring, e.g., an S. pyogenes, S. aureus or S. thermophilus
  • second complementarity domain e.g., a naturally occurring, e.g., an S. pyogenes, S. aureus or S. thermophilus
  • the duplexed region formed by first and second complementarity domains is typically 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 or 22 base pairs in length (excluding any looped out or unpaired nucleotides).
  • the first and second complementarity domains when duplexed, comprise 11 paired nucleotides, for example, in the gRNA sequence (one paired strand underlined, one bolded):
  • the first and second complementarity domains when duplexed, comprise 15 paired nucleotides, for example in the gRNA sequence (one paired strand underlined, one bolded):
  • first and second complementarity domains when duplexed, comprise 16 paired nucleotides, for example in the gRNA sequence (one paired strand underlined, one bolded):
  • first and second complementarity domains when duplexed, comprise 21 paired nucleotides, for example in the gRNA sequence (one paired strand underlined, one bolded):
  • nucleotides are exchanged to remove poly-U tracts, for example in the gRNA sequences (exchanged nucleotides underlined):
  • a modular gRNA can comprise additional sequence, 5′ to the second complementarity domain.
  • the 5′ extension domain is 2 to 10, 2 to 9, 2 to 8, 2 to 7, 2 to 6, 2 to 5, or 2 to 4 nucleotides in length.
  • the 5′ extension domain is 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more nucleotides in length.
  • the 5′ extension domain nucleotides do not comprise modifications, e.g., modifications of the type provided in Section XI.
  • the 5′ extension domain comprises one or more modifications, e.g., modifications that it render it less susceptible to degradation or more bio-compatible, e.g., less immunogenic.
  • the backbone of the 5′ extension domain can be modified with a phosphorothioate, or other modification(s) from Section XI.
  • a nucleotide of the 5′ extension domain can comprise a 2′ modification (e.g., a modification at the 2′ position on ribose), e.g., a 2-acetylation, e.g., a 2′ methylation, or other modification(s) from Section XI.
  • a 2′ modification e.g., a modification at the 2′ position on ribose
  • 2-acetylation e.g., a 2′ methylation
  • the 5′ extension domain can comprise as many as 1, 2, 3, 4, 5, 6, 7 or 8 modifications. In an embodiment, the 5′ extension domain comprises as many as 1, 2, 3, or 4 modifications within 5 nucleotides of its 5′ end, e.g., in a modular gRNA molecule. In an embodiment, the 5′ extension domain comprises as many as 1, 2, 3, or 4 modifications within 5 nucleotides of its 3′ end, e.g., in a modular gRNA molecule.
  • the 5′ extension domain comprises modifications at two consecutive nucleotides, e.g., two consecutive nucleotides that are within 5 nucleotides of the 5′ end of the 5′ extension domain, within 5 nucleotides of the 3′ end of the 5′ extension domain, or more than 5 nucleotides away from one or both ends of the 5′ extension domain. In an embodiment, no two consecutive nucleotides are modified within 5 nucleotides of the 5′ end of the 5′ extension domain, within 5 nucleotides of the 3′ end of the 5′ extension domain, or within a region that is more than 5 nucleotides away from one or both ends of the 5′ extension domain.
  • no nucleotide is modified within 5 nucleotides of the 5′ end of the 5′ extension domain, within 5 nucleotides of the 3′ end of the 5′ extension domain, or within a region that is more than 5 nucleotides away from one or both ends of the 5′ extension domain.
  • Modifications in the 5′ extension domain can be selected so as to not interfere with gRNA molecule efficacy, which can be evaluated by testing a candidate modification in the system described in Section VII.
  • gRNAs having a candidate 5′ extension domain having a selected length, sequence, degree of complementarity, or degree of modification can be evaluated in the system described at Section VII.
  • the candidate 5′ extension domain can be placed, either alone, or with one or more other candidate changes in a gRNA molecule/Cas9 molecule system known to be functional with a selected target and evaluated.
  • the 5′ extension domain has at least 60, 70, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99% homology with, or differs by no more than 1, 2, 3, 4, 5, or 6 nucleotides from, a reference 5′ extension domain, e.g., a naturally occurring, e.g., an S. pyogenes, S. aureus or S. thermophilus, 5′ extension domain, or a 5′ extension domain described herein.
  • a reference 5′ extension domain e.g., a naturally occurring, e.g., an S. pyogenes, S. aureus or S. thermophilus
  • 5′ extension domain or a 5′ extension domain described herein.
  • the linking domain is disposed between the first and second complementarity domains.
  • the two molecules are associated with one another by the complementarity domains.
  • the linking domain is 10+/ ⁇ 5, 20+/ ⁇ 5, 30+/ ⁇ 5, 40+/ ⁇ 5, 50+/ ⁇ 5, 60+/ ⁇ 5, 70+/ ⁇ 5, 80+/ ⁇ 5, 90+/ ⁇ 5, or 100+/ ⁇ 5 nucleotides, in length.
  • the linking domain is 20+/ ⁇ 10, 30+/ ⁇ 10, 40+/ ⁇ 10, 50+/ ⁇ 10, 60+/ ⁇ 10, 70+/ ⁇ 10, 80+/ ⁇ 10, 90+/ ⁇ 10, or 100+/ ⁇ 10 nucleotides, in length.
  • the linking domain is 10 to 100, 10 to 90, 10 to 80, 10 to 70, 10 to 60, 10 to 50, 10 to 40, 10 to 30, 10 to 20 or 10 to 15 nucleotides in length.
  • the linking domain is 20 to 100, 20 to 90, 20 to 80, 20 to 70, 20 to 60, 20 to 50, 20 to 40, 20 to 30, or 20 to 25 nucleotides in length.
  • the linking domain is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16 17, 18, 19, or 20 nucleotides in length.
  • the linking domain is a covalent bond.
  • the linking domain comprises a duplexed region, typically adjacent to or within 1, 2, or 3 nucleotides of the 3′ end of the first complementarity domain and/or the 5-end of the second complementarity domain.
  • the duplexed region can be 20+/ ⁇ 10 base pairs in length.
  • the duplexed region can be 10+/ ⁇ 5, 15+/ ⁇ 5, 20+/ ⁇ 5, or 30+/ ⁇ 5 base pairs in length.
  • the duplexed region can be 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 base pairs in length.
  • sequences forming the duplexed region have exact complementarity with one another, though in an embodiment as many as 1, 2, 3, 4, 5, 6, 7 or 8 nucleotides are not complementary with the corresponding nucleotides.
  • the linking domain nucleotides do not comprise modifications, e.g., modifications of the type provided in Section XI.
  • the linking domain comprises one or more modifications, e.g., modifications that it render it less susceptible to degradation or more bio-compatible, e.g., less immunogenic.
  • the backbone of the linking domain can be modified with a phosphorothioate, or other modification(s) from Section XI.
  • a nucleotide of the linking domain can comprise a 2′ modification (e.g., a modification at the 2′ position on ribose), e.g., a 2-acetylation, e.g., a 2′ methylation, or other modification(s) from Section XI.
  • the linking domain can comprise as many as 1, 2, 3, 4, 5, 6, 7 or 8 modifications.
  • Modifications in a linking domain can be selected so as to not interfere with targeting efficacy, which can be evaluated by testing a candidate modification in the system described in Section VII.
  • gRNAs having a candidate linking domain having a selected length, sequence, degree of complementarity, or degree of modification can be evaluated a system described in Section VII.
  • a candidate linking domain can be placed, either alone, or with one or more other candidate changes in a gRNA molecule/Cas9 molecule system known to be functional with a selected target and evaluated.
  • the linking domain has at least 60, 70, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99% homology with, or differs by no more than 1, 2, 3, 4, 5, or 6 nucleotides from, a reference linking domain, e.g., a linking domain described herein.
  • the proximal domain is 6+/ ⁇ 2, 7+/ ⁇ 2, 8+/ ⁇ 2, 9+/ ⁇ 2, 10+/ ⁇ 2, 11+/ ⁇ 2, 12+/ ⁇ 2, 13+/ ⁇ 2, 14+/ ⁇ 2, 14+/ ⁇ 2, 16+/ ⁇ 2, 17+/ ⁇ 2, 18+/ ⁇ 2, 19+/ ⁇ 2, or 20+/ ⁇ 2 nucleotides in length.
  • the proximal domain is 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25 or 26 nucleotides in length.
  • the proximal domain is 5 to 20, 7, to 18, 9 to 16, or 10 to 14 nucleotides in length.
  • the proximal domain nucleotides do not comprise modifications, e.g., modifications of the type provided in Section XI.
  • the proximal domain comprises one or more modifications, e.g., modifications that it render it less susceptible to degradation or more bio-compatible, e.g., less immunogenic.
  • the backbone of the proximal domain can be modified with a phosphorothioate, or other modification(s) from Section XI.
  • a nucleotide of the proximal domain can comprise a 2′ modification (e.g., a modification at the 2′ position on ribose), e.g., a 2-acetylation, e.g., a 2′ methylation, or other modification(s) from Section XI.
  • a 2′ modification e.g., a modification at the 2′ position on ribose
  • 2-acetylation e.g., a 2′ methylation
  • the proximal domain can comprise as many as 1, 2, 3, 4, 5, 6, 7 or 8 modifications. In an embodiment, the proximal domain comprises as many as 1, 2, 3, or 4 modifications within 5 nucleotides of its 5′ end, e.g., in a modular gRNA molecule. In an embodiment, the target domain comprises as many as 1, 2, 3, or 4 modifications within 5 nucleotides of its 3′ end, e.g., in a modular gRNA molecule.
  • the proximal domain comprises modifications at two consecutive nucleotides, e.g., two consecutive nucleotides that are within 5 nucleotides of the 5′ end of the proximal domain, within 5 nucleotides of the 3′ end of the proximal domain, or more than 5 nucleotides away from one or both ends of the proximal domain. In an embodiment, no two consecutive nucleotides are modified within 5 nucleotides of the 5′ end of the proximal domain, within 5 nucleotides of the 3′ end of the proximal domain, or within a region that is more than 5 nucleotides away from one or both ends of the proximal domain.
  • no nucleotide is modified within 5 nucleotides of the 5′ end of the proximal domain, within 5 nucleotides of the 3′ end of the proximal domain, or within a region that is more than 5 nucleotides away from one or both ends of the proximal domain.
  • Modifications in the proximal domain can be selected so as to not interfere with gRNA molecule efficacy, which can be evaluated by testing a candidate modification in the system described in Section VII.
  • gRNAs having a candidate proximal domain having a selected length, sequence, degree of complementarity, or degree of modification can be evaluated in the system described at Section VII.
  • the candidate proximal domain can be placed, either alone, or with one or more other candidate changes in a gRNA molecule/Cas9 molecule system known to be functional with a selected target and evaluated.
  • the proximal domain has at least 60, 70, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99% homology with, or differs by no more than 1, 2, 3, 4, 5, or 6 nucleotides from, a reference proximal domain, e.g., a naturally occurring, e.g., an S. pyogenes, S. aureus or S. thermophilus , proximal domain, or a proximal domain described herein.
  • the tail domain is 10+/ ⁇ 5, 20+/ ⁇ 5, 30+/ ⁇ 5, 40+/ ⁇ 5, 50+/ ⁇ 5, 60+/ ⁇ 5, 70+/ ⁇ 5, 80+/ ⁇ 5, 90+/ ⁇ 5, or 100+/ ⁇ 5 nucleotides, in length.
  • the tail domain is 20+/ ⁇ 5 nucleotides in length.
  • the tail domain is 20+/ ⁇ 10, 30+/ ⁇ 10, 40+/ ⁇ 10, 50+/ ⁇ 10, 60+/ ⁇ 10, 70+/ ⁇ 10, 80+/ ⁇ 10, 90+/ ⁇ 10, or 100+/ ⁇ 10 nucleotides, in length.
  • the tail domain is 25+/ ⁇ 10 nucleotides in length.
  • the tail domain is 10 to 100, 10 to 90, 10 to 80, 10 to 70, 10 to 60, 10 to 50, 10 to 40, 10 to 30, 10 to 20 or 10 to 15 nucleotides in length.
  • the tail domain is 20 to 100, 20 to 90, 20 to 80, 20 to 70, 20 to 60, 20 to 50, 20 to 40, 20 to 30, or 20 to 25 nucleotides in length.
  • the tail domain is 1 to 20, 1 to 15, 1 to 10, or 1 to 5 nucleotides in length.
  • the tail domain nucleotides do not comprise modifications, e.g., modifications of the type provided in Section XI.
  • the tail domain comprises one or more modifications, e.g., modifications that it render it less susceptible to degradation or more bio-compatible, e.g., less immunogenic.
  • the backbone of the tail domain can be modified with a phosphorothioate, or other modification(s) from Section XI.
  • a nucleotide of the tail domain can comprise a 2′ modification (e.g., a modification at the 2′ position on ribose), e.g., a 2-acetylation, e.g., a 2′ methylation, or other modification(s) from Section XI.
  • a 2′ modification e.g., a modification at the 2′ position on ribose
  • 2-acetylation e.g., a 2′ methylation
  • the tail domain can have as many as 1, 2, 3, 4, 5, 6, 7 or 8 modifications.
  • the target domain comprises as many as 1, 2, 3, or 4 modifications within 5 nucleotides of its 5′ end. In an embodiment, the target domain comprises as many as 1, 2, 3, or 4 modifications within 5 nucleotides of its 3′ end.
  • the tail domain comprises a tail duplex domain, which can form a tail duplexed region.
  • the tail duplexed region can be 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 base pairs in length.
  • a further single stranded domain exists 3′ to the tail duplexed domain.
  • this domain is 3, 4, 5, 6, 7, 8, 9, or 10 nucleotides in length. In an embodiment it is 4 to 6 nucleotides in length.
  • the tail domain has at least 60, 70, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99% homology with, or differs by no more than 1, 2, 3, 4, 5, or 6 nucleotides from, a reference tail domain, e.g., a naturally occurring, e.g., an S. pyogenes, S. aureus or S. thermophilus , tail domain, or a tail domain described herein.
  • a reference tail domain e.g., a naturally occurring, e.g., an S. pyogenes, S. aureus or S. thermophilus
  • tail domain or a tail domain described herein.
  • proximal and tail domain taken together comprise the following sequences:
  • the tail domain comprises the 3′ sequence UUUUUU, e.g., if a U6 promoter is used for transcription.
  • the tail domain comprises the 3′ sequence UUUU, e.g., if an H1 promoter is used for transcription.
  • tail domain comprises variable numbers of 3′ Us depending, e.g., on the termination signal of the pol-III promoter used.
  • the tail domain comprises variable 3′ sequence derived from the DNA template if a T7 promoter is used.
  • the tail domain comprises variable 3′ sequence derived from the DNA template, e.g., if in vitro transcription is used to generate the RNA molecule.
  • the tail domain comprises variable 3′ sequence derived from the DNA template, e.g., if a pol-II promoter is used to drive transcription.
  • Modifications in the tail domain can be selected to not interfere with targeting efficacy, which can be evaluated by testing a candidate modification in the system described in Section VII.
  • gRNAs having a candidate tail domain having a selected length, sequence, degree of complementarity, or degree of modification can be evaluated in the system described in Section VII.
  • the candidate tail domain can be placed, either alone, or with one or more other candidate changes in a gRNA molecule/Cas9 molecule system known to be functional with a selected target and evaluated.
  • the tail domain comprises modifications at two consecutive nucleotides, e.g., two consecutive nucleotides that are within 5 nucleotides of the 5′ end of the tail domain, within 5 nucleotides of the 3′ end of the tail domain, or more than 5 nucleotides away from one or both ends of the tail domain. In an embodiment, no two consecutive nucleotides are modified within 5 nucleotides of the 5′ end of the tail domain, within 5 nucleotides of the 3′ end of the tail domain, or within a region that is more than 5 nucleotides away from one or both ends of the tail domain.
  • no nucleotide is modified within 5 nucleotides of the 5′ end of the tail domain, within 5 nucleotides of the 3′ end of the tail domain, or within a region that is more than 5 nucleotides away from one or both ends of the tail domain.
  • the targeting domain comprises a core domain and optionally a secondary domain, and is 10 to 50 nucleotides in length;
  • the first complementarity domain is 5 to 25 nucleotides in length and, In an embodiment has at least 50, 60, 70, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99% homology with a reference first complementarity domain disclosed herein;
  • the linking domain is 1 to 5 nucleotides in length
  • the second complementarity domain is 5 to 27 nucleotides in length and, in an embodiment has at least 50, 60, 70, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99% homology with a reference second complementarity domain disclosed herein;
  • the proximal domain is 5 to 20 nucleotides in length and, in an embodiment has at least 50, 60, 70, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99% homology with a reference proximal domain disclosed herein; and
  • the tail domain is absent or a nucleotide sequence is 1 to 50 nucleotides in length and, in an embodiment has at least 60, 70, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99% homology with a reference tail domain disclosed herein.
  • a unimolecular, or chimeric, gRNA comprises, preferably from 5′ to 3′:
  • the sequence from (a), (b), or (c) has at least 60, 70, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99% homology with the corresponding sequence of a naturally occurring gRNA, or with a gRNA described herein.
  • proximal and tail domain when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • the targeting domain comprises, has, or consists of, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25 or 26 nucleotides (e.g., 16, 17, 18, 19, 20, 21, 22, 23, 24, 25 or 26 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, or 26 nucleotides in length.
  • the targeting domain comprises, has, or consists of, 16 nucleotides (e.g., 16 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 16 nucleotides in length.
  • the targeting domain comprises, has, or consists of, 17 nucleotides (e.g., 17 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 17 nucleotides in length.
  • the targeting domain comprises, has, or consists of, 18 nucleotides (e.g., 18 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 18 nucleotides in length.
  • the targeting domain comprises, has, or consists of, 19 nucleotides (e.g., 19 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 19 nucleotides in length.
  • the targeting domain comprises, has, or consists of, 20 nucleotides (e.g., 20 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 20 nucleotides in length.
  • the targeting domain comprises, has, or consists of, 21 nucleotides (e.g., 21 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 21 nucleotides in length.
  • the targeting domain comprises, has, or consists of, 22 nucleotides (e.g., 22 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 22 nucleotides in length.
  • the targeting domain comprises, has, or consists of, 23 nucleotides (e.g., 23 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 23 nucleotides in length.
  • the targeting domain comprises, has, or consists of, 24 nucleotides (e.g., 24 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 24 nucleotides in length.
  • the targeting domain comprises, has, or consists of, 25 nucleotides (e.g., 25 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 25 nucleotides in length.
  • the targeting domain comprises, has, or consists of, 26 nucleotides (e.g., 26 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 26 nucleotides in length.
  • the targeting domain comprises, has, or consists of, 16 nucleotides (e.g., 16 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 16 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • the targeting domain comprises, has, or consists of, 16 nucleotides (e.g., 16 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 16 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • the targeting domain comprises, has, or consists of, 16 nucleotides (e.g., 16 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 16 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • 16 nucleotides e.g., 16 consecutive nucleotides having complementarity with the target domain
  • the targeting domain is 16 nucleotides in length
  • the targeting domain comprises, has, or consists of, 17 nucleotides (e.g., 17 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 17 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • the targeting domain comprises, has, or consists of, 17 nucleotides (e.g., 17 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 17 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • the targeting domain comprises, has, or consists of, 17 nucleotides (e.g., 17 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 17 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • the targeting domain comprises, has, or consists of, 18 nucleotides (e.g., 18 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 18 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • the targeting domain comprises, has, or consists of, 18 nucleotides (e.g., 18 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 18 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • the targeting domain comprises, has, or consists of, 18 nucleotides (e.g., 18 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 18 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • 18 nucleotides e.g., 18 consecutive nucleotides having complementarity with the target domain
  • the targeting domain is 18 nucleotides in length
  • the targeting domain comprises, has, or consists of, 19 nucleotides (e.g., 19 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 19 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • the targeting domain comprises, has, or consists of, 19 nucleotides (e.g., 19 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 19 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • the targeting domain comprises, has, or consists of, 19 nucleotides (e.g., 19 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 19 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • 19 nucleotides e.g., 19 consecutive nucleotides having complementarity with the target domain
  • the targeting domain is 19 nucleotides in length
  • the targeting domain comprises, has, or consists of, 20 nucleotides (e.g., 20 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 20 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • the targeting domain comprises, has, or consists of, 20 nucleotides (e.g., 20 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 20 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • the targeting domain comprises, has, or consists of, 20 nucleotides (e.g., 20 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 20 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • the targeting domain comprises, has, or consists of, 21 nucleotides (e.g., 21 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 21 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • the targeting domain comprises, has, or consists of, 21 nucleotides (e.g., 21 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 21 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • the targeting domain comprises, has, or consists of, 21 nucleotides (e.g., 21 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 21 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • the targeting domain comprises, has, or consists of, 22 nucleotides (e.g., 22 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 22 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • the targeting domain comprises, has, or consists of, 22 nucleotides (e.g., 22 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 22 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • the targeting domain comprises, has, or consists of, 22 nucleotides (e.g., 22 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 22 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • the targeting domain comprises, has, or consists of, 23 nucleotides (e.g., 23 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 23 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • the targeting domain comprises, has, or consists of, 23 nucleotides (e.g., 23 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 23 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • the targeting domain comprises, has, or consists of, 23 nucleotides (e.g., 23 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 23 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • the targeting domain comprises, has, or consists of, 24 nucleotides (e.g., 24 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 24 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • the targeting domain comprises, has, or consists of, 24 nucleotides (e.g., 24 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 24 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • the targeting domain comprises, has, or consists of, 24 nucleotides (e.g., 24 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 24 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • the targeting domain comprises, has, or consists of, 25 nucleotides (e.g., 25 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 25 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • the targeting domain comprises, has, or consists of, 25 nucleotides (e.g., 25 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 25 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • the targeting domain comprises, has, or consists of, 25 nucleotides (e.g., 25 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 25 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • the targeting domain comprises, has, or consists of, 26 nucleotides (e.g., 26 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 26 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • the targeting domain comprises, has, or consists of, 26 nucleotides (e.g., 26 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 26 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • the targeting domain comprises, has, or consists of, 26 nucleotides (e.g., 26 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 26 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • the unimolecular, or chimeric, gRNA molecule (comprising a targeting domain, a first complementary domain, a linking domain, a second complementary domain, a proximal domain and, optionally, a tail domain) comprises the following sequence in which the targeting domain is depicted as 20 Ns but could be any sequence and range in length from 16 to 26 nucleotides and in which the gRNA sequence is followed by 6 Us, which serve as a termination signal for the U6 promoter, but which could be either absent or fewer in number: NNNNNNNNNNNNNNNNNNNNNNNNGUUUUAGAGCUAGAAAUAGCAAGUUAAAAUAA GGCUAGUCCGUUAUCAACUUGAAAAAGUGGCACCGAGUCGGUGCUUUUUU (SEQ ID NO: XX).
  • the unimolecular, or chimeric, gRNA molecule is a S. pyogenes gRNA molecule.
  • the unimolecular, or chimeric, gRNA molecule (comprising a targeting domain, a first complementary domain, a linking domain, a second complementary domain, a proximal domain and, optionally, a tail domain) comprises the following sequence in which the targeting domain is depicted as 20 Ns but could be any sequence and range in length from 16 to 26 nucleotides and in which the gRNA sequence is followed by 6 Us, which serve as a termination signal for the U6 promoter, but which could be either absent or fewer in number: NNNNNNNNNNNNNNNNNNNNNNNNGUUUUAGUACUCUGGAAACAGAAUCUACUAAA ACAAGGCAAAAUGCCGUGUUUAUCUCGUCAACUUGUUGGCGAGAUUUUU (SEQ ID NO: XX).
  • the unimolecular, or chimeric, gRNA molecule is a S. aureus gRNA molecule.
  • a modular gRNA comprises:
  • the sequence from (a), (b), or (c) has at least 60, 75, 80, 85, 90, 95, or 99% homology with the corresponding sequence of a naturally occurring gRNA, or with a gRNA described herein.
  • proximal and tail domain when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • the targeting domain comprises, has, or consists of, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25 or 26 nucleotides (e.g., 16, 17, 18, 19, 20, 21, 22, 23, 24, 25 or 26 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 16, 17, 18, 19, 20, 21, 22, 23, 24, 25 or 26 nucleotides in length.
  • the targeting domain comprises, has, or consists of, 16 nucleotides (e.g., 16 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 16 nucleotides in length.
  • the targeting domain comprises, has, or consists of, 17 nucleotides (e.g., 17 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 17 nucleotides in length.
  • the targeting domain comprises, has, or consists of, 18 nucleotides (e.g., 18 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 18 nucleotides in length.
  • the targeting domain comprises, has, or consists of, 19 nucleotides (e.g., 19 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 19 nucleotides in length.
  • the targeting domain comprises, has, or consists of, 20 nucleotides (e.g., 20 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 20 nucleotides in length.
  • the targeting domain comprises, has, or consists of, 21 nucleotides (e.g., 21 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 21 nucleotides in length.
  • the targeting domain comprises, has, or consists of, 22 nucleotides (e.g., 22 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 22 nucleotides in length.
  • the targeting domain comprises, has, or consists of, 23 nucleotides (e.g., 23 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 23 nucleotides in length.
  • the targeting domain comprises, has, or consists of, 24 nucleotides (e.g., 24 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 24 nucleotides in length.
  • the targeting domain comprises, has, or consists of, 25 nucleotides (e.g., 25 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 5 nucleotides in length.
  • the targeting domain comprises, has, or consists of, 26 nucleotides (e.g., 26 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 26 nucleotides in length.
  • the targeting domain comprises, has, or consists of, 16 nucleotides (e.g., 16 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 16 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • the targeting domain comprises, has, or consists of, 16 nucleotides (e.g., 16 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 16 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • the targeting domain comprises, has, or consists of, 16 nucleotides (e.g., 16 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 16 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • 16 nucleotides e.g., 16 consecutive nucleotides having complementarity with the target domain
  • the targeting domain is 16 nucleotides in length
  • the targeting domain has, or consists of, 17 nucleotides (e.g., 17 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 17 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • the targeting domain has, or consists of, 17 nucleotides (e.g., 17 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 17 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • the targeting domain has, or consists of, 17 nucleotides (e.g., 17 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 17 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • the targeting domain has, or consists of, 18 nucleotides (e.g., 18 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 18 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • the targeting domain has, or consists of, 18 nucleotides (e.g., 18 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 18 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • the targeting domain has, or consists of, 18 nucleotides (e.g., 18 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 18 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • the targeting domain comprises, has, or consists of, 19 nucleotides (e.g., 19 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 19 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • the targeting domain comprises, has, or consists of, 19 nucleotides (e.g., 19 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 19 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • the targeting domain comprises, has, or consists of, 19 nucleotides (e.g., 19 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 19 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • 19 nucleotides e.g., 19 consecutive nucleotides having complementarity with the target domain
  • the targeting domain is 19 nucleotides in length
  • the targeting domain comprises, has, or consists of, 20 nucleotides (e.g., 20 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 20 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • the targeting domain comprises, has, or consists of, 20 nucleotides (e.g., 20 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 20 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • the targeting domain comprises, has, or consists of, 20 nucleotides (e.g., 20 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 20 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • the targeting domain comprises, has, or consists of, 21 nucleotides (e.g., 21 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 21 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • the targeting domain comprises, has, or consists of, 21 nucleotides (e.g., 21 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 21 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • the targeting domain comprises, has, or consists of, 21 nucleotides (e.g., 21 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 21 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • the targeting domain comprises, has, or consists of, 22 nucleotides (e.g., 22 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 22 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • the targeting domain comprises, has, or consists of, 22 nucleotides (e.g., 22 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 22 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • the targeting domain comprises, has, or consists of, 22 nucleotides (e.g., 22 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 22 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • the targeting domain comprises, has, or consists of, 23 nucleotides (e.g., 23 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 23 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • the targeting domain comprises, has, or consists of, 23 nucleotides (e.g., 23 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 23 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • the targeting domain comprises, has, or consists of, 23 nucleotides (e.g., 23 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 23 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • the targeting domain comprises, has, or consists of, 24 nucleotides (e.g., 24 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 24 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • the targeting domain comprises, has, or consists of, 24 nucleotides (e.g., 24 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 24 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • the targeting domain comprises, has, or consists of, 24 nucleotides (e.g., 24 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 24 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • the targeting domain comprises, has, or consists of, 25 nucleotides (e.g., 25 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 25 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • the targeting domain comprises, has, or consists of, 25 nucleotides (e.g., 25 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 25 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • the targeting domain comprises, has, or consists of, 25 nucleotides (e.g., 25 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 25 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • the targeting domain comprises, has, or consists of, 26 nucleotides (e.g., 26 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 26 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • the targeting domain comprises, has, or consists of, 26 nucleotides (e.g., 26 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 26 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • the targeting domain comprises, has, or consists of, 26 nucleotides (e.g., 26 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 26 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • methods and compositions discussed herein provide methods and compositions for gene editing by using a gRNA molecule which comprises a polyA tail.
  • a polyA tail of undefined length ranging from 1 to 1000 nucleotide is added enzymatically using a polymerase such as E. coli polyA polymerase (E-PAP).
  • E-PAP E. coli polyA polymerase
  • the polyA tail of a specified length e.g., 1, 5, 10, 20, 30, 40, 50, 60, 100, or 150 nucleotides
  • RNA polymerase e.g., T7 RNA polymerase
  • a polyA tail of defined length (e.g., 1, 5, 10, 20, 30, 40, 50, 60, 100, or 150 nucleotides) is synthesized as a synthetic oligonucleotide and ligated on the 3′ end of the gRNA with either an RNA ligase or a DNA ligase with our without a splinted DNA oligonucleotide complementary to the guide RNA and the polyA oligonucleotide.
  • the entire gRNA including a defined length of polyA tail is made synthetically, in one or several pieces, and ligated together by either an RNA ligase or a DNA ligase with or without a splinted DNA oligonucleotide.
  • Methods for designing gRNAs are described herein, including methods for selecting, designing and validating target domains. Exemplary targeting domains are also provided herein. Targeting Domains discussed herein can be incorporated into the gRNAs described herein.
  • a software tool can be used to optimize the choice of gRNA within a user's target sequence, e.g., to minimize total off-target activity across the genome. Off target activity may be other than cleavage.
  • the tool can identify all off-target sequences (preceding either NAG or NGG PAMs) across the genome that contain up to certain number (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) of mismatched base-pairs.
  • the cleavage efficiency at each off-target sequence can be predicted, e.g., using an experimentally-derived weighting scheme.
  • Each possible gRNA is then ranked according to its total predicted off-target cleavage; the top-ranked gRNAs represent those that are likely to have the greatest on-target and the least off-target cleavage.
  • Other functions e.g., automated reagent design for CRISPR construction, primer design for the on-target Surveyor assay, and primer design for high-throughput detection and quantification of off-target cleavage via next-gen sequencing, can also be included in the tool.
  • Candidate gRNA molecules can be evaluated by art-known methods or as described in Section VII herein.
  • the targeting domains discussed herein can be incorporated into the gRNAs described herein.
  • gRNAs for use with S. pyogenes, S. aureus and N. meningitidis Cas9 enzymes.
  • Guide RNAs for use with S. pyogenes, S. aureus and N. meningitidis Cas9 molecules are identified using a DNA sequence searching algorithm.
  • Guide RNA design is carried out using a custom guide RNA design software based on the public tool cas-offinder (reference:Cas-OFFinder: a fast and versatile algorithm that searches for potential off-target sites of Cas9 RNA-guided endonucleases, Bioinformatics. 2014 Feb. 17. Bae S, Park J, Kim J S. PMID:24463181).
  • Said custom guide RNA design software scores guides after calculating their genome-wide off-target propensity.
  • an aggregate score is calculated for each guide and summarized in a tabular output using a web-interface.
  • the software also identifies all PAM adjacent sequences that differ by 1, 2, 3 or more nucleotides from the selected gRNA sites. Genomic DNA sequence for each gene was obtained from the UCSC Genome browser and sequences were screened for repeat elements using the publically available RepeatMasker program. RepeatMasker searches input DNA sequences for repeated elements and regions of low complexity. The output is a detailed annotation of the repeats present in a given query sequence.
  • gRNAs are ranked into tiers based on their distance to the target site, their orthogonality and presence of a 5′ G (based on identification of close matches in the human genome containing a relevant PAM (e.g., in the case of S. pyogenes , a NGG PAM, in the case of S. aureus , a NNGRRT or NNGRRV PAM, and in the case of N. meningitidis , a NNNNGATT or NNNNGCTT PAM).
  • PAM e.g., in the case of S. pyogenes , a NGG PAM, in the case of S. aureus , a NNGRRT or NNGRRV PAM, and in the case of N. meningitidis , a NNNNGATT or NNNNGCTT PAM.
  • Orthogonality refers to the number of sequences in the human genome that contain a minimum number of mismatches to the target sequence.
  • a “high level of orthogonality” or “good orthogonality” may, for example, refer to 20-mer gRNAs that have no identical sequences in the human genome besides the intended target, nor any sequences that contain one or two mismatches in the target sequence. Targeting domains with good orthogonality are selected to minimize off-target DNA cleavage.
  • gRNAs are identified for both single-gRNA nuclease cleavage and for a dual-gRNA paired “nickase” strategy. Criteria for selecting gRNAs and the determination for which gRNAs can be used for the dual-gRNA paired “nickase” strategy is based on two considerations:
  • the targeting domains discussed herein can be incorporated into the gRNAs described herein.
  • two or more (e.g., three or four) gRNA molecules are used with one Cas9 fusion molecule.
  • at least one Cas9 molecule is from a different species than the other Cas9 molecule(s).
  • one Cas9 molecule can be from one species and the other Cas9 molecule can be from a different species. Both Cas9 species are used to generate a single or double-strand break, as desired.
  • Any of the targeting domains in the tables described herein can be used with a Cas9 nickase molecule to generate a single strand break.
  • any of the targeting domains in the tables described herein can be used with a Cas9 nuclease molecule to generate a double strand break.
  • one Cas9 can be one species
  • the second Cas9 can be from a different species. Both Cas9 species are used to generate a single or double-strand break, as desired.
  • any upstream gRNA described herein may be paired with any downstream gRNA described herein.
  • an upstream gRNA designed for use with one species of Cas9 is paired with a downstream gRNA designed for use from a different species of Cas9, both Cas9 species are used to generate a single or double-strand break, as desired.
  • Cas9 molecules of a variety of species can be used in the methods and compositions described herein. While the S. pyogenes, S. aureus , and S. thermophilus Cas9 molecules are the subject of much of the disclosure herein, Cas9 molecules, derived from, or based on the Cas9 proteins of other species listed herein can be used as well. In other words, while the much of the description herein uses S. pyogenes and S. thermophilus Cas9 molecules, Cas9 molecules from the other species can replace them, e.g., Staphylococcus aureus and Neisseria meningitidis Cas9 molecules.
  • Additional Cas9 species include: Acidovorax avenae, Actinobacillus pleuropneumoniae, Actinobacillus succinogenes, Actinobacillus suis, Actinomyces sp., cycliphilus denitrificans, Aminomonas paucivorans, Bacillus cereus, Bacillus smithii, Bacillus thuringiensis, Bacteroides sp., Blastopirellula marina, Bradyrhizobium sp., Brevibacillus laterosporus, Campylobacter coli, Campylobacter jejuni, Campylobacter lari, Candidatus Puniceispirillum, Clostridium cellulolyticum, Clostridium perfringens, Corynebacterium accolens, Corynebacterium diphtheria, Corynebacterium matruchotii, Dinoroseobacter shibae, Eubacterium dolichum
  • a Cas9 molecule, or Cas9 polypeptide refers to a molecule or a polypeptide that can interact with a guide RNA (gRNA) molecule) and, in concert with the gRNA molecule, localizes to a site which comprises a target domain, and in some embodiments, a PAM sequence.
  • gRNA guide RNA
  • Cas9 molecule and Cas9 polypeptide refer to naturally occurring Cas9 molecules and to engineered, altered, or modified Cas9 molecules or Cas9 polypeptides that differ, e.g., by at least one amino acid residue, from a reference sequence, e.g., the most similar naturally occurring Cas9 molecule or a sequence of Table 100.
  • Crystal structures have been determined for two different naturally occurring bacterial Cas9 molecules (Jinek et al., S CIENCE , 343(6176):1247997, 2014) and for S. pyogenes Cas9 with a guide RNA (e.g., a synthetic fusion of crRNA and tracrRNA) (Nishimasu et al., C ELL , 156:935-949, 2014; and Anders et al., N ATURE , 2014, doi: 10.1038/nature13579).
  • a guide RNA e.g., a synthetic fusion of crRNA and tracrRNA
  • a naturally occurring Cas9 molecule comprises two lobes: a recognition (REC) lobe and a nuclease (NUC) lobe; each of which further comprise domains described herein.
  • the REC lobe comprises the arginine-rich bridge helix (BH), the REC1 domain, and the REC2 domain.
  • the REC lobe does not share structural similarity with other known proteins, indicating that it is a Cas9-specific functional domain.
  • the BH domain is a long ac helix and arginine rich region and comprises amino acids 60-93 of the sequence of S. pyogenes Cas9.
  • the REC1 domain is important for recognition of the repeat:anti-repeat duplex, e.g., of a gRNA or a tracrRNA, and is therefore critical for Cas9 activity by recognizing the target sequence.
  • the REC1 domain comprises two REC1 motifs at amino acids 94 to 179 and 308 to 717 of the sequence of S. pyogenes Cas9. These two REC1 domains, though separated by the REC2 domain in the linear primary structure, assemble in the tertiary structure to form the REC1 domain.
  • the REC2 domain, or parts thereof, may also play a role in the recognition of the repeat:anti-repeat duplex.
  • the REC2 domain comprises amino acids 180-307 of the sequence of S. pyogenes Cas9.
  • the NUC lobe comprises the RuvC domain, the HNH domain, and the PAM-interacting (PI) domain.
  • the RuvC domain shares structural similarity to retroviral integrase superfamily members and cleaves a single strand, e.g., the non-complementary strand of the target nucleic acid molecule.
  • the RuvC domain is assembled from the three split RuvC motifs (RuvC I, RuvCII, and RuvCIII, which are often commonly referred to in the art as RuvCI domain, or N-terminal RuvC domain, RuvCII domain, and RuvCIII domain) at amino acids 1-59, 718-769, and 909-1098, respectively, of the sequence of S. pyogenes Cas9.
  • the three RuvC motifs are linearly separated by other domains in the primary structure, however in the tertiary structure, the three RuvC motifs assemble and form the RuvC domain.
  • the HNH domain shares structural similarity with HNH endonucleases, and cleaves a single strand, e.g., the complementary strand of the target nucleic acid molecule.
  • the HNH domain lies between the RuvC II-III motifs and comprises amino acids 775-908 of the sequence of S. pyogenes Cas9.
  • the PI domain interacts with the PAM of the target nucleic acid molecule, and comprises amino acids 1099-1368 of the sequence of S. pyogenes Cas9.
  • a Cas9 molecule or Cas9 polypeptide comprises an HNH-like domain and a RuvC-like domain.
  • cleavage activity is dependent on a RuvC-like domain and an HNH-like domain.
  • a Cas9 molecule or Cas9 polypeptide e.g., an eaCas9 molecule or eaCas9 polypeptide, can comprise one or more of the following domains: a RuvC-like domain and an HNH-like domain.
  • a Cas9 molecule or Cas9 polypeptide is an eaCas9 molecule or eaCas9 polypeptide and the eaCas9 molecule or eaCas9 polypeptide comprises a RuvC-like domain, e.g., a RuvC-like domain described below, and/or an HNH-like domain, e.g., an HNH-like domain described below.
  • a RuvC-like domain cleaves, a single strand, e.g., the non-complementary strand of the target nucleic acid molecule.
  • the Cas9 molecule or Cas9 polypeptide can include more than one RuvC-like domain (e.g., one, two, three or more RuvC-like domains).
  • a RuvC-like domain is at least 5, 6, 7, 8 amino acids in length but not more than 20, 19, 18, 17, 16 or 15 amino acids in length.
  • the Cas9 molecule or Cas9 polypeptide comprises an N-terminal RuvC-like domain of about 10 to 20 amino acids, e.g., about 15 amino acids in length.
  • Cas9 molecules or Cas9 polypeptide can comprise an N-terminal RuvC-like domain.
  • Exemplary N-terminal RuvC-like domains are described below.
  • an eaCas9 molecule or eaCas9 polypeptide comprises an N-terminal RuvC-like domain comprising an amino acid sequence of formula I:
  • X1 is selected from I, V, M, L and T (e.g., selected from I, V, and L);
  • X2 is selected from T, I, V, S, N, Y, E and L (e.g., selected from T, V, and I);
  • X3 is selected from N, S, G, A, D, T, R, M and F (e.g., A or N);
  • X4 is selected from S, Y, N and F (e.g., S);
  • X5 is selected from V, I, L, C, T and F (e.g., selected from V, I and L);
  • X6 is selected from W, F, V, Y, S and L (e.g., W);
  • X7 is selected from A, S, C, V and G (e.g., selected from A and S);
  • X8 is selected from V, I, L, A, M and H (e.g., selected from V, I, M and L);
  • X9 is selected from any amino acid or is absent (e.g., selected from T, V, I, L, A, F, S, A, Y, M and R, or, e.g., selected from T, V, I, L and A).
  • the N-terminal RuvC-like domain differs from a sequence of SEQ ID NO: EE1, by as many as 1 but no more than 2, 3, 4, or 5 residues.
  • the N-terminal RuvC-like domain is cleavage competent.
  • the N-terminal RuvC-like domain is cleavage incompetent.
  • a eaCas9 molecule or eaCas9 polypeptide comprises an N-terminal RuvC-like domain comprising an amino acid sequence of formula II:
  • X1 is selected from I, V, M, L and T (e.g., selected from I, V, and L);
  • X2 is selected from T, I, V, S, N, Y, E and L (e.g., selected from T, V, and I);
  • X3 is selected from N, S, G, A, D, T, R, M and F (e.g., A or N);
  • X5 is selected from V, I, L, C, T and F (e.g., selected from V, I and L);
  • X6 is selected from W, F, V, Y, S and L (e.g., W);
  • X7 is selected from A, S, C, V and G (e.g., selected from A and S);
  • X8 is selected from V, I, L, A, M and H (e.g., selected from V, I, M and L);
  • X9 is selected from any amino acid or is absent (e.g., selected from T, V, I, L, A, F, S, A, Y, M and R or selected from e.g., T, V, I, L and A).
  • the N-terminal RuvC-like domain differs from a sequence of SEQ ID NO: EE2 by as many as 1 but no more than 2, 3, 4, or 5 residues.
  • the N-terminal RuvC-like domain comprises an amino acid sequence of formula III:
  • X2 is selected from T, I, V, S, N, Y, E and L (e.g., selected from T, V, and I);
  • X3 is selected from N, S, G, A, D, T, R, M and F (e.g., A or N);
  • X8 is selected from V, I, L, A, M and H (e.g., selected from V, I, M and L);
  • X9 is selected from any amino acid or is absent (e.g., selected from T, V, I, L, A, F, S, A, Y, M and R or selected from e.g., T, V, I, L and A).
  • the N-terminal RuvC-like domain differs from a sequence of SEQ ID NO:AA1 by as many as 1 but no more than, 2, 3, 4, or 5 residues.
  • the N-terminal RuvC-like domain comprises an amino acid sequence of formula III:
  • X is a non-polar alkyl amino acid or a hydroxyl amino acid, e.g., X is selected from V, I, L and T.
  • the N-terminal RuvC-like domain differs from a sequence of SEQ ID NO: AA2 by as many as 1 but no more than, 2, 3, 4, or 5 residues.
  • the N-terminal RuvC-like domain differs from a sequence of an N-terminal RuvC like domain disclosed herein, as many as 1 but no more than 2, 3, 4, or 5 residues.
  • the Cas9 molecule or Cas9 polypeptide can comprise one or more additional RuvC-like domains.
  • the Cas9 molecule or Cas9 polypeptide can comprise two additional RuvC-like domains.
  • the additional RuvC-like domain is at least 5 amino acids in length and, e.g., less than 15 amino acids in length, e.g., 5 to 10 amino acids in length, e.g., 8 amino acids in length.
  • An additional RuvC-like domain can comprise an amino acid sequence:
  • X1 is V or H
  • X2 is I, L or V (e.g., I or V);
  • X3 is M or T.
  • the additional RuvC-like domain comprises the amino acid sequence:
  • X2 is I, L or V (e.g., I or V).
  • An additional RuvC-like domain can comprise an amino acid sequence:
  • X1 is H or L
  • X2 is R or V
  • X3 is E or V.
  • the additional RuvC-like domain comprises the amino acid sequence: H-H-A-H-D-A-Y-L (SEQ ID NO:AA6).
  • the additional RuvC-like domain differs from a sequence of SEQ ID NO: AA3, AA4, AA5, AA6 by as many as 1 but no more than 2, 3, 4, or 5 residues.
  • sequence flanking the N-terminal RuvC-like domain is a sequences of formula V:
  • X1′ is selected from K and P
  • X2′ is selected from V, L, I, and F (e.g., V, I and L);
  • X3′ is selected from G, A and S (e.g., G),
  • X4′ is selected from L, I, V and F (e.g., L);
  • X9′ is selected from D, E, N and Q;
  • Z is an N-terminal RuvC-like domain, e.g., as described above.
  • an HNH-like domain cleaves a single stranded complementary domain, e.g., a complementary strand of a double stranded nucleic acid molecule.
  • an HNH-like domain is at least 15, 20, 25 amino acids in length but not more than 40, 35 or 30 amino acids in length, e.g., 20 to 35 amino acids in length, e.g., 25 to 30 amino acids in length. Exemplary HNH-like domains are described below.
  • an eaCas9 molecule or eaCas9 polypeptide comprises an HNH-like domain having an amino acid sequence of formula VI:
  • X1 is selected from D, E, Q and N (e.g., D and E);
  • X2 is selected from L, I, R, Q, V, M and K;
  • X3 is selected from D and E;
  • X4 is selected from I, V, T, A and L (e.g., A, I and V);
  • X5 is selected from V, Y, I, L, F and W (e.g., V, I and L);
  • X6 is selected from Q, H, R, K, Y, I, L, F and W;
  • X7 is selected from S, A, D, T and K (e.g., S and A);
  • X8 is selected from F, L, V, K, Y, M, I, R, A, E, D and Q (e.g., F);
  • X9 is selected from L, R, T, I, V, S, C, Y, K, F and G;
  • X10 is selected from K, Q, Y, T, F, L, W, M, A, E, G, and S;
  • X11 is selected from D, S, N, R, L and T (e.g., D);
  • X12 is selected from D, N and S;
  • X13 is selected from S, A, T, G and R (e.g., S);
  • X14 is selected from I, L, F, S, R, Y, Q, W, D, K and H (e.g., I, L and F);
  • X15 is selected from D, S, I, N, E, A, H, F, L, Q, M, G, Y and V;
  • X16 is selected from K, L, R, M, T and F (e.g., L, R and K);
  • X17 is selected from V, L, I, A and T;
  • X18 is selected from L, I, V and A (e.g., L and I);
  • X19 is selected from T, V, C, E, S and A (e.g., T and V);
  • X20 is selected from R, F, T, W, E, L, N, C, K, V, S, Q, I, Y, H and A;
  • X21 is selected from S, P, R, K, N, A, H, Q, G and L;
  • X22 is selected from D, G, T, N, S, K, A, I, E, L, Q, R and Y;
  • X23 is selected from K, V, A, E, Y, I, C, L, S, T, G, K, M, D and F.
  • a HNH-like domain differs from a sequence of SEQ ID NO: AA8 by at least one but no more than, 2, 3, 4, or 5 residues.
  • the HNH-like domain is cleavage competent.
  • the HNH-like domain is cleavage incompetent.
  • an eaCas9 molecule or eaCas9 polypeptide comprises an HNH-like domain comprising an amino acid sequence of formula VII:
  • X1 is selected from D and E;
  • X2 is selected from L, I, R, Q, V, M and K;
  • X3 is selected from D and E;
  • X4 is selected from I, V, T, A and L (e.g., A, I and V);
  • X5 is selected from V, Y, I, L, F and W (e.g., V, I and L);
  • X6 is selected from Q, H, R, K, Y, I, L, F and W;
  • X8 is selected from F, L, V, K, Y, M, I, R, A, E, D and Q (e.g., F);
  • X9 is selected from L, R, T, I, V, S, C, Y, K, F and G;
  • X10 is selected from K, Q, Y, T, F, L, W, M, A, E, G, and S;
  • X14 is selected from I, L, F, S, R, Y, Q, W, D, K and H (e.g., I, L and F);
  • X15 is selected from D, S, I, N, E, A, H, F, L, Q, M, G, Y and V;
  • X19 is selected from T, V, C, E, S and A (e.g., T and V);
  • X20 is selected from R, F, T, W, E, L, N, C, K, V, S, Q, I, Y, H and A;
  • X21 is selected from S, P, R, K, N, A, H, Q, G and L;
  • X22 is selected from D, G, T, N, S, K, A, I, E, L, Q, R and Y;
  • X23 is selected from K, V, A, E, Y, I, C, L, S, T, G, K, M, D and F.
  • the HNH-like domain differs from a sequence of SEQ ID NO: AA9 by 1, 2, 3, 4, or 5 residues.
  • an eaCas9 molecule or eaCas9 polypeptide comprises an HNH-like domain comprising an amino acid sequence of formula VII:
  • X1 is selected from D and E;
  • X3 is selected from D and E;
  • X6 is selected from Q, H, R, K, Y, I, L and W;
  • X8 is selected from F, L, V, K, Y, M, I, R, A, E, D and Q (e.g., F);
  • X9 is selected from L, R, T, I, V, S, C, Y, K, F and G;
  • X10 is selected from K, Q, Y, T, F, L, W, M, A, E, G, and S;
  • X14 is selected from I, L, F, S, R, Y, Q, W, D, K and H (e.g., I, L and F);
  • X15 is selected from D, S, I, N, E, A, H, F, L, Q, M, G, Y and V;
  • X20 is selected from R, F, T, W, E, L, N, C, K, V, S, Q, I, Y, H and A;
  • X21 is selected from S, P, R, K, N, A, H, Q, G and L;
  • X22 is selected from D, G, T, N, S, K, A, I, E, L, Q, R and Y;
  • X23 is selected from K, V, A, E, Y, I, C, L, S, T, G, K, M, D and F.
  • the HNH-like domain differs from a sequence of SEQ ID NO: AA10 by 1, 2, 3, 4, or 5 residues.
  • an eaCas9 molecule or eaCas9 polypeptide comprises an HNH-like domain having an amino acid sequence of formula VIII:
  • X2 is selected from I and V;
  • X5 is selected from I and V;
  • X7 is selected from A and S;
  • X9 is selected from I and L;
  • X10 is selected from K and T;
  • X12 is selected from D and N;
  • X16 is selected from R, K and L; X19 is selected from T and V;
  • X20 is selected from S and R;
  • X22 is selected from K, D and A;
  • X23 is selected from E, K, G and N (e.g., the eaCas9 molecule or eaCas9 polypeptide can comprise an HNH-like domain as described herein).
  • the HNH-like domain differs from a sequence of SEQ ID NO: AA11 by as many as 1 but no more than 2, 3, 4, or 5 residues.
  • an eaCas9 molecule or eaCas9 polypeptide comprises the amino acid sequence of formula IX:
  • X1′ is selected from K and R;
  • X2′ is selected from V and T;
  • X3′ is selected from G and D;
  • X4′ is selected from E, Q and D;
  • X5′ is selected from E and D;
  • X6′ is selected from D, N and H;
  • X7′ is selected from Y, R and N;
  • X8′ is selected from Q, D and N; X9′ is selected from G and E;
  • X10′ is selected from S and G;
  • X11′ is selected from D and N;
  • Z is an HNH-like domain, e.g., as described above.
  • the eaCas9 molecule or eaCas9 polypeptide comprises an amino acid sequence that differs from a sequence of SEQ ID NO: AA12 by as many as 1 but no more than 2, 3, 4, or 5 residues.
  • the HNH-like domain differs from a sequence of an HNH-like domain disclosed herein, by as many as 1 but no more than 2, 3, 4, or 5 residues.
  • the HNH-like domain differs from a sequence of an HNH-like domain disclosed herein, by as many as 1 but no more than 2, 3, 4, or 5 residues.
  • the Cas9 molecule or Cas9 polypeptide is capable of cleaving a target nucleic acid molecule.
  • Cas9 molecules and Cas9 polypeptides can be engineered to alter nuclease cleavage (or other properties), e.g., to provide a Cas9 molecule or Cas9 polypeptide which is a nickase, or which lacks the ability to cleave target nucleic acid.
  • a Cas9 molecule or Cas9 polypeptide that is capable of cleaving a target nucleic acid molecule is referred to herein as an eaCas9 (an enzymatically active Cas9) molecule or eaCas9 polypeptide.
  • an eaCas9 molecule or eaCas9 polypeptide comprises one or more of the following enzymatic activities:
  • nickase activity i.e., the ability to cleave a single strand, e.g., the non-complementary strand or the complementary strand, of a nucleic acid molecule
  • a double stranded nuclease activity i.e., the ability to cleave both strands of a double stranded nucleic acid and create a double stranded break, which in an embodiment is the presence of two nickase activities;
  • a helicase activity i.e., the ability to unwind the helical structure of a double stranded nucleic acid.
  • an enzymatically active or an eaCas9 molecule or eaCas9 polypeptide cleaves both DNA strands and results in a double stranded break.
  • an eaCas9 molecule or eaCas9 polypeptide cleaves only one strand, e.g., the strand to which the gRNA hybridizes to, or the strand complementary to the strand the gRNA hybridizes with.
  • an eaCas9 molecule or eaCas9 polypeptide comprises cleavage activity associated with an HNH domain.
  • an eaCas9 molecule or eaCas9 polypeptide comprises cleavage activity associated with a RuvC domain. In an embodiment, an eaCas9 molecule or eaCas9 polypeptide comprises cleavage activity associated with an HNH domain and cleavage activity associated with a RuvC domain. In an embodiment, an eaCas9 molecule or eaCas9 polypeptide comprises an active, or cleavage competent, HNH domain and an inactive, or cleavage incompetent, RuvC domain. In an embodiment, an eaCas9 molecule or eaCas9 polypeptide comprises an inactive, or cleavage incompetent, HNH domain and an active, or cleavage competent, RuvC domain.
  • Cas9 molecules or Cas9 polypeptides have the ability to interact with a gRNA molecule, and in conjunction with the gRNA molecule localize to a core target domain, but are incapable of cleaving the target nucleic acid, or incapable of cleaving at efficient rates.
  • Cas9 molecules having no, or no substantial, cleavage activity are referred to herein as an eiCas9 molecule or eiCas9 polypeptide.
  • an eiCas9 molecule or eiCas9 polypeptide can lack cleavage activity or have substantially less, e.g., less than 20, 10, 5, 1 or 0.1% of the cleavage activity of a reference Cas9 molecule or eiCas9 polypeptide, as measured by an assay described herein.
  • a Cas9 molecule or Cas9 polypeptide is a polypeptide that can interact with a guide RNA (gRNA) molecule and, in concert with the gRNA molecule, localizes to a site which comprises a target domain, and in an embodiment, a PAM sequence.
  • gRNA guide RNA
  • the ability of an eaCas9 molecule or eaCas9 polypeptide to interact with and cleave a target nucleic acid is PAM sequence dependent.
  • a PAM sequence is a sequence in the target nucleic acid.
  • cleavage of the target nucleic acid occurs upstream from the PAM sequence.
  • EaCas9 molecules from different bacterial species can recognize different sequence motifs (e.g., PAM sequences).
  • an eaCas9 molecule of S. pyogenes recognizes the sequence motif NGG and directs cleavage of a target nucleic acid sequence 1 to 10, e.g., 3 to 5, base pairs upstream from that sequence.
  • N can be any nucleotide residue, e.g., any of A, G, C or T.
  • Cas9 molecules can be engineered to alter the PAM specificity of the Cas9 molecule.
  • Exemplary naturally occurring Cas9 molecules are described in Chylinski et al. (2013) RNA B IOLOGY 10:5, 727-737.
  • Such Cas9 molecules include Cas9 molecules of a cluster 1 bacterial family, cluster 2 bacterial family, cluster 3 bacterial family, cluster 4 bacterial family, cluster 5 bacterial family, cluster 6 bacterial family, a cluster 7 bacterial family, a cluster 8 bacterial family, a cluster 9 bacterial family, a cluster 10 bacterial family, a cluster 11 bacterial family, a cluster 12 bacterial family, a cluster 13 bacterial family, a cluster 14 bacterial family, a cluster 15 bacterial family, a cluster 16 bacterial family, a cluster 17 bacterial family, a cluster 18 bacterial family, a cluster 19 bacterial family, a cluster 20 bacterial family, a cluster 21 bacterial family, a cluster 22 bacterial family, a cluster 23 bacterial family, a cluster 24 bacterial family, a cluster 25 bacterial family, a cluster 26 bacterial family, a cluster 27 bacterial
  • Exemplary naturally occurring Cas9 molecules include a Cas9 molecule of a cluster 1 bacterial family.
  • Examples include a Cas9 molecule of: S. pyogenes (e.g., strain SF370, MGAS10270, MGAS10750, MGAS2096, MGAS315, MGAS5005, MGAS6180, MGAS9429, NZ131 and SSI-1), S. thermophilus (e.g., strain LMD-9), S. pseudoporcinus (e.g., strain SPIN 20026), S. mutans (e.g., strain UA159, NN2025), S. macacae (e.g., strain NCTC11558), S.
  • S. pyogenes e.g., strain SF370, MGAS10270, MGAS10750, MGAS2096, MGAS315, MGAS5005, MGAS6180, MGAS9429, NZ131 and SSI-1
  • gallolyticus e.g., strain UCN34, ATCC BAA-2069
  • S. equines e.g., strain ATCC 9812, MGCS 124
  • S. dysdalactiae e.g., strain GGS 124
  • S. bovis e.g., strain ATCC 70033
  • S. anginosus e.g., strain F0211
  • S. agalactiae e.g., strain NEM316, A909
  • Listeria monocytogenes e.g., strain F6854
  • Listeria innocua L.
  • Additional exemplary Cas9 molecules are a Cas9 molecule of Neisseria meningitidis (Hou et al., PNAS Early Edition 2013, 1-6 and a S. aureus cas9 molecule.
  • a Cas9 molecule or Cas9 polypeptide e.g., an eaCas9 molecule or eaCas9 polypeptide, comprises an amino acid sequence:
  • Cas9 molecule sequence is identical to any Cas9 molecule sequence described herein, or a naturally occurring Cas9 molecule sequence, e.g., a Cas9 molecule from a species listed herein or described in Chylinski et al. (2013) RNA B IOLOGY 10:5, 727-737; Hou et al., PNAS Early Edition 2013, 1-6.
  • the Cas9 molecule or Cas9 polypeptide comprises one or more of the following activities: a nickase activity; a double stranded cleavage activity (e.g., an endonuclease and/or exonuclease activity); a helicase activity; or the ability, together with a gRNA molecule, to localize to a target nucleic acid.
  • Cas9 molecules and Cas9 polypeptides described herein, e.g., naturally occurring Cas9 molecules, can possess any of a number of properties, including: nuclease activity (e.g., endonuclease and/or exonuclease activity); helicase activity; the ability to associate functionally with a gRNA molecule; and the ability to target (or localize to) a site on a nucleic acid (e.g., PAM recognition and specificity).
  • a Cas9 molecule or Cas9 polypeptide can include all or a subset of these properties.
  • a Cas9 molecule or Cas9 polypeptide has the ability to interact with a gRNA molecule and, in concert with the gRNA molecule, localize to a site in a nucleic acid.
  • Other activities e.g., PAM specificity, cleavage activity, or helicase activity can vary more widely in Cas9 molecules and Cas9 polypeptides.
  • Cas9 molecules include engineered Cas9 molecules and engineered Cas9 polypeptides (engineered, as used in this context, means merely that the Cas9 molecule or Cas9 polypeptide differs from a reference sequences, and implies no process or origin limitation).
  • An engineered Cas9 molecule or Cas9 polypeptide can comprise altered enzymatic properties, e.g., altered nuclease activity (as compared with a naturally occurring or other reference Cas9 molecule) or altered helicase activity.
  • an engineered Cas9 molecule or Cas9 polypeptide can have nickase activity (as opposed to double strand nuclease activity).
  • an engineered Cas9 molecule or Cas9 polypeptide can have an alteration that alters its size, e.g., a deletion of amino acid sequence that reduces its size, e.g., without significant effect on one or more, or any Cas9 activity.
  • an engineered Cas9 molecule or Cas9 polypeptide can comprise an alteration that affects PAM recognition.
  • an engineered Cas9 molecule can be altered to recognize a PAM sequence other than that recognized by the endogenous wild-type PI domain.
  • a Cas9 molecule or Cas9 polypeptide can differ in sequence from a naturally occurring Cas9 molecule but not have significant alteration in one or more Cas9 activities.
  • Cas9 molecules or Cas9 polypeptides with desired properties can be made in a number of ways, e.g., by alteration of a parental, e.g., naturally occurring Cas9 molecule or Cas9 polypeptide, to provide an altered Cas9 molecule or Cas9 polypeptide having a desired property.
  • a parental Cas9 molecule e.g., a naturally occurring or engineered Cas9 molecule
  • Such mutations and differences comprise: substitutions (e.g., conservative substitutions or substitutions of non-essential amino acids), insertions, or deletions.
  • a Cas9 molecule or Cas9 polypeptide can comprises one or more mutations or differences, e.g., at least 1, 2, 3, 4, 5, 10, 15, 20, 30, 40 or 50 mutations but less than 200, 100, or 80 mutations relative to a reference, e.g., a parental Cas9 molecule.
  • a mutation or mutations do not have a substantial effect on a Cas9 activity, e.g. a Cas9 activity described herein. In an embodiment, a mutation or mutations have a substantial effect on a Cas9 activity, e.g. a Cas9 activity described herein.
  • a Cas9 molecule or Cas9 polypeptide comprises a cleavage property that differs from naturally occurring Cas9 molecules, e.g., that differs from the naturally occurring Cas9 molecule having the closest homology.
  • a Cas9 molecule or Cas9 polypeptide can differ from a naturally occurring Cas9 molecule, e.g., a Cas9 molecule of S.
  • pyogenes as follows: its ability to modulate, e.g., decreased or increased, cleavage of a double stranded nucleic acid (endonuclease and/or exonuclease activity), e.g., as compared to a naturally occurring Cas9 molecule (e.g., a Cas9 molecule of S.
  • pyogenes its ability to modulate, e.g., decreased or increased, cleavage of a single strand of a nucleic acid, e.g., a non-complementary strand of a nucleic acid molecule or a complementary strand of a nucleic acid molecule (nickase activity), e.g., as compared to a naturally occurring Cas9 molecule (e.g., a Cas9 molecule of S. pyogenes ); or the ability to cleave a nucleic acid molecule, e.g., a double stranded or single stranded nucleic acid molecule, can be eliminated.
  • exemplary Cas9 activities comprise one or more of PAM specificity, cleavage activity, and helicase activity.
  • a mutation(s) can be present, e.g., in: one or more RuvC domains, e.g., an N-terminal RuvC domain; an HNH domain; a region outside the RuvC domains and the HNH domain.
  • a mutation(s) is present in a RuvC domain.
  • a mutation(s) is present in an HNH domain.
  • mutations are present in both a RuvC domain and an HNH domain.
  • Exemplary mutations that may be made in the RuvC domain or HNH domain with reference to the S. pyogenes Cas9 sequence include: D10A, E762A, H840A, N854A, N863A and/or D986A.
  • Exemplary mutations that may be made in the RuvC domain with reference to the S. aureus Cas9 sequence include: N580A.
  • a Cas9 molecule is an eiCas9 molecule comprising one or more differences in a RuvC domain and/or in an HNH domain as compared to a reference Cas9 molecule, and the eiCas9 molecule does not cleave a nucleic acid, or cleaves with significantly less efficiency than does wild type, e.g., when compared with wild type in a cleavage assay, e.g., as described herein, cuts with less than 50, 25, 10, or 1% of a reference Cas9 molecule, as measured by an assay described herein.
  • a “non-essential” amino acid residue is a residue that can be altered from the wild-type sequence of a Cas9 molecule, e.g., a naturally occurring Cas9 molecule, e.g., an eaCas9 molecule, without abolishing or more preferably, without substantially altering a Cas9 activity (e.g., cleavage activity), whereas changing an “essential” amino acid residue results in a substantial loss of activity (e.g., cleavage activity).
  • a Cas9 molecule comprises a cleavage property that differs from naturally occurring Cas9 molecules, e.g., that differs from the naturally occurring Cas9 molecule having the closest homology.
  • a Cas9 molecule can differ from naturally occurring Cas9 molecules, e.g., a Cas9 molecule of S. aureus, S. pyogenes , or C.
  • jejuni as follows: its ability to modulate, e.g., decreased or increased, cleavage of a double stranded break (endonuclease and/or exonuclease activity), e.g., as compared to a naturally occurring Cas9 molecule (e.g., a Cas9 molecule of S. aureus, S. pyogenes , or C.
  • a naturally occurring Cas9 molecule e.g., a Cas9 molecule of S. aureus, S. pyogenes , or C.
  • jejuni its ability to modulate, e.g., decreased or increased, cleavage of a single strand of a nucleic acid, e.g., a non-complimentary strand of a nucleic acid molecule or a complementary strand of a nucleic acid molecule (nickase activity), e.g., as compared to a naturally occurring Cas9 molecule (e.g., a Cas9 molecule of S. aureus, S. pyogenes , or C. jejuni ); or the ability to cleave a nucleic acid molecule, e.g., a double stranded or single stranded nucleic acid molecule, can be eliminated.
  • a naturally occurring Cas9 molecule e.g., a Cas9 molecule of S. aureus, S. pyogenes , or C. jejuni
  • the ability to cleave a nucleic acid molecule e
  • the altered Cas9 molecule is an eaCas9 molecule comprising one or more of the following activities: cleavage activity associated with a RuvC domain; cleavage activity associated with an HNH domain; cleavage activity associated with an HNH domain and cleavage activity associated with a RuvC domain.
  • the altered Cas9 molecule is an eiCas9 molecule which does not cleave a nucleic acid molecule (either double stranded or single stranded nucleic acid molecules) or cleaves a nucleic acid molecule with significantly less efficiency, e.g., less than 20, 10, 5, 1 or 0.1% of the cleavage activity of a reference Cas9 molecule, e.g., as measured by an assay described herein.
  • the reference Cas9 molecule can be a naturally occurring unmodified Cas9 molecule, e.g., a naturally occurring Cas9 molecule such as a Cas9 molecule of S. pyogenes, S. thermophilus, S. aureus, C.
  • the reference Cas9 molecule is the naturally occurring Cas9 molecule having the closest sequence identity or homology.
  • the eiCas9 molecule lacks substantial cleavage activity associated with a RuvC domain and cleavage activity associated with an HNH domain.
  • the altered Cas9 molecule or Cas9 polypeptide can be a fusion, e.g., of two of more different Cas9 molecules, e.g., of two or more naturally occurring Cas9 molecules of different species.
  • a fragment of a naturally occurring Cas9 molecule of one species can be fused to a fragment of a Cas9 molecule of a second species.
  • a fragment of a Cas9 molecule of S. pyogenes comprising an N-terminal RuvC-like domain can be fused to a fragment of Cas9 molecule of a species other than S. pyogenes (e.g., S. thermophilus ) comprising an HNH-like domain.
  • Naturally occurring Cas9 molecules can recognize specific PAM sequences, for example the PAM recognition sequences described above for, e.g., S. pyogenes, S. thermophilus, S. mutans, S. aureus and N. meningitidis.
  • a Cas9 molecule or Cas9 polypeptide has the same PAM specificities as a naturally occurring Cas9 molecule.
  • a Cas9 molecule or Cas9 polypeptide has a PAM specificity not associated with a naturally occurring Cas9 molecule, or a PAM specificity not associated with the naturally occurring Cas9 molecule to which it has the closest sequence homology.
  • a naturally occurring Cas9 molecule can be altered, e.g., to alter PAM recognition, e.g., to alter the PAM sequence that the Cas9 molecule or Cas9 polypeptide recognizes to decrease off target sites and/or improve specificity; or eliminate a PAM recognition requirement.
  • a Cas9 molecule or Cas9 polypeptide can be altered, e.g., to increase length of PAM recognition sequence and/or improve Cas9 specificity to high level of identity (e.g., 98%, 99% or 100% match between gRNA and a PAM sequence), e.g., to decrease off target sites and increase specificity.
  • the length of the PAM recognition sequence is at least 4, 5, 6, 7, 8, 9, 10 or 15 amino acids in length.
  • the Cas9 specificity requires at least 90%, 95%, 96%, 97%, 98%, 99% or more homology between the gRNA and the PAM sequence.
  • Cas9 molecules or Cas9 polypeptides that recognize different PAM sequences and/or have reduced off-target activity can be generated using directed evolution. Exemplary methods and systems that can be used for directed evolution of Cas9 molecules are described, e.g., in Esvelt et al. (2011) N ATURE 472(7344): 499-503. Candidate Cas9 molecules can be evaluated, e.g., by methods described in Section VII.
  • a synthetic Cas9 molecule or Syn-Cas9 molecule
  • synthetic Cas9 polypeptide or syn-Cas9 polypeptide
  • a synthetic Cas9 molecule refers to a Cas9 molecule or Cas9 polypeptide that comprises a Cas9 core domain from one bacterial species and a functional altered PI domain, i.e., a PI domain other than that naturally associated with the Cas9 core domain, e.g., from a different bacterial species.
  • the altered PI domain recognizes a PAM sequence that is different from the PAM sequence recognized by the naturally-occurring Cas9 from which the Cas9 core domain is derived. In an embodiment, the altered PI domain recognizes the same PAM sequence recognized by the naturally-occurring Cas9 from which the Cas9 core domain is derived, but with different affinity or specificity.
  • a Syn-Cas9 molecule or Syn-Cas9 polypetide can be, respectively, a Syn-eaCas9 molecule or Syn-eaCas9 polypeptide or a Syn-eiCas9 molecule Syn-eiCas9 polypeptide.
  • Syn-Cas9 molecule Syn-Cas9 polypetide comprises:
  • a Cas9 core domain e.g., a Cas9 core domain from Table 100 or 200, e.g., a S. aureus, S. pyogenes , or C. jejuni Cas9 core domain;
  • the RKR motif (the PAM binding motif) of said altered PI domain comprises: differences at 1, 2, or 3 amino acid residues; a difference in amino acid sequence at the first, second, or third position; differences in amino acid sequence at the first and second positions, the first and third positions, or the second and third positions; as compared with the sequence of the RKR motif of the native or endogenous PI domain associated with the Cas9 core domain.
  • the Cas9 core domain comprises the Cas9 core domain from a species X Cas9 from Table 100 and said altered PI domain comprises a PI domain from a species Y Cas9 from Table 100.
  • the RKR motif of the species X Cas9 is other than the RKR motif of the species Y Cas9.
  • the RKR motif of the altered PI domain is selected from XXY, XNG, and XNQ.
  • the altered PI domain has at least 60, 70, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99 or 100% homology with the amino acid sequence of a naturally occurring PI domain of said species Y from Table 100.
  • the altered PI domain differs by no more than 50, 40, 30, 25, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residue from the amino acid sequence of a naturally occurring PI domain of said second species from Table 100.
  • the Cas9 core domain comprises a S. aureus core domain and altered PI domain comprises: an A. denitrificans PI domain; a C. jejuni PI domain; a H. mustelae PI domain; or an altered PI domain of species X PI domain, wherein species X is selected from Table 400 or Table 500.
  • the Cas9 core domain comprises a S. pyogenes core domain and the altered PI domain comprises: an A. denitrificans PI domain; a C. jejuni PI domain; a H. mustelae PI domain; or an altered PI domain of species X PI domain, wherein species X is selected from Table 400 or Table 500.
  • the Cas9 core domain comprises a C. jejuni core domain and the altered PI domain comprises: an A. denitrificans PI domain; a H. mustelae PI domain; or an altered PI domain of species X PI domain, wherein species X is selected from Table 400 or Table 500.
  • the Cas9 molecule further comprises a linker disposed between said Cas9 core domain and said altered PI domain.
  • the linker comprises: a linker described elsewhere herein disposed between the Cas9 core domain and the heterologous PI domain.
  • Tables 400 and 500 Exemplary altered PI domains for use in Syn-Cas9 molecules are described in Tables 400 and 500.
  • the sequences for the 83 Cas9 orthologs referenced in Tables 400 and 500 are provided in Table 100.
  • Table 250 provides the Cas9 orthologs with known PAM sequences and the corresponding RKR motif.
  • a Syn-Cas9 molecule may also be size-optimized, e.g., the Syn-Cas9 molecule comprises one or more deletions, and optionally one or more linkers disposed between the amino acid residues flanking the deletions. In an embodiment, a Syn-Cas9 molecule comprises a REC deletion.
  • Engineered Cas9 molecules and engineered Cas9 polypeptides, as described herein, include a Cas9 molecule or Cas9 polypeptide comprising a deletion that reduces the size of the molecule while still retaining desired Cas9 properties, e.g., essentially native conformation, Cas9 nuclease activity, and/or target nucleic acid molecule recognition.
  • Cas9 molecules or Cas9 polypeptides comprising one or more deletions, and optionally one or more linkers, wherein a linker is disposed between the amino acid residues that flank the deletion.
  • a Cas9 molecule e.g., a S. aureus, S. pyogenes , or C. jejuni , Cas9 molecule, having a deletion is smaller, e.g., has reduced number of amino acids, than the corresponding naturally-occurring Cas9 molecule.
  • the smaller size of the Cas9 molecules allows increased flexibility for delivery methods, and thereby increases utility for genome-editing.
  • a Cas9 molecule can comprise one or more deletions that do not substantially affect or decrease the activity of the resultant Cas9 molecules described herein. Activities that are retained in the Cas9 molecules comprising a deletion as described herein include one or more of the following:
  • a nickase activity i.e., the ability to cleave a single strand, e.g., the non-complementary strand or the complementary strand, of a nucleic acid molecule
  • a double stranded nuclease activity i.e., the ability to cleave both strands of a double stranded nucleic acid and create a double stranded break, which in an embodiment is the presence of two nickase activities;
  • a helicase activity i.e., the ability to unwind the helical structure of a double stranded nucleic acid
  • nucleic acid molecule e.g., a target nucleic acid or a gRNA.
  • Activity of the Cas9 molecules described herein can be assessed using the activity assays described herein or in the art.
  • Suitable regions of Cas9 molecules for deletion can be identified by a variety of methods.
  • Naturally-occurring orthologous Cas9 molecules from various bacterial species e.g., any one of those listed in Table 100, can be modeled onto the crystal structure of S. pyogenes Cas9 (Nishimasu et al. (2014) C ELL , 156: 935-949) to examine the level of conservation across the selected Cas9 orthologs with respect to the three-dimensional conformation of the protein.
  • regions that are located spatially distant from regions involved in Cas9 activity e.g., the interface with a target nucleic acid molecule and/or gRNA, represent regions or domains that are candidates for deletion without substantially affecting or decreasing Cas9 activity.
  • a REC-optimized Cas9 molecule refers to a Cas9 molecule that comprises a deletion in one or both of the REC2 domain and the RE1 CT domain (collectively a REC deletion), wherein the deletion comprises at least 10% of the amino acid residues in the cognate domain.
  • a REC-optimized Cas9 molecule can be an eaCas9 molecule or an eiCas9 molecule.
  • An exemplary REC-optimized Cas9 molecule comprises:
  • a linker is disposed between the amino acid residues that flank the deletion.
  • a Cas9 molecule includes only one deletion, or only two deletions.
  • a Cas9 molecule can comprise a REC2 deletion and a REC1 CT deletion.
  • a Cas9 molecule can comprise a REC2 deletion and a REC1 SUB deletion.
  • the deletion will contain at least 10% of the amino acids in the cognate domain, e.g., a REC2 deletion will include at least 10% of the amino acids in the REC2 domain.
  • a deletion can comprise: at least 10, 20, 30, 40, 50, 60, 70, 80, or 90% of the amino acid residues of its cognate domain; all of the amino acid residues of its cognate domain; an amino acid residue outside its cognate domain; a plurality of amino acid residues outside its cognate domain; the amino acid residue immediately N terminal to its cognate domain; the amino acid residue immediately C terminal to its cognate domain; the amino acid residue immediately N terminal to its cognate and the amino acid residue immediately C terminal to its cognate domain; a plurality of, e.g., up to 5, 10, 15, or 20, amino acid residues N terminal to its cognate domain; a plurality of, e.g., up to 5, 10, 15, or 20, amino acid residues C terminal to its cognate domain; a plurality of, e.g., up to 5, 10, 15, or 20, amino acid residue
  • a deletion does not extend beyond: its cognate domain; the N terminal amino acid residue of its cognate domain; the C terminal amino acid residue of its cognate domain.
  • a REC-optimized Cas9 molecule can include a linker disposed between the amino acid residues that flank the deletion.
  • Linkers for use in generating recombinant proteins, e.g., multi-domain proteins, are known in the art (Chen et al. (2013) A DV . D RUG D ELIVERY R EV . 65:1357-69). Any linkers known in the art that maintain the conformation or native fold of the Cas9 molecule (thereby retaining Cas9 activity) can be used.
  • properties of linkers such as length, hydrophobicity, intrinsic properties of the amino acids residues themselves, and secondary structure should be considered in the context of the goal to maintain native conformation and functional activity of Cas9.
  • linkers Any linkers known in the art that maintain the conformation or native fold of the Cas9 molecule (thereby retaining Cas9 activity) can be used.
  • properties of linkers such as length, hydrophobicity, intrinsic properties of the amino acids residues themselves, and secondary structure should be considered in the context of the goal to maintain native conformation and functional activity of Cas9.
  • a flexible linker can be utilized in the Cas9 molecules described herein.
  • Flexible linkers allow a certain degree of movement and/or interaction within and between the joined domains or regions of the protein.
  • flexible linkers are composed of small, non-polar (e.g., Gly) or polar (e.g., Ser or Thr) amino acids. The small size of these amino acids provides flexibility and allows mobility of the connected domains or regions.
  • Ser or Thr can help maintain the stability of the linker in aqueous solutions by hydrogen bonding with the water molecules, thereby reducing unfavorable interactions between the linker and the other protein moieties.
  • Commonly used flexible linkers are comprised of sequences that primarily consist of Gly and Ser residues.
  • these flexible linkers consist of repeating units of a combination of Gly and Ser residues, e.g., (GGS) x , where the number of repeating units, e.g., x, can be optimized to achieve the appropriate separation of other domains or regions of the protein.
  • a rigid linker may be preferred if there is significant distance between the joined domains or regions, or to maintain a fixed distance between the joined domains or regions of a protein and independent functions of the domains/regions.
  • Rigid linkers often have defined secondary structure, e.g., alpha helix, or other stabilizing interactions, e.g., salt bridges and disulfide bonds.
  • Rigid linkers commonly contain multiple Pro residues, or repeating combinations of Glu-Pro or Lys-Pro because Pro imposes a strong conformation constraint due to its structure.
  • the linker can comprise an amino acid residue, e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acid residues. Typically, the linker will comprises less than 10, 20 or 30 amino acid residues. Typically, the linker is less than 50, 40, 30, 20, 10, or 5% of the length of the deleted sequence.
  • Suitable linkers include: [Gly-Ser] x , wherein x is 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; [Gly-Gly-Ser] x , wherein x is 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; [Gly-Gly-Ser]; [Gly-Ser-Gly-Ser] x , wherein x is 1, 2, 3, 4, or 5; [Gly-Ser-Gly-Ser]; (GSAGSAAGSGEF) x , wherein x is 1, 2, 3 or 4; (SIVAQLSRPDPA) x , wherein x is 1, 2, 3 or 4; or an XTEN sequence, e.g., the XTEN sequence of SEQ ID NO: _, or a sequence that differs therefrom by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 amino acid residues.
  • linker comprises an amino acid sequence other than a sequence within REC2.
  • a REC-optimized Cas9 molecule comprises an amino acid sequence that, other than any REC deletion and associated linker, has at least 50, 55, 60, 65, 70, 75, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% homology with the amino acid sequence of a naturally occurring Cas9, e.g., a Cas9 molecule described in Table 100, e.g., a S. aureus Cas9 molecule, a S. pyogenes Cas9 molecule, or a C. jejuni Cas9 molecule.
  • a REC-optimized Cas9 molecule comprises an amino acid sequence that, other than any REC deletion and associated linker, differs by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25 amino acid residues from the amino acid sequence of a naturally occurring Cas9, e.g., a Cas9 molecule described in Table 100, e.g., a S. aureus Cas9 molecule, a S. pyogenes Cas9 molecule, or a C. jejuni Cas9 molecule.
  • a naturally occurring Cas9 e.g., a Cas9 molecule described in Table 100, e.g., a S. aureus Cas9 molecule, a S. pyogenes Cas9 molecule, or a C. jejuni Cas9 molecule.
  • a REC-optimized Cas9 molecule comprises an amino acid sequence that, other than any REC deletion and associate linker, differs by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25% of the amino acid residues from the amino acid sequence of a naturally occurring Cas9, e.g., a Cas9 molecule described in Table 100, e.g., a S. aureus Cas9 molecule, a S. pyogenes Cas9 molecule, or a C. jejuni Cas9 molecule.
  • sequence comparison typically one sequence acts as a reference sequence, to which test sequences are compared.
  • test and reference sequences are entered into a computer, subsequence coordinates are designated, if necessary, and sequence algorithm program parameters are designated. Default program parameters can be used, or alternative parameters can be designated.
  • sequence comparison algorithm then calculates the percent sequence identities for the test sequences relative to the reference sequence, based on the program parameters. Methods of alignment of sequences for comparison are well known in the art. Optimal alignment of sequences for comparison can be conducted, e.g., by the local homology algorithm of Smith and Waterman (1970) A DV . A PPL . M ATH .
  • BLAST and BLAST 2.0 algorithms Two examples of algorithms that are suitable for determining percent sequence identity and sequence similarity are the BLAST and BLAST 2.0 algorithms, which are described in Altschul et al. (1977) N UC . A CIDS R ES . 25:3389-3402; and Altschul et al. (1990) J. M OL . B IOL . 215: 403-410, respectively.
  • Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information.
  • the percent identity between two amino acid sequences can also be determined using the algorithm of E. Meyers and W. Miller (1988) C OMPUT . A PPL . B IOSCI .
  • the percent identity between two amino acid sequences can be determined using the Needleman and Wunsch (1970) J. M OL . B IOL . 48:444-453 algorithm which has been incorporated into the GAP program in the GCG software package (available at www.gcg.com), using either a Blossom 62 matrix or a PAM250 matrix, and a gap weight of 16, 14, 12, 10, 8, 6, or 4 and a length weight of 1, 2, 3, 4, 5, or 6.
  • Sequence information for exemplary REC deletions are provided for 83 naturally-occurring Cas9 orthologs in Table 100.
  • Exemplary Cas9 core domains are provided in Table 200.
  • Exemplary PI domains e.g., altered PI domains, are provided in Tables 400 and 500.
  • PI Start PI Stop (AA pos) (AA pos) Start and Stop numbers refer to Length RKR the sequences in of PI motif Strain Name Table 100 (AA) (AA) Akkermansia muciniphila ATCC BAA-835 871 1101 231 ALK Ralstonia syzygii R24 821 1062 242 APY Cand.
  • Nucleic acids encoding the Cas9 fusion molecules, the Cas9 molecules or Cas9 polypeptides, e.g., an eaCas9 molecule or eaCas9 polypeptides, are provided herein.
  • a nucleic acid encoding a Cas9 fusion molecule, a Cas9 molecule, or Cas9 polypeptide can be a synthetic nucleic acid sequence.
  • the synthetic nucleic acid molecule can be chemically modified, e.g., as described in Section XI.
  • the mRNA, e.g., coding for a Cas9 fusion molecule, Cas9 molecule, or Cas9 polypeptide, disclosed herein has one or more, e.g., all, of the following properties: it is capped, polyadenylated, substituted with 5-methylcytidine and/or pseudouridine.
  • the synthetic nucleic acid sequence can be codon optimized, e.g., at least one non-common codon or less-common codon has been replaced by a codon that is common in the host cell.
  • the synthetic nucleic acid can direct the synthesis of an optimized messenger mRNA, e.g., optimized for expression in a mammalian expression system, e.g., described herein.
  • a nucleic acid encoding a Cas9 fusion molecule, a Cas9 molecule, or a Cas9 polypeptide may comprise a nuclear localization sequence (NLS). Nuclear localization sequences are known in the art.
  • N. meningitidis Cas9 molecule Provided below is the corresponding amino acid sequence of a N. meningitidis Cas9 molecule.
  • Cas molecules or Cas polypeptides can be used to practice the inventions disclosed herein.
  • Cas molecules of Type II Cas systems are used.
  • Cas molecules of other Cas systems are used.
  • Type I or Type III Cas molecules may be used.
  • Exemplary Cas molecules (and Cas systems) are described, e.g., in Haft et al. (2005) PL O S C OMPUTATIONAL B IOLOGY 1(6): e60, and in Makarova et al. (2011) N ATURE R EVIEW M ICROBIOLOGY 9:467-477, the contents of which are incorporated herein by reference in their entirety.
  • Exemplary Cas molecules (and Cas systems) are also shown in Table 600.
  • a linker covalently connects the Cas9 molecule to the template binding domain (e.g., a DNA binding domain).
  • a linker is a short peptide sequence that connects protein domains. In another embodiment, a linker is a short peptide sequence that connects a protein domain and a nucleic acid (e.g., DNA or RNA). In an embodiment, a linker is a short peptide sequence that connects a protein domain and a small molecule. In an embodiment, a linker is a short peptide sequence that connects two small molecules. Linkers are often composed of flexible residues like glycine and serine so that the adjacent protein domains are free to move relative to one another.
  • the linker has sufficient length and flexibility to allow the Cas9 molecule to bind to a target nucleic acid and simultaneously allow the target binding domain to associate with a nucleic acid template system, e.g., so that one binding event does not sterically block the other, and/or so that two adjacent domains do not sterically interfere with one another.
  • the linker can attach to the C-terminus or N-terminus of the Cas9 molecule.
  • the linker can attach to the C-terminus or N-terminus of the template binding domain.
  • the linker can attach to the C-terminus or N-terminus of the template binding domain partner.
  • the linker can attach to the C-terminus or N-terminus of the template nucleic acid.
  • the linker length is from about 6 to 60 amino acids.
  • the linker may be, e.g., 6-10, 10-15, 15-20, 20-30, 30-40, 40-50, or 50-60 amino acids in length.
  • the linker may be, e.g., at least 6, 7, 8, 9, 10, 15, 20, 30, 40, or 50 amino acids in length.
  • the linker is, e.g., at most 7, 8, 9, 10, 15, 20, 30, 40, 50, or 60 amino acids in length. Ranges comprising any combination of these endpoints are also envisioned.
  • the linker is encoded by a nucleic acid sequence of 6 to 60 nucleotides or base pairs.
  • the nucleic acid may be, e.g., 6-10, 10-15, 15-20, 20-30, 30-40, 40-50, or 50-60 nucleotides in length.
  • the linker may be, e.g., at least 6, 7, 8, 9, 10, 15, 20, 30, 40, or 50 nucleotides in length.
  • the linker is, e.g., at most 7, 8, 9, 10, 15, 20, 30, 40, 50, or 60 nucleotides in length. Ranges comprising any combination of these endpoints are also envisioned.
  • the linker comprises glycine and serine residues. In some embodiments the linker consists of glycine and serine residues.
  • the linker may comprise one of more modules such as GGS, GSGS, GGGS, GGGGS or GGSG.
  • the linker comprises a plurality of modules comprising glycine and serine, e.g., at least 2, 3, 4, 5, 10, or 15 of these modules, and/or at most 3, 4, 5, 10, 15, or 20 of these modules, or any combination of these endpoints.
  • each module in the linker has the same sequence, and in other embodiments, at least two modules in a linker have different sequences from each other.
  • the linker is an XTEN linker or a variation of an XTEN linker such as SGSETPGTSESA, SGSETPGTSESATPES, or SGSETPGTSESATPEGGSGGS.
  • Linkers can comprise a direct bond or an atom such as, e.g., an oxygen (O) or sulfur (S), a unit such as —NR— wherein R is hydrogen or alkyl, —C(O)—, —C(O)O—, —C(O)NH—, SO, SO 2 , —SO 2 NH— or a chain of atoms, such as substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, arylalkyl, heteroarylalkyl.
  • one or more methylenes in the chain of atoms can be replaced with one or more of O, S, S(O), SO 2 , —SO 2 NH—, —NR—, —NR 2 , —C(O)—, —C(O)O—, —C(O)NH—, a cleavable linking group, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, and substituted or unsubstituted heterocyclic.
  • the template binding domain is attached to the Cas9 molecule through a linker that is itself stable under physiological conditions, such as an alkylene chain, and does not result in release of the template binding domain from the Cas9 molecule for at least 2, 3, 4, 5, 10, 15, 24 or 48 hours or for at least 1, 2, 3, 4, 5 or 10 days when administered to a subject.
  • the template binding domain and the Cas9 molecule comprise residues of a functional groups through which reaction and linkage of the template binding domain to the Cas9 molecule was achieved.
  • the functional groups which may be the same or different, terminal or internal, of the template binding domain or Cas9 molecule comprise an amino, acid, imidazole, hydroxyl, thio, acyl halide, —HC ⁇ CH—, —C ⁇ C— group, or derivative thereof.
  • the linker comprises a hydrocarbylene group wherein one or more methylene groups is optionally replaced by a group Y (provided that none of the Y groups are adjacent to each other), wherein each Y, independently for each occurrence, is selected from, substituted or unsubstituted aryl, heteroaryl, cycloalkyl, heterocycloalkyl, or —O—, C( ⁇ X) (wherein X is NR 1 , O or S), —NR 1 —, —NR 1 C(O)—, —C(O)NR 1 —, —S(O) n —, —NR 1 S(O) n —, —S(O) n NR 1 —, —NR 1 C(O)—NR 1 —; and R 1 , independently for each occurrence, represents H or a lower alkyl and wherein n is 0, 1, or 2.
  • the linker comprises an alkylene moiety or a heteroalkylene moiety (e.g., an alkylene glycol moiety such as ethylene glycol).
  • a linker comprises a poly-L-glutamic acid, polylactic acid, poly(ethyleneimine), an oligosaccharide, an amino acid (e.g., glycine), an amino acid chain, or any other suitable linkage.
  • the linker groups can be biologically inactive, such as a PEG, polyglycolic acid, or polylactic acid chain.
  • the linker group represents a derivatized or non-derivatized amino acid (e.g., glycine).
  • the Cas9 molecule attached to the linker may be any Cas9 molecule described herein, e.g., a Cas9 molecule of Section III, e.g., a nickase or a Cas9 molecule capable of making a double stranded break.
  • the template binding domain attached to the linker may be any template binding domain described herein, e.g., a template binding domain of Section V, e.g., a DNA-binding domain.
  • the template binding domain comprises a plurality of domains that bind a template binding domain partner, e.g., DNA-binding molecules.
  • a template binding domain partner e.g., DNA-binding molecules.
  • a first DNA-binding molecule and a second DNA-binding molecule may, on their own, be poor choices because each has a low affinity and optionally also a short recognition sequence that is present in multiple copies in the genome of the cell to be altered.
  • the first and second molecules could give a higher affinity and optionally also a longer recognition sequence that is rarer in the genome of the cell to be altered.
  • the template binding domain comprises a plurality of domains that bind a template binding domain partner, e.g., DNA-binding molecules.
  • the template binding domain may comprise two or more, three or more, four or more, five or more, ten or more, 2-4, 4-6, 6-8, or 8-10 domains that bind a template binding domain partner, e.g., DNA-binding molecules.
  • a template binding domain partner e.g., DNA-binding molecules.
  • the template binding domain can be covalently bound to the Cas9 molecule, e.g., using a linker.
  • the template binding domain and the Cas9 can be part of the same fusion protein.
  • the Cas9 molecule and the template binding domain are part of the same gene and are transcribed to form a single mRNA which is then translated into a single protein.
  • the Cas9 molecule and the template binding domain are produced separately and then joined covalently or non-covalently.
  • a template binding domain which binds non-covalently to the Cas9 molecule is a template-binding domain that comprises a region with specific affinity for Cas9, e.g., an antibody that recognizes Cas9, e.g., an scFv antibody or another type of antibody that contains sufficient CDR sequences for binding to Cas9.
  • the template binding domain is derived from a wild-type protein.
  • the template binding domain may be a fragment of a wild type protein, a mutagenized wild type protein, a mutagenized wild-type protein fragment, a synthetic protein that has been modeled after the three dimensional structure of a naturally-occurring protein.
  • the template binding domain is mutagenized to increase its affinity for a template binding domain partner.
  • the template binding domain is mutagenized to decrease its affinity for a template binding domain partner.
  • the template binding domain is a polypeptide, e.g., a protein or protein domain.
  • This polypeptide can bind to the major groove of a target DNA sequence and/or a minor groove of a target DNA sequence. It can comprise one or more of the following domains: zinc finger, helix-turn-helix, leucine zipper, winged helix, winged helix turn helix, helix-loop-helix, HMG-box, and Wor3 domain. It can bind single stranded DNA or double stranded DNA.
  • the DNA-binding polypeptide is identical in sequence to a wild-type protein, and in other embodiments it comprises one or more mutations, e.g., deletions, relative to a wild-type protein.
  • the DNA-binding polypeptide comprises a mutation relative to a wild-type DNA-binding protein.
  • the DNA-binding polypeptide is optionally mutated to a constitutively active form.
  • the DNA-binding polypeptide can also be mutated to a constitutively active form.
  • the DNA-binding polypeptide carries a deletion relative to a wild-type protein, e.g., a transcriptional activation or repression domain or a catalytic domain is removed. In some embodiments, the DNA-binding polypeptide consists only of the DNA-binding region of the corresponding wild-type DNA-binding protein.
  • the DNA-binding polypeptide recognizes chemically modified DNA, e.g., methylated DNA. In some embodiments, the DNA-binding polypeptide recognizes a chemical modification that is rare in or absent from the genome of the cell to be altered. This can help avoid the DNA-binding polypeptide non-specifically binding to the cell's genome.
  • the DNA-binding polypeptide is, or is derived from, a DNA-binding protein from an operon, e.g., a bacterial operon.
  • the DNA-binding polypeptide may be, e.g., a repressor or an activator in the context of the operon.
  • the DNA-binding polypeptide will not activate or repress transcription in the methods described herein. This can be achieved by, e.g., mutating transcriptional regulation domains, or choosing a DNA-binding polypeptide that does not engage the transcriptional machinery of the cell to be altered.
  • DNA-binding peptide when altering the genome of a human cell, one could choose a DNA-binding peptide from a prokaryote, Archaea, single celled eukaryote, plant, or fungus.
  • DNA-binding proteins from operons, and the nucleotide sequences to which they bind are known in the art (see, e.g., Postle et al. (1984) N UCLEIC A CIDS R ES . 12: 4849-63; Buvinger and Riley (1985) J. B ACTERIOL . 163: 850-7; Laughon and Gesteland (1984) M OL . C ELL B IOL . 4:260-7; Bram et al.
  • DNA-binding proteins from operons are given in the table below.
  • the template-binding domain may comprise one or more of these proteins or polypeptides derived therefrom.
  • the DNA-binding polypeptide is, or is derived from, a transcription factor.
  • the DNA-binding polypeptide may be or be derived from, e.g., a repressor or an activator in its wild-type context.
  • the DNA-binding polypeptide will not activate or repress transcription in the methods described herein. This can be achieved by, e.g., mutating transcriptional regulation domains, such as the trans-activating domain (TAD) or any other domain that binds a transcription co-regulator.
  • TAD trans-activating domain
  • This can also be achieved by choosing a DNA-binding polypeptide that does not engage the transcriptional machinery of the cell to be altered. For example, when altering the genome of a human cell, one could choose a DNA-binding peptide from a prokaryote, Archaea, single celled eukaryote, plant, or fungus.
  • the transcription factor in some embodiments, falls into one or more of several categories as set out here.
  • the transcription factor may be a specific transcription factor and/or an upstream transcription factor. It may be constitutively active or conditionally active. If conditionally active, it may be developmental or signal-dependent.
  • the transcription factor is a resident nuclear factor and/or comprises a nuclear localization signal (NLS).
  • the template-binding domain may comprise one or more of these transcription factors or polypeptides derived therefrom.
  • the DNA-binding polypeptide is derived from an endonuclease.
  • the DNA-binding domain may be a catalytically inactive endonuclease, e.g., may have a substitution in or deletion of the domain that catalyzes DNA cleavage. If the endonuclease has other activities such as DNA modification activity, one may introduce mutations into the other active domains as well.
  • the restriction endonuclease may be, e.g., of Type I; Type II, e.g., Type IIR, Type IIS, or Type IIG; Type III; or Type IV.
  • the endonuclease may be used in combination with other DNA-binding polypeptides, e.g., other endonuclease-derived polypeptides, to achieve higher affinity binding to a longer recognition site.
  • the endonuclease recognizes modified DNA, e.g., methylated DNA, and the template binding domain partner comprises modified DNA.
  • the template-binding domain may comprise one or more of these endonucleases or polypeptides derived therefrom.
  • the DNA-binding polypeptide is, or is derived from, a TAL (transcription activator-like) effector.
  • TAL effectors bind specifically to DNA through a series of 34-amino acid repeats, and engineering of these repeats tailors the specificity of the TAL effector to bind a desired DNA sequence. Details on how to engineer specificity are given in, e.g., U.S. Pat. No. 8,440,431. Briefly, each repeat in the TAL effector has a direct, linear correspondence with one nucleotide in the target site.
  • TAL effector by selecting a first residue at position 12 and a second residue at position 13, in order to have that repeat bind to A, C, G, or T.
  • Different repeats can be assembled to create a binding domain that is customized to recognize the desired target sequence.
  • Table V.4 lists different combinations of amino acid residues that can be used to create repeats with specificity for a given nucleotide in the target binding sequence.
  • the DNA-binding polypeptide is derived from a TALEN (TAL effector nuclease), and is mutated to lack nuclease activity. For example, there may be a substitution in or deletion of the domain that catalyzes DNA cleavage.
  • TALEN TAL effector nuclease
  • the TAL effector is from, or is derived from, a TAL effector in a Xanthomonas bacterium, Ralstonia solanacearum , or Burkholderia rhizoxinica.
  • the template-binding domain may comprise one or more of these TAL effectors and TALENs or polypeptides derived therefrom.
  • the template binding domain is a protein
  • the template binding domain partner is a protein
  • the template binding domain and the template binding domain partner have affinity for each other.
  • the template binding domain is a protein
  • the template-binding domain is derived from a protein in another species than the species of the cell to be altered.
  • the template-binding domain is derived from a protein that has no binding partners that are expressed in the cell type to be altered.
  • the protein-binding polypeptide comprises one of more of the following domains: SH2, SH3, PTB, 14-3-3, FHA, WW, WD40, bromo, chromo, EVH1, PDZ, DD, DED, CARD, BH1-4, CSD, F-box, Hect, RING, ANK, ARM, LIM, EF-hand, MH2.
  • the template-binding domain comprises an antibody with affinity for the template binding domain partner.
  • the template-binding domain is a protein and the template binding domain partner is an antibody with affinity for the template binding domain.
  • the antibody may be, e.g., an scFv or any antibody having sufficient CDR sequences to bind its target.
  • the template-binding domain carries one or more deletions relative to the wild-type protein from which it was derived. For example, there may be a deletion of a catalytic domain.
  • the wild-type protein has multiple protein-binding domains, one or more of these domains, e.g., all but one of these domains, is deleted.
  • the template-binding domain may comprise one or more of these protein-binding domains or polypeptides derived therefrom. It is understood that in some embodiments, the template binding domain is, or is derived from, the protein in the left column and the template binding domain partner is, or is derived from, the protein in the right column. In other embodiments, the template binding domain is, or is derived from, the protein in the right column and the template binding domain partner is, or is derived from, the protein in the left column.
  • the template binding domain is a protein
  • the template binding domain partner is a small molecule.
  • the small molecule is rare or absent in the cell being altered. This helps to avoid nonspecific binding.
  • the template-binding domain carries one or more deletions or substitutions relative to the wild-type protein from which it was derived. For example, there may be a deletion of or substitution within a catalytic domain, a DNA-binding domain, a protein-protein interaction domain, and/or a domain necessary for transcriptional regulation.
  • the template-binding domain may comprise one or more of these small molecule-binding domains or polypeptides derived therefrom.
  • Proteins that bind small molecules Protein Small molecule Avidin or Streptavidin (BBa_K283010 from biotin the Registry of Standard Biological Parts) gyrEC (BBa_K133070 from the Registry of coumermycin Standard Biological Parts) RI7 (BBa_K211001 from the Registry of octanal, heptanal or hexanal Standard Biological Parts) VirA receptor (BBa_K389001 from the acetosyringone Registry of Standard Biological Parts) Penicillin-binding proteins (PBPs), e.g., penicillin or cephalosporin serine type D-alanyl-D-alanine carboxypeptidase/transpeptidase TetR tetracycline ASGPR N-Acetylgalactosamine or galactose
  • PBPs Penicillin-binding proteins
  • the template binding domain partner is double-stranded DNA.
  • the template binding domain partner is double-stranded DNA that is recognized by a DNA-binding protein described above in Section V.1.
  • the template binding domain partner may be, e.g., identical to or derived from a DNA sequence that is bound by a protein in a wild-type context.
  • the template binding domain partner comprises all or part of a transcription factor binding site from an organism other than the organism of the cell being altered.
  • the template binding domain partner comprises all or part of a transcriptional regulation site from an operon, e.g., a bacterial operon.
  • the template binding domain partner is at least 10 nucleotides long, e.g., at least 15, 20, 25, 30, 35, 40, 45, 50, 75, 100, 150, or 200 nucleotides long. In some embodiments, the template binding domain partner is at most 200 nucleotides long, e.g., at most 15, 20, 25, 30, 35, 40, 45, 50, 75, 100, 150, or 200 nucleotides long. In some embodiments, the template binding domain partner is 10-20, 20-30, 30-40, 40-50, 50-75, 75-100, 100-150, or 150-200 nucleotides long.
  • the template binding domain partner comprises palindromic sequences.
  • the template binding domain partner comprises a plurality of shorter sequences, wherein each shorter sequence is bound by a distinct DNA-binding domain.
  • the plurality of shorter sequences are identical, e.g., the template binding domain partner comprises repeats.
  • one or more of, e.g., all of the plurality of shorter sequences are not identical to each other.
  • the template binding domain partner is chemically modified DNA, e.g., as set out in Section XI below.
  • the modification may be, e.g., to one or more bases and/or to the backbone.
  • the chemical modification may do one or more of the following: improve the stability of the DNA, reduce the innate immune response against the DNA, and improve the binding of the template binding domain to the template binding domain partner.
  • the template binding domain partner need not always be the same type of molecule as the template nucleic acid.
  • the template binding domain partner is double stranded, while the template nucleic acid is single stranded.
  • a long single-stranded DNA comprises a hairpin at one end, and the double stranded region of the hairpin comprises the template binding domain partner.
  • the template binding domain partner and the template nucleic acid are both double stranded.
  • the template binding domain partner is derived from a wild-type template binding domain partner.
  • the template binding domain partner may be a fragment of a naturally occurring nucleic acid, a mutagenized nucleic acid, a synthetic nucleic acid modeled after a naturally-ocurring nucleic acid.
  • the template binding domain partner is mutagenized to increase its affinity for a template binding domain.
  • the template binding domain partner is mutagenized to decrease its affinity for a template binding domain.
  • the template binding domain partner is single-stranded DNA.
  • the template binding domain partner is a polypeptide. Any of the proteins and/or polypeptides discussed above in Sections V, may be used in the present invention, as a template binding domain partner. For example, protein-protein interacting pairs are discussed above, e.g., in Table V.6.
  • Candidate Cas9 molecules, candidate Cas9 fusion molecules, candidate gRNA molecules, and candidate Cas9 fusion molecule/gRNA molecule complexes can be evaluated by art-known methods or as described herein. For example, exemplary methods for evaluating the endonuclease activity of Cas9 fusion molecule are described, e.g., in Jinek et al. (2012) S CIENCE 337(6096): 816-821.
  • the methods in this section may be used, e.g., to test various portions of a gRNA, for example, the targeting domain, the first complementarity domain, the linking domain, the second complementarity domain, the proximal domain, or the tail domain. In some embodiments, the methods in this section are tested to determine whether modifications made in one or more of these domains interfere with targeting efficacy.
  • a gRNA with a selected length, sequence, degree of complementarity, or degree of modification, can be evaluated in a system of this section.
  • a Cas9 fusion molecule/gRNA molecule complex to bind to and cleave a target nucleic acid can be evaluated in a plasmid cleavage assay.
  • synthetic or in vitro-transcribed gRNA molecule is pre-annealed prior to the reaction by heating to 95° C. and slowly cooling down to room temperature.
  • Native or restriction digest-linearized plasmid DNA 300 ng ( ⁇ 8 nM) is incubated for 60 min at 37° C.
  • Cas9 protein molecule 50-500 nM
  • gRNA 50-500 nM, 1:1
  • Cas9 plasmid cleavage buffer 20 mM HEPES pH 7.5, 150 mM KCl, 0.5 mM DTT, 0.1 mM EDTA
  • the reactions are stopped with 5 ⁇ DNA loading buffer (30% glycerol, 1.2% SDS, 250 mM EDTA), resolved by a 0.8 or 1% agarose gel electrophoresis and visualized by ethidium bromide staining.
  • the resulting cleavage products indicate whether the Cas9 molecule cleaves both DNA strands, or only one of the two strands.
  • linear DNA products indicate the cleavage of both DNA strands.
  • nicked open circular products indicate that only one of the two strands is cleaved.
  • DNA oligonucleotides (10 pmol) are radiolabeled by incubating with 5 units T4 polynucleotide kinase and ⁇ 3-6 pmol ( ⁇ 20-40 mCi) [ ⁇ -32P]-ATP in 1 ⁇ T4 polynucleotide kinase reaction buffer at 37° C. for 30 min., in a 50 ⁇ L reaction. After heat inactivation (65° C.

Abstract

This application provides improved methods of genome editing. Cas9 molecules can be used to create a break in a genomic region of interest. To increase the likelihood that the break is repaired by HDR (homology-directed repair), the cell can be contacted with molecules that bring a template nucleic acid in close proximity to the break, under conditions that allow the cell to repair the break using the template nucleic acid.

Description

    RELATED APPLICATIONS
  • This application claims priority to U.S. Provisional Patent Application No. 62/062,815, filed on Oct. 10, 2014, and to U.S. Provisional Patent Application No. 62/068,371, filed on Oct. 24, 2014, the entire contents of each of which are expressly incorporated herein by reference.
  • BACKGROUND
  • The CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats)/Cas (CRISPR-associated) system evolved in bacteria and archaea as an adaptive immune system to defend against viral attack. Upon exposure to a virus, short segments of viral DNA are integrated into the CRISPR locus. RNA is transcribed from a portion of the CRISPR locus that includes the viral sequence. That RNA, which contains sequence complimentary to the viral genome, mediates targeting of a Cas9 protein to the sequence in the viral genome. The Cas9 protein cleaves and thereby silences the viral target.
  • Recently, the CRISPR/Cas system has been adapted for genome editing in eukaryotic cells. The introduction of site-specific double strand breaks (DSBs) enables target nucleic acid alteration through homology-directed repair (HDR).
  • SUMMARY
  • This disclosure provides systems and methods for editing a genome, e.g., by correcting a mutation. Using the methods and compositions disclosed herein, a Cas9 fusion molecule or Cas9 system may be used to mediate a break or nick near a target position, e.g., a chromosome position, that one desires to edit. The cell then utilizes one of several repair pathways, e.g., HDR, to repair the break or nick. Provided herein is a Cas9 repair system comprising a Cas9 fusion molecule, a nucleic acid template system, and optionally, at least one gRNA molecule. While not being bound by theory, it is believed that by contacting a cell, or population of cells, with the Cas9 repair systems disclosed herein, the proximity of the template nucleic acid used by the cell to repair a Cas9-mediated cleavage event can be increased, and the frequency of use of a particular DNA repair pathway, e.g., HDR, can be modulated.
  • In one aspect, described herein is a Cas9 fusion molecule comprising a Cas9 molecule linked to a template binding domain.
  • In one embodiment, the template binding domain comprises a specific affinity for a template binding domain partner. In one embodiment, the template binding domain does not comprise substantial affinity for a nucleic acid from one or more of a human, a non-human animal, a mammal, a eukaryote, a plant, or a pathogen.
  • In one embodiment, the template binding domain is not a nuclease, a transcription factor, an antibody or other complementarity determining region (CDR)-based molecule, a protein that alters chromatin, a protein that binds chromatin, a protein that modifies DNA, a DNA methylase, a protein that cleaves DNA, a protein that unwinds DNA, or any combination thereof. In one embodiment, the nuclease is an endonuclease or an exonuclease. In one embodiment, the chromatin comprises a histone.
  • In one embodiment, the template binding domain does not comprise substantial affinity for a nucleic acid from a preselected species. In one embodiment, the preselected species is a human, a non-human animal, a mammal, a eukaryote, a plant, a pathogen, or any combination thereof. In one embodiment, the preselected species is human.
  • In one embodiment, the Cas9 molecule is covalently linked to the template binding domain. In another embodiment, the Cas9 molecule is non-covalently linked to the template binding domain.
  • In one embodiment, the template binding domain is linked to the N-terminus of the Cas9 molecule. In one embodiment, the template binding domain is covalently linked to the N-terminus of the Cas9 molecule. In another embodiment, the template binding domain is non-covalently linked to the N-terminus of the Cas9 molecule.
  • In one embodiment, the template binding domain is linked to the C-terminus of the Cas9 molecule. In one embodiment, the template binding domain is covalently linked to the C-terminus of the Cas9 molecule. In another embodiment, the template binding domain is non-covalently linked to the C-terminus of the Cas9 molecule.
  • In one embodiment, the Cas9 fusion molecule comprises at least two template binding domains.
  • In one embodiment, the template binding domain comprises a protein, a nucleic acid, or a small molecule. In one embodiment, the nucleic acid is a DNA or an RNA. In one embodiment, the template binding domain comprises a protein, wherein the protein comprises a DNA binding domain. In one embodiment, the protein comprises a repressor protein, or a fragment of a repressor protein. In one embodiment, the repressor protein, or the fragment of the repressor protein, is a TetR repressor, or a fragment of the TetR repressor; a LacI repressor, or a fragment of the LacI repressor; a Gal4 repressor, or a fragment of the Gal4 repressor; or a repressor protein C1, or a fragment of the repressor protein C1.
  • In one embodiment, the Cas9 fusion protein comprises a linker between the Cas9 molecule and the template binding domain. In one embodiment, the linker is sufficiently long to allow the Cas9 molecule to bind to a target nucleic acid and the template binding domain to bind to a template binding domain partner without steric interference. In one embodiment, the linker is at least 6, but no longer than 60 amino acids in length. In another embodiment, the linker is at least 18, but no longer than 180 nucleotides in length. In one embodiment, the linker is at least 10, 50, 100, 200, 500, 1000, 2000, 5000, or 10000 Angstroms in length. In one embodiment, the linker is no more than 10, 50, 100, 200, 500, 1000, 2000, 5000, or 10000 Angstroms in length. In one embodiment, the linker comprises a polypeptide. In one embodiment, the linker comprises serine, glycine, or glycine and serine. In one embodiment, the linker comprises a sequence of Table 2 or Table 3.
  • In yet another embodiment, the Cas9 molecule is a Cas9 molecule selected from Table 100. In one embodiment, the Cas9 molecule is a Cas9 molecule selected from Table 600. In one embodiment, the Cas9 molecule is not a wild-type Cas9 molecule.
  • In one embodiment, the Cas9 molecule is an eaCas9 molecule. In one embodiment, the eaCas9 molecule forms a double stranded break in a target nucleic acid. In one embodiment, the eaCas9 molecule comprises N-terminal RuvC-like domain cleavage activity and HNH-like domain cleavage activity. In another embodiment, the eaCas9 molecule forms a single stranded break in a target nucleic acid. In one embodiment, the eaCas9 molecule comprises HNH-like domain cleavage activity but has no, or no significant, N-terminal RuvC-like domain cleavage activity. In one embodiment, the eaCas9 molecule is an HNH-like domain nickase. In one embodiment, the eaCas9 molecule comprises a mutation at an amino acid position corresponding to amino acid position D10 of Streptococcus pyogenes Cas9. In yet another embodiment, the eaCas9 molecule comprises N-terminal RuvC-like domain cleavage activity but has no, or no significant, HNH-like domain cleavage activity. In one embodiment, the eaCas9 molecule is an N-terminal RuvC-like domain nickase. In one embodiment, the eaCas9 molecule comprises a mutation at an amino acid position corresponding to amino acid position H840 or N863 of S. pyogenes Cas9.
  • In another embodiment, the Cas9 molecule is an eiCas9 molecule. In some embodiments, the Cas9 molecule comprises a mutation, e.g., a point mutation, that causes the Cas9 molecule to be inactive, e.g., a mutation that eliminates the Cas9 molecule cleavage activity.
  • In one embodiment, the Cas9 molecule comprises a REC2 deletion, REC1CT deletion, or a REC1SUB deletion, or any combination thereof.
  • In one embodiment, the Cas9 molecule comprises an altered PI domain.
  • In another embodiment, the Cas9 molecule is less than about 1300 amino acids in length. In one embodiment, the Cas9 molecule is less than about 1200, 1100, 1000, 900, or 800 amino acids in length. In one embodiment, the Cas9 molecule is between about 800 to 1300, 900 to 1200, 900 to 1100, or 900 to 1000 amino acids in length.
  • In one embodiment, the Cas9 fusion molecule is substantially purified.
  • In another aspect, described herein is a Cas9 system comprising a Cas9 fusion molecule described herein, and a nucleic acid template system comprising a template binding domain partner and a template nucleic acid.
  • In one embodiment, the template binding domain of the Cas9 fusion molecule is bound to the template binding domain partner. In one embodiment, the template binding domain of the Cas9 fusion molecule is covalently bound to the template binding domain partner. In another embodiment, the template binding domain of the Cas9 fusion molecule is non-covalently bound to the template binding domain partner.
  • In one embodiment, the template binding domain partner is linked to the template nucleic acid. In one embodiment, the template binding domain partner is covalently linked to the template nucleic acid. In another embodiment, the template binding domain partner is non-covalently linked to the template nucleic acid.
  • In one embodiment, the template binding domain partner comprises a protein, a nucleic acid, or a small molecule. In one embodiment, the nucleic acid is a DNA or an RNA. In one embodiment, the template binding domain comprises a protein and the template binding domain partner comprises a protein.
  • In one embodiment, the template binding domain comprises a protein and the template binding domain partner comprises a nucleic acid. In one embodiment, the nucleic acid is a DNA. In one embodiment, the DNA is a double-stranded DNA or a single-stranded DNA. In one embodiment, the nucleic acid is an RNA.
  • In one embodiment, the template binding domain comprises a protein and the template binding domain partner comprises a small molecule.
  • In another embodiment, the template binding domain comprises a small molecule, and the template binding domain partner comprises a protein.
  • In one embodiment, the template binding domain comprises a small molecule, and the template binding domain partner comprises a small molecule.
  • In yet another embodiment, the template binding domain comprises a small molecule, and the template binding domain partner comprises a nucleic acid. In one embodiment, the template binding domain comprises a nucleic acid, and the template binding domain partner comprises a protein. In another embodiment, the template binding domain comprises a nucleic acid, and the template binding domain partner comprises a nucleic acid. In one embodiment, the template binding domain comprises a nucleic acid, and the template binding domain partner comprises a small molecule. In one embodiment, the nucleic acid is an RNA. In another embodiment, the nucleic acid is a DNA. In one embodiment, the DNA is a double-stranded DNA or a single-stranded DNA.
  • In one embodiment, the template binding domain partner is a DNA sequence recognized by a DNA binding protein. In one embodiment, the DNA sequence recognized by the DNA binding protein is selected from a Tet-O sequence, a Lac operon O1 sequence, a UAS sequence, or an Operator L and R sequence. In yet another embodiment, the template binding domain partner comprises a repressor-binding DNA sequence from a bacterial operon. In one embodiment, the protein comprises a TetR repressor, or a fragment of the TetR repressor, and the DNA comprises at least one Tet-O sequence. In one embodiment, the DNA comprises at least two, three, four, five, or six Tet-O sequences. In one embodiment, the DNA comprises a spacer sequence between the Tet-O sequences. In one embodiment, the spacer sequence is at least 17 nucleotides in length. In another embodiment, the spacer sequence is at least 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 nucleotides in length. In one embodiment, the DNA does not comprise a spacer sequence between the Tet-O sequences.
  • In one embodiment, the template binding domain partner comprises a protein that is not a nuclease, a transcription factor, an antibody or other CDR-based molecule, a protein that alters chromatin, a protein that binds chromatin, a protein that modifies DNA, a DNA methylase, a protein that cleaves DNA, a protein that unwinds DNA, or any combination thereof. In one embodiment, the nuclease is an endonuclease or an exonuclease. In one embodiment, the chromatin comprises a histone.
  • In one embodiment, the Cas9 system comprises at least one additional template binding domain and at least one additional template binding domain partner.
  • In one embodiment, the template binding domain partner comprises a linear nucleic acid. In another embodiment, the template binding domain partner comprises a nucleic acid sequence located on a circular nucleic acid.
  • In one embodiment, the template binding domain partner comprises a double stranded nucleic acid sequence. In one embodiment, the template binding domain partner comprises a single stranded nucleic acid sequence.
  • In one embodiment, the template nucleic acid comprises a double stranded nucleic acid sequence or a single stranded nucleic acid sequence.
  • In one embodiment, the nucleic acid template system comprises a double stranded nucleic acid sequence or a single stranded nucleic acid sequence.
  • In one embodiment, the template nucleic acid comprises a linear nucleic acid. In another embodiment, the template nucleic acid is a nucleic acid located on a circular nucleic acid.
  • In one embodiment, the template binding domain partner comprises a double stranded nucleic acid; and the template nucleic acid comprises a double stranded nucleic acid.
  • In one embodiment, the template binding domain partner comprises a double stranded nucleic acid; and the template nucleic acid comprises a single stranded nucleic acid.
  • In one embodiment, the nucleic acid template system is a circular nucleic acid. In another embodiment, the nucleic acid template system is a linear nucleic acid.
  • In one embodiment, the template binding domain partner comprises a single stranded nucleic acid; and the template nucleic acid comprises a single stranded nucleic acid.
  • In another embodiment, the template binding domain partner comprises a single stranded nucleic acid; and the template nucleic acid comprises a double stranded nucleic acid.
  • In one embodiment, the nucleic acid template system is a single stranded nucleic acid, and the template binding domain partner is 5′ of the template nucleic acid. In another embodiment, the nucleic acid template system is a single stranded nucleic acid, and the template binding domain partner is 3′ of the template nucleic acid.
  • In one embodiment, the nucleic acid template system is a double stranded nucleic acid, and the template binding domain partner is 5′ of the template nucleic acid. In another embodiment, the nucleic acid template system is a double stranded nucleic acid, and the template binding domain partner is 3′ of the template nucleic acid.
  • In one embodiment, the template nucleic acid comprises about 50-500 nucleotides of homology with a target nucleic acid. In one embodiment, the template nucleic acid comprises about 100-200 nucleotides of homology with a target nucleic acid. In one embodiment, the template nucleic acid comprises about 500-2000 nucleotides of homology with a target nucleic acid.
  • In one embodiment, the template nucleic acid comprises a human nucleic acid sequence. In one embodiment, the template nucleic acid comprises a wild-type human nucleic acid sequence.
  • In yet another embodiment, the Cas9 system further comprises at least one additional template binding domain partner.
  • In one embodiment, the template nucleic acid lacks repeated elements. In one embodiment, the repeated element is an Alu element or a LINE element.
  • In one embodiment, the Cas9 system further comprises a gRNA. In one embodiment, the gRNA comprises a targeting domain, first and second complementary domains, and a proximal domain. In one embodiment, the gRNA is a chimeric gRNA.
  • In one embodiment, the Cas9 system further comprises at least one additional Cas9 molecule. In some embodiment, the Cas9 molecule is an eaCas9 molecule. In some embodiments, the Cas9 molecule is an eiCas9 molecule. In one embodiment, Cas9 system comprises a Cas9 fusion molecule, comprising an eiCas9 molecule linked to a template binding domain, and a Cas9 molecule. In one embodiment, Cas9 system comprises a Cas9 fusion molecule, comprising an eiCas9 molecule linked to a template binding domain, and an eaCas9 molecule.
  • In another aspect, described herein is a cell, or a population of cells, comprising a Cas9 system described herein.
  • In yet another aspect, described herein is a cell, or a population of cells, comprising a Cas9 fusion molecule described herein.
  • In another aspect, described herein is a nucleic acid encoding a Cas9 fusion molecule described herein. In yet another aspect, described herein is a vector comprising said nucleic acid. In one embodiment, the vector is an AAV vector. In one aspect, described herein is a cell, or a population of cells, comprising said nucleic acid.
  • In another aspect, described herein is a method of altering a nucleic acid at a target position in a cell, or a population of cells, the method comprising contacting the cell with the Cas9 system described herein, wherein the gRNA molecule and Cas9 fusion molecule interact with the nucleic acid, resulting in a cleavage event, wherein the cleavage event is repaired by at least one DNA repair pathway, and wherein the sequence of the nucleic acid after the cleavage event is different than the sequence of the nucleic acid prior to the cleavage event, thereby altering the nucleic acid at the target position in the cell, or in the population of cells.
  • In one embodiment, the method, further comprises contacting the cell, or the population of cells, with a second gRNA molecule, wherein the second gRNA molecule and the Cas9 fusion molecule interact with the nucleic acid, resulting in a second cleavage event.
  • In one embodiment, the at least one DNA repair pathway is selected from the group consisting of resection, mismatch repair (MMR), nucleotide excision repair (NER), base excision repair (BER), canonical non-homologous end joining (canonical NHEJ), alternative non-homologous end joining (ALT-NHEJ), canonical homology directed-repair (canonical HDR), alternative homology directed repair (ALT-HDR), microhomology-mediated end joining (MMEJ), Blunt End Joining, Synthesis Dependent Microhomology Mediated End Joining, single strand annealing (SSA), Holliday junction model or double strand break repair (DSBR), synthesis-dependent strand annealing (SDSA), single strand break repair (SSBR), translesion synthesis repair (TLS), and interstrand crosslink repair (ICL), and DNA/RNA processing.
  • In one embodiment, the at least one DNA repair pathway is canonical homology directed-repair (canonical HDR) or alternative homology directed repair (ALT-HDR).
  • In one embodiment, the cleavage event comprises one or more single strand breaks, one or more double strand breaks, or a combination of single strand breaks and double strand breaks.
  • In one embodiment, the gRNA molecule positions one cleavage event on each strand of the nucleic acid.
  • In yet another embodiment, the gRNA molecule positions the cleavage event on a strand of the nucleic acid that binds to the gRNA molecule.
  • In one embodiment, the second gRNA molecule positions the second cleavage event on a strand of the nucleic acid that binds to the second gRNA molecule.
  • In one embodiment, the Cas9 fusion molecule makes a single strand break in the nucleic acid. In another embodiment, the Cas9 fusion molecule makes a double strand break in the nucleic acid. In one embodiment, the single strand break is made in the strand of the nucleic acid to which a targeting domain of the gRNA molecule is complementary. In another embodiment, the single strand break is made in the strand of the nucleic acid other than the strand to which a targeting domain of the gRNA is complementary.
  • In one embodiment, the target position is in an HBB gene.
  • In one embodiment, the cell, or the population of cells, is a eukaryotic cell, or a population of eukaryotic cells. In one embodiment, the cell, or the population of cells, is a plant cell, or a population of plant cells. In one embodiment, the plant cell, or the population of plant cells, is a monocot plant cell, a dicot plant cell, a population of monocot plant cells, or a population of dicot plant cells.
  • In another embodiment, the cell, or the population of cells, is a mammalian cell, or a population of mammalian cells. In one embodiment, the cell, or the population of cells, is a human cell, or a population of human cells.
  • In one embodiment, the cell, or the population of cells, is a vertebrate, mammalian, rodent, goat, pig, bird, chicken, turkey, cow, horse, sheep, fish, primate, or human cell or population of cells.
  • In one embodiment, the cell, or the population of cells, is a somatic cell, a germ cell, or a prenatal cell or population of cells.
  • In another embodiment, the cell, or the population of cells, is a zygotic cell, a blastocyst, an embryonic cell, a stem cell, a mitotically competent cell, a meiotically competent cell or population of cells.
  • In one embodiment, the cell, or the population of cells, is a T cell, a CD8+ T cell, a CD8+ naïve T cell, a central memory T cell, an effector memory T cell, a CD4+ T cell, a stem cell memory T cell, a helper T cell, a regulatory T cell, a cytotoxic T cell, a natural killer T cell, a Hematopoietic Stem Cell, a long term hematopoietic stem cell, a short term hematopoietic stem cell, a multipotent progenitor cell, a lineage restricted progenitor cell, a lymphoid progenitor cell, a myeloid progenitor cell, a common myeloid progenitor cell, an erythroid progenitor cell, a megakaryocyte erythroid progenitor cell, a monocytic precursor cell, an endocrine precursor cell, an exocrine cell, a fibroblast, a retinal cell, a photoreceptor cell, a rod cell, a cone cell, a retinal pigmented epithelium cell, a trabecular meshwork cell, a cochlear hair cell, an outer hair cell, an inner hair cell, a pulmonary epithelial cell, a bronchial epithelial cell, an alveolar epithelial cell, a pulmonary epithelial progenitor cell, a striated muscle cell, a cardiac muscle cell, a muscle satellite cell, a myocyte, a neuron, a neuronal stem cell, a mesenchymal stem cell, an induced pluripotent stem (iPS) cell, an embryonic stem cell, a monocyte, a megakaryocyte, a neutrophil, an eosinophil, a basophil, a mast cell, a reticulocyte, a B cell, e.g. a progenitor B cell, a Pre B cell, a Pro B cell, a memory B cell, a plasma B cell, a gastrointestinal epithelial cell, a biliary epithelial cell, a pancreatic ductal epithelial cell, an intestinal stem cell, a hepatocyte, a liver stellate cell, a Kupffer cell, an osteoblast, an osteoclast, an adipocyte, a preadipocyte, a pancreatic precursor cell, a pancreatic islet cell, a pancreatic beta cell, a pancreatic alpha cell, a pancreatic delta cell, a pancreatic exocrine cell, a Schwann cell, or an oligodendrocyte, or population of such cells.
  • In one embodiment, the cell, or population of cells, is from a subject suffering from a disease or disorder.
  • In one embodiment, the disease is a blood disease, an immune disease, a neurological disease, a cancer, an infectious disease, a genetic disease, a disorder caused by aberrant mtDNA, a metabolic disease, a disorder caused by aberrant cell cycle, a disorder caused by aberrant angiogenesis, a disorder cause by aberrant DNA damage repair, or a pain disorder.
  • In one embodiment, the cell, or population of cells, is from a subject having at least one mutation at the target position.
  • In one embodiment, the method further comprises isolating the cell, or population of cells, from the subject prior to contacting the cell, or population of cells, with the Cas9 system described herein.
  • In one embodiment, the method further comprises introducing the cell, or the population of cells, into a subject after contacting the cell, or the population of cells, with the Cas9 system described herein.
  • In one embodiment, the contacting the cell, or the population of cells, with the Cas9 system described herein is performed ex vivo. In another embodiment, the contacting the cell, or the population of cells, with the Cas9 system described herein is performed in vivo. In one embodiment, the contacting the cell, or the population of cells, with the Cas9 system described herein is performed in vitro.
  • In one embodiment, the method further comprises sequencing the nucleic acid, or a portion of the nucleic acid, prior to contacting the cell, or the population of cells, with the Cas9 system described herein.
  • In one embodiment, the method further comprises sequencing the nucleic acid, or a portion of the nucleic acid, after the cleavage event.
  • In one embodiment, the cell, or the population of cells, is contacted with the gRNA molecule and the Cas9 fusion molecule as a pre-formed complex.
  • In one aspect, described herein is a cell, or a population of cells, altered by the methods described herein. In another aspect, described herein is a pharmaceutical composition comprising said cell, or said population of cells.
  • In yet another aspect, described herein is a pharmaceutical composition comprising a Cas9 system described herein.
  • In one aspect, described herein is a method of treating a subject comprising administering to the subject the cell, or the population of cells, altered by the methods described herein, or a pharmaceutical composition comprising said cell, or said population of cells.
  • In another aspect, described herein is a method of treating a subject suffering from a disease or disorder, the method comprising contacting a cell, or a population of cells, from the subject with the Cas9 system described herein, wherein the gRNA molecule and the Cas9 fusion molecule interact with a nucleic acid at a target position, resulting in a cleavage event, wherein the cleavage event is repaired by at least one DNA repair pathway, and wherein the sequence of the nucleic acid after the cleavage event is different than the sequence of the nucleic acid prior to the cleavage event, thereby treating the subject suffering from the disease or disorder.
  • In one embodiment, the method further comprises contacting the cell from the subject with a second gRNA molecule, wherein the second gRNA molecule and the Cas9 fusion molecule interact with the nucleic acid, resulting in a second cleavage event.
  • In one embodiment, the contacting the cell is performed ex vivo. In another embodiment, the contacting the cell is performed in vivo.
  • In certain aspects, the present disclosure provides a cell comprising:
      • a Cas9 molecule and a template binding domain, wherein optionally the Cas9 molecule is linked to the template binding domain;
  • wherein said template binding domain comprises specific affinity for a template binding domain partner and lacks substantial affinity for:
  • (i) the endogenous nucleic acid of said cell;
  • (ii) the endogenous chromosomal nucleic acid of said cell; or
  • (iii) the endogenous organellar nucleic acid, e.g., mitochondrial, chloroplast, or both, of said cell.
  • In certain aspects, the present disclosure also provides a cell comprising:
      • (a) a Cas9 molecule and a template binding domain; wherein optionally the Cas9 molecule is linked to the template binding domain; and
      • (b) a gRNA molecule having a targeting domain complementary with a target sequence;
  • wherein said template binding domain comprises specific affinity for a template binding domain partner and lacks substantial affinity for a nucleic acid sequence within 100, 500, 1000, 2000, 5,000, or 10,000 nucleotides of said target sequence.
  • In certain aspects, the present disclosure also provides a kit comprising packaging and at least three of:
      • (a) a Cas9 fusion molecule,
      • (b) a template binding domain,
      • (c) a template binding domain partner, and
      • (d) a template nucleic acid.
  • In the kit, in some embodiments, (a) and (b) form part of a single composition; (a) and (c) form part of a single composition; (a) and (d) form part of a single composition; (b) and (c) form part of a single composition; (b) and (d) form part of a single composition; (c) and (d) form part of a single composition; (a), (b), and (c) form part of a single composition; (a), (b), and (d) form part of a single composition; (a), (c), and (d) form part of a single composition; (b), (c), and (d) form part of a single composition; or (a), (b), (c), and (d) form part of a single composition.
  • In certain aspects, the present disclosure also provides a reaction mixture comprising the cell and a solution.
  • In certain aspects, the present disclosure also provides a reaction mixture wherein the solution is a cell growth medium.
  • In certain aspects, the present disclosure provides a method of altering the structure of a cell, e.g., altering the sequence, of a target nucleic acid of the cell, comprising contacting the cell with the Cas9 system as described herein under conditions that allow for alteration of the structure of the cell, thereby altering the structure of a cell, e.g., altering the sequence of a target nucleic acid.
  • In some embodiments the method, comprises bringing a template nucleic acid in proximity with a target nucleic acid in a cell, e.g., sufficient proximity that the efficiency of altering the cell, e.g., a target nucleic acid of the cell, is increased over the level seen with a non bound template nucleic acid.
  • In some embodiments, the target nucleic acid is cleaved at a position that is:
  • i) at a target position;
  • ii) away from a target position;
  • iii) within 200 nucleotides of the target position;
  • iv) at least 10 nucleotides away from the target position; or
  • v) 10 to 200 nucleotides away from the target position.
  • In some embodiments, the target nucleic acid is cleaved at two positions that are:
  • within 55 nucleotides of one another;
  • at least 25 nucleotides apart; or
  • within 25-55 nucleotides of one another.
  • Other features and advantages of the invention will be apparent from the detailed description, drawings, and from the claims.
  • DESCRIPTION OF THE DRAWINGS
  • FIG. 1 illustrates embodiments where a Cas9 molecule is engineered to selectively associate with a template nucleic acid, and therefore increase the proximity of the template nucleic acid to a target nucleic acid. The Cas9 molecule (which can be, e.g., wild-type or a nickase) is shown associated with a target nucleic acid, e.g., a mutant gene. The Cas9 molecule is associated with a gRNA that targets the Cas9 molecule to the target nucleic acid. The target nucleic acid comprises a target sequence which is recognized by a gRNA, as well as a PAM sequence that is recognized by the Cas9 molecule. The Cas9 molecule is shown linked to a template binding domain (illustrated as a trapezoid) using a linker region (illustrated as a line connecting the Cas9 molecule and the template binding domain). The template binding domain (which can be a DNA binding domain) has affinity for a template binding domain partner (which can be a DNA sequence specifically recognized by the DNA binding domain) which is shown as a shaded rectangle. In this figure, the template binding domain partner is part of a circular double stranded nucleic acid molecule. The circular nucleic acid molecule also comprises a template nucleic acid which is capable of participating in homology-dependent repair of a break in the target nucleic acid. Together, the template nucleic acid and the template binding domain partner form a nucleic acid template system. The figure also shows alternative nucleic acid template systems, one which is a double stranded linear nucleic acid, and one which is a single stranded linear nucleic acid.
  • FIG. 2 depicts exemplary nucleic acid template systems. The template binding domain partner (e.g., a stretch of DNA that a DNA-binding domain can bind) is shown as a shaded box. The template nucleic acid is illustrated as a single line which represents single-stranded DNA, or a double line which represents double stranded DNA. The figure illustrates that the template nucleic acid system can be a circular double stranded DNA molecule like a plasmid; a linear double stranded DNA molecule, or a linear DNA molecule that is single stranded in the template nucleic acid region and double stranded in the template binding domain partner region. When the DNA is linear, the template binding domain partner can be, e.g., on either end of the molecule.
  • FIG. 3 depicts exemplary arrangements of linear nucleic acid template systems. The target nucleic acid (a long double stranded DNA segment like a gene or chromosome) is depicted as a double stranded shaded line with a break. The regions of the target nucleic acid that are homologous to a template nucleic acid are shown as thick solid or dotted lines. The Cas9 molecule is depicted as a shaded circle positioned over the break, and the template binding domain extends from the Cas9 molecule. The nucleic acid template systems are shown below the target nucleic acids, with the template binding domain partner shown as a shaded box and the template nucleic acid as a dotted or solid line. In this figure, the nucleic acid template system can be double stranded, single stranded, or have a single stranded portion and a double stranded portion (e.g., where the template binding domain partner is double stranded and the template nucleic acid is single stranded). The top row shows embodiments where the template nucleic acid has homology on either side of the cut, e.g., approximately 50 to 100 bp or nucleotides both on the left and on the right side of the cut. The homology regions to the left are shown as solid lines and the homology regions to the right are shown as dotted lines. The central row shows embodiments where the template nucleic acid has the majority of the homology on the right side of the cut, e.g., approximately 150-200 bp or nucleotides of homology. The bottom row shows embodiments where the template nucleic acid has the majority of the homology on the left side of the cut, e.g., approximately 150-200 bp or nucleotides of homology. In all the rows, the template binding domain partner can be placed at either end of the nucleic acid template system; in the left column the template binding domain partner is placed at the right of the nucleic acid template system, and in the right column the template binding domain partner is placed at the left of the nucleic acid template system.
  • FIG. 4 depicts exemplary arrangements of circular nucleic acid template systems. The target nucleic acid (a long double stranded DNA segment like a gene or chromosome) is depicted as a double stranded shaded line with a break. The regions of the target nucleic acid that are homologous to a template nucleic acid are shown as thick solid or dotted lines. The Cas9 molecule is depicted as a shaded circle positioned over the break, and the template binding domain extends from the Cas9 molecule. The nucleic acid template systems are shown below the target nucleic acids, with the template binding domain partner is shown as a shaded box and the template nucleic acid as a dotted or solid line. In this figure, the circular nucleic acid template systems are double stranded DNA, but it is understood that the disclosure also contemplates circular molecules that are partly or whole single stranded. The top row shows embodiments where the template nucleic acid has a homology region to the right of the template binding domain partner. The center row shows embodiments where the template nucleic acid has a homology region to the left of the template binding domain partner. The bottom row shows embodiments where the template nucleic acid has a homology region on the opposite side of the circular DNA molecule from the template binding domain partner. In all nine arrangements, the homology region on the template nucleic acid can corresponds to a region on the target nucleic acid, where a solid line is homologous with a solid line and a dotted line is homologous with a dotted line. In the left column, the template nucleic acid has homology to the target nucleic acid on either side of the break. In the central column, the template nucleic acid has homology to the target nucleic acid on the left side of the break. In the right column, the template nucleic acid has homology to the target nucleic acid on the right side of the break. The homology region can be, e.g., 500 to 2000 bp.
  • FIGS. 5A and 5B are schematic representations of the domain organization of S. pyogenes Cas9. FIG. 5A shows the organization of the Cas9 domains, including amino acid positions, in reference to the two lobes of Cas9 (recognition (REC) and nuclease (NUC) lobes). FIG. 5B shows the percent homology of each domain across 83 Cas9 orthologs.
  • FIG. 6 is a schematic drawing of a Cas9 fusion molecule, comprising a Cas9 molecule fused to a template binding domain, bound to a target nucleic acid, and complexed with a nucleic acid template system comprising a template binding domain partner and a template nucleic acid.
  • FIG. 7 is a schematic drawing of a Cas9 system comprising an eaCas9 molecule bound to a target nucleic acid, and a Cas9 fusion molecule, comprising an eiCas9 molecule fused to a template binding domain, bound to a target nucleic acid, and complexed with a nucleic acid template system comprising a template binding domain partner and a template nucleic acid. The Cas9 fusion molecule positions the nucleic acid template system in close proximity to the eaCas9 molecule.
  • FIG. 8A illustrates four exemplary ssDNA nucleic acid template systems comprising a template binding domain partner, comprising three Tet Operator sequences, and a template nucleic acid. V1.1 is an exemplary ssDNA nucleic acid template system comprising a template binding domain partner, comprising three Tet Operator sequences separated by two 17 nucleotide spacers, N terminal of a 109 nucleotide template nucleic acid. V1.2 is an exemplary ssDNA nucleic acid template system comprising a template binding domain partner, comprising three Tet Operator sequences separated by two 17 nucleotide spacers, C terminal of a 109 nucleotide template nucleic acid. V2 is an exemplary ssDNA nucleic acid template system comprising a template binding domain partner, comprising three Tet Operator sequences separated by two 8 nucleotide spacers, N terminal of a 109 nucleotide template nucleic acid. V3 is an exemplary ssDNA nucleic acid template system comprising a template binding domain partner, comprising three Tet Operator sequences adjacent to one another without intervening spacers, N terminal of a 109 nucleotide template nucleic acid.
  • FIG. 8B shows Tet Repressor protein binding activity for four exemplary ssDNA nucleic acid template systems comprising a template binding domain partner, comprising three Tet Operator sequences, and a template nucleic acid (i.e., V1.1, V1.2, V2, and V3) and two control DNA templates (i.e., ssODN 179, and ssODN109. An electrophoretic mobility shift assay using a 10% polyacrylamide gel was performed using 500 nM of recombinant Tet Repressor protein 50 nM of ssDNA nucleic acid template system or control DNA template.
  • FIG. 9 illustrates four exemplary ssDNA nucleic acid template systems comprising a template binding domain partner, comprising three or four Tet Operator sequences, and a template nucleic acid. V4.1 is an exemplary ssDNA nucleic acid template system comprising a template binding domain partner, comprising three Tet Operator sequences separated by three 8 nucleotide spacers, N terminal of a 119 nucleotide template nucleic acid. V4.2 is an exemplary ssDNA nucleic acid template system comprising a template binding domain partner, comprising three Tet Operator sequences separated by three 8 nucleotide spacers, C terminal of a 119 nucleotide template nucleic acid. V5.1 is an exemplary ssDNA nucleic acid template system comprising a template binding domain partner, comprising four Tet Operator sequences separated by three 8 nucleotide spacers, N terminal of a 100 nucleotide template nucleic acid. V5.2 is an exemplary ssDNA nucleic acid template system comprising a template binding domain partner, comprising four Tet Operator sequences separated by three 8 nucleotide spacers, C terminal of a 100 nucleotide template nucleic acid.
  • DETAILED DESCRIPTION Definitions
  • “Amino acids” as used herein encompasses the canonical amino acids as well as analogs thereof.
  • “Amino acid residues that flank a deletion”, as that phrase is used herein, refers to the amino acid residue that immediately precedes the deletion and the amino acid residue that immediately follows the deletion. By way of example, in a sequence CT1-CT2-CT3-CT7-CT8-CT9, wherein CT4-CT5-CT6 is deleted, the flanking amino acid residues are, CT3 and CT7.
  • “Cas9 core domain”, as that term is used herein, refers to a polypeptide that does not include a functional PI domain, e.g., a polypeptide not having an endogenous PI domain, e.g., wherein the endogenous PI domain is deleted (deleted, as used in this context, refers merely to a sequence difference or the absence of amino acid residues and implies no process or origin limitation), or generally, a Cas9 molecule lacking a PI domain. In an embodiment, a Cas9 core domain comprises a REC1 domain, a REC2 domain, a BH domain, a RuvC domain, and an HNH domain. A Cas9 core domain, together with an altered PI domain, comprises a functional Cas9 molecule.
  • “Cas9 fusion molecule”, “Cas9 fusion protein”, or “Cas9 fusion”, as used herein, refers to a chimeric protein comprising a Cas9 molecule, e.g., Cas9 protein or Cas9 polypeptide, or a fragment thereof, linked to a template binding domain. In some embodiments, the template binding domain is a protein or polypeptide. In some embodiments, the template binding domain is a nucleic acid, e.g., DNA or RNA. In some embodiments, the template binding domain is a small molecule. In some embodiments, the Cas9 fusion molecule comprises a Cas9 molecule covalently linked to the template binding domain. For example, the Cas9 fusion protein may be a chimeric protein comprising one or more Cas9 protein domains and a DNA binding domain from a protein disclosed herein. In some embodiments, the Cas9 fusion molecule comprises a Cas9 molecule noncovalently linked to the template binding domain.
  • In an embodiment, a species X Cas9 core domain has at least 20, 30, 40, 50, 60, 70, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% homology with the corresponding sequence of a reference sequence, e.g., a naturally occurring species X Cas9 core domain, e.g., from a Cas9 core domain from Table 100. In an embodiment, each of a REC1 domain, a REC2 domain, a BH domain, a RuvC domain, and/or an HNH domain of a species X Cas9 core domain has, independently, at least 20, 30, 40, 50, 60, 70, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% homology with the corresponding sequence of a reference sequence, e.g., a naturally occurring species X Cas9 core domain, e.g., from a Cas9 core domain from Table 100.
  • “Cas9 molecule” or “Cas9 polypeptide”, as that term is used herein, refers to a polypeptide that can bind (1) a PAM (a protospacer adjacent motif) in a nucleic acid, and (2) a guide RNA (gRNA) molecule. In an embodiment, in concert with the gRNA molecule, a Cas9 molecule or Cas9 polypeptide can localize to a site which comprises a target domain.
  • Cas9 may be a nuclease (an enzyme that cleaves both strands of a double-stranded nucleic acid), a nickase (an enzyme that cleaves one strand of a double-stranded nucleic acid), or an enzymatically inactive (or dead) molecule. A Cas9 molecule having nuclease or nickase activity is referred to as an enzymatically active Cas9 molecule (an eaCas9 molecule). A Cas9 molecule lacking the ability to cleave target nucleic acid is referred to as an enzymatically inactive Cas9 molecule (an eiCas9 molecule). A Cas9 molecule can have the amino acid sequence of a naturally occurring Cas9 molecule or can be an altered, engineered or modified Cas9 molecule, which differs by at least one amino acid residue, from a reference sequence, e.g., the most similar naturally occurring Cas9 molecule, e.g., a Cas9 molecule from Table 100. (The terms altered, engineered or modified, as used in this context, refers merely to a difference from a reference or naturally occurring sequence, and impose no specific process or origin limitations.) For example, an altered, engineered or modified Cas9 molecule can comprise one or more point mutations which alter (e.g., increase, decrease and/or eliminate), one or more Cas9 molecule activities, e.g., a nuclease activity.
  • In an embodiment, a Cas9 molecule meets one or both of the following criteria:
  • it has at least 20, 30, 40, 50, 55, 60, 65, 70, 75, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% homology with, or
  • it differs by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 35, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 150, 200, 250, 300, 350 or 400, amino acid residues from, the amino acid sequence of a reference sequences, e.g., naturally occurring Cas9 molecule, e.g., a Cas9 molecule described in Table 100 herein.
  • In one embodiment, the Cas9 molecule may be a Cas9 deletion, e.g., the Cas9 may comprise a deletion in one or more of the following domains: a REC2, REC1CT, or REC1SUB domain. Except for any REC deletion, a Cas9 molecule meets one or both of the following criteria:
  • it has at least 20, 30, 40, 50, 55, 60, 65, 70, 75, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% homology with, or
  • it differs by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 35, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 150, 200, 250, 300, 350 or 400, amino acid residues from, the amino acid sequence of a reference sequences, e.g., naturally occurring Cas9 molecule, e.g., a Cas9 molecule described in Table 100 herein. Homology except for any REC deletion is determined as follows: a sequence having a deletion is altered by replacing the deleted sequence with the corresponding sequence from the reference sequence, and the altered sequence is compared with the reference sequence.
  • In another embodiment, the Cas9 molecule may be a Cas9 variant, e.g., the Cas9 may comprise an altered PI domain, or other modified amino acid sequence, or the Cas9 may comprise a linker. In an alternate embodiment, except for an altered PI domain or other modified amino acid sequence, a Cas9 molecule meets one or both of the following criteria:
  • it has at least 20, 30, 40, 50, 55, 60, 65, 70, 75, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% homology with, or
  • it differs by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 35, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 150, 200, 250, 300, 350 or 400, amino acid residues from, the amino acid sequence of a reference sequences, e.g., naturally occurring Cas9 molecule, e.g., a Cas9 molecule described in Table 100 herein. Homology except for an altered PI domain, or other modified amino acid sequence is determined as follows: a sequence having an altered PI domain (or other modified amino acid sequence) is altered by restoring the altered PI domain (or other modified amino acid sequence) to the naturally occurring PI domain (or other naturally occurring sequence) from the reference sequence, and the thus altered sequence is compared with the reference sequence.
  • In an alternate embodiment, except for a linker, a Cas9 molecule meets one or both of the following criteria:
  • it has at least 20, 30, 40, 50, 55, 60, 65, 70, 75, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% homology with, or
  • it differs by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 35, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 150, 200, 250, 300, 350 or 400, amino acid residues from, the amino acid sequence of a reference sequences, e.g., naturally occurring Cas9 molecule, e.g., a Cas9 molecule described in Table 100 herein. Homology except for a linker is determined as follows: a sequence having a linker is altered by omitting the linker sequence, and the thus altered sequence is compared with the reference sequence.
  • In another embodiment, each domain of the Cas9 molecule (e.g., the domains named in FIG. 5 or elsewhere herein), including any remaining portion of a REC2, REC1CT, or REC1SUB domain having a deletion or an unaltered portion of a PI domain, will, independently have:
  • at least 20, 30, 40, 50, 55, 60, 65, 70, 75, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% homology with such a domain described herein, e.g., in a species of Table 100. In an embodiment at least 1, 2, 3, 4, 5, of 6 domains will have, independently, at least 50, 60, 70, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% homology with a corresponding domain, while any remaining domains will be absent, or have less homology to their corresponding naturally occurring domains.
  • “PI domain”, as that term is used herein, refers to the region of a Cas9 molecule that interacts with the PAM sequence of a target nucleic acid.
  • “Altered PI domain”, as that term is used herein, refers to a PI domain other than the native or endogenous PI domain associated with the naturally occurring Cas9 molecule. For example, a Cas9 molecule comprises an altered PI domain if its PI domain is other than the PI domain naturally associated with the Cas9 core domain of the Cas9 molecule, or if its PI domain is not a naturally occurring PI domain associated with any Cas9 molecule. (Derived, as used in this sense, is not limited to physical derivation or even derivation from a specific source, and does not require a process limitation, but in some embodiments, includes mere structural similarity). An altered PI domain may have less than 99, 98, 97, 96, 95, 94, 93, 92, 91, 90, 89, 88, 87, 86, 85, 84, 83, 82, 81, 80, 70, 60, 50, 30, 40, 30, 20, or 10% homology with the native or endogenous PI domain of a subject naturally occurring Cas9 molecule from which the Cas9 core domain is derived. An altered PI domain may have a different RKR motif (the PAM recognition sequence) than that of the native or endogenous PI domain of the Cas9 species that supplies the Cas9 core domain. The RKR motif of an altered PI domain may differ from the RKR motif of the native or endogenous PI domain of the Cas9 core domain by 1, 2, or 3 residues. The RKR motif of the altered PI differs at the first position, the second position, the third position, the first and second positions, the first and third positions, the second and third positions, or all three positions, from the RKR motif of the PI endogenous to or naturally associated with the Cas9 core domain. In an embodiment, an altered PI domain is one having greater homology with the PI domain of a reference or donor naturally occurring Cas9 molecule (a heterologous Cas9) that with the native PI domain of a subject Cas9.
  • The terms “homology” or “identity”, as used interchangeably herein, refer to sequence identity between two amino acid sequences or two nucleic acid sequences, with identity being a more strict comparison. The phrases “percent identity or homology” and “% identity or homology” refer to the percentage of sequence identity found in a comparison of two or more amino acid sequences or nucleic acid sequences. Two or more sequences can be anywhere from 0-100% identical, or any value there between. Identity can be determined by comparing a position in each sequence that can be aligned for purposes of comparison to a reference sequence. When a position in the compared sequence is occupied by the same nucleotide base or amino acid, then the molecules are identical at that position. A degree of identity of amino acid sequences is a function of the number of identical amino acids at positions shared by the amino acid sequences. A degree of identity between nucleic acid sequences is a function of the number of identical or matching nucleotides at positions shared by the nucleic acid sequences. A degree of homology of amino acid sequences is a function of the number of amino acids at positions shared by the polypeptide sequences.
  • Calculations of homology or sequence identity between two sequences (the terms are used interchangeably herein) are performed as follows. The sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in one or both of a first and a second amino acid or nucleic acid sequence for optimal alignment and non-homologous sequences can be disregarded for comparison purposes). The optimal alignment is determined as the best score using the GAP program in the GCG software package with a Blossum 62 scoring matrix with a gap penalty of 12, a gap extend penalty of 4, and a frame shift gap penalty of 5. The amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position. The percent identity between the two sequences is a function of the number of identical positions shared by the sequences.
  • “Linker”, as that term is used herein, refers to a molecular entity that may link a Cas9 molecule, or a fragment thereof, and a template binding domain, or may link a template binding domain partner to a template nucleic acid, or may link a Cas9 molecule fragment or domain to another Cas9 molecule fragment or domain. In one embodiment, the linker is a nucleic acid, e.g. an oligonucleotide. In another embodiment, the linker is a small molecule. In a further embodiment, the linker is a polypeptide comprising at least one amino acid. For example, a polypeptide linker may be disposed between sequences or domains of a Cas9 molecule. In an embodiment, the linker is disposed between the amino acid residues that flank a deletion. In an embodiment, the linker is disposed between the amino acid residues of a Cas9 core domain and an altered PI domain. By way of example, in a sequence CT1-CT2-CT3-CT7-CT8-CT9, wherein CT4-CT5-CT6 is deleted, the linker is located immediately C-terminal to the amino acid residue CT3 and immediately N-terminal to the amino acid residue CT7. Preferably, the linker is selected such that the Cas9 molecule exhibits a tertiary structure or folded conformation similar to that of the corresponding naturally occurring Cas9 molecule, such that some Cas9 activity is retained. Suitable linkers are described herein. In some embodiments, the linker comprises a combination of Gly and Ser residues, e.g., (GS)x or (GGS)x, where x is 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10. In other embodiments, the linker comprises a linker comprising the amino acid sequence SGSETPGTSESATPES (SEQ ID NO: _), referred to herein as XTEN linker or XTEN. Alternative linkers include (GSAGSAAGSGEF)x, wherein x is 1, 2, 3 or 4 (SEQ ID NO: _) and (SIVAQLSRPDPA)x, wherein x is 1, 2, 3 or 4 (SEQ ID NO: _). Linkers also include a combination of linkers described herein or known in the art.
  • “REC deletion”, as that term is used herein, refers to a REC2 deletion, a REC1CT deletion, or a REC1SUB deletion.
  • “REC2 deletion”, as that term is used herein, refers to a deletion of at least 10% of the amino acid residues of the REC2 domain.
  • “REC2 domain”, as that term is used herein, refers to a region, in the N terminal half of a naturally occurring Cas9 molecule that is not needed for cleavage or gRNA-mediated targeting. Its length and boundaries differ between Cas9 molecules from various species. In the case of S. aureus, the REC2 domain is about 41 amino acid residues in length and corresponds, approximately, to residues 126 to 166, of S. aureus Cas9. In the case of S. pyogenes, the REC2 domain is about 139 amino acid residues in length and corresponds, approximately, to residues 176 to 314 of S. pyogenes Cas9. In the case of C. jejuni, the REC2 domain is about 45 amino acid residues in length and corresponds, approximately, to residues 137 to 181 of C. jejuni Cas9. These, and the approximate sizes and boundaries of REC2 domains from other species, are provided in Table 100.
  • “REC1CT deletion”, as that term is used herein, refers to a deletion of at least 10% of the amino acid residues of the REC1CT domain.
  • “REC1CT domain”, as that term is used herein, refers to a region, C terminal of the REC1 domain, of a naturally occurring Cas9 polypeptide that is not needed for cleavage or gRNA-mediated targeting. Its length and boundaries differ between Cas9 proteins from various species. In the case of S. aureus, the REC1CT domain is about 146 amino acid residues in length and corresponds, approximately, to residues 288 to 166, of S. aureus Cas9. In the case of S. pyogenes, the REC1CT domain is about 219 amino acid residues in length and corresponds, approximately, to residues 500 to 718 of S. pyogenes Cas9. In the case of C. jejuni, the REC1CT domain is about 134 amino acid residues in length and corresponds, approximately, to residues 305 to 438 of C. jejuni Cas9. These, and the approximate sizes and boundaries of REC1CT domains from other species, are provided in Table 100.
  • “REC1SUB deletion”, as that term is used herein, refers to a deletion of at least 10% of the amino acid residues of the REC1SUB domain.
  • “REC1SUB domain”, as that term is used herein, refers to a region, located within the REC1CT domain, of a naturally occurring Cas9 polypeptide that is not needed for cleavage or gRNA-mediated targeting. Its length and boundaries differ between Cas9 proteins from various species. In the case of S. aureus, the REC1SUB domain is about 57 amino acid residues in length and corresponds, approximately, to residues 296 to 352, of S. aureus Cas9. In the case of S. pyogenes, the REC1SUB domain is about 82 amino acid residues in length and corresponds, approximately, to residues 511 to 592 of S. pyogenes Cas9. In the case of C. jejuni, the REC1SUB domain is about 45 amino acid residues in length and corresponds, approximately, to residues 316 to 360 of C. jejuni Cas9. These, and the approximate sizes and boundaries of REC1SUB domains from other species, are provided in Table 100.
  • “n” as used herein in the context of proteins or Cas9 molecules described herein, refers to the number of amino acid residues that are deleted in a REC2, REC1CT, or REC1SUB deletion, unless otherwise specified.
  • “X” as used herein in the context of an amino acid sequence of a linker sequence, refers to any number of repeating units unless otherwise specified.
  • A disorder “caused by” a mutation, as used herein, refers to a disorder that is made more likely or severe by the presence of the mutation, compared to a subject that does not have the mutation. The mutation need not be the only cause of a disorder, i.e., the disorder can still be caused by the mutation even if other causes, such as environmental factors or lifestyle factors, contribute causally to the disorder. In some embodiments, the disorder is caused by the mutation if the mutation is a medically recognized risk factor for developing the disorder, and/or if a study has found that the mutation correlates with development of the disorder.
  • “Derived from”, as used herein, refers to the source or origin of a molecular entity, e.g., a nucleic acid or protein. The source of a molecular entity may be naturally-occurring, recombinant, unpurified, or a purified molecular entity. For example, a polypeptide that is derived from a second polypeptide comprises an amino acid sequence that is identical or substantially similar, e.g., is more than 50% homologous to, the amino acid sequence of the second protein. The derived molecular entity, e.g., a nucleic acid or protein, can comprise one or more modifications, e.g., one or more amino acid or nucleotide changes.
  • “Domain”, as used herein, is used to describe segments of a protein or nucleic acid. Unless otherwise indicated, a domain is not required to have any specific functional property.
  • As used herein, “HDR”, or homology-directed repair, refers to the process of repairing DNA damage using a homologous nucleic acid (e.g., a sister chromatid or an exogenous nucleic acid). In a normal cell, HDR typically involves a series of steps such as recognition of the break, stabilization of the break, resection, stabilization of single stranded DNA, formation of a DNA crossover intermediate, resolution of the crossover intermediate, and ligation.
  • “Large molecule”, as used herein, refers to a molecule having a molecular weight of at least 2, 3, 5, 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100 kDa. Large molecules include proteins, polypeptides, nucleic acids, biologics, and carbohydrates.
  • “Polypeptide”, as used herein, refers to a polymer of amino acids.
  • “Reference molecule”, e.g., a reference Cas9 molecule or reference gRNA, as used herein, refers to a molecule to which a subject molecule, e.g., a subject Cas9 molecule or a subject gRNA molecule, e.g., a modified or candidate Cas9 molecule, is compared. For example, a Cas9 molecule can be characterized as having no more than 10% of the nuclease activity of a reference Cas9 molecule. Examples of reference Cas9 molecules include naturally occurring unmodified Cas9 molecules, e.g., a naturally occurring Cas9 molecule such as a Cas9 molecule of S. aureus, S. pyogenes, or S. thermophilus. In an embodiment, the reference Cas9 molecule is the naturally occurring Cas9 molecule having the closest sequence identity or homology with the Cas9 molecule to which it is being compared. In an embodiment, the reference Cas9 molecule is a sequence, e.g., a naturally occurring or known sequence, which is the parental form on which a change, e.g., a mutation has been made.
  • “Replacement”, or “replaced”, as used herein with reference to a modification of a molecule does not require a process limitation but merely indicates that the replacement entity is present.
  • “Small molecule”, as used herein, refers to a compound having a molecular weight less than about 2 kDa, e.g., less than about 2 kDa, less than about 1.5 kDa, less than about 1 kDa, or less than about 0.75 kDa.
  • “Steric interference” or “steric hindrance”, as used herein, refers to the restriction or prevention of the binding or interaction of one molecular entity (e.g., a protein or a protein fragment) with another molecular entity (e.g., a nucleic acid or a protein).
  • “Subject”, as used herein, may mean either a human or non-human animal. The term includes, but is not limited to, mammals (e.g., humans, other primates, pigs, rodents (e.g., mice and rats or hamsters), rabbits, guinea pigs, cows, horses, cats, dogs, sheep, and goats. In an embodiment, the subject is a human. In other embodiments, the subject is poultry.
  • “Sufficiently long”, as the term is used herein to refer to linkers, refers to a linker length that does not prevent or restrict the binding, folding, conformation, activity, and/or interaction of one molecular entity (e.g., a protein) with another molecular entity (e.g., a nucleic acid). In one embodiment, the linker is at least 6, but no longer than 60 amino acids in length. In another embodiment, the linker is at least 18, but no longer than 180 nucleotides in length. In one embodiment, the linker is at least 10, 50, 100, 200, 500, 1000, 2000, 5000, or 10000 Angstroms in length. In one embodiment, the linker is no more than 10, 50, 100, 200, 500, 1000, 2000, 5000, or 10000 Angstroms in length.
  • “Treat”, “treating” and “treatment”, as used herein, mean the treatment of a disease in a mammal, e.g., in a human, including (a) inhibiting the disease, i.e., arresting or preventing its development; (b) relieving the disease, i.e., causing regression of the disease state; and (c) curing the disease.
  • “Prevent,” “preventing” and “prevention,” as used herein, means the prevention of a disease in a subject, e.g., a mammal, e.g., in a human, including (a) avoiding or precluding the disease; (2) affecting the predisposition toward the disease, e.g., preventing at least one symptom of the disease or to delay onset of at least one symptom of the disease.
  • “Specific affinity”, or “specifically binds”, or “specific binding”, as used herein, refer to a binding interaction between two or more molecular entities, e.g., a template binding domain and a template binding domain partner, such as, for example, a DNA-binding polypeptide and a DNA molecule, wherein one molecular entity preferentially binds to another molecular entity, but does not substantially bind to other molecular entities present in a molecular milieu, e.g., a heterologous molecular milieu, in a cell or in solution. In some embodiments, the term “specific affinity” refers to a DNA-binding protein or polypeptide that binds to a specific sequence of a nucleic acid molecule. Exemplary protein-protein pairs that specifically bind to each other are provided in Table V.6. Exemplary protein-small molecule pairs that specifically bind to each other are provided in Table V.7. Exemplary protein-DNA sequence pairs that specifically bind to each other are provided in Table V.1.
  • “Substantial affinity”, as the term is used herein, refers to a binding interaction between two or more molecular entities, e.g., a template binding domain and a template binding domain partner, such as, for example, a DNA-binding polypeptide and a DNA molecule, wherein the binding event induces a significant event, change or alteration in the molecular entity being bound, or a significant phenotypic change in a cell in which the binding event occurs.
  • “Target position” or “target nucleic acid” as used herein, refers to a site on a nucleic acid (e.g., a region of a chromosome) that is modified by a Cas9 molecule-dependent process. For example, the target position can be modified by a Cas9 molecule-mediated cleavage of the nucleic acid using a template nucleic acid. In an embodiment, a target position can be a site between two nucleotides, e.g., adjacent nucleotides, on the target nucleic acid into which one or more nucleotides is added. The target position may comprise one or more nucleotides that are altered, e.g., corrected, by a template nucleic acid. In an embodiment, the target position is within a “target sequence” (e.g., the sequence to which the gRNA binds). In an embodiment, a target position is upstream or downstream of a target sequence (e.g., the sequence to which the gRNA binds).
  • The “targeting domain” of the gRNA is complementary to the “target domain” on the target nucleic acid.
  • A “template binding domain partner” as used herein, is a molecule with specific affinity for a template binding domain. The template binding domain partner may be, e.g., DNA, protein, or a small molecule.
  • A “template nucleic acid”, as that term is used herein, refers to a nucleic acid sequence which can be used in conjunction with a Cas9 molecule and a gRNA molecule to alter the structure of a target position. “Template nucleic acid” is used interchangeably with “donor nucleic acid” and “swap nucleic acid” herein. In an embodiment, the target nucleic acid is modified to have some or all of the sequence of the template nucleic acid, typically at or near cleavage site(s). In an embodiment, the template nucleic acid is single stranded. In an alternate embodiment, the template nucleic acid is double stranded. In an embodiment, the template nucleic acid is DNA, e.g., double stranded DNA. In an alternate embodiment, the template nucleic acid is single stranded DNA. In an embodiment, the template nucleic acid is encoded on the same vector backbone, e.g. AAV genome, plasmid DNA, as the Cas9 and gRNA. In an embodiment, the template nucleic acid is excised from a vector backbone in vivo, e.g., it is flanked by gRNA recognition sequences.
  • “Wild type”, as used herein, refers to a gene or polypeptide which has the characteristics, e.g., the nucleotide or amino acid sequence, of a gene or polypeptide from a naturally-occurring source. The term “wild type” typically includes the most frequent observation of a particular gene or polypeptide in a population of organisms found in nature.
  • “X” as used herein in the context of an amino acid sequence, refers to any amino acid (e.g., any of the twenty natural amino acids) unless otherwise specified.
  • A “template binding domain,” as that term is used herein, refers to an entity which, by virtue of its specific affinity for a template binding domain partner, mediates the association of a template nucleic acid with a Cas9 with which the template binding domain is associated. The template binding domain associates with, e.g., by non-covalent or covalent interactions, with a template binding domain partner. The template binding domain partner is associated with, e.g., covalently or non-covalently bound to, the template nucleic acid. In an embodiment, the template binding domain comprises a polypeptide and the template binding domain partner comprises a nucleic acid. Typically, the template binding domain does not cleave nucleic acid.
  • I. gRNA Molecules
  • A gRNA molecule, as that term is used herein, refers to a nucleic acid that promotes the specific targeting or homing of a gRNA molecule/Cas9 molecule complex to a target nucleic acid. Typically, the nucleic acid will incorporate the functions or structure of both crRNA and tracrRNA, e.g., the functions of processed or mature crRNA and of processed or mature tracrRNA. gRNA molecules can be unimolecular (having a single nucleic acid molecule, e.g., which incorporates both crRNA function or structure and the tracrRNA function or structure), sometimes referred to herein as “chimeric” gRNAs, or modular (comprising more than one, and typically two, separate nucleic acid molecules, e.g., where one incorporates the crRNA function or structure and the other incorporates the tracrRNA function or structure). A gRNA molecule comprises a number of domains. The gRNA molecule domains are described in more detail below. Additional details on gRNAs are provided in Section I entitled “gRNA molecules” of PCT Application WO 2015/048577, the entire contents of which are expressly incorporated herein by reference.
  • In an embodiment, a unimolecular, or chimeric, gRNA comprises, preferably from 5′ to 3′: a targeting domain (which is complementary to a target nucleic acid, and which is sometimes referred to as a spacer); a first complementarity domain; a linking domain; a second complementarity domain (which is complementary to the first complementarity domain); a proximal domain; and optionally, a tail domain. In an embodiment, the targeting domain, and first complementarity domain correspond functionally or structurally to elements of a crRNA, e.g., a mature or processed crRNA. In an embodiment, the second complementarity domain, proximal domain, and tail domain correspond functionally or structurally to elements of a tracrRNA, e.g., a processed or mature tracrRNA.
  • In an embodiment, a modular gRNA comprises: a first strand (which corresponds to a crRNA) comprising, preferably from 5′ to 3′; a targeting domain (which is complementary to a target nucleic acid); and a first complementarity domain; and a second strand (which corresponds to a tracrRNA), comprising, preferably from 5′ to 3′: optionally, a 5′ extension domain; a second complementarity domain; a proximal domain; and optionally, a tail domain.
  • The domains are discussed briefly below.
  • The Targeting Domain
  • The targeting domain (which can also be referred to as a “spacer”) comprises a nucleotide sequence that is complementary, e.g., at least 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, or 99% complementary, e.g., fully complementary, to the target sequence on the target nucleic acid. The targeting domain is part of an RNA molecule and will therefore comprise the base uracil (U), while any DNA encoding the gRNA molecule will comprise the base thymine (T). While not wishing to be bound by theory, in an embodiment, it is believed that the complementarity of the targeting domain with the target sequence contributes to specificity of the interaction of the gRNA molecule/Cas9 molecule complex with a target nucleic acid. It is understood that in a targeting domain and target sequence pair, the uracil bases in the targeting domain will pair with the adenine bases in the target sequence. In an embodiment, the target domain itself comprises in the 5′ to 3′ direction, an optional secondary domain, and a core domain. In an embodiment, the core domain is fully complementary with the target sequence. In an embodiment, the targeting domain is 5 to 50 nucleotides in length, e.g., 10 to 30, e.g., 16, 17, 18, 19, 20, 21, 22, 23, 24, 25 or 26, nucleotides in length. The strand of the target nucleic acid with which the targeting domain is complementary is referred to herein as the complementary strand. Some or all of the nucleotides of the targeting domain can have a modification, e.g., a modification found in Section XI herein.
  • In an embodiment, the targeting domain is 16 nucleotides in length.
  • In an embodiment, the targeting domain is 17 nucleotides in length.
  • In an embodiment, the targeting domain is 18 nucleotides in length.
  • In an embodiment, the targeting domain is 19 nucleotides in length.
  • In an embodiment, the targeting domain is 20 nucleotides in length.
  • In an embodiment, the targeting domain is 21 nucleotides in length.
  • In an embodiment, the targeting domain is 22 nucleotides in length.
  • In an embodiment, the targeting domain is 23 nucleotides in length.
  • In an embodiment, the targeting domain is 24 nucleotides in length.
  • In an embodiment, the targeting domain is 25 nucleotides in length.
  • In an embodiment, the targeting domain is 26 nucleotides in length.
  • In an embodiment, the targeting domain comprises 16 nucleotides.
  • In an embodiment, the targeting domain comprises 17 nucleotides.
  • In an embodiment, the targeting domain comprises 18 nucleotides.
  • In an embodiment, the targeting domain comprises 19 nucleotides.
  • In an embodiment, the targeting domain comprises 20 nucleotides.
  • In an embodiment, the targeting domain comprises 21 nucleotides.
  • In an embodiment, the targeting domain comprises 22 nucleotides.
  • In an embodiment, the targeting domain comprises 23 nucleotides.
  • In an embodiment, the targeting domain comprises 24 nucleotides.
  • In an embodiment, the targeting domain comprises 25 nucleotides.
  • In an embodiment, the targeting domain comprises 26 nucleotides.
  • Targeting domains are discussed in more detail below.
  • The First Complementarity Domain
  • The first complementarity domain is complementary with the second complementarity domain, and in an embodiment, has sufficient complementarity to the second complementarity domain to form a duplexed region under at least some physiological conditions. In an embodiment, the first complementarity domain is 5 to 30 nucleotides in length. In an embodiment, the first complementarity domain is 5 to 25 nucleotides in length. In an embodiment, the first complementary domain is 7 to 25 nucleotides in length. In an embodiment, the first complementary domain is 7 to 22 nucleotides in length. In an embodiment, the first complementary domain is 7 to 18 nucleotides in length. In an embodiment, the first complementary domain is 7 to 15 nucleotides in length. In an embodiment, the first complementary domain is 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25 nucleotides in length.
  • In an embodiment, the first complementarity domain comprises 3 subdomains, which, in the 5′ to 3′ direction are: a 5′ subdomain, a central subdomain, and a 3′ subdomain. In an embodiment, the 5′ subdomain is 4 to 9, e.g., 4, 5, 6, 7, 8 or 9 nucleotides in length. In an embodiment, the central subdomain is 1, 2, or 3, e.g., 1, nucleotide in length. In an embodiment, the 3′ subdomain is 3 to 25, e.g., 4 to 22, 4 to 18, or 4 to 10, or 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25 nucleotides in length.
  • The first complementarity domain can share homology with, or be derived from, a naturally occurring first complementarity domain. In an embodiment, it has at least 50% homology with a first complementarity domain disclosed herein, e.g., an S. pyogenes, S. aureus or S. thermophilus, first complementarity domain.
  • Some or all of the nucleotides of the domain can have a modification, e.g., a modification found in Section XI herein.
  • First complementarity domains are discussed in more detail below.
  • The Linking Domain
  • A linking domain serves to link the first complementarity domain with the second complementarity domain of a unimolecular gRNA. The linking domain can link the first and second complementarity domains covalently or non-covalently. In an embodiment, the linkage is covalent. In an embodiment, the linking domain covalently couples the first and second complementarity domains. In an embodiment, the linking domain is, or comprises, a covalent bond interposed between the first complementarity domain and the second complementarity domain. Typically the linking domain comprises one or more, e.g., 2, 3, 4, 5, 6, 7, 8, 9, or 10 nucleotides.
  • In modular gRNA molecules the two molecules are associated by virtue of the hybridization of the complementarity domains.
  • A wide variety of linking domains are suitable for use in unimolecular gRNA molecules. Linking domains can consist of a covalent bond, or be as short as one or a few nucleotides, e.g., 1, 2, 3, 4, or 5 nucleotides in length. In an embodiment, a linking domain is 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, or 25 or more nucleotides in length. In an embodiment, a linking domain is 2 to 50, 2 to 40, 2 to 30, 2 to 20, 2 to 10, or 2 to 5 nucleotides in length. In an embodiment, a linking domain shares homology with, or is derived from, a naturally occurring sequence, e.g., the sequence of a tracrRNA that is 5′ to the second complementarity domain. In an embodiment, the linking domain has at least 50% homology with a linking domain disclosed herein.
  • Some or all of the nucleotides of the domain can have a modification, e.g., a modification found in Section XI herein.
  • Linking domains are discussed in more detail below.
  • The 5′ Extension Domain
  • In an embodiment, a modular gRNA can comprise additional sequence, 5′ to the second complementarity domain, referred to herein as the 5′ extension domain. In an embodiment, the 5′ extension domain is, 2 to 10, 2 to 9, 2 to 8, 2 to 7, 2 to 6, 2 to 5, or 2 to 4, nucleotides in length. In an embodiment, the 5′ extension domain is 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more nucleotides in length.
  • The Second Complementarity Domain
  • The second complementarity domain is complementary with the first complementarity domain, and in an embodiment, has sufficient complementarity to the second complementarity domain to form a duplexed region under at least some physiological conditions. In an embodiment, the second complementarity domain can include sequence that lacks complementarity with the first complementarity domain, e.g., sequence that loops out from the duplexed region.
  • In an embodiment, the second complementarity domain is 5 to 27 nucleotides in length. In an embodiment, it is longer than the first complementarity region. In an embodiment the second complementary domain is 7 to 27 nucleotides in length. In an embodiment, the second complementary domain is 7 to 25 nucleotides in length. In an embodiment, the second complementary domain is 7 to 20 nucleotides in length. In an embodiment, the second complementary domain is 7 to 17 nucleotides in length. In an embodiment, the complementary domain is 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25 or 26 nucleotides in length.
  • In an embodiment, the second complementarity domain comprises 3 subdomains, which, in the 5′ to 3′ direction are: a 5′ subdomain, a central subdomain, and a 3′ subdomain. In an embodiment, the 5′ subdomain is 3 to 25, e.g., 4 to 22, 4 to 18, or 4 to 10, or 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25 nucleotides in length. In an embodiment, the central subdomain is 1, 2, 3, 4 or 5, e.g., 3, nucleotides in length. In an embodiment, the 3′ subdomain is 4 to 9, e.g., 4, 5, 6, 7, 8 or 9 nucleotides in length.
  • In an embodiment, the 5′ subdomain and the 3′ subdomain of the first complementarity domain, are respectively, complementary, e.g., fully complementary, with the 3′ subdomain and the 5′ subdomain of the second complementarity domain.
  • The second complementarity domain can share homology with or be derived from a naturally occurring second complementarity domain. In an embodiment, it has at least 50% homology with a second complementarity domain disclosed herein, e.g., an S. pyogenes, S. aureus or S. thermophilus, first complementarity domain.
  • Some or all of the nucleotides of the domain can have a modification, e.g., a modification found in Section XI herein.
  • A Proximal Domain
  • In an embodiment, the proximal domain is 5 to 20 nucleotides in length. In an embodiment, the proximal domain can share homology with or be derived from a naturally occurring proximal domain. In an embodiment, it has at least 50% homology with a proximal domain disclosed herein, e.g., an S. pyogenes, S. aureus or S. thermophilus, proximal domain.
  • Some or all of the nucleotides of the domain can have a modification, e.g., a modification found in Section XI herein.
  • A Tail Domain
  • A broad spectrum of tail domains are suitable for use in gRNA molecules. In an embodiment, the tail domain is 0 (absent), 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 nucleotides in length. In embodiment, the tail domain nucleotides are from or share homology with sequence from the 5′ end of a naturally occurring tail domain. In an embodiment, the tail domain includes sequences that are complementary to each other and which, under at least some physiological conditions, form a duplexed region.
  • In an embodiment, the tail domain is absent or is 1 to 50 nucleotides in length. In an embodiment, the tail domain can share homology with or be derived from a naturally occurring proximal tail domain. In an embodiment, it has at least 50% homology with a tail domain disclosed herein, e.g., an S. pyogenes, S. aureus or S. thermophilus, tail domain.
  • In an embodiment, the tail domain includes nucleotides at the 3′ end that are related to the method of in vitro or in vivo transcription. When a T7 promoter is used for in vitro transcription of the gRNA, these nucleotides may be any nucleotides present before the 3′ end of the DNA template. When a U6 promoter is used for in vivo transcription, these nucleotides may be the sequence UUUUUU. When alternate pol-III promoters are used, these nucleotides may be various numbers or uracil bases or may include alternate bases.
  • The domains of gRNA molecules are described in more detail below.
  • The Targeting Domain
  • The “targeting domain” of the gRNA is complementary to the “target domain” on the target nucleic acid. The strfnd of the target nucleic acid comprising the nucleotide sequence complementary to the core domain of the gRNA is referred to herein as the “complementary strand” of the target nucleic acid. Guidance on the selection of targeting domains can be found, e.g., in Fu Y et al. (2014) NAT. BIOTECHNOL. 32: 279-84 (doi: 10.1038/nbt.2808) and Sternberg S H et al. (2014) NATURE 507: 62-7 (doi: 10.1038/nature13011).
  • In an embodiment, the targeting domain is 16, 17, 18, 19, 20, 21, 22, 23, 24, 25 or 26 nucleotides in length.
  • In an embodiment, the targeting domain is 16 nucleotides in length.
  • In an embodiment, the targeting domain is 17 nucleotides in length.
  • In an embodiment, the targeting domain is 18 nucleotides in length.
  • In an embodiment, the targeting domain is 19 nucleotides in length.
  • In an embodiment, the targeting domain is 20 nucleotides in length.
  • In an embodiment, the targeting domain is 21 nucleotides in length.
  • In an embodiment, the targeting domain is 22 nucleotides in length.
  • In an embodiment, the targeting domain is 23 nucleotides in length.
  • In an embodiment, the targeting domain is 24 nucleotides in length.
  • In an embodiment, the targeting domain is 25 nucleotides in length.
  • In an embodiment, the targeting domain is 26 nucleotides in length.
  • In an embodiment, the targeting domain comprises 16 nucleotides.
  • In an embodiment, the targeting domain comprises 17 nucleotides.
  • In an embodiment, the targeting domain comprises 18 nucleotides.
  • In an embodiment, the targeting domain comprises 19 nucleotides.
  • In an embodiment, the targeting domain comprises 20 nucleotides.
  • In an embodiment, the targeting domain comprises 21 nucleotides.
  • In an embodiment, the targeting domain comprises 22 nucleotides.
  • In an embodiment, the targeting domain comprises 23 nucleotides.
  • In an embodiment, the targeting domain comprises 24 nucleotides.
  • In an embodiment, the targeting domain comprises 25 nucleotides.
  • In an embodiment, the targeting domain comprises 26 nucleotides.
  • In an embodiment, the targeting domain is 10+/−5, 20+/−5, 30+/−5, 40+/−5, 50+/−5, 60+/−5, 70+/−5, 80+/−5, 90+/−5, or 100+/−5 nucleotides, in length.
  • In an embodiment, the targeting domain is 20+/−5 nucleotides in length.
  • In an embodiment, the targeting domain is 20+/−10, 30+/−10, 40+/−10, 50+/−10, 60+/−10, 70+/−10, 80+/−10, 90+/−10, or 100+/−10 nucleotides, in length.
  • In an embodiment, the targeting domain is 30+/−10 nucleotides in length.
  • In an embodiment, the targeting domain is 10 to 100, 10 to 90, 10 to 80, 10 to 70, 10 to 60, 10 to 50, 10 to 40, 10 to 30, 10 to 20 or 10 to 15 nucleotides in length.
  • In another embodiment, the targeting domain is 20 to 100, 20 to 90, 20 to 80, 20 to 70, 20 to 60, 20 to 50, 20 to 40, 20 to 30, or 20 to 25 nucleotides in length.
  • Typically the targeting domain has full complementarity with the target sequence. In an embodiment the targeting domain has or includes 1, 2, 3, 4, 5, 6, 7 or 8 nucleotides that are not complementary with the corresponding nucleotide of the targeting domain.
  • In an embodiment, the target domain includes 1, 2, 3, 4 or 5 nucleotides that are complementary with the corresponding nucleotide of the targeting domain within 5 nucleotides of its 5′ end. In an embodiment, the target domain includes 1, 2, 3, 4 or 5 nucleotides that are complementary with the corresponding nucleotide of the targeting domain within 5 nucleotides of its 3′ end.
  • In an embodiment, the target domain includes 1, 2, 3, or 4 nucleotides that are not complementary with the corresponding nucleotide of the targeting domain within 5 nucleotides of its 5′ end. In an embodiment, the target domain includes 1, 2, 3, or 4 nucleotides that are not complementary with the corresponding nucleotide of the targeting domain within 5 nucleotides of its 3′ end.
  • In an embodiment, the degree of complementarity, together with other properties of the gRNA, is sufficient to allow targeting of a Cas9 fusion molecule to the target nucleic acid.
  • In an embodiment, the targeting domain comprises two consecutive nucleotides that are not complementary to the target domain (“non-complementary nucleotides”), e.g., two consecutive noncomplementary nucleotides that are within 5 nucleotides of the 5′ end of the targeting domain, within 5 nucleotides of the 3′ end of the targeting domain, or more than 5 nucleotides away from one or both ends of the targeting domain.
  • In an embodiment, no two consecutive nucleotides within 5 nucleotides of the 5′ end of the targeting domain, within 5 nucleotides of the 3′ end of the targeting domain, or within a region that is more than 5 nucleotides away from one or both ends of the targeting domain, are not complementary to the targeting domain.
  • In an embodiment, there are no non-complementary nucleotides within 5 nucleotides of the 5′ end of the targeting domain, within 5 nucleotides of the 3′ end of the targeting domain, or within a region that is more than 5 nucleotides away from one or both ends of the targeting domain.
  • In an embodiment, the targeting domain nucleotides do not comprise modifications, e.g., modifications of the type provided in Section XI. However, in an embodiment, the targeting domain comprises one or more modifications, e.g., modifications that it render it less susceptible to degradation or more bio-compatible, e.g., less immunogenic. By way of example, the backbone of the targeting domain can be modified with a phosphorothioate, or other modification from Section XI. In an embodiment, a nucleotide of the targeting domain can comprise a 2′ modification (e.g., a modification at the 2′ position on ribose), e.g., a 2-acetylation, e.g., a 2′ methylation, or other modification(s) from Section XI.
  • In an embodiment, the targeting domain includes 1, 2, 3, 4, 5, 6, 7 or 8 or more modifications. In an embodiment, the targeting domain includes 1, 2, 3, or 4 modifications within 5 nucleotides of its 5′ end. In an embodiment, the targeting domain comprises as many as 1, 2, 3, or 4 modifications within 5 nucleotides of its 3′ end.
  • In an embodiment, the targeting domain comprises modifications at two consecutive nucleotides, e.g., two consecutive nucleotides that are within 5 nucleotides of the 5′ end of the targeting domain, within 5 nucleotides of the 3′ end of the targeting domain, or more than 5 nucleotides away from one or both ends of the targeting domain.
  • In an embodiment, no two consecutive nucleotides are modified within 5 nucleotides of the 5′ end of the targeting domain, within 5 nucleotides of the 3′ end of the targeting domain, or within a region that is more than 5 nucleotides away from one or both ends of the targeting domain. In an embodiment, no nucleotide is modified within 5 nucleotides of the 5′ end of the targeting domain, within 5 nucleotides of the 3′ end of the targeting domain, or within a region that is more than 5 nucleotides away from one or both ends of the targeting domain.
  • Modifications in the targeting domain can be selected to not interfere with targeting efficacy, which can be evaluated by testing a candidate modification in the system described in Section VII. gRNAs having a candidate targeting domain having a selected length, sequence, degree of complementarity, or degree of modification, can be evaluated in a system in Section VII. The candidate targeting domain can be placed, either alone, or with one or more other candidate changes in a gRNA molecule/Cas9 molecule system known to be functional with a selected target and evaluated.
  • In an embodiment, all of the modified nucleotides are complementary to and capable of hybridizing to corresponding nucleotides present in the target domain. In another embodiment, 1, 2, 3, 4, 5, 6, 7 or 8 or more modified nucleotides are not complementary to or capable of hybridizing to corresponding nucleotides present in the target domain.
  • In an embodiment, the targeting domain comprises, preferably in the 5′-3′ direction: a secondary domain and a core domain. These domains are discussed in more detail below.
  • The Core Domain and Secondary Domain of the Targeting Domain
  • The “core domain” of the targeting domain is complementary to the “core domain target” on the target nucleic acid. In an embodiment, the core domain comprises about 8 to about 13 nucleotides from the 3′ end of the targeting domain (e.g., the most 3′ 8 to 13 nucleotides of the targeting domain).
  • In an embodiment, the core domain of the targeting domain and core domain target, are independently, 6+/−2, 7+/−2, 8+/−2, 9+/−2, 10+/−2, 11+/−2, 12+/−2, 13+/−2, 14+/−2, 15+/−2, or 16+−2, nucleotides in length.
  • In an embodiment, the core domain of the targeting domain and core domain target, are independently, 10+/−2 nucleotides in length.
  • In an embodiment, the core domain of the targeting domain and core domain target, are independently, 10+/−4 nucleotides in length.
  • In an embodiment, the core domain of the targeting domain and core domain target are independently 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18 nucleotides in length.
  • In an embodiment, the core domain of the targeting domain and core domain target are independently 3 to 20, 4 to 20, 5 to 20, 6 to 20, 7 to 20, 8 to 20, 9 to 20 10 to 20 or 15 to 20 nucleotides in length.
  • In an embodiment, the core domain of the targeting domain and core domain target are independently 3 to 15, e.g., 6 to 15, 7 to 14, 7 to 13, 6 to 12, 7 to 12, 7 to 11, 7 to 10, 8 to 14, 8 to 13, 8 to 12, 8 to 11, 8 to 10 or 8 to 9 nucleotides in length.
  • The core domain of the targeting domain is complementary with the core domain target. Typically the core domain has exact complementarity with the core domain target. In an embodiment, the core domain of the targeting domain can have 1, 2, 3, 4 or 5 nucleotides that are not complementary with the corresponding nucleotide of the core domain target. In an embodiment, the degree of complementarity, together with other properties of the gRNA molecule, is sufficient to allow targeting of a Cas9 molecule to the target nucleic acid.
  • The “secondary domain” of the targeting domain of the gRNA is complementary to the “secondary domain target” of the target nucleic acid.
  • In an embodiment, the secondary domain is positioned 5′ to the core domain.
  • In an embodiment, the secondary domain is absent or optional.
  • In an embodiment, if the targeting domain is 26 nucleotides in length and the core domain (counted from the 3′ end of the targeting domain) is 8 to 13 nucleotides in length, the secondary domain is 13 to 18 nucleotides in length.
  • In an embodiment, if the targeting domain is 25 nucleotides in length and the core domain (counted from the 3′ end of the targeting domain) is 8 to 13 nucleotides in length, the secondary domain is 12 to 17 nucleotides in length.
  • In an embodiment, if the targeting domain is 24 nucleotides in length and the core domain (counted from the 3′ end of the targeting domain) is 8 to 13 nucleotides in length, the secondary domain is 11 to 16 nucleotides in length.
  • In an embodiment, if the targeting domain is 23 nucleotides in length and the core domain (counted from the 3′ end of the targeting domain) is 8 to 13 nucleotides in length, the secondary domain is 10 to 15 nucleotides in length.
  • In an embodiment, if the targeting domain is 22 nucleotides in length and the core domain (counted from the 3′ end of the targeting domain) is 8 to 13 nucleotides in length, the secondary domain is 9 to 14 nucleotides in length.
  • In an embodiment, if the targeting domain is 21 nucleotides in length and the core domain (counted from the 3′ end of the targeting domain) is 8 to 13 nucleotides in length, the secondary domain is 8 to 13 nucleotides in length.
  • In an embodiment, if the targeting domain is 20 nucleotides in length and the core domain (counted from the 3′ end of the targeting domain) is 8 to 13 nucleotides in length, the secondary domain is 7 to 12 nucleotides in length.
  • In an embodiment, if the targeting domain is 19 nucleotides in length and the core domain (counted from the 3′ end of the targeting domain) is 8 to 13 nucleotides in length, the secondary domain is 6 to 11 nucleotides in length.
  • In an embodiment, if the targeting domain is 18 nucleotides in length and the core domain (counted from the 3′ end of the targeting domain) is 8 to 13 nucleotides in length, the secondary domain is 5 to 10 nucleotides in length.
  • In an embodiment, if the targeting domain is 17 nucleotides in length and the core domain (counted from the 3′ end of the targeting domain) is 8 to 13 nucleotides in length, the secondary domain is 4 to 9 nucleotides in length.
  • In an embodiment, if the targeting domain is 16 nucleotides in length and the core domain (counted from the 3′ end of the targeting domain) is 8 to 13 nucleotides in length, the secondary domain is 3 to 8 nucleotides in length.
  • In an embodiment, the secondary domain is 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18 nucleotides in length.
  • The secondary domain of the targeting domain is complementary with the secondary domain target. Typically the secondary domain of the targeting domain has exact complementarity with the secondary domain target. In an embodiment the secondary domain of the targeting domain can have 1, 2, 3, 4 or 5 nucleotides that are not complementary with the corresponding nucleotide of the secondary domain target. In an embodiment, the degree of complementarity, together with other properties of the gRNA, is sufficient to allow targeting of a Cas9 fusion molecule to the target nucleic acid.
  • In an embodiment, the core domain nucleotides do not comprise modifications, e.g., modifications of the type provided in Section XI. However, in an embodiment, the core domain comprises one or more modifications, e.g., modifications that it render it less susceptible to degradation or more bio-compatible, e.g., less immunogenic. By way of example, the backbone of the core domain can be modified with a phosphorothioate, or other modification(s) from Section XI. In an embodiment a nucleotide of the core domain can comprise a 2′ modification (e.g., a modification at the 2′ position on ribose), e.g., a 2-acetylation, e.g., a 2′ methylation, or other modification(s) from Section XI. Typically, a core domain will contain no more than 1, 2, or 3 modifications.
  • Modifications in the core domain can be selected to not interfere with targeting efficacy, which can be evaluated by testing a candidate modification in the system described in Section VII. gRNAs having a candidate core domain having a selected length, sequence, degree of complementarity, or degree of modification, can be evaluated in the system described at Section VII. The candidate core domain can be placed, either alone, or with one or more other candidate changes in a gRNA molecule/Cas9 molecule system known to be functional with a selected target and evaluated.
  • In an embodiment, the secondary domain nucleotides do not comprise modifications, e.g., modifications of the type provided in Section XI. However, in an embodiment, the secondary domain comprises one or more modifications, e.g., modifications that render it less susceptible to degradation or more bio-compatible, e.g., less immunogenic. By way of example, the backbone of the secondary domain can be modified with a phosphorothioate, or other modification(s) from Section XI. In an embodiment a nucleotide of the secondary domain can comprise a 2′ modification (e.g., a modification at the 2′ position on ribose), e.g., a 2-acetylation, e.g., a 2′ methylation, or other modification from Section XI. Typically, a secondary domain will contain no more than 1, 2, or 3 modifications.
  • Modifications in the secondary domain can be selected to not interfere with targeting efficacy, which can be evaluated by testing a candidate modification in the system described in Section VII. gRNAs having a candidate secondary domain having a selected length, sequence, degree of complementarity, or degree of modification, can be evaluated in the system described at Section VII. The candidate secondary domain can be placed, either alone, or with one or more other candidate changes in a gRNA molecule/Cas9 molecule system known to be functional with a selected target and evaluated.
  • In an embodiment, (1) the degree of complementarity between the core domain of the targeting domain and its target (i.e., the core domain target), and (2) the degree of complementarity between the secondary domain of the targeting domain and its target (i.e., the secondary domain target), may differ. In an embodiment, (1) may be greater than (2). In an embodiment, (1) may be less than (2). In an embodiment, (1) and (2) are the same, e.g., each may be completely complementary with its target.
  • In an embodiment, (1) the number of modifications (e.g., modifications from Section XI) of the nucleotides of the core domain and (2) the number of modification (e.g., modifications from Section XI) of the nucleotides of the secondary domain, may differ. In an embodiment, (1) may be less than (2). In an embodiment, (1) may be greater than (2). In an embodiment, (1) and (2) may be the same, e.g., each may be free of modifications.
  • The First and Second Complementarity Domains
  • The first complementarity domain is complementary with the second complementarity domain.
  • Typically the first domain does not have exact complementarity with the second complementarity domain. In an embodiment, the first complementarity domain can have 1, 2, 3, 4 or 5 nucleotides that are not complementary with the corresponding nucleotide of the second complementarity domain. In an embodiment, 1, 2, 3, 4, 5 or 6, e.g., 3 nucleotides, will not pair in the duplex, and, e.g., form a non-duplexed or looped-out region. In an embodiment, an unpaired, or loop-out, region, e.g., a loop-out of 3 nucleotides, is present on the second complementarity domain. In an embodiment, the unpaired region begins 1, 2, 3, 4, 5, or 6, e.g., 4, nucleotides from the 5′ end of the second complementarity domain.
  • In an embodiment, the degree of complementarity, together with other properties of the gRNA, is sufficient to allow targeting of a Cas9 molecule to the target nucleic acid.
  • In an embodiment, the first and second complementarity domains are:
  • independently, 6+/−2, 7+/−2, 8+/−2, 9+/−2, 10+/−2, 11+/−2, 12+/−2, 13+/−2, 14+/−2, 15+/−2, 16+/−2, 17+/−2, 18+/−2, 19+/−2, or 20+/−2, 21+/−2, 22+/−2, 23+/−2, or 24+/−2 nucleotides in length;
  • independently, 6, 7, 8, 9, 10, 11, 12, 13, 14, 14, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, or 26, nucleotides in length; or
  • independently, 5 to 24, 5 to 23, 5 to 22, 5 to 21, 5 to 20, 7 to 18, 9 to 16, or 10 to 14 nucleotides in length.
  • In an embodiment, the second complementarity domain is longer than the first complementarity domain, e.g., 2, 3, 4, 5, or 6, e.g., 6, nucleotides longer.
  • In an embodiment, the first and second complementary domains, independently, do not comprise modifications, e.g., modifications of the type provided in Section XI.
  • In an embodiment, the first and second complementary domains, independently, comprise one or more modifications, e.g., modifications that the render the domain less susceptible to degradation or more bio-compatible, e.g., less immunogenic. By way of example, the backbone of the domain can be modified with a phosphorothioate, or other modification(s) from Section XI. In an embodiment a nucleotide of the domain can comprise a 2′ modification (e.g., a modification at the 2′ position on ribose), e.g., a 2-acetylation, e.g., a 2′ methylation, or other modification(s) from Section XI.
  • In an embodiment, the first and second complementary domains, independently, include 1, 2, 3, 4, 5, 6, 7 or 8 or more modifications. In an embodiment, the first and second complementary domains, independently, include 1, 2, 3, or 4 modifications within 5 nucleotides of its 5′ end. In an embodiment, the first and second complementary domains, independently, include as many as 1, 2, 3, or 4 modifications within 5 nucleotides of its 3′ end.
  • In an embodiment, the first and second complementary domains, independently, include modifications at two consecutive nucleotides, e.g., two consecutive nucleotides that are within 5 nucleotides of the 5′ end of the domain, within 5 nucleotides of the 3′ end of the domain, or more than 5 nucleotides away from one or both ends of the domain. In an embodiment, the first and second complementary domains, independently, include no two consecutive nucleotides that are modified, within 5 nucleotides of the 5′ end of the domain, within 5 nucleotides of the 3′ end of the domain, or within a region that is more than 5 nucleotides away from one or both ends of the domain. In an embodiment, the first and second complementary domains, independently, include no nucleotide that is modified within 5 nucleotides of the 5′ end of the domain, within 5 nucleotides of the 3′ end of the domain, or within a region that is more than 5 nucleotides away from one or both ends of the domain.
  • Modifications in a complementarity domain can be selected to not interfere with targeting efficacy, which can be evaluated by testing a candidate modification in the system described in Section VII. gRNAs having a candidate complementarity domain having a selected length, sequence, degree of complementarity, or degree of modification, can be evaluated in the system described in SectionVII. The candidate complementarity domain can be placed, either alone, or with one or more other candidate changes in a gRNA molecule/Cas9 molecule system known to be functional with a selected target and evaluated.
  • In an embodiment, the first complementarity domain has at least 60, 70, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99% homology with, or differs by no more than 1, 2, 3, 4, 5, or 6 nucleotides from, a reference first complementarity domain, e.g., a naturally occurring, e.g., an S. pyogenes, S. aureus or S. thermophilus, first complementarity domain, or a first complementarity domain described herein.
  • In an embodiment, the second complementarity domain has at least 60, 70, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99% homology with, or differs by no more than 1, 2, 3, 4, 5, or 6 nucleotides from, a reference second complementarity domain, e.g., a naturally occurring, e.g., an S. pyogenes, S. aureus or S. thermophilus, second complementarity domain, or a second complementarity domain described herein.
  • The duplexed region formed by first and second complementarity domains is typically 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 or 22 base pairs in length (excluding any looped out or unpaired nucleotides).
  • In an embodiment, the first and second complementarity domains, when duplexed, comprise 11 paired nucleotides, for example, in the gRNA sequence (one paired strand underlined, one bolded):
  • (SEQ ID NO: XX)
    NNNNNNNNNNNNNNNNNNNNGUUUUAGAGCUAGAAAUAGCAAGUUAAAA
    UAAGGCUAGUCCGUUAUCAACUUGAAAAAGUGGCACCGAGUCGGUGC.
  • In an embodiment, the first and second complementarity domains, when duplexed, comprise 15 paired nucleotides, for example in the gRNA sequence (one paired strand underlined, one bolded):
  • (SEQ ID NO: XX)
    NNNNNNNNNNNNNNNNNNNNGUUUUAGAGCUAUGCUGAAAAGCAUAGCAA
    GUUAAAAUAAGGCUAGUCCGUUAUCAACUUGAAAAAGUGGCACCGAGUCG
    GUGC.
  • In an embodiment the first and second complementarity domains, when duplexed, comprise 16 paired nucleotides, for example in the gRNA sequence (one paired strand underlined, one bolded):
  • (SEQ ID NO: XX)
    NNNNNNNNNNNNNNNNNNNNGUUUUAGAGCUAUGCUGGAAACAGCAUAGC
    AAGUUAAAAUAAGGCUAGUCCGUUAUCAACUUGAAAAAGUGGCACCGAGU
    CGGUGC.
  • In an embodiment the first and second complementarity domains, when duplexed, comprise 21 paired nucleotides, for example in the gRNA sequence (one paired strand underlined, one bolded):
  • (SEQ ID NO: XX)
    NNNNNNNNNNNNNNNNNNNNGUUUUAGAGCUAUGCUGUUUUGGAAACAAA
    ACAGCAUAGCAAGUUAAAAUAAGGCUAGUCCGUUAUCAACUUGAAAAAGU
    GGCACCGAGUCGGUGC.
  • In an embodiment, nucleotides are exchanged to remove poly-U tracts, for example in the gRNA sequences (exchanged nucleotides underlined):
  • (SEQ ID NO: XX)
    NNNNNNNNNNNNNNNNNNNNGUAUUAGAGCUAGAAAUAGCAAGUUAAUAU
    AAGGCUAGUCCGUUAUCAACUUGAAAAAGUGGCACCGAGUCGGUGC;
    (SEQ ID NO: XX)
    NNNNNNNNNNNNNNNNNNNNGUUUAAGAGCUAGAAAUAGCAAGUUUAAAU
    AAGGCUAGUCCGUUAUCAACUUGAAAAAGUGGCACCGAGUCGGUGC;
    or
    (SEQ ID NO: XX)
    NNNNNNNNNNNNNNNNNNNNGUAUUAGAGCUAUGCUGUAUUGGAAACAAU
    ACAGCAUAGCAAGUUAAUAUAAGGCUAGUCCGUUAUCAACUUGAAAAAGU
    GGCACCGAGUCGGUGC.
  • The 5′ Extension Domain
  • In an embodiment, a modular gRNA can comprise additional sequence, 5′ to the second complementarity domain. In an embodiment, the 5′ extension domain is 2 to 10, 2 to 9, 2 to 8, 2 to 7, 2 to 6, 2 to 5, or 2 to 4 nucleotides in length. In an embodiment, the 5′ extension domain is 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more nucleotides in length.
  • In an embodiment, the 5′ extension domain nucleotides do not comprise modifications, e.g., modifications of the type provided in Section XI. However, in an embodiment, the 5′ extension domain comprises one or more modifications, e.g., modifications that it render it less susceptible to degradation or more bio-compatible, e.g., less immunogenic. By way of example, the backbone of the 5′ extension domain can be modified with a phosphorothioate, or other modification(s) from Section XI. In an embodiment, a nucleotide of the 5′ extension domain can comprise a 2′ modification (e.g., a modification at the 2′ position on ribose), e.g., a 2-acetylation, e.g., a 2′ methylation, or other modification(s) from Section XI.
  • In an embodiment, the 5′ extension domain can comprise as many as 1, 2, 3, 4, 5, 6, 7 or 8 modifications. In an embodiment, the 5′ extension domain comprises as many as 1, 2, 3, or 4 modifications within 5 nucleotides of its 5′ end, e.g., in a modular gRNA molecule. In an embodiment, the 5′ extension domain comprises as many as 1, 2, 3, or 4 modifications within 5 nucleotides of its 3′ end, e.g., in a modular gRNA molecule.
  • In an embodiment, the 5′ extension domain comprises modifications at two consecutive nucleotides, e.g., two consecutive nucleotides that are within 5 nucleotides of the 5′ end of the 5′ extension domain, within 5 nucleotides of the 3′ end of the 5′ extension domain, or more than 5 nucleotides away from one or both ends of the 5′ extension domain. In an embodiment, no two consecutive nucleotides are modified within 5 nucleotides of the 5′ end of the 5′ extension domain, within 5 nucleotides of the 3′ end of the 5′ extension domain, or within a region that is more than 5 nucleotides away from one or both ends of the 5′ extension domain. In an embodiment, no nucleotide is modified within 5 nucleotides of the 5′ end of the 5′ extension domain, within 5 nucleotides of the 3′ end of the 5′ extension domain, or within a region that is more than 5 nucleotides away from one or both ends of the 5′ extension domain.
  • Modifications in the 5′ extension domain can be selected so as to not interfere with gRNA molecule efficacy, which can be evaluated by testing a candidate modification in the system described in Section VII. gRNAs having a candidate 5′ extension domain having a selected length, sequence, degree of complementarity, or degree of modification, can be evaluated in the system described at Section VII. The candidate 5′ extension domain can be placed, either alone, or with one or more other candidate changes in a gRNA molecule/Cas9 molecule system known to be functional with a selected target and evaluated.
  • In an embodiment, the 5′ extension domain has at least 60, 70, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99% homology with, or differs by no more than 1, 2, 3, 4, 5, or 6 nucleotides from, a reference 5′ extension domain, e.g., a naturally occurring, e.g., an S. pyogenes, S. aureus or S. thermophilus, 5′ extension domain, or a 5′ extension domain described herein.
  • The Linking Domain
  • In a unimolecular gRNA molecule the linking domain is disposed between the first and second complementarity domains. In a modular gRNA molecule, the two molecules are associated with one another by the complementarity domains.
  • In an embodiment, the linking domain is 10+/−5, 20+/−5, 30+/−5, 40+/−5, 50+/−5, 60+/−5, 70+/−5, 80+/−5, 90+/−5, or 100+/−5 nucleotides, in length.
  • In an embodiment, the linking domain is 20+/−10, 30+/−10, 40+/−10, 50+/−10, 60+/−10, 70+/−10, 80+/−10, 90+/−10, or 100+/−10 nucleotides, in length.
  • In an embodiment, the linking domain is 10 to 100, 10 to 90, 10 to 80, 10 to 70, 10 to 60, 10 to 50, 10 to 40, 10 to 30, 10 to 20 or 10 to 15 nucleotides in length.
  • In another embodiment, the linking domain is 20 to 100, 20 to 90, 20 to 80, 20 to 70, 20 to 60, 20 to 50, 20 to 40, 20 to 30, or 20 to 25 nucleotides in length.
  • In an embodiment, the linking domain is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16 17, 18, 19, or 20 nucleotides in length.
  • In and embodiment, the linking domain is a covalent bond.
  • In an embodiment, the linking domain comprises a duplexed region, typically adjacent to or within 1, 2, or 3 nucleotides of the 3′ end of the first complementarity domain and/or the 5-end of the second complementarity domain. In an embodiment, the duplexed region can be 20+/−10 base pairs in length. In an embodiment, the duplexed region can be 10+/−5, 15+/−5, 20+/−5, or 30+/−5 base pairs in length. In an embodiment, the duplexed region can be 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 base pairs in length.
  • Typically the sequences forming the duplexed region have exact complementarity with one another, though in an embodiment as many as 1, 2, 3, 4, 5, 6, 7 or 8 nucleotides are not complementary with the corresponding nucleotides.
  • In an embodiment, the linking domain nucleotides do not comprise modifications, e.g., modifications of the type provided in Section XI. However, in an embodiment, the linking domain comprises one or more modifications, e.g., modifications that it render it less susceptible to degradation or more bio-compatible, e.g., less immunogenic. By way of example, the backbone of the linking domain can be modified with a phosphorothioate, or other modification(s) from Section XI. In an embodiment a nucleotide of the linking domain can comprise a 2′ modification (e.g., a modification at the 2′ position on ribose), e.g., a 2-acetylation, e.g., a 2′ methylation, or other modification(s) from Section XI. In an embodiment, the linking domain can comprise as many as 1, 2, 3, 4, 5, 6, 7 or 8 modifications.
  • Modifications in a linking domain can be selected so as to not interfere with targeting efficacy, which can be evaluated by testing a candidate modification in the system described in Section VII. gRNAs having a candidate linking domain having a selected length, sequence, degree of complementarity, or degree of modification, can be evaluated a system described in Section VII. A candidate linking domain can be placed, either alone, or with one or more other candidate changes in a gRNA molecule/Cas9 molecule system known to be functional with a selected target and evaluated.
  • In an embodiment, the linking domain has at least 60, 70, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99% homology with, or differs by no more than 1, 2, 3, 4, 5, or 6 nucleotides from, a reference linking domain, e.g., a linking domain described herein.
  • The Proximal Domain
  • In an embodiment, the proximal domain is 6+/−2, 7+/−2, 8+/−2, 9+/−2, 10+/−2, 11+/−2, 12+/−2, 13+/−2, 14+/−2, 14+/−2, 16+/−2, 17+/−2, 18+/−2, 19+/−2, or 20+/−2 nucleotides in length.
  • In an embodiment, the proximal domain is 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25 or 26 nucleotides in length.
  • In an embodiment, the proximal domain is 5 to 20, 7, to 18, 9 to 16, or 10 to 14 nucleotides in length.
  • In an embodiment, the proximal domain nucleotides do not comprise modifications, e.g., modifications of the type provided in Section XI. However, in an embodiment, the proximal domain comprises one or more modifications, e.g., modifications that it render it less susceptible to degradation or more bio-compatible, e.g., less immunogenic. By way of example, the backbone of the proximal domain can be modified with a phosphorothioate, or other modification(s) from Section XI. In an embodiment a nucleotide of the proximal domain can comprise a 2′ modification (e.g., a modification at the 2′ position on ribose), e.g., a 2-acetylation, e.g., a 2′ methylation, or other modification(s) from Section XI.
  • In an embodiment, the proximal domain can comprise as many as 1, 2, 3, 4, 5, 6, 7 or 8 modifications. In an embodiment, the proximal domain comprises as many as 1, 2, 3, or 4 modifications within 5 nucleotides of its 5′ end, e.g., in a modular gRNA molecule. In an embodiment, the target domain comprises as many as 1, 2, 3, or 4 modifications within 5 nucleotides of its 3′ end, e.g., in a modular gRNA molecule.
  • In an embodiment, the proximal domain comprises modifications at two consecutive nucleotides, e.g., two consecutive nucleotides that are within 5 nucleotides of the 5′ end of the proximal domain, within 5 nucleotides of the 3′ end of the proximal domain, or more than 5 nucleotides away from one or both ends of the proximal domain. In an embodiment, no two consecutive nucleotides are modified within 5 nucleotides of the 5′ end of the proximal domain, within 5 nucleotides of the 3′ end of the proximal domain, or within a region that is more than 5 nucleotides away from one or both ends of the proximal domain. In an embodiment, no nucleotide is modified within 5 nucleotides of the 5′ end of the proximal domain, within 5 nucleotides of the 3′ end of the proximal domain, or within a region that is more than 5 nucleotides away from one or both ends of the proximal domain.
  • Modifications in the proximal domain can be selected so as to not interfere with gRNA molecule efficacy, which can be evaluated by testing a candidate modification in the system described in Section VII. gRNAs having a candidate proximal domain having a selected length, sequence, degree of complementarity, or degree of modification, can be evaluated in the system described at Section VII. The candidate proximal domain can be placed, either alone, or with one or more other candidate changes in a gRNA molecule/Cas9 molecule system known to be functional with a selected target and evaluated.
  • In an embodiment, the proximal domain has at least 60, 70, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99% homology with, or differs by no more than 1, 2, 3, 4, 5, or 6 nucleotides from, a reference proximal domain, e.g., a naturally occurring, e.g., an S. pyogenes, S. aureus or S. thermophilus, proximal domain, or a proximal domain described herein.
  • The Tail Domain
  • In an embodiment, the tail domain is 10+/−5, 20+/−5, 30+/−5, 40+/−5, 50+/−5, 60+/−5, 70+/−5, 80+/−5, 90+/−5, or 100+/−5 nucleotides, in length.
  • In an embodiment, the tail domain is 20+/−5 nucleotides in length.
  • In an embodiment, the tail domain is 20+/−10, 30+/−10, 40+/−10, 50+/−10, 60+/−10, 70+/−10, 80+/−10, 90+/−10, or 100+/−10 nucleotides, in length.
  • In an embodiment, the tail domain is 25+/−10 nucleotides in length.
  • In an embodiment, the tail domain is 10 to 100, 10 to 90, 10 to 80, 10 to 70, 10 to 60, 10 to 50, 10 to 40, 10 to 30, 10 to 20 or 10 to 15 nucleotides in length.
  • In another embodiment, the tail domain is 20 to 100, 20 to 90, 20 to 80, 20 to 70, 20 to 60, 20 to 50, 20 to 40, 20 to 30, or 20 to 25 nucleotides in length.
  • In an embodiment, the tail domain is 1 to 20, 1 to 15, 1 to 10, or 1 to 5 nucleotides in length.
  • In an embodiment, the tail domain nucleotides do not comprise modifications, e.g., modifications of the type provided in Section XI. However, in an embodiment, the tail domain comprises one or more modifications, e.g., modifications that it render it less susceptible to degradation or more bio-compatible, e.g., less immunogenic. By way of example, the backbone of the tail domain can be modified with a phosphorothioate, or other modification(s) from Section XI. In an embodiment a nucleotide of the tail domain can comprise a 2′ modification (e.g., a modification at the 2′ position on ribose), e.g., a 2-acetylation, e.g., a 2′ methylation, or other modification(s) from Section XI.
  • In an embodiment, the tail domain can have as many as 1, 2, 3, 4, 5, 6, 7 or 8 modifications. In an embodiment, the target domain comprises as many as 1, 2, 3, or 4 modifications within 5 nucleotides of its 5′ end. In an embodiment, the target domain comprises as many as 1, 2, 3, or 4 modifications within 5 nucleotides of its 3′ end.
  • In an embodiment, the tail domain comprises a tail duplex domain, which can form a tail duplexed region. In an embodiment, the tail duplexed region can be 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 base pairs in length. In an embodiment, a further single stranded domain, exists 3′ to the tail duplexed domain. In an embodiment, this domain is 3, 4, 5, 6, 7, 8, 9, or 10 nucleotides in length. In an embodiment it is 4 to 6 nucleotides in length.
  • In an embodiment, the tail domain has at least 60, 70, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99% homology with, or differs by no more than 1, 2, 3, 4, 5, or 6 nucleotides from, a reference tail domain, e.g., a naturally occurring, e.g., an S. pyogenes, S. aureus or S. thermophilus, tail domain, or a tail domain described herein.
  • In an embodiment, the proximal and tail domain, taken together comprise the following sequences:
  • (SEQ ID NO: XX)
    AAGGCUAGUCCGUUAUCAACUUGAAAAAGUGGCACCGAGUCGGUGCU,
    or
    (SEQ ID NO: XX)
    AAGGCUAGUCCGUUAUCAACUUGAAAAAGUGGCACCGAGUCGGUGGUGC,
    or
    (SEQ ID NO: XX)
    AAGGCUAGUCCGUUAUCAACUUGAAAAAGUGGCACCGAGUCGGUGCGGAU
    C,
    or
    (SEQ ID NO: XX)
    AAGGCUAGUCCGUUAUCAACUUGAAAAAGUG,
    or
    (SEQ ID NO: XX)
    AAGGCUAGUCCGUUAUCA,
    or
    (SEQ ID NO: XX)
    AAGGCUAGUCCG.
  • In an embodiment, the tail domain comprises the 3′ sequence UUUUUU, e.g., if a U6 promoter is used for transcription.
  • In an embodiment, the tail domain comprises the 3′ sequence UUUU, e.g., if an H1 promoter is used for transcription.
  • In an embodiment, tail domain comprises variable numbers of 3′ Us depending, e.g., on the termination signal of the pol-III promoter used.
  • In an embodiment, the tail domain comprises variable 3′ sequence derived from the DNA template if a T7 promoter is used.
  • In an embodiment, the tail domain comprises variable 3′ sequence derived from the DNA template, e.g., if in vitro transcription is used to generate the RNA molecule.
  • In an embodiment, the tail domain comprises variable 3′ sequence derived from the DNA template, e.g., if a pol-II promoter is used to drive transcription.
  • Modifications in the tail domain can be selected to not interfere with targeting efficacy, which can be evaluated by testing a candidate modification in the system described in Section VII. gRNAs having a candidate tail domain having a selected length, sequence, degree of complementarity, or degree of modification, can be evaluated in the system described in Section VII. The candidate tail domain can be placed, either alone, or with one or more other candidate changes in a gRNA molecule/Cas9 molecule system known to be functional with a selected target and evaluated.
  • In an embodiment, the tail domain comprises modifications at two consecutive nucleotides, e.g., two consecutive nucleotides that are within 5 nucleotides of the 5′ end of the tail domain, within 5 nucleotides of the 3′ end of the tail domain, or more than 5 nucleotides away from one or both ends of the tail domain. In an embodiment, no two consecutive nucleotides are modified within 5 nucleotides of the 5′ end of the tail domain, within 5 nucleotides of the 3′ end of the tail domain, or within a region that is more than 5 nucleotides away from one or both ends of the tail domain. In an embodiment, no nucleotide is modified within 5 nucleotides of the 5′ end of the tail domain, within 5 nucleotides of the 3′ end of the tail domain, or within a region that is more than 5 nucleotides away from one or both ends of the tail domain.
  • In an embodiment a gRNA has the following structure:
  • 5′ [targeting domain]-[first complementarity domain]-[linking domain]-[second complementarity domain]-[proximal domain]-[tail domain]-3′
  • wherein, the targeting domain comprises a core domain and optionally a secondary domain, and is 10 to 50 nucleotides in length;
  • the first complementarity domain is 5 to 25 nucleotides in length and, In an embodiment has at least 50, 60, 70, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99% homology with a reference first complementarity domain disclosed herein;
  • the linking domain is 1 to 5 nucleotides in length;
  • the second complementarity domain is 5 to 27 nucleotides in length and, in an embodiment has at least 50, 60, 70, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99% homology with a reference second complementarity domain disclosed herein;
  • the proximal domain is 5 to 20 nucleotides in length and, in an embodiment has at least 50, 60, 70, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99% homology with a reference proximal domain disclosed herein; and
  • the tail domain is absent or a nucleotide sequence is 1 to 50 nucleotides in length and, in an embodiment has at least 60, 70, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99% homology with a reference tail domain disclosed herein.
  • Exemplary Chimeric gRNAs
  • In an embodiment, a unimolecular, or chimeric, gRNA comprises, preferably from 5′ to 3′:
      • a targeting domain (which is complementary to a target nucleic acid);
      • a first complementarity domain, e.g., comprising 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, or 26 nucleotides;
      • a linking domain;
      • a second complementarity domain (which is complementary to the first complementarity domain);
      • a proximal domain; and
      • a tail domain,
      • wherein,
      • (a) the proximal and tail domain, when taken together, comprise
      • at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides;
      • (b) there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain; or
      • (c) there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • In an embodiment, the sequence from (a), (b), or (c), has at least 60, 70, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99% homology with the corresponding sequence of a naturally occurring gRNA, or with a gRNA described herein.
  • In an embodiment, the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • In an embodiment, there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • In an embodiment, there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • In an embodiment, the targeting domain comprises, has, or consists of, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25 or 26 nucleotides (e.g., 16, 17, 18, 19, 20, 21, 22, 23, 24, 25 or 26 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, or 26 nucleotides in length.
  • In an embodiment, the targeting domain comprises, has, or consists of, 16 nucleotides (e.g., 16 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 16 nucleotides in length.
  • In an embodiment, the targeting domain comprises, has, or consists of, 17 nucleotides (e.g., 17 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 17 nucleotides in length.
  • In an embodiment, the targeting domain comprises, has, or consists of, 18 nucleotides (e.g., 18 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 18 nucleotides in length.
  • In an embodiment, the targeting domain comprises, has, or consists of, 19 nucleotides (e.g., 19 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 19 nucleotides in length.
  • In an embodiment, the targeting domain comprises, has, or consists of, 20 nucleotides (e.g., 20 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 20 nucleotides in length.
  • In an embodiment, the targeting domain comprises, has, or consists of, 21 nucleotides (e.g., 21 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 21 nucleotides in length.
  • In an embodiment, the targeting domain comprises, has, or consists of, 22 nucleotides (e.g., 22 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 22 nucleotides in length.
  • In an embodiment, the targeting domain comprises, has, or consists of, 23 nucleotides (e.g., 23 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 23 nucleotides in length.
  • In an embodiment, the targeting domain comprises, has, or consists of, 24 nucleotides (e.g., 24 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 24 nucleotides in length.
  • In an embodiment, the targeting domain comprises, has, or consists of, 25 nucleotides (e.g., 25 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 25 nucleotides in length.
  • In an embodiment, the targeting domain comprises, has, or consists of, 26 nucleotides (e.g., 26 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 26 nucleotides in length.
  • In an embodiment, the targeting domain comprises, has, or consists of, 16 nucleotides (e.g., 16 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 16 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • In an embodiment, the targeting domain comprises, has, or consists of, 16 nucleotides (e.g., 16 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 16 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • In an embodiment, the targeting domain comprises, has, or consists of, 16 nucleotides (e.g., 16 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 16 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • In an embodiment, the targeting domain comprises, has, or consists of, 17 nucleotides (e.g., 17 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 17 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • In an embodiment, the targeting domain comprises, has, or consists of, 17 nucleotides (e.g., 17 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 17 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • In an embodiment, the targeting domain comprises, has, or consists of, 17 nucleotides (e.g., 17 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 17 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • In an embodiment, the targeting domain comprises, has, or consists of, 18 nucleotides (e.g., 18 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 18 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • In an embodiment, the targeting domain comprises, has, or consists of, 18 nucleotides (e.g., 18 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 18 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • In an embodiment, the targeting domain comprises, has, or consists of, 18 nucleotides (e.g., 18 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 18 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • In an embodiment, the targeting domain comprises, has, or consists of, 19 nucleotides (e.g., 19 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 19 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • In an embodiment, the targeting domain comprises, has, or consists of, 19 nucleotides (e.g., 19 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 19 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • In an embodiment, the targeting domain comprises, has, or consists of, 19 nucleotides (e.g., 19 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 19 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • In an embodiment, the targeting domain comprises, has, or consists of, 20 nucleotides (e.g., 20 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 20 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • In an embodiment, the targeting domain comprises, has, or consists of, 20 nucleotides (e.g., 20 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 20 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • In an embodiment, the targeting domain comprises, has, or consists of, 20 nucleotides (e.g., 20 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 20 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • In an embodiment, the targeting domain comprises, has, or consists of, 21 nucleotides (e.g., 21 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 21 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • In an embodiment, the targeting domain comprises, has, or consists of, 21 nucleotides (e.g., 21 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 21 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • In an embodiment, the targeting domain comprises, has, or consists of, 21 nucleotides (e.g., 21 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 21 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • In an embodiment, the targeting domain comprises, has, or consists of, 22 nucleotides (e.g., 22 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 22 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • In an embodiment, the targeting domain comprises, has, or consists of, 22 nucleotides (e.g., 22 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 22 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • In an embodiment, the targeting domain comprises, has, or consists of, 22 nucleotides (e.g., 22 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 22 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • In an embodiment, the targeting domain comprises, has, or consists of, 23 nucleotides (e.g., 23 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 23 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • In an embodiment, the targeting domain comprises, has, or consists of, 23 nucleotides (e.g., 23 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 23 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • In an embodiment, the targeting domain comprises, has, or consists of, 23 nucleotides (e.g., 23 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 23 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • In an embodiment, the targeting domain comprises, has, or consists of, 24 nucleotides (e.g., 24 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 24 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • In an embodiment, the targeting domain comprises, has, or consists of, 24 nucleotides (e.g., 24 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 24 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • In an embodiment, the targeting domain comprises, has, or consists of, 24 nucleotides (e.g., 24 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 24 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • In an embodiment, the targeting domain comprises, has, or consists of, 25 nucleotides (e.g., 25 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 25 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • In an embodiment, the targeting domain comprises, has, or consists of, 25 nucleotides (e.g., 25 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 25 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • In an embodiment, the targeting domain comprises, has, or consists of, 25 nucleotides (e.g., 25 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 25 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • In an embodiment, the targeting domain comprises, has, or consists of, 26 nucleotides (e.g., 26 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 26 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • In an embodiment, the targeting domain comprises, has, or consists of, 26 nucleotides (e.g., 26 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 26 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • In an embodiment, the targeting domain comprises, has, or consists of, 26 nucleotides (e.g., 26 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 26 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • In an embodiment, the unimolecular, or chimeric, gRNA molecule (comprising a targeting domain, a first complementary domain, a linking domain, a second complementary domain, a proximal domain and, optionally, a tail domain) comprises the following sequence in which the targeting domain is depicted as 20 Ns but could be any sequence and range in length from 16 to 26 nucleotides and in which the gRNA sequence is followed by 6 Us, which serve as a termination signal for the U6 promoter, but which could be either absent or fewer in number: NNNNNNNNNNNNNNNNNNNNGUUUUAGAGCUAGAAAUAGCAAGUUAAAAUAA GGCUAGUCCGUUAUCAACUUGAAAAAGUGGCACCGAGUCGGUGCUUUUUU (SEQ ID NO: XX). In an embodiment, the unimolecular, or chimeric, gRNA molecule is a S. pyogenes gRNA molecule.
  • In some embodiments, the unimolecular, or chimeric, gRNA molecule (comprising a targeting domain, a first complementary domain, a linking domain, a second complementary domain, a proximal domain and, optionally, a tail domain) comprises the following sequence in which the targeting domain is depicted as 20 Ns but could be any sequence and range in length from 16 to 26 nucleotides and in which the gRNA sequence is followed by 6 Us, which serve as a termination signal for the U6 promoter, but which could be either absent or fewer in number: NNNNNNNNNNNNNNNNNNNNGUUUUAGUACUCUGGAAACAGAAUCUACUAAA ACAAGGCAAAAUGCCGUGUUUAUCUCGUCAACUUGUUGGCGAGAUUUUUU (SEQ ID NO: XX). In an embodiment, the unimolecular, or chimeric, gRNA molecule is a S. aureus gRNA molecule.
  • Exemplary Modular gRNAs
  • In an embodiment, a modular gRNA comprises:
      • a first strand comprising, preferably from 5′ to 3′;
        • a targeting domain, e.g., comprising 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, or 26 nucleotides;
        • a first complementarity domain; and
        • a second strand, comprising, preferably from 5′ to 3′:
        • optionally a 5′ extension domain;
        • a second complementarity domain;
        • a proximal domain; and
        • a tail domain,
      • wherein:
      • (a) the proximal and tail domain, when taken together, comprise
      • at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides;
      • (b) there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain; or
      • (c) there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • In an embodiment, the sequence from (a), (b), or (c), has at least 60, 75, 80, 85, 90, 95, or 99% homology with the corresponding sequence of a naturally occurring gRNA, or with a gRNA described herein.
  • In an embodiment, the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • In an embodiment, there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • In an embodiment, there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • In an embodiment, the targeting domain comprises, has, or consists of, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25 or 26 nucleotides (e.g., 16, 17, 18, 19, 20, 21, 22, 23, 24, 25 or 26 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 16, 17, 18, 19, 20, 21, 22, 23, 24, 25 or 26 nucleotides in length.
  • In an embodiment, the targeting domain comprises, has, or consists of, 16 nucleotides (e.g., 16 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 16 nucleotides in length.
  • In an embodiment, the targeting domain comprises, has, or consists of, 17 nucleotides (e.g., 17 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 17 nucleotides in length.
  • In an embodiment, the targeting domain comprises, has, or consists of, 18 nucleotides (e.g., 18 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 18 nucleotides in length.
  • In an embodiment, the targeting domain comprises, has, or consists of, 19 nucleotides (e.g., 19 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 19 nucleotides in length.
  • In an embodiment, the targeting domain comprises, has, or consists of, 20 nucleotides (e.g., 20 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 20 nucleotides in length.
  • In an embodiment, the targeting domain comprises, has, or consists of, 21 nucleotides (e.g., 21 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 21 nucleotides in length.
  • In an embodiment, the targeting domain comprises, has, or consists of, 22 nucleotides (e.g., 22 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 22 nucleotides in length.
  • In an embodiment, the targeting domain comprises, has, or consists of, 23 nucleotides (e.g., 23 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 23 nucleotides in length.
  • In an embodiment, the targeting domain comprises, has, or consists of, 24 nucleotides (e.g., 24 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 24 nucleotides in length.
  • In an embodiment, the targeting domain comprises, has, or consists of, 25 nucleotides (e.g., 25 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 5 nucleotides in length.
  • In an embodiment, the targeting domain comprises, has, or consists of, 26 nucleotides (e.g., 26 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 26 nucleotides in length.
  • In an embodiment, the targeting domain comprises, has, or consists of, 16 nucleotides (e.g., 16 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 16 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • In an embodiment, the targeting domain comprises, has, or consists of, 16 nucleotides (e.g., 16 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 16 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • In an embodiment, the targeting domain comprises, has, or consists of, 16 nucleotides (e.g., 16 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 16 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • In an embodiment, the targeting domain has, or consists of, 17 nucleotides (e.g., 17 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 17 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • In an embodiment, the targeting domain has, or consists of, 17 nucleotides (e.g., 17 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 17 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • In an embodiment, the targeting domain has, or consists of, 17 nucleotides (e.g., 17 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 17 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • In an embodiment, the targeting domain has, or consists of, 18 nucleotides (e.g., 18 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 18 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • In an embodiment, the targeting domain has, or consists of, 18 nucleotides (e.g., 18 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 18 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • In an embodiment, the targeting domain has, or consists of, 18 nucleotides (e.g., 18 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 18 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • In an embodiment, the targeting domain comprises, has, or consists of, 19 nucleotides (e.g., 19 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 19 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • In an embodiment, the targeting domain comprises, has, or consists of, 19 nucleotides (e.g., 19 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 19 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • In an embodiment, the targeting domain comprises, has, or consists of, 19 nucleotides (e.g., 19 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 19 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • In an embodiment, the targeting domain comprises, has, or consists of, 20 nucleotides (e.g., 20 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 20 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • In an embodiment, the targeting domain comprises, has, or consists of, 20 nucleotides (e.g., 20 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 20 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • In an embodiment, the targeting domain comprises, has, or consists of, 20 nucleotides (e.g., 20 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 20 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • In an embodiment, the targeting domain comprises, has, or consists of, 21 nucleotides (e.g., 21 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 21 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • In an embodiment, the targeting domain comprises, has, or consists of, 21 nucleotides (e.g., 21 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 21 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • In an embodiment, the targeting domain comprises, has, or consists of, 21 nucleotides (e.g., 21 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 21 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • In an embodiment, the targeting domain comprises, has, or consists of, 22 nucleotides (e.g., 22 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 22 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • In an embodiment, the targeting domain comprises, has, or consists of, 22 nucleotides (e.g., 22 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 22 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • In an embodiment, the targeting domain comprises, has, or consists of, 22 nucleotides (e.g., 22 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 22 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • In an embodiment, the targeting domain comprises, has, or consists of, 23 nucleotides (e.g., 23 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 23 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • In an embodiment, the targeting domain comprises, has, or consists of, 23 nucleotides (e.g., 23 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 23 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • In an embodiment, the targeting domain comprises, has, or consists of, 23 nucleotides (e.g., 23 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 23 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • In an embodiment, the targeting domain comprises, has, or consists of, 24 nucleotides (e.g., 24 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 24 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • In an embodiment, the targeting domain comprises, has, or consists of, 24 nucleotides (e.g., 24 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 24 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • In an embodiment, the targeting domain comprises, has, or consists of, 24 nucleotides (e.g., 24 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 24 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • In an embodiment, the targeting domain comprises, has, or consists of, 25 nucleotides (e.g., 25 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 25 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • In an embodiment, the targeting domain comprises, has, or consists of, 25 nucleotides (e.g., 25 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 25 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • In an embodiment, the targeting domain comprises, has, or consists of, 25 nucleotides (e.g., 25 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 25 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • In an embodiment, the targeting domain comprises, has, or consists of, 26 nucleotides (e.g., 26 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 26 nucleotides in length; and the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides.
  • In an embodiment, the targeting domain comprises, has, or consists of, 26 nucleotides (e.g., 26 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 26 nucleotides in length; and there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain.
  • In an embodiment, the targeting domain comprises, has, or consists of, 26 nucleotides (e.g., 26 consecutive nucleotides) having complementarity with the target domain, e.g., the targeting domain is 26 nucleotides in length; and there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain.
  • In another aspect, methods and compositions discussed herein provide methods and compositions for gene editing by using a gRNA molecule which comprises a polyA tail. In one embodiment, a polyA tail of undefined length ranging from 1 to 1000 nucleotide is added enzymatically using a polymerase such as E. coli polyA polymerase (E-PAP). In one embodiment, the polyA tail of a specified length (e.g., 1, 5, 10, 20, 30, 40, 50, 60, 100, or 150 nucleotides) is encoded on a DNA template and transcribed with the gRNA via an RNA polymerase (e.g., T7 RNA polymerase). In one embodiment, a polyA tail of defined length (e.g., 1, 5, 10, 20, 30, 40, 50, 60, 100, or 150 nucleotides) is synthesized as a synthetic oligonucleotide and ligated on the 3′ end of the gRNA with either an RNA ligase or a DNA ligase with our without a splinted DNA oligonucleotide complementary to the guide RNA and the polyA oligonucleotide. In one embodiment, the entire gRNA including a defined length of polyA tail is made synthetically, in one or several pieces, and ligated together by either an RNA ligase or a DNA ligase with or without a splinted DNA oligonucleotide.
  • Additional exemplary gRNAs for use in the present invention are disclosed in International Application WO 2015/048577, the entire contents of which are expressly incorporated herein by reference
  • II. Methods for Designing gRNAs
  • Methods for designing gRNAs are described herein, including methods for selecting, designing and validating target domains. Exemplary targeting domains are also provided herein. Targeting Domains discussed herein can be incorporated into the gRNAs described herein.
  • Methods for selection and validation of target sequences as well as off-target analyses are described, e.g., in Mali et al., 2013 SCIENCE 339(6121): 823-826; Hsu et al. NAT BIOTECHNOL, 31(9): 827-32; Fu et al., 2014 NAT BIOTECHNOL, doi: 10.1038/nbt.2808. PubMed PMID: 24463574; Heigwer et al., 2014 NAT METHODS 11(2):122-3. doi: 10.1038/nmeth.2812. PubMed PMID: 24481216; Bae et al., 2014 BIOINFORMATICS PubMed PMID: 24463181; Xiao A et al., 2014 BIOINFORMATICS PubMed PMID: 24389662. Additional considerations for designing gRNAs are discussed in the section entitled “gRNA Design” in PCT Application WO 2015/048577, the entire contents of which are expressly incorporated herein by reference.
  • For example, a software tool can be used to optimize the choice of gRNA within a user's target sequence, e.g., to minimize total off-target activity across the genome. Off target activity may be other than cleavage. For each possible gRNA choice using S. pyogenes Cas9, the tool can identify all off-target sequences (preceding either NAG or NGG PAMs) across the genome that contain up to certain number (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) of mismatched base-pairs. The cleavage efficiency at each off-target sequence can be predicted, e.g., using an experimentally-derived weighting scheme. Each possible gRNA is then ranked according to its total predicted off-target cleavage; the top-ranked gRNAs represent those that are likely to have the greatest on-target and the least off-target cleavage. Other functions, e.g., automated reagent design for CRISPR construction, primer design for the on-target Surveyor assay, and primer design for high-throughput detection and quantification of off-target cleavage via next-gen sequencing, can also be included in the tool. Candidate gRNA molecules can be evaluated by art-known methods or as described in Section VII herein.
  • The targeting domains discussed herein can be incorporated into the gRNAs described herein.
  • As an example, three strategies are utilized to identify gRNAs for use with S. pyogenes, S. aureus and N. meningitidis Cas9 enzymes.
  • Guide RNAs (gRNAs) for use with S. pyogenes, S. aureus and N. meningitidis Cas9 molecules are identified using a DNA sequence searching algorithm. Guide RNA design is carried out using a custom guide RNA design software based on the public tool cas-offinder (reference:Cas-OFFinder: a fast and versatile algorithm that searches for potential off-target sites of Cas9 RNA-guided endonucleases, Bioinformatics. 2014 Feb. 17. Bae S, Park J, Kim J S. PMID:24463181). Said custom guide RNA design software scores guides after calculating their genome-wide off-target propensity. Typically matches ranging from perfect matches to 7 mismatches are considered for guides ranging in length from 17 to 24. Once the off-target sites are computationally determined, an aggregate score is calculated for each guide and summarized in a tabular output using a web-interface. In addition to identifying potential gRNA sites adjacent to PAM sequences, the software also identifies all PAM adjacent sequences that differ by 1, 2, 3 or more nucleotides from the selected gRNA sites. Genomic DNA sequence for each gene was obtained from the UCSC Genome browser and sequences were screened for repeat elements using the publically available RepeatMasker program. RepeatMasker searches input DNA sequences for repeated elements and regions of low complexity. The output is a detailed annotation of the repeats present in a given query sequence.
  • Following identification, gRNAs are ranked into tiers based on their distance to the target site, their orthogonality and presence of a 5′ G (based on identification of close matches in the human genome containing a relevant PAM (e.g., in the case of S. pyogenes, a NGG PAM, in the case of S. aureus, a NNGRRT or NNGRRV PAM, and in the case of N. meningitidis, a NNNNGATT or NNNNGCTT PAM). Orthogonality refers to the number of sequences in the human genome that contain a minimum number of mismatches to the target sequence. A “high level of orthogonality” or “good orthogonality” may, for example, refer to 20-mer gRNAs that have no identical sequences in the human genome besides the intended target, nor any sequences that contain one or two mismatches in the target sequence. Targeting domains with good orthogonality are selected to minimize off-target DNA cleavage.
  • gRNAs are identified for both single-gRNA nuclease cleavage and for a dual-gRNA paired “nickase” strategy. Criteria for selecting gRNAs and the determination for which gRNAs can be used for the dual-gRNA paired “nickase” strategy is based on two considerations:
      • 1. gRNA pairs should be oriented on the DNA such that PAMs are facing out and cutting with the D10A Cas9 nickase will result in 5′ overhangs.
      • 2. An assumption that cleaving with dual nickase pairs will result in deletion of the entire intervening sequence at a reasonable frequency. However, cleaving with dual nickase pairs can also result in indel mutations at the site of only one of the gRNAs. Candidate pair members can be tested for how efficiently they remove the entire sequence versus causing indel mutations at the site of one gRNA.
  • The targeting domains discussed herein can be incorporated into the gRNAs described herein.
  • In an embodiment, two or more (e.g., three or four) gRNA molecules are used with one Cas9 fusion molecule. In another embodiment, when two or more (e.g., three or four) gRNAs are used with two or more Cas9 fusion molecules, at least one Cas9 molecule is from a different species than the other Cas9 molecule(s). For example, when two gRNA molecules are used with two Cas9 fusion molecules, one Cas9 molecule can be from one species and the other Cas9 molecule can be from a different species. Both Cas9 species are used to generate a single or double-strand break, as desired.
  • Any of the targeting domains in the tables described herein can be used with a Cas9 nickase molecule to generate a single strand break.
  • Any of the targeting domains in the tables described herein can be used with a Cas9 nuclease molecule to generate a double strand break.
  • When two gRNAs designed for use to target two Cas9 molecules, one Cas9 can be one species, the second Cas9 can be from a different species. Both Cas9 species are used to generate a single or double-strand break, as desired.
  • It is contemplated herein that any upstream gRNA described herein may be paired with any downstream gRNA described herein. When an upstream gRNA designed for use with one species of Cas9 is paired with a downstream gRNA designed for use from a different species of Cas9, both Cas9 species are used to generate a single or double-strand break, as desired.
  • III. Cas9 Molecules
  • Cas9 molecules of a variety of species can be used in the methods and compositions described herein. While the S. pyogenes, S. aureus, and S. thermophilus Cas9 molecules are the subject of much of the disclosure herein, Cas9 molecules, derived from, or based on the Cas9 proteins of other species listed herein can be used as well. In other words, while the much of the description herein uses S. pyogenes and S. thermophilus Cas9 molecules, Cas9 molecules from the other species can replace them, e.g., Staphylococcus aureus and Neisseria meningitidis Cas9 molecules. Additional Cas9 species include: Acidovorax avenae, Actinobacillus pleuropneumoniae, Actinobacillus succinogenes, Actinobacillus suis, Actinomyces sp., cycliphilus denitrificans, Aminomonas paucivorans, Bacillus cereus, Bacillus smithii, Bacillus thuringiensis, Bacteroides sp., Blastopirellula marina, Bradyrhizobium sp., Brevibacillus laterosporus, Campylobacter coli, Campylobacter jejuni, Campylobacter lari, Candidatus Puniceispirillum, Clostridium cellulolyticum, Clostridium perfringens, Corynebacterium accolens, Corynebacterium diphtheria, Corynebacterium matruchotii, Dinoroseobacter shibae, Eubacterium dolichum, gamma proteobacterium, Gluconacetobacter diazotrophicus, Haemophilus parainfluenzae, Haemophilus sputorum, Helicobacter canadensis, Helicobacter cinaedi, Helicobacter mustelae, Ilyobacter polytropus, Kingella kingae, Lactobacillus crispatus, Listeria ivanovii, Listeria monocytogenes, Listeriaceae bacterium, Methylocystis sp., Methylosinus trichosporium, Mobiluncus mulieris, Neisseria bacilliformis, Neisseria cinerea, Neisseria flavescens, Neisseria lactamica, Neisseria sp., Neisseria wadsworthii, Nitrosomonas sp., Parvibaculum lavamentivorans, Pasteurella multocida, Phascolarctobacterium succinatutens, Ralstonia syzygii, Rhodopseudomonas palustris, Rhodovulum sp., Simonsiella muelleri, Sphingomonas sp., Sporolactobacillus vineae, Staphylococcus lugdunensis, Streptococcus sp., Subdoligranulum sp., Tistrella mobilis, Treponema sp., or Verminephrobacter eiseniae.
  • A Cas9 molecule, or Cas9 polypeptide, as the term is used herein, refers to a molecule or a polypeptide that can interact with a guide RNA (gRNA) molecule) and, in concert with the gRNA molecule, localizes to a site which comprises a target domain, and in some embodiments, a PAM sequence. Cas9 molecule and Cas9 polypeptide, as those terms are used herein, refer to naturally occurring Cas9 molecules and to engineered, altered, or modified Cas9 molecules or Cas9 polypeptides that differ, e.g., by at least one amino acid residue, from a reference sequence, e.g., the most similar naturally occurring Cas9 molecule or a sequence of Table 100.
  • Cas9 Domains
  • Crystal structures have been determined for two different naturally occurring bacterial Cas9 molecules (Jinek et al., SCIENCE, 343(6176):1247997, 2014) and for S. pyogenes Cas9 with a guide RNA (e.g., a synthetic fusion of crRNA and tracrRNA) (Nishimasu et al., CELL, 156:935-949, 2014; and Anders et al., NATURE, 2014, doi: 10.1038/nature13579).
  • A naturally occurring Cas9 molecule comprises two lobes: a recognition (REC) lobe and a nuclease (NUC) lobe; each of which further comprise domains described herein. The REC lobe comprises the arginine-rich bridge helix (BH), the REC1 domain, and the REC2 domain. The REC lobe does not share structural similarity with other known proteins, indicating that it is a Cas9-specific functional domain. The BH domain is a long ac helix and arginine rich region and comprises amino acids 60-93 of the sequence of S. pyogenes Cas9. The REC1 domain is important for recognition of the repeat:anti-repeat duplex, e.g., of a gRNA or a tracrRNA, and is therefore critical for Cas9 activity by recognizing the target sequence. The REC1 domain comprises two REC1 motifs at amino acids 94 to 179 and 308 to 717 of the sequence of S. pyogenes Cas9. These two REC1 domains, though separated by the REC2 domain in the linear primary structure, assemble in the tertiary structure to form the REC1 domain. The REC2 domain, or parts thereof, may also play a role in the recognition of the repeat:anti-repeat duplex. The REC2 domain comprises amino acids 180-307 of the sequence of S. pyogenes Cas9.
  • The NUC lobe comprises the RuvC domain, the HNH domain, and the PAM-interacting (PI) domain. The RuvC domain shares structural similarity to retroviral integrase superfamily members and cleaves a single strand, e.g., the non-complementary strand of the target nucleic acid molecule. The RuvC domain is assembled from the three split RuvC motifs (RuvC I, RuvCII, and RuvCIII, which are often commonly referred to in the art as RuvCI domain, or N-terminal RuvC domain, RuvCII domain, and RuvCIII domain) at amino acids 1-59, 718-769, and 909-1098, respectively, of the sequence of S. pyogenes Cas9. Similar to the REC1 domain, the three RuvC motifs are linearly separated by other domains in the primary structure, however in the tertiary structure, the three RuvC motifs assemble and form the RuvC domain. The HNH domain shares structural similarity with HNH endonucleases, and cleaves a single strand, e.g., the complementary strand of the target nucleic acid molecule. The HNH domain lies between the RuvC II-III motifs and comprises amino acids 775-908 of the sequence of S. pyogenes Cas9. The PI domain interacts with the PAM of the target nucleic acid molecule, and comprises amino acids 1099-1368 of the sequence of S. pyogenes Cas9.
  • A RuvC-Like Domain and an HNH-Like Domain
  • In an embodiment, a Cas9 molecule or Cas9 polypeptide comprises an HNH-like domain and a RuvC-like domain. In an embodiment, cleavage activity is dependent on a RuvC-like domain and an HNH-like domain. A Cas9 molecule or Cas9 polypeptide, e.g., an eaCas9 molecule or eaCas9 polypeptide, can comprise one or more of the following domains: a RuvC-like domain and an HNH-like domain. In an embodiment, a Cas9 molecule or Cas9 polypeptide is an eaCas9 molecule or eaCas9 polypeptide and the eaCas9 molecule or eaCas9 polypeptide comprises a RuvC-like domain, e.g., a RuvC-like domain described below, and/or an HNH-like domain, e.g., an HNH-like domain described below.
  • RuvC-Like Domains
  • In an embodiment, a RuvC-like domain cleaves, a single strand, e.g., the non-complementary strand of the target nucleic acid molecule. The Cas9 molecule or Cas9 polypeptide can include more than one RuvC-like domain (e.g., one, two, three or more RuvC-like domains). In an embodiment, a RuvC-like domain is at least 5, 6, 7, 8 amino acids in length but not more than 20, 19, 18, 17, 16 or 15 amino acids in length. In an embodiment, the Cas9 molecule or Cas9 polypeptide comprises an N-terminal RuvC-like domain of about 10 to 20 amino acids, e.g., about 15 amino acids in length.
  • N-Terminal RuvC-Like Domains
  • Some naturally occurring Cas9 molecules comprise more than one RuvC-like domain with cleavage being dependent on the N-terminal RuvC-like domain. Accordingly, Cas9 molecules or Cas9 polypeptide can comprise an N-terminal RuvC-like domain. Exemplary N-terminal RuvC-like domains are described below.
  • In an embodiment, an eaCas9 molecule or eaCas9 polypeptide comprises an N-terminal RuvC-like domain comprising an amino acid sequence of formula I:
  • (SEQ ID NO: EE1)
    D-X1-G-X2-X3-X4-X5-G-X6-X7-X8-X9,
  • wherein,
  • X1 is selected from I, V, M, L and T (e.g., selected from I, V, and L);
  • X2 is selected from T, I, V, S, N, Y, E and L (e.g., selected from T, V, and I);
  • X3 is selected from N, S, G, A, D, T, R, M and F (e.g., A or N);
  • X4 is selected from S, Y, N and F (e.g., S);
  • X5 is selected from V, I, L, C, T and F (e.g., selected from V, I and L);
  • X6 is selected from W, F, V, Y, S and L (e.g., W);
  • X7 is selected from A, S, C, V and G (e.g., selected from A and S);
  • X8 is selected from V, I, L, A, M and H (e.g., selected from V, I, M and L); and
  • X9 is selected from any amino acid or is absent (e.g., selected from T, V, I, L, A, F, S, A, Y, M and R, or, e.g., selected from T, V, I, L and A).
  • In an embodiment, the N-terminal RuvC-like domain differs from a sequence of SEQ ID NO: EE1, by as many as 1 but no more than 2, 3, 4, or 5 residues.
  • In embodiment, the N-terminal RuvC-like domain is cleavage competent.
  • In embodiment, the N-terminal RuvC-like domain is cleavage incompetent.
  • In an embodiment, a eaCas9 molecule or eaCas9 polypeptide comprises an N-terminal RuvC-like domain comprising an amino acid sequence of formula II:
  • (SEQ ID NO: EE2)
    D-X1-G-X2-X3-S-X5-G-X6-X7-X8-X9,,
  • wherein
  • X1 is selected from I, V, M, L and T (e.g., selected from I, V, and L);
  • X2 is selected from T, I, V, S, N, Y, E and L (e.g., selected from T, V, and I);
  • X3 is selected from N, S, G, A, D, T, R, M and F (e.g., A or N);
  • X5 is selected from V, I, L, C, T and F (e.g., selected from V, I and L);
  • X6 is selected from W, F, V, Y, S and L (e.g., W);
  • X7 is selected from A, S, C, V and G (e.g., selected from A and S);
  • X8 is selected from V, I, L, A, M and H (e.g., selected from V, I, M and L); and
  • X9 is selected from any amino acid or is absent (e.g., selected from T, V, I, L, A, F, S, A, Y, M and R or selected from e.g., T, V, I, L and A).
  • In an embodiment, the N-terminal RuvC-like domain differs from a sequence of SEQ ID NO: EE2 by as many as 1 but no more than 2, 3, 4, or 5 residues.
  • In an embodiment, the N-terminal RuvC-like domain comprises an amino acid sequence of formula III:
  • (SEQ ID NO: AA1)
    D-I-G-X2-X3-S-V-G-W-A-X8-X9,
  • wherein
  • X2 is selected from T, I, V, S, N, Y, E and L (e.g., selected from T, V, and I);
  • X3 is selected from N, S, G, A, D, T, R, M and F (e.g., A or N);
  • X8 is selected from V, I, L, A, M and H (e.g., selected from V, I, M and L); and
  • X9 is selected from any amino acid or is absent (e.g., selected from T, V, I, L, A, F, S, A, Y, M and R or selected from e.g., T, V, I, L and A).
  • In an embodiment, the N-terminal RuvC-like domain differs from a sequence of SEQ ID NO:AA1 by as many as 1 but no more than, 2, 3, 4, or 5 residues.
  • In an embodiment, the N-terminal RuvC-like domain comprises an amino acid sequence of formula III:
  • (SEQ ID NO: AA2)
    D-I-G-T-N-S-V-G-W-A-V-X,
  • wherein
  • X is a non-polar alkyl amino acid or a hydroxyl amino acid, e.g., X is selected from V, I, L and T.
  • In an embodiment, the N-terminal RuvC-like domain differs from a sequence of SEQ ID NO: AA2 by as many as 1 but no more than, 2, 3, 4, or 5 residues.
  • In an embodiment, the N-terminal RuvC-like domain differs from a sequence of an N-terminal RuvC like domain disclosed herein, as many as 1 but no more than 2, 3, 4, or 5 residues.
  • Additional RuvC-Like Domains
  • In addition to the N-terminal RuvC-like domain, the Cas9 molecule or Cas9 polypeptide, e.g., an eaCas9 molecule or eaCas9 polypeptide, can comprise one or more additional RuvC-like domains. In an embodiment, the Cas9 molecule or Cas9 polypeptide can comprise two additional RuvC-like domains. Preferably, the additional RuvC-like domain is at least 5 amino acids in length and, e.g., less than 15 amino acids in length, e.g., 5 to 10 amino acids in length, e.g., 8 amino acids in length.
  • An additional RuvC-like domain can comprise an amino acid sequence:
  • (SEQ ID NO: AA3)
    I-X1-X2-E-X3-A-R-E,

    wherein
  • X1 is V or H,
  • X2 is I, L or V (e.g., I or V); and
  • X3 is M or T.
  • In an embodiment, the additional RuvC-like domain comprises the amino acid sequence:
  • (SEQ ID NO: AA4)
    I-V-X2-E-M-A-R-E,

    wherein
  • X2 is I, L or V (e.g., I or V).
  • An additional RuvC-like domain can comprise an amino acid sequence:
  • (SEQ ID NO: AA5)
    H-H-A-X1-D-A-X2-X3,

    wherein
  • X1 is H or L;
  • X2 is R or V; and
  • X3 is E or V.
  • In an embodiment, the additional RuvC-like domain comprises the amino acid sequence: H-H-A-H-D-A-Y-L (SEQ ID NO:AA6).
  • In an embodiment, the additional RuvC-like domain differs from a sequence of SEQ ID NO: AA3, AA4, AA5, AA6 by as many as 1 but no more than 2, 3, 4, or 5 residues.
  • In some embodiments, the sequence flanking the N-terminal RuvC-like domain is a sequences of formula V:
  • (SEQ ID NO: AA7)
    K-X1′-Y-X2′-X3′-X4′-Z-T-D-X9′-Y,.
  • wherein
  • X1′ is selected from K and P,
  • X2′ is selected from V, L, I, and F (e.g., V, I and L);
  • X3′ is selected from G, A and S (e.g., G),
  • X4′ is selected from L, I, V and F (e.g., L);
  • X9′ is selected from D, E, N and Q; and
  • Z is an N-terminal RuvC-like domain, e.g., as described above.
  • HNH-Like Domains
  • In an embodiment, an HNH-like domain cleaves a single stranded complementary domain, e.g., a complementary strand of a double stranded nucleic acid molecule. In an embodiment, an HNH-like domain is at least 15, 20, 25 amino acids in length but not more than 40, 35 or 30 amino acids in length, e.g., 20 to 35 amino acids in length, e.g., 25 to 30 amino acids in length. Exemplary HNH-like domains are described below.
  • In an embodiment, an eaCas9 molecule or eaCas9 polypeptide comprises an HNH-like domain having an amino acid sequence of formula VI:
  • (SEQ ID NO: AA8)
    X1-X2-X3-H-X4-X5-P-X6-X7-X8-X9-X10-X11-X12-X13-
    X14-X15-N-X16-X17-X18-X19-X20-X21-X22-X23-N.

    wherein
  • X1 is selected from D, E, Q and N (e.g., D and E);
  • X2 is selected from L, I, R, Q, V, M and K;
  • X3 is selected from D and E;
  • X4 is selected from I, V, T, A and L (e.g., A, I and V);
  • X5 is selected from V, Y, I, L, F and W (e.g., V, I and L);
  • X6 is selected from Q, H, R, K, Y, I, L, F and W;
  • X7 is selected from S, A, D, T and K (e.g., S and A);
  • X8 is selected from F, L, V, K, Y, M, I, R, A, E, D and Q (e.g., F);
  • X9 is selected from L, R, T, I, V, S, C, Y, K, F and G;
  • X10 is selected from K, Q, Y, T, F, L, W, M, A, E, G, and S;
  • X11 is selected from D, S, N, R, L and T (e.g., D);
  • X12 is selected from D, N and S;
  • X13 is selected from S, A, T, G and R (e.g., S);
  • X14 is selected from I, L, F, S, R, Y, Q, W, D, K and H (e.g., I, L and F);
  • X15 is selected from D, S, I, N, E, A, H, F, L, Q, M, G, Y and V;
  • X16 is selected from K, L, R, M, T and F (e.g., L, R and K);
  • X17 is selected from V, L, I, A and T;
  • X18 is selected from L, I, V and A (e.g., L and I);
  • X19 is selected from T, V, C, E, S and A (e.g., T and V);
  • X20 is selected from R, F, T, W, E, L, N, C, K, V, S, Q, I, Y, H and A;
  • X21 is selected from S, P, R, K, N, A, H, Q, G and L;
  • X22 is selected from D, G, T, N, S, K, A, I, E, L, Q, R and Y; and
  • X23 is selected from K, V, A, E, Y, I, C, L, S, T, G, K, M, D and F.
  • In an embodiment, a HNH-like domain differs from a sequence of SEQ ID NO: AA8 by at least one but no more than, 2, 3, 4, or 5 residues.
  • In an embodiment, the HNH-like domain is cleavage competent.
  • In an embodiment, the HNH-like domain is cleavage incompetent.
  • In an embodiment, an eaCas9 molecule or eaCas9 polypeptide comprises an HNH-like domain comprising an amino acid sequence of formula VII:
  • (SEQ ID NO: AA9)
    X1-X2-X3-H-X4-X5-P-X6-S-X8-X9-X10-D-D-S-X14-
    X15-N-K-V-L-X19-X20-X21-X22-X23-N,
  • wherein
  • X1 is selected from D and E;
  • X2 is selected from L, I, R, Q, V, M and K;
  • X3 is selected from D and E;
  • X4 is selected from I, V, T, A and L (e.g., A, I and V);
  • X5 is selected from V, Y, I, L, F and W (e.g., V, I and L);
  • X6 is selected from Q, H, R, K, Y, I, L, F and W;
  • X8 is selected from F, L, V, K, Y, M, I, R, A, E, D and Q (e.g., F);
  • X9 is selected from L, R, T, I, V, S, C, Y, K, F and G;
  • X10 is selected from K, Q, Y, T, F, L, W, M, A, E, G, and S;
  • X14 is selected from I, L, F, S, R, Y, Q, W, D, K and H (e.g., I, L and F);
  • X15 is selected from D, S, I, N, E, A, H, F, L, Q, M, G, Y and V;
  • X19 is selected from T, V, C, E, S and A (e.g., T and V);
  • X20 is selected from R, F, T, W, E, L, N, C, K, V, S, Q, I, Y, H and A;
  • X21 is selected from S, P, R, K, N, A, H, Q, G and L;
  • X22 is selected from D, G, T, N, S, K, A, I, E, L, Q, R and Y; and
  • X23 is selected from K, V, A, E, Y, I, C, L, S, T, G, K, M, D and F.
  • In an embodiment, the HNH-like domain differs from a sequence of SEQ ID NO: AA9 by 1, 2, 3, 4, or 5 residues.
  • In an embodiment, an eaCas9 molecule or eaCas9 polypeptide comprises an HNH-like domain comprising an amino acid sequence of formula VII:
  • (SEQ ID NO: AA10)
    X1-V-X3-H-I-V-P-X6-S-X8-X9-X10-D-D-S-X14-
    X15-N-K-V-L-T-X20-X21-X22-X23-N,
  • wherein
  • X1 is selected from D and E;
  • X3 is selected from D and E;
  • X6 is selected from Q, H, R, K, Y, I, L and W;
  • X8 is selected from F, L, V, K, Y, M, I, R, A, E, D and Q (e.g., F);
  • X9 is selected from L, R, T, I, V, S, C, Y, K, F and G;
  • X10 is selected from K, Q, Y, T, F, L, W, M, A, E, G, and S;
  • X14 is selected from I, L, F, S, R, Y, Q, W, D, K and H (e.g., I, L and F);
  • X15 is selected from D, S, I, N, E, A, H, F, L, Q, M, G, Y and V;
  • X20 is selected from R, F, T, W, E, L, N, C, K, V, S, Q, I, Y, H and A;
  • X21 is selected from S, P, R, K, N, A, H, Q, G and L;
  • X22 is selected from D, G, T, N, S, K, A, I, E, L, Q, R and Y; and
  • X23 is selected from K, V, A, E, Y, I, C, L, S, T, G, K, M, D and F.
  • In an embodiment, the HNH-like domain differs from a sequence of SEQ ID NO: AA10 by 1, 2, 3, 4, or 5 residues.
  • In an embodiment, an eaCas9 molecule or eaCas9 polypeptide comprises an HNH-like domain having an amino acid sequence of formula VIII:
  • (SEQ ID NO: AA11)
    D-X2-D-H-I-X5-P-Q-X7-F-X9-X10-D-X12-S-I-D-
    N-X16-V-L-X19-X20-S-X22-X23-N,
  • wherein
  • X2 is selected from I and V;
  • X5 is selected from I and V;
  • X7 is selected from A and S;
  • X9 is selected from I and L;
  • X10 is selected from K and T;
  • X12 is selected from D and N;
  • X16 is selected from R, K and L; X19 is selected from T and V;
  • X20 is selected from S and R;
  • X22 is selected from K, D and A; and
  • X23 is selected from E, K, G and N (e.g., the eaCas9 molecule or eaCas9 polypeptide can comprise an HNH-like domain as described herein).
  • In an embodiment, the HNH-like domain differs from a sequence of SEQ ID NO: AA11 by as many as 1 but no more than 2, 3, 4, or 5 residues.
  • In an embodiment, an eaCas9 molecule or eaCas9 polypeptide comprises the amino acid sequence of formula IX:
  • (SEQ ID NO: AA12)
    L-Y-Y-L-Q-N-G-X1′-D-M-Y-X2′-X3′-X4′-X5′-L-
    D-I-X6′-X7′-L-S-X8′-Y-Z-N-R-X9′-K-X10′-D-X11′-V-P,
  • wherein
  • X1′ is selected from K and R;
  • X2′ is selected from V and T;
  • X3′ is selected from G and D;
  • X4′ is selected from E, Q and D;
  • X5′ is selected from E and D;
  • X6′ is selected from D, N and H;
  • X7′ is selected from Y, R and N;
  • X8′ is selected from Q, D and N; X9′ is selected from G and E;
  • X10′ is selected from S and G;
  • X11′ is selected from D and N; and
  • Z is an HNH-like domain, e.g., as described above.
  • In an embodiment, the eaCas9 molecule or eaCas9 polypeptide comprises an amino acid sequence that differs from a sequence of SEQ ID NO: AA12 by as many as 1 but no more than 2, 3, 4, or 5 residues.
  • In an embodiment, the HNH-like domain differs from a sequence of an HNH-like domain disclosed herein, by as many as 1 but no more than 2, 3, 4, or 5 residues.
  • In an embodiment, the HNH-like domain differs from a sequence of an HNH-like domain disclosed herein, by as many as 1 but no more than 2, 3, 4, or 5 residues.
  • Cas9 Activities
  • Nuclease and Helicase Activities
  • In an embodiment, the Cas9 molecule or Cas9 polypeptide is capable of cleaving a target nucleic acid molecule. Typically wild type Cas9 molecules cleave both strands of a target nucleic acid molecule. Cas9 molecules and Cas9 polypeptides can be engineered to alter nuclease cleavage (or other properties), e.g., to provide a Cas9 molecule or Cas9 polypeptide which is a nickase, or which lacks the ability to cleave target nucleic acid. A Cas9 molecule or Cas9 polypeptide that is capable of cleaving a target nucleic acid molecule is referred to herein as an eaCas9 (an enzymatically active Cas9) molecule or eaCas9 polypeptide.
  • In an embodiment, an eaCas9 molecule or eaCas9 polypeptide comprises one or more of the following enzymatic activities:
  • a nickase activity, i.e., the ability to cleave a single strand, e.g., the non-complementary strand or the complementary strand, of a nucleic acid molecule;
  • a double stranded nuclease activity, i.e., the ability to cleave both strands of a double stranded nucleic acid and create a double stranded break, which in an embodiment is the presence of two nickase activities;
  • an endonuclease activity;
  • an exonuclease activity; and
  • a helicase activity, i.e., the ability to unwind the helical structure of a double stranded nucleic acid.
  • In an embodiment, an enzymatically active or an eaCas9 molecule or eaCas9 polypeptide cleaves both DNA strands and results in a double stranded break. In an embodiment, an eaCas9 molecule or eaCas9 polypeptide cleaves only one strand, e.g., the strand to which the gRNA hybridizes to, or the strand complementary to the strand the gRNA hybridizes with. In an embodiment, an eaCas9 molecule or eaCas9 polypeptide comprises cleavage activity associated with an HNH domain. In an embodiment, an eaCas9 molecule or eaCas9 polypeptide comprises cleavage activity associated with a RuvC domain. In an embodiment, an eaCas9 molecule or eaCas9 polypeptide comprises cleavage activity associated with an HNH domain and cleavage activity associated with a RuvC domain. In an embodiment, an eaCas9 molecule or eaCas9 polypeptide comprises an active, or cleavage competent, HNH domain and an inactive, or cleavage incompetent, RuvC domain. In an embodiment, an eaCas9 molecule or eaCas9 polypeptide comprises an inactive, or cleavage incompetent, HNH domain and an active, or cleavage competent, RuvC domain.
  • Some Cas9 molecules or Cas9 polypeptides have the ability to interact with a gRNA molecule, and in conjunction with the gRNA molecule localize to a core target domain, but are incapable of cleaving the target nucleic acid, or incapable of cleaving at efficient rates. Cas9 molecules having no, or no substantial, cleavage activity are referred to herein as an eiCas9 molecule or eiCas9 polypeptide. For example, an eiCas9 molecule or eiCas9 polypeptide can lack cleavage activity or have substantially less, e.g., less than 20, 10, 5, 1 or 0.1% of the cleavage activity of a reference Cas9 molecule or eiCas9 polypeptide, as measured by an assay described herein.
  • Targeting and PAMs
  • A Cas9 molecule or Cas9 polypeptide, is a polypeptide that can interact with a guide RNA (gRNA) molecule and, in concert with the gRNA molecule, localizes to a site which comprises a target domain, and in an embodiment, a PAM sequence.
  • In an embodiment, the ability of an eaCas9 molecule or eaCas9 polypeptide to interact with and cleave a target nucleic acid is PAM sequence dependent. A PAM sequence is a sequence in the target nucleic acid. In an embodiment, cleavage of the target nucleic acid occurs upstream from the PAM sequence. EaCas9 molecules from different bacterial species can recognize different sequence motifs (e.g., PAM sequences). In an embodiment, an eaCas9 molecule of S. pyogenes recognizes the sequence motif NGG and directs cleavage of a target nucleic acid sequence 1 to 10, e.g., 3 to 5, base pairs upstream from that sequence. See, e.g., Mali et al., SCIENCE (2013) 339(6121): 823-826. In an embodiment, an eaCas9 molecule of S. thermophilus recognizes the sequence motif NGGNG (SEQ ID NO.: BB1) and/or NNAGAAW (W=A or T) (SEQ ID NO.: BB2) and directs cleavage of a target nucleic acid sequence 1 to 10, e.g., 3 to 5, base pairs upstream from these sequences. See, e.g., Horvath et al., SCIENCE (2010); 327(5962):167-170, and Deveau et al., J. BACTERIOL. 2008; 190(4): 1390-1400. In an embodiment, an eaCas9 molecule of S. mutans recognizes the sequence motif NGG and/or NAAR (R=A or G) (SEQ ID NO.: BB3) and directs cleavage of a target nucleic acid sequence 1 to 10, e.g., 3 to 5 base pairs, upstream from this sequence. See, e.g., Deveau et al., J BACTERIOL 2008; 190(4): 1390-1400. In an embodiment, an eaCas9 molecule of S. aureus recognizes the sequence motif NNGRR (R=A or G) (SEQ ID NO.: BB4) and directs cleavage of a target nucleic acid sequence 1 to 10, e.g., 3 to 5, base pairs upstream from that sequence. In an embodiment, an eaCas9 molecule of S. aureus recognizes the sequence motif NNGRRN (R=A or G) and directs cleavage of a target nucleic acid sequence 1 to 10, e.g., 3 to 5, base pairs upstream from that sequence. In an embodiment, an eaCas9 molecule of S. aureus recognizes the sequence motif NNGRRT (R=A or G) and directs cleavage of a target nucleic acid sequence 1 to 10, e.g., 3 to 5, base pairs upstream from that sequence. In an embodiment, an eaCas9 molecule of S. aureus recognizes the sequence motif NNGRRV (R=A or G) (SEQ ID NO.: BB5) and directs cleavage of a target nucleic acid sequence 1 to 10, e.g., 3 to 5, base pairs upstream from that sequence. In an embodiment, an eaCas9 molecule of N. meningitidis recognizes the sequence motif NNNNGATT (SEQ ID NO.: BB6) or NNNGCTT (R=A or G) (SEQ ID NO: BB7) and directs cleavage of a target nucleic acid sequence 1 to 10, e.g., 3 to 5, base pairs upstream from that sequence. See, e.g., Hou et al. (2013) PROC. NAT'L. ACAD. SCI. USA 110(39):15644-15649. The ability of a Cas9 molecule to recognize a PAM sequence can be determined, e.g., using a transformation assay described in Jinek et al. (2012) SCIENCE 337:816. In the aforementioned embodiments, N can be any nucleotide residue, e.g., any of A, G, C or T.
  • As is discussed herein, Cas9 molecules can be engineered to alter the PAM specificity of the Cas9 molecule.
  • Exemplary naturally occurring Cas9 molecules are described in Chylinski et al. (2013) RNA BIOLOGY 10:5, 727-737. Such Cas9 molecules include Cas9 molecules of a cluster 1 bacterial family, cluster 2 bacterial family, cluster 3 bacterial family, cluster 4 bacterial family, cluster 5 bacterial family, cluster 6 bacterial family, a cluster 7 bacterial family, a cluster 8 bacterial family, a cluster 9 bacterial family, a cluster 10 bacterial family, a cluster 11 bacterial family, a cluster 12 bacterial family, a cluster 13 bacterial family, a cluster 14 bacterial family, a cluster 15 bacterial family, a cluster 16 bacterial family, a cluster 17 bacterial family, a cluster 18 bacterial family, a cluster 19 bacterial family, a cluster 20 bacterial family, a cluster 21 bacterial family, a cluster 22 bacterial family, a cluster 23 bacterial family, a cluster 24 bacterial family, a cluster 25 bacterial family, a cluster 26 bacterial family, a cluster 27 bacterial family, a cluster 28 bacterial family, a cluster 29 bacterial family, a cluster 30 bacterial family, a cluster 31 bacterial family, a cluster 32 bacterial family, a cluster 33 bacterial family, a cluster 34 bacterial family, a cluster 35 bacterial family, a cluster 36 bacterial family, a cluster 37 bacterial family, a cluster 38 bacterial family, a cluster 39 bacterial family, a cluster 40 bacterial family, a cluster 41 bacterial family, a cluster 42 bacterial family, a cluster 43 bacterial family, a cluster 44 bacterial family, a cluster 45 bacterial family, a cluster 46 bacterial family, a cluster 47 bacterial family, a cluster 48 bacterial family, a cluster 49 bacterial family, a cluster 50 bacterial family, a cluster 51 bacterial family, a cluster 52 bacterial family, a cluster 53 bacterial family, a cluster 54 bacterial family, a cluster 55 bacterial family, a cluster 56 bacterial family, a cluster 57 bacterial family, a cluster 58 bacterial family, a cluster 59 bacterial family, a cluster 60 bacterial family, a cluster 61 bacterial family, a cluster 62 bacterial family, a cluster 63 bacterial family, a cluster 64 bacterial family, a cluster 65 bacterial family, a cluster 66 bacterial family, a cluster 67 bacterial family, a cluster 68 bacterial family, a cluster 69 bacterial family, a cluster 70 bacterial family, a cluster 71 bacterial family, a cluster 72 bacterial family, a cluster 73 bacterial family, a cluster 74 bacterial family, a cluster 75 bacterial family, a cluster 76 bacterial family, a cluster 77 bacterial family, or a cluster 78 bacterial family.
  • Exemplary naturally occurring Cas9 molecules include a Cas9 molecule of a cluster 1 bacterial family. Examples include a Cas9 molecule of: S. pyogenes (e.g., strain SF370, MGAS10270, MGAS10750, MGAS2096, MGAS315, MGAS5005, MGAS6180, MGAS9429, NZ131 and SSI-1), S. thermophilus (e.g., strain LMD-9), S. pseudoporcinus (e.g., strain SPIN 20026), S. mutans (e.g., strain UA159, NN2025), S. macacae (e.g., strain NCTC11558), S. gallolyticus (e.g., strain UCN34, ATCC BAA-2069), S. equines (e.g., strain ATCC 9812, MGCS 124), S. dysdalactiae (e.g., strain GGS 124), S. bovis (e.g., strain ATCC 700338), S. anginosus (e.g., strain F0211), S. agalactiae (e.g., strain NEM316, A909), Listeria monocytogenes (e.g., strain F6854), Listeria innocua (L. innocua, e.g., strain Clip11262), Enterococcus italicus (e.g., strain DSM 15952), or Enterococcus faecium (e.g., strain 1,231,408). Additional exemplary Cas9 molecules are a Cas9 molecule of Neisseria meningitidis (Hou et al., PNAS Early Edition 2013, 1-6 and a S. aureus cas9 molecule.
  • In an embodiment, a Cas9 molecule or Cas9 polypeptide, e.g., an eaCas9 molecule or eaCas9 polypeptide, comprises an amino acid sequence:
  • having 60%, 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% homology with;
  • differs at no more than, 2, 5, 10, 15, 20, 30, or 40% of the amino acid residues when compared with;
  • differs by at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 amino acids, but by no more than 100, 80, 70, 60, 50, 40 or 30 amino acids from; or
  • is identical to any Cas9 molecule sequence described herein, or a naturally occurring Cas9 molecule sequence, e.g., a Cas9 molecule from a species listed herein or described in Chylinski et al. (2013) RNA BIOLOGY 10:5, 727-737; Hou et al., PNAS Early Edition 2013, 1-6. In an embodiment, the Cas9 molecule or Cas9 polypeptide comprises one or more of the following activities: a nickase activity; a double stranded cleavage activity (e.g., an endonuclease and/or exonuclease activity); a helicase activity; or the ability, together with a gRNA molecule, to localize to a target nucleic acid.
  • Engineered or Altered Cas9 Molecules and Cas9 Polypeptides
  • Cas9 molecules and Cas9 polypeptides described herein, e.g., naturally occurring Cas9 molecules, can possess any of a number of properties, including: nuclease activity (e.g., endonuclease and/or exonuclease activity); helicase activity; the ability to associate functionally with a gRNA molecule; and the ability to target (or localize to) a site on a nucleic acid (e.g., PAM recognition and specificity). In an embodiment, a Cas9 molecule or Cas9 polypeptide can include all or a subset of these properties. In a typical embodiment, a Cas9 molecule or Cas9 polypeptide has the ability to interact with a gRNA molecule and, in concert with the gRNA molecule, localize to a site in a nucleic acid. Other activities, e.g., PAM specificity, cleavage activity, or helicase activity can vary more widely in Cas9 molecules and Cas9 polypeptides.
  • Cas9 molecules include engineered Cas9 molecules and engineered Cas9 polypeptides (engineered, as used in this context, means merely that the Cas9 molecule or Cas9 polypeptide differs from a reference sequences, and implies no process or origin limitation). An engineered Cas9 molecule or Cas9 polypeptide can comprise altered enzymatic properties, e.g., altered nuclease activity (as compared with a naturally occurring or other reference Cas9 molecule) or altered helicase activity. As discussed herein, an engineered Cas9 molecule or Cas9 polypeptide can have nickase activity (as opposed to double strand nuclease activity). In an embodiment an engineered Cas9 molecule or Cas9 polypeptide can have an alteration that alters its size, e.g., a deletion of amino acid sequence that reduces its size, e.g., without significant effect on one or more, or any Cas9 activity. In an embodiment, an engineered Cas9 molecule or Cas9 polypeptide can comprise an alteration that affects PAM recognition. For example, an engineered Cas9 molecule can be altered to recognize a PAM sequence other than that recognized by the endogenous wild-type PI domain. In an embodiment a Cas9 molecule or Cas9 polypeptide can differ in sequence from a naturally occurring Cas9 molecule but not have significant alteration in one or more Cas9 activities.
  • Cas9 molecules or Cas9 polypeptides with desired properties can be made in a number of ways, e.g., by alteration of a parental, e.g., naturally occurring Cas9 molecule or Cas9 polypeptide, to provide an altered Cas9 molecule or Cas9 polypeptide having a desired property. For example, one or more mutations or differences relative to a parental Cas9 molecule, e.g., a naturally occurring or engineered Cas9 molecule, can be introduced. Such mutations and differences comprise: substitutions (e.g., conservative substitutions or substitutions of non-essential amino acids), insertions, or deletions. In an embodiment, a Cas9 molecule or Cas9 polypeptide can comprises one or more mutations or differences, e.g., at least 1, 2, 3, 4, 5, 10, 15, 20, 30, 40 or 50 mutations but less than 200, 100, or 80 mutations relative to a reference, e.g., a parental Cas9 molecule.
  • In an embodiment, a mutation or mutations do not have a substantial effect on a Cas9 activity, e.g. a Cas9 activity described herein. In an embodiment, a mutation or mutations have a substantial effect on a Cas9 activity, e.g. a Cas9 activity described herein.
  • Non-Cleaving and Modified-Cleavage Cas9 Molecules and Cas9 Polypeptides
  • In an embodiment, a Cas9 molecule or Cas9 polypeptide comprises a cleavage property that differs from naturally occurring Cas9 molecules, e.g., that differs from the naturally occurring Cas9 molecule having the closest homology. For example, a Cas9 molecule or Cas9 polypeptide can differ from a naturally occurring Cas9 molecule, e.g., a Cas9 molecule of S. pyogenes, as follows: its ability to modulate, e.g., decreased or increased, cleavage of a double stranded nucleic acid (endonuclease and/or exonuclease activity), e.g., as compared to a naturally occurring Cas9 molecule (e.g., a Cas9 molecule of S. pyogenes); its ability to modulate, e.g., decreased or increased, cleavage of a single strand of a nucleic acid, e.g., a non-complementary strand of a nucleic acid molecule or a complementary strand of a nucleic acid molecule (nickase activity), e.g., as compared to a naturally occurring Cas9 molecule (e.g., a Cas9 molecule of S. pyogenes); or the ability to cleave a nucleic acid molecule, e.g., a double stranded or single stranded nucleic acid molecule, can be eliminated.
  • Alterations in the Ability to Cleave One or Both Strands of a Target Nucleic Acid
  • In an embodiment, exemplary Cas9 activities comprise one or more of PAM specificity, cleavage activity, and helicase activity. A mutation(s) can be present, e.g., in: one or more RuvC domains, e.g., an N-terminal RuvC domain; an HNH domain; a region outside the RuvC domains and the HNH domain. In an embodiment, a mutation(s) is present in a RuvC domain. In an embodiment, a mutation(s) is present in an HNH domain. In an embodiment, mutations are present in both a RuvC domain and an HNH domain.
  • Exemplary mutations that may be made in the RuvC domain or HNH domain with reference to the S. pyogenes Cas9 sequence include: D10A, E762A, H840A, N854A, N863A and/or D986A. Exemplary mutations that may be made in the RuvC domain with reference to the S. aureus Cas9 sequence include: N580A.
  • In an embodiment, a Cas9 molecule is an eiCas9 molecule comprising one or more differences in a RuvC domain and/or in an HNH domain as compared to a reference Cas9 molecule, and the eiCas9 molecule does not cleave a nucleic acid, or cleaves with significantly less efficiency than does wild type, e.g., when compared with wild type in a cleavage assay, e.g., as described herein, cuts with less than 50, 25, 10, or 1% of a reference Cas9 molecule, as measured by an assay described herein.
  • Whether or not a particular sequence, e.g., a substitution, may affect one or more activity, such as targeting activity, cleavage activity, etc., can be evaluated or predicted, e.g., by evaluating whether the mutation is conservative. In an embodiment, a “non-essential” amino acid residue, as used in the context of a Cas9 molecule, is a residue that can be altered from the wild-type sequence of a Cas9 molecule, e.g., a naturally occurring Cas9 molecule, e.g., an eaCas9 molecule, without abolishing or more preferably, without substantially altering a Cas9 activity (e.g., cleavage activity), whereas changing an “essential” amino acid residue results in a substantial loss of activity (e.g., cleavage activity).
  • In an embodiment, a Cas9 molecule comprises a cleavage property that differs from naturally occurring Cas9 molecules, e.g., that differs from the naturally occurring Cas9 molecule having the closest homology. For example, a Cas9 molecule can differ from naturally occurring Cas9 molecules, e.g., a Cas9 molecule of S. aureus, S. pyogenes, or C. jejuni as follows: its ability to modulate, e.g., decreased or increased, cleavage of a double stranded break (endonuclease and/or exonuclease activity), e.g., as compared to a naturally occurring Cas9 molecule (e.g., a Cas9 molecule of S. aureus, S. pyogenes, or C. jejuni); its ability to modulate, e.g., decreased or increased, cleavage of a single strand of a nucleic acid, e.g., a non-complimentary strand of a nucleic acid molecule or a complementary strand of a nucleic acid molecule (nickase activity), e.g., as compared to a naturally occurring Cas9 molecule (e.g., a Cas9 molecule of S. aureus, S. pyogenes, or C. jejuni); or the ability to cleave a nucleic acid molecule, e.g., a double stranded or single stranded nucleic acid molecule, can be eliminated.
  • In an embodiment, the altered Cas9 molecule is an eaCas9 molecule comprising one or more of the following activities: cleavage activity associated with a RuvC domain; cleavage activity associated with an HNH domain; cleavage activity associated with an HNH domain and cleavage activity associated with a RuvC domain.
  • In an embodiment, the altered Cas9 molecule is an eiCas9 molecule which does not cleave a nucleic acid molecule (either double stranded or single stranded nucleic acid molecules) or cleaves a nucleic acid molecule with significantly less efficiency, e.g., less than 20, 10, 5, 1 or 0.1% of the cleavage activity of a reference Cas9 molecule, e.g., as measured by an assay described herein. The reference Cas9 molecule can be a naturally occurring unmodified Cas9 molecule, e.g., a naturally occurring Cas9 molecule such as a Cas9 molecule of S. pyogenes, S. thermophilus, S. aureus, C. jejuni or N. meningitidis. In an embodiment, the reference Cas9 molecule is the naturally occurring Cas9 molecule having the closest sequence identity or homology. In an embodiment, the eiCas9 molecule lacks substantial cleavage activity associated with a RuvC domain and cleavage activity associated with an HNH domain.
  • In an embodiment, the altered Cas9 molecule or Cas9 polypeptide, e.g., an eaCas9 molecule or eaCas9 polypeptide, can be a fusion, e.g., of two of more different Cas9 molecules, e.g., of two or more naturally occurring Cas9 molecules of different species. For example, a fragment of a naturally occurring Cas9 molecule of one species can be fused to a fragment of a Cas9 molecule of a second species. As an example, a fragment of a Cas9 molecule of S. pyogenes comprising an N-terminal RuvC-like domain can be fused to a fragment of Cas9 molecule of a species other than S. pyogenes (e.g., S. thermophilus) comprising an HNH-like domain.
  • Cas9 Molecules with Altered PAM Recognition or No PAM Recognition
  • Naturally occurring Cas9 molecules can recognize specific PAM sequences, for example the PAM recognition sequences described above for, e.g., S. pyogenes, S. thermophilus, S. mutans, S. aureus and N. meningitidis.
  • In an embodiment, a Cas9 molecule or Cas9 polypeptide has the same PAM specificities as a naturally occurring Cas9 molecule. In an embodiment, a Cas9 molecule or Cas9 polypeptide has a PAM specificity not associated with a naturally occurring Cas9 molecule, or a PAM specificity not associated with the naturally occurring Cas9 molecule to which it has the closest sequence homology. For example, a naturally occurring Cas9 molecule can be altered, e.g., to alter PAM recognition, e.g., to alter the PAM sequence that the Cas9 molecule or Cas9 polypeptide recognizes to decrease off target sites and/or improve specificity; or eliminate a PAM recognition requirement. In an embodiment, a Cas9 molecule or Cas9 polypeptide can be altered, e.g., to increase length of PAM recognition sequence and/or improve Cas9 specificity to high level of identity (e.g., 98%, 99% or 100% match between gRNA and a PAM sequence), e.g., to decrease off target sites and increase specificity. In an embodiment, the length of the PAM recognition sequence is at least 4, 5, 6, 7, 8, 9, 10 or 15 amino acids in length. In an embodiment, the Cas9 specificity requires at least 90%, 95%, 96%, 97%, 98%, 99% or more homology between the gRNA and the PAM sequence. Cas9 molecules or Cas9 polypeptides that recognize different PAM sequences and/or have reduced off-target activity can be generated using directed evolution. Exemplary methods and systems that can be used for directed evolution of Cas9 molecules are described, e.g., in Esvelt et al. (2011) NATURE 472(7344): 499-503. Candidate Cas9 molecules can be evaluated, e.g., by methods described in Section VII.
  • Alterations of the PI domain, which mediates PAM recognition are discussed below.
  • Synthetic Cas9 Molecules and Cas9 Polypeptides with Altered PI Domains
  • Current genome-editing methods are limited in the diversity of target sequences that can be targeted by the PAM sequence that is recognized by the Cas9 molecule utilized. A synthetic Cas9 molecule (or Syn-Cas9 molecule), or synthetic Cas9 polypeptide (or syn-Cas9 polypeptide), as that term is used herein, refers to a Cas9 molecule or Cas9 polypeptide that comprises a Cas9 core domain from one bacterial species and a functional altered PI domain, i.e., a PI domain other than that naturally associated with the Cas9 core domain, e.g., from a different bacterial species.
  • In an embodiment, the altered PI domain recognizes a PAM sequence that is different from the PAM sequence recognized by the naturally-occurring Cas9 from which the Cas9 core domain is derived. In an embodiment, the altered PI domain recognizes the same PAM sequence recognized by the naturally-occurring Cas9 from which the Cas9 core domain is derived, but with different affinity or specificity. A Syn-Cas9 molecule or Syn-Cas9 polypetide can be, respectively, a Syn-eaCas9 molecule or Syn-eaCas9 polypeptide or a Syn-eiCas9 molecule Syn-eiCas9 polypeptide.
  • An exemplary Syn-Cas9 molecule Syn-Cas9 polypetide comprises:
  • a) a Cas9 core domain, e.g., a Cas9 core domain from Table 100 or 200, e.g., a S. aureus, S. pyogenes, or C. jejuni Cas9 core domain; and
  • b) an altered PI domain from a species X Cas9 sequence selected from Tables 400 and 500.
  • In an embodiment, the RKR motif (the PAM binding motif) of said altered PI domain comprises: differences at 1, 2, or 3 amino acid residues; a difference in amino acid sequence at the first, second, or third position; differences in amino acid sequence at the first and second positions, the first and third positions, or the second and third positions; as compared with the sequence of the RKR motif of the native or endogenous PI domain associated with the Cas9 core domain.
  • In an embodiment, the Cas9 core domain comprises the Cas9 core domain from a species X Cas9 from Table 100 and said altered PI domain comprises a PI domain from a species Y Cas9 from Table 100.
  • In an embodiment, the RKR motif of the species X Cas9 is other than the RKR motif of the species Y Cas9.
  • In an embodiment, the RKR motif of the altered PI domain is selected from XXY, XNG, and XNQ.
  • In an embodiment, the altered PI domain has at least 60, 70, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99 or 100% homology with the amino acid sequence of a naturally occurring PI domain of said species Y from Table 100.
  • In an embodiment, the altered PI domain differs by no more than 50, 40, 30, 25, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residue from the amino acid sequence of a naturally occurring PI domain of said second species from Table 100.
  • In an embodiment, the Cas9 core domain comprises a S. aureus core domain and altered PI domain comprises: an A. denitrificans PI domain; a C. jejuni PI domain; a H. mustelae PI domain; or an altered PI domain of species X PI domain, wherein species X is selected from Table 400 or Table 500.
  • In an embodiment, the Cas9 core domain comprises a S. pyogenes core domain and the altered PI domain comprises: an A. denitrificans PI domain; a C. jejuni PI domain; a H. mustelae PI domain; or an altered PI domain of species X PI domain, wherein species X is selected from Table 400 or Table 500.
  • In an embodiment, the Cas9 core domain comprises a C. jejuni core domain and the altered PI domain comprises: an A. denitrificans PI domain; a H. mustelae PI domain; or an altered PI domain of species X PI domain, wherein species X is selected from Table 400 or Table 500.
  • In an embodiment, the Cas9 molecule further comprises a linker disposed between said Cas9 core domain and said altered PI domain.
  • In an embodiment, the linker comprises: a linker described elsewhere herein disposed between the Cas9 core domain and the heterologous PI domain.
  • Exemplary altered PI domains for use in Syn-Cas9 molecules are described in Tables 400 and 500. The sequences for the 83 Cas9 orthologs referenced in Tables 400 and 500 are provided in Table 100. Table 250 provides the Cas9 orthologs with known PAM sequences and the corresponding RKR motif.
  • In an embodiment, a Syn-Cas9 molecule may also be size-optimized, e.g., the Syn-Cas9 molecule comprises one or more deletions, and optionally one or more linkers disposed between the amino acid residues flanking the deletions. In an embodiment, a Syn-Cas9 molecule comprises a REC deletion.
  • Size-Optimized Cas9 Molecules
  • Engineered Cas9 molecules and engineered Cas9 polypeptides, as described herein, include a Cas9 molecule or Cas9 polypeptide comprising a deletion that reduces the size of the molecule while still retaining desired Cas9 properties, e.g., essentially native conformation, Cas9 nuclease activity, and/or target nucleic acid molecule recognition. Provided herein are Cas9 molecules or Cas9 polypeptides comprising one or more deletions, and optionally one or more linkers, wherein a linker is disposed between the amino acid residues that flank the deletion. Methods for identifying suitable deletions in a reference Cas9 molecule, methods for generating Cas9 molecules with a deletion and a linker, and methods for using such Cas9 molecules will be apparent to one of ordinary skill in the art upon review of this document.
  • A Cas9 molecule, e.g., a S. aureus, S. pyogenes, or C. jejuni, Cas9 molecule, having a deletion is smaller, e.g., has reduced number of amino acids, than the corresponding naturally-occurring Cas9 molecule. The smaller size of the Cas9 molecules allows increased flexibility for delivery methods, and thereby increases utility for genome-editing. A Cas9 molecule can comprise one or more deletions that do not substantially affect or decrease the activity of the resultant Cas9 molecules described herein. Activities that are retained in the Cas9 molecules comprising a deletion as described herein include one or more of the following:
  • a nickase activity, i.e., the ability to cleave a single strand, e.g., the non-complementary strand or the complementary strand, of a nucleic acid molecule; a double stranded nuclease activity, i.e., the ability to cleave both strands of a double stranded nucleic acid and create a double stranded break, which in an embodiment is the presence of two nickase activities;
  • an endonuclease activity;
  • an exonuclease activity;
  • a helicase activity, i.e., the ability to unwind the helical structure of a double stranded nucleic acid;
  • and recognition activity of a nucleic acid molecule, e.g., a target nucleic acid or a gRNA.
  • Activity of the Cas9 molecules described herein can be assessed using the activity assays described herein or in the art.
  • Identifying Regions Suitable for Deletion
  • Suitable regions of Cas9 molecules for deletion can be identified by a variety of methods. Naturally-occurring orthologous Cas9 molecules from various bacterial species, e.g., any one of those listed in Table 100, can be modeled onto the crystal structure of S. pyogenes Cas9 (Nishimasu et al. (2014) CELL, 156: 935-949) to examine the level of conservation across the selected Cas9 orthologs with respect to the three-dimensional conformation of the protein. Less conserved or unconserved regions that are located spatially distant from regions involved in Cas9 activity, e.g., the interface with a target nucleic acid molecule and/or gRNA, represent regions or domains that are candidates for deletion without substantially affecting or decreasing Cas9 activity.
  • REC-Optimized Cas9 Molecules
  • A REC-optimized Cas9 molecule, as that term is used herein, refers to a Cas9 molecule that comprises a deletion in one or both of the REC2 domain and the RE1CT domain (collectively a REC deletion), wherein the deletion comprises at least 10% of the amino acid residues in the cognate domain. A REC-optimized Cas9 molecule can be an eaCas9 molecule or an eiCas9 molecule. An exemplary REC-optimized Cas9 molecule comprises:
  • a) a deletion selected from:
      • i) a REC2 deletion;
      • ii) a REC1CT deletion; or
      • iii) a REC1SUB deletion.
  • Optionally, a linker is disposed between the amino acid residues that flank the deletion. In an embodiment a Cas9 molecule includes only one deletion, or only two deletions. A Cas9 molecule can comprise a REC2 deletion and a REC1CT deletion. A Cas9 molecule can comprise a REC2 deletion and a REC1SUB deletion.
  • Generally, the deletion will contain at least 10% of the amino acids in the cognate domain, e.g., a REC2 deletion will include at least 10% of the amino acids in the REC2 domain. A deletion can comprise: at least 10, 20, 30, 40, 50, 60, 70, 80, or 90% of the amino acid residues of its cognate domain; all of the amino acid residues of its cognate domain; an amino acid residue outside its cognate domain; a plurality of amino acid residues outside its cognate domain; the amino acid residue immediately N terminal to its cognate domain; the amino acid residue immediately C terminal to its cognate domain; the amino acid residue immediately N terminal to its cognate and the amino acid residue immediately C terminal to its cognate domain; a plurality of, e.g., up to 5, 10, 15, or 20, amino acid residues N terminal to its cognate domain; a plurality of, e.g., up to 5, 10, 15, or 20, amino acid residues C terminal to its cognate domain; a plurality of, e.g., up to 5, 10, 15, or 20, amino acid residues N terminal to its cognate domain and a plurality of e.g., up to 5, 10, 15, or 20, amino acid residues C terminal to its cognate domain.
  • In an embodiment, a deletion does not extend beyond: its cognate domain; the N terminal amino acid residue of its cognate domain; the C terminal amino acid residue of its cognate domain.
  • A REC-optimized Cas9 molecule can include a linker disposed between the amino acid residues that flank the deletion. Linkers for use in generating recombinant proteins, e.g., multi-domain proteins, are known in the art (Chen et al. (2013) ADV. DRUG DELIVERY REV. 65:1357-69). Any linkers known in the art that maintain the conformation or native fold of the Cas9 molecule (thereby retaining Cas9 activity) can be used. Several properties of linkers, such as length, hydrophobicity, intrinsic properties of the amino acids residues themselves, and secondary structure should be considered in the context of the goal to maintain native conformation and functional activity of Cas9. Any linkers known in the art that maintain the conformation or native fold of the Cas9 molecule (thereby retaining Cas9 activity) can be used. Several properties of linkers, such as length, hydrophobicity, intrinsic properties of the amino acids residues themselves, and secondary structure should be considered in the context of the goal to maintain native conformation and functional activity of Cas9.
  • A flexible linker can be utilized in the Cas9 molecules described herein. Flexible linkers allow a certain degree of movement and/or interaction within and between the joined domains or regions of the protein. Generally, flexible linkers are composed of small, non-polar (e.g., Gly) or polar (e.g., Ser or Thr) amino acids. The small size of these amino acids provides flexibility and allows mobility of the connected domains or regions. Furthermore, the incorporation of Ser or Thr can help maintain the stability of the linker in aqueous solutions by hydrogen bonding with the water molecules, thereby reducing unfavorable interactions between the linker and the other protein moieties. Commonly used flexible linkers are comprised of sequences that primarily consist of Gly and Ser residues. Often, these flexible linkers consist of repeating units of a combination of Gly and Ser residues, e.g., (GGS)x, where the number of repeating units, e.g., x, can be optimized to achieve the appropriate separation of other domains or regions of the protein.
  • In some cases, a rigid linker may be preferred if there is significant distance between the joined domains or regions, or to maintain a fixed distance between the joined domains or regions of a protein and independent functions of the domains/regions. Rigid linkers often have defined secondary structure, e.g., alpha helix, or other stabilizing interactions, e.g., salt bridges and disulfide bonds. Rigid linkers commonly contain multiple Pro residues, or repeating combinations of Glu-Pro or Lys-Pro because Pro imposes a strong conformation constraint due to its structure.
  • The linker can comprise an amino acid residue, e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acid residues. Typically, the linker will comprises less than 10, 20 or 30 amino acid residues. Typically, the linker is less than 50, 40, 30, 20, 10, or 5% of the length of the deleted sequence. Suitable linkers include: [Gly-Ser]x, wherein x is 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; [Gly-Gly-Ser]x, wherein x is 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; [Gly-Gly-Ser]; [Gly-Ser-Gly-Ser]x, wherein x is 1, 2, 3, 4, or 5; [Gly-Ser-Gly-Ser]; (GSAGSAAGSGEF)x, wherein x is 1, 2, 3 or 4; (SIVAQLSRPDPA)x, wherein x is 1, 2, 3 or 4; or an XTEN sequence, e.g., the XTEN sequence of SEQ ID NO: _, or a sequence that differs therefrom by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 amino acid residues. In an embodiment linker comprises an amino acid sequence other than a sequence within REC2.
  • In an embodiment, a REC-optimized Cas9 molecule comprises an amino acid sequence that, other than any REC deletion and associated linker, has at least 50, 55, 60, 65, 70, 75, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% homology with the amino acid sequence of a naturally occurring Cas9, e.g., a Cas9 molecule described in Table 100, e.g., a S. aureus Cas9 molecule, a S. pyogenes Cas9 molecule, or a C. jejuni Cas9 molecule.
  • In an embodiment, a REC-optimized Cas9 molecule comprises an amino acid sequence that, other than any REC deletion and associated linker, differs by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25 amino acid residues from the amino acid sequence of a naturally occurring Cas9, e.g., a Cas9 molecule described in Table 100, e.g., a S. aureus Cas9 molecule, a S. pyogenes Cas9 molecule, or a C. jejuni Cas9 molecule.
  • In an embodiment, a REC-optimized Cas9 molecule comprises an amino acid sequence that, other than any REC deletion and associate linker, differs by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25% of the amino acid residues from the amino acid sequence of a naturally occurring Cas9, e.g., a Cas9 molecule described in Table 100, e.g., a S. aureus Cas9 molecule, a S. pyogenes Cas9 molecule, or a C. jejuni Cas9 molecule.
  • For sequence comparison, typically one sequence acts as a reference sequence, to which test sequences are compared. When using a sequence comparison algorithm, test and reference sequences are entered into a computer, subsequence coordinates are designated, if necessary, and sequence algorithm program parameters are designated. Default program parameters can be used, or alternative parameters can be designated. The sequence comparison algorithm then calculates the percent sequence identities for the test sequences relative to the reference sequence, based on the program parameters. Methods of alignment of sequences for comparison are well known in the art. Optimal alignment of sequences for comparison can be conducted, e.g., by the local homology algorithm of Smith and Waterman (1970) ADV. APPL. MATH. 2: 482c, by the homology alignment algorithm of Needleman and Wunsch, (1970) J. MOL. BIOL. 48:443, by the search for similarity method of Pearson and Lipman, (1988) PROC. NAT'L. ACAD. SCI. USA 85:2444, by computerized implementations of these algorithms (GAP, BESTFIT, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Dr., Madison, Wis.), or by manual alignment and visual inspection (see, e.g., Brent et al., (2003) CURRENT PROTOCOLS IN MOLECULAR BIOLOGY).
  • Two examples of algorithms that are suitable for determining percent sequence identity and sequence similarity are the BLAST and BLAST 2.0 algorithms, which are described in Altschul et al. (1977) NUC. ACIDS RES. 25:3389-3402; and Altschul et al. (1990) J. MOL. BIOL. 215: 403-410, respectively. Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information. The percent identity between two amino acid sequences can also be determined using the algorithm of E. Meyers and W. Miller (1988) COMPUT. APPL. BIOSCI. 4:11-17, which has been incorporated into the ALIGN program (version 2.0), using a PAM120 weight residue table, a gap length penalty of 12 and a gap penalty of 4. In addition, the percent identity between two amino acid sequences can be determined using the Needleman and Wunsch (1970) J. MOL. BIOL. 48:444-453 algorithm which has been incorporated into the GAP program in the GCG software package (available at www.gcg.com), using either a Blossom 62 matrix or a PAM250 matrix, and a gap weight of 16, 14, 12, 10, 8, 6, or 4 and a length weight of 1, 2, 3, 4, 5, or 6.
  • Sequence information for exemplary REC deletions are provided for 83 naturally-occurring Cas9 orthologs in Table 100.
  • The amino acid sequences of exemplary Cas9 molecules from different bacterial species are shown below.
  • TABLE 100
    Amino Acid Sequence of Cas9 Orthologs
    REC2 REC1CT Recsub
    Species/ start stop # AA start stop # AA start stop # AA
    Composite (AA (AA deleted (AA (AA deleted (AA (AA deleted
    ID Amino acid sequence pos) pos) (n) pos) pos) (n) pos) pos) (n)
    Staphylococcus MKRNYILGLDIGITSVGYGIID 126 166 41 296 352 57 296 352 57
    aureus YETRDVIDAGVRLFKEANVENN
    tr|J7RUA5|J7RUA5| EGRRSKRGARRLKRRRRHRIQR
    J7RUA5_STAAU VKKLLFDYNLLTDHSELSGINP
    YEARVKGLSQKLSEEEFSAALL
    HLAKRRGVHNVNEVEEDTGNEL
    STKEQISRNSKALEEKYVAELQ
    LERLKKDGEVRGSINRFKTSDY
    VKEAKQLLKVQKAYHQLDQSFI
    DTYIDLLETRRTYYEGPGEGSP
    FGWKDIKEWYEMLMGHCTYFPE
    ELRSVKYAYNADLYNALNDLNN
    LVITRDENEKLEYYEKFQIIEN
    VFKQKKKPTLKQIAKEILVNEE
    DIKGYRVTSTGKPEFTNLKVYH
    DIKDITARKEIIENAELLDQIA
    KILTIYQSSEDIQEELTNLNSE
    LTQEEIEQISNLKGYTGTHNLS
    LKAINLILDELWHTNDNQIAIF
    NRLKLVPKKVDLSQQKEIPTTL
    VDDFILSPVVKRSFIQSIKVIN
    AIIKKYGLPNDIIIELAREKNS
    KDAQKMINEMQKRNRQTNERIE
    EIIRTTGKENAKYLIEKIKLHD
    MQEGKCLYSLEAIPLEDLLNNP
    FNYEVDHIIPRSVSFDNSFNNK
    VLVKQEENSKKGNRTPFQYLSS
    SDSKISYETFKKHILNLAKGKG
    RISKTKKEYLLEERDINRFSVQ
    KDFINRNLVDTRYATRGLMNLL
    RSYFRVNNLDVKVKSINGGFTS
    FLRRKWKFKKERNKGYKHHAED
    ALIIANADFIFKEWKKLDKAKK
    VMENQMFEEKQAESMPEIETEQ
    EYKEIFITPHQIKHIKDFKDYK
    YSHRVDKKPNRELINDTLYSTR
    KDDKGNTLIVNNLNGLYDKDND
    KLKKLINKSPEKLLMYHHDPQT
    YQKLKLIMEQYGDEKNPLYKYY
    EETGNYLTKYSKKDNGPVIKKI
    KYYGNKLNAHLDITDDYPNSRN
    KVVKLSLKPYRFDVYLDNGVYK
    FVTVKNLDVIKKENYYEVNSKC
    YEEAKKLKKISNQAEFIASFYN
    NDLIKINGELYRVIGVNNDLLN
    RIEVNMIDITYREYLENMNDKR
    PPRIIKTIASKTQSIKKYSTDI
    LGNLYEVKSKKHPQIIKKG
    (SEQ ID NO: 6)
    Streptococcus MDKKYSIGLDIGTNSVGWAVIT 176 314 139 511 592 82 511 592 82
    pyogenes DEYKVPSKKFKVLGNTDRHSIK
    sp|Q99ZW2| KNLIGALLFDSGETAEATRLKR
    CAS9_STRP1 TARRRYTRRKNRICYLQEIFSN
    EMAKVDDSFFHRLEESFLVEED
    KKHERHPIFGNIVDEVAYHEKY
    PTIYHLRKKLVDSTDKADLRLI
    YLALAHMIKFRGHFLIEGDLNP
    DNSDVDKLFIQLVQTYNQLFEE
    NPINASGVDAKAILSARLSKSR
    RLENLIAQLPGEKKNGLFGNLI
    ALSLGLTPNFKSNFDLAEDAKL
    QLSKDTYDDDLDNLLAQIGDQY
    ADLFLAAKNLSDAILLSDILRV
    NTEITKAPLSASMIKRYDEHHQ
    DLTLLKALVRQQLPEKYKEIFF
    DQSKNGYAGYIDGGASQEEFYK
    FIKPILEKMDGTEELLVKLNRE
    DLLRKQRTFDNGSIPHQIHLGE
    LHAILRRQEDFYPFLKDNREKI
    EKILTFRIPYYVGPLARGNSRF
    AWMTRKSEETITPWNFEEVVDK
    GASAQSFIERMTNFDKNLPNEK
    VLPKHSLLYEYFTVYNELTKVK
    YVTEGMRKPAFLSGEQKKAIVD
    LLFKTNRKVTVKQLKEDYFKKI
    ECFDSVEISGVEDRFNASLGTY
    HDLLKIIKDKDFLDNEENEDIL
    EDIVLTLTLFEDREMIEERLKT
    YAHLFDDKVMKQLKRRRYTGWG
    RLSRKLINGIRDKQSGKTILDF
    LKSDGFANRNFMQLIHDDSLTF
    KEDIQKAQVSGQGDSLHEHIAN
    LAGSPAIKKGILQTVKVVDELV
    KVMGRHKPENIVIEMARENQTT
    QKGQKNSRERMKRIEEGIKELG
    SQILKEHPVENTQLQNEKLYLY
    YLQNGRDMYVDQELDINRLSDY
    DVDHIVPQSFLKDDSIDNKVLT
    RSDKNRGKSDNVPSEEVVKKMK
    NYWRQLLNAKLITQRKFDNLTK
    AERGGLSELDKAGFIKRQLVET
    RQITKHVAQILDSRMNTKYDEN
    DKLIREVKVITLKSKLVSDFRK
    DFQFYKVREINNYHHAHDAYLN
    AVVGTALIKKYPKLESEFVYGD
    YKVYDVRKMIAKSEQEIGKATA
    KYFFYSNIMNFFKTEITLANGE
    IRKRPLIETNGETGEIVWDKGR
    DFATVRKVLSMPQVNIVKKTEV
    QTGGFSKESILPKRNSDKLIAR
    KKDWDPKKYGGFDSPTVAYSVL
    VVAKVEKGKSKKLKSVKELLGI
    TIMERSSFEKNPIDFLEAKGYK
    EVKKDLIIKLPKYSLFELENGR
    KRMLASAGELQKGNELALPSKY
    VNFLYLASHYEKLKGSPEDNEQ
    KQLFVEQHKHYLDEIIEQISEF
    SKRVILADANLDKVLSAYNKHR
    DKPIREQAENIIHLFTLTNLGA
    PAAFKYFDTTIDRKRYTSTKEV
    LDATLIHQSITGLYETRIDLSQ
    LGGD (SEQ ID NO: 7)
    Campylobacter MARILAFDIGISSIGWAFSEND 137 181 45 316 360 45 316 360 45
    jejuni ELKDCGVRIFTKVENPKTGESL
    NCTC 11168 ALPRRLARSARKRLARRKARLN
    gi|218563121| HLKHLIANEFKLNYEDYQSFDE
    ref|YP_002344900.1 SLAKAYKGSLISPYELRFRALN
    ELLSKQDFARVILHIAKRRGYD
    DIKNSDDKEKGAILKAIKQNEE
    KLANYQSVGEYLYKEYFQKFKE
    NSKEFTNVRNKKESYERCIAQS
    FLKDELKLIFKKQREFGFSFSK
    KFEEEVLSVAFYKRALKDFSHL
    VGNCSFFTDEKRAPKNSPLAFM
    FVALTRIINLLNNLKNTEGILY
    TKDDLNALLNEVLKNGTLTYKQ
    TKKLLGLSDDYEFKGEKGTYFI
    EFKKYKEFIKALGEHNLSQDDL
    NEIAKDITLIKDEIKLKKALAK
    YDLNQNQIDSLSKLEFKDHLNI
    SFKALKLVTPLMLEGKKYDEAC
    NELNLKVAINEDKKDFLPAFNE
    TYYKDEVTNPVVLRAIKEYRKV
    LNALLKKYGKVHKINIELAREV
    GKNHSQRAKIEKEQNENYKAKK
    DAELECEKLGLKINSKNILKLR
    LFKEQKEFCAYSGEKIKISDLQ
    DEKMLEIDHIYPYSRSFDDSYM
    NKVLVFTKQNQEKLNQTPFEAF
    GNDSAKWQKIEVLAKNLPTKKQ
    KRILDKNYKDKEQKNFKDRNLN
    DTRYIARLVLNYTKDYLDFLPL
    SDDENTKLNDTQKGSKVHVEAK
    SGMLTSALRHTWGFSAKDRNNH
    LHHAIDAVIIAYANNSIVKAFS
    DFKKEQESNSAELYAKKISELD
    YKNKRKFFEPFSGFRQKVLDKI
    DEIFVSKPERKKPSGALHEETF
    RKEEEFYQSYGGKEGVLKALEL
    GKIRKVNGKIVKNGDMFRVDIF
    KHKKTNKFYAVPIYTMDFALKV
    LPNKAVARSKKGEIKDWILMDE
    NYEFCFSLYKDSLILIQTKDMQ
    EPEFVYYNAFTSSTVSLIVSKH
    DNKFETLSKNQKILFKNANEKE
    VIAKSIGIQNLKVFEKYIVSAL
    GEVTKAEFRQREDFKK (SEQ
    ID NO: 8)
    Bacteroides MKRILGLDLGTNSIGWALVNEA 148 339 192 524 617 84 524 617 84
    fragilis NCTC ENKDERSSIVKLGVRVNPLTVD
    9343 ELTNFEKGKSITTNADRTLKRG
    gi|60683389| MRRNLQRYKLRRETLTEVLKEH
    ref|YP_213533.1| KLITEDTILSENGNRTTFETYR
    LRAKAVTEEISLEEFARVLLMI
    NKKRGYKSSRKAKGVEEGTLID
    GMDIARELYNNNLTPGELCLQL
    LDAGKKFLPDFYRSDLQNELDR
    IWEKQKEYYPEILTDVLKEELR
    GKKRDAVWAICAKYFVWKENYT
    EWNKEKGKTEQQEREHKLEGIY
    SKRKRDEAKRENLQWRVNGLKE
    KLSLEQLVIVFQEMNTQINNSS
    GYLGAISDRSKELYFNKQTVGQ
    YQMEMLDKNPNASLRNMVFYRQ
    DYLDEFNMLWEKQAVYHKELTE
    ELKKEIRDIIIFYQRRLKSQKG
    LIGFCEFESRQIEVDIDGKKKI
    KTVGNRVISRSSPLFQEFKIWQ
    ILNNIEVTVVGKKRKRRKLKEN
    YSALFEELNDAEQLELNGSRRL
    CQEEKELLAQELFIRDKMTKSE
    VLKLLFDNPQELDLNFKTIDGN
    KTGYALFQAYSKMIEMSGHEPV
    DFKKPVEKVVEYIKAVFDLLNW
    NTDILGFNSNEELDNQPYYKLW
    HLLYSFEGDNTPTGNGRLIQKM
    TELYGFEKEYATILANVSFQDD
    YGSLSAKAIHKILPHLKEGNRY
    DVACVYAGYRHSESSLTREEIA
    NKVLKDRLMLLPKNSLHNPVVE
    KILNQMVNVINVIIDIYGKPDE
    IRVELARELKKNAKEREELTKS
    IAQTTKAHEEYKTLLQTEFGLT
    NVSRTDILRYKLYKELESCGYK
    TLYSNTYISREKLFSKEFDIEH
    IIPQARLFDDSFSNKTLEARSV
    NIEKGNKTAYDFVKEKFGESGA
    DNSLEHYLNNIEDLFKSGKISK
    TKYNKLKMAEQDIPDGFIERDL
    RNTQYIAKKALSMLNEISHRVV
    ATSGSVTDKLREDWQLIDVMKE
    LNWEKYKALGLVEYFEDRDGRQ
    IGRIKDWTKRNDHRHHAMDALT
    VAFTKDVFIQYFNNKNASLDPN
    ANEHAIKNKYFQNGRAIAPMPL
    REFRAEAKKHLENTLISIKAKN
    KVITGNINKTRKKGGVNKNMQQ
    TPRGQLHLETIYGSGKQYLTKE
    EKVNASFDMRKIGTVSKSAYRD
    ALLKRLYENDNDPKKAFAGKNS
    LDKQPIWLDKEQMRKVPEKVKI
    VTLEAIYTIRKEISPDLKVDKV
    IDVGVRKILIDRLNEYGNDAKK
    AFSNLDKNPIWLNKEKGISIKR
    VTISGISNAQSLHVKKDKDGKP
    ILDENGRNIPVDFVNTGNNHHV
    AVYYRPVIDKRGQLVVDEAGNP
    KYELEEVVVSFFEAVTRANLGL
    PIIDKDYKTTEGWQFLFSMKQN
    EYFVFPNEKTGFNPKEIDLLDV
    ENYGLISPNLFRVQKFSLKNYV
    FRHHLETTIKDTSSILRGITWI
    DFRSSKGLDTIVKVRVNHIGQI
    VSVGEY (SEQ ID NO: 9)
    Bifidobacterium MSRKNYVDDYAISLDIGNASVG 173 335 163 516 607 87 516 607 87
    bifidum WSAFTPNYRLVRAKGHELIGVR
    S17 LFDPADTAESRRMARTTRRRYS
    gi|310286728| RRRWRLRLLDALFDQALSEIDP
    ref|YP_003937986. SFLARRKYSWVHPDDENNADCW
    YGSVLFDSNEQDKRFYEKYPTI
    YHLRKALMEDDSQHDIREIYLA
    IHHMVKYRGNFLVEGTLESSNA
    FKEDELLKLLGRITRYEMSEGE
    QNSDIEQDDENKLVAPANGQLA
    DALCATRGSRSMRVDNALEALS
    AVNDLSREQRAIVKAIFAGLEG
    NKLDLAKIFVSKEFSSENKKIL
    GIYFNKSDYEEKCVQIVDSGLL
    DDEEREFLDRMQGQYNAIALKQ
    LLGRSTSVSDSKCASYDAHRAN
    WNLIKLQLRTKENEKDINENYG
    ILVGWKIDSGQRKSVRGESAYE
    NMRKKANVFFKKMIETSDLSET
    DKNRLIHDIEEDKLFPIQRDSD
    NGVIPHQLHQNELKQIIKKQGK
    YYPFLLDAFEKDGKQINKIEGL
    LTFRVPYFVGPLVVPEDLQKSD
    NSENHWMVRKKKGEITPWNFDE
    MVDKDASGRKFIERLVGTDSYL
    LGEPTLPKNSLLYQEYEVLNEL
    NNVRLSVRTGNHWNDKRRMRLG
    REEKTLLCQRLFMKGQTVTKRT
    AENLLRKEYGRTYELSGLSDES
    KFTSSLSTYGKMCRIFGEKYVN
    EHRDLMEKIVELQTVFEDKETL
    LHQLRQLEGISEADCALLVNTH
    YTGWGRLSRKLLTTKAGECKIS
    DDFAPRKHSIIEIMRAEDRNLM
    EIITDKQLGFSDWIEQENLGAE
    NGSSLMEVVDDLRVSPKVKRGI
    IQSIRLIDDISKAVGKRPSRIF
    LELADDIQPSGRTISRKSRLQD
    LYRNANLGKEFKGIADELNACS
    DKDLQDDRLFLYYTQLGKDMYT
    GEELDLDRLSSAYDIDHIIPQA
    VTQNDSIDNRVLVARAENARKT
    DSFTYMPQIADRMRNFWQILLD
    NGLISRVKFERLTRQNEFSERE
    KERFVQRSLVETRQIMKNVATL
    MRQRYGNSAAVIGLNAELTKEM
    HRYLGFSHKNRDINDYHHAQDA
    LCVGIAGQFAANRGFFADGEVS
    DGAQNSYNQYLRDYLRGYREKL
    SAEDRKQGRAFGFIVGSMRSQD
    EQKRVNPRTGEVVWSEEDKDYL
    RKVMNYRKMLVTQKVGDDFGAL
    YDETRYAATDPKGIKGIPFDGA
    KQDTSLYGGFSSAKPAYAVLIE
    SKGKTRLVNVTMQEYSLLGDRP
    SDDELRKVLAKKKSEYAKANIL
    LRHVPKMQLIRYGGGLMVIKSA
    GELNNAQQLWLPYEEYCYFDDL
    SQGKGSLEKDDLKKLLDSILGS
    VQCLYPWHRFTEEELADLHVAF
    DKLPEDEKKNVITGIVSALHAD
    AKTANLSIVGMTGSWRRMNNKS
    GYTFSDEDEFIFQSPSGLFEKR
    VTVGELKRKAKKEVNSKYRTNE
    KRLPTLSGASQP (SEQ ID
    NO: 10)
    Veillonella METQTSNQLITSHLKDYPKQDY 185 339 155 574 663 79 574 663 79
    atypica ACS- FVGLDIGTNSVGWAVTNTSYEL
    134-V-Col7a LKFHSHKMWGSRLFEEGESAVT
    gi|303229466| RRGFRSMRRRLERRKLRLKLLE
    ref|ZP_07316256.1 ELFADAMAQVDSTFFIRLHESK
    YHYEDKTTGHSSKHILFIDEDY
    TDQDYFTEYPTIYHLRKDLMEN
    GTDDIRKLFLAVHHILKYRGNF
    LYEGATFNSNAFTFEDVLKQAL
    VNITFNCFDTNSAISSISNILM
    ESGKTKSDKAKAIERLVDTYTV
    FDEVNTPDKPQKEQVKEDKKTL
    KAFANLVLGLSANLIDLFGSVE
    DIDDDLKKLQIVGDTYDEKRDE
    LAKVWGDEIHIIDDCKSVYDAI
    ILMSIKEPGLTISQSKVKAFDK
    HKEDLVILKSLLKLDRNVYNEM
    FKSDKKGLHNYVHYIKQGRTEE
    TSCSREDFYKYTKKIVEGLADS
    KDKEYILNEIELQTLLPLQRIK
    DNGVIPYQLHLEELKVILDKCG
    PKFPFLHTVSDGFSVTEKLIKM
    LEFRIPYYVGPLNTHHNIDNGG
    FSWAVRKQAGRVTPWNFEEKID
    REKSAAAFIKNLTNKCTYLFGE
    DVLPKSSLLYSEFMLLNELNNV
    RIDGKALAQGVKQHLIDSIFKQ
    DHKKMTKNRIELFLKDNNYITK
    KHKPEITGLDGEIKNDLTSYRD
    MVRILGNNFDVSMAEDIITDIT
    IFGESKKMLRQTLRNKFGSQLN
    DETIKKLSKLRYRDWGRLSKKL
    LKGIDGCDKAGNGAPKTIIELM
    RNDSYNLMEILGDKFSFMECIE
    EENAKLAQGQVVNPHDIIDELA
    LSPAVKRAVWQALRIVDEVAHI
    KKALPSRIFVEVARTNKSEKKK
    KDSRQKRLSDLYSAIKKDDVLQ
    SGLQDKEFGALKSGLANYDDAA
    LRSKKLYLYYTQMGRCAYTGNI
    IDLNQLNTDNYDIDHIYPRSLT
    KDDSFDNLVLCERTANAKKSDI
    YPIDNRIQTKQKPFWAFLKHQG
    LISERKYERLTRIAPLTADDLS
    GFIARQLVETNQSVKATTTLLR
    RLYPDIDVVFVKAENVSDFRHN
    NNFIKVRSLNHHHHAKDAYLNI
    VVGNVYHEKFTRNFRLFFKKNG
    ANRTYNLAKMFNYDVICTNAQD
    GKAWDVKTSMNTVKKMMASNDV
    RVTRRLLEQSGALADATIYKAS
    VAAKAKDGAYIGMKTKYSVFAD
    VTKYGGMTKIKNAYSIIVQYTG
    KKGEEIKEIVPLPIYLINRNAT
    DIELIDYVKSVIPKAKDISIKY
    RKLCINQLVKVNGFYYYLGGKT
    NDKIYIDNAIELVVPHDIATYI
    KLLDKYDLLRKENKTLKASSIT
    TSIYNINTSTVVSLNKVGIDVF
    DYFMSKLRTPLYMKMKGNKVDE
    LSSTGRSKFIKMTLEEQSIYLL
    EVLNLLTNSKTTFDVKPLGITG
    SRSTIGVKIHNLDEFKIINESI
    TGLYSNEVTIV (SEQ ID
    NO: 11)
    Lactobacillus MTKLNQPYGIGLDIGSNSIGFA 169 320 152 559 645 78 559 645 78
    rhamnosus VVDANSHLLRLKGETAIGARLF
    GG REGQSAADRRGSRTTRRRLSRT
    gi|258509199| RWRLSFLRDFFAPHITKIDPDF
    ref|YP_003171950.1 FLRQKYSEISPKDKDRFKYEKR
    LFNDRTDAEFYEDYPSMYHLRL
    HLMTHTHKADPREIFLAIHHIL
    KSRGHFLTPGAAKDFNTDKVDL
    EDIFPALTEAYAQVYPDLELTF
    DLAKADDFKAKLLDEQATPSDT
    QKALVNLLLSSDGEKEIVKKRK
    QVLTEFAKAITGLKTKFNLALG
    TEVDEADASNWQFSMGQLDDKW
    SNIETSMTDQGTEIFEQIQELY
    RARLLNGIVPAGMSLSQAKVAD
    YGQHKEDLELFKTYLKKLNDHE
    LAKTIRGLYDRYINGDDAKPFL
    REDFVKALTKEVTAHPNEVSEQ
    LLNRMGQANFMLKQRTKANGAI
    PIQLQQRELDQIIANQSKYYDW
    LAAPNPVEAHRWKMPYQLDELL
    NFHIPYYVGPLITPKQQAESGE
    NVFAWMVRKDPSGNITPYNFDE
    KVDREASANTFIQRMKTTDTYL
    IGEDVLPKQSLLYQKYEVLNEL
    NNVRINNECLGTDQKQRLIREV
    FERHSSVTIKQVADNLVAHGDF
    ARRPEIRGLADEKRFLSSLSTY
    HQLKEILHEAIDDPTKLLDIEN
    IITWSTVFEDHTIFETKLAEIE
    WLDPKKINELSGIRYRGWGQFS
    RKLLDGLKLGNGHTVIQELMLS
    NHNLMQILADETLKETMTELNQ
    DKLKTDDIEDVINDAYTSPSNK
    KALRQVLRVVEDIKHAANGQDP
    SWLFIETADGTGTAGKRTQSRQ
    KQIQTVYANAAQELIDSAVRGE
    LEDKIADKASFTDRLVLYFMQG
    GRDIYTGAPLNIDQLSHYDIDH
    ILPQSLIKDDSLDNRVLVNATI
    NREKNNVFASTLFAGKMKATWR
    KWHEAGLISGRKLRNLMLRPDE
    IDKFAKGFVARQLVETRQIIKL
    TEQIAAAQYPNTKIIAVKAGLS
    HQLREELDFPKNRDVNHYHHAF
    DAFLAARIGTYLLKRYPKLAPF
    FTYGEFAKVDVKKFREFNFIGA
    LTHAKKNIIAKDTGEIVWDKER
    DIRELDRIYNFKRMLITHEVYF
    ETADLFKQTIYAAKDSKERGGS
    KQLIPKKQGYPTQVYGGYTQES
    GSYNALVRVAEADTTAYQVIKI
    SAQNASKIASANLKSREKGKQL
    LNEIVVKQLAKRRKNWKPSANS
    FKIVIPRFGMGTLFQNAKYGLF
    MVNSDTYYRNYQELWLSRENQK
    LLKKLFSIKYEKTQMNHDALQV
    YKAIIDQVEKFFKLYDINQFRA
    KLSDAIERFEKLPINTDGNKIG
    KTETLRQILIGLQANGTRSNVK
    NLGIKTDLGLLQVGSGIKLDKD
    TQIVYQSPSGLFKRRIPLADL
    (SEQ ID NO: 12)
    Filifactor MTKEYYLGLDVGTNSVGWAVTD 166 314 149 508 592 76 508 592 76
    alocis ATCC SQYNLCKFKKKDMWGIRLFESA
    35896 NTAKDRRLQRGNRRRLERKKQR
    gi|374307738| IDLLQEIFSPEICKIDPTFFIR
    ref|YP_005054169.1 LNESRLHLEDKSNDFKYPLFIE
    KDYSDIEYYKEFPTIFHLRKHL
    IESEEKQDIRLIYLALHNIIKT
    RGHFLIDGDLQSAKQLRPILDT
    FLLSLQEEQNLSVSLSENQKDE
    YEEILKNRSIAKSEKVKKLKNL
    FEISDELEKEEKKAQSAVIENF
    CKFIVGNKGDVCKFLRVSKEEL
    EIDSFSFSEGKYEDDIVKNLEE
    KVPEKVYLFEQMKAMYDWNILV
    DILETEEYISFAKVKQYEKHKT
    NLRLLRDIILKYCTKDEYNRMF
    NDEKEAGSYTAYVGKLKKNNKK
    YWIEKKRNPEEFYKSLGKLLDK
    IEPLKEDLEVLTMMIEECKNHT
    LLPIQKNKDNGVIPHQVHEVEL
    KKILENAKKYYSFLTETDKDGY
    SVVQKIESIFRFRIPYYVGPLS
    TRHQEKGSNVWMVRKPGREDRI
    YPWNMEEIIDFEKSNENFITRM
    TNKCTYLIGEDVLPKHSLLYSK
    YMVLNELNNVKVRGKKLPTSLK
    QKVFEDLFENKSKVTGKNLLEY
    LQIQDKDIQIDDLSGFDKDFKT
    SLKSYLDFKKQIFGEEIEKESI
    QNMIEDIIKWITIYGNDKEMLK
    RVIRANYSNQLTEEQMKKITGF
    QYSGWGNFSKMFLKGISGSDVS
    TGETFDIITAMWETDNNLMQIL
    SKKFTFMDNVEDFNSGKVGKID
    KITYDSTVKEMFLSPENKRAVW
    QTIQVAEEIKKVMGCEPKKIFI
    EMARGGEKVKKRTKSRKAQLLE
    LYAACEEDCRELIKEIEDRDER
    DFNSMKLFLYYTQFGKCMYSGD
    DIDINELIRGNSKWDRDHIYPQ
    SKIKDDSIDNLVLVNKTYNAKK
    SNELLSEDIQKKMHSFWLSLLN
    KKLITKSKYDRLTRKGDFTDEE
    LSGFIARQLVETRQSTKAIADI
    FKQIYSSEVVYVKSSLVSDFRK
    KPLNYLKSRRVNDYHHAKDAYL
    NIVVGNVYNKKFTSNPIQWMKK
    NRDTNYSLNKVFEHDVVINGEV
    IWEKCTYHEDTNTYDGGTLDRI
    RKIVERDNILYTEYAYCEKGEL
    FNATIQNKNGNSTVSLKKGLDV
    KKYGGYFSANTSYFSLIEFEDK
    KGDRARHIIGVPIYIANMLEHS
    PSAFLEYCEQKGYQNVRILVEK
    IKKNSLLIINGYPLRIRGENEV
    DTSFKRAIQLKLDQKNYELVRN
    IEKFLEKYVEKKGNYPIDENRD
    HITHEKMNQLYEVLLSKMKKFN
    KKGMADPSDRIEKSKPKFIKLE
    DLIDKINVINKMLNLLRCDNDT
    KADLSLIELPKNAGSFVVKKNT
    IGKSKIILVNQSVTGLYENRRE
    L (SEQ ID NO: 13)
    Oenococcus MARDYSVGLDIGTSSVGWAAID 169 317 149 555 639 80 555 639 80
    kitaharae NKYHLIRAKSKNLIGVRLFDSA
    DSM 17330 VTAEKRRGYRTTRRRLSRRHWR
    gi|366983953| LRLLNDIFAGPLTDFGDENFLA
    gb|EHN59352.1| RLKYSWVHPQDQSNQAHFAAGL
    LFDSKEQDKDFYRKYPTIYHLR
    LALMNDDQKHDLREVYLAIHHL
    VKYRGHFLIEGDVKADSAFDVH
    TFADAIQRYAESNNSDENLLGK
    IDEKKLSAALTDKHGSKSQRAE
    TAETAFDILDLQSKKQIQAILK
    SVVGNQANLMAIFGLDSSAISK
    DEQKNYKFSFDDADIDEKIADS
    EALLSDTEFEFLCDLKAAFDGL
    TLKMLLGDDKTVSAAMVRRFNE
    HQKDWEYIKSHIRNAKNAGNGL
    YEKSKKFDGINAAYLALQSDNE
    DDRKKAKKIFQDEISSADIPDD
    VKADFLKKIDDDQFLPIQRTKN
    NGTIPHQLHRNELEQIIEKQGI
    YYPFLKDTYQENSHELNKITAL
    INFRVPYYVGPLVEEEQKIADD
    GKNIPDPTNHWMVRKSNDTITP
    WNLSQVVDLDKSGRRFIERLTG
    TDTYLIGEPTLPKNSLLYQKFD
    VLQELNNIRVSGRRLDIRAKQD
    AFEHLFKVQKTVSATNLKDFLV
    QAGYISEDTQIEGLADVNGKNF
    NNALTTYNYLVSVLGREFVENP
    SNEELLEEITELQTVFEDKKVL
    RRQLDQLDGLSDHNREKLSRKH
    YTGWGRISKKLLTTKIVQNADK
    IDNQTFDVPRMNQSIIDTLYNT
    KMNLMEIINNAEDDFGVRAWID
    KQNTTDGDEQDVYSLIDELAGP
    KEIKRGIVQSFRILDDITKAVG
    YAPKRVYLEFARKTQESHLTNS
    RKNQLSTLLKNAGLSELVTQVS
    QYDAAALQNDRLYLYFLQQGKD
    MYSGEKLNLDNLSNYDIDHIIP
    QAYTKDNSLDNRVLVSNITNRR
    KSDSSNYLPALIDKMRPFWSVL
    SKQGLLSKHKFANLTRTRDFDD
    MEKERFTARSLVETRQIIKNVA
    SLIDSHEGGETKAVAIRSSLTA
    DMRRYVDIPKNRDINDYHHAFD
    ALLFSTVGQYTENSGLMKKGQL
    SDSAGNQYNRYIKEWIHAARLN
    AQSQRVNPFGFVVGSMRNAAPG
    KLNPETGEITPEENADWSIADL
    DYLHKVMNFRKITVTRRLKDQK
    GQLYDESRYPSVLHDAKSKASI
    NFDKHKPVDLYGGFSSAKPAYA
    ALIKEKNKFRLVNVLRQWTYSD
    KNSEDYILEQIRGKYPKAEMVL
    SHIPYGQLVKKDGALVTISSAT
    ELHNFEQLWLPLADYKLINTLL
    KTKEDNLVDILHNRLDLPEMTI
    ESAFYKAFDSILSFAFNRYALH
    QNALVKLQAHRDDFNALNYEDK
    QQTLERILDALHASPASSDLKK
    INLSSGEGRLFSPSHFTLADTD
    EFIFQSVTGLFSTQKTVAQLYQ
    ETK (SEQ ID NO: 14)
    Fructobacillus MVYDVGLDIGTGSVGWVALDEN 168 314 147 488 571 76 488 571 76
    fructosus GKLARAKGKNLVGVRLFDTAQT
    KCTC 3544 AADRRGFRTTRRRLSRRKWRLR
    gi|339625081| LLDELFSAEINEIDSSFFQRLK
    ref|ZP_08660870.1 YSYVHPKDEENKAHYYGGYLFP
    TEEETKKFHRSYPTIYHLRQEL
    MAQPNKRFDIREIYLAIHHLVK
    YRGHFLSSQEKITIGSTYNPED
    LANAIEVYADEKGLSWELNNPE
    QLTEIISGEAGYGLNKSMKADE
    ALKLFEFDNNQDKVAIKTLLAG
    LTGNQIDFAKLFGKDISDKDEA
    KLWKLKLDDEALEEKSQTILSQ
    LTDEEIELFHAVVQAYDGFVLI
    GLLNGADSVSAAMVQLYDQHRE
    DRKLLKSLAQKAGLKHKRFSEI
    YEQLALATDEATIKNGISTARE
    LVEESNLSKEVKEDTLRRLDEN
    EFLPKQRTKANSVIPHQLHLAE
    LQKILQNQGQYYPFLLDTFEKE
    DGQDNKIEELLRFRIPYYVGPL
    VTKKDVEHAGGDADNHWVERNE
    GFEKSRVTPWNFDKVFNRDKAA
    RDFIERLTGNDTYLIGEKTLPQ
    NSLRYQLFTVLNELNNVRVNGK
    KFDSKTKADLINDLFKARKTVS
    LSALKDYLKAQGKGDVTITGLA
    DESKFNSSLSSYNDLKKTFDAE
    YLENEDNQETLEKIIEIQTVFE
    DSKIASRELSKLPLDDDQVKKL
    SQTHYTGWGRLSEKLLDSKIID
    ERGQKVSILDKLKSTSQNFMSI
    INNDKYGVQAWITEQNTGSSKL
    TFDEKVNELTTSPANKRGIKQS
    FAVLNDIKKAMKEEPRRVYLEF
    AREDQTSVRSVPRYNQLKEKYQ
    SKSLSEEAKVLKKTLDGNKNKM
    SDDRYFLYFQQQGKDMYTGRPI
    NFERLSQDYDIDHIIPQAFTKD
    DSLDNRVLVSRPENARKSDSFA
    YTDEVQKQDGSLWTSLLKSGFI
    NRKKYERLTKAGKYLDGQKTGF
    IARQLVETRQIIKNVASLIEGE
    YENSKAVAIRSEITADMRLLVG
    IKKHREINSFHHAFDALLITAA
    GQYMQNRYPDRDSTNVYNEFDR
    YTNDYLKNLRQLSSRDEVRRLK
    SFGFVVGTMRKGNEDWSEENTS
    YLRKVMMFKNILTTKKTEKDRG
    PLNKETIFSPKSGKKLIPLNSK
    RSDTALYGGYSNVYSAYMTLVR
    ANGKNLLIKIPISIANQIEVGN
    LKINDYIVNNPAIKKFEKILIS
    KLPLGQLVNEDGNLIYLASNEY
    RHNAKQLWLSTTDADKIASISE
    NSSDEELLEAYDILTSENVKNR
    FPFFKKDIDKLSQVRDEFLDSD
    KRIAVIQTILRGLQIDAAYQAP
    VKIISKKVSDWHKLQQSGGIKL
    SDNSEMIYQSATGIFETRVKIS
    DLL (SEQ ID NO: 15)
    Catenibacterium IVDYCIGLDLGTGSVGWAVVDM 173 318 146 511 594 78 511 594 78
    mitsuokai NHRLMKRNGKHLWGSRLFSNAE
    DSM 15897 TAANRRASRSIRRRYNKRRERI
    gi|224543312| RLLRAILQDMVLEKDPTFFIRL
    ref|ZP_03683851.1 EHTSFLDEEDKAKYLGTDYKDN
    YNLFIDEDFNDYTYYHKYPTIY
    HLRKALCESTEKADPRLIYLAL
    HHIVKYRGNFLYEGQKFNMDAS
    NIEDKLSDIFTQFTSFNNIPYE
    DDEKKNLEILEILKKPLSKKAK
    VDEVMTLIAPEKDYKSAFKELV
    TGIAGNKMNVTKMILCEPIKQG
    DSEIKLKFSDSNYDDQFSEVEK
    DLGEYVEFVDALHNVYSWVELQ
    TIMGATHTDNASISEAMVSRYN
    KHHDDLKLLKDCIKNNVPNKYF
    DMFRNDSEKSKGYYNYINRPSK
    APVDEFYKYVKKCIEKVDTPEA
    KQILNDIELENFLLKQNSRTNG
    SVPYQMQLDEMIKIIDNQAEYY
    PILKEKREQLLSILTFRIPYYF
    GPLNETSEHAWIKRLEGKENQR
    ILPWNYQDIVDVDATAEGFIKR
    MRSYCTYFPDEEVLPKNSLIVS
    KYEVYNELNKIRVDDKLLEVDV
    KNDIYNELFMKNKTVTEKKLKN
    WLVNNQCCSKDAEIKGFQKENQ
    FSTSLTPWIDFTNIFGKIDQSN
    FDLIENIIYDLTVFEDKKIMKR
    RLKKKYALPDDKVKQILKLKYK
    DWSRLSKKLLDGIVADNRFGSS
    VTVLDVLEMSRLNLMEIINDKD
    LGYAQMIEEATSCPEDGKFTYE
    EVERLAGSPALKRGIWQSLQIV
    EEITKVMKCRPKYIYIEFERSE
    EAKERTESKIKKLENVYKDLDE
    QTKKEYKSVLEELKGFDNTKKI
    SSDSLFLYFTQLGKCMYSGKKL
    DIDSLDKYQIDHIVPQSLVKDD
    SFDNRVLVVPSENQRKLDDLVV
    PFDIRDKMYRFWKLLFDHELIS
    PKKFYSLIKTEYTERDEERFIN
    RQLVETRQITKNVTQIIEDHYS
    TTKVAAIRANLSHEFRVKNHIY
    KNRDINDYHHAHDAYIVALIGG
    FMRDRYPNMHDSKAVYSEYMKM
    FRKNKNDQKRWKDGFVINSMNY
    PYEVDGKLIWNPDLINEIKKCF
    YYKDCYCTTKLDQKSGQLFNLT
    VLSNDAHADKGVTKAVVPVNKN
    RSDVHKYGGFSGLQYTIVAIEG
    QKKKGKKTELVKKISGVPLHLK
    AASINEKINYIEEKEGLSDVRI
    IKDNIPVNQMIEMDGGEYLLTS
    PTEYVNARQLVLNEKQCALIAD
    IYNAIYKQDYDNLDDILMIQLY
    IELTNKMKVLYPAYRGIAEKFE
    SMNENYVVISKEEKANIIKQML
    IVMHRGPQNGNIVYDDFKISDR
    IGRLKTKNHNLNNIVFISQSPT
    GIYTKKYKL (SEQ ID NO:
    16)
    Finegoldia MKSEKKYYIGLDVGTNSVGWAV 168 313 146 452 534 77 452 534 77
    magna ATCC TDEFYNILRAKGKDLWGVRLFE
    29328 KADTAANTRIFRSGRRRNDRKG
    gi|169823755| MRLQILREIFEDEIKKVDKDFY
    ref|YP_001691366.1 DRLDESKFWAEDKKVSGKYSLF
    NDKNFSDKQYFEKFPTIFHLRK
    YLMEEHGKVDIRYYFLAINQMM
    KRRGHFLIDGQISHVTDDKPLK
    EQLILLINDLLKIELEEELMDS
    IFEILADVNEKRTDKKNNLKEL
    IKGQDFNKQEGNILNSIFESIV
    TGKAKIKNIISDEDILEKIKED
    NKEDFVLTGDSYEENLQYFEEV
    LQENITLFNTLKSTYDFLILQS
    ILKGKSTLSDAQVERYDEHKKD
    LEILKKVIKKYDEDGKLFKQVF
    KEDNGNGYVSYIGYYLNKNKKI
    TAKKKISNIEFTKYVKGILEKQ
    CDCEDEDVKYLLGKIEQENFLL
    KQISSINSVIPHQIHLFELDKI
    LENLAKNYPSFNNKKEEFTKIE
    KIRKTFTFRIPYYVGPLNDYHK
    NNGGNAWIFRNKGEKIRPWNFE
    KIVDLHKSEEEFIKRMLNQCTY
    LPEETVLPKSSILYSEYMVLNE
    LNNLRINGKPLDTDVKLKLIEE
    LFKKKTKVTLKSIRDYMVRNNF
    ADKEDFDNSEKNLEIASNMKSY
    IDFNNILEDKFDVEMVEDLIEK
    ITIHTGNKKLLKKYIEETYPDL
    SSSQIQKIINLKYKDWGRLSRK
    LLDGIKGTKKETEKTDTVINFL
    RNSSDNLMQIIGSQNYSFNEYI
    DKLRKKYIPQEISYEVVENLYV
    SPSVKKMIWQVIRVTEEITKVM
    GYDPDKIFIEMAKSEEEKKTTI
    SRKNKLLDLYKAIKKDERDSQY
    EKLLTGLNKLDDSDLRSRKLYL
    YYTQMGRDMYTGEKIDLDKLFD
    STHYDKDHIIPQSMKKDDSIIN
    NLVLVNKNANQTTKGNIYPVPS
    SIRNNPKIYNYWKYLMEKEFIS
    KEKYNRLIRNTPLTNEELGGFI
    NRQLVETRQSTKAIKELFEKFY
    QKSKIIPVKASLASDLRKDMNT
    LKSREVNDLHHAHDAFLNIVAG
    DVWNREFTSNPINYVKENREGD
    KVKYSLSKDFTRPRKSKGKVIW
    TPEKGRKLIVDTLNKPSVLISN
    ESHVKKGELFNATIAGKKDYKK
    GKIYLPLKKDDRLQDVSKYGGY
    KAINGAFFFLVEHTKSKKRIRS
    IELFPLHLLSKFYEDKNTVLDY
    AINVLQLQDPKIIIDKINYRTE
    IIIDNFSYLISTKSNDGSITVK
    PNEQMYWRVDEISNLKKIENKY
    KKDAILTEEDRKIMESYIDKIY
    QQFKAGKYKNRRTTDTIIEKYE
    IIDLDTLDNKQLYQLLVAFISL
    SYKTSNNAVDFTVIGLGTECGK
    PRITNLPDNTYLVYKSITGIYE
    KRIRIK (SEQ ID NO: 17)
    Coriobacterium MKLRGIEDDYSIGLDMGTSSVG 175 318 144 511 592 82 511 592 82
    glomeran WAVTDERGTLAHFKRKPTWGSR
    sPW2 LFREAQTAAVARMPRGQRRRYV
    gi|328956315| RRRWRLDLLQKLFEQQMEQADP
    ref|YP_004373648.1 DFFIRLRQSRLLRDDRAEEHAD
    YRWPLFNDCKFTERDYYQRFPT
    IYHVRSWLMETDEQADIRLIYL
    ALHNIVKHRGNFLREGQSLSAK
    SARPDEALNHLRETLRVWSSER
    GFECSIADNGSILAMLTHPDLS
    PSDRRKKIAPLFDVKSDDAAAD
    KKLGIALAGAVIGLKTEFKNIF
    GDFPCEDSSIYLSNDEAVDAVR
    SACPDDCAELFDRLCEVYSAYV
    LQGLLSYAPGQTISANMVEKYR
    RYGEDLALLKKLVKIYAPDQYR
    MFFSGATYPGTGIYDAAQARGY
    TKYNLGPKKSEYKPSESMQYDD
    FRKAVEKLFAKTDARADERYRM
    MMDRFDKQQFLRRLKTSDNGSI
    YHQLHLEELKAIVENQGRFYPF
    LKRDADKLVSLVSFRIPYYVGP
    LSTRNARTDQHGENRFAWSERK
    PGMQDEPIFPWNWESIIDRSKS
    AEKFILRMTGMCTYLQQEPVLP
    KSSLLYEEFCVLNELNGAHWSI
    DGDDEHRFDAADREGIIEELFR
    RKRTVSYGDVAGWMERERNQIG
    AHVCGGQGEKGFESKLGSYIFF
    CKDVFKVERLEQSDYPMIERII
    LWNTLFEDRKILSQRLKEEYGS
    RLSAEQIKTICKKRFTGWGRLS
    EKFLTGITVQVDEDSVSIMDVL
    REGCPVSGKRGRAMVMMEILRD
    EELGFQKKVDDFNRAFFAENAQ
    ALGVNELPGSPAVRRSLNQSIR
    IVDEIASIAGKAPANIFIEVTR
    DEDPKKKGRRTKRRYNDLKDAL
    EAFKKEDPELWRELCETAPNDM
    DERLSLYFMQRGKCLYSGRAID
    IHQLSNAGIYEVDHIIPRTYVK
    DDSLENKALVYREENQRKTDML
    LIDPEIRRRMSGYWRMLHEAKL
    IGDKKFRNLLRSRIDDKALKGF
    IARQLVETGQMVKLVRSLLEAR
    YPETNIISVKASISHDLRTAAE
    LVKCREANDFHHAHDAFLACRV
    GLFIQKRHPCVYENPIGLSQVV
    RNYVRQQADIFKRCRTIPGSSG
    FIVNSFMTSGFDKETGEIFKDD
    WDAEAEVEGIRRSLNFRQCFIS
    RMPFEDHGVFWDATIYSPRAKK
    TAALPLKQGLNPSRYGSFSREQ
    FAYFFIYKARNPRKEQTLFEFA
    QVPVRLSAQIRQDENALERYAR
    ELAKDQGLEFIRIERSKILKNQ
    LIEIDGDRLCITGKEEVRNACE
    LAFAQDEMRVIRMLVSEKPVSR
    ECVISLFNRILLHGDQASRRLS
    KQLKLALLSEAFSEASDNVQRN
    VVLGLIAIFNGSTNMVNLSDIG
    GSKFAGNVRIKYKKELASPKVN
    VHLIDQSVTGMFERRTKIGL
    (SEQ ID NO: 18)
    Eubacterium MENKQYYIGLDVGTNSVGWAVT 169 310 142 552 633 76 552 633 76
    yurii ATCC DTSYNLLRAKGKDMWGARLFEK
    43715 ANTAAERRTKRTSRRRSEREKA
    gi|306821691| RKAMLKELFADEINRVDPSFFI
    ref|ZP_07455288.1 RLEESKFFLDDRSENNRQRYTL
    FNDATFTDKDYYEKYKTIFHLR
    SALINSDEKFDVRLVFLAILNL
    FSHRGHFLNASLKGDGDIQGMD
    VFYNDLVESCEYFEIELPRITN
    IDNFEKILSQKGKSRTKILEEL
    SEELSISKKDKSKYNLIKLISG
    LEASVVELYNIEDIQDENKKIK
    IGFRESDYEESSLKVKEIIGDE
    YFDLVERAKSVHDMGLLSNIIG
    NSKYLCEARVEAYENHHKDLLK
    IKELLKKYDKKAYNDMFRKMTD
    KNYSAYVGSVNSNIAKERRSVD
    KRKIEDLYKYIEDTALKNIPDD
    NKDKIEILEKIKLGEFLKKQLT
    ASNGVIPNQLQSRELRAILKKA
    ENYLPFLKEKGEKNLTVSEMII
    QLFEFQIPYYVGPLDKNPKKDN
    KANSWAKIKQGGRILPWNFEDK
    VDVKGSRKEFIEKMVRKCTYIS
    DEHTLPKQSLLYEKFMVLNEIN
    NIKIDGEKISVEAKQKIYNDLF
    VKGKKVSQKDIKKELISLNIMD
    KDSVLSGTDTVCNAYLSSIGKF
    TGVFKEEINKQSIVDMIEDIIF
    LKTVYGDEKRFVKEEIVEKYGD
    EIDKDKIKRILGFKFSNWGNLS
    KSFLELEGADVGTGEVRSIIQS
    LWETNFNLMELLSSRFTYMDEL
    EKRVKKLEKPLSEWTIEDLDDM
    YLSSPVKRMIWQSMKIVDEIQT
    VIGYAPKRIFVEMTRSEGEKVR
    TKSRKDRLKELYNGIKEDSKQW
    VKELDSKDESYFRSKKMYLYYL
    QKGRCMYSGEVIELDKLMDDNL
    YDIDHIYPRSFVKDDSLDNLVL
    VKKEINNRKQNDPITPQIQASC
    QGFWKILHDQGFMSNEKYSRLT
    RKTQEFSDEEKLSFINRQIVET
    GQATKCMAQILQKSMGEDVDVV
    FSKARLVSEFRHKFELFKSRLI
    NDFHHANDAYLNIVVGNSYFVK
    FTRNPANFIKDARKNPDNPVYK
    YHMDRFFERDVKSKSEVAWIGQ
    SEGNSGTIVIVKKTMAKNSPLI
    TKKVEEGHGSITKETIVGVKEI
    KFGRNKVEKADKTPKKPNLQAY
    RPIKTSDERLCNILRYGGRTSI
    SISGYCLVEYVKKRKTIRSLEA
    IPVYLGRKDSLSEEKLLNYFRY
    NLNDGGKDSVSDIRLCLPFIST
    NSLVKIDGYLYYLGGKNDDRIQ
    LYNAYQLKMKKEEVEYIRKIEK
    AVSMSKFDEIDREKNPVLTEEK
    NIELYNKIQDKFENTVFSKRMS
    LVKYNKKDLSFGDFLKNKKSKF
    EEIDLEKQCKVLYNIIFNLSNL
    KEVDLSDIGGSKSTGKCRCKKN
    ITNYKEFKLIQQSITGLYSCEK
    DLMTI (SEQ ID NO: 19)
    Peptoniphilus MKNLKEYYIGLDIGTASVGWAV 171 311 141 535 615 76 535 615 76
    duerdenii TDESYNIPKFNGKKMWGVRLFD
    ATCC BAA- DAKTAEERRTQRGSRRRLNRRK
    1640 ERINLLQDLFATEISKVDPNFF
    gi|304438954| LRLDNSDLYREDKDEKLKSKYT
    ref|ZP_07398877.1 LFNDKDFKDRDYHKKYPTIHHL
    IMDLIEDEGKKDIRLLYLACHY
    LLKNRGHFIFEGQKFDTKNSFD
    KSINDLKIHLRDEYNIDLEFNN
    EDLIEIITDTTLNKTNKKKELK
    NIVGDTKFLKAISAIMIGSSQK
    LVDLFEDGEFEETTVKSVDFST
    TAFDDKYSEYEEALGDTISLLN
    ILKSIYDSSILENLLKDADKSK
    DGNKYISKAFVKKFNKHGKDLK
    TLKRIIKKYLPSEYANIFRNKS
    INDNYVAYTKSNITSNKRTKAS
    KFTKQEDFYKFIKKHLDTIKET
    KLNSSENEDLKLIDEMLTDIEF
    KTFIPKLKSSDNGVIPYQLKLM
    ELKKILDNQSKYYDFLNESDEY
    GTVKDKVESIMEFRIPYYVGPL
    NPDSKYAWIKRENTKITPWNFK
    DIVDLDSSREEFIDRLIGRCTY
    LKEEKVLPKASLIYNEFMVLNE
    LNNLKLNEFLITEEMKKAIFEE
    LFKTKKKVTLKAVSNLLKKEFN
    LTGDILLSGTDGDFKQGLNSYI
    DFKNIIGDKVDRDDYRIKIEEI
    IKLIVLYEDDKTYLKKKIKSAY
    KNDFTDDEIKKIAALNYKDWGR
    LSKRFLTGIEGVDKTTGEKGSI
    IYFMREYNLNLMELMSGHYTFT
    EEVEKLNPVENRELCYEMVDEL
    YLSPSVKRMLWQSLRVVDEIKR
    IIGKDPKKIFIEMARAKEAKNS
    RKESRKNKLLEFYKFGKKAFIN
    EIGEERYNYLLNEINSEEESKF
    RWDNLYLYYTQLGRCMYSLEPI
    DLADLKSNNIYDQDHIYPKSKI
    YDDSLENRVLVKKNLNHEKGNQ
    YPIPEKVLNKNAYGFWKILFDK
    GLIGQKKYTRLTRRTPFEEREL
    AEFIERQIVETRQATKETANLL
    KNICQDSEIVYSKAENASRFRQ
    EFDIIKCRTVNDLHHMHDAYLN
    IVVGNVYNTKFTKNPLNFIKDK
    DNVRSYNLENMFKYDVVRGSYT
    AWIADDSEGNVKAATIKKVKRE
    LEGKNYRFTRMSYIGTGGLYDQ
    NLMRKGKGQIPQKENTNKSNIE
    KYGGYNKASSAYFALIESDGKA
    GRERTLETIPIMVYNQEKYGNT
    EAVDKYLKDNLELQDPKILKDK
    IKINSLIKLDGFLYNIKGKTGD
    SLSIAGSVQLIVNKEEQKLIKK
    MDKFLVKKKDNKDIKVTSFDNI
    KEEELIKLYKTLSDKLNNGIYS
    NKRNNQAKNISEALDKFKEISI
    EEKIDVLNQIILLFQSYNNGCN
    LKSIGLSAKTGVVFIPKKLNYK
    ECKLINQSITGLFENEVDLLNL
    (SEQ ID NO: 20)
    Acidaminococcus MGKMYYLGLDIGTNSVGYAVTD 167 306 140 511 591 75 511 591 75
    sp. D21 PSYHLLKFKGEPMWGAHVFAAG
    gi|227824983| NQSAERRSFRTSRRRLDRRQQR
    ref|ZP_03989815.1 VKLVQEIFAPVISPIDPRFFIR
    LHESALWRDDVAETDKHIFFND
    PTYTDKEYYSDYPTIHHLIVDL
    MESSEKHDPRLVYLAVAWLVAH
    RGHFLNEVDKDNIGDVLSFDAF
    YPEFLAFLSDNGVSPWVCESKA
    LQATLLSRNSVNDKYKALKSLI
    FGSQKPEDNFDANISEDGLIQL
    LAGKKVKVNKLFPQESNDASFT
    LNDKEDAIEEILGTLTPDECEW
    IAHIRRLFDWAIMKHALKDGRT
    ISESKVKLYEQHHHDLTQLKYF
    VKTYLAKEYDDIFRNVDSETTK
    NYVAYSYHVKEVKGTLPKNKAT
    QEEFCKYVLGKVKNIECSEADK
    VDFDEMIQRLTDNSFMPKQVSG
    ENRVIPYQLYYYELKTILNKAA
    SYLPFLTQCGKDAISNQDKLLS
    IMTFRIPYFVGPLRKDNSEHAW
    LERKAGKIYPWNFNDKVDLDKS
    EEAFIRRMTNTCTYYPGEDVLP
    LDSLIYEKFMILNEINNIRIDG
    YPISVDVKQQVFGLFEKKRRVT
    VKDIQNLLLSLGALDKHGKLTG
    IDTTIHSNYNTYHHFKSLMERG
    VLTRDDVERIVERMTYSDDTKR
    VRLWLNNNYGTLTADDVKHISR
    LRKHDFGRLSKMFLTGLKGVHK
    ETGERASILDFMWNTNDNLMQL
    LSECYTFSDEITKLQEAYYAKA
    QLSLNDFLDSMYISNAVKRPIY
    RTLAVVNDIRKACGTAPKRIFI
    EMARDGESKKKRSVTRREQIKN
    LYRSIRKDFQQEVDFLEKILEN
    KSDGQLQSDALYLYFAQLGRDM
    YTGDPIKLEHIKDQSFYNIDHI
    YPQSMVKDDSLDNKVLVQSEIN
    GEKSSRYPLDAAIRNKMKPLWD
    AYYNHGLISLKKYQRLTRSTPF
    TDDEKWDFINRQLVETRQSTKA
    LAILLKRKFPDTEIVYSKAGLS
    SDFRHEFGLVKSRNINDLHHAK
    DAFLAIVTGNVYHERFNRRWFM
    VNQPYSVKTKTLFTHSIKNGNF
    VAWNGEEDLGRIVKMLKQNKNT
    IHFTRFSFDRKEGLFDIQPLKA
    STGLVPRKAGLDVVKYGGYDKS
    TAAYYLLVRFTLEDKKTQHKLM
    MIPVEGLYKARIDHDKEFLTDY
    AQTTISEILQKDKQKVINIMFP
    MGTRHIKLNSMISIDGFYLSIG
    GKSSKGKSVLCHAMVPLIVPHK
    IECYIKAMESFARKFKENNKLR
    IVEKFDKITVEDNLNLYELFLQ
    KLQHNPYNKFFSTQFDVLTNGR
    STFTKLSPEEQVQTLLNILSIF
    KTCRSSGCDLKSINGSAQAARI
    MISADLTGLSKKYSDIRLVEQS
    ASGLFVSKSQNLLEYL (SEQ
    ID NO: 21)
    Lactobacillus MTKKEQPYNIGLDIGTSSVGWA 171 310 140 542 621 85 542 621 85
    farciminis VTNDNYDLLNIKKKNLWGVRLF
    KCTC 3681 EEAQTAKETRLNRSTRRRYRRR
    gi|336394882| KNRINWLNEIFSEELAKTDPSF
    ref|ZP_08576281.1 LIRLQNSWVSKKDPDRKRDKYN
    LFIDGPYTDKEYYREFPTIFHL
    RKELILNKDKADIRLIYLALHN
    ILKYRGNFTYEHQKFNISNLNN
    NLSKELIELNQQLIKYDISFPD
    DCDWNHISDILIGRGNATQKSS
    NILKDFTLDKETKKLLKEVINL
    ILGNVAHLNTIFKTSLTKDEEK
    LNFSGKDIESKLDDLDSILDDD
    QFTVLDAANRIYSTITLNEILN
    GESYFSMAKVNQYENHAIDLCK
    LRDMWHTTKNEEAVEQSRQAYD
    DYINKPKYGTKELYTSLKKFLK
    VALPTNLAKEAEEKISKGTYLV
    KPRNSENGVVPYQLNKIEMEKI
    IDNQSQYYPFLKENKEKLLSIL
    SFRIPYYVGPLQSAEKNPFAWM
    ERKSNGHARPWNFDEIVDREKS
    SNKFIRRMTVTDSYLVGEPVLP
    KNSLIYQRYEVLNELNNIRITE
    NLKTNPIGSRLTVETKQRIYNE
    LFKKYKKVTVKKLTKWLIAQGY
    YKNPILIGLSQKDEFNSTLTTY
    LDMKKIFGSSFMEDNKNYDQIE
    ELIEWLTIFEDKQILNEKLHSS
    KYSYTPDQIKKISNMRYKGWGR
    LSKKILMDITTETNTPQLLQLS
    NYSILDLMWATNNNFISIMSND
    KYDFKNYIENHNLNKNEDQNIS
    DLVNDIHVSPALKRGITQSIKI
    VQEIVKFMGHAPKHIFIEVTRE
    TKKSEITTSREKRIKRLQSKLL
    NKANDFKPQLREYLVPNKKIQE
    ELKKHKNDLSSERIMLYFLQNG
    KSLYSEESLNINKLSDYQVDHI
    LPRTYIPDDSLENKALVLAKEN
    QRKADDLLLNSNVIDRNLERWT
    YMLNNNMIGLKKFKNLTRRVIT
    DKDKLGFIHRQLVQTSQMVKGV
    ANILDNMYKNQGTTCIQARANL
    STAFRKALSGQDDTYHFKHPEL
    VKNRNVNDFHHAQDAYLASFLG
    TYRLRRFPTNEMLLMNGEYNKF
    YGQVKELYSKKKKLPDSRKNGF
    IISPLVNGTTQYDRNTGEIIWN
    VGFRDKILKIFNYHQCNVTRKT
    EIKTGQFYDQTIYSPKNPKYKK
    LIAQKKDMDPNIYGGFSGDNKS
    SITIVKIDNNKIKPVAIPIRLI
    NDLKDKKTLQNWLEENVKHKKS
    IQIIKNNVPIGQIIYSKKVGLL
    SLNSDREVANRQQLILPPEHSA
    LLRLLQIPDEDLDQILAFYDKN
    ILVEILQELITKMKKFYPFYKG
    EREFLIANIENFNQATTSEKVN
    SLEELITLLHANSTSAHLIFNN
    IEKKAFGRKTHGLTLNNTDFIY
    QSVTGLYETRIHIE (SEQ ID
    NO: 22)
    Streptococcus MTKFNKNYSIGLDIGVSSVGYA 185 324 140 411 490 85 411 490 85
    sanguinis VVTEDYRVPAFKFKVLGNTEKE
    SK49 KIKKNLIGSTTFVSAQPAKGTR
    gi|422884106| VFRVNRRRIDRRNHRITYLRDI
    ref|ZP_16930555.1 FQKEIEKVDKNFYRRLDESFRV
    LGDKSEDLQIKQPFFGDKELET
    AYHKKYPTIYHLRKHLADADKN
    SPVADIREVYMAISHILKYRGH
    FLTLDKINPNNINMQNSWIDFI
    ESCQEVFDLEISDESKNIADIF
    KSSENRQEKVKKILPYFQQELL
    KKDKSIFKQLLQLLFGLKTKFK
    DCFELEEEPDLNFSKENYDENL
    ENFLGSLEEDFSDVFAKLKVLR
    DTILLSGMLTYTGATHARFSAT
    MVERYEEHRKDLQRFKFFIKQN
    LSEQDYLDIFGRKTQNGFDVDK
    ETKGYVGYITNKMVLTNPQKQK
    TIQQNFYDYISGKITGIEGAEY
    FLNKISDGTFLRKLRTSDNGAI
    PNQIHAYELEKIIERQGKDYPF
    LLENKDKLLSILTFKIPYYVGP
    LAKGSNSRFAWIKRATSSDILD
    DNDEDTRNGKIRPWNYQKLINM
    DETRDAFITNLIGNDIILLNEK
    VLPKRSLIYEEVMLQNELTRVK
    YKDKYGKAHFFDSELRQNIING
    LFKNNSKRVNAKSLIKYLSDNH
    KDLNAIEIVSGVEKGKSFNSTL
    KTYNDLKTIFSEELLDSEIYQK
    ELEEIIKVITVFDDKKSIKNYL
    TKFFGHLEILDEEKINQLSKLR
    YSGWGRYSAKLLLDIRDEDTGF
    NLLQFLRNDEENRNLTKLISDN
    TLSFEPKIKDIQSKSTIEDDIF
    DEIKKLAGSPAIKRGILNSIKI
    VDELVQIIGYPPHNIVIEMARE
    NMTTEEGQKKAKTRKTKLESAL
    KNIENSLLENGKVPHSDEQLQS
    EKLYLYYLQNGKDMYTLDKTGS
    PAPLYLDQLDQYEVDHIIPYSF
    LPIDSIDNKVLTHRENNQQKLN
    NIPDKETVANMKPFWEKLYNAK
    LISQTKYQRLTTSERTPDGVLT
    ESMKAGFIERQLVETRQIIKHV
    ARILDNRFSDTKIITLKSQLIT
    NFRNTFHIAKIRELNDYHHAHD
    AYLAVVVGQTLLKVYPKLAPEL
    IYGHHAHFNRHEENKATLRKHL
    YSNIMRFFNNPDSKVSKDIWDC
    NRDLPIIKDVIYNSQINFVKRT
    MIKKGAFYNQNPVGKFNKQLAA
    NNRYPLKTKALCLDTSIYGGYG
    PMNSALSIIIIAERFNEKKGKI
    ETVKEFHDIFIIDYEKFNNNPF
    QFLNDTSENGFLKKNNINRVLG
    FYRIPKYSLMQKIDGTRMLFES
    KSNLHKATQFKLTKTQNELFFH
    MKRLLTKSNLMDLKSKSAIKES
    QNFILKHKEEFDNISNQLSAFS
    QKMLGNTTSLKNLIKGYNERKI
    KEIDIRDETIKYFYDNFIKMFS
    FVKSGAPKDINDFFDNKCTVAR
    MRPKPDKKLLNATLIHQSITGL
    YETRIDLSKLGED (SEQ ID
    NO: 23)
    Coprococcus MKQEYFLGLDMGTGSLGWAVTD 172 310 139 556 634 76 556 634 76
    catus GD-7 STYQVMRKHGKALWGTRLFESA
    gi|291520705| STAEERRMFRTARRRLDRRNWR
    emb|CBK78998.1| IQVLQEIFSEEISKVDPGFFLR
    MKESKYYPEDKRDAEGNCPELP
    YALFVDDNYTDKNYHKDYPTIY
    HLRKMLMETTEIPDIRLVYLVL
    HHMMKHRGHFLLSGDISQIKEF
    KSTFEQLIQNIQDEELEWHISL
    DDAAIQFVEHVLKDRNLTRSTK
    KSRLIKQLNAKSACEKAILNLL
    SGGTVKLSDIFNNKELDESERP
    KVSFADSGYDDYIGIVEAELAE
    QYYIIASAKAVYDWSVLVEILG
    NSVSISEAKIKVYQKHQADLKT
    LKKIVRQYMTKEDYKRVFVDTE
    EKLNNYSAYIGMTKKNGKKVDL
    KSKQCTQADFYDFLKKNVIKVI
    DHKEITQEIESEIEKENFLPKQ
    VTKDNGVIPYQVHDYELKKILD
    NLGTRMPFIKENAEKIQQLFEF
    RIPYYVGPLNRVDDGKDGKFTW
    SVRKSDARIYPWNFTEVIDVEA
    SAEKFIRRMTNKCTYLVGEDVL
    PKDSLVYSKFMVLNELNNLRLN
    GEKISVELKQRIYEELFCKYRK
    VTRKKLERYLVIEGIAKKGVEI
    TGIDGDFKASLTAYHDFKERLT
    DVQLSQRAKEAIVLNVVLFGDD
    KKLLKQRLSKMYPNLTTGQLKG
    ICSLSYQGWGRLSKTFLEEITV
    PAPGTGEVWNIMTALWQTNDNL
    MQLLSRNYGFTNEVEEFNTLKK
    ETDLSYKTVDELYVSPAVKRQI
    WQTLKVVKEIQKVMGNAPKRVF
    VEMAREKQEGKRSDSRKKQLVE
    LYRACKNEERDWITELNAQSDQ
    QLRSDKLFLYYIQKGRCMYSGE
    TIQLDELWDNTKYDIDHIYPQS
    KTMDDSLNNRVLVKKNYNAIKS
    DTYPLSLDIQKKMMSFWKMLQQ
    QGFITKEKYVRLVRSDELSADE
    LAGFIERQIVETRQSTKAVATI
    LKEALPDTEIVYVKAGNVSNFR
    QTYELLKVREMNDLHHAKDAYL
    NIVVGNAYFVKFTKNAAWFIRN
    NPGRSYNLKRMFEFDIERSGEI
    AWKAGNKGSIVTVKKVMQKNNI
    LVTRKAYEVKGGLFDQQIMKKG
    KGQVPIKGNDERLADIEKYGGY
    NKAAGTYFMLVKSLDKKGKEIR
    TIEFVPLYLKNQIEINHESAIQ
    YLAQERGLNSPEILLSKIKIDT
    LFKVDGFKMWLSGRTGNQLIFK
    GANQLILSHQEAAILKGVVKYV
    NRKNENKDAKLSERDGMTEEKL
    LQLYDTFLDKLSNTVYSIRLSA
    QIKTLTEKRAKFIGLSNEDQCI
    VLNEILHMFQCQSGSANLKLIG
    GPGSAGILVMNNNITACKQISV
    INQSPTGIYEKEIDLIKL
    (SEQ ID NO: 24)
    Streptococcus MKKPYSIGLDIGTNSVGWAVVT 176 314 139 392 470 84 392 470 84
    mutans DDYKVPAKKMKVLGNTDKSHIE
    UA159 KNLLGALLFDSGNTAEDRRLKR
    gi|24379809| TARRRYTRRRNRILYLQEIFSE
    ref|NP_721764.1| EMGKVDDSFFHRLEDSFLVTED
    KRGERHPIFGNLEEEVKYHENF
    PTIYHLRQYLADNPEKVDLRLV
    YLALAHIIKFRGHFLIEGKFDT
    RNNDVQRLFQEFLAVYDNTFEN
    SSLQEQNVQVEEILTDKISKSA
    KKDRVLKLFPNEKSNGRFAEFL
    KLIVGNQADFKKHFELEEKAPL
    QFSKDTYEEELEVLLAQIGDNY
    AELFLSAKKLYDSILLSGILTV
    TDVGTKAPLSASMIQRYNEHQM
    DLAQLKQFIRQKLSDKYNEVFS
    DVSKDGYAGYIDGKTNQEAFYK
    YLKGLLNKIEGSGYFLDKIERE
    DFLRKQRTFDNGSIPHQIHLQE
    MRAIIRRQAEFYPFLADNQDRI
    EKLLTFRIPYYVGPLARGKSDF
    AWLSRKSADKITPWNFDEIVDK
    ESSAEAFINRMTNYDLYLPNQK
    VLPKHSLLYEKFTVYNELTKVK
    YKTEQGKTAFFDANMKQEIFDG
    VFKVYRKVTKDKLMDFLEKEFD
    EFRIVDLTGLDKENKVFNASYG
    TYHDLCKILDKDFLDNSKNEKI
    LEDIVLTLTLFEDREMIRKRLE
    NYSDLLTKEQVKKLERRHYTGW
    GRLSAELIHGIRNKESRKTILD
    YLIDDGNSNRNFMQLINDDALS
    FKEEIAKAQVIGETDNLNQVVS
    DIAGSPAIKKGILQSLKIVDEL
    VKIMGHQPENIVVEMARENQFT
    NQGRRNSQQRLKGLTDSIKEFG
    SQILKEHPVENSQLQNDRLFLY
    YLQNGRDMYTGEELDIDYLSQY
    DIDHIIPQAFIKDNSIDNRVLT
    SSKENRGKSDDVPSKDVVRKMK
    SYWSKLLSAKLITQRKFDNLTK
    AERGGLTDDDKAGFIKRQLVET
    RQITKHVARILDERFNTETDEN
    NKKIRQVKIVTLKSNLVSNFRK
    EFELYKVREINDYHHAHDAYLN
    AVIGKALLGVYPQLEPEFVYGD
    YPHFHGHKENKATAKKFFYSNI
    MNFFKKDDVRTDKNGEIIWKKD
    EHISNIKKVLSYPQVNIVKKVE
    EQTGGFSKESILPKGNSDKLIP
    RKTKKFYWDTKKYGGFDSPIVA
    YSILVIADIEKGKSKKLKTVKA
    LVGVTIMEKMTFERDPVAFLER
    KGYRNVQEENIIKLPKYSLFKL
    ENGRKRLLASARELQKGNEIVL
    PNHLGTLLYHAKNIHKVDEPKH
    LDYVDKHKDEFKELLDVVSNFS
    KKYTLAEGNLEKIKELYAQNNG
    EDLKELASSFINLLTFTAIGAP
    ATFKFFDKNIDRKRYTSTTEIL
    NATLIHQSITGLYETRIDLNKL
    GGD (SEQ ID NO: 25)
    Streptococcus MDKKYSIGLDIGTNSVGWAVIT 176 314 139 523 600 82 523 600 82
    pyogenes DEYKVPSKKFKVLGNTDRHSIK
    M1 GAS KNLIGALLFDSGETAEATRLKR
    gi|13622193| TARRRYTRRKNRICYLQEIFSN
    gb|AAK33936.1| EMAKVDDSFFHRLEESFLVEED
    KKHERHPIFGNIVDEVAYHEKY
    PTIYHLRKKLVDSTDKADLRLI
    YLALAHMIKFRGHFLIEGDLNP
    DNSDVDKLFIQLVQTYNQLFEE
    NPINASGVDAKAILSARLSKSR
    RLENLIAQLPGEKKNGLFGNLI
    ALSLGLTPNFKSNFDLAEDAKL
    QLSKDTYDDDLDNLLAQIGDQY
    ADLFLAAKNLSDAILLSDILRV
    NTEITKAPLSASMIKRYDEHHQ
    DLTLLKALVRQQLPEKYKEIFF
    DQSKNGYAGYIDGGASQEEFYK
    FIKPILEKMDGTEELLVKLNRE
    DLLRKQRTFDNGSIPHQIHLGE
    LHAILRRQEDFYPFLKDNREKI
    EKILTFRIPYYVGPLARGNSRF
    AWMTRKSEETITPWNFEEVVDK
    GASAQSFIERMTNFDKNLPNEK
    VLPKHSLLYEYFTVYNELTKVK
    YVTEGMRKPAFLSGEQKKAIVD
    LLFKTNRKVTVKQLKEDYFKKI
    ECFDSVEISGVEDRFNASLGTY
    HDLLKIIKDKDFLDNEENEDIL
    EDIVLTLTLFEDREMIEERLKT
    YAHLFDDKVMKQLKRRRYTGWG
    RLSRKLINGIRDKQSGKTILDF
    LKSDGFANRNFMQLIHDDSLTF
    KEDIQKAQVSGQGDSLHEHIAN
    LAGSPAIKKGILQTVKVVDELV
    KVMGRHKPENIVIEMARENQTT
    QKGQKNSRERMKRIEEGIKELG
    SQILKEHPVENTQLQNEKLYLY
    YLQNGRDMYVDQELDINRLSDY
    DVDHIVPQSFLKDDSIDNKVLT
    RSDKNRGKSDNVPSEEVVKKMK
    NYWRQLLNAKLITQRKFDNLTK
    AERGGLSELDKAGFIKRQLVET
    RQITKHVAQILDSRMNTKYDEN
    DKLIREVKVITLKSKLVSDFRK
    DFQFYKVREINNYHHAHDAYLN
    AVVGTALIKKYPKLESEFVYGD
    YKVYDVRKMIAKSEQEIGKATA
    KYFFYSNIMNFFKTEITLANGE
    IRKRPLIETNGETGEIVWDKGR
    DFATVRKVLSMPQVNIVKKTEV
    QTGGFSKESILPKRNSDKLIAR
    KKDWDPKKYGGFDSPTVAYSVL
    VVAKVEKGKSKKLKSVKELLGI
    TIMERSSFEKNPIDFLEAKGYK
    EVKKDLIIKLPKYSLFELENGR
    KRMLASAGELQKGNELALPSKY
    VNFLYLASHYEKLKGSPEDNEQ
    KQLFVEQHKHYLDEIIEQISEF
    SKRVILADANLDKVLSAYNKHR
    DKPIREQAENIIHLFTLTNLGA
    PAAFKYFDTTIDRKRYTSTKEV
    LDATLIHQSITGLYETRIDLSQ
    LGGD (SEQ ID NO: 26)
    Streptococcus MTKPYSIGLDIGTNSVGWAVTT 176 314 139 481 558 81 481 558 81
    thermophilus DNYKVPSKKMKVLGNTSKKYIK
    LMD-9 KNLLGVLLFDSGITAEGRRLKR
    gi|116628213| TARRRYTRRRNRILYLQEIFST
    ref|YP_820832.1| EMATLDDAFFQRLDDSFLVPDD
    KRDSKYPIFGNLVEEKAYHDEF
    PTIYHLRKYLADSTKKADLRLV
    YLALAHMIKYRGHFLIEGEFNS
    KNNDIQKNFQDFLDTYNAIFES
    DLSLENSKQLEEIVKDKISKLE
    KKDRILKLFPGEKNSGIFSEFL
    KLIVGNQADFRKCFNLDEKASL
    HFSKESYDEDLETLLGYIGDDY
    SDVFLKAKKLYDAILLSGFLTV
    TDNETEAPLSSAMIKRYNEHKE
    DLALLKEYIRNISLKTYNEVFK
    DDTKNGYAGYIDGKTNQEDFYV
    YLKKLLAEFEGADYFLEKIDRE
    DFLRKQRTFDNGSIPYQIHLQE
    MRAILDKQAKFYPFLAKNKERI
    EKILTFRIPYYVGPLARGNSDF
    AWSIRKRNEKITPWNFEDVIDK
    ESSAEAFINRMTSFDLYLPEEK
    VLPKHSLLYETFNVYNELTKVR
    FIAESMRDYQFLDSKQKKDIVR
    LYFKDKRKVTDKDIIEYLHAIY
    GYDGIELKGIEKQFNSSLSTYH
    DLLNIINDKEFLDDSSNEAIIE
    EIIHTLTIFEDREMIKQRLSKF
    ENIFDKSVLKKLSRRHYTGWGK
    LSAKLINGIRDEKSGNTILDYL
    IDDGISNRNFMQLIHDDALSFK
    KKIQKAQIIGDEDKGNIKEVVK
    SLPGSPAIKKGILQSIKIVDEL
    VKVMGGRKPESIVVEMARENQY
    TNQGKSNSQQRLKRLEKSLKEL
    GSKILKENIPAKLSKIDNNALQ
    NDRLYLYYLQNGKDMYTGDDLD
    IDRLSNYDIDHIIPQAFLKDNS
    IDNKVLVSSASNRGKSDDVPSL
    EVVKKRKTFWYQLLKSKLISQR
    KFDNLTKAERGGLSPEDKAGFI
    QRQLVETRQITKHVARLLDEKF
    NNKKDENNRAVRTVKIITLKST
    LVSQFRKDFELYKVREINDFHH
    AHDAYLNAVVASALLKKYPKLE
    PEFVYGDYPKYNSFRERKSATE
    KVYFYSNIMNIFKKSISLADGR
    VIERPLIEVNEETGESVWNKES
    DLATVRRVLSYPQVNVVKKVEE
    QNHGLDRGKPKGLFNANLSSKP
    KPNSNENLVGAKEYLDPKKYGG
    YAGISNSFTVLVKGTIEKGAKK
    KITNVLEFQGISILDRINYRKD
    KLNFLLEKGYKDIELIIELPKY
    SLFELSDGSRRMLASILSTNNK
    RGEIHKGNQIFLSQKFVKLLYH
    AKRISNTINENHRKYVENHKKE
    FEELFYYILEFNENYVGAKKNG
    KLLNSAFQSWQNHSIDELCSSF
    IGPTGSERKGLFELTSRGSAAD
    FEFLGVKIPRYRDYTPSSLLKD
    ATLIHQSVTGLYETRIDLAKLG
    EG (SEQ ID NO: 27)
    Fusobacterium MKKQKFSDYYLGFDIGTNSVGW 171 308 138 537 614 76 537 614 76
    nucleatum CVTDLDYNVLRFNKKDMWGSRL
    ATCC49256 FDEAKTAAERRVQRNSRRRLKR
    gi|34762592| RKWRLNLLEEIFSDEIMKIDSN
    ref|ZP_00143587.1| FFRRLKESSLWLEDKNSKEKFT
    LFNDDNYKDYDFYKQYPTIFHL
    RDELIKNPEKKDIRLIYLALHS
    IFKSRGHFLFEGQNLKEIKNFE
    TLYNNLISFLEDNGINKSIDKD
    NIEKLEKIICDSGKGLKDKEKE
    FKGIFNSDKQLVAIFKLSVGSS
    VSLNDLFDTDEYKKEEVEKEKI
    SFREQIYEDDKPIYYSILGEKI
    ELLDIAKSFYDFMVLNNILSDS
    NYISEAKVKLYEEHKKDLKNLK
    YIIRKYNKENYDKLFKDKNENN
    YPAYIGLNKEKDKKEVVEKSRL
    KIDDLIKVIKGYLPKPERIEEK
    DKTIFNEILNKIELKTILPKQR
    ISDNGTLPYQIHEVELEKILEN
    QSKYYDFLNYEENGVSTKDKLL
    KTFKFRIPYYVGPLNSYHKDKG
    GNSWIVRKEEGKILPWNFEQKV
    DIEKSAEEFIKRMTNKCTYLNG
    EDVIPKDSFLYSEYIILNELNK
    VQVNDEFLNEENKRKIIDELFK
    ENKKVSEKKFKEYLLVNQIANR
    TVELKGIKDSFNSNYVSYIKFK
    DIFGEKLNLDIYKEISEKSILW
    KCLYGDDKKIFEKKIKNEYGDI
    LNKDEIKKINSFKFNTWGRLSE
    KLLTGIEFINLETGECYSSVME
    ALRRTNYNLMELLSSKFTLQES
    IDNENKEMNEVSYRDLIEESYV
    SPSLKRAILQTLKIYEEIKKIT
    GRVPKKVFIEMARGGDESMKNK
    KIPARQEQLKKLYDSCGNDIAN
    FSIDIKEMKNSLSSYDNNSLRQ
    KKLYLYYLQFGKCMYTGREIDL
    DRLLQNNDTYDIDHIYPRSKVI
    KDDSFDNLVLVLKNENAEKSNE
    YPVKKEIQEKMKSFWRFLKEKN
    FISDEKYKRLTGKDDFELRGFM
    ARQLVNVRQTTKEVGKILQQIE
    PEIKIVYSKAEIASSFREMFDF
    IKVRELNDTHHAKDAYLNIVAG
    NVYNTKFTEKPYRYLQEIKENY
    DVKKIYNYDIKNAWDKENSLEI
    VKKNMEKNTVNITRFIKEEKGE
    LFNLNPIKKGETSNEIISIKPK
    LYDGKDNKLNEKYGYYTSLKAA
    YFIYVEHEKKNKKVKTFERITR
    IDSTLIKNEKNLIKYLVSQKKL
    LNPKIIKKIYKEQTLIIDSYPY
    TFTGVDSNKKVELKNKKQLYLE
    KKYEQILKNALKFVEDNQGETE
    ENYKFIYLKKRNNNEKNETIDA
    VKERYNIEFNEMYDKFLEKLSS
    KDYKNYINNKLYTNFLNSKEKF
    KKLKLWEKSLILREFLKIFNKN
    TYGKYEIKDSQTKEKLFSFPED
    TGRIRLGQSSLGNNKELLEESV
    TGLFVKKIKL (SEQ ID NO:
    28)
    Planococcus MKNYTIGLDIGVASVGWVCIDE 162 299 138 538 614 94 538 614 94
    antarcticus NYKILNYNNRHAFGVHEFESAE
    DSM 14505 SAAGRRLKRGMRRRYNRRKKRL
    gi|389815359| QLLQSLFDSYITDSGFFSKTDS
    ref|ZP_10206685.1 QHFWKNNNEFENRSLTEVLSSL
    RISSRKYPTIYHLRSDLIESNK
    KMDLRLVYLALHNLVKYRGHFL
    QEGNWSEAASAEGMDDQLLELV
    TRYAELENLSPLDLSESQWKAA
    ETLLLNRNLTKTDQSKELTAMF
    GKEYEPFCKLVAGLGVSLHQLF
    PSSEQALAYKETKTKVQLSNEN
    VEEVMELLLEEESALLEAVQPF
    YQQVVLYELLKGETYVAKAKVS
    AFKQYQKDMASLKNLLDKTFGE
    KVYRSYFISDKNSQREYQKSHK
    VEVLCKLDQFNKEAKFAETFYK
    DLKKLLEDKSKTSIGTTEKDEM
    LRIIKAIDSNQFLQKQKGIQNA
    AIPHQNSLYEAEKILRNQQAHY
    PFITTEWIEKVKQILAFRIPYY
    IGPLVKDTTQSPFSWVERKGDA
    PITPWNFDEQIDKAASAEAFIS
    RMRKTCTYLKGQEVLPKSSLTY
    ERFEVLNELNGIQLRTTGAESD
    FRHRLSYEMKCWIIDNVFKQYK
    TVSTKRLLQELKKSPYADELYD
    EHTGEIKEVFGTQKENAFATSL
    SGYISMKSILGAVVDDNPAMTE
    ELIYWIAVFEDREILHLKIQEK
    YPSITDVQRQKLALVKLPGWGR
    FSRLLIDGLPLDEQGQSVLDHM
    EQYSSVFMEVLKNKGFGLEKKI
    QKMNQHQVDGTKKIRYEDIEEL
    AGSPALKRGIWRSVKIVEELVS
    IFGEPANIVLEVAREDGEKKRT
    KSRKDQWEELTKTTLKNDPDLK
    SFIGEIKSQGDQRFNEQRFWLY
    VTQQGKCLYTGKALDIQNLSMY
    EVDHILPQNFVKDDSLDNLALV
    MPEANQRKNQVGQNKMPLEIIE
    ANQQYAMRTLWERLHELKLISS
    GKLGRLKKPSFDEVDKDKFIAR
    QLVETRQIIKHVRDLLDERFSK
    SDIHLVKAGIVSKFRRFSEIPK
    IRDYNNKHHAMDALFAAALIQS
    ILGKYGKNFLAFDLSKKDRQKQ
    WRSVKGSNKEFFLFKNFGNLRL
    QSPVTGEEVSGVEYMKHVYFEL
    PWQTTKMTQTGDGMFYKESIFS
    PKVKQAKYVSPKTEKFVHDEVK
    NHSICLVEFTFMKKEKEVQETK
    FIDLKVIEHHQFLKEPESQLAK
    FLAEKETNSPIIHARIIRTIPK
    YQKIWIEHFPYYFISTRELHNA
    RQFEISYELMEKVKQLSERSSV
    EELKIVFGLLIDQMNDNYPIYT
    KSSIQDRVQKFVDTQLYDFKSF
    EIGFEELKKAVAANAQRSDTFG
    SRISKKPKPEEVAIGYESITGL
    KYRKPRSVVGTKR (SEQ ID
    NO: 29)
    Treponema MKKEIKDYFLGLDVGTGSVGWA 169 305 137 524 600 81 524 600 81
    denticola VTDTDYKLLKANRKDLWGMRCF
    ATCC 35405 ETAETAEVRRLHRGARRRIERR
    gi|42525843| KKRIKLLQELFSQEIAKTDEGF
    ref|NP_970941.1| FQRMKESPFYAEDKTILQENTL
    FNDKDFADKTYHKAYPTINHLI
    KAWIENKVKPDPRLLYLACHNI
    IKKRGHFLFEGDFDSENQFDTS
    IQALFEYLREDMEVDIDADSQK
    VKEILKDSSLKNSEKQSRLNKI
    LGLKPSDKQKKAITNLISGNKI
    NFADLYDNPDLKDAEKNSISFS
    KDDFDALSDDLASILGDSFELL
    LKAKAVYNCSVLSKVIGDEQYL
    SFAKVKIYEKHKTDLTKLKNVI
    KKHFPKDYKKVFGYNKNEKNNN
    NYSGYVGVCKTKSKKLIINNSV
    NQEDFYKFLKTILSAKSEIKEV
    NDILTEIETGTFLPKQISKSNA
    EIPYQLRKMELEKILSNAEKHF
    SFLKQKDEKGLSHSEKIIMLLT
    FKIPYYIGPINDNHKKFFPDRC
    WVVKKEKSPSGKTTPWNFFDHI
    DKEKTAEAFITSRTNFCTYLVG
    ESVLPKSSLLYSEYTVLNEINN
    LQIIIDGKNICDIKLKQKIYED
    LFKKYKKITQKQISTFIKHEGI
    CNKTDEVIILGIDKECTSSLKS
    YIELKNIFGKQVDEISTKNMLE
    EIIRWATIYDEGEGKTILKTKI
    KAEYGKYCSDEQIKKILNLKFS
    GWGRLSRKFLETVTSEMPGFSE
    PVNIITAMRETQNNLMELLSSE
    FTFTENIKKINSGFEDAEKQFS
    YDGLVKPLFLSPSVKKMLWQTL
    KLVKEISHITQAPPKKIFIEMA
    KGAELEPARTKTRLKILQDLYN
    NCKNDADAFSSEIKDLSGKIEN
    EDNLRLRSDKLYLYYTQLGKCM
    YCGKPIEIGHVFDTSNYDIDHI
    YPQSKIKDDSISNRVLVCSSCN
    KNKEDKYPLKSEIQSKQRGFWN
    FLQRNNFISLEKLNRLTRATPI
    SDDETAKFIARQLVETRQATKV
    AAKVLEKMFPETKIVYSKAETV
    SMFRNKFDIVKCREINDFHHAH
    DAYLNIVVGNVYNTKFTNNPWN
    FIKEKRDNPKIADTYNYYKVFD
    YDVKRNNITAWEKGKTIITVKD
    MLKRNTPIYTRQAACKKGELFN
    QTIMKKGLGQHPLKKEGPFSNI
    SKYGGYNKVSAAYYTLIEYEEK
    GNKIRSLETIPLYLVKDIQKDQ
    DVLKSYLTDLLGKKEFKILVPK
    IKINSLLKINGFPCHITGKTND
    SFLLRPAVQFCCSNNEVLYFKK
    IIRFSEIRSQREKIGKTISPYE
    DLSFRSYIKENLWKKTKNDEIG
    EKEFYDLLQKKNLEIYDMLLTK
    HKDTIYKKRPNSATIDILVKGK
    EKFKSLIIENQFEVILEILKLF
    SATRNVSDLQHIGGSKYSGVAK
    IGNKISSLDNCILIYQSITGIF
    EKRIDLLKV (SEQ ID NO:
    30)
    Solobacterium MEGQMKNNGNNLQQGNYYLGLD 179 314 136 544 619 77 544 619 77
    moorei VGTSSVGWAVTDTDYNVLKFRG
    F0204 KSMWGARLFDEASTAEERRTHR
    gi|320528778| GNRRRLARRKYRLLLLEQLFEK
    ref|ZP_08029929.1 EIRKIDDNFFVRLHESNLWADD
    KSKPSKFLLFNDTNFTDKDYLK
    KYPTIYHLRSDLIHNSTEHDIR
    LVFLALHHLIKYRGHFIYDNSA
    NGDVKTLDEAVSDFEEYLNEND
    IEFNIENKKEFINVLSDKHLTK
    KEKKISLKKLYGDITDSENINI
    SVLIEMLSGSSISLSNLFKDIE
    FDGKQNLSLDSDIEETLNDVVD
    ILGDNIDLLIHAKEVYDIAVLT
    SSLGKHKYLCDAKVELFEKNKK
    DLMILKKYIKKNHPEDYKKIFS
    SPTEKKNYAAYSQTNSKNVCSQ
    EEFCLFIKPYIRDMVKSENEDE
    VRIAKEVEDKSFLTKLKGTNNS
    VVPYQIHERELNQILKNIVAYL
    PFMNDEQEDISVVDKIKLIFKF
    KIPYYVGPLNTKSTRSWVYRSD
    EKIYPWNFSNVIDLDKTAHEFM
    NRLIGRCTYTNDPVLPMDSLLY
    SKYNVLNEINPIKVNGKAIPVE
    VKQAIYTDLFENSKKKVTRKSI
    YIYLLKNGYIEKEDIVSGIDIE
    IKSKLKSHHDFTQIVQENKCTP
    EEIERIIKGILVYSDDKSMLRR
    WLKNNIKGLSENDVKYLAKLNY
    KEWGRLSKTLLTDIYTINPEDG
    EACSILDIMWNTNATLMEILSN
    EKYQFKQNIENYKAENYDEKQN
    LHEELDDMYISPAARRSIWQAL
    RIVDEIVDIKKSAPKKIFIEMA
    REKKSAMKKKRTESRKDTLLEL
    YKSCKSQADGFYDEELFEKLSN
    ESNSRLRRDQLYLYYTQMGRSM
    YTGKRIDFDKLINDKNTYDIDH
    IYPRSKIKDDSITNRVLVEKDI
    NGEKTDIYPISEDIRQKMQPFW
    KILKEKGLINEEKYKRLTRNYE
    LTDEELSSFVARQLVETQQSTK
    ALATLLKKEYPSAKIVYSKAGN
    VSEFRNRKDKELPKFREINDLH
    HAKDAYLNIVVGNVYDTKFTEK
    FFNNIRNENYSLKRVFDFSVPG
    AWDAKGSTFNTIKKYMAKNNPI
    IAFAPYEVKGELFDQQIVPKGK
    GQFPIKQGKDIEKYGGYNKLSS
    AFLFAVEYKGKKARERSLETVY
    IKDVELYLQDPIKYCESVLGLK
    EPQIIKPKILMGSLFSINNKKL
    VVTGRSGKQYVCHHIYQLSIND
    EDSQYLKNIAKYLQEEPDGNIE
    RQNILNITSVNNIKLFDVLCTK
    FNSNTYEIILNSLKNDVNEGRE
    KFSELDILEQCNILLQLLKAFK
    CNRESSNLEKLNNKKQAGVIVI
    PHLFTKCSVFKVIHQSITGLFE
    KEMDLLK (SEQ ID NO:
    31)
    Staphylococcus MGRKPYILSLDIGTGSVGYACM 164 299 136 531 606 92 531 606 92
    pseudintermedius DKGFNVLKYHDKDALGVYLFDG
    ED99 ALTAQERRQFRTSRRRKNRRIK
    gi|323463801| RLGLLQELLAPLVQNPNFYQFQ
    gb|ADX75954.1| RQFAWKNDNMDFKNKSLSEVLS
    FLGYESKKYPTIYHLQEALLLK
    DEKFDPELIYMALYHLVKYRGH
    FLFDHLKIENLTNNDNMHDFVE
    LIETYENLNNIKLNLDYEKTKV
    IYEILKDNEMTKNDRAKRVKNM
    EKKLEQFSIMLLGLKFNEGKLF
    NHADNAEELKGANQSHTFADNY
    EENLTPFLTVEQSEFIERANKI
    YLSLTLQDILKGKKSMAMSKVA
    AYDKFRNELKQVKDIVYKADST
    RTQFKKIFVSSKKSLKQYDATP
    NDQTFSSLCLFDQYLIRPKKQY
    SLLIKELKKIIPQDSELYFEAE
    NDTLLKVLNTTDNASIPMQINL
    YEAETILRNQQKYHAEITDEMI
    EKVLSLIQFRIPYYVGPLVNDH
    TASKFGWMERKSNESIKPWNFD
    EVVDRSKSATQFIRRMTNKCSY
    LINEDVLPKNSLLYQEMEVLNE
    LNATQIRLQTDPKNRKYRMMPQ
    IKLFAVEHIFKKYKTVSHSKFL
    EIMLNSNHRENFMNHGEKLSIF
    GTQDDKKFASKLSSYQDMTKIF
    GDIEGKRAQIEEIIQWITIFED
    KKILVQKLKECYPELTSKQINQ
    LKKLNYSGWGRLSEKLLTHAYQ
    GHSIIELLRHSDENFMEILTND
    VYGFQNFIKEENQVQSNKIQHQ
    DIANLTTSPALKKGIWSTIKLV
    RELTSIFGEPEKIIMEFATEDQ
    QKGKKQKSRKQLWDDNIKKNKL
    KSVDEYKYIIDVANKLNNEQLQ
    QEKLWLYLSQNGKCMYSGQSID
    LDALLSPNATKHYEVDHIFPRS
    FIKDDSIDNKVLVIKKMNQTKG
    DQVPLQFIQQPYERIAYWKSLN
    KAGLISDSKLHKLMKPEFTAMD
    KEGFIQRQLVETRQISVHVRDF
    LKEEYPNTKVIPMKAKMVSEFR
    KKFDIPKIRQMNDAHHAIDAYL
    NGVVYHGAQLAYPNVDLFDFNF
    KWEKVREKWKALGEFNTKQKSR
    ELFFFKKLEKMEVSQGERLISK
    IKLDMNHFKINYSRKLANIPQQ
    FYNQTAVSPKTAELKYESNKSN
    EVVYKGLTPYQTYVVAIKSVNK
    KGKEKMEYQMIDHYVFDFYKFQ
    NGNEKELALYLAQRENKDEVLD
    AQIVYSLNKGDLLYINNHPCYF
    VSRKEVINAKQFELTVEQQLSL
    YNVMNNKETNVEKLLIEYDFIA
    EKVINEYHHYLNSKLKEKRVRT
    FFSESNQTHEDFIKALDELFKV
    VTASATRSDKIGSRKNSMTHRA
    FLGKGKDVKIAYTSISGLKTTK
    PKSLFKLAESRNEL (SEQ ID
    NO: 32)
    Flavobacterium MAKILGLDLGTNSIGWAVVERE 162 286 125 538 613 63 538 613 63
    branchiophilum NIDFSLIDKGVRIFSEGVKSEK
    FL-15 GIESSRAAERTGYRSARKIKYR
    gi|347536497| RKLRKYETLKVLSLNRMCPLSI
    ref|YP_004843922.1 EEVEEWKKSGFKDYPLNPEFLK
    WLSTDEESNVNPYFFRDRASKH
    KVSLFELGRAFYHIAQRRGFLS
    NRLDQSAEGILEEHCPKIEAIV
    EDLISIDEISTNITDYFFETGI
    LDSNEKNGYAKDLDEGDKKLVS
    LYKSLLAILKKNESDFENCKSE
    IIERLNKKDVLGKVKGKIKDIS
    QAMLDGNYKTLGQYFYSLYSKE
    KIRNQYTSREEHYLSEFITICK
    VQGIDQINEEEKINEKKFDGLA
    KDLYKAIFFQRPLKSQKGLIGK
    CSFEKSKSRCAISHPDFEEYRM
    WTYLNTIKIGTQSDKKLRFLTQ
    DEKLKLVPKFYRKNDFNFDVLA
    KELIEKGSSFGFYKSSKKNDFF
    YWFNYKPTDTVAACQVAASLKN
    AIGEDWKTKSFKYQTINSNKEQ
    VSRTVDYKDLWHLLTVATSDVY
    LYEFAIDKLGLDEKNAKAFSKT
    KLKKDFASLSLSAINKILPYLK
    EGLLYSHAVFVANIENIVDENI
    WKDEKQRDYIKTQISEIIENYT
    LEKSRFEIINGLLKEYKSENED
    GKRVYYSKEAEQSFENDLKKKL
    VLFYKSNEIENKEQQETIFNEL
    LPIFIQQLKDYEFIKIQRLDQK
    VLIFLKGKNETGQIFCTEEKGT
    AEEKEKKIKNRLKKLYHPSDIE
    KFKKKIIKDEFGNEKIVLGSPL
    TPSIKNPMAMRALHQLRKVLNA
    LILEGQIDEKTIIHIEMARELN
    DANKRKGIQDYQNDNKKFREDA
    IKEIKKLYFEDCKKEVEPTEDD
    ILRYQLWMEQNRSEIYEEGKNI
    SICDIIGSNPAYDIEHTIPRSR
    SQDNSQMNKTLCSQRFNREVKK
    QSMPIELNNHLEILPRIAHWKE
    EADNLTREIEIISRSIKAAATK
    EIKDKKIRRRHYLTLKRDYLQG
    KYDRFIWEEPKVGFKNSQIPDT
    GIITKYAQAYLKSYFKKVESVK
    GGMVAEFRKIWGIQESFIDENG
    MKHYKVKDRSKHTHHTIDAITI
    ACMTKEKYDVLAHAWTLEDQQN
    KKEARSIIEASKPWKTFKEDLL
    KIEEEILVSHYTPDNVKKQAKK
    IVRVRGKKQFVAEVERDVNGKA
    VPKKAASGKTIYKLDGEGKKLP
    RLQQGDTIRGSLHQDSIYGAIK
    NPLNTDEIKYVIRKDLESIKGS
    DVESIVDEVVKEKIKEAIANKV
    LLLSSNAQQKNKLVGTVWMNEE
    KRIAINKVRIYANSVKNPLHIK
    EHSLLSKSKHVHKQKVYGQNDE
    NYAMAIYELDGKRDFELINIFN
    LAKLIKQGQGFYPLHKKKEIKG
    KIVFVPIEKRNKRDVVLKRGQQ
    VVFYDKEVENPKDISEIVDFKG
    RIYIIEGLSIQRIVRPSGKVDE
    YGVIMLRYFKEARKADDIKQDN
    FKPDGVFKLGENKPTRKMNHQF
    TAFVEGIDFKVLPSGKFEKI
    (SEQ ID NO: 33)
    Ignavibacterium MEFKKVLGLDIGTNSIGCALLS 223 329 107 357 432 90 357 432 90
    album LPKSIQDYGKGGRLEWLTSRVI
    JCM 16511 PLDADYMKAFIDGKNGLPQVIT
    gi|385811609| PAGKRRQKRGSRRLKHRYKLRR
    ref|YP_005848005.1 SRLIRVFKTLNWLPEDFPLDNP
    KRIKETISTEGKFSFRISDYVP
    ISDESYREFYREFGYPENEIEQ
    VIEEINFRRKTKGKNKNPMIKL
    LPEDWVVYYLRKKALIKPTTKE
    ELIRIIYLFNQRRGFKSSRKDL
    TETAILDYDEFAKRLAEKEKYS
    AENYETKFVSITKVKEVVELKT
    DGRKGKKRFKVILEDSRIEPYE
    IERKEKPDWEGKEYTFLVTQKL
    EKGKFKQNKPDLPKEEDWALCT
    TALDNRMGSKHPGEFFFDELLK
    AFKEKRGYKIRQYPVNRWRYKK
    ELEFIWTKQCQLNPELNNLNIN
    KEILRKLATVLYPSQSKFFGPK
    IKEFENSDVLHIISEDIIYYQR
    DLKSQKSLISECRYEKRKGIDG
    EIYGLKCIPKSSPLYQEFRIWQ
    DIHNIKVIRKESEVNGKKKINI
    DETQLYINENIKEKLFELFNSK
    DSLSEKDILELISLNIINSGIK
    ISKKEEETTHRINLFANRKELK
    GNETKSRYRKVFKKLGFDGEYI
    LNHPSKLNRLWHSDYSNDYADK
    EKTEKSILSSLGWKNRNGKWEK
    SKNYDVFNLPLEVAKAIANLPP
    LKKEYGSYSALAIRKMLVVMRD
    GKYWQHPDQIAKDQENTSLMLF
    DKNLIQLTNNQRKVLNKYLLTL
    AEVQKRSTLIKQKLNEIEHNPY
    KLELVSDQDLEKQVLKSFLEKK
    NESDYLKGLKTYQAGYLIYGKH
    SEKDVPIVNSPDELGEYIRKKL
    PNNSLRNPIVEQVIRETIFIVR
    DVWKSFGIIDEIHIELGRELKN
    NSEERKKTSESQEKNFQEKERA
    RKLLKELLNSSNFEHYDENGNK
    IFSSFTVNPNPDSPLDIEKFRI
    WKNQSGLTDEELNKKLKDEKIP
    TEIEVKKYILWLTQKCRSPYTG
    KIIPLSKLFDSNVYEIEHIIPR
    SKMKNDSTNNLVICELGVNKAK
    GDRLAANFISESNGKCKFGEVE
    YTLLKYGDYLQYCKDTFKYQKA
    KYKNLLATEPPEDFIERQINDT
    RYIGRKLAELLTPVVKDSKNII
    FTIGSITSELKITWGLNGVWKD
    ILRPRFKRLESIINKKLIFQDE
    DDPNKYHFDLSINPQLDKEGLK
    RLDHRHHALDATIIAATTREHV
    RYLNSLNAADNDEEKREYFLSL
    CNHKIRDFKLPWENFTSEVKSK
    LLSCVVSYKESKPILSDPFNKY
    LKWEYKNGKWQKVFAIQIKNDR
    WKAVRRSMFKEPIGTVWIKKIK
    EVSLKEAIKIQAIWEEVKNDPV
    RKKKEKYIYDDYAQKVIAKIVQ
    ELGLSSSMRKQDDEKLNKFINE
    AKVSAGVNKNLNTTNKTIYNLE
    GRFYEKIKVAEYVLYKAKRMPL
    NKKEYIEKLSLQKMFNDLPNFI
    LEKSILDNYPEILKELESDNKY
    IIEPHKKNNPVNRLLLEHILEY
    HNNPKEAFSTEGLEKLNKKAIN
    KIGKPIKYITRLDGDINEEEIF
    RGAVFETDKGSNVYFVMYENNQ
    TKDREFLKPNPSISVLKAIEHK
    NKIDFFAPNRLGFSRIILSPGD
    LVYVPTNDQYVLIKDNSSNETI
    INWDDNEFISNRIYQVKKFTGN
    SCYFLKNDIASLILSYSASNGV
    GEFGSQNISEYSVDDPPIRIKD
    VCIKIRVDRLGNVRPL (SEQ
    ID NO: 34)
    Bergeyella MKHILGLDLGTNSIGWALIERN 165 261 97 529 604 56 529 604 56
    zoohelcum IEEKYGKIIGMGSRIVPMGAEL
    ATCC 43767 SKFEQGQAQTKNADRRTNRGAR
    gi|423317190| RLNKRYKQRRNKLIYILQKLDM
    ref|ZP_17295095.1 LPSQIKLKEDFSDPNKIDKITI
    LPISKKQEQLTAFDLVSLRVKA
    LTEKVGLEDLGKIIYKYNQLRG
    YAGGSLEPEKEDIFDEEQSKDK
    KNKSFIAFSKIVFLGEPQEEIF
    KNKKLNRRAIIVETEEGNFEGS
    TFLENIKVGDSLELLINISASK
    SGDTITIKLPNKTNWRKKMENI
    ENQLKEKSKEMGREFYISEFLL
    ELLKENRWAKIRNNTILRARYE
    SEFEAIWNEQVKHYPFLENLDK
    KTLIEIVSFIFPGEKESQKKYR
    ELGLEKGLKYIIKNQVVFYQRE
    LKDQSHLISDCRYEPNEKAIAK
    SHPVFQEYKVWEQINKLIVNTK
    IEAGTNRKGEKKYKYIDRPIPT
    ALKEWIFEELQNKKEITFSAIF
    KKLKAEFDLREGIDFLNGMSPK
    DKLKGNETKLQLQKSLGELWDV
    LGLDSINRQIELWNILYNEKGN
    EYDLTSDRTSKVLEFINKYGNN
    IVDDNAEETAIRISKIKFARAY
    SSLSLKAVERILPLVRAGKYFN
    NDFSQQLQSKILKLLNENVEDP
    FAKAAQTYLDNNQSVLSEGGVG
    NSIATILVYDKHTAKEYSHDEL
    YKSYKEINLLKQGDLRNPLVEQ
    IINEALVLIRDIWKNYGIKPNE
    IRVELARDLKNSAKERATIHKR
    NKDNQTINNKIKETLVKNKKEL
    SLANIEKVKLWEAQRHLSPYTG
    QPIPLSDLFDKEKYDVDHIIPI
    SRYFDDSFTNKVISEKSVNQEK
    ANRTAMEYFEVGSLKYSIFTKE
    QFIAHVNEYFSGVKRKNLLATS
    IPEDPVQRQIKDTQYIAIRVKE
    ELNKIVGNENVKTTTGSITDYL
    RNHWGLTDKFKLLLKERYEALL
    ESEKFLEAEYDNYKKDFDSRKK
    EYEEKEVLFEEQELTREEFIKE
    YKENYIRYKKNKLIIKGWSKRI
    DHRHHAIDALIVACTEPAHIKR
    LNDLNKVLQDWLVEHKSEFMPN
    FEGSNSELLEEILSLPENERTE
    IFTQIEKFRAIEMPWKGFPEQV
    EQKLKEIIISHKPKDKLLLQYN
    KAGDRQIKLRGQLHEGTLYGIS
    QGKEAYRIPLTKFGGSKFATEK
    NIQKIVSPFLSGFIANHLKEYN
    NKKEEAFSAEGIMDLNNKLAQY
    RNEKGELKPHTPISTVKIYYKD
    PSKNKKKKDEEDLSLQKLDREK
    AFNEKLYVKTGDNYLFAVLEGE
    IKTKKTSQIKRLYDIISFFDAT
    NFLKEEFRNAPDKKTFDKDLLF
    RQYFEERNKAKLLFTLKQGDFV
    YLPNENEEVILDKESPLYNQYW
    GDLKERGKNIYVVQKFSKKQIY
    FIKHTIADIIKKDVEFGSQNCY
    ETVEGRSIKENCFKLEIDRLGN
    IVKVIKR (SEQ ID NO: 35)
    Nitrobacter MHVEIDFPHFSRGDSHLAMNKN 169 253 85 536 611 48 536 611 48
    hamburgensis EILRGSSVLYRLGLDLGSNSLG
    X14 WFVTHLEKRGDRHEPVALGPGG
    gi|92109262| VRIFPDGRDPQSGTSNAVDRRM
    ref|YP_571550.1| ARGARKRRDRFVERRKELIAAL
    IKYNLLPDDARERRALEVLDPY
    ALRKTALTDTLPAHHVGRALFH
    LNQRRGFQSNRKTDSKQSEDGA
    IKQAASRLATDKGNETLGVFFA
    DMHLRKSYEDRQTAIRAELVRL
    GKDHLTGNARKKIWAKVRKRLF
    GDEVLPRADAPHGVRARATITG
    TKASYDYYPTRDMLRDEFNAIW
    AGQSAHHATITDEARTEIEHII
    FYQRPLKPAIVGKCTLDPATRP
    FKEDPEGYRAPWSHPLAQRFRI
    LSEARNLEIRDTGKGSRRLTKE
    QSDLVVAALLANREVKFDKLRT
    LLKLPAEARFNLESDRRAALDG
    DQTAARLSDKKGFNKAWRGFPP
    ERQIAIVARLEETEDENELIAW
    LEKECALDGAAAARVANTTLPD
    GHCRLGLRAIKKIVPIMQDGLD
    EDGVAGAGYHIAAKRAGYDHAK
    LPTGEQLGRLPYYGQWLQDAVV
    GSGDARDQKEKQYGQFPNPTVH
    IGLGQLRRVVNDLIDKYGPPTE
    ISIEFTRALKLSEQQKAERQRE
    QRRNQDKNKARAEELAKFGRPA
    NPRNLLKMRLWEELAHDPLDRK
    CVYTGEQISIERLLSDEVDIDH
    ILPVAMTLDDSPANKIICMRYA
    NRHKRKQTPSEAFGSSPTLQGH
    RYNWDDIAARATGLPRNKRWRF
    DANAREEFDKRGGFLARQLNET
    GWLARLAKQYLGAVTDPNQIWV
    VPGRLTSMLRGKWGLNGLLPSD
    NYAGVQDKAEEFLASTDDMEFS
    GVKNRADHRHHAIDGLVTALTD
    RSLLWKMANAYDEEHEKFVIEP
    PWPTMRDDLKAALEKMVVSHKP
    DHGIEGKLHEDSAYGFVKPLDA
    TGLKEEEAGNLVYRKAIESLNE
    NEVDRIRDIQLRTIVRDHVNVE
    KTKGVALADALRQLQAPSDDYP
    QFKHGLRHVRILKKEKGDYLVP
    IANRASGVAYKAYSAGENFCVE
    VFETAGGKWDGEAVRRFDANKK
    NAGPKIAHAPQWRDANEGAKLV
    MRIHKGDLIRLDHEGRARIMVV
    HRLDAAAGRFKLADHNETGNLD
    KRHATNNDIDPFRWLMASYNTL
    KKLAAVPVRVDELGRVWRVMPN
    (SEQ ID NO: 36)
    Odoribacter METTLGIDLGTNSIGLALVDQE 164 242 79 535 610 63 535 610 63
    laneus YIT EHQILYSGVRIFPEGINKDTIG
    12061 LGEKEESRNATRRAKRQMRRQY
    gi|374384763| FRKKLRKAKLLELLIAYDMCPL
    ref|ZP_09642280.1 KPEDVRRWKNWDKQQKSTVRQF
    PDTPAFREWLKQNPYELRKQAV
    TEDVTRPELGRILYQMIQRRGF
    LSSRKGKEEGKIFTGKDRMVGI
    DETRKNLQKQTLGAYLYDIAPK
    NGEKYRFRTERVRARYTLRDMY
    IREFEIIWQRQAGHLGLAHEQA
    TRKKNIFLEGSATNVRNSKLIT
    HLQAKYGRGHVLIEDTRITVTF
    QLPLKEVLGGKIEIEEEQLKFK
    SNESVLFWQRPLRSQKSLLSKC
    VFEGRNFYDPVHQKWIIAGPTP
    APLSHPEFEEFRAYQFINNIIY
    GKNEHLTAIQREAVFELMCTES
    KDFNFEKIPKHLKLFEKFNFDD
    TTKVPACTTISQLRKLFPHPVW
    EEKREEIWHCFYFYDDNTLLFE
    KLQKDYALQTNDLEKIKKIRLS
    ESYGNVSLKAIRRINPYLKKGY
    AYSTAVLLGGIRNSFGKRFEYF
    KEYEPEIEKAVCRILKEKNAEG
    EVIRKIKDYLVHNRFGFAKNDR
    AFQKLYHHSQAITTQAQKERLP
    ETGNLRNPIVQQGLNELRRTVN
    KLLATCREKYGPSFKFDHIHVE
    MGRELRSSKTEREKQSRQIREN
    EKKNEAAKVKLAEYGLKAYRDN
    IQKYLLYKEIEEKGGTVCCPYT
    GKTLNISHTLGSDNSVQIEHII
    PYSISLDDSLANKTLCDATFNR
    EKGELTPYDFYQKDPSPEKWGA
    SSWEEIEDRAFRLLPYAKAQRF
    IRRKPQESNEFISRQLNDTRYI
    SKKAVEYLSAICSDVKAFPGQL
    TAELRHLWGLNNILQSAPDITF
    PLPVSATENHREYYVITNEQNE
    VIRLFPKQGETPRTEKGELLLT
    GEVERKVFRCKGMQEFQTDVSD
    GKYWRRIKLSSSVTWSPLFAPK
    PISADGQIVLKGRIEKGVFVCN
    QLKQKLKTGLPDGSYWISLPVI
    SQTFKEGESVNNSKLTSQQVQL
    FGRVREGIFRCHNYQCPASGAD
    GNFWCTLDTDTAQPAFTPIKNA
    PPGVGGGQIILTGDVDDKGIFH
    ADDDLHYELPASLPKGKYYGIF
    TVESCDPTLIPIELSAPKTSKG
    ENLIEGNIWVDEHTGEVRFDPK
    KNREDQRHHAIDAIVIALSSQS
    LFQRLSTYNARRENKKRGLDST
    EHFPSPWPGFAQDVRQSVVPLL
    VSYKQNPKTLCKISKTLYKDGK
    KIHSCGNAVRGQLHKETVYGQR
    TAPGATEKSYHIRKDIRELKTS
    KHIGKVVDITIRQMLLKHLQEN
    YHIDITQEFNIPSNAFFKEGVY
    RIFLPNKHGEPVPIKKIRMKEE
    LGNAERLKDNINQYVNPRNNHH
    VMIYQDADGNLKEEIVSFWSVI
    ERQNQGQPIYQLPREGRNIVSI
    LQINDTFLIGLKEEEPEVYRND
    LSTLSKHLYRVQKLSGMYYTFR
    HHLASTLNNEREEFRIQSLEAW
    KRANPVKVQIDEIGRITFLNGP
    LC (SEQ ID NO: 37)
    Legionella MESSQILSPIGIDLGGKFTGVC 164 239 76 402 476 67 402 476 67
    pneumophila LSHLEAFAELPNHANTKYSVIL
    str. Paris IDHNNFQLSQAQRRATRHRVRN
    gi|54296138| KKRNQFVKRVALQLFQHILSRD
    ref|YP_122507.1| LNAKEETALCHYLNNRGYTYVD
    TDLDEYIKDETTINLLKELLPS
    ESEHNFIDWFLQKMQSSEFRKI
    LVSKVEEKKDDKELKNAVKNIK
    NFITGFEKNSVEGHRHRKVYFE
    NIKSDITKDNQLDSIKKKIPSV
    CLSNLLGHLSNLQWKNLHRYLA
    KNPKQFDEQTFGNEFLRMLKNF
    RHLKGSQESLAVRNLIQQLEQS
    QDYISILEKTPPEITIPPYEAR
    TNTGMEKDQSLLLNPEKLNNLY
    PNWRNLIPGIIDAHPFLEKDLE
    HTKLRDRKRIISPSKQDEKRDS
    YILQRYLDLNKKIDKFKIKKQL
    SFLGQGKQLPANLIETQKEMET
    HFNSSLVSVLIQIASAYNKERE
    DAAQGIWFDNAFSLCELSNINP
    PRKQKILPLLVGAILSEDFINN
    KDKWAKFKIFWNTHKIGRTSLK
    SKCKEIEEARKNSGNAFKIDYE
    EALNHPEHSNNKALIKIIQTIP
    DIIQAIQSHLGHNDSQALIYHN
    PFSLSQLYTILETKRDGFHKNC
    VAVTCENYWRSQKTEIDPEISY
    ASRLPADSVRPFDGVLARMMQR
    LAYEIAMAKWEQIKHIPDNSSL
    LIPIYLEQNRFEFEESFKKIKG
    SSSDKTLEQAIEKQNIQWEEKF
    QRIINASMNICPYKGASIGGQG
    EIDHIYPRSLSKKHFGVIFNSE
    VNLIYCSSQGNREKKEEHYLLE
    HLSPLYLKHQFGTDNVSDIKNF
    ISQNVANIKKYISFHLLTPEQQ
    KAARHALFLDYDDEAFKTITKF
    LMSQQKARVNGTQKFLGKQIME
    FLSTLADSKQLQLEFSIKQITA
    EEVHDHRELLSKQEPKLVKSRQ
    QSFPSHAIDATLTMSIGLKEFP
    QFSQELDNSWFINHLMPDEVHL
    NPVRSKEKYNKPNISSTPLFKD
    SLYAERFIPVWVKGETFAIGFS
    EKDLFEIKPSNKEKLFTLLKTY
    STKNPGESLQELQAKSKAKWLY
    FPINKTLALEFLHHYFHKEIVT
    PDDTTVCHFINSLRYYTKKESI
    TVKILKEPMPVLSVKFESSKKN
    VLGSFKHTIALPATKDWERLFN
    HPNFLALKANPAPNPKEFNEFI
    RKYFLSDNNPNSDIPNNGHNIK
    PQKHKAVRKVFSLPVIPGNAGT
    MMRIRRKDNKGQPLYQLQTIDD
    TPSMGIQINEDRLVKQEVLMDA
    YKTRNLSTIDGINNSEGQAYAT
    FDNWLTLPVSTFKPEIIKLEMK
    PHSKTRRYIRITQSLADFIKTI
    DEALMIKPSDSIDDPLNMPNEI
    VCKNKLFGNELKPRDGKMKIVS
    TGKIVTYEFESDSTPQWIQTLY
    VTQLKKQP (SEQ ID NO:
    38)
    Bacteroides MKKIVGLDLGTNSIGWALINAY 198 269 72 530 604 83 530 604 83
    sp. 203 INKEHLYGIEACGSRIIPMDAA
    gi|301311869| ILGNFDKGNSISQTADRTSYRG
    ref|ZP_07217791.1 IRRLRERHLLRRERLHRILDLL
    GFLPKHYSDSLNRYGKFLNDIE
    CKLPWVKDETGSYKFIFQESFK
    EMLANFTEHHPILIANNKKVPY
    DWTIYYLRKKALTQKISKEELA
    WILLNFNQKRGYYQLRGEEEET
    PNKLVEYYSLKVEKVEDSGERK
    GKDTWYNVHLENGMIYRRTSNI
    PLDWEGKTKEFIVTTDLEADGS
    PKKDKEGNIKRSFRAPKDDDWT
    LIKKKTEADIDKIKMTVGAYIY
    DTLLQKPDQKIRGKLVRTIERK
    YYKNELYQILKTQSEFHEELRD
    KQLYIACLNELYPNNEPRRNSI
    STRDFCHLFIEDIIFYQRPLKS
    KKSLIDNCPYEENRYIDKESGE
    IKHASIKCIAKSHPLYQEFRLW
    QFIVNLRIYRKETDVDVTQELL
    PTEADYVTLFEWLNEKKEIDQK
    AFFKYPPFGFKKTTSNYRWNYV
    EDKPYPCNETHAQIIARLGKAH
    IPKAFLSKEKEETLWHILYSIE
    DKQEIEKALHSFANKNNLSEEF
    IEQFKNFPPFKKEYGSYSAKAI
    KKLLPLMRMGKYWSIENIDNGT
    RIRINKIIDGEYDENIRERVRQ
    KAINLTDITHFRALPLWLACYL
    VYDRHSEVKDIVKWKTPKDIDL
    YLKSFKQHSLRNPIVEQVITET
    LRTVRDIWQQVGHIDEIHIELG
    REMKNPADKRARMSQQMIKNEN
    TNLRIKALLTEFLNPEFGIENV
    RPYSPSQQDLLRIYEEGVLNSI
    LELPEDIGIILGKFNQTDTLKR
    PTRSEILRYKLWLEQKYRSPYT
    GEMIPLSKLFTPAYEIEHIIPQ
    SRYFDDSLSNKVICESEINKLK
    DRSLGYEFIKNHHGEKVELAFD
    KPVEVLSVEAYEKLVHESYSHN
    RSKMKKLLMEDIPDQFIERQLN
    DSRYISKVVKSLLSNIVREENE
    QEAISKNVIPCTGGITDRLKKD
    WGINDVWNKIVLPRFIRLNELT
    ESTRFTSINTNNTMIPSMPLEL
    QKGFNKKRIDHRHHAMDAIIIA
    CANRNIVNYLNNVSASKNTKIT
    RRDLQTLLCHKDKTDNNGNYKW
    VIDKPWETFTQDTLTALQKITV
    SFKQNLRVINKTTNHYQHYENG
    KKIVSNQSKGDSWAIRKSMHKE
    TVHGEVNLRMIKTVSFNEALKK
    PQAIVEMDLKKKILAMLELGYD
    TKRIKNYFEENKDTWQDINPSK
    IKVYYFTKETKDRYFAVRKPID
    TSFDKKKIKESITDTGIQQIML
    RHLETKDNDPTLAFSPDGIDEM
    NRNILILNKGKKHQPIYKVRVY
    EKAEKFTVGQKGNKRTKFVEAA
    KGTNLFFAIYETEEIDKDTKKV
    IRKRSYSTIPLNVVIERQKQGL
    SSAPEDENGNLPKYILSPNDLV
    YVPTQEEINKGEVVMPIDRDRI
    YKMVDSSGITANFIPASTANLI
    FALPKATAEIYCNGENCIQNEY
    GIGSPQSKNQKAITGEMVKEIC
    FPIKVDRLGNIIQVGSCILTN
    (SEQ ID NO: 39)
    Akkermansia MSRSLTFSFDIGYASIGWAVIA 136 202 67 348 418 62 348 418 62
    muciniphila SASHDDADPSVCGCGTVLFPKD
    ATCC BAA- DCQAFKRREYRRLRRNIRSRRV
    835 RIERIGRLLVQAQIITPEMKET
    gi|187736489| SGHPAPFYLASEALKGHRTLAP
    ref|YP_001878601. IELWHVLRWYAHNRGYDNNASW
    SNSLSEDGGNGEDTERVKHAQD
    LMDKHGTATMAETICRELKLEE
    GKADAPMEVSTPAYKNLNTAFP
    RLIVEKEVRRILELSAPLIPGL
    TAEIIELIAQHHPLTTEQRGVL
    LQHGIKLARRYRGSLLFGQLIP
    RFDNRIISRCPVTWAQVYEAEL
    KKGNSEQSARERAEKLSKVPTA
    NCPEFYEYRMARILCNIRADGE
    PLSAEIRRELMNQARQEGKLTK
    ASLEKAISSRLGKETETNVSNY
    FTLHPDSEEALYLNPAVEVLQR
    SGIGQILSPSVYRIAANRLRRG
    KSVTPNYLLNLLKSRGESGEAL
    EKKIEKESKKKEADYADTPLKP
    KYATGRAPYARTVLKKVVEEIL
    DGEDPTRPARGEAHPDGELKAH
    DGCLYCLLDTDSSVNQHQKERR
    LDTMTNNHLVRHRMLILDRLLK
    DLIQDFADGQKDRISRVCVEVG
    KELTTFSAMDSKKIQRELTLRQ
    KSHTDAVNRLKRKLPGKALSAN
    LIRKCRIAMDMNWTCPFTGATY
    GDHELENLELEHIVPHSFRQSN
    ALSSLVLTWPGVNRMKGQRTGY
    DFVEQEQENPVPDKPNLHICSL
    NNYRELVEKLDDKKGHEDDRRR
    KKKRKALLMVRGLSHKHQSQNH
    EAMKEIGMTEGMMTQSSHLMKL
    ACKSIKTSLPDAHIDMIPGAVT
    AEVRKAWDVFGVFKELCPEAAD
    PDSGKILKENLRSLTHLHHALD
    ACVLGLIPYIIPAHHNGLLRRV
    LAMRRIPEKLIPQVRPVANQRH
    YVLNDDGRMMLRDLSASLKENI
    REQLMEQRVIQHVPADMGGALL
    KETMQRVLSVDGSGEDAMVSLS
    KKKDGKKEKNQVKASKLVGVFP
    EGPSKLKALKAAIEIDGNYGVA
    LDPKPVVIRHIKVFKRIMALKE
    QNGGKPVRILKKGMLIHLTSSK
    DPKHAGVWRIESIQDSKGGVKL
    DLQRAHCAVPKNKTHECNWREV
    DLISLLKKYQMKRYPTSYTGTP
    R (SEQ ID NO: 40)
    Prevotella MTQKVLGLDLGTNSIGSAVRNL 184 250 67 357 425 78 357 425 78
    sp. C561 DLSDDLQWQLEFFSSDIFRSSV
    gi|345885718| NKESNGREYSLAAQRSAHRRSR
    ref|ZP_08837074.1 GLNEVRRRRLWATLNLLIKHGF
    CPMSSESLMRWCTYDKRKGLFR
    EYPIDDKDFNAWILLDFNGDGR
    PDYSSPYQLRRELVTRQFDFEQ
    PIERYKLGRALYHIAQHRGFKS
    SKGETLSQQETNSKPSSTDEIP
    DVAGAMKASEEKLSKGLSTYMK
    EHNLLTVGAAFAQLEDEGVRVR
    NNNDYRAIRSQFQHEIETIFKF
    QQGLSVESELYERLISEKKNVG
    TIFYKRPLRSQRGNVGKCTLER
    SKPRCAIGHPLFEKFRAWTLIN
    NIKVRMSVDTLDEQLPMKLRLD
    LYNECFLAFVRTEFKFEDIRKY
    LEKRLGIHFSYNDKTINYKDST
    SVAGCPITARFRKMLGEEWESF
    RVEGQKERQAHSKNNISFHRVS
    YSIEDIWHFCYDAEEPEAVLAF
    AQETLRLERKKAEELVRIWSAM
    PQGYAMLSQKAIRNINKILMLG
    LKYSDAVILAKVPELVDVSDEE
    LLSIAKDYYLVEAQVNYDKRIN
    SIVNGLIAKYKSVSEEYRFADH
    NYEYLLDESDEKDIIRQIENSL
    GARRWSLMDANEQTDILQKVRD
    RYQDFFRSHERKFVESPKLGES
    FENYLTKKFPMVEREQWKKLYH
    PSQITIYRPVSVGKDRSVLRLG
    NPDIGAIKNPTVLRVLNTLRRR
    VNQLLDDGVISPDETRVVVETA
    RELNDANRKWALDTYNRIRHDE
    NEKIKKILEEFYPKRDGISTDD
    IDKARYVIDQREVDYFTGSKTY
    NKDIKKYKFWLEQGGQCMYTGR
    TINLSNLFDPNAFDIEHTIPES
    LSFDSSDMNLTLCDAHYNRFIK
    KNHIPTDMPNYDKAITIDGKEY
    PAITSQLQRWVERVERLNRNVE
    YWKGQARRAQNKDRKDQCMREM
    HLWKMELEYWKKKLERFTVTEV
    TDGFKNSQLVDTRVITRHAVLY
    LKSIFPHVDVQRGDVTAKFRKI
    LGIQSVDEKKDRSLHSHHAIDA
    TTLTIIPVSAKRDRMLELFAKI
    EEINKMLSFSGSEDRTGLIQEL
    EGLKNKLQMEVKVCRIGHNVSE
    IGTFINDNIIVNHHIKNQALTP
    VRRRLRKKGYIVGGVDNPRWQT
    GDALRGEIHKASYYGAITQFAK
    DDEGKVLMKEGRPQVNPTIKFV
    IRRELKYKKSAADSGFASWDDL
    GKAIVDKELFALMKGQFPAETS
    FKDACEQGIYMIKKGKNGMPDI
    KLHHIRHVRCEAPQSGLKIKEQ
    TYKSEKEYKRYFYAAVGDLYAM
    CCYTNGKIREFRIYSLYDVSCH
    RKSDIEDIPEFITDKKGNRLML
    DYKLRTGDMILLYKDNPAELYD
    LDNVNLSRRLYKINRFESQSNL
    VLMTHHLSTSKERGRSLGKTVD
    YQNLPESIRSSVKSLNFLIMGE
    NRDFVIKNGKIIFNHR (SEQ
    ID NO: 41)
    Wolinella MLVSPISVDLGGKNTGFFSFTD 157 218 36 401 468 60 401 468 60
    succinogenes SLDNSQSGTVIYDESFVLSQVG
    DSM 1740 RRSKRHSKRNNLRNKLVKRLFL
    gi|34557932| LILQEHHGLSIDVLPDEIRGLF
    ref|NP_907747.1| NKRGYTYAGFELDEKKKDALES
    DTLKEFLSEKLQSIDRDSDVED
    FLNQIASNAESFKDYKKGFEAV
    FASATHSPNKKLELKDELKSEY
    GENAKELLAGLRVTKEILDEFD
    KQENQGNLPRAKYFEELGEYIA
    TNEKVKSFFDSNSLKLTDMTKL
    IGNISNYQLKELRRYFNDKEME
    KGDIWIPNKLHKITERFVRSWH
    PKNDADRQRRAELMKDLKSKEI
    MELLTTTEPVMTIPPYDDMNNR
    GAVKCQTLRLNEEYLDKHLPNW
    RDIAKRLNHGKFNDDLADSTVK
    GYSEDSTLLHRLLDTSKEIDIY
    ELRGKKPNELLVKTLGQSDANR
    LYGFAQNYYELIRQKVRAGIWV
    PVKNKDDSLNLEDNSNMLKRCN
    HNPPHKKNQIHNLVAGILGVKL
    DEAKFAEFEKELWSAKVGNKKL
    SAYCKNIEELRKTHGNTFKIDI
    EELRKKDPAELSKEEKAKLRLT
    DDVILNEWSQKIANFFDIDDKH
    RQRFNNLFSMAQLHTVIDTPRS
    GFSSTCKRCTAENRFRSETAFY
    NDETGEFHKKATATCQRLPADT
    QRPFSGKIERYIDKLGYELAKI
    KAKELEGMEAKEIKVPIILEQN
    AFEYEESLRKSKTGSNDRVINS
    KKDRDGKKLAKAKENAEDRLKD
    KDKRIKAFSSGICPYCGDTIGD
    DGEIDHILPRSHTLKIYGTVFN
    PEGNLIYVHQKCNQAKADSIYK
    LSDIKAGVSAQWIEEQVANIKG
    YKTFSVLSAEQQKAFRYALFLQ
    NDNEAYKKVVDWLRTDQSARVN
    GTQKYLAKKIQEKLTKMLPNKH
    LSFEFILADATEVSELRRQYAR
    QNPLLAKAEKQAPSSHAIDAVM
    AFVARYQKVFKDGTPPNADEVA
    KLAMLDSWNPASNEPLTKGLST
    NQKIEKMIKSGDYGQKNMREVF
    GKSIFGENAIGERYKPIVVQEG
    GYYIGYPATVKKGYELKNCKVV
    TSKNDIAKLEKIIKNQDLISLK
    ENQYIKIFSINKQTISELSNRY
    FNMNYKNLVERDKEIVGLLEFI
    VENCRYYTKKVDVKFAPKYIHE
    TKYPFYDDWRRFDEAWRYLQEN
    QNKTSSKDRFVIDKSSLNEYYQ
    PDKNEYKLDVDTQPIWDDFCRW
    YFLDRYKTANDKKSIRIKARKT
    FSLLAESGVQGKVFRAKRKIPT
    GYAYQALPMDNNVIAGDYANIL
    LEANSKTLSLVPKSGISIEKQL
    DKKLDVIKKTDVRGLAIDNNSF
    FNADFDTHGIRLIVENTSVKVG
    NFPISAIDKSAKRMIFRALFEK
    EKGKRKKKTTISFKESGPVQDY
    LKVFLKKIVKIQLRTDGSISNI
    VVRKNAADFTLSFRSEHIQKLL
    K (SEQ ID NO: 42)
    Alicyclobacillus MAYRLGLDIGITSVGWAVVALE 142 196 55 416 482 61 416 482 61
    hesperidum KDESGLKPVRIQDLGVRIFDKA
    URH17-3-68 EDSKTGASLALPRREARSARRR
    gi|403744858| TRRRRHRLWRVKRLLEQHGILS
    ref|ZP_10953934.1 MEQIEALYAQRTSSPDVYALRV
    AGLDRCLIAEEIARVLIHIAHR
    RGFQSNRKSEIKDSDAGKLLKA
    VQENENLMQSKGYRTVAEMLVS
    EATKTDAEGKLVHGKKHGYVSN
    VRNKAGEYRHTVSRQAIVDEVR
    KIFAAQRALGNDVMSEELEDSY
    LKILCSQRNFDDGPGGDSPYGH
    GSVSPDGVRQSIYERMVGSCTF
    ETGEKRAPRSSYSFERFQLLTK
    VVNLRIYRQQEDGGRYPCELTQ
    TERARVIDCAYEQTKITYGKLR
    KLLDMKDTESFAGLTYGLNRSR
    NKTEDTVFVEMKFYHEVRKALQ
    RAGVFIQDLSIETLDQIGWILS
    VWKSDDNRRKKLSTLGLSDNVI
    EELLPLNGSKFGHLSLKAIRKI
    LPFLEDGYSYDVACELAGYQFQ
    GKTEYVKQRLLPPLGEGEVTNP
    VVRRALSQAIKVVNAVIRKHGS
    PESIHIELARELSKNLDERRKI
    EKAQKENQKNNEQIKDEIREIL
    GSAHVTGRDIVKYKLFKQQQEF
    CMYSGEKLDVTRLFEPGYAEVD
    HIIPYGISFDDSYDNKVLVKTE
    QNRQKGNRTPLEYLRDKPEQKA
    KFIALVESIPLSQKKKNHLLMD
    KRAIDLEQEGFRERNLSDTRYI
    TRALMNHIQAWLLFDETASTRS
    KRVVCVNGAVTAYMRARWGLTK
    DRDAGDKHHAADAVVVACIGDS
    LIQRVTKYDKFKRNALADRNRY
    VQQVSKSEGITQYVDKETGEVF
    TWESFDERKFLPNEPLEPWPFF
    RDELLARLSDDPSKNIRAIGLL
    TYSETEQIDPIFVSRMPTRKVT
    GAAHKETIRSPRIVKVDDNKGT
    EIQVVVSKVALTELKLTKDGEI
    KDYFRPEDDPRLYNTLRERLVQ
    FGGDAKAAFKEPVYKISKDGSV
    RTPVRKVKIQEKLTLGVPVHGG
    RGIAENGGMVRIDVFAKGGKYY
    FVPIYVADVLKRELPNRLATAH
    KPYSEWRVVDDSYQFKFSLYPN
    DAVMIKPSREVDITYKDRKEPV
    GCRIMYFVSANIASASISLRTH
    DNSGELEGLGIQGLEVFEKYVV
    GPLGDTHPVYKERRMPFRVERK
    MN (SEQ ID NO: 43)
    Caenispirillum MPVLSPLSPNAAQGRRRWSLAL 161 214 54 330 393 68 330 393 68
    salinarum DIGEGSIGWAVAEVDAEGRVLQ
    AK4 LTGTGVTLFPSAWSNENGTYVA
    gi|427429481| HGAADRAVRGQQQRHDSRRRRL
    ref|ZP_18919511.1 AGLARLCAPVLERSPEDLKDLT
    RTPPKADPRAIFFLRADAARRP
    LDGPELFRVLHHMAAHRGIRLA
    ELQEVDPPPESDADDAAPAATE
    DEDGTRRAAADERAFRRLMAEH
    MHRHGTQPTCGEIMAGRLRETP
    AGAQPVTRARDGLRVGGGVAVP
    TRALIEQEFDAIRAIQAPRHPD
    LPWDSLRRLVLDQAPIAVPPAT
    PCLFLEELRRRGETFQGRTITR
    EAIDRGLTVDPLIQALRIRETV
    GNLRLHERITEPDGRQRYVPRA
    MPELGLSHGELTAPERDTLVRA
    LMHDPDGLAAKDGRIPYTRLRK
    LIGYDNSPVCFAQERDTSGGGI
    TVNPTDPLMARWIDGWVDLPLK
    ARSLYVRDVVARGADSAALARL
    LAEGAHGVPPVAAAAVPAATAA
    ILESDIMQPGRYSVCPWAAEAI
    LDAWANAPTEGFYDVTRGLFGF
    APGEIVLEDLRRARGALLAHLP
    RTMAAARTPNRAAQQRGPLPAY
    ESVIPSQLITSLRRAHKGRAAD
    WSAADPEERNPFLRTWTGNAAT
    DHILNQVRKTANEVITKYGNRR
    GWDPLPSRITVELAREAKHGVI
    RRNEIAKENRENEGRRKKESAA
    LDTFCQDNTVSWQAGGLPKERA
    ALRLRLAQRQEFFCPYCAERPK
    LRATDLFSPAETEIDHVIERRM
    GGDGPDNLVLAHKDCNNAKGKK
    TPHEHAGDLLDSPALAALWQGW
    RKENADRLKGKGHKARTPREDK
    DFMDRVGWRFEEDARAKAEENQ
    ERRGRRMLHDTARATRLARLYL
    AAAVMPEDPAEIGAPPVETPPS
    PEDPTGYTAIYRTISRVQPVNG
    SVTHMLRQRLLQRDKNRDYQTH
    HAEDACLLLLAGPAVVQAFNTE
    AAQHGADAPDDRPVDLMPTSDA
    YHQQRRARALGRVPLATVDAAL
    ADIVMPESDRQDPETGRVHWRL
    TRAGRGLKRRIDDLTRNCVILS
    RPRRPSETGTPGALHNATHYGR
    REITVDGRTDTVVTQRMNARDL
    VALLDNAKIVPAARLDAAAPGD
    TILKEICTEIADRHDRVVDPEG
    THARRWISARLAALVPAHAEAV
    ARDIAELADLDALADADRTPEQ
    EARRSALRQSPYLGRAISAKKA
    DGRARAREQEILTRALLDPHWG
    PRGLRHLIMREARAPSLVRIRA
    NKTDAFGRPVPDAAVWVKTDGN
    AVSQLWRLTSVVTDDGRRIPLP
    KPIEKRIEISNLEYARLNGLDE
    GAGVTGNNAPPRPLRQDIDRLT
    PLWRDHGTAPGGYLGTAVGELE
    DKARSALRGKAMRQTLTDAGIT
    AEAGWRLDSEGAVCDLEVAKGD
    TVKKDGKTYKVGVITQGIFGMP
    VDAAGSAPRTPEDCEKFEEQYG
    IKPWKAKGIPLA (SEQ ID
    NO: 44)
    Eubacterium MNYTEKEKLFMKYILALDIGIA 133 185 53 322 384 60 322 384 60
    rectale ATCC SVGWAILDKESETVIEAGSNIF
    33656 PEASAADNQLRRDMRGAKRNNR
    gi|238924075| RLKTRINDFIKLWENNNLSIPQ
    ref|YP_002937591.1 FKSTEIVGLKVRAITEEITLDE
    LYLILYSYLKHRGISYLEDALD
    DTVSGSSAYANGLKLNAKELET
    HYPCEIQQERLNTIGKYRGQSQ
    IINENGEVLDLSNVFTIGAYRK
    EIQRVFEIQKKYHPELTDEFCD
    GYMLIFNRKRKYYEGPGNEKSR
    TDYGRFTTKLDANGNYITEDNI
    FEKLIGKCSVYPDELRAAAASY
    TAQEYNVLNDLNNLTINGRKLE
    ENEKHEIVERIKSSNTINMRKI
    ISDCMGENIDDFAGARIDKSGK
    EIFHKFEVYNKMRKALLEIGID
    ISNYSREELDEIGYIMTINTDK
    EAMMEAFQKSWIDLSDDVKQCL
    INMRKTNGALFNKWQSFSLKIM
    NELIPEMYAQPKEQMTLLTEMG
    VTKGTQEEFAGLKYIPVDVVSE
    DIFNPVVRRSVRISFKILNAVL
    KKYKALDTIVIEMPRDRNSEEQ
    KKRINDSQKLNEKEMEYIEKKL
    AVTYGIKLSPSDFSSQKQLSLK
    LKLWNEQDGICLYSGKTIDPND
    IINNPQLFEIDHIIPRSISFDD
    ARSNKVLVYRSENQKKGNQTPY
    YYLTHSHSEWSFEQYKATVMNL
    SKKKEYAISRKKIQNLLYSEDI
    TKMDVLKGFINRNINDTSYASR
    LVLNTIQNFFMANEADTKVKVI
    KGSYTHQMRCNLKLDKNRDESY
    SHHAVDAMLIGYSELGYEAYHK
    LQGEFIDFETGEILRKDMWDEN
    MSDEVYADYLYGKKWANIRNEV
    VKAEKNVKYWHYVMRKSNRGLC
    NQTIRGTREYDGKQYKINKLDI
    RTKEGIKVFAKLAFSKKDSDRE
    RLLVYLNDRRTFDDLCKIYEDY
    SDAANPFVQYEKETGDIIRKYS
    KKHNGPRIDKLKYKDGEVGACI
    DISHKYGFEKGSKKVILESLVP
    YRMDVYYKEENHSYYLVGVKQS
    DIKFEKGRNVIDEEAYARILVN
    EKMIQPGQSRADLENLGFKFKL
    SFYKNDIIEYEKDGKIYTERLV
    SRTMPKQRNYIETKPIDKAKFE
    KQNLVGLGKTKFIKKYRYDILG
    NKYSCSEEKFTSFC (SEQ ID
    NO: 45)
    Mycoplasma MLRLYCANNLVLNNVQNLWKYL 187 239 53 319 381 80 319 381 80
    synoviae 53 LLLIFDKKIIFLFKIKVILIRR
    gi|71894592| YMENNNKEKIVIGFDLGVASVG
    ref|YP_278700.1| WSIVNAETKEVIDLGVRLFSEP
    EKADYRRAKRTTRRLLRRKKFK
    REKFHKLILKNAEIFGLQSRNE
    ILNVYKDQSSKYRNILKLKINA
    LKEEIKPSELVWILRDYLQNRG
    YFYKNEKLTDEFVSNSFPSKKL
    HEHYEKYGFFRGSVKLDNKLDN
    KKDKAKEKDEEEESDAKKESEE
    LIFSNKQWINEIVKVFENQSYL
    TESFKEEYLKLFNYVRPFNKGP
    GSKNSRTAYGVFSTDIDPETNK
    FKDYSNIWDKTIGKCSLFEEEI
    RAPKNLPSALIFNLQNEICTIK
    NEFTEFKNWWLNAEQKSEILKF
    VFTELFNWKDKKYSDKKFNKNL
    QDKIKKYLLNFALENFNLNEEI
    LKNRDLENDTVLGLKGVKYYEK
    SNATADAALEFSSLKPLYVFIK
    FLKEKKLDLNYLLGLENTEILY
    FLDSIYLAISYSSDLKERNEWF
    KKLLKELYPKIKNNNLEIIENV
    EDIFEITDQEKFESFSKTHSLS
    REAFNHIIPLLLSNNEGKNYES
    LKHSNEELKKRTEKAELKAQQN
    QKYLKDNFLKEALVPLSVKTSV
    LQAIKIFNQIIKNFGKKYEISQ
    VVIEMARELTKPNLEKLLNNAT
    NSNIKILKEKLDQTEKFDDFTK
    KKFIDKIENSVVFRNKLFLWFE
    QDRKDPYTQLDIKINEIEDETE
    IDHVIPYSKSADDSWFNKLLVK
    KSTNQLKKNKTVWEYYQNESDP
    EAKWNKFVAWAKRIYLVQKSDK
    ESKDNSEKNSIFKNKKPNLKFK
    NITKKLFDPYKDLGFLARNLND
    TRYATKVFRDQLNNYSKHHSKD
    DENKLFKVVCMNGSITSFLRKS
    MWRKNEEQVYRFNFWKKDRDQF
    FHHAVDASIIAIFSLLTKTLYN
    KLRVYESYDVQRREDGVYLINK
    ETGEVKKADKDYWKDQHNFLKI
    RENAIEIKNVLNNVDFQNQVRY
    SRKANTKLNTQLFNETLYGVKE
    FENNFYKLEKVNLFSRKDLRKF
    ILEDLNEESEKNKKNENGSRKR
    ILTEKYIVDEILQILENEEFKD
    SKSDINALNKYMDSLPSKFSEF
    FSQDFINKCKKENSLILTFDAI
    KHNDPKKVIKIKNLKFFREDAT
    LKNKQAVHKDSKNQIKSFYESY
    KCVGFIWLKNKNDLEESIFVPI
    NSRVIHFGDKDKDIFDFDSYNK
    EKLLNEINLKRPENKKFNSINE
    IEFVKFVKPGALLLNFENQQIY
    YISTLESSSLRAKIKLLNKMDK
    GKAVSMKKITNPDEYKIIEHVN
    PLGINLNWTKKLENNN (SEQ
    ID NO: 46)
    Porphyromonas MLMSKHVLGLDLGVGSIGWCLI 150 202 53 309 371 60 309 371 60
    sp. oral ALDAQGDPAEILGMGSRVVPLN
    taxon 279 NATKAIEAFNAGAAFTASQERT
    str. F0450 ARRTMRRGFARYQLRRYRLRRE
    gi|402847315| LEKVGMLPDAALIQLPLLELWE
    ref|ZP_10895610.1 LRERAATAGRRLTLPELGRVLC
    HINQKRGYRHVKSDAAAIVGDE
    GEKKKDSNSAYLAGIRANDEKL
    QAEHKTVGQYFAEQLRQNQSES
    PTGGISYRIKDQIFSRQCYIDE
    YDQIMAVQRVHYPDILTDEFIR
    MLRDEVIFMQRPLKSCKHLVSL
    CEFEKQERVMRVQQDDGKGGWQ
    LVERRVKFGPKVAPKSSPLFQL
    CCIYEAVNNIRLTRPNGSPCDI
    TPEERAKIVAHLQSSASLSFAA
    LKKLLKEKALIADQLTSKSGLK
    GNSTRVALASALQPYPQYHHLL
    DMELETRMMTVQLTDEETGEVT
    EREVAVVTDSYVRKPLYRLWHI
    LYSIEEREAMRRALITQLGMKE
    EDLDGGLLDQLYRLDFVKPGYG
    NKSAKFICKLLPQLQQGLGYSE
    ACAAVGYRHSNSPTSEEITERT
    LLEKIPLLQRNELRQPLVEKIL
    NQMINLVNALKAEYGIDEVRVE
    LARELKMSREERERMARNNKDR
    EERNKGVAAKIRECGLYPTKPR
    IQKYMLWKEAGRQCLYCGRSIE
    EEQCLREGGMEVEHIIPKSVLY
    DDSYGNKTCACRRCNKEKGNRT
    ALEYIRAKGREAEYMKRINDLL
    KEKKISYSKHQRLRWLKEDIPS
    DFLERQLRLTQYISRQAMAILQ
    QGIRRVSASEGGVTARLRSLWG
    YGKILHTLNLDRYDSMGETERV
    SREGEATEELHITNWSKRMDHR
    HHAIDALVVACTRQSYIQRLNR
    LSSEFGREDKKKEDQEAQEQQA
    TETGRLSNLERWLTQRPHFSVR
    TVSDKVAEILISYRPGQRVVTR
    GRNIYRKKMADGREVSCVQRGV
    LVPRGELMEASFYGKILSQGRV
    RIVKRYPLHDLKGEVVDPHLRE
    LITTYNQELKSREKGAPIPPLC
    LDKDKKQEVRSVRCYAKTLSLD
    KAIPMCFDEKGEPTAFVKSASN
    HHLALYRTPKGKLVESIVTFWD
    AVDRARYGIPLVITHPREVMEQ
    VLQRGDIPEQVLSLLPPSDWVF
    VDSLQQDEMVVIGLSDEELQRA
    LEAQNYRKISEHLYRVQKMSSS
    YYVFRYHLETSVADDKNTSGRI
    PKFHRVQSLKAYEERNIRKVRV
    DLLGRISLL (SEQ ID NO:
    47)
    Streptococcus MSDLVLGLDIGIGSVGVGILNK 127 178 139 424 486 81 424 486 81
    thermophilus VTGEIIHKNSRIFPAAQAENNL
    LMD-9 VRRTNRQGRRLARRKKHRRVRL
    gi|116627542| NRLFEESGLITDFTKISINLNP
    ref|YP_820161.1| YQLRVKGLTDELSNEELFIALK
    NMVKHRGISYLDDASDDGNSSV
    GDYAQIVKENSKQLETKTPGQI
    QLERYQTYGQLRGDFTVEKDGK
    KHRLINVFPTSAYRSEALRILQ
    TQQEFNPQITDEFINRYLEILT
    GKRKYYHGPGNEKSRTDYGRYR
    TSGETLDNIFGILIGKCTFYPD
    EFRAAKASYTAQEFNLLNDLNN
    LTVPTETKKLSKEQKNQIINYV
    KNEKAMGPAKLFKYIAKLLSCD
    VADIKGYRIDKSGKAEIHTFEA
    YRKMKTLETLDIEQMDRETLDK
    LAYVLTLNTEREGIQEALEHEF
    ADGSFSQKQVDELVQFRKANSS
    IFGKGWHNFSVKLMMELIPELY
    ETSEEQMTILTRLGKQKTTSSS
    NKTKYIDEKLLTEEIYNPVVAK
    SVRQAIKIVNAAIKEYGDFDNI
    VIEMARETNEDDEKKAIQKIQK
    ANKDEKDAAMLKAANQYNGKAE
    LPHSVFHGHKQLATKIRLWHQQ
    GERCLYTGKTISIHDLINNSNQ
    FEVDHILPLSITFDDSLANKVL
    VYATANQEKGQRTPYQALDSMD
    DAWSFRELKAFVRESKTLSNKK
    KEYLLTEEDISKFDVRKKFIER
    NLVDTRYASRVVLNALQEHFRA
    HKIDTKVSVVRGQFTSQLRRHW
    GIEKTRDTYHHHAVDALIIAAS
    SQLNLWKKQKNTLVSYSEDQLL
    DIETGELISDDEYKESVFKAPY
    QHFVDTLKSKEFEDSILFSYQV
    DSKFNRKISDATIYATRQAKVG
    KDKADETYVLGKIKDIYTQDGY
    DAFMKIYKKDKSKFLMYRHDPQ
    TFEKVIEPILENYPNKQINEKG
    KEVPCNPFLKYKEEHGYIRKYS
    KKGNGPEIKSLKYYDSKLGNHI
    DITPKDSNNKVVLQSVSPWRAD
    VYFNKTTGKYEILGLKYADLQF
    EKGTGTYKISQEKYNDIKKKEG
    VDSDSEFKFTLYKNDLLLVKDT
    ETKEQQLFRFLSRTMPKQKHYV
    ELKPYDKQKFEGGEALIKVLGN
    VANSGQCKKGLGKSNISIYKVR
    TDVLGNQHIIKNEGDKPKLDF
    (SEQ ID NO: 48)
    Roseburia MNAEHGKEGLLIMEENFQYRIG 154 204 51 318 380 69 318 380 69
    inulinivorans LDIGITSVGWAVLQNNSQDEPV
    DSM 16841 RITDLGVRIFDVAENPKNGDAL
    gi|225377804| AAPRRDARTTRRRLRRRRHRLE
    ref|ZP_03755025.1 RIKFLLQENGLIEMDSFMERYY
    KGNLPDVYQLRYEGLDRKLKDE
    ELAQVLIHIAKHRGFRSTRKAE
    TKEKEGGAVLKATTENQKIMQE
    KGYRTVGEMLYLDEAFHTECLW
    NEKGYVLTPRNRPDDYKHTILR
    SMLVEEVHAIFAAQRAHGNQKA
    TEGLEEAYVEIMTSQRSFDMGP
    GLQPDGKPSPYAMEGFGDRVGK
    CTFEKDEYRAPKATYTAELFVA
    LQKINHTKLIDEFGTGRFFSEE
    ERKTIIGLLLSSKELKYGTIRK
    KLNIDPSLKFNSLNYSAKKEGE
    TEEERVLDTEKAKFASMFWTYE
    YSKCLKDRTEEMPVGEKADLFD
    RIGEILTAYKNDDSRSSRLKEL
    GLSGEEIDGLLDLSPAKYQRVS
    LKAMRKMQPYLEDGLIYDKACE
    AAGYDFRALNDGNKKHLLKGEE
    INAIVNDITNPVVKRSVSQTIK
    VINAIIQKYGSPQAVNIELARE
    MSKNFQDRTNLEKEMKKRQQEN
    ERAKQQIIELGKQNPTGQDILK
    YRLWNDQGGYCLYSGKKIPLEE
    LFDGGYDIDHILPYSITFDDSY
    RNKVLVTAQENRQKGNRTPYEY
    FGADEKRWEDYEASVRLLVRDY
    KKQQKLLKKNFTEEERKEFKER
    NLNDTKYITRVVYNMIRQNLEL
    EPFNHPEKKKQVWAVNGAVTSY
    LRKRWGLMQKDRSTDRHHAMDA
    VVIACCTDGMIHKISRYMQGRE
    LAYSRNFKFPDEETGEILNRDN
    FTREQWDEKFGVKVPLPWNSFR
    DELDIRLLNEDPKNFLLTHADV
    QRELDYPGWMYGEEESPIEEGR
    YINYIRPLFVSRMPNHKVTGSA
    HDATIRSARDYETRGVVITKVP
    LTDLKLNKDNEIEGYYDKDSDR
    LLYQALVRQLLLHGNDGKKAFA
    EDFHKPKADGTEGPVVRKVKIE
    KKQTSGVMVRGGTGIAANGEMV
    RIDVFRENGKYYFVPVYTADVV
    RKVLPNRAATHTKPYSEWRVMD
    DANFVFSLYSRDLIHVKSKKDI
    KTNLVNGGLLLQKEIFAYYTGA
    DIATASIAGFANDSNFKFRGLG
    IQSLEIFEKCQVDILGNISVVR
    HENRQEFH (SEQ ID NO:
    49)
    Methylosinus MRVLGLDAGIASLGWALIEIEE 144 193 50 426 488 64 426 488 64
    trichosporium SNRGELSQGTIIGAGTWMFDAP
    OB3b EEKTQAGAKLKSEQRRTFRGQR
    gi|296446027| RVVRRRRQRMNEVRRILHSHGL
    ref|ZP_06887976.1 LPSSDRDALKQPGLDPWRIRAE
    ALDRLLGPVELAVALGHIARHR
    GFKSNSKGAKTNDPADDTSKMK
    RAVNETREKLARFGSAAKMLVE
    DESFVLRQTPTKNGASEIVRRF
    RNREGDYSRSLLRDDLAAEMRA
    LFTAQARFQSAIATADLQTAFT
    KAAFFQRPLQDSEKLVGPCPFE
    VDEKRAPKRGYSFELFRFLSRL
    NHVTLRDGKQERTLTRDELALA
    AADFGAAAKVSFTALRKKLKLP
    ETTVFVGVKADEESKLDVVARS
    GKAAEGTARLRSVIVDALGELA
    WGALLCSPEKLDKIAEVISFRS
    DIGRISEGLAQAGCNAPLVDAL
    TAAASDGRFDPFTGAGHISSKA
    ARNILSGLRQGMTYDKACCAAD
    YDHTASRERGAFDVGGHGREAL
    KRILQEERISRELVGSPTARKA
    LIESIKQVKAIVERYGVPDRIH
    VELARDVGKSIEEREEITRGIE
    KRNRQKDKLRGLFEKEVGRPPQ
    DGARGKEELLRFELWSEQMGRC
    LYTDDYISPSQLVATDDAVQVD
    HILPWSRFADDSYANKTLCMAK
    ANQDKKGRTPYEWFKAEKTDTE
    WDAFIVRVEALADMKGFKKRNY
    KLRNAEEAAAKFRNRNLNDTRW
    ACRLLAEALKQLYPKGEKDKDG
    KERRRVFSRPGALTDRLRRAWG
    LQWMKKSTKGDRIPDDRHHALD
    AIVIAATTESLLQRATREVQEI
    EDKGLHYDLVKNVTPPWPGFRE
    QAVEAVEKVFVARAERRRARGK
    AHDATIRHIAVREGEQRVYERR
    KVAELKLADLDRVKDAERNARL
    IEKLRNWIEAGSPKDDPPLSPK
    GDPIFKVRLVTKSKVNIALDTG
    NPKRPGTVDRGEMARVDVFRKA
    SKKGKYEYYLVPIYPHDIATMK
    TPPIRAVQAYKPEDEWPEMDSS
    YEFCWSLVPMTYLQVISSKGEI
    FEGYYRGMNRSVGAIQLSAHSN
    SSDVVQGIGARTLTEFKKFNVD
    RFGRKHEVERELRTWRGETWRG
    KAYI (SEQ ID NO: 50)
    Ruminococcus MGNYYLGLDVGIGSIGWAVINI 139 187 49 351 412 55 351 412 55
    albus 8 EKKRIEDFNVRIFKSGEIQEKN
    gi|325677756| RNSRASQQCRRSRGLRRLYRRK
    ref|ZP_08157403.1 SHRKLRLKNYLSIIGLTTSEKI
    DYYYETADNNVIQLRNKGLSEK
    LTPEEIAACLIHICNNRGYKDF
    YEVNVEDIEDPDERNEYKEEHD
    SIVLISNLMNEGGYCTPAEMIC
    NCREFDEPNSVYRKFHNSAASK
    NHYLITRHMLVKEVDLILENQS
    KYYGILDDKTIAKIKDIIFAQR
    DFEIGPGKNERFRRFTGYLDSI
    GKCQFFKDQERGSRFTVIADIY
    AFVNVLSQYTYTNNRGESVFDT
    SFANDLINSALKNGSMDKRELK
    AIAKSYHIDISDKNSDTSLTKC
    FKYIKVVKPLFEKYGYDWDKLI
    ENYTDTDNNVLNRIGIVLSQAQ
    TPKRRREKLKALNIGLDDGLIN
    ELTKLKLSGTANVSYKYMQGSI
    EAFCEGDLYGKYQAKFNKEIPD
    IDENAKPQKLPPFKNEDDCEFF
    KNPVVFRSINETRKLINAIIDK
    YGYPAAVNIETADELNKTFEDR
    AIDTKRNNDNQKENDRIVKEII
    ECIKCDEVHARHLIEKYKLWEA
    QEGKCLYSGETITKEDMLRDKD
    KLFEVDHIVPYSLILDNTINNK
    ALVYAEENQKKGQRTPLMYMNE
    AQAADYRVRVNTMFKSKKCSKK
    KYQYLMLPDLNDQELLGGWRSR
    NLNDTRYICKYLVNYLRKNLRF
    DRSYESSDEDDLKIRDHYRVFP
    VKSRFTSMFRRWWLNEKTWGRY
    DKAELKKLTYLDHAADAIIIAN
    CRPEYVVLAGEKLKLNKMYHQA
    GKRITPEYEQSKKACIDNLYKL
    FRMDRRTAEKLLSGHGRLTPII
    PNLSEEVDKRLWDKNIYEQFWK
    DDKDKKSCEELYRENVASLYKG
    DPKFASSLSMPVISLKPDHKYR
    GTITGEEAIRVKEIDGKLIKLK
    RKSISEITAESINSIYTDDKIL
    IDSLKTIFEQADYKDVGDYLKK
    TNQHFFTTSSGKRVNKVTVIEK
    VPSRWLRKEIDDNNFSLLNDSS
    YYCIELYKDSKGDNNLQGIAMS
    DIVHDRKTKKLYLKPDFNYPDD
    YYTHVMYIFPGDYLRIKSTSKK
    SGEQLKFEGYFISVKNVNENSF
    RFISDNKPCAKDKRVSITKKDI
    VIKLAVDLMGKVQGENNGKGIS
    CGEPLSLLKEKN (SEQ ID
    NO: 51)
    Bifidobacterium MLSRQLLGASHLARPVSYSYNV 183 230 48 370 431 44 370 431 44
    longum QDNDVHCSYGERCFMRGKRYRI
    DJO10A GIDVGLNSVGLAAVEVSDENSP
    gi|189440764| VRLLNAQSVIHDGGVDPQKNKE
    ref|YP_001955845. AITRKNMSGVARRTRRMRRRKR
    ERLHKLDMLLGKFGYPVIEPES
    LDKPFEEWHVRAELATRYIEDD
    ELRRESISIALRHMARHRGWRN
    PYRQVDSLISDNPYSKQYGELK
    EKAKAYNDDATAAEEESTPAQL
    VVAMLDAGYAEAPRLRWRTGSK
    KPDAEGYLPVRLMQEDNANELK
    QIFRVQRVPADEWKPLFRSVFY
    AVSPKGSAEQRVGQDPLAPEQA
    RALKASLAFQEYRIANVITNLR
    IKDASAELRKLTVDEKQSIYDQ
    LVSPSSEDITWSDLCDFLGFKR
    SQLKGVGSLTEDGEERISSRPP
    RLTSVQRIYESDNKIRKPLVAW
    WKSASDNEHEAMIRLLSNTVDI
    DKVREDVAYASAIEFIDGLDDD
    ALTKLDSVDLPSGRAAYSVETL
    QKLTRQMLTTDDDLHEARKTLF
    NVTDSWRPPADPIGEPLGNPSV
    DRVLKNVNRYLMNCQQRWGNPV
    SVNIEHVRSSFSSVAFARKDKR
    EYEKNNEKRSIFRSSLSEQLRA
    DEQMEKVRESDLRRLEAIQRQN
    GQCLYCGRTITFRTCEMDHIVP
    RKGVGSTNTRTNFAAVCAECNR
    MKSNTPFAIWARSEDAQTRGVS
    LAEAKKRVTMFTFNPKSYAPRE
    VKAFKQAVIARLQQTEDDAAID
    NRSIESVAWMADELHRRIDWYF
    NAKQYVNSASIDDAEAETMKTT
    VSVFQGRVTASARRAAGIEGKI
    HFIGQQSKTRLDRRHHAVDASV
    IAMMNTAAAQTLMERESLRESQ
    RLIGLMPGERSWKEYPYEGTSR
    YESFHLWLDNMDVLLELLNDAL
    DNDRIAVMQSQRYVLGNSIAHD
    ATIHPLEKVPLGSAMSADLIRR
    ASTPALWCALTRLPDYDEKEGL
    PEDSHREIRVHDTRYSADDEMG
    FFASQAAQIAVQEGSADIGSAI
    HHARVYRCWKTNAKGVRKYFYG
    MIRVFQTDLLRACHDDLFTVPL
    PPQSISMRYGEPRVVQALQSGN
    AQYLGSLVVGDEIEMDFSSLDV
    DGQIGEYLQFFSQFSGGNLAWK
    HWVVDGFFNQTQLRIRPRYLAA
    EGLAKAFSDDVVPDGVQKIVTK
    QGWLPPVNTASKTAVRIVRRNA
    FGEPRLSSAHHMPCSWQWRHE
    (SEQ ID NO: 52)
    Enterococcus MYSIGLDLGISSVGWSVIDERT 123 170 48 327 387 60 327 387 60
    faecalis GNVIDLGVRLFSAKNSEKNLER
    TX0012 RTNRGGRRLIRRKTNRLKDAKK
    gi|315149830| ILAAVGFYEDKSLKNSCPYQLR
    gb|EFT93846.1| VKGLTEPLSRGEIYKVTLHILK
    KRGISYLDEVDTEAAKESQDYK
    EQVRKNAQLLTKYTPGQIQLQR
    LKENNRVKTGINAQGNYQLNVF
    KVSAYANELATILKTQQAFYPN
    ELTDDWIALFVQPGIAEEAGLI
    YRKRPYYHGPGNEANNSPYGRW
    SDFQKTGEPATNIFDKLIGKDF
    QGELRASGLSLSAQQYNLLNDL
    TNLKIDGEVPLSSEQKEYILTE
    LMTKEFTRFGVNDVVKLLGVKK
    ERLSGWRLDKKGKPEIHTLKGY
    RNWRKIFAEAGIDLATLPTETI
    DCLAKVLTLNTEREGIENTLAF
    ELPELSESVKLLVLDRYKELSQ
    SISTQSWHRFSLKTLHLLIPEL
    MNATSEQNTLLEQFQLKSDVRK
    RYSEYKKLPTKDVLAEIYNPTV
    NKTVSQAFKVIDALLVKYGKEQ
    IRYITIEMPRDDNEEDEKKRIK
    ELHAKNSQRKNDSQSYFMQKSG
    WSQEKFQTTIQKNRRFLAKLLY
    YYEQDGICAYTGLPISPELLVS
    DSTEIDHIIPISISLDDSINNK
    VLVLSKANQVKGQQTPYDAWMD
    GSFKKINGKFSNWDDYQKWVES
    RHFSHKKENNLLETRNIFDSEQ
    VEKFLARNLNDTRYASRLVLNT
    LQSFFTNQETKVRVVNGSFTHT
    LRKKWGADLDKTRETHHHHAVD
    ATLCAVTSFVKVSRYHYAVKEE
    TGEKVMREIDFETGEIVNEMSY
    WEFKKSKKYERKTYQVKWPNFR
    EQLKPVNLHPRIKFSHQVDRKA
    NRKLSDATIYSVREKTEVKTLK
    SGKQKITTDEYTIGKIKDIYTL
    DGWEAFKKKQDKLLMKDLDEKT
    YERLLSIAETTPDFQEVEEKNG
    KVKRVKRSPFAVYCEENDIPAI
    QKYAKKNNGPLIRSLKYYDGKL
    NKHINITKDSQGRPVEKTKNGR
    KVTLQSLKPYRYDIYQDLETKA
    YYTVQLYYSDLRFVEGKYGITE
    KEYMKKVAEQTKGQVVRFCFSL
    QKNDGLEIEWKDSQRYDVRFYN
    FQSANSINFKGLEQEMMPAENQ
    FKQKPYNNGAINLNIAKYGKEG
    KKLRKFNTDILGKKHYLFYEKE
    PKNIIK (SEQ ID NO: 53)
    Mycoplasma MYFYKNKENKLNKKVVLGLDLG 179 226 48 314 374 79 314 374 79
    mobile 163K IASVGWCLTDISQKEDNKFPII
    gi|47458868| LHGVRLFETVDDSDDKLLNETR
    ref|YP_015730.1| RKKRGQRRRNRRLFTRKRDFIK
    YLIDNNIIELEFDKNPKILVRN
    FIEKYINPFSKNLELKYKSVTN
    LPIGFHNLRKAAINEKYKLDKS
    ELIVLLYFYLSLRGAFFDNPED
    TKSKEMNKNEIEIFDKNESIKN
    AEFPIDKIIEFYKISGKIRSTI
    NLKFGHQDYLKEIKQVFEKQNI
    DFMNYEKFAMEEKSFFSRIRNY
    SEGPGNEKSFSKYGLYANENGN
    PELIINEKGQKIYTKIFKTLWE
    SKIGKCSYDKKLYRAPKNSFSA
    KVFDITNKLTDWKHKNEYISER
    LKRKILLSRFLNKDSKSAVEKI
    LKEENIKFENLSEIAYNKDDNK
    INLPIINAYHSLTTIFKKHLIN
    FENYLISNENDLSKLMSFYKQQ
    SEKLFVPNEKGSYEINQNNNVL
    HIFDAISNILNKFSTIQDRIRI
    LEGYFEFSNLKKDVKSSEIYSE
    IAKLREFSGTSSLSFGAYYKFI
    PNLISEGSKNYSTISYEEKALQ
    NQKNNFSHSNLFEKTWVEDLIA
    SPTVKRSLRQTMNLLKEIFKYS
    EKNNLEIEKIVVEVTRSSNNKH
    ERKKIEGINKYRKEKYEELKKV
    YDLPNENTTLLKKLWLLRQQQG
    YDAYSLRKIEANDVINKPWNYD
    IDHIVPRSISFDDSFSNLVIVN
    KLDNAKKSNDLSAKQFIEKIYG
    IEKLKEAKENWGNWYLRNANGK
    AFNDKGKFIKLYTIDNLDEFDN
    SDFINRNLSDTSYITNALVNHL
    TFSNSKYKYSVVSVNGKQTSNL
    RNQIAFVGIKNNKETEREWKRP
    EGFKSINSNDFLIREEGKNDVK
    DDVLIKDRSFNGHHAEDAYFIT
    IISQYFRSFKRIERLNVNYRKE
    TRELDDLEKNNIKFKEKASFDN
    FLLINALDELNEKLNQMRFSRM
    VITKKNTQLFNETLYSGKYDKG
    KNTIKKVEKLNLLDNRTDKIKK
    IEEFFDEDKLKENELTKLHIFN
    HDKNLYETLKIIWNEVKIEIKN
    KNLNEKNYFKYFVNKKLQEGKI
    SFNEWVPILDNDFKIIRKIRYI
    KFSSEEKETDEIIFSQSNFLKI
    DQRQNFSFHNTLYWVQIWVYKN
    QKDQYCFISIDARNSKFEKDEI
    KINYEKLKTQKEKLQIINEEPI
    LKINKGDLFENEEKELFYIVGR
    DEKPQKLEIKYILGKKIKDQKQ
    IQKPVKKYFPNWKKVNLTYMGE
    IFKK (SEQ ID NO: 54)
    Actinomyces MDNKNYRIGIDVGLNSIGFCAV 147 193 47 358 418 40 358 418 40
    coleocanis EVDQHDTPLGFLNLSVYRHDAG
    DSM 15436 IDPNGKKTNTTRLAMSGVARRT
    gi|227494853| RRLFRKRKRRLAALDRFIEAQG
    ref|ZP_03925169.1 WTLPDHADYKDPYTPWLVRAEL
    AQTPIRDENDLHEKLAIAVRHI
    ARHRGWRSPWVPVRSLHVEQPP
    SDQYLALKERVEAKTLLQMPEG
    ATPAEMVVALDLSVDVNLRPKN
    REKTDTRPENKKPGFLGGKLMQ
    SDNANELRKIAKIQGLDDALLR
    ELIELVFAADSPKGASGELVGY
    DVLPGQHGKRRAEKAHPAFQRY
    RIASIVSNLRIRHLGSGADERL
    DVETQKRVFEYLLNAKPTADIT
    WSDVAEEIGVERNLLMGTATQT
    ADGERASAKPPVDVTNVAFATC
    KIKPLKEWWLNADYEARCVMVS
    ALSHAEKLTEGTAAEVEVAEFL
    QNLSDEDNEKLDSFSLPIGRAA
    YSVDSLERLTKRMIENGEDLFE
    ARVNEFGVSEDWRPPAEPIGAR
    VGNPAVDRVLKAVNRYLMAAEA
    EWGAPLSVNIEHVREGFISKRQ
    AVEIDRENQKRYQRNQAVRSQI
    ADHINATSGVRGSDVTRYLAIQ
    RQNGECLYCGTAITFVNSEMDH
    IVPRAGLGSTNTRDNLVATCER
    CNKSKSNKPFAVWAAECGIPGV
    SVAEALKRVDFWIADGFASSKE
    HRELQKGVKDRLKRKVSDPEID
    NRSMESVAWMARELAHRVQYYF
    DEKHTGTKVRVFRGSLTSAARK
    ASGFESRVNFIGGNGKTRLDRR
    HHAMDAATVAMLRNSVAKTLVL
    RGNIRASERAIGAAETWKSFRG
    ENVADRQIFESWSENMRVLVEK
    FNLALYNDEVSIFSSLRLQLGN
    GKAHDDTITKLQMHKVGDAWSL
    TEIDRASTPALWCALTRQPDFT
    WKDGLPANEDRTIIVNGTHYGP
    LDKVGIFGKAAASLLVRGGSVD
    IGSAIHHARIYRIAGKKPTYGM
    VRVFAPDLLRYRNEDLFNVELP
    PQSVSMRYAEPKVREAIREGKA
    EYLGWLVVGDELLLDLSSETSG
    QIAELQQDFPGTTHWTVAGFFS
    PSRLRLRPVYLAQEGLGEDVSE
    GSKSIIAGQGWRPAVNKVFGSA
    MPEVIRRDGLGRKRRFSYSGLP
    VSWQG (SEQ ID NO: 55)
    Dinoroseobacter MRLGLDIGTSSIGWWLYETDGA 138 184 47 338 398 48 338 398 48
    shibae GSDARITGVVDGGVRIFSDGRD
    DFL 12 PKSGASLAVDRRAARAMRRRRD
    gi|159042956| RYLRRRATLMKVLAETGLMPAD
    ref|YP_001531750.1 PAEAKALEALDPFALRAAGLDE
    PLPLPHLGRALFHLNQRRGFKS
    NRKTDRGDNESGKIKDATARLD
    MEMMANGARTYGEFLHKRRQKA
    TDPRHVPSVRTRLSIANRGGPD
    GKEEAGYDFYPDRRHLEEEFHK
    LWAAQGAHHPELTETLRDLLFE
    KIFFQRPLKEPEVGLCLFSGHH
    GVPPKDPRLPKAHPLTQRRVLY
    ETVNQLRVTADGREARPLTREE
    RDQVIHALDNKKPTKSLSSMVL
    KLPALAKVLKLRDGERFTLETG
    VRDAIACDPLRASPAHPDRFGP
    RWSILDADAQWEVISRIRRVQS
    DAEHAALVDWLTEAHGLDRAHA
    EATAHAPLPDGYGRLGLTATTR
    ILYQLTADVVTYADAVKACGWH
    HSDGRTGECFDRLPYYGEVLER
    HVIPGSYHPDDDDITRFGRITN
    PTVHIGLNQLRRLVNRIIETHG
    KPHQIVVELARDLKKSEEQKRA
    DIKRIRDTTEAAKKRSEKLEEL
    EIEDNGRNRMLLRLWEDLNPDD
    AMRRFCPYTGTRISAAMIFDGS
    CDVDHILPYSRTLDDSFPNRTL
    CLREANRQKRNQTPWQAWGDTP
    HWHAIAANLKNLPENKRWRFAP
    DAMTRFEGENGFLDRALKDTQY
    LARISRSYLDTLFTKGGHVWVV
    PGRFTEMLRRHWGLNSLLSDAG
    RGAVKAKNRTDHRHHAIDAAVI
    AATDPGLLNRISRAAGQGEAAG
    QSAELIARDTPPPWEGFRDDLR
    VRLDRIIVSHRADHGRIDHAAR
    KQGRDSTAGQLHQETAYSIVDD
    IHVASRTDLLSLKPAQLLDEPG
    RSGQVRDPQLRKALRVATGGKT
    GKDFENALRYFASKPGPYQAIR
    RVRIIKPLQAQARVPVPAQDPI
    KAYQGGSNHLFEIWRLPDGEIE
    AQVITSFEAHTLEGEKRPHPAA
    KRLLRVHKGDMVALERDGRRVV
    GHVQKMDIANGLFIVPHNEANA
    DTRNNDKSDPFKWIQIGARPAI
    ASGIRRVSVDEIGRLRDGGTRP
    I (SEQ ID NO: 56)
    Actinomyces MLHCIAVIRVPPSEEPGFFETH 183 228 46 349 409 40 349 409 40
    sp. oral ADSCALCHHGCMTYAANDKAIR
    taxon
    180 YRVGIDVGLRSIGFCAVEVDDE
    str F0310 DHPIRILNSVVHVHDAGTGGPG
    gi|315605738| ETESLRKRSGVAARARRRGRAE
    ref|ZP_07880770.1 KQRLKKLDVLLEELGWGVSSNE
    LLDSHAPWHIRKRLVSEYIEDE
    TERRQCLSVAMAHIARHRGWRN
    SFSKVDTLLLEQAPSDRMQGLK
    ERVEDRTGLQFSEEVTQGELVA
    TLLEHDGDVTIRGFVRKGGKAT
    KVHGVLEGKYMQSDLVAELRQI
    CRTQRVSETTFEKLVLSIFHSK
    EPAPSAARQRERVGLDELQLAL
    DPAAKQPRAERAHPAFQKFKVV
    ATLANMRIREQSAGERSLTSEE
    LNRVARYLLNHTESESPTWDDV
    ARKLEVPRHRLRGSSRASLETG
    GGLTYPPVDDTTVRVMSAEVDW
    LADWWDCANDESRGHMIDAISN
    GCGSEPDDVEDEEVNELISSAT
    AEDMLKLELLAKKLPSGRVAYS
    LKTLREVTAAILETGDDLSQAI
    TRLYGVDPGWVPTPAPIEAPVG
    NPSVDRVLKQVARWLKFASKRW
    GVPQTVNIEHTREGLKSASLLE
    EERERWERFEARREIRQKEMYK
    RLGISGPFRRSDQVRYEILDLQ
    DCACLYCGNEINFQTFEVDHII
    PRVDASSDSRRTNLAAVCHSCN
    SAKGGLAFGQWVKRGDCPSGVS
    LENAIKRVRSWSKDRLGLTEKA
    MGKRKSEVISRLKTEMPYEEFD
    GRSMESVAWMAIELKKRIEGYF
    NSDRPEGCAAVQVNAYSGRLTA
    CARRAAHVDKRVRLIRLKGDDG
    HHKNRFDRRNHAMDALVIALMT
    PAIARTIAVREDRREAQQLTRA
    FESWKNFLGSEERMQDRWESWI
    GDVEYACDRLNELIDADKIPVT
    ENLRLRNSGKLHADQPESLKKA
    RRGSKRPRPQRYVLGDALPADV
    INRVTDPGLWTALVRAPGFDSQ
    LGLPADLNRGLKLRGKRISADF
    PIDYFPTDSPALAVQGGYVGLE
    FHHARLYRIIGPKEKVKYALLR
    VCAIDLCGIDCDDLFEVELKPS
    SISMRTADAKLKEAMGNGSAKQ
    IGWLVLGDEIQIDPTKFPKQSI
    GKFLKECGPVSSWRVSALDTPS
    KITLKPRLLSNEPLLKTSRVGG
    HESDLVVAECVEKIMKKTGWVV
    EINALCQSGLIRVIRRNALGEV
    RTSPKSGLPISLNLR (SEQ
    ID NO: 57)
    Alcanivorax MRYRVGLDLGTASVGAAVFSMD 139 183 45 344 404 61 344 404 61
    sp. W11-5 EQGNPMELIWHYERLFSEPLVP
    gi|407803669| DMGQLKPKKAARRLARQQRRQI
    ref|ZP_11150502.1 DRRASRLRRIAIVSRRLGIAPG
    RNDSGVHGNDVPTLRAMAVNER
    IELGQLRAVLLRMGKKRGYGGT
    FKAVRKVGEAGEVASGASRLEE
    EMVALASVQNKDSVTVGEYLAA
    RVEHGLPSKLKVAANNEYYAPE
    YALFRQYLGLPAIKGRPDCLPN
    MYALRHQIEHEFERIWATQSQF
    HDVMKDHGVKEEIRNAIFFQRP
    LKSPADKVGRCSLQTNLPRAPR
    AQIAAQNFRIEKQMADLRWGMG
    RRAEMLNDHQKAVIRELLNQQK
    ELSFRKIYKELERAGCPGPEGK
    GLNMDRAALGGRDDLSGNTTLA
    AWRKLGLEDRWQELDEVTQIQV
    INFLADLGSPEQLDTDDWSCRF
    MGKNGRPRNFSDEFVAFMNELR
    MTDGFDRLSKMGFEGGRSSYSI
    KALKALTEWMIAPHWRETPETH
    RVDEEAAIRECYPESLATPAQG
    GRQSKLEPPPLTGNEVVDVALR
    QVRHTINMMIDDLGSVPAQIVV
    EMAREMKGGVTRRNDIEKQNKR
    FASERKKAAQSIEENGKTPTPA
    RILRYQLWIEQGHQCPYCESNI
    SLEQALSGAYTNFEHILPRTLT
    QIGRKRSELVLAHRECNDEKGN
    RTPYQAFGHDDRRWRIVEQRAN
    ALPKKSSRKTRLLLLKDFEGEA
    LTDESIDEFADRQLHESSWLAK
    VTTQWLSSLGSDVYVSRGSLTA
    ELRRRWGLDTVIPQVRFESGMP
    VVDEEGAEITPEEFEKFRLQWE
    GHRVTREMRTDRRPDKRIDHRH
    HLVDAIVTALTSRSLYQQYAKA
    WKVADEKQRHGRVDVKVELPMP
    ILTIRDIALEAVRSVRISHKPD
    RYPDGRFFEATAYGIAQRLDER
    SGEKVDWLVSRKSLTDLAPEKK
    SIDVDKVRANISRIVGEAIRLH
    ISNIFEKRVSKGMTPQQALREP
    IEFQGNILRKVRCFYSKADDCV
    RIEHSSRRGHHYKMLLNDGFAY
    MEVPCKEGILYGVPNLVRPSEA
    VGIKRAPESGDFIRFYKGDTVK
    NIKTGRVYTIKQILGDGGGKLI
    LTPVTETKPADLLSAKWGRLKV
    GGRNIHLLRLCAE (SEQ ID
    NO: 58)
    Aminomonas MIGEHVRGGCLFDDHWTPNWGA 134 178 45 341 401 63 341 401 63
    paucivorans FRLPNTVRTFTKAENPKDGSSL
    DSM 12260 AEPRRQARGLRRRLRRKTQRLE
    gi|312879015| DLRRLLAKEGVLSLSDLETLFR
    ref|ZP_07738815.1 ETPAKDPYQLRAEGLDRPLSFP
    EWVRVLYHITKHRGFQSNRRNP
    VEDGQERSRQEEEGKLLSGVGE
    NERLLREGGYRTAGEMLARDPK
    FQDHRRNRAGDYSHTLSRSLLL
    EEARRLFQSQRTLGNPHASSNL
    EEAFLHLVAFQNPFASGEDIRN
    KAGHCSLEPDQIRAPRRSASAE
    TFMLLQKTGNLRLIHRRTGEER
    PLTDKEREQIHLLAWKQEKVTH
    KTLRRHLEIPEEWLFTGLPYHR
    SGDKAEEKLFVHLAGIHEIRKA
    LDKGPDPAVWDTLRSRRDLLDS
    IADTLTFYKNEDEILPRLESLG
    LSPENARALAPLSFSGTAHLSL
    SALGKLLPHLEEGKSYTQARAD
    AGYAAPPPDRHPKLPPLEEADW
    RNPVVFRALTQTRKVVNALVRR
    YGPPWCIHLETARELSQPAKVR
    RRIETEQQANEKKKQQAEREFL
    DIVGTAPGPGDLLKMRLWREQG
    GFCPYCEEYLNPTRLAEPGYAE
    MDHILPYSRSLDNGWHNRVLVH
    GKDNRDKGNRTPFEAFGGDTAR
    WDRLVAWVQASHLSAPKKRNLL
    REDFGEEAERELKDRNLTDTRF
    ITKTAATLLRDRLTFHPEAPKD
    PVMTLNGRLTAFLRKQWGLHKN
    RKNGDLHHALDAAVLAVASRSF
    VYRLSSHNAAWGELPRGREAEN
    GFSLPYPAFRSEVLARLCPTRE
    EILLRLDQGGVGYDEAFRNGLR
    PVFVSRAPSRRLRGKAHMETLR
    SPKWKDHPEGPRTASRIPLKDL
    NLEKLERMVGKDRDRKLYEALR
    ERLAAFGGNGKKAFVAPFRKPC
    RSGEGPLVRSLRIFDSGYSGVE
    LRDGGEVYAVADHESMVRVDVY
    AKKNRFYLVPVYVADVARGIVK
    NRAIVAHKSEEEWDLVDGSFDF
    RFSLFPGDLVEIEKKDGAYLGY
    YKSCHRGDGRLLLDRHDRMPRE
    SDCGTFYVSTRKDVLSMSKYQV
    DPLGEIRLVGSEKPPFVL
    (SEQ ID NO: 59)
    Mycoplasma MEKKRKVTLGFDLGIASVGWAI 139 183 45 319 379 76 319 379 76
    canis PG 14 VDSETNQVYKLGSRLFDAPDTN
    gi|384393286| LERRTQRGTRRLLRRRKYRNQK
    gb|EIE39736.1 FYNLVKRTEVFGLSSREAIENR
    FRELSIKYPNIIELKTKALSQE
    VCPDEIAWILHDYLKNRGYFYD
    EKETKEDFDQQTVESMPSYKLN
    EFYKKYGYFKGALSQPTESEMK
    DNKDLKEAFFFDFSNKEWLKEI
    NYFFNVQKNILSETFIEEFKKI
    FSFTRDISKGPGSDNMPSPYGI
    FGEFGDNGQGGRYEHIWDKNIG
    KCSIFTNEQRAPKYLPSALIFN
    FLNELANIRLYSTDKKNIQPLW
    KLSSVDKLNILLNLFNLPISEK
    KKKLTSTNINDIVKKESIKSIM
    ISVEDIDMIKDEWAGKEPNVYG
    VGLSGLNIEESAKENKFKFQDL
    KILNVLINLLDNVGIKFEFKDR
    NDIIKNLELLDNLYLFLIYQKE
    SNNKDSSIDLFIAKNESLNIEN
    LKLKLKEFLLGAGNEFENHNSK
    THSLSKKAIDEILPKLLDNNEG
    WNLEAIKNYDEEIKSQIEDNSS
    LMAKQDKKYLNDNFLKDAILPP
    NVKVTFQQAILIFNKIIQKFSK
    DFEIDKVVIELAREMTQDQEND
    ALKGIAKAQKSKKSLVEERLEA
    NNIDKSVFNDKYEKLIYKIFLW
    ISQDFKDPYTGAQISVNEIVNN
    KVEIDHIIPYSLCFDDSSANKV
    LVHKQSNQEKSNSLPYEYIKQG
    HSGWNWDEFTKYVKRVFVNNVD
    SILSKKERLKKSENLLTASYDG
    YDKLGFLARNLNDTRYATILFR
    DQLNNYAEHHLIDNKKMFKVIA
    MNGAVTSFIRKNMSYDNKLRLK
    DRSDFSHHAYDAAIIALFSNKT
    KTLYNLIDPSLNGIISKRSEGY
    WVIEDRYTGEIKELKKEDWTSI
    KNNVQARKIAKEIEEYLIDLDD
    EVFFSRKTKRKTNRQLYNETIY
    GIATKTDEDGITNYYKKEKFSI
    LDDKDIYLRLLREREKFVINQS
    NPEVIDQIIEIIESYGKENNIP
    SRDEAINIKYTKNKINYNLYLK
    QYMRSLTKSLDQFSEEFINQMI
    ANKTFVLYNPTKNTTRKIKFLR
    LVNDVKINDIRKNQVINKFNGK
    NNEPKAFYENINSLGAIVFKNS
    ANNFKTLSINTQIAIFGDKNWD
    IEDFKTYNMEKIEKYKEIYGID
    KTYNFHSFIFPGTILLDKQNKE
    FYYISSIQTVRDIIEIKFLNKI
    EFKDENKNQDTSKTPKRLMFGI
    KSIMNNYEQVDISPFGINKKIF
    E (SEQ ID NO: 60)
    Lactobacillus MGYRIGLDVGITSTGYAVLKTD 141 184 44 328 387 61 328 387 61
    coryniformis KNGLPYKILTLDSVIYPRAENP
    KCTC 3535 QTGASLAEPRRIKRGLRRRTRR
    gi|336393381| TKFRKQRTQQLFIHSGLLSKPE
    ref|ZP_08574780.1 IEQILATPQAKYSVYELRVAGL
    DRRLTNSELFRVLYFFIGHRGF
    KSNRKAELNPENEADKKQMGQL
    LNSIEEIRKAIAEKGYRTVGEL
    YLKDPKYNDHKRNKGYIDGYLS
    TPNRQMLVDEIKQILDKQRELG
    NEKLTDEFYATYLLGDENRAGI
    FQAQRDFDEGPGAGPYAGDQIK
    KMVGKDIFEPTEDRAAKATYTF
    QYFNLLQKMTSLNYQNTTGDTW
    HTLNGLDRQAIIDAVFAKAEKP
    TKTYKPTDFGELRKLLKLPDDA
    RFNLVNYGSLQTQKEIETVEKK
    TRFVDFKAYHDLVKVLPEEMWQ
    SRQLLDHIGTALTLYSSDKRRR
    RYFAEELNLPAELIEKLLPLNF
    SKFGHLSIKSMQNIIPYLEMGQ
    VYSEATTNTGYDFRKKQISKDT
    IREEITNPVVRRAVTKTIKIVE
    QIIRRYGKPDGINIELARELGR
    NFKERGDIQKRQDKNRQTNDKI
    AAELTELGIPVNGQNIIRYKLH
    KEQNGVDPYTGDQIPFERAFSE
    GYEVDHIIPYSISWDDSYTNKV
    LTSAKCNREKGNRIPMVYLANN
    EQRLNALTNIADNIIRNSRKRQ
    KLLKQKLSDEELKDWKQRNIND
    TRFITRVLYNYFRQAIEFNPEL
    EKKQRVLPLNGEVTSKIRSRWG
    FLKVREDGDLHHAIDATVIAAI
    TPKFIQQVTKYSQHQEVKNNQA
    LWHDAEIKDAEYAAEAQRMDAD
    LFNKIFNGFPLPWPEFLDELLA
    RISDNPVEMMKSRSWNTYTPIE
    IAKLKPVFVVRLANHKISGPAH
    LDTIRSAKLFDEKGIVLSRVSI
    TKLKINKKGQVATGDGIYDPEN
    SNNGDKVVYSAIRQALEAHNGS
    GELAFPDGYLEYVDHGTKKLVR
    KVRVAKKVSLPVRLKNKAAADN
    GSMVRIDVFNTGKKFVFVPIYI
    KDTVEQVLPNKAIARGKSLWYQ
    ITESDQFCFSLYPGDMVHIESK
    TGIKPKYSNKENNTSVVPIKNF
    YGYFDGADIATASILVRAHDSS
    YTARSIGIAGLLKFEKYQVDYF
    GRYHKVHEKKRQLFVKRDE
    (SEQ ID NO: 61)
    Elusimicrobium MQKNINTKQNHIYIKQAQKIKE 177 219 43 322 381 47 322 381 47
    minutum KLGDKPYRIGLDLGVGSIGFAI
    Pei191 VSMEENDGNVLLPKEIIMVGSR
    gi|187250660| IFKASAGAADRKLSRGQRNNHR
    ref|YP_001875142.1 HTRERMRYLWKVLAEQKLALPV
    PADLDRKENSSEGETSAKRFLG
    DVLQKDIYELRVKSLDERLSLQ
    ELGYVLYHIAGHRGSSAIRTFE
    NDSEEAQKENTENKKIAGNIKR
    LMAKKNYRTYGEYLYKEFFENK
    EKHKREKISNAANNHKFSPTRD
    LVIKEAEAILKKQAGKDGFHKE
    LTEEYIEKLTKAIGYESEKLIP
    ESGFCPYLKDEKRLPASHKLNE
    ERRLWETLNNARYSDPIVDIVT
    GEITGYYEKQFTKEQKQKLFDY
    LLTGSELTPAQTKKLLGLKNTN
    FEDIILQGRDKKAQKIKGYKLI
    KLESMPFWARLSEAQQDSFLYD
    WNSCPDEKLLTEKLSNEYHLTE
    EEIDNAFNEIVLSSSYAPLGKS
    AMLIILEKIKNDLSYTEAVEEA
    LKEGKLTKEKQAIKDRLPYYGA
    VLQESTQKIIAKGFSPQFKDKG
    YKTPHTNKYELEYGRIANPVVH
    QTLNELRKLVNEIIDILGKKPC
    EIGLETARELKKSAEDRSKLSR
    EQNDNESNRNRIYEIYIRPQQQ
    VIITRRENPRNYILKFELLEEQ
    KSQCPFCGGQISPNDIINNQAD
    IEHLFPIAESEDNGRNNLVISH
    SACNADKAKRSPWAAFASAAKD
    SKYDYNRILSNVKENIPHKAWR
    FNQGAFEKFIENKPMAARFKTD
    NSYISKVAHKYLACLFEKPNII
    CVKGSLTAQLRMAWGLQGLMIP
    FAKQLITEKESESFNKDVNSNK
    KIRLDNRHHALDAIVIAYASRG
    YGNLLNKMAGKDYKINYSERNW
    LSKILLPPNNIVWENIDADLES
    FESSVKTALKNAFISVKHDHSD
    NGELVKGTMYKIFYSERGYTLT
    TYKKLSALKLTDPQKKKTPKDF
    LETALLKFKGRESEMKNEKIKS
    AIENNKRLFDVIQDNLEKAKKL
    LEEENEKSKAEGKKEKNINDAS
    IYQKAISLSGDKYVQLSKKEPG
    KFFAISKPTPTTTGYGYDTGDS
    LCVDLYYDNKGKLCGEIIRKID
    AQQKNPLKYKEQGFTLFERIYG
    GDILEVDFDIHSDKNSFRNNTG
    SAPENRVFIKVGTFTEITNNNI
    QIWFGNIIKSTGGQDDSFTINS
    MQQYNPRKLILSSCGFIKYRSP
    ILKNKEG (SEQ ID NO:
    62)
    Neisseria MAAFKPNPINYILGLDIGIASV 147 189 43 360 419 61 360 419 61
    meningitidis GWAMVEIDEDENPICLIDLGVR
    Z2491 VFERAEVPKTGDSLAMARRLAR
    gi|218767588| SVRRLTRRRAHRLLRARRLLKR
    ref|YP_002342100.1 EGVLQAADFDENGLIKSLPNTP
    WQLRAAALDRKLTPLEWSAVLL
    HLIKHRGYLSQRKNEGETADKE
    LGALLKGVADNAHALQTGDFRT
    PAELALNKFEKESGHIRNQRGD
    YSHTFSRKDLQAELILLFEKQK
    EFGNPHVSGGLKEGIETLLMTQ
    RPALSGDAVQKMLGHCTFEPAE
    PKAAKNTYTAERFIWLTKLNNL
    RILEQGSERPLTDTERATLMDE
    PYRKSKLTYAQARKLLGLEDTA
    FFKGLRYGKDNAEASTLMEMKA
    YHAISRALEKEGLKDKKSPLNL
    SPELQDEIGTAFSLFKTDEDIT
    GRLKDRIQPEILEALLKHISFD
    KFVQISLKALRRIVPLMEQGKR
    YDEACAEIYGDHYGKKNTEEKI
    YLPPIPADEIRNPVVLRALSQA
    RKVINGVVRRYGSPARIHIETA
    REVGKSFKDRKEIEKRQEENRK
    DREKAAAKFREYFPNFVGEPKS
    KDILKLRLYEQQHGKCLYSGKE
    INLGRLNEKGYVEIDHALPFSR
    TWDDSFNNKVLVLGSENQNKGN
    QTPYEYFNGKDNSREWQEFKAR
    VETSRFPRSKKQRILLQKFDED
    GFKERNLNDTRYVNRFLCQFVA
    DRMRLTGKGKKRVFASNGQITN
    LLRGFWGLRKVRAENDRHHALD
    AVVVACSTVAMQQKITRFVRYK
    EMNAFDGKTIDKETGEVLHQKT
    HFPQPWEFFAQEVMIRVFGKPD
    GKPEFEEADTPEKLRTLLAEKL
    SSRPEAVHEYVTPLFVSRAPNR
    KMSGQGHMETVKSAKRLDEGVS
    VLRVPLTQLKLKDLEKMVNRER
    EPKLYEALKARLEAHKDDPAKA
    FAEPFYKYDKAGNRTQQVKAVR
    VEQVQKTGVWVRNHNGIADNAT
    MVRVDVFEKGDKYYLVPIYSWQ
    VAKGILPDRAVVQGKDEEDWQL
    IDDSFNFKFSLHPNDLVEVITK
    KARMFGYFASCHRGTGNINIRI
    HDLDHKIGKNGILEGIGVKTAL
    SFQKYQIDELGKEIRPCRLKKR
    PPVR (SEQ ID NO: 63)
    Pasteurella MQTTNLSYILGLDLGIASVGWA 139 181 43 319 378 61 319 378 61
    multocida VVEINENEDPIGLIDVGVRIFE
    str. Pm70 RAEVPKTGESLALSRRLARSTR
    gi|15602992| RLIRRRAHRLLLAKRFLKREGI
    ref|NP_246064.1| LSTIDLEKGLPNQAWELRVAGL
    ERRLSAIEWGAVLLHLIKHRGY
    LSKRKNESQTNNKELGALLSGV
    AQNHQLLQSDDYRTPAELALKK
    FAKEEGHIRNQRGAYTHTFNRL
    DLLAELNLLFAQQHQFGNPHCK
    EHIQQYMTELLMWQKPALSGEA
    ILKMLGKCTHEKNEFKAAKHTY
    SAERFVWLTKLNNLRILEDGAE
    RALNEEERQLLINHPYEKSKLT
    YAQVRKLLGLSEQAIFKHLRYS
    KENAESATFMELKAWHAIRKAL
    ENQGLKDTWQDLAKKPDLLDEI
    GTAFSLYKTDEDIQQYLTNKVP
    NSVINALLVSLNFDKFIELSLK
    SLRKILPLMEQGKRYDQACREI
    YGHHYGEANQKTSQLLPAIPAQ
    EIRNPVVLRTLSQARKVINAII
    RQYGSPARVHIETGRELGKSFK
    ERREIQKQQEDNRTKRESAVQK
    FKELFSDFSSEPKSKDILKFRL
    YEQQHGKCLYSGKEINIHRLNE
    KGYVEIDHALPFSRTWDDSFNN
    KVLVLASENQNKGNQTPYEWLQ
    GKINSERWKNFVALVLGSQCSA
    AKKQRLLTQVIDDNKFIDRNLN
    DTRYIARFLSNYIQENLLLVGK
    NKKNVFTPNGQITALLRSRWGL
    IKARENNNRHHALDAIVVACAT
    PSMQQKITRFIRFKEVHPYKIE
    NRYEMVDQESGEIISPHFPEPW
    AYFRQEVNIRVFDNHPDTVLKE
    MLPDRPQANHQFVQPLFVSRAP
    TRKMSGQGHMETIKSAKRLAEG
    ISVLRIPLTQLKPNLLENMVNK
    EREPALYAGLKARLAEFNQDPA
    KAFATPFYKQGGQQVKAIRVEQ
    VQKSGVLVRENNGVADNASIVR
    TDVFIKNNKFFLVPIYTWQVAK
    GILPNKAIVAHKNEDEWEEMDE
    GAKFKFSLFPNDLVELKTKKEY
    FFGYYIGLDRATGNISLKEHDG
    EISKGKDGVYRVGVKLALSFEK
    YQVDELGKNRQICRPQQRQPVR
    (SEQ ID NO: 64)
    Rhodovulum MGIRFAFDLGTNSIGWAVWRTG 141 183 43 319 378 48 319 378 48
    sp. PH10 PGVFGEDTAASLDGSGVLIFKD
    gi|402849997| GRNPKDGQSLATMRRVPRQSRK
    ref|ZP_10898214.1 RRDRFVLRRRDLLAALRKAGLF
    PVDVEEGRRLAATDPYHLRAKA
    LDESLTPHEMGRVIFHLNQRRG
    FRSNRKADRQDREKGKIAEGSK
    RLAETLAATNCRTLGEFLWSRH
    RGTPRTRSPTRIRMEGEGAKAL
    YAFYPTREMVRAEFERLWTAQS
    RFAPDLLTPERHEEIAGILFRQ
    RDLAPPKIGCCTFEPSERRLPR
    ALPSVEARGIYERLAHLRITTG
    PVSDRGLTRPERDVLASALLAG
    KSLTFKAVRKTLKILPHALVNF
    EEAGEKGLDGALTAKLLSKPDH
    YGAAWHGLSFAEKDTFVGKLLD
    EADEERLIRRLVTENRLSEDAA
    RRCASIPLADGYGRLGRTANTE
    ILAALVEETDETGTVVTYAEAV
    RRAGERTGRNWHHSDERDGVIL
    DRLPYYGEILQRHVVPGSGEPE
    EKNEAARWGRLANPTVHIGLNQ
    LRKVVNRLIAAHGRPDQIVVEL
    ARELKLNREQKERLDRENRKNR
    EENERRTAILAEHGQRDTAENK
    IRLRLFEEQARANAGIALCPYT
    GRAIGIAELFTSEVEIDHILPV
    SLTLDDSLANRVLCRREANREK
    RRQTPFQAFGATPAWNDIVARA
    AKLPPNKRWRFDPAALERFERE
    GGFLGRQLNETKYLSRLAKIYL
    GKICDPDRVYVTPGTLTGLLRA
    RWGLNSILSDSNFKNRSDHRHH
    AVDAVVIGVLTRGMIQRIAHDA
    ARAEDQDLDRVFRDVPVPFEDF
    RDHVRERVSTITVAVKPEHGKG
    GALHEDTSYGLVPDTDPNAALG
    NLVVRKPIRSLTAGEVDRVRDR
    ALRARLGALAAPFRDESGRVRD
    AKGLAQALEAFGAENGIRRVRI
    LKPDASVVTIADRRTGVPYRAV
    APGENHHVDIVQMRDGSWRGFA
    ASVFEVNRPGWRPEWEVKKLGG
    KLVMRLHKGDMVELSDKDGQRR
    VKVVQQIEISANRVRLSPHNDG
    GKLQDRHADADDPFRWDLATIP
    LLKDRGCVAVRVDPIGVVTLRR
    SNV (SEQ ID NO: 65)
    Eubacterium MMEVFMGRLVLGLDIGITSVGF 131 172 42 303 361 59 303 361 59
    dolichum GIIDLDESEIVDYGVRLFKEGT
    DSM 3991 AAENETRRTKRGGRRLKRRRVT
    gi|160915782| RREDMLHLLKQAGIISTSFHPL
    ref|ZP_02077990.1 NNPYDVRVKGLNERLNGEELAT
    ALLHLCKHRGSSVETIEDDEAK
    AKEAGETKKVLSMNDQLLKSGK
    YVCEIQKERLRTNGHIRGHENN
    FKTRAYVDEAFQILSHQDLSNE
    LKSAIITIISRKRMYYDGPGGP
    LSPTPYGRYTYFGQKEPIDLIE
    KMRGKCSLFPNEPRAPKLAYSA
    ELFNLLNDLNNLSIEGEKLTSE
    QKAMILKIVHEKGKITPKQLAK
    EVGVSLEQIRGFRIDTKGSPLL
    SELTGYKMIREVLEKSNDEHLE
    DHVFYDEIAEILTKTKDIEGRK
    KQISELSSDLNEESVHQLAGLT
    KFTAYHSLSFKALRLINEEMLK
    TELNQMQSITLFGLKQNNELSV
    KGMKNIQADDTAILSPVAKRAQ
    RETFKVVNRLREIYGEFDSWV
    EMAREKNSEEQRKAIRERQKFF
    EMRNKQVADIIGDDRKINAKLR
    EKLVLYQEQDGKTAYSLEPIDL
    KLLIDDPNAYEVDHIIPISISL
    DDSITNKVLVTHRENQEKGNLT
    PISAFVKGRFTKGSLAQYKAYC
    LKLKEKNIKTNKGYRKKVEQYL
    LNENDIYKYDIQKEFINRNLVD
    TSYASRVVLNTLTTYFKQNEIP
    TKVFTVKGSLTNAFRRKINLKK
    DRDEDYGHHAIDALIIASMPKM
    RLLSTIFSRYKIEDIYDESTGE
    VFSSGDDSMYYDDRYFAFIASL
    KAIKVRKFSHKIDTKPNRSVAD
    ETIYSTRVIDGKEKVVKKYKDI
    YDPKFTALAEDILNNAYQEKYL
    MALHDPQTFDQIVKVVNYYFEE
    MSKSEKYFTKDKKGRIKISGMN
    PLSLYRDEHGMLKKYSKKGDGP
    AITQMKYFDGVLGNHIDISAHY
    QVRDKKVVLQQISPYRTDFYYS
    KENGYKFVTIRYKDVRWSEKKK
    KYVIDQQDYAMKKAEKKIDDTY
    EFQFSMHRDELIGITKAEGEAL
    IYPDETWHNFNFFFHAGETPEI
    LKFTATNNDKSNKIEVKPIHCY
    CKMRLMPTISKKIVRIDKYATD
    VVGNLYKVKKNTLKFEFD
    (SEQ ID NO: 66)
    Nitratifractor MKKILGVDLGITSFGYAILQET 143 184 42 347 404 61 347 404 61
    salsuginis GKDLYRCLDNSVVMRNNPYDEK
    DSM 16511 SGESSQSIRSTQKSMRRLIEKR
    gi|319957206| KKRIRCVAQTMERYGILDYSET
    ref|YP_004168469.1 MKINDPKNNPIKNRWQLRAVDA
    WKRPLSPQELFAIFAHMAKHRG
    YKSIATEDLIYELELELGLNDP
    EKESEKKADERRQVYNALRHLE
    ELRKKYGGETIAQTIHRAVEAG
    DLRSYRNHDDYEKMIRREDIEE
    EIEKVLLRQAELGALGLPEEQV
    SELIDELKACITDQEMPTIDES
    LFGKCTFYKDELAAPAYSYLYD
    LYRLYKKLADLNIDGYEVTQED
    REKVIEWVEKKIAQGKNLKKIT
    HKDLRKILGLAPEQKIFGVEDE
    RIVKGKKEPRTFVPFFFLADIA
    KFKELFASIQKHPDALQIFREL
    AEILQRSKTPQEALDRLRALMA
    GKGIDTDDRELLELFKNKRSGT
    RELSHRYILEALPLFLEGYDEK
    EVQRILGFDDREDYSRYPKSLR
    HLHLREGNLFEKEENPINNHAV
    KSLASWALGLIADLSWRYGPFD
    EIILETTRDALPEKIRKEIDKA
    MREREKALDKIIGKYKKEFPSI
    DKRLARKIQLWERQKGLDLYSG
    KVINLSQLLDGSADIEHIVPQS
    LGGLSTDYNTIVTLKSVNAAKG
    NRLPGDWLAGNPDYRERIGMLS
    EKGLIDWKKRKNLLAQSLDEIY
    TENTHSKGIRATSYLEALVAQV
    LKRYYPFPDPELRKNGIGVRMI
    PGKVTSKTRSLLGIKSKSRETN
    FHHAEDALILSTLTRGWQNRLH
    RMLRDNYGKSEAELKELWKKYM
    PHIEGLTLADYIDEAFRRFMSK
    GEESLFYRDMFDTIRSISYWVD
    KKPLSASSHKETVYSSRHEVPT
    LRKNILEAFDSLNVIKDRHKLT
    TEEFMKRYDKEIRQKLWLHRIG
    NTNDESYRAVEERATQIAQILT
    RYQLMDAQNDKEIDEKFQQALK
    ELITSPIEVTGKLLRKMRFVYD
    KLNAMQIDRGLVETDKNMLGIH
    ISKGPNEKLIFRRMDVNNAHEL
    QKERSGILCYLNEMLFIFNKKG
    LIHYGCLRSYLEKGQGSKYIAL
    FNPRFPANPKAQPSKFTSDSKI
    KQVGIGSATGIIKAHLDLDGHV
    RSYEVFGTLPEGSIEWFKEESG
    YGRVEDDPHH (SEQ ID NO:
    67)
    Rhodospirillum MRPIEPWILGLDIGTDSLGWAV 139 180 42 314 371 55 314 371 55
    rubrum FSCEEKGPPTAKELLGGGVRLF
    ATCC 11170 DSGRDAKDHTSRQAERGAFRRA
    gi|83591793| RRQTRTWPWRRDRLIALFQAAG
    ref|YP_425545.1| LTPPAAETRQIALALRREAVSR
    PLAPDALWAALLHLAHHRGFRS
    NRIDKRERAAAKALAKAKPAKA
    TAKATAPAKEADDEAGFWEGAE
    AALRQRMAASGAPTVGALLADD
    LDRGQPVRMRYNQSDRDGVVAP
    TRALIAEELAEIVARQSSAYPG
    LDWPAVTRLVLDQRPLRSKGAG
    PCAFLPGEDRALRALPTVQDFI
    IRQTLANLRLPSTSADEPRPLT
    DEEHAKALALLSTARFVEWPAL
    RRALGLKRGVKFTAETERNGAK
    QAARGTAGNLTEAILAPLIPGW
    SGWDLDRKDRVFSDLWAARQDR
    SALLALIGDPRGPTRVTEDETA
    EAVADAIQIVLPTGRASLSAKA
    ARAIAQAMAPGIGYDEAVTLAL
    GLHHSHRPRQERLARLPYYAAA
    LPDVGLDGDPVGPPPAEDDGAA
    AEAYYGRIGNISVHIALNETRK
    IVNALLHRHGPILRLVMVETTR
    ELKAGADERKRMIAEQAERERE
    NAEIDVELRKSDRWMANARERR
    QRVRLARRQNNLCPYTSTPIGH
    ADLLGDAYDIDHVIPLARGGRD
    SLDNMVLCQSDANKTKGDKTPW
    EAFHDKPGWIAQRDDFLARLDP
    QTAKALAWRFADDAGERVARKS
    AEDEDQGFLPRQLTDTGYIARV
    ALRYLSLVTNEPNAVVATNGRL
    TGLLRLAWDITPGPAPRDLLPT
    PRDALRDDTAARRFLDGLTPPP
    LAKAVEGAVQARLAALGRSRVA
    DAGLADALGLTLASLGGGGKNR
    ADHRHHFIDAAMIAVTTRGLIN
    QINQASGAGRILDLRKWPRTNF
    EPPYPTFRAEVMKQWDHIHPSI
    RPAHRDGGSLHAATVFGVRNRP
    DARVLVQRKPVEKLFLDANAKP
    LPADKIAEIIDGFASPRMAKRF
    KALLARYQAAHPEVPPALAALA
    VARDPAFGPRGMTANTVIAGRS
    DGDGEDAGLITPFRANPKAAVR
    TMGNAVYEVWEIQVKGRPRWTH
    RVLTRFDRTQPAPPPPPENARL
    VMRLRRGDLVYWPLESGDRLFL
    VKKMAVDGRLALWPARLATGKA
    TALYAQLSCPNINLNGDQGYCV
    QSAEGIRKEKIRTTSCTALGRL
    RLSKKAT (SEQ ID NO:
    68)
    Clostridium MKYTLGLDVGIASVGWAVIDKD 137 176 40 320 376 61 320 376 61
    cellulolyticum NNKIIDLGVRCFDKAEESKTGE
    H10 SLATARRIARGMRRRISRRSQR
    gi|220930482| LRLVKKLFVQYEIIKDSSEFNR
    ref|YP_002507391.1 IFDTSRDGWKDPWELRYNALSR
    ILKPYELVQVLTHITKRRGFKS
    NRKEDLSTTKEGVVITSIKNNS
    EMLRTKNYRTIGEMIFMETPEN
    SNKRNKVDEYIHTIAREDLLNE
    IKYIFSIQRKLGSPFVTEKLEH
    DFLNIWEFQRPFASGDSILSKV
    GKCTLLKEELRAPTSCYTSEYF
    GLLQSINNLVLVEDNNTLTLNN
    DQRAKIIEYAHFKNEIKYSEIR
    KLLDIEPEILFKAHNLTHKNPS
    GNNESKKFYEMKSYHKLKSTLP
    TDIWGKLHSNKESLDNLFYCLT
    VYKNDNEIKDYLQANNLDYLIE
    YIAKLPTFNKFKHLSLVAMKRI
    IPFMEKGYKYSDACNMAELDFT
    GSSKLEKCNKLTVEPIIENVTN
    PVVIRALTQARKVINAIIQKYG
    LPYMVNIELAREAGMTRQDRDN
    LKKEHENNRKAREKISDLIRQN
    GRVASGLDILKWRLWEDQGGRC
    AYSGKPIPVCDLLNDSLTQIDH
    IYPYSRSMDDSYMNKVLVLTDE
    NQNKRSYTPYEVWGSTEKWEDF
    EARIYSMHLPQSKEKRLLNRNF
    ITKDLDSFISRNLNDTRYISRF
    LKNYIESYLQFSNDSPKSCVVC
    VNGQCTAQLRSRWGLNKNREES
    DLHHALDAAVIACADRKIIKEI
    TNYYNERENHNYKVKYPLPWHS
    FRQDLMETLAGVFISRAPRRKI
    TGPAHDETIRSPKHFNKGLTSV
    KIPLTTVTLEKLETMVKNTKGG
    ISDKAVYNVLKNRLIEHNNKPL
    KAFAEKIYKPLKNGTNGAIIRS
    IRVETPSYTGVFRNEGKGISDN
    SLMVRVDVFKKKDKYYLVPIYV
    AHMIKKELPSKAIVPLKPESQW
    ELIDSTHEFLFSLYQNDYLVIK
    TKKGITEGYYRSCHRGTGSLSL
    MPHFANNKNVKIDIGVRTAISI
    EKYNVDILGNKSIVKGEPRRGM
    EKYNSFKSN (SEQ ID NO:
    69)
    Helicobacter MIRTLGIDIGIASIGWAVIEGE 148 187 40 298 354 48 298 354 48
    mustelae YTDKGLENKEIVASGVRVFTKA
    12198 ENPKNKESLALPRTLARSARRR
    gi|291276265| NARKKGRIQQVKHYLSKALGLD
    ref|YP_003516037.1 LECFVQGEKLATLFQTSKDFLS
    PWELRERALYRVLDKEELARVI
    LHIAKRRGYDDITYGVEDNDSG
    KIKKAIAENSKRIKEEQCKTIG
    EMMYKLYFQKSLNVRNKKESYN
    RCVGRSELREELKTIFQIQQEL
    KSPWVNEELIYKLLGNPDAQSK
    QEREGLIFYQRPLKGFGDKIGK
    CSHIKKGENSPYRACKHAPSAE
    EFVALTKSINFLKNLTNRHGLC
    FSQEDMCVYLGKILQEAQKNEK
    GLTYSKLKLLLDLPSDFEFLGL
    DYSGKNPEKAVFLSLPSTFKLN
    KITQDRKTQDKIANILGANKDW
    EAILKELESLQLSKEQIQTIKD
    AKLNFSKHINLSLEALYHLLPL
    MREGKRYDEGVEILQERGIFSK
    PQPKNRQLLPPLSELAKEESYF
    DIPNPVLRRALSEFRKVVNALL
    EKYGGFHYFHIELTRDVCKAKS
    ARMQLEKINKKNKSENDAASQL
    LEVLGLPNTYNNRLKCKLWKQQ
    EEYCLYSGEKITIDHLKDQRAL
    QIDHAFPLSRSLDDSQSNKVLC
    LTSSNQEKSNKTPYEWLGSDEK
    KWDMYVGRVYSSNFSPSKKRKL
    TQKNFKERNEEDFLARNLVDTG
    YIGRVTKEYIKHSLSFLPLPDG
    KKEHIRIISGSMTSTMRSFWGV
    QEKNRDHHLHHAQDAIIIACIE
    PSMIQKYTTYLKDKETHRLKSH
    QKAQILREGDHKLSLRWPMSNF
    KDKIQESIQNIIPSHHVSHKVT
    GELHQETVRTKEFYYQAFGGEE
    GVKKALKFGKIREINQGIVDNG
    AMVRVDIFKSKDKGKFYAVPIY
    TYDFAIGKLPNKAIVQGKKNGI
    IKDWLEMDENYEFCFSLFKNDC
    IKIQTKEMQEAVLAIYKSTNSA
    KATIELEHLSKYALKNEDEEKM
    FTDTDKEKNKTMTRESCGIQGL
    KVFQKVKLSVLGEVLEHKPRNR
    QNIALKTTPKHV (SEQ ID
    NO: 70)
    Ilyobacter MKYSIGLDIGIASVGWSVINKD 134 173 40 462 517 63 462 517 63
    polytropus KERIEDMGVRIFQKAENPKDGS
    DSM 2926 SLASSRREKRGSRRRNRRKKHR
    gi|310780384| LDRIKNILCESGLVKKNEIEKI
    ref|YP_003968716.1 YKNAYLKSPWELRAKSLEAKIS
    NKEIAQILLHIAKRRGFKSFRK
    TDRNADDTGKLLSGIQENKKIM
    EEKGYLTIGDMVAKDPKFNTHV
    RNKAGSYLFSFSRKLLEDEVRK
    IQAKQKELGNTHFTDDVLEKYI
    EVFNSQRNFDEGPSKPSPYYSE
    IGQIAKMIGNCTFESSEKRTAK
    NTWSGERFVFLQKLNNFRIVGL
    SGKRPLTEEERDIVEKEVYLKK
    EVRYEKLRKILYLKEEERFGDL
    NYSKDEKQDKKTEKTKFISLIG
    NYTIKKLNLSEKLKSEIEEDKS
    KLDKIIEILTFNKSDKTIESNL
    KKLELSREDIEILLSEEFSGTL
    NLSLKAIKKILPYLEKGLSYNE
    ACEKADYDYKNNGIKFKRGELL
    PVVDKDLIANPVVLRAISQTRK
    VVNAIIRKYGTPHTIHVEVARD
    LAKSYDDRQTIIKENKKRELEN
    EKTKKFISEEFGIKNVKGKLLL
    KYRLYQEQEGRCAYSRKELSLS
    EVILDESMTDIDHIIPYSRSMD
    DSYSNKVLVLSGENRKKSNLLP
    KEYFDRQGRDWDTFVLNVKAMK
    IHPRKKSNLLKEKFTREDNKDW
    KSRALNDTRYISRFVANYLENA
    LEYRDDSPKKRVFMIPGQLTAQ
    LRARWRLNKVRENGDLHHALDA
    AVVAVTDQKAINNISNISRYKE
    LKNCKDVIPSIEYHADEETGEV
    YFEEVKDTRFPMPWSGFDLELQ
    KRLESENPREEFYNLLSDKRYL
    GWFNYEEGFIEKLRPVFVSRMP
    NRGVKGQAHQETIRSSKKISNQ
    IAVSKKPLNSIKLKDLEKMQGR
    DTDRKLYEALKNRLEEYDDKPE
    KAFAEPFYKPTNSGKRGPLVRG
    IKVEEKQNVGVYVNGGQASNGS
    MVRIDVFRKNGKFYTVPIYVHQ
    TLLKELPNRAINGKPYKDWDLI
    DGSFEFLYSFYPNDLIEIEFGK
    SKSIKNDNKLTKTEIPEVNLSE
    VLGYYRGMDTSTGAATIDTQDG
    KIQMRIGIKTVKNIKKYQVDVL
    GNVYKVKREKRQTF (SEQ ID
    NO: 71)
    Sphaerochaeta MSKKVSRRYEEQAQEICQRLGS 163 202 40 335 389 45 335 389 45
    globus RPYSIGLDLGVGSIGVAVAAYD
    str. Buddy PIKKQPSDLVFVSSRIFIPSTG
    gi|325972003| AAERRQKRGQRNSLRHRANRLK
    ref|YP_004248194.1 FLWKLLAERNLMLSYSEQDVPD
    PARLRFEDAVVRANPYELRLKG
    LNEQLTLSELGYALYHIANHRG
    SSSVRTFLDEEKSSDDKKLEEQ
    QAMTEQLAKEKGISTFIEVLTA
    FNTNGLIGYRNSESVKSKGVPV
    PTRDIISNEIDVLLQTQKQFYQ
    EILSDEYCDRIVSAILFENEKI
    VPEAGCCPYFPDEKKLPRCHFL
    NEERRLWEAINNARIKMPMQEG
    AAKRYQSASFSDEQRHILFHIA
    RSGTDITPKLVQKEFPALKTSI
    IVLQGKEKAIQKIAGFRFRRLE
    EKSFWKRLSEEQKDDFFSAWTN
    TPDDKRLSKYLMKHLLLTENEV
    VDALKTVSLIGDYGPIGKTATQ
    LLMKHLEDGLTYTEALERGMET
    GEFQELSVWEQQSLLPYYGQIL
    TGSTQALMGKYWHSAFKEKRDS
    EGFFKPNTNSDEEKYGRIANPV
    VHQTLNELRKLMNELITILGAK
    PQEITVELARELKVGAEKREDI
    IKQQTKQEKEAVLAYSKYCEPN
    NLDKRYIERFRLLEDQAFVCPY
    CLEHISVADIAAGRADVDHIFP
    RDDTADNSYGNKVVAHRQCNDI
    KGKRTPYAAFSNTSAWGPIMHY
    LDETPGMWRKRRKFETNEEEYA
    KYLQSKGFVSRFESDNSYIAKA
    AKEYLRCLFNPNNVTAVGSLKG
    METSILRKAWNLQGIDDLLGSR
    HWSKDADTSPTMRKNRDDNRHH
    GLDAIVALYCSRSLVQMINTMS
    EQGKRAVEIEAMIPIPGYASEP
    NLSFEAQRELFRKKILEFMDLH
    AFVSMKTDNDANGALLKDTVYS
    ILGADTQGEDLVFVVKKKIKDI
    GVKIGDYEEVASAIRGRITDKQ
    PKWYPMEMKDKIEQLQSKNEAA
    LQKYKESLVQAAAVLEESNRKL
    IESGKKPIQLSEKTISKKALEL
    VGGYYYLISNNKRTKTFVVKEP
    SNEVKGFAFDTGSNLCLDFYHD
    AQGKLCGEIIRKIQAMNPSYKP
    AYMKQGYSLYVRLYQGDVCELR
    ASDLTEAESNLAKTTHVRLPNA
    KPGRTFVIIITFTEMGSGYQIY
    FSNLAKSKKGQDTSFTLTTIKN
    YDVRKVQLSSAGLVRYVSPLLV
    DKIEKDEVALCGE (SEQ ID
    NO: 72)
    Staphylococcus MNQKFILGLDIGITSVGYGLID 128 167 40 337 391 57 337 391 57
    lugdunensis YETKNIIDAGVRLFPEANVENN
    M23590 EGRRSKRGSRRLKRRRIHRLER
    gi|315659848| VKKLLEDYNLLDQSQIPQSTNP
    ref|ZP_07912707.1 YAIRVKGLSEALSKDELVIALL
    HIAKRRGIHKIDVIDSNDDVGN
    ELSTKEQLNKNSKLLKDKFVCQ
    IQLERMNEGQVRGEKNRFKTAD
    IIKEIIQLLNVQKNFHQLDENF
    INKYIELVEMRREYFEGPGKGS
    PYGWEGDPKAWYETLMGHCTYF
    PDELRSVKYAYSADLFNALNDL
    NNLVIQRDGLSKLEYHEKYHII
    ENVFKQKKKPTLKQIANEINVN
    PEDIKGYRITKSGKPQFTEFKL
    YHDLKSVLFDQSILENEDVLDQ
    IAEILTIYQDKDSIKSKLTELD
    ILLNEEDKENIAQLTGYTGTHR
    LSLKCIRLVLEEQWYSSRNQME
    IFTHLNIKPKKINLTAANKIPK
    AMIDEFILSPVVKRTFGQAINL
    INKIIEKYGVPEDIIIELAREN
    NSKDKQKFINEMQKKNENTRKR
    INEIIGKYGNQNAKRLVEKIRL
    HDEQEGKCLYSLESIPLEDLLN
    NPNHYEVDHIIPRSVSFDNSYH
    NKVLVKQSENSKKSNLTPYQYF
    NSGKSKLSYNQFKQHILNLSKS
    QDRISKKKKEYLLEERDINKFE
    VQKEFINRNLVDTRYATRELTN
    YLKAYFSANNMNVKVKTINGSF
    TDYLRKVWKFKKERNHGYKHHA
    EDALIIANADFLFKENKKLKAV
    NSVLEKPEIESKQLDIQVDSED
    NYSEMFIIPKQVQDIKDFRNFK
    YSHRVDKKPNRQLINDTLYSTR
    KKDNSTYIVQTIKDIYAKDNTT
    LKKQFDKSPEKFLMYQHDPRTF
    EKLEVIMKQYANEKNPLAKYHE
    ETGEYLTKYSKKNNGPIVKSLK
    YIGNKLGSHLDVTHQFKSSTKK
    LVKLSIKPYRFDVYLTDKGYKF
    ITISYLDVLKKDNYYYIPEQKY
    DKLKLGKAIDKNAKFIASFYKN
    DLIKLDGEIYKIIGVNSDTRNM
    IELDLPDIRYKEYCELNNIKGE
    PRIKKTIGKKVNSIEKLTTDVL
    GNVFTNTQYTKPQLLFKRGN
    (SEQ ID NO: 73)
    Treponema MIMKLEKWRLGLDLGTNSIGWS 144 183 40 328 382 63 328 382 63
    sp. JC4 VFSLDKDNSVQDLIDMGVRIFS
    gi|384109266| DGRDPKTKEPLAVARRTARSQR
    ref|ZP_10010146.1 KLIYRRKLRRKQVFKFLQEQGL
    FPKTKEECMTLKSLNPYELRIK
    ALDEKLEPYELGRALFNLAVRR
    GFKSNRKDGSREEVSEKKSPDE
    IKTQADMQTHLEKAIKENGCRT
    ITEFLYKNQGENGGIRFAPGRM
    TYYPTRKMYEEEFNLIRSKQEK
    YYPQVDWDDIYKAIFYQRPLKP
    QQRGYCIYENDKERTFKAMPCS
    QKLRILQDIGNLAYYEGGSKKR
    VELNDNQDKVLYELLNSKDKVT
    FDQMRKALCLADSNSFNLEENR
    DFLIGNPTAVKMRSKNRFGKLW
    DEIPLEEQDLIIETIITADEDD
    AVYEVIKKYDLTQEQRDFIVKN
    TILQSGTSMLCKEVSEKLVKRL
    EEIADLKYHEAVESLGYKFADQ
    TVEKYDLLPYYGKVLPGSTMEI
    DLSAPETNPEKHYGKISNPTVH
    VALNQTRVVVNALIKEYGKPSQ
    IAIELSRDLKNNVEKKAEIARK
    QNQRAKENIAINDTISALYHTA
    FPGKSFYPNRNDRMKYRLWSEL
    GLGNKCIYCGKGISGAELFTKE
    IEIEHILPFSRTLLDAESNLTV
    AHSSCNAFKAERSPFEAFGTNP
    SGYSWQEIIQRANQLKNTSKKN
    KFSPNAMDSFEKDSSFIARQLS
    DNQYIAKAALRYLKCLVENPSD
    VWTTNGSMTKLLRDKWEMDSIL
    CRKFTEKEVALLGLKPEQIGNY
    KKNRFDHRHHAIDAVVIGLTDR
    SMVQKLATKNSHKGNRIEIPEF
    PILRSDLIEKVKNIVVSFKPDH
    GAEGKLSKETLLGKIKLHGKET
    FVCRENIVSLSEKNLDDIVDEI
    KSKVKDYVAKHKGQKIEAVLSD
    FSKENGIKKVRCVNRVQTPIEI
    TSGKISRYLSPEDYFAAVIWEI
    PGEKKTFKAQYIRRNEVEKNSK
    GLNVVKPAVLENGKPHPAAKQV
    CLLHKDDYLEFSDKGKMYFCRI
    AGYAATNNKLDIRPVYAVSYCA
    DWINSTNETMLTGYWKPTPTQN
    WVSVNVLFDKQKARLVTVSPIG
    RVFRK (SEQ ID NO: 74)
    uncultured MSSKAIDSLEQLDLFKPQEYTL 154 193 40 313 365 55 313 365 55
    delta GLDLGIKSIGWAILSGERIANA
    proteobacterium GVYLFETAEELNSTGNKLISKA
    HF007007E19 AERGRKRRIRRMLDRKARRGRH
    gi|297182908| IRYLLEREGLPTDELEEVVVHQ
    gb|ADI19058.1| SNRTLWDVRAEAVERKLTKQEL
    AAVLFHLVRHRGYFPNTKKLPP
    DDESDSADEEQGKINRATSRLR
    EELKASDCKTIGQFLAQNRDRQ
    RNREGDYSNLMARKLVFEEALQ
    ILAFQRKQGHELSKDFEKTYLD
    VLMGQRSGRSPKLGNCSLIPSE
    LRAPSSAPSTEWFKFLQNLGNL
    QISNAYREEWSIDAPRRAQIID
    ACSQRSTSSYWQIRRDFQIPDE
    YRFNLVNYERRDPDVDLQEYLQ
    QQERKTLANFRNWKQLEKIIGT
    GHPIQTLDEAARLITLIKDDEK
    LSDQLADLLPEASDKAITQLCE
    LDFTTAAKISLEAMYRILPHMN
    QGMGFFDACQQESLPEIGVPPA
    GDRVPPFDEMYNPVVNRVLSQS
    RKLINAVIDEYGMPAKIRVELA
    RDLGKGRELRERIKLDQLDKSK
    QNDQRAEDFRAEFQQAPRGDQS
    LRYRLWKEQNCTCPYSGRMIPV
    NSVLSEDTQIDHILPISQSFDN
    SLSNKVLCFTEENAQKSNRTPF
    EYLDAADFQRLEAISGNWPEAK
    RNKLLHKSFGKVAEEWKSRALN
    DTRYLTSALADHLRHHLPDSKI
    QTVNGRITGYLRKQWGLEKDRD
    KHTHHAVDAIVVACTTPAIVQQ
    VTLYHQDIRRYKKLGEKRPTPW
    PETFRQDVLDVEEEIFITRQPK
    KVSGGIQTKDTLRKHRSKPDRQ
    RVALTKVKLADLERLVEKDASN
    RNLYEHLKQCLEESGDQPTKAF
    KAPFYMPSGPEAKQRPILSKVT
    LLREKPEPPKQLTELSGGRRYD
    SMAQGRLDIYRYKPGGKRKDEY
    RVVLQRMIDLMRGEENVHVFQK
    GVPYDQGPEIEQNYTFLFSLYF
    DDLVEFQRSADSEVIRGYYRTF
    NIANGQLKISTYLEGRQDFDFF
    GANRLAHFAKVQVNLLGKVIK
    (SEQ ID NO: 75)
    Alicycliphilus MRSLRYRLALDLGSTSLGWALF 140 178 39 317 366 48 317 366 48
    denitrificans RLDACNRPTAVIKAGVRIFSDG
    K601 RNPKDGSSLAVTRRAARAMRRR
    gi|330822845| RDRLLKRKTRMQAKLVEHGFFP
    ref|YP_004386148.1 ADAGKRKALEQLNPYALRAKGL
    QEALLPGEFARALFHINQRRGF
    KSNRKTDKKDNDSGVLKKAIGQ
    LRQQMAEQGSRTVGEYLWTRLQ
    QGQGVRARYREKPYTTEEGKKR
    IDKSYDLYIDRAMIEQEFDALW
    AAQAAFNPTLFHEAARADLKDT
    LLHQRPLRPVKPGRCTLLPEEE
    RAPLALPSTQRFRIHQEVNHLR
    LLDENLREVALTLAQRDAVVTA
    LETKAKLSFEQIRKLLKLSGSV
    QFNLEDAKRTELKGNATSAALA
    RKELFGAAWSGFDEALQDEIVW
    QLVTEEGEGALIAWLQTHTGVD
    EARAQAIVDVSLPEGYGNLSRK
    ALARIVPALRAAVITYDKAVQA
    AGFDHHSQLGFEYDASEVEDLV
    HPETGEIRSVFKQLPYYGKALQ
    RHVAFGSGKPEDPDEKRYGKIA
    NPTVHIGLNQVRMVVNALIRRY
    GRPTEVVIELARDLKQSREQKV
    EAQRRQADNQRRNARIRRSIAE
    VLGIGEERVRGSDIQKWICWEE
    LSFDAADRRCPYSGVQISAAML
    LSDEVEVEHILPFSKTLDDSLN
    NRTVAMRQANRIKRNRTPWDAR
    AEFEAQGWSYEDILQRAERMPL
    RKRYRFAPDGYERWLGDDKDFL
    ARALNDTRYLSRVAAEYLRLVC
    PGTRVIPGQLTALLRGKFGLND
    VLGLDGEKNRNDHRHHAVDACV
    IGVTDQGLMQRFATASAQARGD
    GLTRLVDGMPMPWPTYRDHVER
    AVRHIWVSHRPDHGFEGAMMEE
    TSYGIRKDGSIKQRRKADGSAG
    REISNLIRIHEATQPLRHGVSA
    DGQPLAYKGYVGGSNYCIEITV
    NDKGKWEGEVISTFRAYGVVRA
    GGMGRLRNPHEGQNGRKLIMRL
    VIGDSVRLEVDGAERTMRIVKI
    SGSNGQIFMAPIHEANVDARNT
    DKQDAFTYTSKYAGSLQKAKTR
    RVTISPIGEVRDPGFKG (SEQ
    ID NO: 76)
    Azospirillum MARPAFRAPRREHVNGWTPDPH 205 243 39 342 389 46 342 389 46
    sp. B510 RISKPFFILVSWHLLSRVVIDS
    gi|288957741| SSGCFPGTSRDHTDKFAEWECA
    ref|YP_003448082.1 VQPYRLSFDLGTNSIGWGLLNL
    DRQGKPREIRALGSRIFSDGRD
    PQDKASLAVARRLARQMRRRRD
    RYLTRRTRLMGALVRFGLMPAD
    PAARKRLEVAVDPYLARERATR
    ERLEPFEIGRALFHLNQRRGYK
    PVRTATKPDEEAGKVKEAVERL
    EAAIAAAGAPTLGAWFAWRKTR
    GETLRARLAGKGKEAAYPFYPA
    RRMLEAEFDTLWAEQARHHPDL
    LTAEAREILRHRIFHQRPLKPP
    PVGRCTLYPDDGRAPRALPSAQ
    RLRLFQELASLRVIHLDLSERP
    LTPAERDRIVAFVQGRPPKAGR
    KPGKVQKSVPFEKLRGLLELPP
    GTGFSLESDKRPELLGDETGAR
    IAPAFGPGWTALPLEEQDALVE
    LLLTEAEPERAIAALTARWALD
    EATAAKLAGATLPDFHGRYGRR
    AVAELLPVLERETRGDPDGRVR
    PIRLDEAVKLLRGGKDHSDFSR
    EGALLDALPYYGAVLERHVAFG
    TGNPADPEEKRVGRVANPTVHI
    ALNQLRHLVNAILARHGRPEEI
    VIELARDLKRSAEDRRREDKRQ
    ADNQKRNEERKRLILSLGERPT
    PRNLLKLRLWEEQGPVENRRCP
    YSGETISMRMLLSEQVDIDHIL
    PFSVSLDDSAANKVVCLREANR
    IKRNRSPWEAFGHDSERWAGIL
    ARAEALPKNKRWRFAPDALEKL
    EGEGGLRARHLNDTRHLSRLAV
    EYLRCVCPKVRVSPGRLTALLR
    RRWGIDAILAEADGPPPEVPAE
    TLDPSPAEKNRADHRHHALDAV
    VIGCIDRSMVQRVQLAAASAER
    EAAAREDNIRRVLEGFKEEPWD
    GFRAELERRARTIVVSHRPEHG
    IGGALHKETAYGPVDPPEEGFN
    LVVRKPIDGLSKDEINSVRDPR
    LRRALIDRLAIRRRDANDPATA
    LAKAAEDLAAQPASRGIRRVRV
    LKKESNPIRVEHGGNPSGPRSG
    GPFHKLLLAGEVHHVDVALRAD
    GRRWVGHWVTLFEAHGGRGADG
    AAAPPRLGDGERFLMRLHKGDC
    LKLEHKGRVRVMQVVKLEPSSN
    SVVVVEPHQVKTDRSKHVKISC
    DQLRARGARRVTVDPLGRVRVH
    APGARVGIGGDAGRTAMEPAED
    IS (SEQ ID NO: 77)
    Bradyrhizobium MKRTSLRAYRLGVDLGANSLGW 143 181 39 323 370 48 323 370 48
    sp. FVVWLDDHGQPEGLGPGGVRIF
    BTAi1 PDGRNPQSKQSNAAGRRLARSA
    gi|148255343| RRRRDRYLQRRGKLMGLLVKHG
    ref|YP_001239928.1 LMPADEPARKRLECLDPYGLRA
    KALDEVLPLHHVGRALFHLNQR
    RGLFANRAIEQGDKDASAIKAA
    AGRLQTSMQACGARTLGEFLNR
    RHQLRATVRARSPVGGDVQARY
    EFYPTRAMVDAEFEAIWAAQAP
    HHPTMTAEAHDTIREAIFSQRA
    MKRPSIGKCSLDPATSQDDVDG
    FRCAWSHPLAQRFRIWQDVRNL
    AVVETGPTSSRLGKEDQDKVAR
    ALLQTDQLSFDEIRGLLGLPSD
    ARFNLESDRRDHLKGDATGAIL
    SARRHFGPAWHDRSLDRQIDIV
    ALLESALDEAAIIASLGTTHSL
    DEAAAQRALSALLPDGYCRLGL
    RAIKRVLPLMEAGRTYAEAASA
    AGYDHALLPGGKLSPTGYLPYY
    GQWLQNDVVGSDDERDTNERRW
    GRLPNPTVHIGIGQLRRVVNEL
    IRWHGPPAEITVELTRDLKLSP
    RRLAELEREQAENQRKNDKRTS
    LLRKLGLPASTHNLLKLRLWDE
    QGDVASECPYTGEAIGLERLVS
    DDVDIDHLIPFSISWDDSAANK
    VVCMRYANREKGNRTPFEAFGH
    RQGRPYDWADIAERAARLPRGK
    RWRFGPGARAQFEELGDFQARL
    LNETSWLARVAKQYLAAVTHPH
    RIHVLPGRLTALLRATWELNDL
    LPGSDDRAAKSRKDHRHHAIDA
    LVAALTDQALLRRMANAHDDTR
    RKIEVLLPWPTFRIDLETRLKA
    MLVSHKPDHGLQARLHEDTAYG
    TVEHPETEDGANLVYRKTFVDI
    SEKEIDRIRDRRLRDLVRAHVA
    GERQQGKTLKAAVLSFAQRRDI
    AGHPNGIRHVRLTKSIKPDYLV
    PIRDKAGRIYKSYNAGENAFVD
    ILQAESGRWIARATTVFQANQA
    NESHDAPAAQPIMRVFKGDMLR
    IDHAGAEKFVKIVRLSPSNNLL
    YLVEHHQAGVFQTRHDDPEDSF
    RWLFASFDKLREWNAELVRIDT
    LGQPWRRKRGLETGSEDATRIG
    WTRPKKWP (SEQ ID NO:
    78)
    Parvibaculum MERIFGFDIGTTSIGFSVIDYS 138 176 39 327 374 58 327 374 58
    lavamentivorans STQSAGNIQRLGVRIFPEARDP
    DS-1 DGTPLNQQRRQKRMMRRQLRRR
    gi|154250555| RIRRKALNETLHEAGFLPAYGS
    ref|YP_001411379.1 ADWPVVMADEPYELRRRGLEEG
    LSAYEFGRAIYHLAQHRHFKGR
    ELEESDTPDPDVDDEKEAANER
    AATLKALKNEQTTLGAWLARRP
    PSDRKRGIHAHRNVVAEEFERL
    WEVQSKFHPALKSEEMRARISD
    TIFAQRPVFWRKNTLGECRFMP
    GEPLCPKGSWLSQQRRMLEKLN
    NLAIAGGNARPLDAEERDAILS
    KLQQQASMSWPGVRSALKALYK
    QRGEPGAEKSLKFNLELGGESK
    LLGNALEAKLADMFGPDWPAHP
    RKQEIRHAVHERLWAADYGETP
    DKKRVIILSEKDRKAHREAAAN
    SFVADFGITGEQAAQLQALKLP
    TGWEPYSIPALNLFLAELEKGE
    RFGALVNGPDWEGWRRTNFPHR
    NQPTGEILDKLPSPASKEERER
    ISQLRNPTVVRTQNELRKVVNN
    LIGLYGKPDRIRIEVGRDVGKS
    KREREEIQSGIRRNEKQRKKAT
    EDLIKNGIANPSRDDVEKWILW
    KEGQERCPYTGDQIGFNALFRE
    GRYEVEHIWPRSRSFDNSPRNK
    TLCRKDVNIEKGNRMPFEAFGH
    DEDRWSAIQIRLQGMVSAKGGT
    GMSPGKVKRFLAKTMPEDFAAR
    QLNDTRYAAKQILAQLKRLWPD
    MGPEAPVKVEAVTGQVTAQLRK
    LWTLNNILADDGEKTRADHRHH
    AIDALTVACTHPGMTNKLSRYW
    QLRDDPRAEKPALTPPWDTIRA
    DAEKAVSEIVVSHRVRKKVSGP
    LHKETTYGDTGTDIKTKSGTYR
    QFVTRKKIESLSKGELDEIRDP
    RIKEIVAAHVAGRGGDPKKAFP
    PYPCVSPGGPEIRKVRLTSKQQ
    LNLMAQTGNGYADLGSNHHIAI
    YRLPDGKADFEIVSLFDASRRL
    AQRNPIVQRTRADGASFVMSLA
    AGEAIMIPEGSKKGIWIVQGVW
    ASGQVVLERDTDADHSTTTRPM
    PNPILKDDAKKVSIDPIGRVRP
    SND (SEQ ID NO: 79)
    Prevotella MNKRILGLDTGTNSLGWAVVDW 170 208 39 328 375 61 328 375 61
    timonensis DEHAQSYELIKYGDVIFQEGVK
    CRIS 5C-B1 IEKGIESSKAAERSGYKAIRKQ
    gi|282880052| YFRRRLRKIQVLKVLVKYHLCP
    ref|ZP_06288774.1 YLSDDDLRQWHLQKQYPKSDEL
    MLWQRTSDEEGKNPYYDRHRCL
    HEKLDLTVEADRYTLGRALYHL
    TQRRGFLSNRLDTSADNKEDGV
    VKSGISQLSTEMEEAGCEYLGD
    YFYKLYDAQGNKVRIRQRYTDR
    NKHYQHEFDAICEKQELSSELI
    EDLQRAIFFQLPLKSQRHGVGR
    CTFERGKPRCADSHPDYEEFRM
    LCFVNNIQVKGPHDLELRPLTY
    EEREKIEPLFFRKSKPNFDFED
    IAKALAGKKNYAWIHDKEERAY
    KFNYRMTQGVPGCPTIAQLKSI
    FGDDWKTGIAETYTLIQKKNGS
    KSLQEMVDDVWNVLYSFSSVEK
    LKEFAHHKLQLDEESAEKFAKI
    KLSHSFAALSLKAIRKFLPFLR
    KGMYYTHASFFANIPTIVGKEI
    WNKEQNRKYIMENVGELVFNYQ
    PKHREVQGTIEMLIKDFLANNF
    ELPAGATDKLYHPSMIETYPNA
    QRNEFGILQLGSPRTNAIRNPM
    AMRSLHILRRVVNQLLKESIID
    ENTEVHVEYARELNDANKRRAI
    ADRQKEQDKQHKKYGDEIRKLY
    KEETGKDIEPTQTDVLKFQLWE
    EQNHHCLYTGEQIGITDFIGSN
    PKFDIEHTIPQSVGGDSTQMNL
    TLCDNRFNREVKKAKLPTELAN
    HEEILTRIEPWKNKYEQLVKER
    DKQRTFAGMDKAVKDIRIQKRH
    KLQMEIDYWRGKYERFTMTEVP
    EGFSRRQGTGIGLISRYAGLYL
    KSLFHQADSRNKSNVYVVKGVA
    TAEFRKMWGLQSEYEKKCRDNH
    SHHCMDAITIACIGKREYDLMA
    EYYRMEETFKQGRGSKPKFSKP
    WATFTEDVLNIYKNLLVVHDTP
    NNMPKHTKKYVQTSIGKVLAQG
    DTARGSLHLDTYYGAIERDGEI
    RYVVRRPLSSFTKPEELENIVD
    ETVKRTIKEAIADKNFKQAIAE
    PIYMNEEKGILIKKVRCFAKSV
    KQPINIRQHRDLSKKEYKQQYH
    VMNENNYLLAIYEGLVKNKVVR
    EFEIVSYIEAAKYYKRSQDRNI
    FSSIVPTHSTKYGLPLKTKLLM
    GQLVLMFEENPDEIQVDNTKDL
    VKRLYKVVGIEKDGRIKFKYHQ
    EARKEGLPIFSTPYKNNDDYAP
    IFRQSINNINILVDGIDFTIDI
    LGKVTLKE (SEQ ID NO:
    80)
    Bacillus MNYKMGLDIGIASVGWAVINLD 134 171 38 401 448 63 401 448 63
    smithii 7 3 LKRIEDLGVRIFDKAEHPQNGE
    47FAA SLALPRRIARSARRRLRRRKHR
    gi|365156657| LERIRRLLVSENVLTKEEMNLL
    ref|ZP_09352959.1 FKQKKQIDVWQLRVDALERKLN
    NDELARVLLHLAKRRGFKSNRK
    SERNSKESSEFLKNIEENQSIL
    AQYRSVGEMIVKDSKFAYHKRN
    KLDSYSNMIARDDLEREIKLIF
    EKQREFNNPVCTERLEEKYLNI
    WSSQRPFASKEDIEKKVGFCTF
    EPKEKRAPKATYTFQSFIVWEH
    INKLRLVSPDETRALTEIERNL
    LYKQAFSKNKMTYYDIRKLLNL
    SDDIHFKGLLYDPKSSLKQIEN
    IRFLELDSYHKIRKCIENVYGK
    DGIRMFNETDIDTFGYALTIFK
    DDEDIVAYLQNEYITKNGKRVS
    NLANKVYDKSLIDELLNLSFSK
    FAHLSMKAIRNILPYMEQGEIY
    SKACELAGYNFTGPKKKEKALL
    LPVIPNIANPVVMRALTQSRKV
    VNAIIKKYGSPVSIHIELARDL
    SHSFDERKKIQKDQTENRKKNE
    TAIKQLIEYELTKNPTGLDIVK
    FKLWSEQQGRCMYSLKPIELER
    LLEPGYVEVDHILPYSRSLDDS
    YANKVLVLTKENREKGNHTPVE
    YLGLGSERWKKFEKFVLANKQF
    SKKKKQNLLRLRYEETEEKEFK
    ERNLNDTRYISKFFANFIKEHL
    KFADGDGGQKVYTINGKITAHL
    RSRWDFNKNREESDLHHAVDAV
    IVACATQGMIKKITEFYKAREQ
    NKESAKKKEPIFPQPWPHFADE
    LKARLSKFPQESIEAFALGNYD
    RKKLESLRPVFVSRMPKRSVTG
    AAHQETLRRCVGIDEQSGKIQT
    AVKTKLSDIKLDKDGHFPMYQK
    ESDPRTYEAIRQRLLEHNNDPK
    KAFQEPLYKPKKNGEPGPVIRT
    VKIIDTKNKVVHLDGSKTVAYN
    SNIVRTDVFEKDGKYYCVPVYT
    MDIMKGTLPNKAIEANKPYSEW
    KEMTEEYTFQFSLFPNDLVRIV
    LPREKTIKTSTNEEIIIKDIFA
    YYKTIDSATGGLELISHDRNFS
    LRGVGSKTLKRFEKYQVDVLGN
    IHKVKGEKRVGLAAPTNQKKGK
    TVDSLQSVSD (SEQ ID NO:
    81)
    Cand. MRRLGLDLGTNSIGWCLLDLGD 135 172 38 344 391 53 344 391 53
    Puniceispirillum DGEPVSIFRTGARIFSDGRDPK
    marinum SLGSLKATRREARLTRRRRDRF
    IMCC1322 IQRQKNLINALVKYGLMPADEI
    gi|294086111| QRQALAYKDPYPIRKKALDEAI
    ref|YP_003552871.1 DPYEMGRAIFHINQRRGFKSNR
    KSADNEAGVVKQSIADLEMKLG
    EAGARTIGEFLADRQATNDTVR
    ARRLSGTNALYEFYPDRYMLEQ
    EFDTLWAKQAAFNPSLYIEAAR
    ERLKEIVFFQRKLKPQEVGRCI
    FLSDEDRISKALPSFQRFRIYQ
    ELSNLAWIDHDGVAHRITASLA
    LRDHLFDELEHKKKLTFKAMRA
    ILRKQGVVDYPVGFNLESDNRD
    HLIGNLTSCIMRDAKKMIGSAW
    DRLDEEEQDSFILMLQDDQKGD
    DEVRSILTQQYGLSDDVAEDCL
    DVRLPDGHGSLSKKAIDRILPV
    LRDQGLIYYDAVKEAGLGEANL
    YDPYAALSDKLDYYGKALAGHV
    MGASGKFEDSDEKRYGTISNPT
    VHIALNQVRAVVNELIRLHGKP
    DEVVIEIGRDLPMGADGKRELE
    RFQKEGRAKNERARDELKKLGH
    IDSRESRQKFQLWEQLAKEPVD
    RCCPFTGKMMSISDLFSDKVEI
    EHLLPFSLTLDDSMANKTVCFR
    QANRDKGNRAPFDAFGNSPAGY
    DWQEILGRSQNLPYAKRWRFLP
    DAMKRFEADGGFLERQLNDTRY
    ISRYTTEYISTIIPKNKIWVVT
    GRLTSLLRGFWGLNSILRGHNT
    DDGTPAKKSRDDHRHHAIDAIV
    VGMTSRGLLQKVSKAARRSEDL
    DLTRLFEGRIDPWDGFRDEVKK
    HIDAIIVSHRPRKKSQGALHND
    TAYGIVEHAENGASTVVHRVPI
    TSLGKQSDIEKVRDPLIKSALL
    NETAGLSGKSFENAVQKWCADN
    SIKSLRIVETVSIIPITDKEGV
    AYKGYKGDGNAYMDIYQDPTSS
    KWKGEIVSRFDANQKGFIPSWQ
    SQFPTARLIMRLRINDLLKLQD
    GEIEEIYRVQRLSGSKILMAPH
    TEANVDARDRDKNDTFKLTSKS
    PGKLQSASARKVHISPTGLIRE
    G (SEQ ID NO: 82)
    Barnesiella MKNILGLDLGLSSIGWSVIREN 140 176 37 371 417 60 371 417 60
    intestinihominis SEEQELVAMGSRVVSLTAAELS
    YIT SFTQGNGVSINSQRTQKRTQRK
    11860 GYDRYQLRRTLLRNKLDTLGML
    gi|404487228| PDDSLSYLPKLQLWGLRAKAVT
    ref|ZP_11022414.1 QRIELNELGRVLLHLNQKRGYK
    SIKSDFSGDKKITDYVKTVKTR
    YDELKEMRLTIGELFFRRLTEN
    AFFRCKEQVYPRQAYVEEFDCI
    MNCQRKFYPDILTDETIRCIRD
    EIIYYQRPLKSCKYLVSRCEFE
    KRFYLNAAGKKTEAGPKVSPRT
    SPLFQVCRLWESINNIVVKDRR
    NEIVFISAEQRAALFDFLNTHE
    KLKGSDLLKLLGLSKTYGYRLG
    EQFKTGIQGNKTRVEIERALGN
    YPDKKRLLQFNLQEESSSMVNT
    ETGEIIPMISLSFEQEPLYRLW
    HVLYSIDDREQLQSVLRQKFGI
    DDDEVLERLSAIDLVKAGFGNK
    SSKAIRRILPFLQLGMNYAEAC
    EAAGYNHSNNYTKAENEARALL
    DRLPAIKKNELRQPVVEKILNQ
    MVNVVNALMEKYGRFDEIRVEL
    ARELKQSKEERSNTYKSINKNQ
    RENEQIAKRIVEYGVPTRSRIQ
    KYKMWEESKHCCIYCGQPVDVG
    DFLRGFDVEVEHIIPKSLYFDD
    SFANKVCSCRSCNKEKNNRTAY
    DYMKSKGEKALSDYVERVNTMY
    TNNQISKTKWQNLLTPVDKISI
    DFIDRQLRESQYIARKAKEILT
    SICYNVTATSGSVTSFLRHVWG
    WDTVLHDLNFDRYKKVGLTEVI
    EVNHRGSVIRREQIKDWSKRFD
    HRHHAIDALTIACTKQAYIQRL
    NNLRAEEGPDFNKMSLERYIQS
    QPHFSVAQVREAVDRILVSFRA
    GKRAVTPGKRYIRKNRKRISVQ
    SVLIPRGALSEESVYGVIHVWE
    KDEQGHVIQKQRAVMKYPITSI
    NREMLDKEKVVDKRIHRILSGR
    LAQYNDNPKEAFAKPVYIDKEC
    RIPIRTVRCFAKPAINTLVPLK
    KDDKGNPVAWVNPGNNHHVAIY
    RDEDGKYKERTVTFWEAVDRCR
    VGIPAIVTQPDTIWDNILQRND
    ISENVLESLPDVKWQFVLSLQQ
    NEMFILGMNEEDYRYAMDQQDY
    ALLNKYLYRVQKLSKSDYSFRY
    HTETSVEDKYDGKPNLKLSMQM
    GKLKRVSIKSLLGLNPHKVHIS
    VLGEIKEIS (SEQ ID NO:
    83)
    Ralstonia MAEKQHRWGLDIGTNSIGWAVI 140 176 37 395 440 50 395 440 50
    syzygii R24 ALIEGRPAGLVATGSRIFSDGR
    gi|344171927| NPKDGSSLAVERRGPRQMRRRR
    emb|CCA84553.1| DRYLRRRDRFMQALINVGLMPG
    DAAARKALVTENPYVLRQRGLD
    QALTLPEFGRALFHLNQRRGFQ
    SNRKTDRATAKESGKVKNAIAA
    FRAGMGNARTVGEALARRLEDG
    RPVRARMVGQGKDEHYELYIAR
    EWIAQEFDALWASQQRFHAEVL
    ADAARDRLRAILLFQRKLLPVP
    VGKCFLEPNQPRVAAALPSAQR
    FRLMQELNHLRVMTLADKRERP
    LSFQERNDLLAQLVARPKCGFD
    MLRKIVFGANKEAYRFTIESER
    RKELKGCDTAAKLAKVNALGTR
    WQALSLDEQDRLVCLLLDGEND
    AVLADALREHYGLTDAQIDTLL
    GLSFEDGHMRLGRSALLRVLDA
    LESGRDEQGLPLSYDKAVVAAG
    YPAHTADLENGERDALPYYGEL
    LWRYTQDAPTAKNDAERKFGKI
    ANPTVHIGLNQLRKLVNALIQR
    YGKPAQIVVELARNLKAGLEEK
    ERIKKQQTANLERNERIRQKLQ
    DAGVPDNRENRLRMRLFEELGQ
    GNGLGTPCIYSGRQISLQRLFS
    NDVQVDHILPFSKTLDDSFANK
    VLAQHDANRYKGNRGPFEAFGA
    NRDGYAWDDIRARAAVLPRNKR
    NRFAETAMQDWLHNETDFLARQ
    LTDTAYLSRVARQYLTAICSKD
    DVYVSPGRLTAMLRAKWGLNRV
    LDGVMEEQGRPAVKNRDDHRHH
    AIDAVVIGATDRAMLQQVATLA
    ARAREQDAERLIGDMPTPWPNF
    LEDVRAAVARCVVSHKPDHGPE
    GGLHNDTAYGIVAGPFEDGRYR
    VRHRVSLFDLKPGDLSNVRCDA
    PLQAELEPIFEQDDARAREVAL
    TALAERYRQRKVWLEELMSVLP
    IRPRGEDGKTLPDSAPYKAYKG
    DSNYCYELFINERGRWDGELIS
    TFRANQAAYRRFRNDPARFRRY
    TAGGRPLLMRLCINDYIAVGTA
    AERTIFRVVKMSENKITLAEHF
    EGGTLKQRDADKDDPFKYLTKS
    PGALRDLGARRIFVDLIGRVLD
    PGIKGD (SEQ ID NO: 84)
    Wolinella MIERILGVDLGISSLGWAIVEY 145 180 36 348 392 60 348 392 60
    succinogenes DKDDEAANRIIDCGVRLFTAAE
    DSM 1740 TPKKKESPNKARREARGIRRVL
    gi|34557790| NRRRVRMNMIKKLFLRAGLIQD
    ref|NP_907605.1| VDLDGEGGMFYSKANRADVWEL
    RHDGLYRLLKGDELARVLIHIA
    KHRGYKFIGDDEADEESGKVKK
    AGVVLRQNFEAAGCRTVGEWLW
    RERGANGKKRNKHGDYEISIHR
    DLLVEEVEAIFVAQQEMRSTIA
    TDALKAAYREIAFFVRPMQRIE
    KMVGHCTYFPEERRAPKSAPTA
    EKFIAISKFFSTVIIDNEGWEQ
    KIIERKTLEELLDFAVSREKVE
    FRHLRKFLDLSDNEIFKGLHYK
    GKPKTAKKREATLFDPNEPTEL
    EFDKVEAEKKAWISLRGAAKLR
    EALGNEFYGRFVALGKHADEAT
    KILTYYKDEGQKRRELTKLPLE
    AEMVERLVKIGFSDFLKLSLKA
    IRDILPAMESGARYDEAVLMLG
    VPHKEKSAILPPLNKTDIDILN
    PTVIRAFAQFRKVANALVRKYG
    AFDRVHFELAREINTKGEIEDI
    KESQRKNEKERKEAADWIAETS
    FQVPLTRKNILKKRLYIQQDGR
    CAYTGDVIELERLFDEGYCEID
    HILPRSRSADDSFANKVLCLAR
    ANQQKTDRTPYEWFGHDAARWN
    AFETRTSAPSNRVRTGKGKIDR
    LLKKNFDENSEMAFKDRNLNDT
    RYMARAIKTYCEQYWVFKNSHT
    KAPVQVRSGKLTSVLRYQWGLE
    SKDRESHTHHAVDAIIIAFSTQ
    GMVQKLSEYYRFKETHREKERP
    KLAVPLANFRDAVEEATRIENT
    ETVKEGVEVKRLLISRPPRARV
    TGQAHEQTAKPYPRIKQVKNKK
    KWRLAPIDEEKFESFKADRVAS
    ANQKNFYETSTIPRVDVYHKKG
    KFHLVPIYLHEMVLNELPNLSL
    GTNPEAMDENFFKFSIFKDDLI
    SIQTQGTPKKPAKIIMGYFKNM
    HGANMVLSSINNSPCEGFTCTP
    VSMDKKHKDKCKLCPEENRIAG
    RCLQGFLDYWSQEGLRPPRKEF
    ECDQGVKFALDVKKYQIDPLGY
    YYEVKQEKRLGTIPQMRSAKKL
    VKK (SEQ ID NO: 86)
    Mycoplasma MNNSIKSKPEVTIGLDLGVGSV 144 177 34 373 416 71 373 416 71
    gallisepticum GWAIVDNETNIIHHLGSRLFSQ
    str. F AKTAEDRRSFRGVRRLIRRRKY
    gi|284931710| KLKRFVNLIWKYNSYFGFKNKE
    gb|ADC31648.1| DILNNYQEQQKLHNTVLNLKSE
    ALNAKIDPKALSWILHDYLKNR
    GHFYEDNRDFNVYPTKELAKYF
    DKYGYYKGIIDSKEDNDNKLEE
    ELTKYKFSNKHWLEEVKKVLSN
    QTGLPEKFKEEYESLFSYVRNY
    SEGPGSINSVSPYGIYHLDEKE
    GKVVQKYNNIWDKTIGKCNIFP
    DEYRAPKNSPIAMIFNEINELS
    TIRSYSIYLTGWFINQEFKKAY
    LNKLLDLLIKTNGEKPIDARQF
    KKLREETIAESIGKETLKDVEN
    EEKLEKEDHKWKLKGLKLNTNG
    KIQYNDLSSLAKFVHKLKQHLK
    LDFLLEDQYATLDKINFLQSLF
    VYLGKHLRYSNRVDSANLKEFS
    DSNKLFERILQKQKDGLFKLFE
    QTDKDDEKILAQTHSLSTKAML
    LAITRMTNLDNDEDNQKNNDKG
    WNFEAIKNFDQKFIDITKKNNN
    LSLKQNKRYLDDRFINDAILSP
    GVKRILREATKVFNAILKQFSE
    EYDVTKVVIELARELSEEKELE
    NTKNYKKLIKKNGDKISEGLKA
    LGISEDEIKDILKSPTKSYKFL
    LWLQQDHIDPYSLKEIAFDDIF
    TKTEKFEIDHIIPYSISFDDSS
    SNKLLVLAESNQAKSNQTPYEF
    ISSGNAGIKWEDYEAYCRKFKD
    GDSSLLDSTQRSKKFAKMMKTD
    TSSKYDIGFLARNLNDTRYATI
    VFRDALEDYANNHLVEDKPMFK
    VVCINGSVTSFLRKNFDDSSYA
    KKDRDKNIHHAVDASIISIFSN
    ETKTLFNQLTQFADYKLFKNTD
    GSWKKIDPKTGVVTEVTDENWK
    QIRVRNQVSEIAKVIEKYIQDS
    NIERKARYSRKIENKTNISLFN
    DTVYSAKKVGYEDQIKRKNLKT
    LDIHESAKENKNSKVKRQFVYR
    KLVNVSLLNNDKLADLFAEKED
    ILMYRANPWVINLAEQIFNEYT
    ENKKIKSQNVFEKYMLDLTKEF
    PEKFSEFLVKSMLRNKTAIIYD
    DKKNIVHRIKRLKMLSSELKEN
    KLSNVIIRSKNQSGTKLSYQDT
    INSLALMIMRSIDPTAKKQYIR
    VPLNTLNLHLGDHDFDLHNMDA
    YLKKPKFVKYLKANEIGDEYKP
    WRVLTSGTLLIHKKDKKLMYIS
    SFQNLNDVIEIKNLIETEYKEN
    DDSDSKKKKKANRFLMTLSTIL
    NDYILLDAKDNFDILGLSKNRI
    DEILNSKLGLDKIVK (SEQ
    ID NO: 87)
    Acidothermus MGGSEVGTVPVTWRLGVDVGER 150 182 33 341 380 58 341 380 58
    cellulolyticus SIGLAAVSYEEDKPKEILAAVS
    11B WIHDGGVGDERSGASRLALRGM
    gi|117929158| ARRARRLRRFRRARLRDLDMLL
    ref|YP_873709.1| SELGWTPLPDKNVSPVDAWLAR
    KRLAEEYVVDETERRRLLGYAV
    SHMARHRGWRNPWTTIKDLKNL
    PQPSDSWERTRESLEARYSVSL
    EPGTVGQWAGYLLQRAPGIRLN
    PTQQSAGRRAELSNATAFETRL
    RQEDVLWELRCIADVQGLPEDV
    VSNVIDAVFCQKRPSVPAERIG
    RDPLDPSQLRASRACLEFQEYR
    IVAAVANLRIRDGSGSRPLSLE
    ERNAVIEALLAQTERSLTWSDI
    ALEILKLPNESDLTSVPEEDGP
    SSLAYSQFAPFDETSARIAEFI
    AKNRRKIPTFAQWWQEQDRTSR
    SDLVAALADNSIAGEEEQELLV
    HLPDAELEALEGLALPSGRVAY
    SRLTLSGLTRVMRDDGVDVHNA
    RKTCFGVDDNWRPPLPALHEAT
    GHPVVDRNLAILRKFLSSATMR
    WGPPQSIVVELARGASESRERQ
    AEEEAARRAHRKANDRIRAELR
    ASGLSDPSPADLVRARLLELYD
    CHCMYCGAPISWENSELDHIVP
    RTDGGSNRHENLAITCGACNKE
    KGRRPFASWAETSNRVQLRDVI
    DRVQKLKYSGNMYWTRDEFSRY
    KKSVVARLKRRTSDPEVIQSIE
    STGYAAVALRDRLLSYGEKNGV
    AQVAVFRGGVTAEARRWLDISI
    ERLFSRVAIFAQSTSTKRLDRR
    HHAVDAVVLTTLTPGVAKTLAD
    ARSRRVSAEFWRRPSDVNRHST
    EEPQSPAYRQWKESCSGLGDLL
    ISTAARDSIAVAAPLRLRPTGA
    LHEETLRAFSEHTVGAAWKGAE
    LRRIVEPEVYAAFLALTDPGGR
    FLKVSPSEDVLPADENRHIVLS
    DRVLGPRDRVKLFPDDRGSIRV
    RGGAAYIASFHHARVFRWGSSH
    SPSFALLRVSLADLAVAGLLRD
    GVDVFTAELPPWTPAWRYASIA
    LVKAVESGDAKQVGWLVPGDEL
    DFGPEGVTTAAGDLSMFLKYFP
    ERHWVVTGFEDDKRINLKPAFL
    SAEQAEVLRTERSDRPDTLTEA
    GEILAQFFPRCWRATVAKVLCH
    PGLTVIRRTALGQPRWRRGHLP
    YSWRPWSADPWSGGTP (SEQ
    ID NO: 88)
    Mycoplasma MHNKKNITIGFDLGIASIGWAI 156 184 29 381 420 62 381 420 62
    ovipneumoniae IDSTTSKILDWGTRTFEERKTA
    SC01 NERRAFRSTRRNIRRKAYRNQR
    gi|363542550| FINLILKYKDLFELKNISDIQR
    ref|ZP_09312133.1 ANKKDTENYEKIISFFTEIYKK
    CAAKHSNILEVKVKALDSKIEK
    LDLIWILHDYLENRGFFYDLEE
    ENVADKYEGIEHPSILLYDFFK
    KNGFFKSNSSIPKDLGGYSFSN
    LQWVNEIKKLFEVQEINPEFSE
    KFLNLFTSVRDYAKGPGSEHSA
    SEYGIFQKDEKGKVFKKYDNIW
    DKTIGKCSFFVEENRSPVNYPS
    YEIFNLLNQLINLSTDLKTTNK
    KIWQLSSNDRNELLDELLKVKE
    KAKIISISLKKNEIKKIILKDF
    GFEKSDIDDQDTIEGRKIIKEE
    PTTKLEVTKHLLATIYSHSSDS
    NWININNILEFLPYLDAICIIL
    DREKSRGQDEVLKKLTEKNIFE
    VLKIDREKQLDFVKSIFSNTKF
    NFKKIGNFSLKAIREFLPKMFE
    QNKNSEYLKWKDEEIRRKWEEQ
    KSKLGKTDKKTKYLNPRIFQDE
    IISPGTKNTFEQAVLVLNQIIK
    KYSKENIIDAIIIESPREKNDK
    KTIEEIKKRNKKGKGKTLEKLF
    QILNLENKGYKLSDLETKPAKL
    LDRLRFYHQQDGIDLYTLDKIN
    IDQLINGSQKYEIEHIIPYSMS
    YDNSQANKILTEKAENLKKGKL
    IASEYIKRNGDEFYNKYYEKAK
    ELFINKYKKNKKLDSYVDLDED
    SAKNRFRFLTLQDYDEFQVEFL
    ARNLNDTRYSTKLFYHALVEHF
    ENNEFFTYIDENSSKHKVKIST
    IKGHVTKYFRAKPVQKNNGPNE
    NLNNNKPEKIEKNRENNEHHAV
    DAAIVAIIGNKNPQIANLLTLA
    DNKTDKKFLLHDENYKENIETG
    ELVKIPKFEVDKLAKVEDLKKI
    IQEKYEEAKKHTAIKFSRKTRT
    ILNGGLSDETLYGFKYDEKEDK
    YFKIIKKKLVTSKNEELKKYFE
    NPFGKKADGKSEYTVLMAQSHL
    SEFNKLKEIFEKYNGFSNKTGN
    AFVEYMNDLALKEPTLKAEIES
    AKSVEKLLYYNFKPSDQFTYHD
    NINNKSFKRFYKNIRIIEYKSI
    PIKFKILSKHDGGKSFKDTLFS
    LYSLVYKVYENGKESYKSIPVT
    SQMRNFGIDEFDFLDENLYNKE
    KLDIYKSDFAKPIPVNCKPVFV
    LKKGSILKKKSLDIDDFKETKE
    TEEGNYYFISTISKRFNRDTAY
    GLKPLKLSVVKPVAEPSTNPIF
    KEYIPIHLDELGNEYPVKIKEH
    TDDEKLMCTIK (SEQ ID
    NO: 89)
  • If any of the above Cas9 sequences are fused with a peptide or polypeptide at the C-terminus, it is understood that the stop codon will be removed.
  • Exemplary PAM sequences and their corresponding RKR motifs are provided in Table 250.
  • TABLE 250
    Identified PAM sequences and corresponding
    RKR motifs.
    RKR
    PAM sequence motif
    Strain Name (NA) (AA)
    Streptococcus pyogenes NGG RKR
    Streptococcus mutans NGG RKR
    Streptococcus thermophilus A NGGNG RYR
    Treponema denticola NAAAAN VAK
    Streptococcus thermophilus B NNAAAAW IYK
    Campylobacter jejuni NNNNACA NLK
    Pasteurella multocida GNNNCNNA KDG
    Neisseria meningitidis NNNNGATT or IGK
    NNGRRT
    (R = A or G)
    Staphylococcus aureus NNGRR NDK
    (R = A or G)
  • Exemplary Cas9 core domains are provided in Table 200.
  • TABLE 200
    Amino Acid Sequence of Cas9 Core Domains
    Cas9 Start Cas9 Stop
    (AA pos) (AA pos)
    Start and Stop numbers refer to
    Strain Name the sequence in Table 100
    Staphylococcus aureus 1 772
    Streptococcus pyogenes 1 1099
    Campulobacter jejuni 1 741
  • Exemplary PI domains, e.g., altered PI domains, are provided in Tables 400 and 500.
  • TABLE 400
    Altered PI Domains
    Length RKR
    PI Start PI Stop of PI motif
    Strain Name (AA pos) (AA pos) (AA) (AA)
    Alicycliphilus 837 1029 193 --Y
    denitrificans K601
    Start and Stop
    numbers refer to
    the sequences in
    Table 100
    Campylobacter jejuni 741  984 244 -NG
    NCTC 11168
    Helicobacter mustelae 12198 771 1024 254 -NQ
  • TABLE 500
    Other Altered PI Domains
    PI Start PI Stop
    (AA pos) (AA pos)
    Start and Stop
    numbers refer to Length RKR
    the sequences in of PI motif
    Strain Name Table 100 (AA) (AA)
    Akkermansia muciniphila ATCC BAA-835 871 1101 231 ALK
    Ralstonia syzygii R24 821 1062 242 APY
    Cand. Puniceispirillum marinum IMCC1322 815 1035 221 AYK
    Fructobacillus fructosus KCTC 3544 1074 1323 250 DGN
    Eubacterium yurii ATCC 43715 1107 1391 285 DGY
    Eubacterium dolichum DSM 3991 779 1096 318 DKK
    Dinoroseobacter shibae DFL 12 851 1079 229 DPI
    Clostridium cellulolyticum H10 767 1021 255 EGK
    Pasteurella multocida str. Pm70 815 1056 242 ENN
    Mycoplasma canis PG 14 907 1233 327 EPK
    Porphyromonas sp. oral taxon 279 str. F0450 935 1197 263 EPT
    Filifactor alocis ATCC 35896 1094 1365 272 EVD
    Aminomonas paucivorans DSM 12260 801 1052 252 EVY
    Wolinella succinogenes DSM 1740 1034 1409 376 EYK
    Oenococcus kitaharae DSM 17330 1119 1389 271 GAL
    CoriobacteriumglomeransPW2 1126 1384 259 GDR
    Peptoniphilus duerdenii ATCC BAA-1640 1091 1364 274 GDS
    Bifidobacterium bifidum S17 1138 1420 283 GGL
    Alicyclobacillus hesperidum URH17-3-68 876 1146 271 GGR
    Roseburia inulinivorans DSM 16841 895 1152 258 GGT
    Actinomyces coleocanis DSM 15436 843 1105 263 GKK
    Odoribacter laneus YIT 12061 1103 1498 396 GKV
    Coprococcus catus GD-7 1063 1338 276 GNQ
    Enterococcus faecalis TX0012 829 1150 322 GRK
    Bacillus smithii 7 3 47FAA 809 1088 280 GSK
    Legionella pneumophila str. Paris 1021 1372 352 GTM
    Bacteroides fragilis NCTC 9343 1140 1436 297 IPV
    Mycoplasma ovipneumoniae SC01 923 1265 343 IRI
    Actinomyces sp. oral taxon 180 str. F0310 895 1181 287 KEK
    Treponema sp. JC4 832 1062 231 KIS
    Fusobacteriumnucleatum ATCC49256 1073 1374 302 KKV
    Lactobacillus farciminis KCTC 3681 1101 1356 256 KKV
    Nitratifractor salsuginis DSM 16511 840 1132 293 KMR
    Lactobacillus coryniformis KCTC 3535 850 1119 270 KNK
    Mycoplasma mobile 163K 916 1236 321 KNY
    Flavobacterium branchiophilum FL-15 1182 1473 292 KQK
    Prevotella timonensis CRIS 5C-B1 957 1218 262 KQQ
    Methylosinus trichosporium OB3b 830 1082 253 KRP
    Prevotella sp. C561 1099 1424 326 KRY
    Mycoplasma gallisepticum str. F 911 1269 359 KTA
    Lactobacillus rhamnosus GG 1077 1363 287 KYG
    Wolinella succinogenes DSM 1740 811 1059 249 LPN
    Streptococcus thermophilus LMD-9 1099 1388 290 MLA
    Treponema denticola ATCC 35405 1092 1395 304 NDS
    Bergeyella zoohelcum ATCC 43767 1098 1415 318 NEK
    Veillonella atypica ACS-134-V-Col7a 1107 1398 292 NGF
    Neisseria meningitidis Z2491 835 1082 248 NHN
    Ignavibacterium album JCM 16511 1296 1688 393 NKK
    Ruminococcus albus 8 853 1156 304 NNF
    Streptococcus thermophilus LMD-9 811 1121 311 NNK
    Barnesiella intestinihominis YIT 11860 871 1153 283 NPV
    Azospirillum sp. B510 911 1168 258 PFH
    Rhodospirillum rubrum ATCC 11170 863 1173 311 PRG
    Planococcus antarcticus DSM 14505 1087 1333 247 PYY
    Staphylococcus pseudintermedius ED99 1073 1334 262 QIV
    Alcanivorax sp. W11-5 843 1113 271 RIE
    Bradyrhizobium sp. BTAi1 811 1064 254 RIY
    Streptococcus pyogenes M1 GAS 1099 1368 270 RKR
    Streptococcus mutans UA159 1078 1345 268 RKR
    Streptococcus pyogenes 1099 1368 270 RKR
    Bacteroides sp. 20 3 1147 1517 371 RNI
    S. aureus 772 1053 282 RNK
    Solobacterium moorei F0204 1062 1327 266 RSG
    Finegoldia magna ATCC 29328 1081 1348 268 RTE
    uncultured delta proteobacterium HF0070 07E19 770 1011 242 SGG
    Acidaminococcus sp. D21 1064 1358 295 SIG
    Eubacterium rectale ATCC 33656 824 1114 291 SKK
    Caenispirillum salinarum AK4 1048 1442 395 SLV
    Acidothermus cellulolyticus 11B 830 1138 309 SPS
    Catenibacterium mitsuokai DSM 15897 1068 1329 262 SPT
    Parvibaculum lavamentivorans DS-1 827 1037 211 TGN
    Staphylococcus lugdunensis M23590 772 1054 283 TKK
    Streptococcus sanguinis SK49 1123 1421 299 TRM
    Elusimicrobium minutum Pei191 910 1195 286 TTG
    Nitrobacter hamburgensis X14 914 1166 253 VAY
    Mycoplasma synoviae 53 991 1314 324 VGF
    Sphaerochaeta globus str. Buddy 877 1179 303 VKG
    Ilyobacter polytropus DSM 2926 837 1092 256 VNG
    Rhodovulum sp. PH10 821 1059 239 VPY
    Bifidobacterium longum DJO10A 904 1187 284 VRK
  • Nucleic Acids Encoding Cas9 Fusion Molecules
  • Nucleic acids encoding the Cas9 fusion molecules, the Cas9 molecules or Cas9 polypeptides, e.g., an eaCas9 molecule or eaCas9 polypeptides, are provided herein.
  • In an embodiment, a nucleic acid encoding a Cas9 fusion molecule, a Cas9 molecule, or Cas9 polypeptide, can be a synthetic nucleic acid sequence. For example, the synthetic nucleic acid molecule can be chemically modified, e.g., as described in Section XI. In an embodiment, the mRNA, e.g., coding for a Cas9 fusion molecule, Cas9 molecule, or Cas9 polypeptide, disclosed herein, has one or more, e.g., all, of the following properties: it is capped, polyadenylated, substituted with 5-methylcytidine and/or pseudouridine.
  • In addition, or alternatively, the synthetic nucleic acid sequence can be codon optimized, e.g., at least one non-common codon or less-common codon has been replaced by a codon that is common in the host cell. For example, the synthetic nucleic acid can direct the synthesis of an optimized messenger mRNA, e.g., optimized for expression in a mammalian expression system, e.g., described herein.
  • In addition, or alternatively, a nucleic acid encoding a Cas9 fusion molecule, a Cas9 molecule, or a Cas9 polypeptide, may comprise a nuclear localization sequence (NLS). Nuclear localization sequences are known in the art.
  • Provided below is an exemplary codon optimized nucleic acid sequence encoding a Cas9 molecule of S. pyogenes.
  • (SEQ ID NO: DD1)
    ATGGATAAAA AGTACAGCAT CGGGCTGGAC ATCGGTACAA
    ACTCAGTGGG GTGGGCCGTG ATTACGGACG AGTACAAGGT
    ACCCTCCAAA AAATTTAAAG TGCTGGGTAA CACGGACAGA
    CACTCTATAA AGAAAAATCT TATTGGAGCC TTGCTGTTCG
    ACTCAGGCGA GACAGCCGAA GCCACAAGGT TGAAGCGGAC
    CGCCAGGAGG CGGTATACCA GGAGAAAGAA CCGCATATGC
    TACCTGCAAG AAATCTTCAG TAACGAGATG GCAAAGGTTG
    ACGATAGCTT TTTCCATCGC CTGGAAGAAT CCTTTCTTGT
    TGAGGAAGAC AAGAAGCACG AACGGCACCC CATCTTTGGC
    AATATTGTCG ACGAAGTGGC ATATCACGAA AAGTACCCGA
    CTATCTACCA CCTCAGGAAG AAGCTGGTGG ACTCTACCGA
    TAAGGCGGAC CTCAGACTTA TTTATTTGGC ACTCGCCCAC
    ATGATTAAAT TTAGAGGACA TTTCTTGATC GAGGGCGACC
    TGAACCCGGA CAACAGTGAC GTCGATAAGC TGTTCATCCA
    ACTTGTGCAG ACCTACAATC AACTGTTCGA AGAAAACCCT
    ATAAATGCTT CAGGAGTCGA CGCTAAAGCA ATCCTGTCCG
    CGCGCCTCTC AAAATCTAGA AGACTTGAGA ATCTGATTGC
    TCAGTTGCCC GGGGAAAAGA AAAATGGATT GTTTGGCAAC
    CTGATCGCCC TCAGTCTCGG ACTGACCCCA AATTTCAAAA
    GTAACTTCGA CCTGGCCGAA GACGCTAAGC TCCAGCTGTC
    CAAGGACACA TACGATGACG ACCTCGACAA TCTGCTGGCC
    CAGATTGGGG ATCAGTACGC CGATCTCTTT TTGGCAGCAA
    AGAACCTGTC CGACGCCATC CTGTTGAGCG ATATCTTGAG
    AGTGAACACC GAAATTACTA AAGCACCCCT TAGCGCATCT
    ATGATCAAGC GGTACGACGA GCATCATCAG GATCTGACCC
    TGCTGAAGGC TCTTGTGAGG CAACAGCTCC CCGAAAAATA
    CAAGGAAATC TTCTTTGACC AGAGCAAAAA CGGCTACGCT
    GGCTATATAG ATGGTGGGGC CAGTCAGGAG GAATTCTATA
    AATTCATCAA GCCCATTCTC GAGAAAATGG ACGGCACAGA
    GGAGTTGCTG GTCAAACTTA ACAGGGAGGA CCTGCTGCGG
    AAGCAGCGGA CCTTTGACAA CGGGTCTATC CCCCACCAGA
    TTCATCTGGG CGAACTGCAC GCAATCCTGA GGAGGCAGGA
    GGATTTTTAT CCTTTTCTTA AAGATAACCG CGAGAAAATA
    GAAAAGATTC TTACATTCAG GATCCCGTAC TACGTGGGAC
    CTCTCGCCCG GGGCAATTCA CGGTTTGCCT GGATGACAAG
    GAAGTCAGAG GAGACTATTA CACCTTGGAA CTTCGAAGAA
    GTGGTGGACA AGGGTGCATC TGCCCAGTCT TTCATCGAGC
    GGATGACAAA TTTTGACAAG AACCTCCCTA ATGAGAAGGT
    GCTGCCCAAA CATTCTCTGC TCTACGAGTA CTTTACCGTC
    TACAATGAAC TGACTAAAGT CAAGTACGTC ACCGAGGGAA
    TGAGGAAGCC GGCATTCCTT AGTGGAGAAC AGAAGAAGGC
    GATTGTAGAC CTGTTGTTCA AGACCAACAG GAAGGTGACT
    GTGAAGCAAC TTAAAGAAGA CTACTTTAAG AAGATCGAAT
    GTTTTGACAG TGTGGAAATT TCAGGGGTTG AAGACCGCTT
    CAATGCGTCA TTGGGGACTT ACCATGATCT TCTCAAGATC
    ATAAAGGACA AAGACTTCCT GGACAACGAA GAAAATGAGG
    ATATTCTCGA AGACATCGTC CTCACCCTGA CCCTGTTCGA
    AGACAGGGAA ATGATAGAAG AGCGCTTGAA AACCTATGCC
    CACCTCTTCG ACGATAAAGT TATGAAGCAG CTGAAGCGCA
    GGAGATACAC AGGATGGGGA AGATTGTCAA GGAAGCTGAT
    CAATGGAATT AGGGATAAAC AGAGTGGCAA GACCATACTG
    GATTTCCTCA AATCTGATGG CTTCGCCAAT AGGAACTTCA
    TGCAACTGAT TCACGATGAC TCTCTTACCT TCAAGGAGGA
    CATTCAAAAG GCTCAGGTGA GCGGGCAGGG AGACTCCCTT
    CATGAACACA TCGCGAATTT GGCAGGTTCC CCCGCTATTA
    AAAAGGGCAT CCTTCAAACT GTCAAGGTGG TGGATGAATT
    GGTCAAGGTA ATGGGCAGAC ATAAGCCAGA AAATATTGTG
    ATCGAGATGG CCCGCGAAAA CCAGACCACA CAGAAGGGCC
    AGAAAAATAG TAGAGAGCGG ATGAAGAGGA TCGAGGAGGG
    CATCAAAGAG CTGGGATCTC AGATTCTCAA AGAACACCCC
    GTAGAAAACA CACAGCTGCA GAACGAAAAA TTGTACTTGT
    ACTATCTGCA GAACGGCAGA GACATGTACG TCGACCAAGA
    ACTTGATATT AATAGACTGT CCGACTATGA CGTAGACCAT
    ATCGTGCCCC AGTCCTTCCT GAAGGACGAC TCCATTGATA
    ACAAAGTCTT GACAAGAAGC GACAAGAACA GGGGTAAAAG
    TGATAATGTG CCTAGCGAGG AGGTGGTGAA AAAAATGAAG
    AACTACTGGC GACAGCTGCT TAATGCAAAG CTCATTACAC
    AACGGAAGTT CGATAATCTG ACGAAAGCAG AGAGAGGTGG
    CTTGTCTGAG TTGGACAAGG CAGGGTTTAT TAAGCGGCAG
    CTGGTGGAAA CTAGGCAGAT CACAAAGCAC GTGGCGCAGA
    TTTTGGACAG CCGGATGAAC ACAAAATACG ACGAAAATGA
    TAAACTGATA CGAGAGGTCA AAGTTATCAC GCTGAAAAGC
    AAGCTGGTGT CCGATTTTCG GAAAGACTTC CAGTTCTACA
    AAGTTCGCGA GATTAATAAC TACCATCATG CTCACGATGC
    GTACCTGAAC GCTGTTGTCG GGACCGCCTT GATAAAGAAG
    TACCCAAAGC TGGAATCCGA GTTCGTATAC GGGGATTACA
    AAGTGTACGA TGTGAGGAAA ATGATAGCCA AGTCCGAGCA
    GGAGATTGGA AAGGCCACAG CTAAGTACTT CTTTTATTCT
    AACATCATGA ATTTTTTTAA GACGGAAATT ACCCTGGCCA
    ACGGAGAGAT CAGAAAGCGG CCCCTTATAG AGACAAATGG
    TGAAACAGGT GAAATCGTCT GGGATAAGGG CAGGGATTTC
    GCTACTGTGA GGAAGGTGCT GAGTATGCCA CAGGTAAATA
    TCGTGAAAAA AACCGAAGTA CAGACCGGAG GATTTTCCAA
    GGAAAGCATT TTGCCTAAAA GAAACTCAGA CAAGCTCATC
    GCCCGCAAGA AAGATTGGGA CCCTAAGAAA TACGGGGGAT
    TTGACTCACC CACCGTAGCC TATTCTGTGC TGGTGGTAGC
    TAAGGTGGAA AAAGGAAAGT CTAAGAAGCT GAAGTCCGTG
    AAGGAACTCT TGGGAATCAC TATCATGGAA AGATCATCCT
    TTGAAAAGAA CCCTATCGAT TTCCTGGAGG CTAAGGGTTA
    CAAGGAGGTC AAGAAAGACC TCATCATTAA ACTGCCAAAA
    TACTCTCTCT TCGAGCTGGA AAATGGCAGG AAGAGAATGT
    TGGCCAGCGC CGGAGAGCTG CAAAAGGGAA ACGAGCTTGC
    TCTGCCCTCC AAATATGTTA ATTTTCTCTA TCTCGCTTCC
    CACTATGAAA AGCTGAAAGG GTCTCCCGAA GATAACGAGC
    AGAAGCAGCT GTTCGTCGAA CAGCACAAGC ACTATCTGGA
    TGAAATAATC GAACAAATAA GCGAGTTCAG CAAAAGGGTT
    ATCCTGGCGG ATGCTAATTT GGACAAAGTA CTGTCTGCTT
    ATAACAAGCA CCGGGATAAG CCTATTAGGG AACAAGCCGA
    GAATATAATT CACCTCTTTA CACTCACGAA TCTCGGAGCC
    CCCGCCGCCT TCAAATACTT TGATACGACT ATCGACCGGA
    AACGGTATAC CAGTACCAAA GAGGTCCTCG ATGCCACCCT
    CATCCACCAG TCAATTACTG GCCTGTACGA AACACGGATC
    GACCTCTCTC AACTGGGCGG CGACTAG
  • Provided below is the corresponding amino acid sequence of a S. pyogenes Cas9 molecule.
  • (SEQ ID NO: DD2)
    MDKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIKKNLIGA
    LLFDSGETAEATRLKRTARRRYTRRKNRICYLQEIFSNEMAKVDDSFFHR
    LEESFLVEEDKKHERHPIFGNIVDEVAYHEKYPTIYHLRKKLVDSTDKAD
    LRLIYLALAHMIKFRGHFLIEGDLNPDNSDVDKLFIQLVQTYNQLFEENP
    INASGVDAKAILSARLSKSRRLENLIAQLPGEKKNGLFGNLIALSLGLTP
    NFKSNFDLAEDAKLQLSKDTYDDDLDNLLAQIGDQYADLFLAAKNLSDAI
    LLSDILRVNTEITKAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEI
    FFDQSKNGYAGYIDGGASQEEFYKFIKPILEKMDGTEELLVKLNREDLLR
    KQRTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKILTFRIPY
    YVGPLARGNSRFAWMTRKSEETITPWNFEEVVDKGASAQSFIERMTNFDK
    NLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRKPAFLSGEQKKAIVD
    LLFKTNRKVTVKQLKEDYFKKIECFDSVEISGVEDRFNASLGTYHDLLKI
    IKDKDFLDNEENEDILEDIVLTLTLFEDREMIEERLKTYAHLFDDKVMKQ
    LKRRRYTGWGRLSRKLINGIRDKQSGKTILDFLKSDGFANRNFMQLIHDD
    SLTFKEDIQKAQVSGQGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKV
    MGRHKPENIVIEMARENQTTQKGQKNSRERMKRIEEGIKELGSQILKEHP
    VENTQLQNEKLYLYYLQNGRDMYVDQELDINRLSDYDVDHIVPQSFLKDD
    SIDNKVLTRSDKNRGKSDNVPSEEVVKKMKNYWRQLLNAKLITQRKFDNL
    TKAERGGLSELDKAGFIKRQLVETRQITKHVAQILDSRMNTKYDENDKLI
    REVKVITLKSKLVSDFRKDFQFYKVREINNYHHAHDAYLNAVVGTALIKK
    YPKLESEFVYGDYKVYDVRKMIAKSEQEIGKATAKYFFYSNIMNFFKTEI
    TLANGEIRKRPLIETNGETGEIVWDKGRDFATVRKVLSMPQVNIVKKTEV
    QTGGFSKESILPKRNSDKLIARKKDWDPKKYGGFDSPTVAYSVLVVAKVE
    KGKSKKLKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPK
    YSLFELENGRKRMLASAGELQKGNELALPSKYVNFLYLASHYEKLKGSPE
    DNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANLDKVLSAYNKHRDK
    PIREQAENIIHLFTLTNLGAPAAFKYFDTTIDRKRYTSTKEVLDATLIHQ
    SITGLYETRIDLSQLGGD*
  • Provided below is an exemplary codon optimized nucleic acid sequence encoding a Cas9 molecule of N. meningitidis.
  • (SEQ ID NO: DD3)
    ATGGCCGCCTTCAAGCCCAACCCCATCAACTACATCCTGGGCCTGGACAT
    CGGCATCGCCAGCGTGGGCTGGGCCATGGTGGAGATCGACGAGGACGAGA
    ACCCCATCTGCCTGATCGACCTGGGTGTGCGCGTGTTCGAGCGCGCTGAG
    GTGCCCAAGACTGGTGACAGTCTGGCTATGGCTCGCCGGCTTGCTCGCTC
    TGTTCGGCGCCTTACTCGCCGGCGCGCTCACCGCCTTCTGCGCGCTCGCC
    GCCTGCTGAAGCGCGAGGGTGTGCTGCAGGCTGCCGACTTCGACGAGAAC
    GGCCTGATCAAGAGCCTGCCCAACACTCCTTGGCAGCTGCGCGCTGCCGC
    TCTGGACCGCAAGCTGACTCCTCTGGAGTGGAGCGCCGTGCTGCTGCACC
    TGATCAAGCACCGCGGCTACCTGAGCCAGCGCAAGAACGAGGGCGAGACC
    GCCGACAAGGAGCTGGGTGCTCTGCTGAAGGGCGTGGCCGACAACGCCCA
    CGCCCTGCAGACTGGTGACTTCCGCACTCCTGCTGAGCTGGCCCTGAACA
    AGTTCGAGAAGGAGAGCGGCCACATCCGCAACCAGCGCGGCGACTACAGC
    CACACCTTCAGCCGCAAGGACCTGCAGGCCGAGCTGATCCTGCTGTTCGA
    GAAGCAGAAGGAGTTCGGCAACCCCCACGTGAGCGGCGGCCTGAAGGAGG
    GCATCGAGACCCTGCTGATGACCCAGCGCCCCGCCCTGAGCGGCGACGCC
    GTGCAGAAGATGCTGGGCCACTGCACCTTCGAGCCAGCCGAGCCCAAGGC
    CGCCAAGAACACCTACACCGCCGAGCGCTTCATCTGGCTGACCAAGCTGA
    ACAACCTGCGCATCCTGGAGCAGGGCAGCGAGCGCCCCCTGACCGACACC
    GAGCGCGCCACCCTGATGGACGAGCCCTACCGCAAGAGCAAGCTGACCTA
    CGCCCAGGCCCGCAAGCTGCTGGGTCTGGAGGACACCGCCTTCTTCAAGG
    GCCTGCGCTACGGCAAGGACAACGCCGAGGCCAGCACCCTGATGGAGATG
    AAGGCCTACCACGCCATCAGCCGCGCCCTGGAGAAGGAGGGCCTGAAGGA
    CAAGAAGAGTCCTCTGAACCTGAGCCCCGAGCTGCAGGACGAGATCGGCA
    CCGCCTTCAGCCTGTTCAAGACCGACGAGGACATCACCGGCCGCCTGAAG
    GACCGCATCCAGCCCGAGATCCTGGAGGCCCTGCTGAAGCACATCAGCTT
    CGACAAGTTCGTGCAGATCAGCCTGAAGGCCCTGCGCCGCATCGTGCCCC
    TGATGGAGCAGGGCAAGCGCTACGACGAGGCCTGCGCCGAGATCTACGGC
    GACCACTACGGCAAGAAGAACACCGAGGAGAAGATCTACCTGCCTCCTAT
    CCCCGCCGACGAGATCCGCAACCCCGTGGTGCTGCGCGCCCTGAGCCAGG
    CCCGCAAGGTGATCAACGGCGTGGTGCGCCGCTACGGCAGCCCCGCCCGC
    ATCCACATCGAGACCGCCCGCGAGGTGGGCAAGAGCTTCAAGGACCGCAA
    GGAGATCGAGAAGCGCCAGGAGGAGAACCGCAAGGACCGCGAGAAGGCCG
    CCGCCAAGTTCCGCGAGTACTTCCCCAACTTCGTGGGCGAGCCCAAGAGC
    AAGGACATCCTGAAGCTGCGCCTGTACGAGCAGCAGCACGGCAAGTGCCT
    GTACAGCGGCAAGGAGATCAACCTGGGCCGCCTGAACGAGAAGGGCTACG
    TGGAGATCGACCACGCCCTGCCCTTCAGCCGCACCTGGGACGACAGCTTC
    AACAACAAGGTGCTGGTGCTGGGCAGCGAGAACCAGAACAAGGGCAACCA
    GACCCCCTACGAGTACTTCAACGGCAAGGACAACAGCCGCGAGTGGCAGG
    AGTTCAAGGCCCGCGTGGAGACCAGCCGCTTCCCCCGCAGCAAGAAGCAG
    CGCATCCTGCTGCAGAAGTTCGACGAGGACGGCTTCAAGGAGCGCAACCT
    GAACGACACCCGCTACGTGAACCGCTTCCTGTGCCAGTTCGTGGCCGACC
    GCATGCGCCTGACCGGCAAGGGCAAGAAGCGCGTGTTCGCCAGCAACGGC
    CAGATCACCAACCTGCTGCGCGGCTTCTGGGGCCTGCGCAAGGTGCGCGC
    CGAGAACGACCGCCACCACGCCCTGGACGCCGTGGTGGTGGCCTGCAGCA
    CCGTGGCCATGCAGCAGAAGATCACCCGCTTCGTGCGCTACAAGGAGATG
    AACGCCTTCGACGGTAAAACCATCGACAAGGAGACCGGCGAGGTGCTGCA
    CCAGAAGACCCACTTCCCCCAGCCCTGGGAGTTCTTCGCCCAGGAGGTGA
    TGATCCGCGTGTTCGGCAAGCCCGACGGCAAGCCCGAGTTCGAGGAGGCC
    GACACCCCCGAGAAGCTGCGCACCCTGCTGGCCGAGAAGCTGAGCAGCCG
    CCCTGAGGCCGTGCACGAGTACGTGACTCCTCTGTTCGTGAGCCGCGCCC
    CCAACCGCAAGATGAGCGGTCAGGGTCACATGGAGACCGTGAAGAGCGCC
    AAGCGCCTGGACGAGGGCGTGAGCGTGCTGCGCGTGCCCCTGACCCAGCT
    GAAGCTGAAGGACCTGGAGAAGATGGTGAACCGCGAGCGCGAGCCCAAGC
    TGTACGAGGCCCTGAAGGCCCGCCTGGAGGCCCACAAGGACGACCCCGCC
    AAGGCCTTCGCCGAGCCCTTCTACAAGTACGACAAGGCCGGCAACCGCAC
    CCAGCAGGTGAAGGCCGTGCGCGTGGAGCAGGTGCAGAAGACCGGCGTGT
    GGGTGCGCAACCACAACGGCATCGCCGACAACGCCACCATGGTGCGCGTG
    GACGTGTTCGAGAAGGGCGACAAGTACTACCTGGTGCCCATCTACAGCTG
    GCAGGTGGCCAAGGGCATCCTGCCCGACCGCGCCGTGGTGCAGGGCAAGG
    ACGAGGAGGACTGGCAGCTGATCGACGACAGCTTCAACTTCAAGTTCAGC
    CTGCACCCCAACGACCTGGTGGAGGTGATCACCAAGAAGGCCCGCATGTT
    CGGCTACTTCGCCAGCTGCCACCGCGGCACCGGCAACATCAACATCCGCA
    TCCACGACCTGGACCACAAGATCGGCAAGAACGGCATCCTGGAGGGCATC
    GGCGTGAAGACCGCCCTGAGCTTCCAGAAGTACCAGATCGACGAGCTGGG
    CAAGGAGATCCGCCCCTGCCGCCTGAAGAAGCGCCCTCCTGTGCGCTAA
  • Provided below is the corresponding amino acid sequence of a N. meningitidis Cas9 molecule.
  • (SEQ ID NO: DD4)
    MAAFKPNPINYILGLDIGIASVGWAMVEIDEDENPICLIDLGVRVFERAE
    VPKTGDSLAMARRLARSVRRLTRRRAHRLLRARRLLKREGVLQAADFDEN
    GLIKSLPNTPWQLRAAALDRKLTPLEWSAVLLHLIKHRGYLSQRKNEGET
    ADKELGALLKGVADNAHALQTGDFRTPAELALNKFEKESGHIRNQRGDYS
    HTFSRKDLQAELILLFEKQKEFGNPHVSGGLKEGIETLLMTQRPALSGDA
    VQKMLGHCTFEPAEPKAAKNTYTAERFIWLTKLNNLRILEQGSERPLTDT
    ERATLMDEPYRKSKLTYAQARKLLGLEDTAFFKGLRYGKDNAEASTLMEM
    KAYHAISRALEKEGLKDKKSPLNLSPELQDEIGTAFSLFKTDEDITGRLK
    DRIQPEILEALLKHISFDKFVQISLKALRRIVPLMEQGKRYDEACAEIYG
    DHYGKKNTEEKIYLPPIPADEIRNPVVLRALSQARKVINGVVRRYGSPAR
    IHIETAREVGKSFKDRKEIEKRQEENRKDREKAAAKFREYFPNFVGEPKS
    KDILKLRLYEQQHGKCLYSGKEINLGRLNEKGYVEIDHALPFSRTWDDSF
    NNKVLVLGSENQNKGNQTPYEYFNGKDNSREWQEFKARVETSRFPRSKKQ
    RILLQKFDEDGFKERNLNDTRYVNRFLCQFVADRMRLTGKGKKRVFASNG
    QITNLLRGFWGLRKVRAENDRHHALDAVVVACSTVAMQQKITRFVRYKEM
    NAFDGKTIDKETGEVLHQKTHFPQPWEFFAQEVMIRVFGKPDGKPEFEEA
    DTPEKLRTLLAEKLSSRPEAVHEYVTPLFVSRAPNRKMSGQGHMETVKSA
    KRLDEGVSVLRVPLTQLKLKDLEKMVNREREPKLYEALKARLEAHKDDPA
    KAFAEPFYKYDKAGNRTQQVKAVRVEQVQKTGVWVRNHNGIADNATMVRV
    DVFEKGDKYYLVPIYSWQVAKGILPDRAVVQGKDEEDWQLIDDSFNFKFS
    LHPNDLVEVITKKARMFGYFASCHRGTGNINIRIHDLDHKIGKNGILEGI
    GVKTALSFQKYQIDELGKEIRPCRLKKRPPVR*
  • Provided below is an amino acid sequence of a S. aureus Cas9 molecule.
  • (SEQ ID NO: DD5)
    MKRNYILGLDIGITSVGYGIIDYETRDVIDAGVRLFKEANVENNEGRRSK
    RGARRLKRRRRHRIQRVKKLLFDYNLLTDHSELSGINPYEARVKGLSQKL
    SEEEFSAALLHLAKRRGVHNVNEVEEDTGNELSTKEQISRNSKALEEKYV
    AELQLERLKKDGEVRGSINRFKTSDYVKEAKQLLKVQKAYHQLDQSFIDT
    YIDLLETRRTYYEGPGEGSPFGWKDIKEWYEMLMGHCTYFPEELRSVKYA
    YNADLYNALNDLNNLVITRDENEKLEYYEKFQIIENVFKQKKKPTLKQIA
    KEILVNEEDIKGYRVTSTGKPEFTNLKVYHDIKDITARKEIIENAELLDQ
    IAKILTIYQSSEDIQEELTNLNSELTQEEIEQISNLKGYTGTHNLSLKAI
    NLILDELWHTNDNQIAIFNRLKLVPKKVDLSQQKEIPTTLVDDFILSPVV
    KRSFIQSIKVINAIIKKYGLPNDIIIELAREKNSKDAQKMINEMQKRNRQ
    TNERIEEIIRTTGKENAKYLIEKIKLHDMQEGKCLYSLEAIPLEDLLNNP
    FNYEVDHIIPRSVSFDNSFNNKVLVKQEENSKKGNRTPFQYLSSSDSKIS
    YETFKKHILNLAKGKGRISKTKKEYLLEERDINRFSVQKDFINRNLVDTR
    YATRGLMNLLRSYFRVNNLDVKVKSINGGFTSFLRRKWKFKKERNKGYKH
    HAEDALIIANADFIFKEWKKLDKAKKVMENQMFEEKQAESMPEIETEQEY
    KEIFITPHQIKHIKDFKDYKYSHRVDKKPNRELINDTLYSTRKDDKGNTL
    IVNNLNGLYDKDNDKLKKLINKSPEKLLMYHHDPQTYQKLKLIMEQYGDE
    KNPLYKYYEETGNYLTKYSKKDNGPVIKKIKYYGNKLNAHLDITDDYPNS
    RNKVVKLSLKPYRFDVYLDNGVYKFVTVKNLDVIKKENYYEVNSKCYEEA
    KKLKKISNQAEFIASFYNNDLIKINGELYRVIGVNNDLLNRIEVNMIDIT
    YREYLENMNDKRPPRIIKTIASKTQSIKKYSTDILGNLYEVKSKKHPQII
    KKG*
  • Provided below is an exemplary codon optimized nucleic acid sequence encoding a Cas9 molecule of S. aureus Cas9.
  • (SEQ ID NO: DD6)
    ATGAAAAGGAACTACATTCTGGGGCTGGACATCGGGATTACAAGCGTGGG
    GTATGGGATTATTGACTATGAAACAAGGGACGTGATCGACGCAGGCGTCA
    GACTGTTCAAGGAGGCCAACGTGGAAAACAATGAGGGACGGAGAAGCAAG
    AGGGGAGCCAGGCGCCTGAAACGACGGAGAAGGCACAGAATCCAGAGGGT
    GAAGAAACTGCTGTTCGATTACAACCTGCTGACCGACCATTCTGAGCTGA
    GTGGAATTAATCCTTATGAAGCCAGGGTGAAAGGCCTGAGTCAGAAGCTG
    TCAGAGGAAGAGTTTTCCGCAGCTCTGCTGCACCTGGCTAAGCGCCGAGG
    AGTGCATAACGTCAATGAGGTGGAAGAGGACACCGGCAACGAGCTGTCTA
    CAAAGGAACAGATCTCACGCAATAGCAAAGCTCTGGAAGAGAAGTATGTC
    GCAGAGCTGCAGCTGGAACGGCTGAAGAAAGATGGCGAGGTGAGAGGGTC
    AATTAATAGGTTCAAGACAAGCGACTACGTCAAAGAAGCCAAGCAGCTGC
    TGAAAGTGCAGAAGGCTTACCACCAGCTGGATCAGAGCTTCATCGATACT
    TATATCGACCTGCTGGAGACTCGGAGAACCTACTATGAGGGACCAGGAGA
    AGGGAGCCCCTTCGGATGGAAAGACATCAAGGAATGGTACGAGATGCTGA
    TGGGACATTGCACCTATTTTCCAGAAGAGCTGAGAAGCGTCAAGTACGCT
    TATAACGCAGATCTGTACAACGCCCTGAATGACCTGAACAACCTGGTCAT
    CACCAGGGATGAAAACGAGAAACTGGAATACTATGAGAAGTTCCAGATCA
    TCGAAAACGTGTTTAAGCAGAAGAAAAAGCCTACACTGAAACAGATTGCT
    AAGGAGATCCTGGTCAACGAAGAGGACATCAAGGGCTACCGGGTGACAAG
    CACTGGAAAACCAGAGTTCACCAATCTGAAAGTGTATCACGATATTAAGG
    ACATCACAGCACGGAAAGAAATCATTGAGAACGCCGAACTGCTGGATCAG
    ATTGCTAAGATCCTGACTATCTACCAGAGCTCCGAGGACATCCAGGAAGA
    GCTGACTAACCTGAACAGCGAGCTGACCCAGGAAGAGATCGAACAGATTA
    GTAATCTGAAGGGGTACACCGGAACACACAACCTGTCCCTGAAAGCTATC
    AATCTGATTCTGGATGAGCTGTGGCATACAAACGACAATCAGATTGCAAT
    CTTTAACCGGCTGAAGCTGGTCCCAAAAAAGGTGGACCTGAGTCAGCAGA
    AAGAGATCCCAACCACACTGGTGGACGATTTCATTCTGTCACCCGTGGTC
    AAGCGGAGCTTCATCCAGAGCATCAAAGTGATCAACGCCATCATCAAGAA
    GTACGGCCTGCCCAATGATATCATTATCGAGCTGGCTAGGGAGAAGAACA
    GCAAGGACGCACAGAAGATGATCAATGAGATGCAGAAACGAAACCGGCAG
    ACCAATGAACGCATTGAAGAGATTATCCGAACTACCGGGAAAGAGAACGC
    AAAGTACCTGATTGAAAAAATCAAGCTGCACGATATGCAGGAGGGAAAGT
    GTCTGTATTCTCTGGAGGCCATCCCCCTGGAGGACCTGCTGAACAATCCA
    TTCAACTACGAGGTCGATCATATTATCCCCAGAAGCGTGTCCTTCGACAA
    TTCCTTTAACAACAAGGTGCTGGTCAAGCAGGAAGAGAACTCTAAAAAGG
    GCAATAGGACTCCTTTCCAGTACCTGTCTAGTTCAGATTCCAAGATCTCT
    TACGAAACCTTTAAAAAGCACATTCTGAATCTGGCCAAAGGAAAGGGCCG
    CATCAGCAAGACCAAAAAGGAGTACCTGCTGGAAGAGCGGGACATCAACA
    GATTCTCCGTCCAGAAGGATTTTATTAACCGGAATCTGGTGGACACAAGA
    TACGCTACTCGCGGCCTGATGAATCTGCTGCGATCCTATTTCCGGGTGAA
    CAATCTGGATGTGAAAGTCAAGTCCATCAACGGCGGGTTCACATCTTTTC
    TGAGGCGCAAATGGAAGTTTAAAAAGGAGCGCAACAAAGGGTACAAGCAC
    CATGCCGAAGATGCTCTGATTATCGCAAATGCCGACTTCATCTTTAAGGA
    GTGGAAAAAGCTGGACAAAGCCAAGAAAGTGATGGAGAACCAGATGTTCG
    AAGAGAAGCAGGCCGAATCTATGCCCGAAATCGAGACAGAACAGGAGTAC
    AAGGAGATTTTCATCACTCCTCACCAGATCAAGCATATCAAGGATTTCAA
    GGACTACAAGTACTCTCACCGGGTGGATAAAAAGCCCAACAGAGAGCTGA
    TCAATGACACCCTGTATAGTACAAGAAAAGACGATAAGGGGAATACCCTG
    ATTGTGAACAATCTGAACGGACTGTACGACAAAGATAATGACAAGCTGAA
    AAAGCTGATCAACAAAAGTCCCGAGAAGCTGCTGATGTACCACCATGATC
    CTCAGACATATCAGAAACTGAAGCTGATTATGGAGCAGTACGGCGACGAG
    AAGAACCCACTGTATAAGTACTATGAAGAGACTGGGAACTACCTGACCAA
    GTATAGCAAAAAGGATAATGGCCCCGTGATCAAGAAGATCAAGTACTATG
    GGAACAAGCTGAATGCCCATCTGGACATCACAGACGATTACCCTAACAGT
    CGCAACAAGGTGGTCAAGCTGTCACTGAAGCCATACAGATTCGATGTCTA
    TCTGGACAACGGCGTGTATAAATTTGTGACTGTCAAGAATCTGGATGTCA
    TCAAAAAGGAGAACTACTATGAAGTGAATAGCAAGTGCTACGAAGAGGCT
    AAAAAGCTGAAAAAGATTAGCAACCAGGCAGAGTTCATCGCCTCCTTTTA
    CAACAACGACCTGATTAAGATCAATGGCGAACTGTATAGGGTCATCGGGG
    TGAACAATGATCTGCTGAACCGCATTGAAGTGAATATGATTGACATCACT
    TACCGAGAGTATCTGGAAAACATGAATGATAAGCGCCCCCCTCGAATTAT
    CAAAACAATTGCCTCTAAGACTCAGAGTATCAAAAAGTACTCAACCGACA
    TTCTGGGAAACCTGTATGAGGTGAAGAGCAAAAAGCACCCTCAGATTATC
    AAAAAGGGC
  • If any of the above Cas9 sequences are fused with a peptide or polypeptide at the C-terminus, it is understood that the stop codon will be removed.
  • Other Cas Molecules and Cas Polypeptides
  • Various types of Cas molecules or Cas polypeptides can be used to practice the inventions disclosed herein. In some embodiments, Cas molecules of Type II Cas systems are used. In other embodiments, Cas molecules of other Cas systems are used. For example, Type I or Type III Cas molecules may be used. Exemplary Cas molecules (and Cas systems) are described, e.g., in Haft et al. (2005) PLOS COMPUTATIONAL BIOLOGY 1(6): e60, and in Makarova et al. (2011) NATURE REVIEW MICROBIOLOGY 9:467-477, the contents of which are incorporated herein by reference in their entirety. Exemplary Cas molecules (and Cas systems) are also shown in Table 600.
  • TABLE 600
    Cas Systems
    Structure of Families
    Gene System type Name from encoded protein (and superfamily)
    name or subtype Haft et al.§ (PDB accessions) of encoded protein#** Representatives
    cas1 Type I cas1 3GOD, 3LFX COG1518 SERP2463,
    Type II and 2YZS SPy1047 and ygbT
    Type III
    cas2 Type I cas2 2IVY, 2I8E COG1343 and SERP2462,
    Type II and 3EXC COG3512 SPy1048, SPy1723
    Type III (N-terminal
    domain) and ygbF
    cas3′ Type I‡‡ cas3 NA COG1203 APE1232 and ygcB
    cas3″ Subtype I-A NA NA COG2254 APE1231 and
    Subtype I-B BH0336
    cas4 Subtype I-A cas4 and NA COG1468 APE1239 and
    Subtype I-B csa1 BH0340
    Subtype I-C
    Subtype I-D
    Subtype II-B
    cas5 Subtype I-A cas5a, cas5d, 3KG4 COG1688 APE1234, BH0337,
    Subtype I-B cas5e, cas5h, (RAMP) devS and ygcI
    Subtype I-C cas5p, cas5t
    Subtype I-E and cmx5
    cas6 Subtype I-A cas6 and 3I4H COG1583 and PF1131 and slr7014
    Subtype I-B cmx6 COG5551
    Subtype I-D (RAMP)
    Subtype III-A
    Subtype III-B
    cas6e Subtype I-E cse3 1WJ9 (RAMP) ygcH
    cas6f Subtype I-F csy4 2XLJ (RAMP) y1727
    cas7 Subtype I-A csa2, csd2, NA COG1857 and devR and ygcJ
    Subtype I-B cse4, csh2, COG3649
    Subtype I-C csp1 and (RAMP)
    Subtype I-E cst2
    cas8a1 Subtype I-A‡‡ cmx1, cst1, NA BH0338-like LA3191§§ and
    csx8, csx13 PG2018§§
    and CXXC-
    CXXC
    cas8a2 Subtype I-A‡‡ csa4 and NA PH0918 AF0070, AF1873,
    csx9 MJ0385, PF0637,
    PH0918 and
    SSO1401
    cas8b Subtype I-B‡‡ csh1 and NA BH0338-like MTH1090 and
    TM1802 TM1802
    cas8c Subtype I-C‡‡ csd1 and NA BH0338-like BH0338
    csp2
    cas9 Type II‡‡ csn1 and NA COG3513 FTN_0757 and
    csx12 SPy1046
    cas10 Type III‡‡ cmr2, csm1 NA COG1353 MTH326,
    and csx11 Rv2823c§§ and
    TM1794§§
    cas10d Subtype I-D‡‡ csc3 NA COG1353 slr7011
    csy1 Subtype I-F‡‡ csy1 NA y1724-like y1724
    csy2 Subtype I-F csy2 NA (RAMP) y1725
    csy3 Subtype I-F csy3 NA (RAMP) y1726
    cse1 Subtype I-E‡‡ cse1 NA YgcL-like ygcL
    cse2 Subtype I-E cse2 2ZCA YgcK-like ygcK
    csc1 Subtype I-D csc1 NA alr1563-like alr1563
    (RAMP)
    csc2 Subtype I-D csc1 and NA COG1337 slr7012
    csc2 (RAMP)
    csa5 Subtype I-A csa5 NA AF1870 AF1870, MJ0380,
    PF0643 and
    SSO1398
    csn2 Subtype II-A csn2 NA SPy1049-like SPy1049
    csm2 Subtype III-A‡‡ csm2 NA COG1421 MTH1081 and
    SERP2460
    csm3 Subtype III-A csc2 and NA COG1337 MTH1080 and
    csm3 (RAMP) SERP2459
    csm4 Subtype III-A csm4 NA COG1567 MTH1079 and
    (RAMP) SERP2458
    csm5 Subtype III-A csm5 NA COG1332 MTH1078 and
    (RAMP) SERP2457
    csm6 Subtype III-A APE2256 2WTE COG1517 APE2256 and
    and csm6 SSO1445
    cmr1 Subtype III-B cmr1 NA COG1367 PF1130
    (RAMP)
    cmr3 Subtype III-B cmr3 NA COG1769 PF1128
    (RAMP)
    cmr4 Subtype III-B cmr4 NA COG1336 PF1126
    (RAMP)
    cmr5 Subtype III-B‡‡ cmr5 2ZOP and COG3337 MTH324 and
    2OEB PF1125
    cmr6 Subtype III-B cmr6 NA COG1604 PF1124
    (RAMP)
    csb1 Subtype I-U GSU0053 NA (RAMP) Balac_1306 and
    GSU0053
    csb2 Subtype I-U§§ NA NA (RAMP) Balac_1305 and
    GSU0054
    csb3 Subtype I-U NA NA (RAMP) Balac_1303§§
    csx17 Subtype I-U NA NA NA Btus_2683
    csx14 Subtype I-U NA NA NA GSU0052
    csx10 Subtype I-U csx10 NA (RAMP) Caur_2274
    csx16 Subtype III-U VVA1548 NA NA VVA1548
    csaX Subtype III-U csaX NA NA SSO1438
    csx3 Subtype III-U csx3 NA NA AF1864
    csx1 Subtype III-U csa3, csx1, 1XMX and COG1517 and MJ1666, NE0113,
    csx2, DXTHG, 2I71 COG4006 PF1127 and
    NE0113 and TM1812
    TIGR02710
    csx15 Unknown NA NA TTE2665 TTE2665
    csf1 Type U csf1 NA NA AFE_1038
    csf2 Type U csf2 NA (RAMP) AFE_1039
    csf3 Type U csf3 NA (RAMP) AFE_1040
    csf4 Type U csf4 NA NA AFE_1037
  • IV. Linkers to Connect Cas9 Molecules or Cas9 Polypeptides to a Template Binding Domain
  • In an embodiment, a linker covalently connects the Cas9 molecule to the template binding domain (e.g., a DNA binding domain).
  • In an embodiment, a linker is a short peptide sequence that connects protein domains. In another embodiment, a linker is a short peptide sequence that connects a protein domain and a nucleic acid (e.g., DNA or RNA). In an embodiment, a linker is a short peptide sequence that connects a protein domain and a small molecule. In an embodiment, a linker is a short peptide sequence that connects two small molecules. Linkers are often composed of flexible residues like glycine and serine so that the adjacent protein domains are free to move relative to one another. In certain embodiments, the linker has sufficient length and flexibility to allow the Cas9 molecule to bind to a target nucleic acid and simultaneously allow the target binding domain to associate with a nucleic acid template system, e.g., so that one binding event does not sterically block the other, and/or so that two adjacent domains do not sterically interfere with one another.
  • The linker can attach to the C-terminus or N-terminus of the Cas9 molecule.
  • The linker can attach to the C-terminus or N-terminus of the template binding domain.
  • The linker can attach to the C-terminus or N-terminus of the template binding domain partner.
  • The linker can attach to the C-terminus or N-terminus of the template nucleic acid.
  • In some embodiments, the linker length is from about 6 to 60 amino acids. The linker may be, e.g., 6-10, 10-15, 15-20, 20-30, 30-40, 40-50, or 50-60 amino acids in length. The linker may be, e.g., at least 6, 7, 8, 9, 10, 15, 20, 30, 40, or 50 amino acids in length. In other embodiments, the linker is, e.g., at most 7, 8, 9, 10, 15, 20, 30, 40, 50, or 60 amino acids in length. Ranges comprising any combination of these endpoints are also envisioned.
  • In some embodiments, the linker is encoded by a nucleic acid sequence of 6 to 60 nucleotides or base pairs. The nucleic acid may be, e.g., 6-10, 10-15, 15-20, 20-30, 30-40, 40-50, or 50-60 nucleotides in length. The linker may be, e.g., at least 6, 7, 8, 9, 10, 15, 20, 30, 40, or 50 nucleotides in length. In some embodiments, the linker is, e.g., at most 7, 8, 9, 10, 15, 20, 30, 40, 50, or 60 nucleotides in length. Ranges comprising any combination of these endpoints are also envisioned.
  • In some embodiments, the linker comprises glycine and serine residues. In some embodiments the linker consists of glycine and serine residues. For instance, the linker may comprise one of more modules such as GGS, GSGS, GGGS, GGGGS or GGSG. In some embodiments, the linker comprises a plurality of modules comprising glycine and serine, e.g., at least 2, 3, 4, 5, 10, or 15 of these modules, and/or at most 3, 4, 5, 10, 15, or 20 of these modules, or any combination of these endpoints. In some embodiments, each module in the linker has the same sequence, and in other embodiments, at least two modules in a linker have different sequences from each other.
  • In some embodiments, the linker is an XTEN linker or a variation of an XTEN linker such as SGSETPGTSESA, SGSETPGTSESATPES, or SGSETPGTSESATPEGGSGGS.
  • Additional information on the XTEN linker may be found in Schellenberger et al. (2009), NATURE BIOTECHNOLOGY 27: 1186-1190, the entire contents of which are incorporated herein by reference.
  • Exemplary linker modules are given in Table 2:
  • GGS
    GSGS
    GGGS
    GGGGS
    GGSG
    SGSETPGTSESA
    SGSETPGTSESATPES
    SGSETPGTSESATPEGGSGGS
  • Additional exemplary linker modules are given in Table 3:
  • Name Description Length (nt)
    BBa_J176131 PLrigid 60
    BBa_J18920 2aa GS linker 6
    BBa_J18921 6aa [GS]x linker 18
    BBa_J18922 10aa [GS]x linker 30
    BBa_K105012 10 aa flexible protein domain linker 30
    BBa_K133132 8 aa protein domain linker 24
    BBa_K1486003 flexible linker 2x (GGGS) 24
    BBa_K1486004 flexible linker 2x (GGGGS) 30
    BBa_K1486037 linker 39
    BBa_K157009 Split fluorophore linker; Freiburg 51
    standard
    BBa_K157013 15 aa flexible glycine-serine protein 45
    domain linker; Freiburg standard
    BBa_K243004 Short Linker (Gly-Gly-Ser-Gly) 12
    BBa_K243005 Middle Linker (Gly-Gly-Ser-Gly)x2 24
    BBa_K243006 Long Linker (Gly-Gly-Ser-Gly)x3 36
    BBa_K243029 GSAT Linker 108
    BBa_K243030 SEG 108
    BBa_K404300 SEG-Linker 108
    BBa_K404301 GSAT-Linker 108
    BBa_K404303 Z-EGFR-1907_Short-Linker 192
    BBa_K404304 Z-EGFR-1907_Middle-Linker 204
    BBa_K404305 Z-EGFR-1907_Long-Linker 216
    BBa_K404306 Z-EGFR-1907_SEG-Linker 288
    BBa_K416001 (Gly4Ser)3 Flexible Peptide Linker 45
    BBa_K648005 Short Fusion Protein Linker: GGSG 12
    with standard 25 prefix/suffix
    BBa_K648006 Long 10AA Fusion Protein Linker 30
    with Standard 25 Prefix/Suffix
    BBa_K648007 Medium 6AA Fusion Protein Linker: 18
    GGSGGS with Standard 25
    Prefix/Suffix
  • Linkers can comprise a direct bond or an atom such as, e.g., an oxygen (O) or sulfur (S), a unit such as —NR— wherein R is hydrogen or alkyl, —C(O)—, —C(O)O—, —C(O)NH—, SO, SO2, —SO2NH— or a chain of atoms, such as substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, arylalkyl, heteroarylalkyl. In some embodiments, one or more methylenes in the chain of atoms can be replaced with one or more of O, S, S(O), SO2, —SO2NH—, —NR—, —NR2, —C(O)—, —C(O)O—, —C(O)NH—, a cleavable linking group, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, and substituted or unsubstituted heterocyclic.
  • In some embodiments, the template binding domain is attached to the Cas9 molecule through a linker that is itself stable under physiological conditions, such as an alkylene chain, and does not result in release of the template binding domain from the Cas9 molecule for at least 2, 3, 4, 5, 10, 15, 24 or 48 hours or for at least 1, 2, 3, 4, 5 or 10 days when administered to a subject. In some embodiments, the template binding domain and the Cas9 molecule comprise residues of a functional groups through which reaction and linkage of the template binding domain to the Cas9 molecule was achieved. In some embodiments, the functional groups, which may be the same or different, terminal or internal, of the template binding domain or Cas9 molecule comprise an amino, acid, imidazole, hydroxyl, thio, acyl halide, —HC═CH—, —C≡C— group, or derivative thereof. In some embodiments, the linker comprises a hydrocarbylene group wherein one or more methylene groups is optionally replaced by a group Y (provided that none of the Y groups are adjacent to each other), wherein each Y, independently for each occurrence, is selected from, substituted or unsubstituted aryl, heteroaryl, cycloalkyl, heterocycloalkyl, or —O—, C(═X) (wherein X is NR1, O or S), —NR1—, —NR1C(O)—, —C(O)NR1—, —S(O)n—, —NR1S(O)n—, —S(O)nNR1—, —NR1C(O)—NR1—; and R1, independently for each occurrence, represents H or a lower alkyl and wherein n is 0, 1, or 2.
  • In some embodiments, the linker comprises an alkylene moiety or a heteroalkylene moiety (e.g., an alkylene glycol moiety such as ethylene glycol). In some embodiments, a linker comprises a poly-L-glutamic acid, polylactic acid, poly(ethyleneimine), an oligosaccharide, an amino acid (e.g., glycine), an amino acid chain, or any other suitable linkage. The linker groups can be biologically inactive, such as a PEG, polyglycolic acid, or polylactic acid chain. In certain embodiments, the linker group represents a derivatized or non-derivatized amino acid (e.g., glycine).
  • The Cas9 molecule attached to the linker may be any Cas9 molecule described herein, e.g., a Cas9 molecule of Section III, e.g., a nickase or a Cas9 molecule capable of making a double stranded break. The template binding domain attached to the linker may be any template binding domain described herein, e.g., a template binding domain of Section V, e.g., a DNA-binding domain.
  • V. Template Binding Domains
  • In some embodiments, the template binding domain comprises a plurality of domains that bind a template binding domain partner, e.g., DNA-binding molecules. For instance, a first DNA-binding molecule and a second DNA-binding molecule may, on their own, be poor choices because each has a low affinity and optionally also a short recognition sequence that is present in multiple copies in the genome of the cell to be altered. However, when combined, the first and second molecules could give a higher affinity and optionally also a longer recognition sequence that is rarer in the genome of the cell to be altered. Accordingly, in some embodiments, the template binding domain comprises a plurality of domains that bind a template binding domain partner, e.g., DNA-binding molecules. For instance, the template binding domain may comprise two or more, three or more, four or more, five or more, ten or more, 2-4, 4-6, 6-8, or 8-10 domains that bind a template binding domain partner, e.g., DNA-binding molecules.
  • The template binding domain can be covalently bound to the Cas9 molecule, e.g., using a linker. The template binding domain and the Cas9 can be part of the same fusion protein. In some embodiments, the Cas9 molecule and the template binding domain are part of the same gene and are transcribed to form a single mRNA which is then translated into a single protein. In other embodiments, the Cas9 molecule and the template binding domain are produced separately and then joined covalently or non-covalently. An example of a template binding domain which binds non-covalently to the Cas9 molecule is a template-binding domain that comprises a region with specific affinity for Cas9, e.g., an antibody that recognizes Cas9, e.g., an scFv antibody or another type of antibody that contains sufficient CDR sequences for binding to Cas9. In some embodiments, the template binding domain is derived from a wild-type protein. For example, the template binding domain may be a fragment of a wild type protein, a mutagenized wild type protein, a mutagenized wild-type protein fragment, a synthetic protein that has been modeled after the three dimensional structure of a naturally-occurring protein. In some embodiments, the template binding domain is mutagenized to increase its affinity for a template binding domain partner. In some embodiments, the template binding domain is mutagenized to decrease its affinity for a template binding domain partner.
  • V.1 Template Binding Domains that are DNA-Binding Polypeptides
  • In some embodiments, the template binding domain is a polypeptide, e.g., a protein or protein domain. This polypeptide can bind to the major groove of a target DNA sequence and/or a minor groove of a target DNA sequence. It can comprise one or more of the following domains: zinc finger, helix-turn-helix, leucine zipper, winged helix, winged helix turn helix, helix-loop-helix, HMG-box, and Wor3 domain. It can bind single stranded DNA or double stranded DNA. In some embodiments, the DNA-binding polypeptide is identical in sequence to a wild-type protein, and in other embodiments it comprises one or more mutations, e.g., deletions, relative to a wild-type protein.
  • In some embodiments, the DNA-binding polypeptide comprises a mutation relative to a wild-type DNA-binding protein. For example, if the wild-type DNA-binding protein must bind a ligand or co-activator before it can bind DNA, the DNA-binding polypeptide is optionally mutated to a constitutively active form. Similarly, if the wild-type DNA-binding protein is incapable of binding to DNA in the presence of a ligand or co-activator before it can bind DNA, the DNA-binding polypeptide can also be mutated to a constitutively active form. In some embodiments, the DNA-binding polypeptide carries a deletion relative to a wild-type protein, e.g., a transcriptional activation or repression domain or a catalytic domain is removed. In some embodiments, the DNA-binding polypeptide consists only of the DNA-binding region of the corresponding wild-type DNA-binding protein.
  • In some embodiments, the DNA-binding polypeptide recognizes chemically modified DNA, e.g., methylated DNA. In some embodiments, the DNA-binding polypeptide recognizes a chemical modification that is rare in or absent from the genome of the cell to be altered. This can help avoid the DNA-binding polypeptide non-specifically binding to the cell's genome.
  • Several exemplary DNA binding proteins are given below.
  • Operon
  • In some embodiments, the DNA-binding polypeptide is, or is derived from, a DNA-binding protein from an operon, e.g., a bacterial operon. The DNA-binding polypeptide may be, e.g., a repressor or an activator in the context of the operon. Generally, the DNA-binding polypeptide will not activate or repress transcription in the methods described herein. This can be achieved by, e.g., mutating transcriptional regulation domains, or choosing a DNA-binding polypeptide that does not engage the transcriptional machinery of the cell to be altered. For example, when altering the genome of a human cell, one could choose a DNA-binding peptide from a prokaryote, Archaea, single celled eukaryote, plant, or fungus. DNA-binding proteins from operons, and the nucleotide sequences to which they bind, are known in the art (see, e.g., Postle et al. (1984) NUCLEIC ACIDS RES. 12: 4849-63; Buvinger and Riley (1985) J. BACTERIOL. 163: 850-7; Laughon and Gesteland (1984) MOL. CELL BIOL. 4:260-7; Bram et al. (1986) EMBO J. 5: 603-8; Von Wilcken-Bergmann & Muller-Hill (1982) PROC. NAT'L. ACAD. SCI. 79: 2427-31; Heinrich et al. (1989) NUCLEIC ACIDS RES. 17: 7681-92; Osborne et al. (1989) NUCLEIC ACIDS RES. 17: 7671-80; Singleton et al. (1980) NUCLEIC ACIDS RES. 8: 1551-60; Widdowson et al. (1996) ANTIMICROB. AGENTS CHEMOTHER. 40: 2891-93; Oehler et al. (1994) EMBO J. 13: 3348-55; Bailone and Galibert (1980) NUCLEIC ACIDS RES. 8: 2147-64; and, Staacke et al. (1990) EMBO J. 9: 1963-7).
  • Exemplary DNA-binding proteins from operons are given in the table below. The template-binding domain may comprise one or more of these proteins or polypeptides derived therefrom.
  • TABLE V.1
    DNA-binding proteins from operons
    DNA sequence recognized by the template
    Template binding domain binding domain
    TetR represser Tet-O
    LacI represser Lac operon 01
    Gal4 represser UAS
    Represser protein C1 Operator L and R
    Trp represser Trp operator
  • Transcription Factors
  • In some embodiments, the DNA-binding polypeptide is, or is derived from, a transcription factor. The DNA-binding polypeptide may be or be derived from, e.g., a repressor or an activator in its wild-type context. Generally, the DNA-binding polypeptide will not activate or repress transcription in the methods described herein. This can be achieved by, e.g., mutating transcriptional regulation domains, such as the trans-activating domain (TAD) or any other domain that binds a transcription co-regulator. This can also be achieved by choosing a DNA-binding polypeptide that does not engage the transcriptional machinery of the cell to be altered. For example, when altering the genome of a human cell, one could choose a DNA-binding peptide from a prokaryote, Archaea, single celled eukaryote, plant, or fungus.
  • The transcription factor, in some embodiments, falls into one or more of several categories as set out here. The transcription factor may be a specific transcription factor and/or an upstream transcription factor. It may be constitutively active or conditionally active. If conditionally active, it may be developmental or signal-dependent. In some embodiments, the transcription factor is a resident nuclear factor and/or comprises a nuclear localization signal (NLS).
  • Exemplary transcription factors are given in the table below. The template-binding domain may comprise one or more of these transcription factors or polypeptides derived therefrom.
  • TABLE V.2
    Transcription factors
    Template binding domain
    Yeast transcription factors
    FHL1
    ROX1
    CMR3
    SUT2
    GAL4
    USV1
    AFT2
    CUP9
    TBF1
    GCR1
    MET31
    ECM23
    RDR1
    HAP5
    TYE7
    YRM1
    YRR1
    AZF1
    CIN5
    MSN1
    MSN1
    INO4
    HAL9
    HAL9
    YAP7
    YAP7
    DAL82
    RAP1
    SKO1
    FKH2
    CRZ1
    RGM1
    CEP3
    MCM1
    MSN2
    MAC1
    STB4
    SOK2
    ARG81
    ORC1
    YOX1
    YAP1
    LEU3
    LEU3
    SFP1
    HAP1
    ECM22
    ECM22
    ACE2
    CHA4
    GAT3
    BAS1
    ABF1
    HAP4
    MSN4
    PHD1
    PHD1
    RGT1
    RSF2
    CBF1
    GZF3
    ZAP1
    YAP5
    GAT4
    FKH1
    XBP1
    CST6
    SKN7
    STB5
    NDT80
    STE12
    STP2
    RIM101
    YAP3
    YAP3
    HAP2
    MIG2
    TOS8
    AFT1
    MIG1
    PDR1
    PHO4
    HAC1
    GAT1
    RPH1
    SPT15
    COM2
    SWI4
    DOT6
    GLN3
    MIG3
    GCN4
    URC2
    STP1
    YHP1
    CAD1
    CAD1
    ARO80
    SUM1
    RSC3
    YAP6
    MET32
    ADR1
    UPC2
    UME6
    STB3
    SWI5
    INO2
    GIS1
    NRG1
    LYS14
    LYS14
    UGA3
    PHO2
    MBP1
    RPN4
    RDS1
    HCM1
    MATALPHA2
    REI1
    THI2
    TBS1
    TBS1
    TEC1
    NRG2
    REB1
    EDS1
    TOD6
    HAP3
    Transcription factor families found, e.g., in plants
    ABI3VP1 family
    CAMTA family
    LFY family
    SBP family
    Alfin-like family
    CCAAT family
    LIM family
    Sigma70-like family
    AP2-EREBP family
    CPP family
    LOB family
    SRS family
    ARF family
    CSD family
    MADS family
    TAZ family
    ARR-B family
    DBP family
    mTERF family
    TCP family
    BBR/BPC family
    E2F-DP family
    MYB family
    Tify family
    BES1 family
    EIL family
    MYB-related family
    TIG family
    bHLH family
    FAR1 family
    NAC family
    Trihelix family
    BSD family
    FHA family
    NOZZLE family
    TUB family
    bZIP family
    G2-like family
    OFP family
    ULT family
    C2C2-CO-like family
    GeBP family
    Orphans family
    VARL family
    C2C2-Dof family
    GRAS family
    PBF-2-like family
    VOZ family
    C2C2-GATA family
    GRF family
    PLATZ family
    WRKY family
    C2C2-YABBY family
    HB family
    RWP-RK family
    zf-HD family
    C2H2 family
    HRT family
    S1Fa-like family
    Zn-clus family
    C3H family
    HSF family
    SAP family
  • Endonucleases
  • In some embodiments, the DNA-binding polypeptide is derived from an endonuclease. The DNA-binding domain may be a catalytically inactive endonuclease, e.g., may have a substitution in or deletion of the domain that catalyzes DNA cleavage. If the endonuclease has other activities such as DNA modification activity, one may introduce mutations into the other active domains as well.
  • The restriction endonuclease may be, e.g., of Type I; Type II, e.g., Type IIR, Type IIS, or Type IIG; Type III; or Type IV.
  • In some embodiments where the endonuclease has a short recognition sequence, it may be used in combination with other DNA-binding polypeptides, e.g., other endonuclease-derived polypeptides, to achieve higher affinity binding to a longer recognition site.
  • In some embodiments, the endonuclease recognizes modified DNA, e.g., methylated DNA, and the template binding domain partner comprises modified DNA.
  • Exemplary restriction endonucleases are given in the table below. The template-binding domain may comprise one or more of these endonucleases or polypeptides derived therefrom.
  • TABLE V.3
    Endonucleases
    Restriction endonucleases
    AatII
    AbaSI
    Acc65I
    AccI
    AciI
    AclI
    AcuI
    AfeI
    AflII
    AflIII
    AgeI
    AhdI
    AleI
    AluI
    AlwI
    AlwNI
    ApaI
    ApaLI
    ApeKI
    ApoI
    AscI
    AseI
    AsiSI
    AvaI
    AvaII
    AvrII
    BaeGI
    BaeI
    BamHI
    BanI
    BanII
    BbsI
    BbvCI
    BbvI
    BccI
    BceAI
    BcgI
    BciVI
    BclI
    BcoDI
    BfaI
    BfuAI
    BfuCI
    BglI
    BglII
    BlpI
    BmgBI
    BmrI
    BmtI
    BpmI
    Bpu10I
    BpuEI
    BsaAI
    BsaBI
    BsaHI
    BsaI
    BsaJI
    BsaWI
    BsaXI
    BseRI
    BseYI
    BsgI
    BsiEI
    BsiHKAI
    BsiWI
    BslI
    BsmAI
    BsmBI
    BsmFI
    BsmI
    BsoBI
    Bsp1286I
    BspCNI
    BspDI
    BspEI
    BspHI
    BspMI
    BspQI
    BsrBI
    BsrDI
    BsrFI
    BsrGI
    BsrI
    BssHII
    BssKI
    BssSI
    BstAPI
    BstBI
    BstEII
    BstNI
    BstUI
    BstXI
    BstYI
    BstZ17I
    Bsu36I
    BtgI
    BtgZI
    BtsCI
    BtsI
    BtsIMutI
    Cac8I
    ClaI
    CspCI
    CviAII
    CviKI-1
    CviQI
    DdeI
    DpnI
    DpnII
    DraI
    DraIII
    DrdI
    EaeI
    EagI
    EarI
    EciI
    Eco53kI
    EcoNI
    EcoO109I
    EcoP15I
    EcoRI
    EcoRV
    FatI
    FauI
    Fnu4HI
    FokI
    FseI
    FspEI
    FspI
    HaeII
    HaeIII
    HgaI
    HhaI
    HincII
    HindIII
    HinfI
    HinP1I
    HpaI
    HpaII
    HphI
    Hpy166II
    Hpy188I
    Hpy188III
    Hpy99I
    HpyAV
    HpyCH4III
    HpyCH4IV
    HpyCH4V
    I-CeuI
    I-SceI
    KasI
    KpnI
    LpnPI
    MboI
    MboII
    MfeI
    MluCI
    MluI
    MlyI
    MmeI
    MnlI
    MscI
    MseI
    MslI
    MspA1I
    MspI
    MspJI
    MwoI
    NaeI
    NarI
    Nb.BbvCI
    Nb.BsmI
    Nb.BsrDI
    Nb.BtsI
    NciI
    NcoI
    NdeI
    NgoMIV
    NheI
    NlaIII
    NlaIV
    NmeAIII
    NotI
    NruI
    NsiI
    NspI
    Nt.AlwI
    Nt.BbvCI
    Nt.BsmAI
    Nt.BspQI
    Nt.BstNBI
    Nt.CviPII
    PacI
    PaeR7I
    PciI
    PflFI
    PflMI
    PI-PspI
    PI-SceI
    PleI
    PluTI
    PmeI
    PmlI
    PpuMI
    PshAI
    PsiI
    PspGI
    PspOMI
    PspXI
    PstI
    PvuI
    PvuII
    RsaI
    RsrII
    SacI
    SacII
    SalI
    SapI
    Sau3AI
    Sau96I
    SbfI
    ScaI
    ScrFI
    SexAI
    SfaNI
    SfcI
    SfiI
    SfoI
    SgrAI
    SmaI
    SmlI
    SnaBI
    SpeI
    Sphl
    SphI
    SspI
    StuI
    StyD4I
    StyI
    SwaI
    TaqαI
    Tfil
    TliI
    TseI
    Tsp45I
    Tsp509I
    TspMI
    TspRI
    Tth111I
    XbaI
    XcmI
    XhoI
    XmaI
    XmnI
    ZraI
  • TAL Effectors
  • In some embodiments, the DNA-binding polypeptide is, or is derived from, a TAL (transcription activator-like) effector. TAL effectors bind specifically to DNA through a series of 34-amino acid repeats, and engineering of these repeats tailors the specificity of the TAL effector to bind a desired DNA sequence. Details on how to engineer specificity are given in, e.g., U.S. Pat. No. 8,440,431. Briefly, each repeat in the TAL effector has a direct, linear correspondence with one nucleotide in the target site. Accordingly, one can readily engineer a TAL effector by selecting a first residue at position 12 and a second residue at position 13, in order to have that repeat bind to A, C, G, or T. Different repeats can be assembled to create a binding domain that is customized to recognize the desired target sequence. Table V.4 lists different combinations of amino acid residues that can be used to create repeats with specificity for a given nucleotide in the target binding sequence.
  • In some embodiments, the DNA-binding polypeptide is derived from a TALEN (TAL effector nuclease), and is mutated to lack nuclease activity. For example, there may be a substitution in or deletion of the domain that catalyzes DNA cleavage.
  • In some embodiments, the TAL effector is from, or is derived from, a TAL effector in a Xanthomonas bacterium, Ralstonia solanacearum, or Burkholderia rhizoxinica.
  • Exemplary TAL effectors and TALENs are given in the table below. The template-binding domain may comprise one or more of these TAL effectors and TALENs or polypeptides derived therefrom.
  • TABLE V.5
    Publications describing TAL effectors and TALENs
    Morbitzer, R. et al. (2010) “Regulation of selected genome loci using de novo-engineered
    transcription activator-like effector (TALE)-type transcription factors,” PROCEEDINGS OF THE
    NATIONAL ACADEMY OF SCIENCES 107 (50): 21617-22. Bibcode: 2010PNAS..10721617M.
    doi: 10.1073/pnas.1013133107. PMC 3003021. PMID 21106758
    Boch J. et al. (2009) “Breaking the code of DNA binding specificity of TAL-type III
    effectors,” SCIENCE 326 (5959): 1509-12. Bibcode: 2009Sci . . . 326.1509B.
    doi: 10.1126/science.1178811
    Li, T. et al. (2011) “Modularly assembled designer TAL effector nucleases for targeted gene
    knockout and gene replacement in eukaryotes,” NUCLEIC ACIDS RESEARCH 39: 6315-25.
    doi: 10.1093/nar/gkr188
    Mahfouz, M. M. et al. (2011) “De novo-engineered transcription activator-like effector
    (TALE) hybrid nuclease with novel DNA binding specificity creates double-strand breaks,”
    PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES 108 (6): 2623-8.
    doi: 10.1073/pnas.1019533108
    Cermak, T. et al. (2011) “Efficient design and assembly of custom TALEN and other TAL
    effector-based constructs for DNA targeting,” NUCLEIC ACIDS RESEARCH 39 (12): e82.
    doi: 10.1093/nar/gkr218. PMC 3130291
    Huang, P. et al. (2011) “Heritable gene targeting in zebrafish using customized TALENs,”
    NATURE BIOTECHNOLOGY 29 (8): 699-700. doi: 10.1038/nbt.1939
    Sander, J. D. et al. (2011) “Targeted gene disruption in somatic zebrafish cells using
    engineered TALENs,” NATURE BIOTECHNOLOGY 29 (8): 697-8. doi: 10.1038/nbt.1934
    Tesson, L. et al. (2011) “Knockout rats generated by embryo microinjection of TALENs,”
    NATURE BIOTECHNOLOGY 29 (8): 695-6. doi: 10.1038/nbt.1940
  • V.2 Template Binding Domains that are Protein-Binding Polypeptides
  • In some embodiments, the template binding domain is a protein, and the template binding domain partner is a protein, and the template binding domain and the template binding domain partner have affinity for each other. Generally, when the template binding domain is a protein, it lacks substantial affinity for other proteins present in the cell to be altered. This helps to avoid nonspecific binding. In some embodiments, the template-binding domain is derived from a protein in another species than the species of the cell to be altered. In some embodiments, the template-binding domain is derived from a protein that has no binding partners that are expressed in the cell type to be altered.
  • In some embodiments, the protein-binding polypeptide comprises one of more of the following domains: SH2, SH3, PTB, 14-3-3, FHA, WW, WD40, bromo, chromo, EVH1, PDZ, DD, DED, CARD, BH1-4, CSD, F-box, Hect, RING, ANK, ARM, LIM, EF-hand, MH2.
  • In some embodiments, the template-binding domain comprises an antibody with affinity for the template binding domain partner. Conversely, in some embodiments, the template-binding domain is a protein and the template binding domain partner is an antibody with affinity for the template binding domain. The antibody may be, e.g., an scFv or any antibody having sufficient CDR sequences to bind its target.
  • In some embodiments, the template-binding domain carries one or more deletions relative to the wild-type protein from which it was derived. For example, there may be a deletion of a catalytic domain. In some embodiments, the wild-type protein has multiple protein-binding domains, one or more of these domains, e.g., all but one of these domains, is deleted.
  • Exemplary protein-binding domains are given in the table below. The template-binding domain may comprise one or more of these protein-binding domains or polypeptides derived therefrom. It is understood that in some embodiments, the template binding domain is, or is derived from, the protein in the left column and the template binding domain partner is, or is derived from, the protein in the right column. In other embodiments, the template binding domain is, or is derived from, the protein in the right column and the template binding domain partner is, or is derived from, the protein in the left column.
  • TABLE V.6
    Protein-protein interaction domains
    Protein or domain Binding partner
    TE33 Fab L chain (BBa_K126000 from the B subunit of cholera toxin
    Registry of Standard Biological Parts)
    protein ZSPA-1 (BBa_K103004 from the Staphylococcal protein A
    Registry of Standard Biological Parts)
    RGD (BBa_K133059 from the Registry of integrins
    Standard Biological Parts)
    Cdc4 (found in yeast; comprises F-box Sic1 CDK inhibitor; Skp1,
    domain) Rbx1
    Grr1 (found in yeast; comprises F-box Cyclin (CLN) 1, 2; Skp1,
    domain) Rbx1
    TrCp (found in yeast; comprises F-box IkB(NFkB regulator); Skp1,
    domain) Rbx1
  • V.3 Template Binding Domains that are Small Molecule-Binding Polypeptides
  • In some embodiments, the template binding domain is a protein, and the template binding domain partner is a small molecule. Generally, when the template binding domain has affinity for a small molecule, the small molecule is rare or absent in the cell being altered. This helps to avoid nonspecific binding.
  • In some embodiments, the template-binding domain carries one or more deletions or substitutions relative to the wild-type protein from which it was derived. For example, there may be a deletion of or substitution within a catalytic domain, a DNA-binding domain, a protein-protein interaction domain, and/or a domain necessary for transcriptional regulation.
  • Exemplary small molecule-binding domains are given in the table below. The template-binding domain may comprise one or more of these small molecule-binding domains or polypeptides derived therefrom.
  • TABLE V.7
    Proteins that bind small molecules
    Protein Small molecule
    Avidin or Streptavidin (BBa_K283010 from biotin
    the Registry of Standard Biological Parts)
    gyrEC (BBa_K133070 from the Registry of coumermycin
    Standard Biological Parts)
    RI7 (BBa_K211001 from the Registry of octanal, heptanal or hexanal
    Standard Biological Parts)
    VirA receptor (BBa_K389001 from the acetosyringone
    Registry of Standard Biological Parts)
    Penicillin-binding proteins (PBPs), e.g., penicillin or cephalosporin
    serine type D-alanyl-D-alanine
    carboxypeptidase/transpeptidase
    TetR tetracycline
    ASGPR N-Acetylgalactosamine
    or galactose
  • VI. Template Binding Domain Partner
  • VI.1 Template Binding Domain Partners that are Nucleic Acids
  • Double Stranded DNA
  • In some embodiments, the template binding domain partner is double-stranded DNA. For instance, in some embodiments, the template binding domain partner is double-stranded DNA that is recognized by a DNA-binding protein described above in Section V.1.
  • The template binding domain partner may be, e.g., identical to or derived from a DNA sequence that is bound by a protein in a wild-type context. In some embodiments, the template binding domain partner comprises all or part of a transcription factor binding site from an organism other than the organism of the cell being altered. In some embodiments, the template binding domain partner comprises all or part of a transcriptional regulation site from an operon, e.g., a bacterial operon.
  • In some embodiments, the template binding domain partner is at least 10 nucleotides long, e.g., at least 15, 20, 25, 30, 35, 40, 45, 50, 75, 100, 150, or 200 nucleotides long. In some embodiments, the template binding domain partner is at most 200 nucleotides long, e.g., at most 15, 20, 25, 30, 35, 40, 45, 50, 75, 100, 150, or 200 nucleotides long. In some embodiments, the template binding domain partner is 10-20, 20-30, 30-40, 40-50, 50-75, 75-100, 100-150, or 150-200 nucleotides long.
  • In some embodiments, the template binding domain partner comprises palindromic sequences.
  • In some embodiments, the template binding domain partner comprises a plurality of shorter sequences, wherein each shorter sequence is bound by a distinct DNA-binding domain. In some embodiments, the plurality of shorter sequences are identical, e.g., the template binding domain partner comprises repeats. In other embodiments, one or more of, e.g., all of the plurality of shorter sequences are not identical to each other.
  • In some embodiments, the template binding domain partner is chemically modified DNA, e.g., as set out in Section XI below. The modification may be, e.g., to one or more bases and/or to the backbone. The chemical modification may do one or more of the following: improve the stability of the DNA, reduce the innate immune response against the DNA, and improve the binding of the template binding domain to the template binding domain partner.
  • The template binding domain partner need not always be the same type of molecule as the template nucleic acid. For instance, in some embodiments, the template binding domain partner is double stranded, while the template nucleic acid is single stranded. In some such embodiments, a long single-stranded DNA comprises a hairpin at one end, and the double stranded region of the hairpin comprises the template binding domain partner. In other embodiments, the template binding domain partner and the template nucleic acid are both double stranded. In some embodiments, the template binding domain partner is derived from a wild-type template binding domain partner. For example, the template binding domain partner may be a fragment of a naturally occurring nucleic acid, a mutagenized nucleic acid, a synthetic nucleic acid modeled after a naturally-ocurring nucleic acid. In some embodiments, the template binding domain partner is mutagenized to increase its affinity for a template binding domain. In some embodiments, the template binding domain partner is mutagenized to decrease its affinity for a template binding domain.
  • Single Stranded DNA
  • In some embodiments, the template binding domain partner is single-stranded DNA.
  • VI.2 Template Binding Domain Partners that are Polypeptides
  • In some embodiments, the template binding domain partner is a polypeptide. Any of the proteins and/or polypeptides discussed above in Sections V, may be used in the present invention, as a template binding domain partner. For example, protein-protein interacting pairs are discussed above, e.g., in Table V.6.
  • VII. Functional Analysis of Candidate Molecules
  • Candidate Cas9 molecules, candidate Cas9 fusion molecules, candidate gRNA molecules, and candidate Cas9 fusion molecule/gRNA molecule complexes, can be evaluated by art-known methods or as described herein. For example, exemplary methods for evaluating the endonuclease activity of Cas9 fusion molecule are described, e.g., in Jinek et al. (2012) SCIENCE 337(6096): 816-821.
  • The methods in this section may be used, e.g., to test various portions of a gRNA, for example, the targeting domain, the first complementarity domain, the linking domain, the second complementarity domain, the proximal domain, or the tail domain. In some embodiments, the methods in this section are tested to determine whether modifications made in one or more of these domains interfere with targeting efficacy. A gRNA with a selected length, sequence, degree of complementarity, or degree of modification, can be evaluated in a system of this section.
  • Binding and Cleavage Assay: Testing the Endonuclease Activity of Cas9 Fusion Molecule
  • The ability of a Cas9 fusion molecule/gRNA molecule complex to bind to and cleave a target nucleic acid can be evaluated in a plasmid cleavage assay. In this assay, synthetic or in vitro-transcribed gRNA molecule is pre-annealed prior to the reaction by heating to 95° C. and slowly cooling down to room temperature. Native or restriction digest-linearized plasmid DNA (300 ng (˜8 nM)) is incubated for 60 min at 37° C. with purified Cas9 protein molecule (50-500 nM) and gRNA (50-500 nM, 1:1) in a Cas9 plasmid cleavage buffer (20 mM HEPES pH 7.5, 150 mM KCl, 0.5 mM DTT, 0.1 mM EDTA) with or without 10 mM MgCl2. The reactions are stopped with 5×DNA loading buffer (30% glycerol, 1.2% SDS, 250 mM EDTA), resolved by a 0.8 or 1% agarose gel electrophoresis and visualized by ethidium bromide staining. The resulting cleavage products indicate whether the Cas9 molecule cleaves both DNA strands, or only one of the two strands. For example, linear DNA products indicate the cleavage of both DNA strands. Nicked open circular products indicate that only one of the two strands is cleaved.
  • Alternatively, the ability of a Cas9 fusion molecule/gRNA molecule complex to bind to and cleave a target nucleic acid can be evaluated in an oligonucleotide DNA cleavage assay. In this assay, DNA oligonucleotides (10 pmol) are radiolabeled by incubating with 5 units T4 polynucleotide kinase and ˜3-6 pmol (˜20-40 mCi) [γ-32P]-ATP in 1× T4 polynucleotide kinase reaction buffer at 37° C. for 30 min., in a 50 μL reaction. After heat inactivation (65° C. for 20 min), reactions are purified through a column to remove unincorporated label. Duplex substrates (100 nM) are generated by annealing labeled oligonucleotides with equimolar amounts of unlabeled complementary oligonucleotide at 95° C. for 3 min., followed by slow cooling to room temperature. For cleavage assays, gRNA molecules are annealed by heating to 95° C. for 30 s, followed by slow cooling to room temperature. Cas9 fusion molecule (500 nM final concentration) is pre-incubated with the annealed gRNA molecules (500 nM) in cleavage assay buffer (20 mM HEPES pH 7.5, 100 mM KCl, 5 mM MgCl2, 1 mM DTT, 5% glycerol) in a total volume of 9 μl. Reactions are initiated by the addition of 1 μl target DNA (10 nM) and incubated for 1 h at 37° C. Reactions are quenched by the addition of 20 μl of loading dye (5 mM EDTA, 0.025% SDS, 5% glycerol in formamide) and heated to 95° C. for 5 min. Cleavage products are resolved on 12% denaturing polyacrylamide gels containing 7 M urea and visualized by phosphorimaging. The resulting cleavage products indicate that whether the complementary strand, the non-complementary strand, or both, are cleaved.
  • One or both of these assays can be used to evaluate the suitability of a candidate gRNA molecule, a candidate Cas9 molecule, or a candidate Cas9 fusion molecule.
  • Binding Assay: Testing the Binding of Cas9 Fusion Molecule to Target DNA
  • Exemplary methods for evaluating the binding of a Cas9 fusion molecule to target DNA are described, e.g., in Jinek et al. (2012) SCIENCE 337(6096):816-821.
  • For example, in an electrophoretic mobility shift assay, target DNA duplexes are formed by mixing of each strand (10 nmol) in deionized water, heating to 95° C. for 3 min. and slow cooling to room temperature. All DNAs are purified on 8% native gels containing 1×TBE. DNA bands are visualized by UV shadowing, excised, and eluted by soaking gel pieces in DEPC-treated H2O. Eluted DNA is ethanol precipitated and dissolved in DEPC-treated H2O. DNA samples are 5′ end labeled with [γ-32P]-ATP using T4 polynucleotide kinase for 30 min. at 37° C. Polynucleotide kinase is heat denatured at 65° C. for 20 min., and unincorporated radiolabel is removed using a column. Binding assays are performed in buffer containing 20 mM HEPES pH 7.5, 100 mM KCl, 5 mM MgCl2, 1 mM DTT and 10% glycerol in a total volume of 10 μl. Cas9 fusion molecule is programmed with equimolar amounts of pre-annealed gRNA molecule and titrated from 100 pM to 1 μM. Radiolabeled DNA is added to a final concentration of 20 pM. Samples are incubated for 1 h at 37° C. and resolved at 4° C. on an 8% native polyacrylamide gel containing 1×TBE and 5 mM MgCl2. Gels are dried and DNA visualized by phosphorimaging.
  • Differential Scanning Flourimetry (DSF)
  • The thermostability of Cas9 molecule-gRNA ribonucleoprotein (RNP) complexes, e.g., a Cas9 fusion molecule-gRNA RNP complex, can be measured via DSF. This technique measures the thermostability of a protein, which can increase under favorable conditions such as the addition of a binding RNA molecule, e.g., a gRNA.
  • The assay is performed using two different protocols, one to test the best stoichiometric ratio of gRNA:Cas9 protein and another to determine the best solution conditions for RNP formation.
  • To determine the best solution to form RNP complexes, a 2 uM solution of Cas9 in water+10× SYPRO Orange® (Life Technologies cat#S-6650) and dispensed into a 384 well plate. An equimolar amount of gRNA diluted in solutions with varied pH and salt is then added. After incubating at room temperature for 10 min. and brief centrifugation to remove any bubbles, a Bio-Rad CFX384™ Real-Time System C1000 Touch™ Thermal Cycler with the Bio-Rad CFX Manager software is used to run a gradient from 20° C. to 90° C. with a 1° C. increase in temperature every 10 seconds.
  • The second assay consists of mixing various concentrations of gRNA with 2 uM Cas9 molecule in optimal buffer from the assay above and incubating at RT for 10 min in a 384 well plate. An equal volume of optimal buffer+10× SYPRO Orange® (Life Techonologies cat#S-6650) is added and the plate sealed with Microseal® B adhesive (MSB-1001). Following brief centrifugation to remove any bubbles, a Bio-Rad CFX384™ Real-Time System C1000 Touch™ Thermal Cycler with the Bio-Rad CFX Manager software is used to run a gradient from 20° C. to 90° C. with a 10 increase in temperature every 10 seconds.
  • VIII. Genome Editing Approaches
  • Mutations may be corrected, and undesirable nucleic acid sequences may be cleaved, using one of the approaches discussed herein. In an embodiment, a mutation in a target nucleic acid is corrected by homology directed repair (HDR) using a template nucleic acid (see Section VIII.1).
  • VIII.1 HDR Repair and Template Nucleic Acids
  • As described herein, nuclease-induced homology directed repair (HDR) can be used to alter a target sequence and correct (e.g., repair or edit) a mutation in the genome. While not wishing to be bound by theory, it is believed that alteration of the target sequence occurs by homology-directed repair (HDR) with a donor template or template nucleic acid. For example, the donor template or the template nucleic acid provides for alteration of the target position. It is contemplated that a plasmid donor can be used as a template for homologous recombination. It is further contemplated that a single stranded donor template can be used as a template for alteration of the target position by alternate methods of homology directed repair (e.g., single strand annealing) between the target position and the donor template.
  • Donor template-effected alteration of a target position depends on cleavage by a Cas9 molecule. Cleavage by Cas9 can comprise a nick, a double strand break, or two single strand breaks, e.g., one on each strand of the target nucleic acid. In an embodiment, a mutation can be corrected by either a single double-strand break or two single strand breaks. In an embodiment, a mutation can be corrected by (1) a single double-strand break, (2) two single strand breaks, (3) two double stranded breaks with a break occurring on each side of the target position, (4) one double stranded break and two single strand breaks with the double strand break and two single strand breaks occurring on each side of the target position or (5) four single stranded breaks with a pair of single stranded breaks occurring on each side of the target position.
  • Additional details on template nucleic acids are provided in Section IV entitled “Template Nucleic Acids” in PCT Application WO 2015/048577, the entire contents of which are expressly incorporated herein by reference.
  • Double Strand Break Mediated Correction
  • In an embodiment, double strand cleavage is effected by a Cas9 molecule having cleavage activity associated with an HNH-like domain and cleavage activity associated with a RuvC-like domain, e.g., an N-terminal RuvC-like domain, e.g., a wild type Cas9 molecule. Such embodiments require only a single gRNA.
  • Single Strand Break Mediated Correction
  • In other embodiments, two single strand breaks, or nicks, are effected by a Cas9 molecule having nickase activity, e.g., cleavage activity associated with an HNH-like domain or cleavage activity associated with an N-terminal RuvC-like domain. Such embodiments usually require two gRNAs, one for placement of each single strand break. In an embodiment, the Cas9 molecule having nickase activity cleaves the strand to which the gRNA hybridizes, but not the strand that is complementary to the strand to which the gRNA hybridizes. In an embodiment, the Cas9 molecule having nickase activity does not cleave the strand to which the gRNA hybridizes, but rather cleaves the strand that is complementary to the strand to which the gRNA hybridizes.
  • In an embodiment, the nickase has HNH activity, e.g., a Cas9 molecule having the RuvC activity inactivated, e.g., a Cas9 molecule having a mutation at D10, e.g., the D10A mutation. D10A inactivates RuvC; therefore, the Cas9 nickase has (only) HNH activity and will cut on the strand to which the gRNA hybridizes (e.g., the complementary strand, which does not have the NGG PAM on it). In other embodiments, a Cas9 molecule having an H840, e.g., an H840A, mutation can be used as a nickase. H840A inactivates HNH; therefore, the Cas9 nickase has (only) RuvC activity and cuts on the non-complementary strand (e.g., the strand that has the NGG PAM and whose sequence is identical to the gRNA). In other embodiments, a Cas9 molecule having an N863, e.g., the N863A mutation, mutation can be used as a nickase. N863A inactivates HNH therefore the Cas9 nickase has (only) RuvC activity and cuts on the non-complementary strand (the strand that has the NGG PAM and whose sequence is identical to the gRNA). In other embodiments, a Cas9 molecule having an N580, e.g., the N580A mutation, mutation can be used as a nickase. N580A inactivates HNH therefore the Cas9 nickase has (only) RuvC activity and cuts on the non-complementary strand (the strand that has the NGG PAM and whose sequence is identical to the gRNA). In an embodiment, in which a nickase and two gRNAs are used to position two single strand nicks, one nick is on the + strand and one nick is on the − strand of the target nucleic acid. The PAMs can be outwardly facing. The gRNAs can be selected such that the gRNAs are separated by, from about 0-50, 0-100, or 0-200 nucleotides. In an embodiment, there is no overlap between the target sequences that are complementary to the targeting domains of the two gRNAs. In an embodiment, the gRNAs do not overlap and are separated by as much as 50, 100, or 200 nucleotides. In an embodiment, the use of two gRNAs can increase specificity, e.g., by decreasing off-target binding (Ran et al. 2013, CELL 154:1380-1389).
  • In an embodiment, a single nick can be used to induce HDR. It is contemplated herein that a single nick can be used to increase the ratio of HR to NHEJ at a given cleavage site. In an embodiment, a single strand break is formed in the strand of the target nucleic acid to which the targeting domain of said gRNA is complementary. In another embodiment, a single strand break is formed in the strand of the target nucleic acid other than the strand to which the targeting domain of said gRNA is complementary.
  • Placement of the Double Strand or Single Strand Breaks Relative to the Target Position
  • The double strand break or single strand break in one of the strands should be sufficiently close to target position such that an alteration is produced in the desired region, e.g., correction of a mutation occurs. In an embodiment, the distance is not more than 50, 100, 200, 300, 350 or 400 nucleotides. While not wishing to be bound by theory, in some embodiments, it is believed that the break should be sufficiently close to target position such that the target position is within the region that is subject to exonuclease-mediated removal during end resection. If the distance between the target position and a break is too great, the mutation or other sequence desired to be altered may not be included in the end resection and, therefore, may not be corrected, as donor sequence, either exogenously provided donor sequence or endogenous genomic donor sequence, in some embodiments is only used to correct sequence within the end resection region.
  • In an embodiment, the targeting domain is configured such that a cleavage event, e.g., a double strand or single strand break, is positioned within 1, 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 150 or 200 nucleotides of the region desired to be altered, e.g., a mutation. The break, e.g., a double strand or single strand break, can be positioned upstream or downstream of the region desired to be altered, e.g., a mutation. In some embodiments, a break is positioned within the region desired to be altered, e.g., within a region defined by at least two mutant nucleotides. In some embodiments, a break is positioned immediately adjacent to the region desired to be altered, e.g., immediately upstream or downstream of a mutation.
  • In an embodiment, a single strand break is accompanied by an additional single strand break, positioned by a second gRNA molecule, as discussed below. For example, the targeting domains bind configured such that a cleavage event, e.g., the two single strand breaks, are positioned within 1, 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 150 or 200 nucleotides of a target position. In an embodiment, the first and second gRNA molecules are configured such, that when guiding a Cas9 nickase, a single strand break will be accompanied by an additional single strand break, positioned by a second gRNA, sufficiently close to one another to result in alteration of the desired region. In an embodiment, the first and second gRNA molecules are configured such that a single strand break positioned by said second gRNA is within 10, 20, 30, 40, or 50 nucleotides of the break positioned by said first gRNA molecule, e.g., when the Cas9 is a nickase. In an embodiment, the two gRNA molecules are configured to position cuts at the same position, or within a few nucleotides of one another, on different strands, e.g., essentially mimicking a double strand break.
  • In an embodiment, in which a gRNA (unimolecular (or chimeric) or modular gRNA) and Cas9 fusion molecule induce a double strand break for the purpose of inducing HDR-mediated correction, the cleavage site is between 0-200 bp (e.g., 0-175, 0 to 150, 0 to 125, 0 to 100, 0 to 75, 0 to 50, 0 to 25, 25 to 200, 25 to 175, 25 to 150, 25 to 125, 25 to 100, 25 to 75, 25 to 50, 50 to 200, 50 to 175, 50 to 150, 50 to 125, 50 to 100, 50 to 75, 75 to 200, 75 to 175, 75 to 150, 75 to 125, 75 to 100 bp) away from the target position. In an embodiment, the cleavage site is between 0-100 bp (e.g., 0 to 75, 0 to 50, 0 to 25, 25 to 100, 25 to 75, 25 to 50, 50 to 100, 50 to 75 or 75 to 100 bp) away from the target position.
  • In an embodiment, the targeting domain of a gRNA molecule is configured to position a cleavage event sufficiently far from a preselected nucleotide, e.g., the nucleotide of a coding region, such that the nucleotide is not altered. In an embodiment, the targeting domain of a gRNA molecule is configured to position an intronic cleavage event sufficiently far from an intron/exon border, or naturally occurring splice signal, to avoid alteration of the exonic sequence or unwanted splicing events. The gRNA molecule may be a first, second, third and/or fourth gRNA molecule, as described herein.
  • Placement of a First Break and a Second Break Relative to Each Other
  • In an embodiment, a double strand break can be accompanied by an additional double strand break, positioned by a second gRNA molecule, as is discussed below.
  • In an embodiment, a double strand break can be accompanied by two additional single strand breaks, positioned by a second gRNA molecule and a third gRNA molecule.
  • In an embodiment, a first and second single strand breaks can be accompanied by two additional single strand breaks positioned by a third gRNA molecule and a fourth gRNA molecule.
  • When two or more gRNAs are used to position two or more cleavage events, e.g., double strand or single strand breaks, in a target nucleic acid, it is contemplated that the two or more cleavage events may be made by the same or different Cas9 molecules. For example, when two gRNAs are used to position two double stranded breaks, a single Cas9 molecule may be used to create both double stranded breaks. When two or more gRNAs are used to position two or more single stranded breaks (nicks), a single Cas9 nickase may be used to create the two or more nicks. When two or more gRNAs are used to position at least one double stranded break and at least one single stranded break, two Cas9 proteins may be used, e.g., one Cas9 nuclease and one Cas9 nickase. It is contemplated that when two or more Cas9 proteins are used that the two or more Cas9 proteins may be delivered sequentially to control specificity of a double stranded versus a single stranded break at the desired position in the target nucleic acid.
  • In some embodiments, the targeting domain of the first gRNA molecule and the targeting domain of the second gRNA molecules are complementary to opposite strands of the target nucleic acid molecule. In some embodiments, the gRNA molecule and the second gRNA molecule are configured such that the PAMs are oriented outward.
  • In an embodiment, in which two gRNAs (independently, unimolecular (or chimeric) or modular gRNA) complexing with Cas9 nickases induce two single strand breaks for the purpose of inducing HDR-mediated correction, the closer nick is between 0-200 bp (e.g., 0-175, 0 to 150, 0 to 125, 0 to 100, 0 to 75, 0 to 50, 0 to 25, 25 to 200, 25 to 175, 25 to 150, 25 to 125, 25 to 100, 25 to 75, 25 to 50, 50 to 200, 50 to 175, 50 to 150, 50 to 125, 50 to 100, 50 to 75, 75 to 200, 75 to 175, 75 to 150, 75 to 125, 75 to 100 bp) away from the target position and the two nicks will ideally be within 25-65 bp of each other (e.g., 25 to 50, 25 to 45, 25 to 40, 25 to 35, 25 to 30, 30 to 55, 30 to 50, 30 to 45, 30 to 40, 30 to 35, 35 to 55, 35 to 50, 35 to 45, 35 to 40, 40 to 55, 40 to 50, 40 to 45 bp, 45 to 50 bp, 50 to 55 bp, 55 to 60 bp, 60 to 65 bp) and no more than 100 bp away from each other (e.g., no more than 90, 80, 70, 60, 50, 40, 30, 20, 10 or 5 bp away from each other). In an embodiment, the cleavage site is between 0-100 bp (e.g., 0 to 75, 0 to 50, 0 to 25, 25 to 100, 25 to 75, 25 to 50, 50 to 100, 50 to 75 or 75 to 100 bp) away from the target position.
  • In one embodiment, two gRNAs, e.g., independently, unimolecular (or chimeric) or modular gRNA, are configured to position a double-strand break on both sides of a target position. In an alternate embodiment, three gRNAs, e.g., independently, unimolecular (or chimeric) or modular gRNA, are configured to position a double strand break (i.e., one gRNA complexes with a Cas9 nuclease) and two single strand breaks or paired single stranded breaks (i.e., two gRNAs complex with Cas9 nickases) on either side of the target position. In another embodiment, four gRNAs, e.g., independently, unimolecular (or chimeric) or modular gRNA, are configured to generate two pairs of single stranded breaks (i.e., two pairs of two gRNAs complex with Cas9 nickases) on either side of the target position. The double strand break(s) or the closer of the two single strand nicks in a pair will ideally be within 0-500 bp of the target position (e.g., no more than 450, 400, 350, 300, 250, 200, 150, 100, 50 or 25 bp from the target position). When nickases are used, the two nicks in a pair are, in some embodiments, within 25-65 bp of each other (e.g., between 25 to 55, 25 to 45, 25 to 40, 25 to 35, 25 to 30, 50 to 55, 45 to 55, 40 to 55, 35 to 55, 30 to 55, 30 to 50, 35 to 50, 40 to 50, 45 to 50, 35 to 45, 40 to 45 bp, 45 to 50 bp, 50 to 55 bp, 55 to 60 bp, or 60 to 65 bp) and no more than 100 bp away from each other (e.g., no more than 90, 80, 70, 60, 50, 40, 30, 20 or 10 bp).
  • When two gRNAs are used to target Cas9 molecules to breaks, different combinations of Cas9 molecules are envisioned. In some embodiments, a first gRNA is used to target a first Cas9 molecule to a first target position, and a second gRNA is used to target a second Cas9 molecule to a second target position. In some embodiments, the first Cas9 molecule creates a nick on the first strand of the target nucleic acid, and the second Cas9 molecule creates a nick on the opposite strand, resulting in a double stranded break (e.g., a blunt ended cut or a cut with overhangs).
  • Different combinations of nickases can be chosen to target one single stranded break to one strand and a second single stranded break to the opposite strand. When choosing a combination, one can take into account that there are nickases having one active RuvC-like domain, and nickases having one active HNH domain. In an embodiment, a RuvC-like domain cleaves the non-complementary strand of the target nucleic acid molecule. In an embodiment, an HNH-like domain cleaves a single stranded complementary domain, e.g., a complementary strand of a double stranded nucleic acid molecule. Generally, if both Cas9 molecules have the same active domain (e.g., both have an active RuvC domain or both have an active HNH domain), one will choose two gRNAs that bind to opposite strands of the target. In more detail, in some embodiments, a first gRNA is complementary with a first strand of the target nucleic acid and binds a nickase having an active RuvC-like domain and causes that nickase to cleave the strand that is non-complementary to that first gRNA, i.e., a second strand of the target nucleic acid; and a second gRNA is complementary with a second strand of the target nucleic acid and binds a nickase having an active RuvC-like domain and causes that nickase to cleave the strand that is non-complementary to that second gRNA, i.e., the first strand of the target nucleic acid. Conversely, in some embodiments, a first gRNA is complementary with a first strand of the target nucleic acid and binds a nickase having an active HNH domain and causes that nickase to cleave the strand that is complementary to that first gRNA, i.e., a first strand of the target nucleic acid; and a second gRNA is complementary with a second strand of the target nucleic acid and binds a nickase having an active HNH domain and causes that nickase to cleave the strand that is complementary to that second gRNA, i.e., the second strand of the target nucleic acid. In another arrangement, if one Cas9 molecule has an active RuvC-like domain and the other Cas9 molecule has an active HNH domain, the gRNAs for both Cas9 molecules can be complementary to the same strand of the target nucleic acid, so that the Cas9 molecule with the active RuvC-like domain will cleave the non-complementary strand and the Cas9 molecule with the HNH domain will cleave the complementary strand, resulting in a double stranded break.
  • Exemplary Template Nucleic Acids
  • A template nucleic acid, as that term is used herein, refers to a nucleic acid sequence which can be used in conjunction with a Cas9 fusion molecule and a gRNA molecule to alter the structure of a target position.
  • Target position, as used herein, refers to a site on a target nucleic acid (e.g., the chromosome) that is modified by a Cas9 molecule-dependent process, e.g., a Cas9 fusion molecule-dependent process. For example, the target position can be a modified Cas9 fusion molecule cleavage of the target nucleic acid and template nucleic acid directed modification, e.g., correction, of the target position. In an embodiment, a target position can be a site between two nucleotides, e.g., adjacent nucleotides, on the target nucleic acid into which one or more nucleotides are added. The target position may comprise one or more nucleotides that are altered, e.g., corrected, by a template nucleic acid. In an embodiment, the target position is within a target sequence (e.g., the sequence to which the gRNA binds). In an embodiment, a target position is upstream or downstream of a target sequence (e.g., the sequence to which the gRNA binds).
  • In an embodiment, the target nucleic acid is modified to have the some or all of the sequence of the template nucleic acid, typically at or near cleavage site(s). In an embodiment, the template nucleic acid is single stranded. In an alternate embodiment, the template nucleic acid is double stranded. In an embodiment, the template nucleic acid is DNA, e.g., double stranded DNA. In an alternate embodiment, the template nucleic acid is single stranded DNA. In an embodiment, the template nucleic acid is encoded on the same vector backbone, e.g., AAV genome or plasmid DNA, as the Cas9 fusion molecule and gRNA. In an embodiment, the template nucleic acid is excised from a vector backbone in vivo, e.g., it is flanked by gRNA recognition sequences. In an embodiment, the template nucleic acid comprises endogenous genomic sequence.
  • In an embodiment, the template nucleic acid alters the structure of the target position by participating in a homology directed repair event. In an embodiment, the template nucleic acid alters the sequence of the target position. In an embodiment, the template nucleic acid results in the incorporation of a modified, or non-naturally occurring base into the target nucleic acid.
  • Typically, the template sequence undergoes a breakage mediated or catalyzed recombination with the target sequence. In an embodiment, the template nucleic acid includes sequence that corresponds to a site on the target sequence that is cleaved by an eaCas9 mediated cleavage event. In an embodiment, the template nucleic acid includes sequence that corresponds to both, a first site on the target sequence that is cleaved in a first Cas9 mediated event, and a second site on the target sequence that is cleaved in a second Cas9 mediated event.
  • In an embodiment, the template nucleic acid can include sequence which results in an alteration in the coding sequence of a translated sequence, e.g., one which results in the substitution of one amino acid for another in a protein product, e.g., transforming a mutant allele into a wild type allele, transforming a wild type allele into a mutant allele, and/or introducing a stop codon, insertion of an amino acid residue, deletion of an amino acid residue, or a nonsense mutation.
  • In other embodiments, the template nucleic acid can include sequence which results in an alteration in a non-coding sequence, e.g., an alteration in an exon or in a 5′ or 3′ non-translated or non-transcribed region. Such alterations include an alteration in a control element, e.g., a promoter, enhancer, and an alteration in a cis-acting or trans-acting control element.
  • A template nucleic acid having homology with a target position in a gene, e.g., a gene described herein, can be used to alter the structure of a target sequence. The template sequence can be used to alter an unwanted structure, e.g., an unwanted or mutant nucleotide.
  • In an embodiment, the template nucleic acid is a single stranded nucleic acid. In another embodiment, the template nucleic acid is a double stranded nucleic acid. In some embodiments, the template nucleic acid comprises a nucleotide sequence, e.g., of one or more nucleotides, that will be added to or will template a change in the target nucleic acid. In other embodiments, the template nucleic acid comprises a nucleotide sequence that may be used to modify the target position. In other embodiments, the template nucleic acid comprises a nucleotide sequence, e.g., of one or more nucleotides, that corresponds to wild type sequence of the target nucleic acid, e.g., of the target position.
  • The template nucleic acid may comprise a replacement sequence. In some embodiments, the template nucleic acid comprises a 5′ homology arm. In other embodiments, the template nucleic acid comprises a 3′ homology arm.
  • In some embodiments, the template nucleic acid is linear double stranded DNA. The length may be, e.g., about 50-500 base pairs. The length may be, e.g., about 150-200 base pairs, e.g., about 150, 160, 170, 180, 190, or 200 base pairs. The length may be, e.g., at least 150, 160, 170, 180, 190, or 200 base pairs. In some embodiments, the length is no greater than 150, 160, 170, 180, 190, or 200 base pairs. In some embodiments, a double stranded template nucleic acid has a length of about 160 base pairs, e.g., about 155-165, 150-170, 140-180, 130-190, 120-200, 110-210, 100-220, 90-230, or 80-240 base pairs.
  • The template nucleic acid can be linear single stranded DNA. In some embodiments, the template nucleic acid is (i) linear single stranded DNA that can anneal to the nicked strand of the target nucleic acid, (ii) linear single stranded DNA that can anneal to the intact strand of the target nucleic acid, (iii) linear single stranded DNA that can anneal to the transcribed strand of the target nucleic acid, (iv) linear single stranded DNA that can anneal to the non-transcribed strand of the target nucleic acid, or more than one of the preceding. The length may be, e.g., 50-500 nucleotides. The length may be, e.g., about 50-500 nucleotides. The length may be, e.g., about 150-200 nucleotides, e.g., about 150, 160, 170, 180, 190, or 200 nucleotides. The length may be, e.g., at least 150, 160, 170, 180, 190, or 200 nucleotides. In some embodiments, the length is no greater than 150, 160, 170, 180, 190, or 200 nucleotides. In some embodiments, a single stranded template nucleic acid has a length of about 160 nucleotides, e.g., about 155-165, 150-170, 140-180, 130-190, 120-200, 110-210, 100-220, 90-230, or 80-240 nucleotides.
  • In some embodiments, the template nucleic acid is circular double stranded DNA, e.g., a plasmid. In some embodiments, the template nucleic acid comprises about 500 to 1000 base pairs of homology on either side of the replacement sequence and/or the nick. In some embodiments, the template nucleic acid comprises about 300, 400, 500, 600, 700, 800, 900, 1000, 1500, or 2000 base pairs of homology 5′ of the nick or replacement sequence, 3′ of the nick or replacement sequence, or both 5′ and 3′ of the nick or replacement sequence. In some embodiments, the template nucleic acid comprises at least 300, 400, 500, 600, 700, 800, 900, 1000, 1500, or 2000 base pairs of homology 5′ of the nick or replacement sequence, 3′ of the nick or replacement sequence, or both 5′ and 3′ of the nick or replacement sequence. In some embodiments, the template nucleic acid comprises no more than 300, 400, 500, 600, 700, 800, 900, 1000, 1500, or 2000 base pairs of homology 5′ of the nick or replacement sequence, 3′ of the nick or replacement sequence, or both 5′ and 3′ of the nick or replacement sequence.
  • In some embodiments, the template nucleic acid is an adenovirus vector, e.g., an AAV vector, e.g., a ssDNA molecule of a length and sequence that allows it to be packaged in an AAV capsid. The vector may be, e.g., less than 5 kb and may contain an ITR sequence that promotes packaging into the capsid. The vector may be integration-deficient. In some embodiments, the template nucleic acid comprises about 150 to 1000 nucleotides of homology on either side of the replacement sequence and/or the nick. In some embodiments, the template nucleic acid comprises about 100, 150, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1500, or 2000 nucleotides 5′ of the nick or replacement sequence, 3′ of the nick or replacement sequence, or both 5′ and 3′ of the nick or replacement sequence. In some embodiments, the template nucleic acid comprises at least 100, 150, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1500, or 2000 nucleotides 5′ of the nick or replacement sequence, 3′ of the nick or replacement sequence, or both 5′ and 3′ of the nick or replacement sequence. In some embodiments, the template nucleic acid comprises at most 100, 150, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1500, or 2000 nucleotides 5′ of the nick or replacement sequence, 3′ of the nick or replacement sequence, or both 5′ and 3′ of the nick or replacement sequence.
  • In some embodiments, the template nucleic acid is a lentiviral vector, e.g., an IDLV (integration deficiency lentivirus). In some embodiments, the template nucleic acid comprises about 500 to 1000 base pairs of homology on either side of the replacement sequence and/or the nick. In some embodiments, the template nucleic acid comprises about 300, 400, 500, 600, 700, 800, 900, 1000, 1500, or 2000 base pairs of homology 5′ of the nick or replacement sequence, 3′ of the nick or replacement sequence, or both 5′ and 3′ of the nick or replacement sequence. In some embodiments, the template nucleic acid comprises at least 300, 400, 500, 600, 700, 800, 900, 1000, 1500, or 2000 base pairs of homology 5′ of the nick or replacement sequence, 3′ of the nick or replacement sequence, or both 5′ and 3′ of the nick or replacement sequence. In some embodiments, the template nucleic acid comprises no more than 300, 400, 500, 600, 700, 800, 900, 1000, 1500, or 2000 base pairs of homology 5′ of the nick or replacement sequence, 3′ of the nick or replacement sequence, or both 5′ and 3′ of the nick or replacement sequence.
  • In an embodiment, the template nucleic acid comprises one or more mutations, e.g., silent mutations, that prevent Cas9 from recognizing and cleaving the template nucleic acid. The template nucleic acid may comprise, e.g., at least 1, 2, 3, 4, 5, 10, 20, or 30 silent mutations relative to the corresponding sequence in the genome of the cell to be altered. In an embodiment, the template nucleic acid comprises at most 2, 3, 4, 5, 10, 20, 30, or 50 silent mutations relative to the corresponding sequence in the genome of the cell to be altered.
  • In an embodiment, the template nucleic acid alters the structure of the target position by participating in a homology directed repair event. In an embodiment, the template nucleic acid alters the sequence of the target position. In an embodiment, the template nucleic acid results in the incorporation of a modified, or non-naturally occurring base into the target nucleic acid.
  • Typically, the template sequence undergoes a breakage mediated or catalyzed recombination with the target sequence. In an embodiment, the template nucleic acid includes sequence that corresponds to a site on the target sequence that is cleaved by an eaCas9 mediated cleavage event. In an embodiment, the template nucleic acid includes sequence that corresponds to both, a first site on the target sequence that is cleaved in a first Cas9 mediated event, and a second site on the target sequence that is cleaved in a second Cas9 mediated event.
  • In an embodiment, the template nucleic acid can include sequence which results in an alteration in the coding sequence of a translated sequence, e.g., one which results in the substitution of one amino acid for another in a protein product, e.g., transforming a mutant allele into a wild type allele, transforming a wild type allele into a mutant allele, and/or introducing a stop codon, insertion of an amino acid residue, deletion of an amino acid residue, or a nonsense mutation. In an embodiment, the template nucleic acid can include sequence which results in an alteration in a coding sequence, e.g., an alteration in an exon.
  • In other embodiments, the template nucleic acid can include sequence which results in an alteration in a non-coding sequence, e.g., an alteration in an intron or in a 5′ or 3′ non-translated or non-transcribed region. Such alterations include an alteration in a control element, e.g., a promoter, enhancer, and an alteration in a cis-acting or trans-acting control element.
  • A template nucleic acid having homology with a target position can be used to alter the structure of a target sequence. The template sequence can be used to alter an unwanted structure, e.g., an unwanted or mutant nucleotide.
  • Length of the Homology Arms
  • The homology arm should extend at least as far as the region in which end resection may occur, e.g., in order to allow the resected single stranded overhang to find a complementary region within the donor template. The overall length could be limited by parameters such as plasmid size or viral packaging limits. In an embodiment, a homology arm does not extend into repeated elements, e.g., ALU elements or LINE elements.
  • Exemplary homology arm lengths include a least 50, 100, 250, 500, 750, 1000, 2000, 3000, 4000, or 5000 nucleotides. In some embodiments, the homology arm length is 50-100, 100-250, 250-500, 500-750, 750-1000, 1000-2000, 2000-3000, 3000-4000, or 4000-5000 nucleotides.
  • A template nucleic acid typically comprises the following components:
  • [5′ homology arm]-[replacement sequence]-[3′ homology arm].
  • The homology arms provide for recombination into the chromosome, thus replacing the undesired element, e.g., a mutation or signature, with the replacement sequence. In an embodiment, the homology arms flank the most distal cleavage sites.
  • In an embodiment, the 3′ end of the 5′ homology arm is the position next to the 5′ end of the replacement sequence. In an embodiment, the 5′ homology arm can extend at least 10, 20, 30, 40, 50, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1500, 2000, 3000, 4000, or 5000 nucleotides 5′ from the 5′ end of the replacement sequence.
  • In an embodiment, the 5′ end of the 3′ homology arm is the position next to the 3′ end of the replacement sequence. In an embodiment, the 3′ homology arm can extend at least 10, 20, 30, 40, 50, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1500, 2000, 3000, 4000, or 5000 nucleotides 3′ from the 3′ end of the replacement sequence.
  • In an embodiment, to correct a mutation, the homology arms, e.g., the 5′ and 3′ homology arms, may each comprise about 1000 base pairs (bp) of sequence flanking the most distal gRNAs (e.g., 1000 bp of sequence on either side of the mutation).
  • It is contemplated herein that one or both homology arms may be shortened to avoid including certain sequence repeat elements, e.g., Alu element or LINE elements. For example, a 5′ homology arm may be shortened to avoid a sequence repeat element. In other embodiments, a 3′ homology arm may be shortened to avoid a sequence repeat element. In some embodiments, both the 5′ and the 3′ homology arms may be shortened to avoid including certain sequence repeat elements.
  • It is contemplated herein that template nucleic acids for correcting a mutation may be designed for use as a single-stranded oligonucleotide, e.g., a single-stranded oligodeoxynucleotide (ssODN). When using a ssODN, 5′ and 3′ homology arms may range up to about 200 base pairs (bp) in length, e.g., at least 25, 50, 75, 100, 125, 150, 175, or 200 bp in length. Longer homology arms are also contemplated for ssODNs as improvements in oligonucleotide synthesis continue to be made. In some embodiments, a longer homology arm is made by a method other than chemical synthesis, e.g., by denaturing a long double stranded nucleic acid and purifying one of the strands, e.g., by affinity for a strand-specific sequence anchored to a solid substrate.
  • Exemplary Arrangements of Linear Nucleic Acid Template Systems
  • In an embodiment, the nucleic acid template system is double stranded. In an embodiment, the nucleic acid template system is single stranded. In an embodiment, the nucleic acid template system comprises a single stranded portion and a double stranded portion. For example, the template binding domain partner is double stranded and the template nucleic acid is single stranded.
  • In an embodiment, the template nucleic acid comprises about 50 to 500 base pairs. In an embodiment, the template nucleic acid comprises about 50 to 100, e.g., 55 to 95, 60 to 90, 65 to 85, or 70 to 80, base pairs, homology on either side of the nick and/or replacement sequence. In an embodiment, the template nucleic acid comprises about 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100 base pairs homology 5′ of the nick or replacement sequence, 3′ of the nick or replacement sequence, or both 5′ and 3′ of the nick or replacement sequences.
  • In an embodiment, the template nucleic acid comprises about 150 to 200, e.g., 155 to 195, 160 to 190, 165 to 185, or 170 to 180, base pairs homology 3′ of the nick and/or replacement sequence. In an embodiment, the template nucleic acid comprises about 150, 155, 160, 165, 170, 175, 180, 185, 190, 195, or 200 base pairs homology 3′ of the nick or replacement sequence. In an embodiment, the template nucleic acid comprises less than about 100, 90, 80, 70, 60, 50, 40, 30, 20, 15, or 10 base pairs homology 5′ of the nick or replacement sequence.
  • In an embodiment, the template nucleic acid comprises about 150 to 200, e.g., 155 to 195, 160 to 190, 165 to 185, or 170 to 180, base pairs homology 5′ of the nick and/or replacement sequence. In an embodiment, the template nucleic acid comprises about 150, 155, 160, 165, 170, 175, 180, 185, 190, 195, or 200 base pairs homology 5′ of the nick or replacement sequence. In an embodiment, the template nucleic acid comprises less than about 100, 90, 80, 70, 60, 50, 40, 30, 20, 15, or 10 base pairs homology 3′ of the nick or replacement sequence.
  • In an embodiment, the 5′ of the homology region is the position next or adjacent (e.g., within 10, 20, 30, 40, or 50 base pairs) to the 3′ of the template binding domain partner. In an embodiment, the 3′ of the homology region is the position next or adjacent (e.g., within 10, 20, 30, 40, or 50 base pairs) to the 5′ of the template binding domain partner. For example, the template binding domain partner can be placed at the right of the nucleic acid template system, or at the left of the nucleic acid template system, as shown in FIG. 3.
  • Exemplary Arrangements of Circular Nucleic Acid Template Systems
  • In an embodiment, the nucleic acid template system is double stranded. In an embodiment, the nucleic acid template system is double stranded comprises a single stranded portion and a double stranded portion. In an embodiment, the nucleic acid template system is single stranded.
  • In an embodiment, the template nucleic acid comprises about 500 to 2000, e.g., 700 to 1900, 800 to 1800, 900 to 1700, 900 to 1600, 1000 to 1500, 1100 to 1400, or 1200 to 1300 base pairs, homology on either side of the nick and/or replacement sequence. In an embodiment, the template nucleic acid comprises about 500, 600, 700, 800, 900, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900, or 2000 base pairs homology 5′ of the nick or replacement sequence, 3′ of the nick or replacement sequence, or both 5′ and 3′ of the nick or replacement sequences.
  • In an embodiment, the template nucleic acid comprises about 500 to 2000, e.g., 700 to 1900, 800 to 1800, 900 to 1700, 900 to 1600, 1000 to 1500, 1100 to 1400, or 1200 to 1300, base pairs homology 3′ of the nick and/or replacement sequence. In an embodiment, the template nucleic acid comprises about 500, 600, 700, 800, 900, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900, or 2000 base pairs homology 3′ of the nick or replacement sequence. In an embodiment, the template nucleic acid comprises less than about 500, 400, 300, 200, 100, or 50 base pairs homology 5′ of the nick or replacement sequence.
  • In an embodiment, the template nucleic acid comprises about 500 to 2000, e.g., 700 to 1900, 800 to 1800, 900 to 1700, 900 to 1600, 1000 to 1500, 1100 to 1400, or 1200 to 1300, base pairs homology 5′ of the nick and/or replacement sequence. In an embodiment, the template nucleic acid comprises about 500, 600, 700, 800, 900, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900, or 2000 base pairs homology 5′ of the nick or replacement sequence. In an embodiment, the template nucleic acid comprises less than about 500, 400, 300, 200, 100, or 50 base pairs homology 3′ of the nick or replacement sequence.
  • In an embodiment, the 5′ of the homology region is the position next or adjacent (e.g., within 10, 20, 30, 40, or 50 base pairs) to the 3′ of the template binding domain partner. In an embodiment, the 3′ of the homology region is the position next or adjacent (e.g., within 10, 20, 30, 40, or 50 base pairs) to the 5′ of the template binding domain partner.
  • In an embodiment, the 5′ of the homology region is the position at least 100, 250, 500, 1000, 1500, 2000, or 2500 base pairs downstream from the 3′ of the template binding domain partner. In an embodiment, the 3′ of the homology region is the position at least 100, 250, 500, 1000, 1500, 2000, or 2500 base pairs upstream from the 5′ of the template binding domain partner.
  • In an embodiment, the 5′ of the homology region is the position next or adjacent (e.g., within 10, 20, 30, 40, or 50 base pairs) to the 3′ of the template binding domain partner, and the 3′ of the homology region is the position at least 100, 250, 500, 1000, 1500, 2000, or 2500 base pairs upstream from the 5′ of the template binding domain partner. In an embodiment, the 3′ of the homology region is the position next or adjacent (e.g., within 10, 20, 30, 40, or 50 base pairs) to the 5′ of the template binding domain partner, and the 5′ of the homology region is the position at least 100, 250, 500, 1000, 1500, 2000, or 2500 base pairs downstream from the 3′ of the template binding domain partner.
  • In an embodiment, the 5′ of the homology region is the position at least 100, 250, 500, 1000, 1500, 2000, or 2500 base pairs downstream from the 3′ of the template binding domain partner, and the 3′ of the homology region is the position at least 100, 250, 500, 1000, 1500, 2000, or 2500 base pairs upstream from the 5′ of the template binding domain partner.
  • For example, the template nucleic acid can comprise a homology region to the right of the template binding domain partner, to the left of the template binding domain partner, or on the opposite side of the circular nucleic acid molecule from the template binding domain partner, as shown in FIG. 4.
  • Methods of Promoting Break Repair by an HDR Pathway
  • In another aspect, disclosed herein is a method of altering a cell, e.g., altering the structure, e.g., altering the sequence, of a target nucleic acid of a cell, comprising contacting the cell with: (a) a gRNA that targets a target position, e.g., a gRNA as described herein; (b) a Cas9 molecule, e.g., a Cas9 fusion molecule as described herein; (c) a template nucleic acid, (d) a template binding domain, and (e) a template binding domain partner.
  • In some embodiments, the method comprises contacting the cell with (a) and (b), or with (a) and (c), or with (a) and (d), or with (a) and (e), or with (b) and (c), or with (b) and (d), or with (b) and (e), or with (c) and (d), or with (c) and (e), or with (d) and (e).
  • In some embodiments, the method comprises contacting the cell with (a), (b), and (c); with (a), (b), and (d); with (a), (b), and (e); with (b), (c), and (d); or with (b), (c), and (e); with (c), (d), and (e).
  • In some embodiments, the method comprises contacting said cell with (a), (b), (c) and (d); with (a), (b), (c), and (e); with (a), (b), (d), and (e); with (a), (c), (d), and (e); or with (b), (c), (d), and (e).
  • The contacting may be performed ex vivo and the contacted cell may be returned to the subject's body after the contacting step. In other embodiments, the contacting step may be performed in vivo.
  • In some embodiments, contacting a cell with a Cas9 fusion molecule comprises contacting the cell with a nucleic acid encoding the Cas9 fusion molecule and allowing the cell to produce the Cas9 fusion molecule. In some embodiments, contacting a cell with a gRNA comprises contacting the cell with DNA that can direct transcription of the gRNA, and allowing the cell to produce the gRNA.
  • In some embodiments, the method of altering a cell as described herein comprises acquiring knowledge of the presence of an undesired sequence in said cell, prior to the contacting step. Acquiring knowledge of the sequence of the undesired sequence in the cell may be by DNA sequencing.
  • In some embodiments, the contacting step of the method comprises contacting the cell with a nucleic acid, e.g., a vector, e.g., an AAV vector, that expresses or comprises at least one of (a), (b), (c), (d), and (e). In some embodiments, the contacting step of the method comprises contacting the cell with a nucleic acid, e.g., a vector, e.g., an AAV vector, that expresses or comprises each of (a), (b), (c), (d), and (e). In some embodiments, the contacting step of the method comprises contacting the cell with a nucleic acid, e.g., a vector, e.g., an AAV vector, that expresses or comprises two, three, or four of (a), (b), (c), (d), and (e). In some embodiments, the contacting step of the method comprises contacting the cell with a nucleic acid, e.g., a vector, e.g., an AAV vector, that expresses or comprises each of (a) and (b).
  • In an embodiment, contacting comprises contacting the cell with a nucleic acid, e.g., a vector, e.g., an AAV vector, e.g., an AAV2 vector, a modified AAV2 vector, an AAV3 vector, a modified AAV3 vector, an AAV6 vector, a modified AAV6 vector, an AAV8 vector or an AAV9 vector.
  • In an embodiment, contacting comprises delivering to the cell a Cas9 fusion molecule of (b) and a template binding domain of (d), as a protein or an mRNA, and a nucleic acid which encodes or comprises (a), (c) and (e).
  • In an embodiment, contacting comprises delivering to the cell a Cas9 fusion molecule of (b), as a protein or an mRNA, said gRNA of (a), as an RNA, and optionally (c) as a nucleic acid.
  • In some embodiments, a subject is treated by inducing a Cas9-mediated break at a target position, wherein the target position causes or exacerbates a disease or disorder, and administering one or more of (a), (b), (c), (d), and (e), wherein the break is repaired by HDR.
  • The method of treating a subject may comprise contacting the subject (or a cell from the subject) with (c) a template nucleic acid. A template nucleic acid is used when the method of treating a subject uses HDR to alter the sequence of the target nucleic acid of the subject.
  • In an embodiment, the method comprises acquiring knowledge of an undesired sequence in said subject, e.g., by DNA sequencing.
  • In an embodiment, the method comprises correcting an undesired sequence by HDR.
  • When the method comprises correcting an undesired sequence by HDR, a Cas9 fusion molecule of (b), at least one guide RNA, e.g., a guide RNA of (a) and a template nucleic acid (c) can be included in the contacting step.
  • In an embodiment, a cell of the subject is contacted ex vivo with (a), (b), (c), (d), and (e). In an embodiment, said cell is returned to the subject's body.
  • In an embodiment, a cell of the subject is contacted is in vivo with one or more of, e.g., all of, (a), (b) (c), (d), and (e).
  • In an embodiment, the cell of the subject is contacted in vivo by intravenous delivery of one or more of, e.g., all of, (a), (b), (c), (d), and (e).
  • In an embodiment, contacting comprises contacting the subject with a nucleic acid, e.g., a vector, e.g., an AAV vector, described herein, e.g., a nucleic acid that encodes or comprises at least one of, e.g., all of, (a), (b), (c), (d), and (e).
  • In an embodiment, contacting comprises delivering to said subject said Cas9 fusion molecule of (b), as a protein or mRNA, and one or more nucleic acid which encodes or comprises at least one of, e.g., all of, (a), (c), (d), and (e)
  • In an embodiment, contacting comprises delivering to the subject the Cas9 fusion molecule of (b), as a protein or mRNA, the gRNA of (a), as an RNA, a nucleic acid of (c) as a DNA, the template binding domain of (d) as an RNA, and a nucleic acid of (e) as a DNA.
  • In an embodiment, contacting comprises delivering to the subject the gRNA of (a), as an RNA, a nucleic acid that encodes the Cas9 fusion molecule of (b), and a nucleic acid of (c).
  • In an embodiment, a cell of the subject is contacted ex vivo with (a), (b), (c), (d), and (e). In an embodiment, said cell is returned to the subject's body.
  • In an embodiment, contacting comprises contacting the subject with a nucleic acid, e.g., a vector, e.g., an AAV vector, described herein, e.g., a nucleic acid that encodes or comprises at least one of, e.g., all of, (a), (b), (c), (d), and (e).
  • In an embodiment, contacting comprises delivering to said subject the Cas9 fusion molecule of (b), as a protein or mRNA, and a nucleic acid which encodes or comprises one or more of (a), (c), (d), and (e).
  • In an embodiment, contacting comprises delivering to the subject the Cas9 fusion molecule of (b), as a protein or mRNA, the gRNA of (a), as an RNA, and the template nucleic acid of (c) as a DNA.
  • VIII.2 Examples of gRNAs in Genome Editing Methods
  • gRNA molecules as described herein can be used with Cas9 fusion molecules that generate a double strand break or a single strand break to alter the sequence of a target nucleic acid, e.g., a target position or target genetic signature. gRNA molecules useful in these methods are described below.
  • In some embodiments, the gRNA is used in making double stranded breaks. In an embodiment, the gRNA, e.g., a chimeric gRNA, is configured such that it comprises one or more of the following properties:
      • a) it can position, e.g., when targeting a Cas9 fusion molecule that makes double strand breaks, a double strand break (i) within 50, 100, 150 or 200 nucleotides of a target position, or (ii) sufficiently close that the target position is within the region of end resection;
      • b) it has a targeting domain of at least 17 nucleotides, e.g., a targeting domain of (i) 17, (ii) 18, or (iii) 20 nucleotides; and
      • c)
  • (i) the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides, e.g., at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides from a naturally occurring S. aureus, S. thermophilus, or N. meningitidis tail and proximal domain, or a sequence that differs by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 nucleotides therefrom;
  • (ii) there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain, e.g., at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides from the corresponding sequence of a naturally occurring S. aureus, S. thermophilus, or N. meningitidis gRNA, or a sequence that differs by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 nucleotides therefrom;
  • (iii) there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain, e.g., at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides from the corresponding sequence of a naturally occurring S. aureus, S. thermophilus, or N. meningitidis gRNA, or a sequence that differs by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 nucleotides therefrom;
  • iv) the tail domain is at least 10, 15, 20, 25, 30, 35 or 40 nucleotides in length, e.g., it comprises at least 10, 15, 20, 25, 30, 35 or 40 nucleotides from a naturally occurring S. aureus, S. thermophilus, or N. meningitidis tail domain; or a sequence that differs by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 nucleotides therefrom; or
  • (v) the tail domain comprises 15, 20, 25, 30, 35, 40 nucleotides or all of the corresponding portions of a naturally occurring tail domain, e.g., a naturally occurring S. aureus, S. thermophilus, or N. meningitidis tail domain.
  • In an embodiment, the gRNA is configured such that it comprises properties: a and b(i).
  • In an embodiment, the gRNA is configured such that it comprises properties: a and b(ii).
  • In an embodiment, the gRNA is configured such that it comprises properties: a and b(iii).
  • In an embodiment, the gRNA is configured such that it comprises properties: a and c.
  • In an embodiment, the gRNA is configured such that in comprises properties: a, b, and c.
  • In an embodiment, the gRNA is configured such that in comprises properties: a(i), b(i), and c(i).
  • In an embodiment, the gRNA is configured such that in comprises properties: a(i), b(i), and c(ii).
  • In an embodiment, the gRNA is configured such that in comprises properties: a(i), b(iii), and c(i).
  • In an embodiment, the gRNA is configured such that in comprises properties: a(i), b(iii), and c(ii).
  • In some embodiments, the gRNA is used in making single stranded breaks. In an embodiment, the gRNA, e.g., a chimeric gRNA, is configured such that it comprises one or more of the following properties:
      • a) it can position, e.g., when targeting a Cas9 fusion molecule that makes single strand breaks, a single strand break (i) within 50, 100, 150 or 200 nucleotides of a target position, or (ii) sufficiently close that the target position is within the region of end resection;
      • b) it has a targeting domain of at least 17 nucleotides, e.g., a targeting domain of (i) 17, (ii) 18, or (iii) 20 nucleotides; and
      • c)
  • (i) the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides, e.g., at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides from a naturally occurring S. aureus, S. thermophilus, or N. meningitidis tail and proximal domain, or a sequence that differs by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 nucleotides therefrom;
  • (ii) there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain, e.g., at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides from the corresponding sequence of a naturally occurring S. aureus, S. thermophilus, or N. meningitidis gRNA, or a sequence that differs by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 nucleotides therefrom;
  • (iii) there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain, e.g., at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides from the corresponding sequence of a naturally occurring S. aureus, S. thermophilus, or N. meningitidis gRNA, or a sequence that differs by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 nucleotides therefrom;
  • iv) the tail domain is at least 10, 15, 20, 25, 30, 35 or 40 nucleotides in length, e.g., it comprises at least 10, 15, 20, 25, 30, 35 or 40 nucleotides from a naturally occurring S. aureus, S. thermophilus, or N. meningitidis tail domain; or, a sequence that differs by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 nucleotides therefrom; or
  • (v) the tail domain comprises 15, 20, 25, 30, 35, 40 nucleotides or all of the corresponding portions of a naturally occurring tail domain, e.g., a naturally occurring S. aureus, S. thermophilus, or N. meningitidis tail domain.
  • In an embodiment, the gRNA is configured such that it comprises properties: a and b(i).
  • In an embodiment, the gRNA is configured such that it comprises properties: a and b(ii).
  • In an embodiment, the gRNA is configured such that it comprises properties: a and b(iii).
  • In an embodiment, the gRNA is configured such that it comprises properties: a and c.
  • In an embodiment, the gRNA is configured such that in comprises properties: a, b, and c.
  • In an embodiment, the gRNA is configured such that in comprises properties: a(i), b(i), and c(i).
  • In an embodiment, the gRNA is configured such that in comprises properties: a(i), b(i), and c(ii).
  • In an embodiment, the gRNA is configured such that in comprises properties: a(i), b(iii), and c(i).
  • In an embodiment, the gRNA is configured such that in comprises properties: a(i), b(iii), and c(ii).
  • In an embodiment, the gRNA is used with a Cas9 nickase molecule having HNH activity, e.g., a Cas9 molecule having the RuvC activity inactivated, e.g., a Cas9 molecule having a mutation at D10, e.g., the D10A mutation.
  • In an embodiment, the gRNA is used with a Cas9 nickase molecule having RuvC activity, e.g., a Cas9 molecule having the HNH activity inactivated, e.g., a Cas9 molecule having a mutation at H840, e.g., a H840A.
  • In an embodiment, a pair of gRNAs, e.g., a pair of chimeric gRNAs, comprising a first and a second gRNA, is configured such that they comprises one or more of the following properties:
      • a) one or both of the gRNAs can position, e.g., when targeting a Cas9 molecule that makes single strand breaks, a single strand break within (i) 50, 100, 150 or 200 nucleotides of a target position, or (ii) sufficiently close that the target position is within the region of end resection;
      • b) one or both have a targeting domain of at least 17 nucleotides, e.g., a targeting domain of (i) 17 or (ii) 18 nucleotides;
      • c) for one or both:
  • (i) the proximal and tail domain, when taken together, comprise at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides, e.g., at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides from a naturally occurring S. aureus, S. thermophilus, or N. meningitidis tail and proximal domain, or a sequence that differs by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 nucleotides therefrom;
  • (ii) there are at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides 3′ to the last nucleotide of the second complementarity domain, e.g., at least 15, 18, 20, 25, 30, 31, 35, 40, 45, 49, 50, or 53 nucleotides from the corresponding sequence of a naturally occurring S. aureus, S. thermophilus, or N. meningitidis gRNA, or a sequence that differs by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 nucleotides therefrom;
  • (iii) there are at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides 3′ to the last nucleotide of the second complementarity domain that is complementary to its corresponding nucleotide of the first complementarity domain, e.g., at least 16, 19, 21, 26, 31, 32, 36, 41, 46, 50, 51, or 54 nucleotides from the corresponding sequence of a naturally occurring S. aureus, S. thermophilus, or N. meningitidis gRNA, or a sequence that differs by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 nucleotides therefrom;
  • iv) the tail domain is at least 10, 15, 20, 25, 30, 35 or 40 nucleotides in length, e.g., it comprises at least 10, 15, 20, 25, 30, 35 or 40 nucleotides from a naturally occurring S. aureus, S. thermophilus, or N. meningitidis tail domain; or a sequence that differs by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 nucleotides therefrom; or
  • (v) the tail domain comprises 15, 20, 25, 30, 35, 40 nucleotides or all of the corresponding portions of a naturally occurring tail domain, e.g., a naturally occurring S. aureus, S. thermophilus, or N. meningitidis tail domain;
      • d) the gRNAs are configured such that, when hybridized to target nucleic acid, they are separated by 0-50, 0-100, 0-200, at least 10, at least 20, at least 30 or at least 50 nucleotides;
      • e) the breaks made by the first gRNA and second gRNA are on different strands; and
      • f) the PAMs are facing outwards.
  • In an embodiment, one or both of the gRNAs is configured such that it comprises properties: a and b(i).
  • In an embodiment, one or both of the gRNAs is configured such that it comprises properties: a and b(ii).
  • In an embodiment, one or both of the gRNAs is configured such that it comprises properties: a and b(iii).
  • In an embodiment, one or both of the gRNAs configured such that it comprises properties: a and c.
  • In an embodiment, one or both of the gRNAs is configured such that it comprises properties: a, b, and c.
  • In an embodiment, one or both of the gRNAs is configured such that it comprises properties: a(i), b(i), and c(i).
  • In an embodiment, one or both of the gRNAs is configured such that it comprises properties: a(i), b(i), and c(ii).
  • In an embodiment, one or both of the gRNAs is configured such that it comprises properties: a(i), b(i), c, and d.
  • In an embodiment, one or both of the gRNAs is configured such that it comprises properties: a(i), b(i), c, and e.
  • In an embodiment, one or both of the gRNAs is configured such that it comprises properties: a(i), b(i), c, and f.
  • In an embodiment, one or both of the gRNAs is configured such that it comprises properties: a(i), b(i), c, d, and e.
  • In an embodiment, one or both of the gRNAs is configured such that it comprises properties: a(i), b(i), c, d, and f.
  • In an embodiment, one or both of the gRNAs is configured such that it comprises properties: a(i), b(i), c, d, e, and f.
  • In an embodiment, one or both of the gRNAs is configured such that it comprises properties: a(i), b(iii), and c(i).
  • In an embodiment, one or both of the gRNAs is configured such that it comprises properties: a(i), b(iii), and c(ii).
  • In an embodiment, one or both of the gRNAs is configured such that it comprises properties: a(i), b(iii), c, and d.
  • In an embodiment, one or both of the gRNAs is configured such that it comprises properties: a(i), b(iii), c, and e.
  • In an embodiment, one or both of the gRNAs is configured such that it comprises properties: a(i), b(iii), c, and f.
  • In an embodiment, one or both of the gRNAs is configured such that it comprises properties: a(i), b(iii), c, d, and e.
  • In an embodiment, one or both of the gRNAs is configured such that it comprises properties: a(i), b(iii), c, d, and f.
  • In an embodiment, one or both of the gRNAs is configured such that it comprises properties: a(i), b(iii), c, d, e, and f.
  • In an embodiment, the gRNAs are used with a Cas9 nickase molecule having HNH activity, e.g., a Cas9 molecule having the RuvC activity inactivated, e.g., a Cas9 molecule having a mutation at D10, e.g., the D10A mutation.
  • In an embodiment, the gRNAs are used with a Cas9 nickase molecule having RuvC activity, e.g., a Cas9 molecule having the HNH activity inactivated, e.g., a Cas9 molecule having a mutation at H840, e.g., a H840A.
  • IX. Target Cells
  • Cas9 molecules and gRNA molecules, e.g., a Cas9 fusion molecule/gRNA molecule complex, as disclosed herein, can be used to manipulate, e.g., to edit a target nucleic acid, in a wide variety of cells. Additional details on types of cells that can be manipulated may be found in the section entitled “VIIA. TARGETS: CELLS” of PCT Application WO 2015/048577, the entire contents of which are expressly incorporated herein by reference.
  • In some embodiments, a cell is manipulated by editing (e.g., introducing a mutation or correcting) one or more target genes, e.g., as described herein. In an embodiment, a cell is manipulated by editing one or more non-coding sequences, e.g., an alteration in an intron or in a 5′ or 3′ non-translated or non-transcribed region. In an embodiment, a cell is manipulated by editing the sequence of a control element, e.g., a promoter, enhancer, or a cis-acting or trans-acting control element. In an embodiment, a cell is manipulated by editing one or more coding sequences, e.g., an alteration in an exon.
  • In some embodiments, a cell is manipulated in vitro. In other embodiments, a cell is manipulated ex vivo. In some embodiments, a cell is manipulated in vivo. In some embodiments, the expression of one or more target genes (e.g., one or more target genes described herein) is modulated, e.g., in vivo. In other embodiments, the expression of one or more target genes (e.g., one or more target genes described herein) is modulated, e.g., ex vivo. In other embodiments, the expression of one or more target genes (e.g., one or more target genes described herein) is modulated, e.g., in vitro.
  • In some embodiments, the cells are manipulated (e.g., converted or differentiated) from one cell type to another. In some embodiments, a pancreatic cell is manipulated into a beta islet cell. In some embodiments, a fibroblast is manipulated into an iPS cell. In some embodiments, a preadipocyte is manipulated into a brown fat cell. Other exemplary cells include, e.g., muscle cells, neural cells, leukocytes, and lymphocytes.
  • In some embodiments, a nucleic acid at one or more target positions in a cell, or a population of cells, is altered e.g., as described herein. In some embodiments, a nucleic acid at one or more target positions in a cell, or a population of cells (e.g., target positions at one or more genes described herein) is altered, e.g., in vivo. In other embodiments, a nucleic acid at one or more target positions in a cell, or a population of cells (e.g., target positions at one or more genes described herein) is altered, e.g., ex vivo. The Cas9 fusion molecule, nucleic acid template system, and/or gRNA molecules described herein can be delivered to a cell or to a population of cells.
  • In some embodiments, the cell, or the population of cells, is a T cell, a CD8+ T cell, a CD8+ naïve T cell, a central memory T cell, an effector memory T cell, a CD4+ T cell, a stem cell memory T cell, a helper T cell, a regulatory T cell, a cytotoxic T cell, a natural killer T cell, a hematopoietic stem cell, a long term hematopoietic stem cell, a short term hematopoietic stem cell, a multipotent progenitor cell, a lineage restricted progenitor cell, a lymphoid progenitor cell, a pancreatic progenitor cell, an endocrine progenitor cell, an exocrine progenitor cell, a myeloid progenitor cell, a common myeloid progenitor cell, an erythroid progenitor cell, a megakaryocyte erythroid progenitor cell, a monocytic precursor cell, an endocrine precursor cell, an exocrine cell, a fibroblast, a hepatoblast, a myoblast, a macrophage, an islet beta-cell, a cardiomyocyte, a blood cell, a ductal cell, an acinar cell, an alpha cell, a beta cell, a delta cell, a PP cell, a cholangiocyte, a retinal cell, a photoreceptor cell, a rod cell, a cone cell, a retinal pigmented epithelium cell, a trabecular meshwork cell, a cochlear hair cell, an outer hair cell, an inner hair cell, a pulmonary epithelial cell, a bronchial epithelial cell, an alveolar epithelial cell, a pulmonary epithelial progenitor cell, a striated muscle cell, a cardiac muscle cell, a muscle satellite cell, a myocyte, a neuron, a neuronal stem cell, a mesenchymal stem cell, an induced pluripotent stem (iPS) cell, an embryonic stem cell, a monocyte, a megakaryocyte, a neutrophil, an eosinophil, a basophil, a mast cell, a reticulocyte, a B cell, e.g. a progenitor B cell, a Pre B cell, a Pro B cell, a memory B cell, a plasma B cell, a gastrointestinal epithelial cell, a biliary epithelial cell, a pancreatic ductal epithelial cell, an intestinal stem cell, a hepatocyte, a liver stellate cell, a Kupffer cell, an osteoblast, an osteoclast, an adipocyte (e.g., a brown adipocyte, or a white adipocyte), a preadipocyte, a pancreatic precursor cell, a pancreatic islet cell, a pancreatic beta cell, a pancreatic alpha cell, a pancreatic delta cell, a pancreatic exocrine cell, a Schwann cell, or an oligodendrocyte, or a population of such cells.
  • In some embodiments, the cell, or the population of cells, is a mammalian cell, e.g., a human cell, a mouse cell, a rat cell, a sheep cell, a cow cell, a pig cell, a horse cell, a goat cell, a dog cell or a cat cell, or a population of mammalian cells. In one embodiment, the cell is a human cell.
  • In an embodiment, the cell, or population of cells, is manipulated ex vivo by altering a nucleic acid at one or more target positions, and administered to a subject. A cell, or population of cells, to be altered according to the methods disclosed herein, may include a stem cell such as, by way of example, an embryonic stem cell, an induced pluripotent stem cell or a neuronal stem cell, or a population of such cells. In an embodiment, the cell, or population of cells, is an induced pluripotent stem (iPS) cell or a cell derived from an iPS cell, or a population of such cells, altered to correct a mutation and differentiated into a clinically relevant cell, or population of cells.
  • In some embodiments, the cell is a cell from a disease-causing organism, e.g., a bacterium, fungus, protozoan, or parasite. In some embodiments, the cell is a cell infected with a disease-causing organism (e.g., a virus, fungus, protozoan, or parasite).
  • In some embodiments, the cell is situated in the body of a subject. In such instances, the cell might be the subject's own cells or might be a cell of a disease-causing organism. In this case, a gRNA molecule, a Cas9 fusion molecule, and a nucleic acid template system, may be administered to the subject as pharmaceutical compositions. In some embodiments, the subject is a mammal, e.g., a human, a farm animal (e.g., a cow, a pig, a horse, or a goat), or a companion animal (e.g., a dog or a cat).
  • In some embodiments, the subject suffers from a disease caused by a target position in a nucleic acid, e.g., a particular mutation, of a cell, or population of cells.
  • In some embodiments, the cell, or population of cells, is a diseased or mutant-bearing cell, or population of cells. Such cells can be altered to treat the disease, e.g., to correct a mutation, or to alter the phenotype of the cell, or population of cells, e.g., to inhibit the growth of a cancer cell or a population of cancer cells, e.g., a tumor. For example, a cell, or a population of cells, is associated with one or more diseases or conditions describe herein. In some embodiments, the cell is a cancer stem cell. In some embodiments, the cancer cell is selected from lung cancer cells, breast cancer cells, skin cancer cells, brain cancer cells, pancreatic cancer cells, hematopoietic cancer cells, liver cancer cells, kidney cancer cells, and ovarian cancer cells.
  • In some embodiments, the cell is characterized by a disorder caused by aberrant mtDNA. This disorder may be, e.g., a mtDNA depletion syndrome (e.g., Alpers or early infantile hepatocerebral syndromes) or a mtDNA deletion disorder (e.g., progressive external ophthalmoplegia (PEO), ataxia-neuropathy, or mitochondrial neurogastrointestinal encephalomyopathy (MNGIE)).
  • In some embodiments, the cell, or population of cells, is a normal cell or a population of normal cells.
  • In some embodiments, the cell, or population of cells, is a stem cell or a progenitor cell (e.g., iPS, embryonic, hematopoietic, adipose, germline, lung, or neural stem or progenitor cells), or a population of such cells.
  • The cells may also be treated at a time when they are not situated in the body of a subject. In some embodiments, a cell, or a population of cells, is treated ex vivo to avoid exposing a patient to an agent or agents that cause undesirable side effects. In some embodiments, treating cells ex vivo allows a user to select a sub-population of cells to administer to the patient. The sub-population may be, e.g., cells having a nucleic acid that was successfully altered, or cells having a desired phenotype, such as minimal undesired alterations to DNA, or a phenotype that indicates the nucleic acid was successfully altered.
  • In some embodiments, the cell, or population of cells, is not situated in a subject's body and the cell, or population of cells, is modified for research or manufacturing purposes. In some embodiments, the cell, or population of cells, is suitable for producing a recombinant biological product. For example, the cell, or population of cells, can be a CHO cell or a fibroblast. In one embodiment, the cell, or population of cells, is a cell, or population of cells, that has been engineered to express a protein.
  • In some embodiments, the cell, or population of cells, is actively dividing. In some embodiments, the cell is in G2 phase. In some embodiments, the population of cells comprises cells that are in G2 phase. In some embodiments, the cell is in G1 phase. In some embodiments, the population of cells comprises cells that are in G1 phase. In some embodiments, the cell is in S phase. In some embodiments, the population of cells comprises cells that are in S phase.
  • The technology described herein can be used to edit numerous types of genomes, including plant genomes. The CRISPR/Cas system has been used for plant genome editing, as has been described in, e.g., Belhaj et al., PLANT METHODS 9:39, 2013. Accordingly, in certain embodiments, the cell, or the population of cells, is a plant cell, e.g., a monocot plant cell, or a dicot plant cell, or a population of plant cell. In certain embodiments, the plant is a crop, e.g., a food crop. In certain embodiments, the plant is rice (e.g., Orzya sativa), maize (e.g., Zea mays), wheat (e.g., Triticum aestivum), soy (e.g., Glycine max), potato (e.g., Solanum tuberosum), a species of Nicotiana, a species of Arabidopsis e.g., Arabidopsis thaliana, cassava, sweet potato, sorghum, yam, plantain, or a citrus plant. In some embodiments, the plant is a pesticide-resistant plant, e.g., a plant that expresses one or more genes that confer resistance to a pesticide. In some embodiments, the plant is herbicide-resistant plant, e.g., a plant that expresses one or more genes that confer resistance to a herbicide. The herbicide may be, e.g., Roundup® (also known as glyphosate or N-(phosphonomethyl)glycine). In some embodiments, the plant produces a pesticide, e.g., Bt.
  • In some embodiments, the components used in the methods described herein (e.g., a Cas9 fusion molecule, a nucleic acid template system, and/or a gRNA) are introduced into the plant cell via protoplast transformation or agroinfiltration.
  • In some embodiments, after genome editing using the methods described herein, seeds are screened and a desired sub-population of seeds are selected. The sub-population may be, e.g., cells having a nucleic acid that was successfully altered, or cells having a desired phenotype such as minimal undesired alterations to DNA, or a phenotype that indicates the nucleic acid was successfully altered.
  • X. Delivery, Formulations and Routes of Administration
  • The components, e.g., a Cas9 fusion molecule, a nucleic acid template system, and/or a gRNA molecule can be delivered or formulated in a variety of forms, see, e.g., Tables 4-5. When a Cas9 fusion molecule, nucleic acid template system component, and/or a gRNA component is encoded by DNA for delivery, the DNA will typically but not necessarily include a control region, e.g., comprising a promoter, to effect expression. Useful promoters for Cas9 fusion molecule sequences include CMV, EF-1a, MSCV, PGK, CAG control promoters. Useful promoters for gRNAs include H1, EF-1a and U6 promoters. Promoters with similar or dissimilar strengths can be selected to tune the expression of components. Sequences encoding a Cas9 fusion molecule can comprise a nuclear localization signal (NLS), e.g., an SV40 NLS. In an embodiment a promoter for a Cas9 fusion molecule or a gRNA molecule can be, independently, inducible, tissue specific, or cell specific.
  • Table 4 provides non-limiting examples of the form in which the components can be delivered to a target cell.
  • TABLE 4
    Elements
    Template
    Cas9 Fusion gRNA Nucleic Acid
    Molecule(s) Molecule(s) System Comments
    DNA DNA DNA In some embodiments, a Cas9 fusion
    molecule, e.g., an eaCas9 fusion molecule,
    and a gRNA are transcribed from DNA. In
    this embodiment, they are encoded on
    separate molecules. In this embodiment, the
    donor template is provided as a separate DNA
    molecule. In some embodiments, the
    template binding domain is encoded by DNA,
    optionally on the same nucleic acid as the
    Cas9 fusion molecule. In some embodiments,
    the template binding domain partner is
    provided as DNA, and is optionally part of the
    same nucleic acid as the donor template.
    DNA DNA In an embodiment, a Cas9 fusion molecule,
    e.g., an eaCas9 fusion molecule, and a gRNA
    are transcribed from DNA. In this
    embodiment, they are encoded on separate
    molecules. In this embodiment, the donor
    template is provided on the same DNA
    molecule that encodes the gRNA. In some
    embodiments, the template binding domain is
    encoded in DNA, optionally on the same
    nucleic acid as the Cas9 fusion molecule. In
    some embodiments, the template binding
    domain partner is provided as DNA, and is
    optionally part of the same nucleic acid as the
    donor template.
    DNA DNA In some embodiments, a Cas9 fusion
    molecule, e.g., an eaCas9 fusion molecule,
    and a gRNA are transcribed from DNA, here
    from a single molecule. In this embodiment,
    the donor template is provided as a separate
    DNA molecule. In some embodiments, the
    template binding domain is encoded in DNA,
    optionally on the same nucleic acid as the
    Cas9 fusion molecule. In some embodiments,
    the template binding domain partner is
    provided as DNA, and is optionally part of the
    same nucleic acid as the donor template.
    DNA DNA DNA In some embodiments, a Cas9 fusion
    molecule, e.g., an eaCas9 fusion molecule,
    and a gRNA are transcribed from DNA. In
    this embodiment, they are encoded on
    separate molecules. In this embodiment, the
    donor template is provided on the same DNA
    molecule that encodes the Cas9 fusion
    molecule. In some embodiments, the
    template binding domain is encoded in DNA,
    optionally on the same nucleic acid as the
    Cas9 fusion molecule. In some embodiments,
    the template binding domain partner is
    provided as DNA, and is optionally part of the
    same nucleic acid as the donor template.
    DNA RNA DNA In some embodiments, a Cas9 fusion
    molecule, e.g., an eaCas9 fusion molecule, is
    transcribed from DNA, and a gRNA is
    provided as in vitro transcribed or synthesized
    RNA. In this embodiment, the donor template
    is provided as a separate DNA molecule. In
    some embodiments, the template binding
    domain is encoded in DNA, optionally on the
    same nucleic acid as the Cas9 fusion
    molecule. In some embodiments, the
    template binding domain partner is provided
    as DNA, and is optionally part of the same
    nucleic acid as the donor template.
    DNA RNA DNA In some embodiments, a Cas9 fusion
    molecule, e.g., an eaCas9 fusion molecule, is
    transcribed from DNA, and a gRNA is
    provided as in vitro transcribed or synthesized
    RNA. In this embodiment, the donor template
    is provided on the same DNA molecule that
    encodes the Cas9 fusion molecule. In some
    embodiments, the template binding domain is
    encoded in DNA, optionally on the same
    nucleic acid as the Cas9 fusion molecule. In
    some embodiments, the template binding
    domain partner is provided as DNA, and is
    optionally part of the same nucleic acid as the
    donor template.
    mRNA RNA DNA In some embodiments, a Cas9 fusion
    molecule, e.g., an eaCas9 fusion molecule, is
    translated from in vitro transcribed mRNA,
    and a gRNA is provided as in vitro
    transcribed or synthesized RNA. In this
    embodiment, the donor template is provided
    as a DNA molecule. In some embodiments,
    the template binding domain is translated
    form in vitro transcribed mRNA, and is
    optionally part of the same nucleic acid as the
    Cas9 fusion molecule. In some embodiments,
    the template binding domain partner is
    provided as DNA, and is optionally part of the
    same nucleic acid as the donor template.
    mRNA DNA DNA In some embodiments, a Cas9 fusion
    molecule, e.g., an eaCas9 fusion molecule, is
    translated from in vitro transcribed mRNA,
    and a gRNA is transcribed from DNA. In this
    embodiment, the donor template is provided
    as a separate DNA molecule. In some
    embodiments, the template binding domain is
    translated form in vitro transcribed mRNA,
    and is optionally part of the same nucleic acid
    as the Cas9 fusion molecule. In some
    embodiments, the template binding domain
    partner is provided as DNA, and is optionally
    part of the same nucleic acid as the donor
    template.
    mRNA DNA In some embodiments, a Cas9 fusion
    molecule, e.g., an eaCas9 fusion molecule, is
    translated from in vitro transcribed mRNA,
    and a gRNA is transcribed from DNA. In this
    embodiment, the donor template is provided
    on the same DNA molecule that encodes the
    gRNA. In some embodiments, the template
    binding domain is translated form in vitro
    transcribed mRNA, and is optionally part of
    the same nucleic acid as the Cas9 fusion
    molecule. In some embodiments, the
    template binding domain partner is provided
    as DNA, and is optionally part of the same
    nucleic acid as the donor template.
    Protein DNA DNA In some embodiments, a Cas9 fusion
    molecule, e.g., an eaCas9 fusion molecule, is
    provided as a protein, and a gRNA is
    transcribed from DNA. In this embodiment,
    the donor template is provided as a separate
    DNA molecule. In some embodiments, the
    template binding domain is provided as a
    protein, and is optionally covalently linked to,
    e.g., fused to, the Cas9 fusion molecule. In
    some embodiments, the template binding
    domain partner is provided as DNA, and is
    optionally part of the same nucleic acid as the
    donor template.
    Protein DNA In some embodiments, a Cas9 fusion
    molecule, e.g., an eaCas9 fusion molecule, is
    provided as a protein, and a gRNA is
    transcribed from DNA. In this embodiment,
    the donor template is provided on the same
    DNA molecule that encodes the gRNA. In
    some embodiments, the template binding
    domain is provided as a protein, and is
    optionally covalently linked to, e.g., fused to,
    the Cas9 molecule. In some embodiments,
    the template binding domain partner is
    provided as DNA, and is optionally part of the
    same nucleic acid as the donor template.
    Protein RNA DNA In some embodiments, a Cas9 fusion
    molecule, e.g., an eaCas9 fusion molecule is
    provided as a protein, and a gRNA is provided
    as transcribed or synthesized RNA. In this
    embodiment, the donor template is provided
    as a DNA molecule. In some embodiments,
    the template binding domain is provided as a
    protein, and is optionally covalently linked to,
    e.g., fused to, the Cas9 fusion molecule. In
    some embodiments, the template binding
    domain partner is provided as DNA, and is
    optionally part of the same nucleic acid as the
    donor template.
  • Table 5 summarizes various delivery methods for the components of a Cas9 system, e.g., the Cas9 fusion molecule, the nucleic acid template system, and a gRNA molecule, as described herein.
  • TABLE 5
    Delivery
    into Non- Duration Type of
    Dividing of Genome Molecule
    Delivery Vector/Mode Cells Expression Integration Delivered
    Physical (e.g., YES Transient NO Nucleic Acids
    electroporation, particle gun, and Proteins
    Calcium Phosphate
    transfection)
    Viral Retrovirus NO Stable YES RNA
    Lentivirus YES Stable YES/NO with RNA
    modifications
    Adenovirus YES Transient NO DNA
    Adeno- YES Stable NO DNA
    Associated
    Virus (AAV)
    Vaccinia Virus YES Very NO DNA
    Transient
    Herpes Simplex YES Stable NO DNA
    Virus
    Non-Viral Cationic YES Transient Depends on Nucleic Acids
    Liposomes what is and Proteins
    delivered
    Polymeric YES Transient Depends on Nucleic Acids
    Nanoparticles what is and Proteins
    delivered
    Biological Attenuated YES Transient NO Nucleic Acids
    Non-Viral Bacteria
    Delivery Engineered YES Transient NO Nucleic Acids
    Vehicles Bacteriophages
    Mammalian YES Transient NO Nucleic Acids
    Virus-like
    Particles
    Biological YES Transient NO Nucleic Acids
    liposomes:
    Erythrocyte
    Ghosts and
    Exosomes

    DNA-Based Delivery of a Cas9 Fusion Molecule, Nucleic Acid Template System and/or a gRNA Molecule
  • DNA encoding Cas9 fusion molecules (e.g., eaCas9 fusion molecules), gRNA molecules, template nucleic acids, template binding domains, and/or template binding domain partners can be administered to subjects or delivered into cells by any appropriate method, e.g., by art-known methods or as described herein. For example, Cas9 fusion molecule-encoding and/or gRNA-encoding DNA, a template binding domain partner, and a template nucleic acid can be delivered, e.g., by vectors (e.g., viral or non-viral vectors), non-vector based methods (e.g., using naked DNA or DNA complexes), or a combination thereof.
  • In some embodiments, the nucleic acid, e.g., Cas9 fusion molecule- and/or gRNA-encoding DNA is delivered by a vector (e.g., viral vector/virus or plasmid).
  • In one embodiment, a vector can comprise a sequence that encodes a Cas9 fusion molecule, a gRNA molecule, and a nucleic acid template system. In one embodiment, a vector can comprise a sequence encoding a signal peptide (e.g., for nuclear localization, nucleolar localization, mitochondrial localization), fused, e.g., to a Cas9 fusion molecule sequence. For example, a vector can comprise a nuclear localization sequence (e.g., from SV40) fused to the sequence encoding the Cas9 fusion molecule.
  • One or more regulatory/control elements, e.g., a promoter, an enhancer, an intron, a polyadenylation signal, a Kozak consensus sequence, internal ribosome entry sites (IRES), a 2A sequence, and splice acceptor or donor, can be included in the vectors. In some embodiments, the promoter is recognized by RNA polymerase II (e.g., a CMV promoter).
  • In some embodiments, the vector or delivery vehicle is a viral vector (e.g., for generation of recombinant viruses). In some embodiments, the virus is a DNA virus (e.g., dsDNA or ssDNA virus). In other embodiments, the virus is an RNA virus (e.g., an ssRNA virus). Exemplary viral vectors/viruses include, e.g., retroviruses, lentiviruses, adenovirus, adeno-associated virus (AAV), vaccinia viruses, poxviruses, and herpes simplex viruses.
  • In some embodiments, the virus infects dividing cells. In other embodiments, the virus infects non-dividing cells. In some embodiments, the virus infects both dividing and non-dividing cells. In some embodiments, the virus can integrate into the host genome. In some embodiments, the virus is engineered to have reduced immunity, e.g., in human. In some embodiments, the virus is replication-competent. In other embodiments, the virus is replication-defective, e.g., having one or more coding regions for the genes necessary for additional rounds of virion replication and/or packaging replaced with other genes or deleted. In some embodiments, the virus causes transient expression of the Cas9 fusion molecule and/or the gRNA molecule. In other embodiments, the virus causes long-lasting, e.g., at least 1 week, 2 weeks, 1 month, 2 months, 3 months, 6 months, 9 months, 1 year, 2 years, or permanent expression, of the Cas9 fusion molecule and/or the gRNA molecule. The packaging capacity of the viruses may vary, e.g., from at least about 4 kb to at least about 30 kb, e.g., at least about 5 kb, 10 kb, 15 kb, 20 kb, 25 kb, 30 kb, 35 kb, 40 kb, 45 kb, or 50 kb.
  • In some embodiments, the Cas9 fusion molecule-, gRNA-, and/or template binding domain-encoding DNA and/or the nucleic acid template system is delivered by a recombinant adenovirus. In some embodiments, the adenovirus is engineered to have reduced immunity in human.
  • In some embodiments, the Cas9 fusion molecule-, gRNA-, and/or template binding domain-encoding DNA and/or the nucleic acid template system is delivered by a recombinant AAV. In some embodiments, the AAV can incorporate its genome into that of a host cell, e.g., a target cell as described herein. In some embodiments, the AAV is a self-complementary adeno-associated virus (scAAV), e.g., a scAAV that packages both strands which anneal together to form double stranded DNA. AAV serotypes that may be used in the disclosed methods, include AAV1, AAV2, modified AAV2 (e.g., modifications at Y444F, Y500F, Y730F and/or S662V), AAV3, modified AAV3 (e.g., modifications at Y705F, Y731F and/or T492V), AAV4, AAV5, AAV6, modified AAV6 (e.g., modifications at S663V and/or T492V), AAV8, AAV 8.2, AAV9, AAV rhl0, and pseudotyped AAV, such as AAV2/8, AAV2/5 and AAV2/6 can also be used in the disclosed methods.
  • In some embodiments, the Cas9 fusion molecule-gRNA-, and/or template binding domain-encoding DNA, and/or the nucleic acid template system is delivered by a non-vector based method (e.g., using naked DNA or DNA complexes). For example, the DNA can be delivered, e.g., by organically modified silica or silicate (Ormosil), electroporation, gene gun, sonoporation, magnetofection, lipid-mediated transfection, dendrimers, inorganic nanoparticles, calcium phosphates, or a combination thereof.
  • In an embodiment, the delivery vehicle is a non-viral vector. In an embodiment, the non-viral vector is an inorganic nanoparticle. Exemplary inorganic nanoparticles include, e.g., magnetic nanoparticles (e.g., Fe3MnO2) and silica. The outer surface of the nanoparticle can be conjugated with a positively charged polymer (e.g., polyethylenimine, polylysine, polyserine) which allows for attachment (e.g., conjugation or entrapment) of payload. In an embodiment, the non-viral vector is an organic nanoparticle (e.g., entrapment of the payload inside the nanoparticle). Exemplary organic nanoparticles include, e.g., SNALP liposomes that contain cationic lipids together with neutral helper lipids which are coated with polyethylene glycol (PEG) and protamine and nucleic acid complex coated with lipid coating.
  • Exemplary lipids for gene transfer are shown below in Table 6.
  • TABLE 6
    Lipids Used for Gene Transfer
    Lipid Abbreviation Feature
    1,2-Dioleoyl-sn-glycero-3-phosphatidylcholine DOPC Helper
    1,2-Dioleoyl-sn-glycero-3-phosphatidylethanolamine DOPE Helper
    Cholesterol Helper
    N-[1-(2,3-Dioleyloxy)prophyl]N,N,N-trimethylammonium chloride DOTMA Cationic
    1,2-Dioleoyloxy-3-trimethylammonium-propane DOTAP Cationic
    Dioctadecylamidoglycylspermine DOGS Cationic
    N-(3-Aminopropyl)-N,N-dimethyl-2,3-bis(dodecyloxy)-1- GAP-DLRIE Cationic
    propanaminium bromide
    Cetyltrimethylammonium bromide CTAB Cationic
    6-Lauroxyhexyl ornithinate LHON Cationic
    1-(2,3-Dioleoyloxypropyl)-2,4,6-trimethylpyridinium 2Oc Cationic
    2,3-Dioleyloxy-N-[2(sperminecarboxamido-ethyl]-N,N-dimethyl-1- DOSPA Cationic
    propanaminium trifluoroacetate
    1,2-Dioleyl-3-trimethylammonium-propane DOPA Cationic
    N-(2-Hydroxyethyl)-N,N-dimethyl-2,3-bis(tetradecyloxy)-1- MDRIE Cationic
    propanaminium bromide
    Dimyristooxypropyl dimethyl hydroxyethyl ammonium bromide DMRI Cationic
    3β-[N-(N′,N′-Dimethylaminoethane)-carbamoyl]cholesterol DC-Chol Cationic
    Bis-guanidium-tren-cholesterol BGTC Cationic
    1,3-Diodeoxy-2-(6-carboxy-spermyl)-propylamide DOSPER Cationic
    Dimethyloctadecylammonium bromide DDAB Cationic
    Dioctadecylamidoglicylspermidin DSL Cationic
    rac-[(2,3-Dioctadecyloxypropyl)(2-hydroxyethyl)]-dimethylammonium CLIP-1 Cationic
    chloride
    rac-[2(2,3-Dihexadecyloxypropyl- CLIP-6 Cationic
    oxymethyloxy)ethyl]trimethylammonium bromide
    Ethyldimyristoylphosphatidylcholine EDMPC Cationic
    1,2-Distearyloxy-N,N-dimethyl-3-aminopropane DSDMA Cationic
    1,2-Dimyristoyl-trimethylammonium propane DMTAP Cationic
    O,O′-Dimyristyl-N-lysyl aspartate DMKE Cationic
    1,2-Distearoyl-sn-glycero-3-ethylphosphocholine DSEPC Cationic
    N-Palmitoyl D-erythro-sphingosyl carbamoyl-spermine CCS Cationic
    N-t-Butyl-N0-tetradecyl-3-tetradecylaminopropionamidine diC14-amidine Cationic
    Octadecenolyoxy[ethyl-2-heptadecenyl-3 hydroxyethyl] imidazolinium DOTIM Cationic
    chloride
    N1-Cholesteryloxycarbonyl-3,7-diazanonane-1,9-diamine CDAN Cationic
    2-(3-[Bis(3-amino-propyl)-amino]propylamino)-N- RPR209120 Cationic
    ditetradecylcarbamoylme-ethyl-acetamide
    1,2-dilinoleyloxy-3-dimethylaminopropane DLinDMA Cationic
    2,2-dilinoleyl-4-dimethylaminoethyl-[1,3]-dioxolane DLin-KC2-DMA Cationic
    dilinoleyl-methyl-4-dimethylaminobutyrate DLin-MC3-DMA Cationic
  • Exemplary polymers for gene transfer are shown below in Table 7.
  • TABLE 7
    Polymers Used for Gene Transfer
    Polymer Abbreviation
    Poly(ethylene)glycol PEG
    Polyethylenimine PEI
    Dithiobis(succinimidylpropionate) DSP
    Dimethyl-3,3′-dithiobispropionimidate DTBP
    Poly(ethylene imine)biscarbamate PEIC
    Poly(L-lysine) PLL
    Histidine modified PLL
    Poly(N-vinylpyrrolidone) PVP
    Poly(propylenimine) PPI
    Poly(amidoamine) PAMAM
    Poly(amidoethylenimine) SS-PAEI
    Triethylenetetramine TETA
    Poly(β-aminoester)
    Poly(4-hydroxy-L-proline ester) PHP
    Poly(allylamine)
    Poly(α-[4-aminobutyl]-L-glycolic acid) PAGA
    Poly(D,L-lactic-co-glycolic acid) PLGA
    Poly(N-ethyl-4-vinylpyridinium bromide)
    Poly(phosphazene)s PPZ
    Poly(phosphoester)s PPE
    Poly(phosphoramidate)s PPA
    Poly(N-2-hydroxypropylmethacrylamide) pHPMA
    Poly (2-(dimethylamino)ethyl methacrylate) pDMAEMA
    Poly(2-aminoethyl propylene phosphate) PPE-EA
    Chitosan
    Galactosylated chitosan
    N-Dodacylated chitosan
    Histone
    Collagen
    Dextran-spermine D-SPM
  • In an embodiment, the vehicle has targeting modifications to increase target cell update of nanoparticles and liposomes, e.g., cell specific antigens, monoclonal antibodies, single chain antibodies, aptamers, polymers, sugars, and cell penetrating peptides. In an embodiment, the vehicle uses fusogenic and endosome-destabilizing peptides/polymers. In an embodiment, the vehicle undergoes acid-triggered conformational changes (e.g., to accelerate endosomal escape of the cargo). In an embodiment, a stimuli-cleavable polymer is used, e.g., for release in a cellular compartment. For example, disulfide-based cationic polymers that are cleaved in the reducing cellular environment can be used.
  • In an embodiment, the delivery vehicle is a biological non-viral delivery vehicle.
  • In an embodiment, one or more nucleic acid molecules (e.g., a DNA molecule or a nucleic acid template system) other than the components of a Cas9 system, e.g., the Cas9 fusion molecule component and/or the gRNA molecule component described herein, are delivered. In an embodiment, the nucleic acid molecule is delivered at the same time as one or more of the components of the Cas9 system are delivered. In an embodiment, the nucleic acid molecule is delivered before or after (e.g., less than about 30 minutes, 1 hour, 2 hours, 3 hours, 6 hours, 9 hours, 12 hours, 1 day, 2 days, 3 days, 1 week, 2 weeks, or 4 weeks) one or more of the components of the Cas9 system are delivered. In an embodiment, the nucleic acid molecule is delivered by a different means than one or more of the components of the Cas9 system, e.g., the Cas9 fusion molecule component and/or the gRNA molecule component, are delivered. The nucleic acid molecule can be delivered by any of the delivery methods described herein. For example, the nucleic acid molecule can be delivered by a viral vector, e.g., an integration-deficient lentivirus, and the Cas9 fusion molecule component and/or the gRNA molecule component can be delivered by electroporation, e.g., such that the toxicity caused by nucleic acids (e.g., DNA) is reduced. In an embodiment, the nucleic acid molecule encodes a therapeutic protein, e.g., a protein described herein. In an embodiment, the nucleic acid molecule encodes an RNA molecule, e.g., an RNA molecule described herein. In some embodiments, the nucleic acid is a nucleic acid template system capable of participating in HDR.
  • Delivery of RNA Encoding a Cas9 Fusion Molecule
  • RNA encoding Cas9 fusion molecules (e.g., mRNA encoding an eaCas9 fusion molecules), gRNA molecules, and/or template binding domains can be delivered into cells, or populations of cells, described herein, by any appropriate method, including art-known methods or methods described herein. For example, Cas9 fusion molecule-, gRNA-, and/or template binding domain-encoding RNA, e.g., mRNA can be delivered, e.g., by microinjection, electroporation, lipid-mediated transfection, peptide-mediated delivery, or a combination thereof.
  • Delivery of Cas9 Fusion Molecule Protein
  • Cas9 fusion molecules (e.g., eaCas9 fusion molecules) and/or template binding domains can be delivered into cells by any appropriate method, including art-known methods or methods described herein. For example, the protein molecules can be delivered, e.g., by microinjection, electroporation, lipid-mediated transfection, peptide-mediated delivery, or a combination thereof. Delivery can be accompanied by DNA encoding a gRNA, or by a gRNA, a nucleic acid template system, and/or DNA encoding a template binding domain partner.
  • Routes of Administration
  • Systemic modes of administration include oral and parenteral routes. Parenteral routes include, by way of example, intravenous, intrarterial, intraosseous, intramuscular, intradermal, subcutaneous, intranasal and intraperitoneal routes. Components administered systemically may be modified or formulated to target the components to the desired cell type.
  • Local modes of administration include, by way of example, intrathecal, intracerebroventricular, intraparenchymal (e.g., localized intraparenchymal delivery to the striatum (e.g., into the caudate or into the putamen)), cerebral cortex, precentral gyrus, hippocampus (e.g., into the dentate gyrus or CA3 region), temporal cortex, amygdala, frontal cortex, thalamus, cerebellum, medulla, hypothalamus, tectum, tegmentum or substantia nigra intraocular, intraorbital, subconjuctival, intravitreal, subretinal or transscleral routes. In an embodiment, significantly smaller amounts of the components (compared with systemic approaches) may exert an effect when administered locally (for example, intraparenchymal or intravitreal) compared to when administered systemically (for example, intravenously). Local modes of administration can reduce or eliminate the incidence of potentially toxic side effects that may occur when therapeutically effective amounts of a component are administered systemically.
  • Administration may be provided as a periodic bolus or as continuous infusion from an internal reservoir or from an external reservoir (for example, from an intravenous bag). Components may be administered locally, for example, by continuous release from a sustained release drug delivery device.
  • Bi-Modal or Differential Delivery of Components
  • Separate delivery of the components of a Cas9 system, e.g., the Cas9 fusion molecule component, the gRNA molecule component, the template binding domain, the template binding domain partner, and/or the nucleic acid acid template system, and more particularly, delivery of the components by differing modes, can enhance performance, e.g., by improving tissue specificity and safety.
  • In an embodiment, one or more of the Cas9 fusion molecule, the gRNA molecule, the template binding domain, the template binding domain partner, and/or the nucleic acid template system, are delivered by different modes, or as sometimes referred to herein as differential modes. Different or differential modes, as used herein, refer to modes of delivery that confer different pharmacodynamic or pharmacokinetic properties on the subject component molecule, e.g., a Cas9 fusion molecule, gRNA molecule, nucleic acid template system, template binding domain, and/or template binding domain partner. For example, the modes of delivery can result in different tissue distribution, different half-life, or different temporal distribution, e.g., in a selected compartment, tissue, or organ. In many embodiments, the components are delivered so that one or more of, e.g., all of, a Cas9 fusion molecule, gRNA molecule, nucleic acid template system, template binding domain, and template binding domain partner will be present in the same cell at the same time.
  • In some embodiments, two gRNAs are delivered to a cell so that a first nickase will make a first single stranded break and a second nickase will make a second single stranded break. In such embodiments, the two gRNAs and other components (e.g., the Cas9 fusion molecule) are delivered such that the two breaks are made at substantially the same time. In some embodiments this comprises the second break being formed before the first break engages with machinery specific to the SSBR (single stranded break repair) pathway, and in some embodiments, it comprises the second break being formed before the first break is repaired. More generally, when one desires to make two or more breaks in a target nucleic acid, the gRNAs and other components can be delivered such that the two or more breaks are made at substantially the same time.
  • Some modes of delivery, e.g., delivery by a nucleic acid vector that persists in a cell, or in progeny of a cell, e.g., by autonomous replication or insertion into cellular nucleic acid, result in more persistent expression of and presence of a component. Examples include viral, e.g., adeno-associated virus or lentivirus, delivery.
  • By way of example, the components, e.g., a Cas9 fusion molecule, a gRNA molecule, nucleic acid template system, template binding domain, and template binding domain partner can be delivered by modes that differ in terms of resulting half-life or persistent of the delivered component the body, or in a particular compartment, tissue or organ. In an embodiment, one or more of, e.g., all of, a gRNA molecule, nucleic acid template system, template binding domain, and template binding domain partner can be delivered by such modes. The Cas9 fusion molecule component can be delivered by a mode which results in less persistence or less exposure to the body or a particular compartment or tissue or organ.
  • More generally, in an embodiment, a first mode of delivery is used to deliver a first component and a second mode of delivery is used to deliver a second component. The first mode of delivery confers a first pharmacodynamic or pharmacokinetic property. The first pharmacodynamic property can be, e.g., distribution, persistence, or exposure, of the component, or of a nucleic acid that encodes the component, in the body, a compartment, tissue or organ. The second mode of delivery confers a second pharmacodynamic or pharmacokinetic property. The second pharmacodynamic property can be, e.g., distribution, persistence, or exposure, of the component, or of a nucleic acid that encodes the component, in the body, a compartment, tissue or organ.
  • In an embodiment, the first pharmacodynamic or pharmacokinetic property, e.g., distribution, persistence or exposure, is more limited than the second pharmacodynamic or pharmacokinetic property.
  • In an embodiment, the first mode of delivery is selected to optimize, e.g., minimize, a pharmacodynamic or pharmacokinetic property, e.g., distribution, persistence or exposure.
  • In an embodiment, the second mode of delivery is selected to optimize, e.g., maximize, a pharmacodynamic or pharmacokinetic property, e.g., distribution, persistence or exposure.
  • In an embodiment, the first mode of delivery comprises the use of a relatively persistent element, e.g., a nucleic acid, e.g., a plasmid or viral vector, e.g., an AAV or lentivirus. As such vectors are relatively persistent product transcribed from them would be relatively persistent.
  • In an embodiment, the second mode of delivery comprises a relatively transient element, e.g., an RNA or protein.
  • In an embodiment, the first component comprises a gRNA, template nucleic acid, template binding domain, or template binding domain partner and the delivery mode is relatively persistent, e.g., the gRNA is transcribed from a plasmid or viral vector, e.g., an AAV or lentivirus. Transcription of these genes would be of little physiological consequence because the genes generally do not encode for a protein product, and the gRNAs are incapable of acting in isolation. The second component, a Cas9 fusion molecule, is delivered in a transient manner, for example as mRNA or as protein, ensuring that the full Cas9 fusion molecule/gRNA molecule complex is only present and active for a short period of time.
  • Furthermore, the components can be delivered in different molecular form or with different delivery vectors that complement one another to enhance safety and tissue specificity.
  • Use of differential delivery modes can enhance performance, safety and efficacy. For example, the likelihood of an eventual off-target modification can be reduced. Delivery of immunogenic components, e.g., Cas9 fusion molecules, by less persistent modes can reduce immunogenicity, as peptides from the bacterially-derived Cas enzyme are displayed on the surface of the cell by MHC molecules. A two-part delivery system can alleviate these drawbacks.
  • Differential delivery modes can be used to deliver components to different, but overlapping target regions. The formation active complex is minimized outside the overlap of the target regions. Thus, in an embodiment, a first component, e.g., a gRNA molecule is delivered by a first delivery mode that results in a first spatial, e.g., tissue, distribution. A second component, e.g., a Cas9 fusion molecule is delivered by a second delivery mode that results in a second spatial, e.g., tissue, distribution. In an embodiment, the first mode comprises a first element selected from a liposome, nanoparticle, e.g., polymeric nanoparticle, and a nucleic acid, e.g., viral vector. The second mode comprises a second element selected from the group. In an embodiment, the first mode of delivery comprises a first targeting element, e.g., a cell specific receptor or an antibody, and the second mode of delivery does not include that element. In embodiment, the second mode of delivery comprises a second targeting element, e.g., a second cell specific receptor or second antibody.
  • When the Cas9 fusion molecule is delivered in a virus delivery vector, a liposome, or polymeric nanoparticle, there is the potential for delivery to and therapeutic activity in multiple tissues, when it may be desirable to only target a single tissue. A two-part delivery system can resolve this challenge and enhance tissue specificity. If the gRNA molecule and the Cas9 fusion molecule are packaged in separated delivery vehicles with distinct but overlapping tissue tropism, the fully functional complex is only formed in the tissue that is targeted by both vectors.
  • Ex Vivo Delivery
  • In some embodiments, components described in Table 4 are introduced into cells which are then introduced into the subject. Methods of introducing the components can include, e.g., any of the delivery methods described in Table 5.
  • In some embodiments, the cells are contacted with a Cas9 fusion molecule (or a nucleic acid encoding it) ex vivo. In some embodiments, the cells are contacted with a gRNA (or a nucleic acid encoding it) ex vivo. In some embodiment, the cells are contacted with a template nucleic acid ex vivo. In some embodiment, the cells are contacted with a template binding domain (or a nucleic acid encoding it) ex vivo. In some embodiment, the cells are contacted with a template binding domain partner ex vivo. In some embodiments, the cells are contacted with the nucleic acid template system ex vivo. In some embodiments, the cells are contacted with two, three, four, or all of the preceding compositions (or nucleic acids encoding them) ex vivo. In some embodiments, the cells are contacted with one or more of the preceding components (or nucleic acids encoding them), and one or more remaining components are administered to the patient.
  • XI. Modified Nucleosides, Nucleotides, and Nucleic Acids
  • Modified nucleosides and modified nucleotides can be present in nucleic acids, e.g., particularly gRNA, a template binding domain partner, and/or a template nucleic acid, but also other forms of DNA or RNA, e.g., mRNA. As described herein, “nucleoside” is defined as a compound containing a five-carbon sugar molecule (a pentose such as deoxyribose or ribose) or derivative thereof, and an organic base (purine or pyrimidine, or a derivative thereof). As described herein, “nucleotide” is defined as a nucleoside further comprising a phosphate group.
  • Modified nucleosides and nucleotides can include one or more of:
  • (i) alteration, e.g., replacement, of one or both of the non-linking phosphate oxygens and/or of one or more of the linking phosphate oxygens in the phosphodiester backbone linkage;
  • (ii) alteration, e.g., replacement, of a constituent of the ribose or deoxyribose sugar, e.g., of the 2′ hydroxyl on the sugar;
  • (iii) wholesale replacement of the phosphate moiety with “dephospho” linkers;
  • (iv) modification or replacement of a naturally occurring nucleobase;
  • (v) replacement or modification of the ribose- or deoxyribose-phosphate backbone;
  • (vi) modification of the 3′ end or 5′ end of the oligonucleotide, e.g., removal, modification or replacement of a terminal phosphate group or conjugation of a moiety; and
  • (vii) modification of the sugar.
  • The modifications listed above can be combined to provide modified nucleosides and nucleotides that can have two, three, four, or more modifications. For example, a modified nucleoside or nucleotide can have a modified sugar and a modified nucleobase. In an embodiment, every base of a gRNA, a template binding domain partner, or template nucleic acid is modified, e.g., all bases have a modified phosphate group, e.g., all are phosphorothioate groups. In an embodiment, all, or substantially all, of the phosphate groups of a unimolecular or modular gRNA molecule, a template binding domain partner, or template nucleic acid are replaced with phosphorothioate groups.
  • In an embodiment, modified nucleotides, e.g., nucleotides having modifications as described herein, can be incorporated into a nucleic acid, e.g., a “modified nucleic acid.” In some embodiments, the modified nucleic acids comprise one, two, three or more modified nucleotides. In some embodiments, at least 5% (e.g., at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or about 100%) of the positions in a modified nucleic acid are a modified nucleotides.
  • Unmodified nucleic acids can be prone to degradation by, e.g., cellular nucleases. For example, nucleases can hydrolyze nucleic acid phosphodiester bonds. Accordingly, in one aspect the modified nucleic acids described herein can contain one or more modified nucleosides or nucleotides, e.g., to introduce stability toward nucleases.
  • In some embodiments, the modified nucleosides, modified nucleotides, and modified nucleic acids described herein can exhibit a reduced innate immune response when introduced into a population of cells, both in vivo and ex vivo. The term “innate immune response” includes a cellular response to exogenous nucleic acids, including single stranded nucleic acids, generally of viral or bacterial origin, which involves the induction of cytokine expression and release, particularly the interferons, and cell death. In some embodiments, the modified nucleosides, modified nucleotides, and modified nucleic acids described herein can disrupt binding of a major groove interacting partner with the nucleic acid. In some embodiments, the modified nucleosides, modified nucleotides, and modified nucleic acids described herein can exhibit a reduced innate immune response when introduced into a population of cells, both in vivo and ex vivo, and also disrupt binding of a major groove interacting partner with the nucleic acid.
  • In some embodiments, a template nucleic acid comprises modifications, e.g., modified nucleotides, modifications to the backbone, and other modifications described herein. In some embodiments, the modification improves the stability of the template nucleic acid, e.g., by increasing its resistance to endonucleases and/or exonucleases.
  • In some embodiments, a template nucleic acid that comprises modifications is double stranded, e.g., is double stranded DNA. In some such embodiments, all the modifications are confined to one strand. In other embodiments, modifications are present on both strands. Modifications may be present in the 5′ homology arm, the 3′ homology arm, or the replacement sequence, or any combination thereof. In some embodiments, modifications are present in one or both homology arms but not the replacement sequence.
  • In some embodiments, a template nucleic acid that comprises modifications is single stranded, e.g., is single stranded DNA.
  • Definitions of Chemical Groups
  • As used herein, “alkyl” is meant to refer to a saturated hydrocarbon group which is straight-chained or branched. Example alkyl groups include methyl (Me), ethyl (Et), propyl (e.g., n-propyl and isopropyl), butyl (e.g., n-butyl, isobutyl, t-butyl), pentyl (e.g., n-pentyl, isopentyl, neopentyl), and the like. An alkyl group can contain from 1 to about 20, from 2 to about 20, from 1 to about 12, from 1 to about 8, from 1 to about 6, from 1 to about 4, or from 1 to about 3 carbon atoms.
  • As used herein, “aryl” refers to monocyclic or polycyclic (e.g., having 2, 3 or 4 fused rings) aromatic hydrocarbons such as, for example, phenyl, naphthyl, anthracenyl, phenanthrenyl, indanyl, indenyl, and the like. In some embodiments, aryl groups have from 6 to about 20 carbon atoms.
  • As used herein, “alkenyl” refers to an aliphatic group containing at least one double bond.
  • As used herein, “alkynyl” refers to a straight or branched hydrocarbon chain containing 2-12 carbon atoms and characterized in having one or more triple bonds. Examples of alkynyl groups include, but are not limited to, ethynyl, propargyl, and 3-hexynyl.
  • As used herein, “arylalkyl” or “aralkyl” refers to an alkyl moiety in which an alkyl hydrogen atom is replaced by an aryl group. Aralkyl includes groups in which more than one hydrogen atom has been replaced by an aryl group. Examples of “arylalkyl” or “aralkyl” include benzyl, 2-phenylethyl, 3-phenylpropyl, 9-fluorenyl, benzhydryl, and trityl groups.
  • As used herein, “cycloalkyl” refers to a cyclic, bicyclic, tricyclic, or polycyclic non-aromatic hydrocarbon groups having 3 to 12 carbons. Examples of cycloalkyl moieties include, but are not limited to, cyclopropyl, cyclopentyl, and cyclohexyl.
  • As used herein, “heterocyclyl” refers to a monovalent radical of a heterocyclic ring system. Representative heterocyclyls include, without limitation, tetrahydrofuranyl, tetrahydrothienyl, pyrrolidinyl, pyrrolidonyl, piperidinyl, pyrrolinyl, piperazinyl, dioxanyl, dioxolanyl, diazepinyl, oxazepinyl, thiazepinyl, and morpholinyl.
  • As used herein, “heteroaryl” refers to a monovalent radical of a heteroaromatic ring system. Examples of heteroaryl moieties include, but are not limited to, imidazolyl, oxazolyl, thiazolyl, triazolyl, pyrrolyl, furanyl, indolyl, thiophenyl pyrazolyl, pyridinyl, pyrazinyl, pyridazinyl, pyrimidinyl, indolizinyl, purinyl, naphthyridinyl, quinolyl, and pteridinyl.
  • Phosphate Backbone Modifications
  • The Phosphate Group
  • In some embodiments, the phosphate group of a modified nucleotide can be modified by replacing one or more of the oxygens with a different substituent. Further, the modified nucleotide, e.g., modified nucleotide present in a modified nucleic acid, can include the wholesale replacement of an unmodified phosphate moiety with a modified phosphate as described herein. In some embodiments, the modification of the phosphate backbone can include alterations that result in either an uncharged linker or a charged linker with unsymmetrical charge distribution.
  • Examples of modified phosphate groups include phosphorothioate, phosphoroselenates, borano phosphates, borano phosphate esters, hydrogen phosphonates, phosphoroamidates, alkyl or aryl phosphonates and phosphotriesters. In some embodiments, one of the non-bridging phosphate oxygen atoms in the phosphate backbone moiety can be replaced by any of the following groups: sulfur (S), selenium (Se), BR3 (wherein R can be, e.g., hydrogen, alkyl, or aryl), C (e.g., an alkyl group, an aryl group, and the like), H, NR2 (wherein R can be, e.g., hydrogen, alkyl, or aryl), or OR (wherein R can be, e.g., alkyl or aryl). The phosphorous atom in an unmodified phosphate group is achiral. However, replacement of one of the non-bridging oxygens with one of the above atoms or groups of atoms can render the phosphorous atom chiral; that is to say that a phosphorous atom in a phosphate group modified in this way is a stereogenic center. The stereogenic phosphorous atom can possess either the “R” configuration (herein Rp) or the “S” configuration (herein Sp).
  • Phosphorodithioates have both non-bridging oxygens replaced by sulfur. The phosphorus center in the phosphorodithioates is achiral which precludes the formation of oligoribonucleotide (or oligodeoxyribonucleotide) diastereomers. In some embodiments, modifications to one or both non-bridging oxygens can also include the replacement of the non-bridging oxygens with a group independently selected from S, Se, B, C, H, N, and OR (R can be, e.g., alkyl or aryl).
  • The phosphate linker can also be modified by replacement of a bridging oxygen, (i.e., the oxygen that links the phosphate to the nucleoside), with nitrogen (bridged phosphoroamidates), sulfur (bridged phosphorothioates) and carbon (bridged methylenephosphonates). The replacement can occur at either linking oxygen or at both of the linking oxygens.
  • Replacement of the Phosphate Group
  • The phosphate group can be replaced by non-phosphorus containing connectors. In some embodiments, the charge phosphate group can be replaced by a neutral moiety.
  • Examples of moieties which can replace the phosphate group can include, without limitation, e.g., methyl phosphonate, hydroxylamino, siloxane, carbonate, carboxymethyl, carbamate, amide, thioether, ethylene oxide linker, sulfonate, sulfonamide, thioformacetal, formacetal, oxime, methyleneimino, methylenemethylimino, methylenehydrazo, methylenedimethylhydrazo and methyleneoxymethylimino.
  • Replacement of the Ribophosphate Backbone
  • Scaffolds that can mimic nucleic acids can also be constructed wherein the phosphate linker and ribose sugar are replaced by nuclease resistant nucleoside or nucleotide surrogates. In some embodiments, the nucleobases can be tethered by a surrogate backbone. Examples can include, without limitation, the morpholino, cyclobutyl, pyrrolidine and peptide nucleic acid (PNA) nucleoside surrogates.
  • Sugar Modifications
  • The modified nucleosides and modified nucleotides can include one or more modifications to the sugar group. For example, the 2′ hydroxyl group (OH) can be modified or replaced with a number of different “oxy” or “deoxy” substituents. In some embodiments, modifications to the 2′ hydroxyl group can enhance the stability of the nucleic acid since the hydroxyl can no longer be deprotonated to form a 2′-alkoxide ion. The 2′-alkoxide can catalyze degradation by intramolecular nucleophilic attack on the linker phosphorus atom.
  • Examples of “oxy”-2′ hydroxyl group modifications can include alkoxy or aryloxy (OR, wherein “R” can be, e.g., alkyl, cycloalkyl, aryl, aralkyl, heteroaryl or a sugar); polyethyleneglycols (PEG), O(CH2CH2O)nCH2CH2OR wherein R can be, e.g., H or optionally substituted alkyl, and n can be an integer from 0 to 20 (e.g., from 0 to 4, from 0 to 8, from 0 to 10, from 0 to 16, from 1 to 4, from 1 to 8, from 1 to 10, from 1 to 16, from 1 to 20, from 2 to 4, from 2 to 8, from 2 to 10, from 2 to 16, from 2 to 20, from 4 to 8, from 4 to 10, from 4 to 16, and from 4 to 20). In some embodiments, the “oxy”-2′ hydroxyl group modification can include “locked” nucleic acids (LNA) in which the 2′ hydroxyl can be connected, e.g., by a C1-6 alkylene or C1-6 heteroalkylene bridge, to the 4′ carbon of the same ribose sugar, where exemplary bridges can include methylene, propylene, ether, or amino bridges; O-amino (wherein amino can be, e.g., NH2; alkylamino, dialkylamino, heterocyclyl, arylamino, diarylamino, heteroarylamino, or diheteroarylamino, ethylenediamine, or polyamino) and aminoalkoxy, O(CH2)n-amino, (wherein amino can be, e.g., NH2; alkylamino, dialkylamino, heterocyclyl, arylamino, diarylamino, heteroarylamino, or diheteroarylamino, ethylenediamine, or polyamino). In some embodiments, the “oxy”-2′ hydroxyl group modification can include the methoxyethyl group (MOE), (OCH2CH2OCH3, e.g., a PEG derivative).
  • “Deoxy” modifications can include hydrogen (i.e. deoxyribose sugars, e.g., at the overhang portions of partially ds RNA); halo (e.g., bromo, chloro, fluoro, or iodo); amino (wherein amino can be, e.g., NH2; alkylamino, dialkylamino, heterocyclyl, arylamino, diarylamino, heteroarylamino, diheteroarylamino, or amino acid); NH(CH2CH2NH)nCH2CH2-amino (wherein amino can be, e.g., as described herein), —NHC(O)R (wherein R can be, e.g., alkyl, cycloalkyl, aryl, aralkyl, heteroaryl or sugar), cyano; mercapto; alkyl-thio-alkyl; thioalkoxy; and alkyl, cycloalkyl, aryl, alkenyl and alkynyl, which may be optionally substituted with e.g., an amino as described herein.
  • The sugar group can also contain one or more carbons that possess the opposite stereochemical configuration than that of the corresponding carbon in ribose. Thus, a modified nucleic acid can include nucleotides containing e.g., arabinose, as the sugar. The nucleotide “monomer” can have an alpha linkage at the 1′ position on the sugar, e.g., alpha-nucleosides. The modified nucleic acids can also include “abasic” sugars, which lack a nucleobase at C-1′. These abasic sugars can also be further modified at one or more of the constituent sugar atoms. The modified nucleic acids can also include one or more sugars that are in the L form, e.g. L-nucleosides.
  • Generally, RNA includes the sugar group ribose, and DNA includes the sugar group deoxyribose, each of which is a 5-membered ring having an oxygen. Exemplary modified nucleosides and modified nucleotides can include, without limitation, replacement of the oxygen in the ribose or deoxyribose ring (e.g., with sulfur (S), selenium (Se), or alkylene, such as, e.g., methylene or ethylene); addition of a double bond (e.g., to replace ribose with cyclopentenyl or cyclohexenyl); ring contraction of ribose (e.g., to form a 4-membered ring of cyclobutane or oxetane); ring expansion of ribose (e.g., to form a 6- or 7-membered ring having an additional carbon or heteroatom, such as for example, anhydrohexitol, altritol, mannitol, cyclohexanyl, cyclohexenyl, and morpholino that also has a phosphoramidate backbone). In some embodiments, the modified nucleotides can include multicyclic forms (e.g., tricyclo; and “unlocked” forms, such as glycol nucleic acid (GNA) (e.g., R-GNA or S-GNA, where ribose is replaced by glycol units attached to phosphodiester bonds), threose nucleic acid (TNA, where ribose is replaced with α-L-threofuranosyl-(3′→2′)).
  • Modifications on the Nucleobase
  • The modified nucleosides and modified nucleotides described herein, which can be incorporated into a modified nucleic acid, can include a modified nucleobase. Examples of nucleobases include, but are not limited to, adenine (A), guanine (G), cytosine (C), and uracil (U). These nucleobases can be modified or wholly replaced to provide modified nucleosides and modified nucleotides that can be incorporated into modified nucleic acids. The nucleobase of the nucleotide can be independently selected from a purine, a pyrimidine, a purine or pyrimidine analog. In some embodiments, the nucleobase can include, for example, naturally-occurring and synthetic derivatives of a base.
  • Uracil
  • In some embodiments, the modified nucleobase is a modified uracil. Exemplary nucleobases and nucleosides having a modified uracil include without limitation pseudouridine (ψ), pyridin-4-one ribonucleoside, 5-aza-uridine, 6-aza-uridine, 2-thio-5-aza-uridine, 2-thio-uridine (s2U), 4-thio-uridine (s4U), 4-thio-pseudouridine, 2-thio-pseudouridine, 5-hydroxy-uridine (ho5U), 5-amino allyl-uridine, 5-halo-uridine (e.g., 5-iodo-uridine or 5-bromo-uridine), 3-methyl-uridine (m3U), 5-methoxy-uridine (mo5U), uridine 5-oxyacetic acid (cmo5U), uridine 5-oxyacetic acid methyl ester (mcmo5U), 5-carboxymethyl-uridine (cm U), 1-carboxymethyl-pseudouridine, 5-carboxyhydroxymethyl-uridine (chm5U), 5-carboxyhydroxymethyl-uridine methyl ester (mchm5U), 5-methoxycarbonylmethyl-uridine (mcm5U), 5-methoxycarbonylmethyl-2-thio-uridine (mcm5s2U), 5-aminomethyl-2-thio-uridine (nm5s2U), 5-methylaminomethyl-uridine (mnm5U), 5-methylaminomethyl-2-thio-uridine (mnm5s2U), 5-methylaminomethyl-2-seleno-uridine (mnm5se2U), 5-carbamoylmethyl-uridine (ncm5U), 5-carboxymethylaminomethyl-uridine (cmnm5U), 5-carboxymethylaminomethyl-2-thio-uridine (cmnm5s2U), 5-propynyl-uridine, 1-propynyl-pseudouridine, 5-taurinomethyl-uridine (τcm5U), 1-taurinomethyl-pseudouridine, 5-taurinomethyl-2-thio-uridine(τm5 s2U), 1-taurinomethyl-4-thio-pseudouridine, 5-methyl-uridine (m5U, i.e., having the nucleobase deoxythymine), 1-methyl-pseudouridine (m1ψ), 5-methyl-2-thio-uridine (m5s2U), 1-methyl-4-thio-pseudouridine (m1s4ψ), 4-thio-1-methyl-pseudouridine, 3-methyl-pseudouridine (m3ψ), 2-thio-1-methyl-pseudouridine, 1-methyl-1-deaza-pseudouridine, 2-thio-1-methyl-1-deaza-pseudouridine, dihydrouridine (D), dihydropseudouridine, 5,6-dihydrouridine, 5-methyl-dihydrouridine (m5D), 2-thio-dihydrouridine, 2-thio-dihydropseudouridine, 2-methoxy-uridine, 2-methoxy-4-thio-uridine, 4-methoxy-pseudouridine, 4-methoxy-2-thio-pseudouridine, N1-methyl-pseudouridine, 3-(3-amino-3-carboxypropyl)uridine (acp3U), 1-methyl-3-(3-amino-3-carboxypropyl)pseudouridine (acp3ψ), 5-(isopentenylaminomethyl)uridine (inm5U), 5-(isopentenylaminomethyl)-2-thio-uridine (inm5s2U), α-thio-uridine, 2′-O-methyl-uridine (Um), 5,2′-O-dimethyl-uridine (m5Um), 2′-O-methyl-pseudouridine (ψm), 2-thio-2′-O-methyl-uridine (s2Um), 5-methoxycarbonylmethyl-2′-O-methyl-uridine (mcm5Um), 5-carbamoylmethyl-2′-O-methyl-uridine (ncm5Um), 5-carboxymethylaminomethyl-2′-O-methyl-uridine (cmnm5Um), 3,2′-O-dimethyl-uridine (m3Um), 5-(isopentenylaminomethyl)-2′-O-methyl-uridine (inm5Um), 1-thio-uridine, deoxythymidine, 2′-F-ara-uridine, 2′-F-uridine, 2′-OH-ara-uridine, 5-(2-carbomethoxyvinyl) uridine, 5-[3-(1-E-propenylamino)uridine, pyrazolo[3,4-d]pyrimidines, xanthine, and hypoxanthine.
  • Thymine
  • In some embodiments, the modified nucleobase is a modified thymine. Thymine differs from uracil in that thymine has a methyl group on carbon 5 of the 6-carbon ring, while uracil has a hydrogen in that position. In some embodiments, the modified thymine is derived from one of the modified uracils described in the previous paragraph, but having said methyl group instead of a hydrogen.
  • Cytosine
  • In some embodiments, the modified nucleobase is a modified cytosine. Exemplary nucleobases and nucleosides having a modified cytosine include without limitation 5-aza-cytidine, 6-aza-cytidine, pseudoisocytidine, 3-methyl-cytidine (m3C), N4-acetyl-cytidine (act), 5-formyl-cytidine (f5C), N4-methyl-cytidine (m4C), 5-methyl-cytidine (m5C), 5-halo-cytidine (e.g., 5-iodo-cytidine), 5-hydroxymethyl-cytidine (hm5C), 1-methyl-pseudoisocytidine, pyrrolo-cytidine, pyrrolo-pseudoisocytidine, 2-thio-cytidine (s2C), 2-thio-5-methyl-cytidine, 4-thio-pseudoisocytidine, 4-thio-1-methyl-pseudoisocytidine, 4-thio-1-methyl-1-deaza-pseudoisocytidine, 1-methyl-1-deaza-pseudoisocytidine, zebularine, 5-aza-zebularine, 5-methyl-zebularine, 5-aza-2-thio-zebularine, 2-thio-zebularine, 2-methoxy-cytidine, 2-methoxy-5-methyl-cytidine, 4-methoxy-pseudoisocytidine, 4-methoxy-1-methyl-pseudoisocytidine, lysidine (k2C), α-thio-cytidine, 2′-O-methyl-cytidine (Cm), 5,2′-O-dimethyl-cytidine (m5Cm), N4-acetyl-2′-O-methyl-cytidine (ac4Cm), N4,2′-O-dimethyl-cytidine (m4Cm), 5-formyl-2′-O-methyl-cytidine (f5Cm), N4,N4,2′-O-trimethyl-cytidine (m4 2Cm), 1-thio-cytidine, 2′-F-ara-cytidine, 2′-F-cytidine, and 2′-OH-ara-cytidine.
  • Adenine
  • In some embodiments, the modified nucleobase is a modified adenine. Exemplary nucleobases and nucleosides having a modified adenine include without limitation 2-amino-purine, 2,6-diaminopurine, 2-amino-6-halo-purine (e.g., 2-amino-6-chloro-purine), 6-halo-purine (e.g., 6-chloro-purine), 2-amino-6-methyl-purine, 8-azido-adenosine, 7-deaza-adenine, 7-deaza-8-aza-adenine, 7-deaza-2-amino-purine, 7-deaza-8-aza-2-amino-purine, 7-deaza-2,6-diaminopurine, 7-deaza-8-aza-2,6-diaminopurine, 1-methyl-adenosine (m A), 2-methyl-adenine (m2A), N6-methyl-adeno sine (m6A), 2-methylthio-N6-methyl-adeno sine (ms2m6A), N6-isopentenyl-adeno sine (i6A), 2-methylthio-N6-isopentenyl-adeno sine (ms2i6A), N6-(cis-hydroxyisopentenyl)adenosine (io6A), 2-methylthio-N6-(cis-hydroxyisopentenyl)adeno sine (ms2io6A), N6-glycinylcarbamoyl-adeno sine (g6A), N6-threonylcarbamoyl-adeno sine (t6A), N6-methyl-N6-threonylcarbamoyl-adenosine (m6t6A), 2-methylthio-N6-threonylcarbamoyl-adenosine (ms2g6A), N6,N6-dimethyl-adenosine (m6 2A), N6-hydroxynorvalylcarbamoyl-adenosine (hn6A), 2-methylthio-N6-hydroxynorvalylcarbamoyl-adenosine (ms2hn6A), N6-acetyl-adenosine (ac6A), 7-methyl-adenine, 2-methylthio-adenine, 2-methoxy-adenine, α-thio-adenosine, 2′-O-methyl-adenosine (Am), N6,2′-O-dimethyl-adenosine (m6Am), N6-Methyl-2′-deoxyadenosine, N6,N6,2′-O-trimethyl-adenosine (m6 2Am), 1,2′-O-dimethyl-adenosine (m1Am), 2′-O-ribosyladenosine (phosphate) (Ar(p)), 2-amino-N6-methyl-purine, 1-thio-adenosine, 8-azido-adenosine, 2′-F-ara-adenosine, 2′-F-adenosine, 2′-OH-ara-adenosine, and N6-(19-amino-pentaoxanonadecyl)-adenosine.
  • Guanine
  • In some embodiments, the modified nucleobase is a modified guanine. Exemplary nucleobases and nucleosides having a modified guanine include without limitation inosine (I), 1-methyl-inosine (m1I), wyosine (imG), methylwyosine (mimG), 4-demethyl-wyosine (imG-14), isowyosine (imG2), wybutosine (yW), peroxywybutosine (o2yW), hydroxywybutosine (OHyW), undermodified hydroxywybutosine (OHyW*), 7-deaza-guanosine, queuosine (Q), epoxyqueuosine (oQ), galactosyl-queuosine (galQ), mannosyl-queuosine (manQ), 7-cyano-7-deaza-guanosine (preQ0), 7-aminomethyl-7-deaza-guanosine (preQ1), archaeosine (G+), 7-deaza-8-aza-guanosine, 6-thio-guanosine, 6-thio-7-deaza-guanosine, 6-thio-7-deaza-8-aza-guanosine, 7-methyl-guanosine (m7G), 6-thio-7-methyl-guanosine, 7-methyl-inosine, 6-methoxy-guanosine, 1-methyl-guanosine (m′G), N2-methyl-guanosine (m2G), N2,N2-dimethyl-guanosine (m2 2G), N2,7-dimethyl-guano sine (m2,7G), N2,N2,7-dimethyl-guanosine (m2,2,7G), 8-oxo-guanosine, 7-methyl-8-oxo-guanosine, 1-meth thio-guanosine, N2-methyl-6-thio-guanosine, N2,N2-dimethyl-6-thio-guanosine, α-thio-guanosine, 2′-O-methyl-guanosine (Gm), N2-methyl-2′-O-methyl-guanosine (m2Gm), N2,N2-dimethyl-2′-O-methyl-guanosine (m2 2Gm), 1-methyl-2′-O-methyl-guanosine (m′Gm), N2,7-dimethyl-2′-O-methyl-guanosine (m2,7Gm), 2′-O-methyl-inosine (Im), 1,2′-O-dimethyl-inosine (m′Im), O6-phenyl-2′-deoxyinosine, 2′-O-ribosylguanosine (phosphate) (Gr(p)), 1-thio-guanosine, O6-methyl-guanosine, O6-Methyl-2′-deoxyguanosine, 2′-F-ara-guanosine, and 2′-F-guanosine.
  • Modified gRNAs
  • In some embodiments, the modified nucleic acids can be modified gRNAs. In some embodiments, gRNAs can be modified at the 3′ end. In this embodiment, the gRNAs can be modified at the 3′ terminal U ribose. For example, the two terminal hydroxyl groups of the U ribose can be oxidized to aldehyde groups and a concomitant opening of the ribose ring to afford a modified nucleoside as shown below:
  • Figure US20180250424A1-20180906-C00001
  • wherein “U” can be an unmodified or modified uridine.
  • In another embodiment, the 3′ terminal U can be modified with a 2′3′ cyclic phosphate as shown below:
  • Figure US20180250424A1-20180906-C00002
  • wherein “U” can be an unmodified or modified uridine.
  • In some embodiments, the gRNA molecules may contain 3′ nucleotides which can be stabilized against degradation, e.g., by incorporating one or more of the modified nucleotides described herein. In this embodiment, e.g., uridines can be replaced with modified uridines, e.g., 5-(2-amino)propyl uridine, and 5-bromo uridine, or with any of the modified uridines described herein; adenosines and guanosines can be replaced with modified adenosines and guanosines, e.g., with modifications at the 8-position, e.g., 8-bromo guanosine, or with any of the modified adenosines or guanosines described herein. In some embodiments, deaza nucleotides, e.g., 7-deaza-adenosine, can be incorporated into the gRNA. In some embodiments, O- and N-alkylated nucleotides, e.g., N6-methyl andenosine, can be incorporated into the gRNA. In some embodiments, sugar-modified ribonucleotides can be incorporated, e.g., wherein the 2′ OH-group is replaced by a group selected from H, —OR, —R (wherein R can be, e.g., alkyl, cycloalkyl, aryl, aralkyl, heteroaryl or sugar), halo, —SH, —SR (wherein R can be, e.g., alkyl, cycloalkyl, aryl, aralkyl, heteroaryl or sugar), amino (wherein amino can be, e.g., NH2; alkylamino, dialkylamino, heterocyclyl, arylamino, diarylamino, heteroarylamino, diheteroarylamino, or amino acid); or cyano (—CN). In some embodiments, the phosphate backbone can be modified as described herein, e.g., with a phosphothioate group. In some embodiments, the nucleotides in the overhang region of the gRNA can each independently be a modified or unmodified nucleotide including, but not limited to 2′-sugar modified, such as, 2-F 2′-O-methyl, thymidine (T), 2′-O-methoxyethyl-5-methyluridine (Teo), 2′-O-methoxyethyladenosine (Aeo), 2′-O-methoxyethyl-5-methylcytidine (m5Ceo), and any combinations thereof.
  • In an embodiment, one or more or all of the nucleotides in single stranded RNA molecule, e.g., a gRNA molecule, are deoxynucleotides.
  • Modified Nucleic Acid Template Systems
  • In some embodiments, the nucleic acid template system comprises chemical modifications. These modifications may, e.g., increase the stability or half-life of the nucleic acid or reduce the innate immune response to the nucleic acid. In some embodiments, the template binding domain partner comprises chemical modifications; in some embodiments the template nucleic acid comprises chemical modifications; and in some embodiments, both the template binding domain partner and the template nucleic acid comprise chemical modifications.
  • In some embodiments, the nucleic acid template system can be modified at one or two 3′ ends. In this embodiment, the nucleic acid template system can be modified at the 3′ nucleotide. For example, the two terminal hydroxyl groups of the 3′-most sugar can be oxidized to aldehyde groups and a concomitant opening of the ring to afford a modified nucleoside, analogous to the first ribonucleotide shown in the previous section entitled “Modified gRNAs”.
  • In another embodiment, the 3′ terminal sugar can be modified with a 2′3′ cyclic phosphate, analogous to the second ribonucleotide shown in the previous section entitled “Modified gRNAs”.
  • In some embodiments, the nucleic acid template system may contain 3′ nucleotides which can be stabilized against degradation, e.g., by incorporating one or more of the modified nucleotides described herein. In this embodiment, e.g., thymines can be replaced with any of the modified thymines described herein; adenosines and guanosines can be replaced with modified adenosines and guanosines, e.g., with modifications at the 8-position, e.g., 8-bromo guanosine, or with any of the modified adenosines or guanosines described herein. In some embodiments, deaza nucleotides, e.g., 7-deaza-adenosine, can be incorporated into the nucleic acid template system. In some embodiments, O- and N-alkylated nucleotides, e.g., N6-methyl andenosine, can be incorporated into the nucleic acid template system. In some embodiments, sugar-modified deoxyribonucleotides can be incorporated, e.g., wherein the 2′ H-group is replaced by a group selected from OH, —OR, —R (wherein R can be, e.g., alkyl, cycloalkyl, aryl, aralkyl, heteroaryl or sugar), halo, —SH, —SR (wherein R can be, e.g., alkyl, cycloalkyl, aryl, aralkyl, heteroaryl or sugar), amino (wherein amino can be, e.g., NH2; alkylamino, dialkylamino, heterocyclyl, arylamino, diarylamino, heteroarylamino, diheteroarylamino, or amino acid); or cyano (—CN). In some embodiments, the phosphate backbone can be modified as described herein, e.g., with a phosphothioate group. In some embodiments, the nucleic acid template system comprises an overhang region, and the nucleotides in the overhang region can each independently be a modified or unmodified nucleotide including, but not limited to 2′-sugar modified, such as, 2-F 2′-O-methyl, 2′-O-methoxyethyl-5-methyluridine (Teo), 2′-O-methoxyethyladenosine (Aeo), 2′-O-methoxyethyl-5-methylcytidine (m5Ceo), and any combinations thereof.
  • miRNA Binding Sites
  • MicroRNAs (miRNAs) are naturally occurring cellular 19-25 nucleotide long noncoding RNAs. They bind to nucleic acid molecules having an appropriate miRNA binding site, e.g., in the 3′ UTR of an mRNA, and down-regulate gene expression. While not wishing to be bound by theory it is believed that the down regulation is either by reducing nucleic acid molecule stability or by inhibiting translation. An RNA species disclosed herein, e.g., an mRNA encoding Cas9 fusion molecule can comprise an miRNA binding site, e.g., in its 3′UTR. The miRNA binding site can be selected to promote down regulation of expression is a selected cell type. By way of example, the incorporation of a binding site for miR-122, a microRNA abundant in liver, can inhibit the expression of the gene of interest in the liver.
  • XII. Nucleic Acids; Kits; Methods of Production
  • In some aspects, disclosed herein is a nucleic acid, e.g., an isolated or non-naturally occurring nucleic acid, e.g., DNA, that comprises a sequence that encodes a gRNA molecule comprising a targeting domain that is complementary with a target domain as disclosed herein. In an embodiment, the nucleic acid encodes a gRNA molecule, e.g., a first gRNA molecule, comprising a targeting domain configured to provide a cleavage event, e.g., a double strand break or a single strand break, sufficiently close to a region desired to be altered to allow alteration, e.g., alteration associated with HDR of the region desired to be altered.
  • A nucleic acid disclosed herein may comprise (a) a sequence that encodes a gRNA molecule comprising a targeting domain that is complementary with a target domain as disclosed herein; (b) a sequence that encodes a Cas9 fusion molecule; and further comprises (c) (i) a sequence that encodes a second gRNA molecule described herein having a targeting domain that is complementary to a second target domain, and optionally, (ii) a sequence that encodes a third gRNA molecule described herein having a targeting domain that is complementary to a third target domain; and optionally, (iii) a sequence that encodes a fourth gRNA molecule described herein having a targeting domain that is complementary to a fourth target domain.
  • In some embodiments, when a region that is desired to be altered is corrected by HDR, the nucleic acid encodes (a) a sequence that encodes a gRNA molecule comprising a targeting domain that is complementary with a target domain gene as disclosed herein; (b) a sequence that encodes a Cas9 fusion molecule; (c) a template nucleic acid, (d) a template binding domain, and (e) a template binding domain partner.
  • In some embodiments, one or more of, e.g., all of, (a), (b), (c), (d), and (e) are present on the same nucleic acid molecule, e.g., the same vector, e.g., the same viral vector, e.g., the same adeno-associated virus (AAV) vector. In an embodiment, the nucleic acid molecule is an AAV vector. Exemplary AAV vectors that may be used in any of the described compositions and methods include an AAV2 vector, a modified AAV2 vector, an AAV3 vector, a modified AAV3 vector, an AAV6 vector, a modified AAV6 vector, an AAV8 vector and an AAV9 vector.
  • In other embodiments, a subset of (a), (b), (c), (d), and (e) is present on a first nucleic acid molecule, e.g. a first vector, e.g., a first viral vector, e.g., a first AAV vector; and the remainder of (a), (b), (c), (d), and (e) is present on a second nucleic acid molecule, e.g., a second vector, e.g., a second vector, e.g., a second AAV vector. The first and second nucleic acid molecules may be AAV vectors. In many embodiments, the Cas9 fusion molecule of (b) and the template binding domain of (d) are present on the same nucleic acid molecule, e.g., a viral vector, e.g., an AAV vector. In many embodiments, the template nucleic acid of (c) and the template binding domain partner of (e) are present on the same nucleic acid molecule, e.g., a viral vector, e.g., an AAV vector.
  • In some embodiments, all of (a), (b), (c), (d), and (e) are on the same vector, e.g., the same AAV vector. In some embodiments, all of (a), (b), (c), (d), and (e) are on different vectors, e.g., one or more AAV vectors. In some embodiments, two or more, e.g., two, three, or four of (a), (b), (c), (d), and (e) are on the same vector, e.g., an AAV vector, and the remainder are on one or more other vectors, e.g., one or more AAV vectors. In some embodiments, (a) and (b) are on the same vector, e.g., an AAV vector; (a) and (c) are on the same vector, e.g., an AAV vector, (a) and (d) are on the same vector, e.g., an AAV vector, (b) and (c) are on the same vector, e.g., an AAV vector, (b) and (d) are on the same vector, e.g., an AAV vector, or (c) and (d) are on the same vector, e.g., an AAV vector.
  • The nucleic acids described herein may comprise a promoter operably linked to the sequence that encodes the gRNA molecule of (a), e.g., a promoter described herein. The nucleic acid may further comprise a second promoter operably linked to the sequence that encodes the second, third and/or fourth gRNA molecule of (e), e.g., a promoter described herein. The promoter and second promoter differ from one another. In some embodiments, the promoter and second promoter are the same.
  • The nucleic acids described herein may further comprise a promoter operably linked to the sequence that encodes the Cas9 fusion molecule of (b), e.g., a promoter described herein.
  • The nucleic acids described herein may further comprise a promoter operably linked to the sequence that encodes the template binding domain of (d), e.g., a promoter described herein. In some embodiments, the template binding domain of (d) and the Cas9 fusion molecule of (b) are controlled by the same promoter, e.g., they can form a fusion protein.
  • In another aspect, disclosed herein is a kit comprising one or more, e.g., all of the following:
  • (a) gRNA molecule described herein, or nucleic acid that encodes the gRNA;
  • (b) a Cas9 fusion molecule, e.g., a Cas9 fusion molecule described herein, or a nucleic acid or mRNA that encodes the Cas9 fusion molecule;
  • (c) a template nucleic acid; and
  • (d) a template binding domain, and
  • (e) a template binding domain partner.
  • In an embodiment, the kit comprises a nucleic acid comprising both (c) and (e).
  • In an embodiment, the kit comprises nucleic acid, e.g., an AAV vector, that encodes one or more of (a), (b), (c), (d), and (e).
  • XIII. Methods of Treatment
  • A genetic disease is caused by a mutation in the patient's genome. Often, the mutation results in a change in a protein, e.g., an amino acid substitution or a truncation. Genetic diseases can be dominant, i.e., one mutant gene is sufficient to cause the disease, or recessive, where a patient with one copy of the mutant gene is an asymptomatic carrier, and two copies of the mutant gene are necessary for the disease to result.
  • Disclosed herein are the approaches to treat or prevent genetic diseases, using the compositions and methods described herein.
  • One approach to treat or prevent genetic diseases is to repair (i.e., correct) one or more mutations in the disease-causing gene by HDR. In this approach, mutant allele(s) are corrected and restored to wild type state. While not wishing to be bound by theory, it is believed that correction of the mutation to the corresponding wild-type sequence restores wild type protein production within the relevant cell type. The method described herein can be performed in all cell types.
  • In an embodiment, one mutant allele is repaired in the subject. For example, in a patient with an autosomal dominant genetic disease, the sole mutant allele in the cell is corrected so that the cell becomes wild-type at both loci. As another example, in a patient with an autosomal recessive genetic disease, one of the two mutant alleles in the cell is corrected, and so the cell becomes heterozygous, which is sufficient for normal functioning. As a recessive genetic disease only displays a phenotype when both alleles are mutated, repair of a single allele is adequate for a cure. In another embodiment, both mutant alleles are repaired in the subject. In either situation, the subjects can be cured of disease.
  • Correction of a mutation in the relevant gene may be performed prior to disease onset (e.g., prior to the appearance of symptoms) or after disease onset, for instance, early in the disease course.
  • In an embodiment, the method comprises initiating treatment of a subject prior to disease onset. In an embodiment, the method comprises initiating treatment of a subject after disease onset. In an embodiment, the method comprises initiating treatment of a subject well after disease onset, e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 24, or 36 months after onset of the disease. While not wishing to be bound by theory it is believed that this may be effective if subjects did not present to physician until well into the course of illness.
  • In an embodiment, the method comprises initiating treatment of a subject in an advanced stage of disease.
  • Overall, initiation of treatment for subjects at all stages of disease is expected to prevent negative consequences of disease and be of benefit to subjects.
  • In an embodiment, the method comprises initiating treatment of a subject prior to disease expression. In an embodiment, the method comprises initiating treatment of a subject in an early stage of disease, e.g., when a subject has tested positive for the disease but has no signs or symptoms associated with the disease.
  • In an embodiment, the method comprises initiating treatment of a subject who has tested positive for the mutation underlying the disease, based on diagnosis via electrophoresis, genotyping, family history or other diagnostic criteria.
  • EXAMPLES
  • The following Examples are merely illustrative and are not intended to limit the scope or content of the invention in any way.
  • Example 1: Enhancing HDR-Mediated Alteration of a Target Nucleic Acid in Cultured Cells Using a Cas9 Fusion Protein
  • This study demonstrates an increased rate of HDR-mediated nucleic acid modification at a target site in cultured cells by administering to the cells a Cas9 fusion protein, one or more gRNAs, and an HDR donor nucleic acid template. The Cas9 fusion protein is a Cas9 protein fused to a polypeptide comprising the DNA binding domain derived from a sequence-specific DNA binding protein, e.g., one of the DNA binding proteins listed in Tables V.1-V.4 of Section V, above. The Cas9 fusion protein (or, in control cells, Cas9 protein lacking the fusion domain) is administered as a DNA expression vector, an mRNA or a protein. The one or more gRNAs are administered as DNA expression vectors or RNA molecules. The donor template is provided as a plasmid DNA, a linear double-stranded DNA, a single-stranded oligonucleotide or viral genomic DNA, as described above, e.g., in FIGS. 1-4. The donor template contains one or more copies of a nucleotide sequence that can be bound by the sequence-specific DNA binding domain that is fused to the Cas9 polypeptide. The Cas9 fusion protein (or control Cas9), gRNAs and donor template are introduced into the cells by methods known in the art, such as viral transduction, non-viral transfection or electroporation.
  • At various time points, cells are harvested in order to assess the rate of nucleic acid modification by HDR. This is achieved by 1) isolating genomic DNA from the Cas9 control and Cas9 fusion protein treated cells; 2) PCR amplifying the DNA encompassing the region targeted for modification; 3) sequencing the amplified DNA products; and 4) determining the frequency of HDR-mediated alteration by dividing the number of sequence reads containing the donor template-specified sequence by the total number of sequence reads comprising the targeted region. Treatment of cells with a Cas9 fusion protein is shown to increase the rate of HDR-mediated nucleic acid modification.
  • Example 2: Enhancing HDR-Mediated Alteration of a Target Nucleic Acid in a Tissue of an Animal Using a Cas9 Fusion Protein
  • This study demonstrates an increased rate of HDR-mediated nucleic acid modification at a target site in a tissue of an animal by administering to the animal a Cas9 fusion protein, one or more gRNAs, and a nucleic acid template system, e.g., an HDR donor nucleic acid template. The Cas9 fusion protein is a Cas9 protein fused to a polypeptide comprising the DNA binding domain derived from a sequence-specific DNA binding protein, e.g. one of the DNA binding proteins listed in Tables V.1-V.4 of Section V, above. The Cas9 fusion protein (or, in a control cohort of animals, Cas9 protein lacking the fusion domain) is administered as a DNA expression vector, an mRNA or a protein. The one or more gRNAs are administered as DNA expression vectors or RNA molecules. The donor template is provided as a plasmid DNA, a linear double-stranded DNA, a single-stranded oligonucleotide or viral genomic DNA, as described above, e.g., in FIGS. 1-4. The donor template contains one or more copies of a nucleotide sequence that can be bound by the sequence-specific DNA binding domain that is fused to the Cas9 polypeptide. The Cas9 fusion protein (or control Cas9), gRNAs and nucleic acid template system are introduced into the animal by local or systemic administration of viral vectors or non-viral delivery vehicles.
  • At various time points, tissues are harvested in order to assess the rate of nucleic acid modification by HDR. This is achieved by 1) isolating genomic DNA from the Cas9 control and Cas9 fusion protein treated cells; 2) PCR amplifying the DNA encompassing the region targeted for modification; 3) sequencing the amplified DNA products; and 4) determining the frequency of HDR-mediated alteration by dividing the number of sequence reads containing the donor template-specified sequence by the total number of sequence reads comprising the targeted region. Treatment of cells with a Cas9 fusion protein is shown to increase the rate of HDR-mediated nucleic acid modification.
  • Example 3: Constructing a Cas9 Fusion Protein
  • To generate a Cas9 fusion protein, established molecular biology techniques are used to ligate a nucleotide sequence encoding a DNA binding domain derived from a sequence-specific DNA binding protein, e.g., one of the DNA binding proteins listed in Tables V.1-V.4 of Section V, above, in-frame to a nucleotide sequence encoding a Cas9 protein. A nucleotide sequence encoding a linker peptide may be inserted in-frame between the nucleotide sequences encoding the Cas9 protein and the DNA binding domain. For expression in bacteria, cultured cells, or animal tissues, the nucleotide sequence encoding the Cas9-DNA binding domain fusion molecule is operably linked to one or more transcriptional control elements, e.g., promoter and/or enhancer elements, which enable expression in the relevant bacteria, cultured cells, or animal tissue. The Cas9 fusion protein can be purified from the bacteria, cultured cells, or animal tissue using established biochemical techniques. To generate mRNA encoding the Cas9 fusion protein, the nucleotide sequence encoding the Cas9-DNA binding domain fusion is operably linked to a promoter, e.g., a bacteriophage promoter, e.g., a T7 RNA polymerase promoter enabling in vitro transcription of mRNA encoding the Cas9-DNA binding domain fusion molecule.
  • Example 4: Optimization of the Tet Repressor Binding Domain
  • In this study, the Tet Repressor (TetR) Operon system was selected the optimal composition of the template binding domain partner assessed to achieve strong binding and ensure sufficient length of a locus-specific DNA donor.
  • The nucleotide sequence to which the Tet Repressor protein binds, typically consists of several repeats (5 or more) of the Tet Operator (TetO) 19 base pair sequence, each followed by a 17 bp spacer. Here, we evaluated the binding of the Tet repressor protein to several Tet Operator sequence-containing variants that included spacer sequences of differing sizes, or no spacer sequences. Specifically, we investigated whether only 3 repeats of the Tet Operator sequence were sufficient for binding of the Tet repressor protein, and whether shrinking the spacer sequence would still allow for efficient Tet repressor protein binding. The conditions tested (including the sequences) are indicated in Table 8, and visually depicted in FIG. 8A.
  • TABLE 8
    Template
    DNA ID Description Sequence
    ssODN-179 ssODN-179 TGCTTCTGACACAACTGTGTTCACTAGCAA
    (control) CCTCAAACAGACACCATGGTGCATCTGACT
    CGTGTGGAGAAGTCGGCCGTTACTGCCCTG
    CAGGGCAAGCTTAACGTGGATGAAGTTCGT
    GGTGAGGCCCTGGGCAGGTTGGTATCAAGG
    TTACAAGACAGGTTTAAGGAGACCAATAG
    ssODN-109 ssODN-109 AACAGACACCATGGTGCATCTGACTCGTGT
    (control) GGAGAAGTCGGCCGTTACTGCCCTGCAGGG
    CAAGCTTAACGTGGATGAAGTTCGTGGTGA
    GGCCCTGGGCAGGTTGGTA
    V1.1 (3xTetO, 17 bp TCCCTATCAGTGATAGAGAACGTATGTCGA
    spacer)-ssODN-109 GTTTACTCCCTATCAGTGATAGAGAACGTA
    TGTCGAGTTTACTCCCTATCAGTGATAGAG
    AAACAGACACCATGGTGCATCTGACTCGTG
    TGGAGAAGTCGGCCGTTACTGCCCTGCAGG
    GCAAGCTTAACGTGGATGAAGTTCGTGGTG
    AGGCCCTGGGCAGGTTGGTA
    V1.2 ssODN-109-(3xTetO, AACAGACACCATGGTGCATCTGACTCGTGT
    17 bp spacer) GGAGAAGTCGGCCGTTACTGCCCTGCAGGG
    CAAGCTTAACGTGGATGAAGTTCGTGGTGA
    GGCCCTGGGCAGGTTGGTATCCCTATCAGT
    GATAGAGAACGTATGTCGAGTTTACTCCCT
    ATCAGTGATAGAGAACGTATGTCGAGTTTA
    CTCCCTATCAGTGATAGAGA
    V2 (3xTetO, 8 bp TCCCTATCAGTGATAGAGAACGTATGTTCC
    spacer)-ssODN-109 CTATCAGTGATAGAGAACGTATGTTCCCTA
    TCAGTGATAGAGAACGTATGTAACAGACAC
    CATGGTGCATCTGACTCGTGTGGAGAAGTC
    GGCCGTTACTGCCCTGCAGGGCAAGCTTAA
    CGTGGATGAAGTTCGTGGTGAGGCCCTGGG
    CAGGTTGGTA
    V3 (3xTetO, no spacer)- TCCCTATCAGTGATAGAGATCCCTATCAGT
    ssODN-109 GATAGAGATCCCTATCAGTGATAGAGAAAC
    AGACACCATGGTGCATCTGACTCGTGTGGA
    GAAGTCGGCCGTTACTGCCCTGCAGGGCAA
    GCTTAACGTGGATGAAGTTCGTGGTGAGGC
    CCTGGGCAGGTTGGTA
  • To test the binding capacity of the Tet Operator sequence-containing variants experimentally, 500 nM of recombinant TetR protein (Imgen BioSciences, Inc.; Cat. No. P-1002-0.5 mg) was incubated with 50 nM of template DNA in binding buffer (50 mM HEPES, 20% glycerol, 300 mM NaCl, pH 7.7) and protein binding to the Tet Operator sequence-containing variants was analyzed using an electrophoretic mobility shift assay using a 10% polyacrylamide gel (FIG. 8B).
  • As expected, no gel shift was observed for control templates lacking the Tet operator sequence (ssODN-179, ssODN109). In contrast, complete gel shift (indicating binding of Tet repressor protein to the Tet Operator sequence) was observed for templates V1.1, V1.2, and V2. Moreover, partial gel shift was observed for template V3, indicating some, but not complete, binding of TetR protein. These data indicate that Tet Operator sequences separated by a shorter 8 bp spacer are indeed functional, and that three Tet Operator repeats are sufficient for Tet Repressor protein binding to occur. Based on these results, novel templates, with shortened spacer sequences (8 bp spacer sequences), were designed, which allowed for increased donor template sequence length (by 10 bp, see V4.1 and V4.2), or for the inclusion of an additional Tet Operator sequence attached to the donor template sequence (V5.1 and V5.2), as shown in Table 9 and FIG. 9.
  • Based on these findings, Cas9 fusion molecules comprising a Cas9 molecule, e.g., wild type Cas9 molecule, linked to a Tet Repressor molecule, may be generated using established molecular biology techniques. In addition, a nucleic acid template system comprising a template binding domain partner comprising, e.g., three Tet Operator sequences, organized in one or more configurations described above, and a template nucleic acid, may also be generated. U20S cells may be electroporated with 200 ng of gRNA targeting the HBB endogenous locus (gRNA #8 (GTAACGGCAGACTTCTCCTC) and gRNA #15 (AAGGTGAACGTGGATGAAGT)), 750 ng of plasmid encoding a Cas9 fusion molecule, comprising, e.g., a wild type Cas9 or mutant Cas9 (D10A or N863A) fused to wild type Tet Repressor molecule, or to a mutant Tet Repressor molecule as a negative control. Simultaneously, the cells may also be electroporated with 25 pmol of the aforementioned optimized single stranded oligo (SSODN). Cells are collected 6 days after electroporation and genomic DNA extracted. PCR amplification of the HBB locus is performed and amplicons are subcloned into Topo Blunt Vector. For each condition, 96 colonies are sequenced using Sanger sequencing and homology-directed repair efficacy is assessed.
  • TABLE 9
    Template ID Description Sequence
    V4.1 3xTetO (8bp spacer)- TCCCTATCAGTGATAGAGAACGTATGTTCCC
    ssODN-119 TATCAGTGATAGAGAACGTATGTTCCCTATC
    AGTGATAGAGAACGTATGTCCTCAAACAGA
    CACCATGGTGCATCTGACTCGTGTGGAGAAG
    TCGGCCGTTACTGCCCTGCAGGGCAAGCTTA
    ACGTGGATGAAGTTCGTGGTGAGGCCCTGG
    GCAGGTTGGTATCAAG
    V4.2 ssODN-119-3xTetO CCTCAAACAGACACCATGGTGCATCTGACTC
    (8bp spacer) GTGTGGAGAAGTCGGCCGTTACTGCCCTGCA
    GGGCAAGCTTAACGTGGATGAAGTTCGTGG
    TGAGGCCCTGGGCAGGTTGGTATCAAGTCCC
    TATCAGTGATAGAGAACGTATGTTCCCTATC
    AGTGATAGAGAACGTATGTTCCCTATCAGTG
    ATAGAGAACGTATGT
    V5.1 (4xTetO (8bp spacer)- TCCCTATCAGTGATAGAGAACGTATGTTCCC
    ssODN-100 TATCAGTGATAGAGAACGTATGTTCCCTATC
    AGTGATAGAGAACGTATGTTCCCTATCAGTG
    ATAGAGAACACCATGGTGCATCTGACTCGTG
    TGGAGAAGTCGGCCGTTACTGCCCTGCAGG
    GCAAGCTTAACGTGGATGAAGTTCGTGGTG
    AGGCCCTGGGCAGGT
    V5.2 ssODN-100-4xTetO ACACCATGGTGCATCTGACTCGTGTGGAGAA
    (8bp spacer) GTCGGCCGTTACTGCCCTGCAGGGCAAGCTT
    AACGTGGATGAAGTTCGTGGTGAGGCCCTG
    GGCAGGTTCCCTATCAGTGATAGAGAACGT
    ATGTTCCCTATCAGTGATAGAGAACGTATGT
    TCCCTATCAGTGATAGAGAACGTATGTTCCC
    TATCAGTGATAGAGA
  • Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described below. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting.
  • Headings, including numeric and alphabetical headings and subheadings, are for organization and presentation and are not intended to be limiting.
  • INCORPORATION BY REFERENCE
  • All publications, patents, and patent applications mentioned herein are hereby incorporated by reference in their entirety as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference. In case of conflict, the present application, including any definitions herein, will control.
  • EQUIVALENTS
  • Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.
  • Other embodiments are within the following claims.

Claims (52)

1. A Cas9 fusion molecule comprising:
a Cas9 molecule linked to
a template binding domain.
2. The Cas9 fusion molecule of claim 1, wherein the template binding domain comprises a specific affinity for a template binding domain partner.
3.-9. (canceled)
10. The Cas9 fusion molecule of claim 1, wherein the Cas9 molecule is covalently or non-covalently linked to the template binding domain.
11.-17. (canceled)
18. The Cas9 fusion molecule of claim 1, wherein said Cas9 fusion molecule comprises at least two template binding domains.
19. The Cas9 fusion molecule of claim 1, wherein the template binding domain comprises a protein, a nucleic acid, or a small molecule.
20. (canceled)
21. The Cas9 fusion molecule of claim 1, wherein the template binding domain comprises a protein, and wherein the protein comprises a DNA binding domain.
22. The Cas9 fusion molecule of claim 21, wherein the protein comprises a repressor protein, or a fragment of a repressor protein.
23. The Cas9 fusion molecule of claim 22, wherein the repressor protein, or the fragment of the repressor protein, is a TetR repressor, or a fragment of the TetR repressor; a LacI repressor, or a fragment of the LacI repressor; a Gal4 repressor, or a fragment of the Gal4 repressor; or a repressor protein C1, or a fragment of the repressor protein C1.
24. The Cas9 fusion molecule of claim 1, further comprising a linker between the Cas9 molecule and the template binding domain.
25.-34. (canceled)
35. The Cas9 fusion molecule of claim 1, wherein the Cas9 molecule is an eaCas9 molecule or an eiCas9 molecule.
36.-51. (canceled)
52. A Cas9 system comprising a Cas9 fusion molecule of claim 1 and a nucleic acid template system comprising:
a template binding domain partner and
a template nucleic acid.
53. The Cas9 system of claim 52, wherein the template binding domain of the Cas9 fusion molecule is bound to the template binding domain partner.
54. The Cas9 system of claim 53, wherein the template binding domain of the Cas9 fusion molecule is covalently or non-covalently bound to the template binding domain partner.
55. (canceled)
56. The Cas9 system of claim 52, wherein the template binding domain partner is linked to the template nucleic acid.
57.-65. (canceled)
66. The Cas9 system of claim 52, wherein the template binding domain comprises a protein, a small molecule, or a nucleic acid, and wherein the template binding domain partner comprises a small molecule, a protein, or a nucleic acid.
67.-75. (canceled)
76. The Cas9 system of claim 52, wherein the template binding domain partner is a DNA sequence recognized by a DNA binding protein.
77. The Cas9 system of claim 76, wherein the DNA sequence recognized by the DNA binding protein is selected from a Tet-O sequence, a Lac operon O1 sequence, a UAS sequence, or an Operator L and R sequence.
78. (canceled)
79. The Cas9 system of claim 76, wherein the DNA binding protein comprises a TetR repressor, or a fragment of the TetR repressor, and the DNA comprises at least one Tet-O sequence.
80.-93. (canceled)
94. The Cas9 system of claim 52, wherein the nucleic acid template system comprises a double stranded nucleic acid sequence or a single stranded nucleic acid sequence.
95.-106. (canceled)
107. The Cas9 system of claim 52, wherein the template nucleic acid comprises about 50-500 nucleotides of homology with a target nucleic acid.
108.-114. (canceled)
115. The Cas9 system of claim 52, further comprising a gRNA.
116.-117. (canceled)
118. A cell, or a population of cells, comprising the Cas9 system of claim 52.
119. A cell, or a population of cells, comprising the Cas9 fusion molecule of claim 1.
120. A nucleic acid encoding the Cas9 fusion molecule of claim 1.
121. A vector comprising a nucleic acid of claim 120.
122.-123. (canceled)
124. A method of altering a nucleic acid at a target position in a cell, or a population of cells, the method comprising contacting the cell with the Cas9 system of claim 115,
wherein the gRNA molecule and Cas9 fusion molecule interact with the nucleic acid, resulting in a cleavage event, wherein the cleavage event is repaired by at least one DNA repair pathway, and
wherein the sequence of the nucleic acid after the cleavage event is different than the sequence of the nucleic acid prior to the cleavage event,
thereby altering the nucleic acid at the target position in the cell, or in the population of cells.
125. The method of claim 124, further comprising contacting the cell, or the population of cells, with a second gRNA molecule,
wherein the second gRNA molecule and the Cas9 fusion molecule interact with the nucleic acid, resulting in a second cleavage event.
126.-145. (canceled)
146. The method of claim 125, wherein the cell, or population of cells, is from a subject suffering from a disease or disorder selected from the group consisting of a blood disease, an immune disease, a neurological disease, a cancer, an infectious disease, a genetic disease, a disorder caused by aberrant mtDNA, a metabolic disease, a disorder caused by aberrant cell cycle, a disorder caused by aberrant angiogenesis, a disorder cause by aberrant DNA damage repair, or a pain disorder.
147. (canceled)
148. The method of claim 125, wherein the cell, or population of cells, is from a subject having at least one mutation at the target position.
149.-156. (canceled)
157. A cell, or a population of cells, altered by the method of claim 125.
158. A pharmaceutical composition comprising the cell, or the population of cells, of claim 157.
159. A pharmaceutical composition comprising the Cas9 system of claim 115.
160. (canceled)
161. A method of treating a subject suffering from a disease or disorder, the method comprising contacting a cell, or a population of cells, from the subject with the Cas9 system of claim 115,
wherein the gRNA molecule and the Cas9 fusion molecule interact with a nucleic acid at a target position, resulting in a cleavage event,
wherein the cleavage event is repaired by at least one DNA repair pathway, and
wherein the sequence of the nucleic acid after the cleavage event is different than the sequence of the nucleic acid prior to the cleavage event,
thereby treating the subject suffering from the disease or disorder.
162.-164. (canceled)
US15/518,105 2014-10-10 2015-10-09 Compositions and methods for promoting homology directed repair Abandoned US20180250424A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/518,105 US20180250424A1 (en) 2014-10-10 2015-10-09 Compositions and methods for promoting homology directed repair

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201462062815P 2014-10-10 2014-10-10
US201462068371P 2014-10-24 2014-10-24
US15/518,105 US20180250424A1 (en) 2014-10-10 2015-10-09 Compositions and methods for promoting homology directed repair
PCT/US2015/055002 WO2016057961A1 (en) 2014-10-10 2015-10-09 Compositions and methods for promoting homology directed repair

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2015/055002 A-371-Of-International WO2016057961A1 (en) 2014-10-10 2015-10-09 Compositions and methods for promoting homology directed repair

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US17/393,575 Division US20220072160A1 (en) 2014-10-10 2021-08-04 Compositions and methods for promoting homology directed repair

Publications (1)

Publication Number Publication Date
US20180250424A1 true US20180250424A1 (en) 2018-09-06

Family

ID=54540174

Family Applications (2)

Application Number Title Priority Date Filing Date
US15/518,105 Abandoned US20180250424A1 (en) 2014-10-10 2015-10-09 Compositions and methods for promoting homology directed repair
US17/393,575 Pending US20220072160A1 (en) 2014-10-10 2021-08-04 Compositions and methods for promoting homology directed repair

Family Applications After (1)

Application Number Title Priority Date Filing Date
US17/393,575 Pending US20220072160A1 (en) 2014-10-10 2021-08-04 Compositions and methods for promoting homology directed repair

Country Status (5)

Country Link
US (2) US20180250424A1 (en)
EP (1) EP3204496A1 (en)
AU (2) AU2015330699B2 (en)
CA (1) CA2963693A1 (en)
WO (1) WO2016057961A1 (en)

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20180127785A1 (en) * 2015-05-14 2018-05-10 University Of Southern California Optimized gene editing utilizing a recombinant endonuclease system
CN111004816A (en) * 2019-11-28 2020-04-14 中国农业大学 Exogenous gene site-directed knock-in method based on Cas12a technology
US10704021B2 (en) 2012-03-15 2020-07-07 Flodesign Sonics, Inc. Acoustic perfusion devices
US10785574B2 (en) 2017-12-14 2020-09-22 Flodesign Sonics, Inc. Acoustic transducer driver and controller
WO2020198675A1 (en) * 2019-03-27 2020-10-01 Research Institute At Nationwide Children's Hospital Generation of chimeric antigen receptor (car)-primary nk cells for cancer immunotherapy using a combination of cas9/rnp and aav viruses
CN112154211A (en) * 2018-03-25 2020-12-29 基因系链公司 Modified nucleic acid editing system for binding donor DNA
WO2021016043A1 (en) * 2019-07-19 2021-01-28 Inari Agriculture, Inc. Improved homology dependent repair genome editing
US10975368B2 (en) 2014-01-08 2021-04-13 Flodesign Sonics, Inc. Acoustophoresis device with dual acoustophoretic chamber
CN113667017A (en) * 2021-07-28 2021-11-19 上海南方模式生物科技股份有限公司 Method capable of improving homologous recombination efficiency of CRISPR/Cas9 system and application
WO2021252498A1 (en) * 2020-06-12 2021-12-16 New England Biolabs, Inc. Compositions and methods for detecting molecular targets on chromosomal dna
US11214789B2 (en) 2016-05-03 2022-01-04 Flodesign Sonics, Inc. Concentration and washing of particles with acoustics
US11236313B2 (en) 2016-04-13 2022-02-01 Editas Medicine, Inc. Cas9 fusion molecules, gene editing systems, and methods of use thereof
US11268144B2 (en) * 2015-09-24 2022-03-08 Sigma-Aldrich Co. Llc Methods and reagents for molecular proximity detection using RNA-guided nucleic acid binding proteins
US11377651B2 (en) 2016-10-19 2022-07-05 Flodesign Sonics, Inc. Cell therapy processes utilizing acoustophoresis
US11597924B2 (en) 2016-03-25 2023-03-07 Editas Medicine, Inc. Genome editing systems comprising repair-modulating enzyme molecules and methods of their use
US11667911B2 (en) 2015-09-24 2023-06-06 Editas Medicine, Inc. Use of exonucleases to improve CRISPR/CAS-mediated genome editing
US11680268B2 (en) 2014-11-07 2023-06-20 Editas Medicine, Inc. Methods for improving CRISPR/Cas-mediated genome-editing
US11708572B2 (en) 2015-04-29 2023-07-25 Flodesign Sonics, Inc. Acoustic cell separation techniques and processes
US11866726B2 (en) 2017-07-14 2024-01-09 Editas Medicine, Inc. Systems and methods for targeted integration and genome editing and detection thereof using integrated priming sites

Families Citing this family (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP6261500B2 (en) 2011-07-22 2018-01-17 プレジデント アンド フェローズ オブ ハーバード カレッジ Evaluation and improvement of nuclease cleavage specificity
SG11201508028QA (en) 2013-04-16 2015-10-29 Regeneron Pharma Targeted modification of rat genome
US20150044192A1 (en) 2013-08-09 2015-02-12 President And Fellows Of Harvard College Methods for identifying a target site of a cas9 nuclease
US9359599B2 (en) 2013-08-22 2016-06-07 President And Fellows Of Harvard College Engineered transcription activator-like effector (TALE) domains and uses thereof
US9340799B2 (en) 2013-09-06 2016-05-17 President And Fellows Of Harvard College MRNA-sensing switchable gRNAs
US9388430B2 (en) 2013-09-06 2016-07-12 President And Fellows Of Harvard College Cas9-recombinase fusion proteins and uses thereof
US9526784B2 (en) 2013-09-06 2016-12-27 President And Fellows Of Harvard College Delivery system for functional nucleases
JP6174811B2 (en) 2013-12-11 2017-08-02 リジェネロン・ファーマシューティカルズ・インコーポレイテッドRegeneron Pharmaceuticals, Inc. Methods and compositions for targeted genomic modification
US9840699B2 (en) 2013-12-12 2017-12-12 President And Fellows Of Harvard College Methods for nucleic acid editing
WO2016022363A2 (en) 2014-07-30 2016-02-11 President And Fellows Of Harvard College Cas9 proteins including ligand-dependent inteins
RU2706965C2 (en) 2014-10-15 2019-11-21 Регенерон Фармасьютикалз, Инк. Methods and compositions for producing or maintaining pluripotent cells
RU2734770C2 (en) 2014-11-21 2020-10-23 Регенерон Фармасьютикалз, Инк. Methods and compositions for targeted genetic modification using paired guide rnas
CA2969847A1 (en) 2014-12-10 2016-06-16 Regents Of The University Of Minnesota Genetically modified cells, tissues, and organs for treating disease
US10190106B2 (en) * 2014-12-22 2019-01-29 Univesity Of Massachusetts Cas9-DNA targeting unit chimeras
EP3328399B1 (en) 2015-07-31 2023-12-27 Regents of the University of Minnesota Modified cells and methods of therapy
US20190225955A1 (en) 2015-10-23 2019-07-25 President And Fellows Of Harvard College Evolved cas9 proteins for gene editing
CN107513538A (en) * 2016-06-17 2017-12-26 北京大学 Gene knockout method
BR112019000055A2 (en) * 2016-07-05 2019-04-02 The Johns Hopkins University compositions and methods comprising enhancements to the crispr guide using the h1 promoter
KR102547316B1 (en) 2016-08-03 2023-06-23 프레지던트 앤드 펠로우즈 오브 하바드 칼리지 Adenosine nucleobase editing agents and uses thereof
AU2017308889B2 (en) 2016-08-09 2023-11-09 President And Fellows Of Harvard College Programmable Cas9-recombinase fusion proteins and uses thereof
US11542509B2 (en) 2016-08-24 2023-01-03 President And Fellows Of Harvard College Incorporation of unnatural amino acids into proteins using base editing
KR20240007715A (en) 2016-10-14 2024-01-16 프레지던트 앤드 펠로우즈 오브 하바드 칼리지 Aav delivery of nucleobase editors
WO2018075664A1 (en) 2016-10-18 2018-04-26 Regents Of The University Of Minnesota Tumor infiltrating lymphocytes and methods of therapy
US20180245065A1 (en) 2016-11-01 2018-08-30 Novartis Ag Methods and compositions for enhancing gene editing
WO2018118585A1 (en) * 2016-12-22 2018-06-28 Agenovir Corporation Antiviral compositions
US10745677B2 (en) 2016-12-23 2020-08-18 President And Fellows Of Harvard College Editing of CCR5 receptor gene to protect against HIV infection
TW201839136A (en) 2017-02-06 2018-11-01 瑞士商諾華公司 Compositions and methods for the treatment of hemoglobinopathies
US11898179B2 (en) 2017-03-09 2024-02-13 President And Fellows Of Harvard College Suppression of pain by gene editing
EP3592777A1 (en) 2017-03-10 2020-01-15 President and Fellows of Harvard College Cytosine to guanine base editor
US11268082B2 (en) 2017-03-23 2022-03-08 President And Fellows Of Harvard College Nucleobase editors comprising nucleic acid programmable DNA binding proteins
US11834670B2 (en) * 2017-04-19 2023-12-05 Global Life Sciences Solutions Usa Llc Site-specific DNA modification using a donor DNA repair template having tandem repeat sequences
CN110785179A (en) * 2017-04-21 2020-02-11 西雅图儿童医院(Dba西雅图儿童研究所) Therapeutic genome editing in Wiskott-Aldrich syndrome and X-linked thrombocytopenia
US11560566B2 (en) 2017-05-12 2023-01-24 President And Fellows Of Harvard College Aptazyme-embedded guide RNAs for use with CRISPR-Cas9 in genome editing and transcriptional activation
WO2019006418A2 (en) 2017-06-30 2019-01-03 Intima Bioscience, Inc. Adeno-associated viral vectors for gene therapy
WO2019003193A1 (en) 2017-06-30 2019-01-03 Novartis Ag Methods for the treatment of disease with gene editing systems
CN111183226A (en) 2017-07-11 2020-05-19 西格马-奥尔德里奇有限责任公司 Use of nucleosome interacting protein domains to enhance targeted genomic modifications
WO2019023680A1 (en) 2017-07-28 2019-01-31 President And Fellows Of Harvard College Methods and compositions for evolving base editors using phage-assisted continuous evolution (pace)
WO2019139645A2 (en) 2017-08-30 2019-07-18 President And Fellows Of Harvard College High efficiency base editors comprising gam
US11795443B2 (en) 2017-10-16 2023-10-24 The Broad Institute, Inc. Uses of adenosine base editors
US20210180053A1 (en) 2017-11-01 2021-06-17 Novartis Ag Synthetic rnas and methods of use
JP2021523745A (en) 2018-05-16 2021-09-09 シンテゴ コーポレイション Methods and systems for guide RNA design and use
WO2019232494A2 (en) * 2018-06-01 2019-12-05 Synthego Corporation Methods and systems for determining editing outcomes from repair of targeted endonuclease mediated cuts
AU2020206997A1 (en) * 2019-01-07 2021-08-26 Crisp-Hr Therapeutics, Inc. A non-toxic Cas9 enzyme and application thereof
CN111718418B (en) * 2019-03-19 2021-08-27 华东师范大学 Fusion protein for enhancing gene editing and application thereof
CA3130488A1 (en) 2019-03-19 2020-09-24 David R. Liu Methods and compositions for editing nucleotide sequences
WO2020187272A1 (en) * 2019-03-19 2020-09-24 上海邦耀生物科技有限公司 Fusion protein for gene therapy and application thereof
GB2614813A (en) 2020-05-08 2023-07-19 Harvard College Methods and compositions for simultaneous editing of both strands of a target double-stranded nucleotide sequence

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8993233B2 (en) * 2012-12-12 2015-03-31 The Broad Institute Inc. Engineering and optimization of systems, methods and compositions for sequence manipulation with functional domains

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BR112012014080A2 (en) 2009-12-10 2015-10-27 Univ Iowa State Res Found method for modifying genetic material, method for generating a nucleic acid, effector endonuclease monomer such, method for generating an aninal, method for generating a plant, method for directed genetic recombination, nucleic acid, expression cassette, and host cell
EP3741868A1 (en) * 2013-03-15 2020-11-25 The General Hospital Corporation Rna-guided targeting of genetic and epigenomic regulatory proteins to specific genomic loci
US20160237455A1 (en) 2013-09-27 2016-08-18 Editas Medicine, Inc. Crispr-related methods and compositions

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8993233B2 (en) * 2012-12-12 2015-03-31 The Broad Institute Inc. Engineering and optimization of systems, methods and compositions for sequence manipulation with functional domains
US8999641B2 (en) * 2012-12-12 2015-04-07 The Broad Institute Inc. Engineering and optimization of systems, methods and compositions for sequence manipulation with functional domains

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10704021B2 (en) 2012-03-15 2020-07-07 Flodesign Sonics, Inc. Acoustic perfusion devices
US10975368B2 (en) 2014-01-08 2021-04-13 Flodesign Sonics, Inc. Acoustophoresis device with dual acoustophoretic chamber
US11680268B2 (en) 2014-11-07 2023-06-20 Editas Medicine, Inc. Methods for improving CRISPR/Cas-mediated genome-editing
US11708572B2 (en) 2015-04-29 2023-07-25 Flodesign Sonics, Inc. Acoustic cell separation techniques and processes
US11535871B2 (en) * 2015-05-14 2022-12-27 University Of Southern California Optimized gene editing utilizing a recombinant endonuclease system
US20180127785A1 (en) * 2015-05-14 2018-05-10 University Of Southern California Optimized gene editing utilizing a recombinant endonuclease system
US11667911B2 (en) 2015-09-24 2023-06-06 Editas Medicine, Inc. Use of exonucleases to improve CRISPR/CAS-mediated genome editing
US11268144B2 (en) * 2015-09-24 2022-03-08 Sigma-Aldrich Co. Llc Methods and reagents for molecular proximity detection using RNA-guided nucleic acid binding proteins
US11597924B2 (en) 2016-03-25 2023-03-07 Editas Medicine, Inc. Genome editing systems comprising repair-modulating enzyme molecules and methods of their use
US11236313B2 (en) 2016-04-13 2022-02-01 Editas Medicine, Inc. Cas9 fusion molecules, gene editing systems, and methods of use thereof
US11214789B2 (en) 2016-05-03 2022-01-04 Flodesign Sonics, Inc. Concentration and washing of particles with acoustics
US11377651B2 (en) 2016-10-19 2022-07-05 Flodesign Sonics, Inc. Cell therapy processes utilizing acoustophoresis
US11866726B2 (en) 2017-07-14 2024-01-09 Editas Medicine, Inc. Systems and methods for targeted integration and genome editing and detection thereof using integrated priming sites
US10785574B2 (en) 2017-12-14 2020-09-22 Flodesign Sonics, Inc. Acoustic transducer driver and controller
CN112154211A (en) * 2018-03-25 2020-12-29 基因系链公司 Modified nucleic acid editing system for binding donor DNA
US11339385B2 (en) * 2018-03-25 2022-05-24 GeneTether, Inc. Modified nucleic acid editing systems for tethering donor DNA
WO2020198675A1 (en) * 2019-03-27 2020-10-01 Research Institute At Nationwide Children's Hospital Generation of chimeric antigen receptor (car)-primary nk cells for cancer immunotherapy using a combination of cas9/rnp and aav viruses
WO2021016043A1 (en) * 2019-07-19 2021-01-28 Inari Agriculture, Inc. Improved homology dependent repair genome editing
CN111004816A (en) * 2019-11-28 2020-04-14 中国农业大学 Exogenous gene site-directed knock-in method based on Cas12a technology
US11492667B2 (en) 2020-06-12 2022-11-08 New England Biolabs, Inc. Compositions and methods for detecting molecular targets on chromosomal DNA
WO2021252498A1 (en) * 2020-06-12 2021-12-16 New England Biolabs, Inc. Compositions and methods for detecting molecular targets on chromosomal dna
CN113667017A (en) * 2021-07-28 2021-11-19 上海南方模式生物科技股份有限公司 Method capable of improving homologous recombination efficiency of CRISPR/Cas9 system and application

Also Published As

Publication number Publication date
CA2963693A1 (en) 2016-04-14
WO2016057961A1 (en) 2016-04-14
US20220072160A1 (en) 2022-03-10
EP3204496A1 (en) 2017-08-16
AU2015330699A1 (en) 2017-05-11
AU2015330699B2 (en) 2021-12-02
AU2022201038A1 (en) 2022-03-10
WO2016057961A8 (en) 2016-05-12

Similar Documents

Publication Publication Date Title
US20220072160A1 (en) Compositions and methods for promoting homology directed repair
US20230026726A1 (en) Crispr/cas-related methods and compositions for treating sickle cell disease
US11597924B2 (en) Genome editing systems comprising repair-modulating enzyme molecules and methods of their use
US20190062734A1 (en) Grna fusion molecules, gene editing systems, and methods of use thereof
US20220325326A1 (en) Evaluation of cas9 molecule/guide rna molecule complexes
US20230025279A1 (en) Crispr/cas-related methods and compositions for treating leber's congenital amaurosis 10 (lca10)
US20210380987A1 (en) Crispr/cas-related methods and compositions for treating cystic fibrosis
EP3132030B1 (en) Crispr-cas-related methods, compositions and components for cancer immunotherapy
AU2022202645A1 (en) Methods for improving crispr/cas-mediated genome-editing
US20170007679A1 (en) Crispr/cas-related methods and compositions for treating hiv infection and aids
EP3540061A1 (en) Crispr/cas-related methods and compositions for treating primary open angle glaucoma
AU2016326711A1 (en) Use of exonucleases to improve CRISPR/Cas-mediated genome editing
WO2015148860A1 (en) Crispr/cas-related methods and compositions for treating beta-thalassemia
US20240132877A1 (en) Genome editing systems comprising repair-modulating enzyme molecules and methods of their use

Legal Events

Date Code Title Description
AS Assignment

Owner name: EDITAS MEDICINE, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:COTTA-RAMUSINO, CECILIA;REEL/FRAME:042301/0611

Effective date: 20151015

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: ADVISORY ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

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