[go: up one dir, main page]

WO2025101679A1 - Effector proteins and uses thereof - Google Patents

Effector proteins and uses thereof Download PDF

Info

Publication number
WO2025101679A1
WO2025101679A1 PCT/US2024/054826 US2024054826W WO2025101679A1 WO 2025101679 A1 WO2025101679 A1 WO 2025101679A1 US 2024054826 W US2024054826 W US 2024054826W WO 2025101679 A1 WO2025101679 A1 WO 2025101679A1
Authority
WO
WIPO (PCT)
Prior art keywords
sequence
nucleic acid
effector protein
composition
effector
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.)
Pending
Application number
PCT/US2024/054826
Other languages
French (fr)
Inventor
Adam L. Garske
Stepan TYMOSHENKO
Shiaki Arnett MINAMI
Rohan Grover
Fnu YUNANDA
Yasaman HEIDARIAN
Paula GONCALVES CERQUEIRA
Stacey Akie Shiigi BOYARSKIY
Srijay RAJAN
Benjamin Julius RAUCH
Aaron DELOUGHERY
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.)
Mammoth Biosciences Inc
Original Assignee
Mammoth Biosciences Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Mammoth Biosciences Inc filed Critical Mammoth Biosciences Inc
Publication of WO2025101679A1 publication Critical patent/WO2025101679A1/en
Pending legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/102Mutagenizing nucleic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/90Stable introduction of foreign DNA into chromosome
    • 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 [RNase]; Deoxyribonucleases [DNase]
    • C12N9/222Clustered regularly interspaced short palindromic repeats [CRISPR]-associated [CAS] enzymes
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/09Fusion polypeptide containing a localisation/targetting motif containing a nuclear localisation signal
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • 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 [CRISPR]

Definitions

  • the present disclosure relates generally to polypeptides, such as effector proteins, compositions of such polypeptides and guide nucleic acids, systems, and methods of using such polypeptides and compositions, including detecting and editing target nucleic acids.
  • effector proteins are engineered.
  • TABLE 1 provides exemplary engineered effector proteins and TABLE 2 shows the parent sequence that was the basis for engineering as well as the engineering strategy corresponding to each effector protein in TABLE 1.
  • TABLE 6 provides exemplary effector proteins that may be engineered.
  • a domain of an effector protein in TABLE 6 or a portion thereof may be replaced with a sequence described in TABLE 2.
  • TABLE 8 also provides exemplary engineered effector proteins.
  • compositions, systems, and methods comprise guide nucleic acids or uses thereof.
  • a guide nucleic acid comprises a handle sequence provided in TABLE 7.
  • Compositions, systems, and methods disclosed herein may leverage nucleic acid modification activities, such as nucleic acid editing.
  • Nucleic acid editing may comprise: insertion, deletion, substitution, or a combination thereof of one or more nucleotides in a target nucleic acid.
  • Nucleic acid editing may comprise cleavage activity, such as cis cleavage activity, nickase activity, and/or nuclease activity. Modification activities may also include epigenetic modifications of nucleic acids.
  • compositions, systems and methods are useful for the treatment of a disease or disorder.
  • the disease or disorder may be associated with a target nucleic acid.
  • the disease or disorder may be associated with one or more mutations in the target nucleic acid.
  • the present disclosure provides systems or compositions comprising: an effector protein, or a nucleic acid encoding the effector protein, comprising an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to a sequence selected from TABLE 1, TABLE 6, and TABLE 8.
  • the system or composition comprises an engineered guide nucleic acid, or a nucleic acid encoding the guide nucleic acid, wherein the engineered guide nucleic acid comprises a protein binding sequence and a spacer sequence.
  • the protein binding sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 858.
  • the protein binding sequence comprises a handle sequence comprising a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to a sequence selected from TABLE 7.
  • the effector protein comprises an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%.
  • the handle sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 859 or 860.
  • the effector protein comprises an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 624
  • the handle sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 884.
  • the effector protein comprises an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 713
  • the handle sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 885.
  • the effector protein comprises an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 833
  • the handle sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 886.
  • the effector protein comprises an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 691
  • the handle sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 884.
  • the effector protein comprises an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 774
  • the handle sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 888.
  • the effector protein comprises an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 179 and 889-910, and the protein binding sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 858.
  • the spacer sequence is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical, complementary, or reverse complementary to a eukaryotic sequence. In some embodiments, the spacer sequence is linked to a 5’ end of the protein binding sequence.
  • the effector protein comprises about 100, about 120, about 140, about 160, about 180, about 200, about 220, about 240, about 260, about 280, about 300, about 320, about 340, about 360, about 380, about 400, about 420, about 440, about 460, about 480, about 500, or about 520 contiguous amino acids of a sequence selected from TABLE 1, TABLE 6, and TABLE 8.
  • the length of the effector protein is less than 600, less than 580, less than 560, less than 540, less than 520, less than 500, less than 480, less than 460, less than 440, less than 420, or less than 400 contiguous amino acids; and at least 300, at least 320, at least 340, at least 360, at least 380 contiguous amino acids.
  • the amino acid sequence is less than 100% identical to the sequence selected from TABLE 1, TABLE 6, and TABLE 8, and wherein not more than 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 amino acid substitutions in the amino acid sequence are non-conservative amino acid substitutions relative to the respective sequence selected from TABLE 1, TABLE 6, and TABLE 8.
  • the effector protein comprises at least one amino acid substitution in a RuvC domain, an HNH domain, or a combination thereof.
  • the effector protein comprises an amino acid sequence that is at least 90%, at least 95%, at least 97%, at least 98%, or at least 99% identical to SEQ ID NO: 624, wherein the effector protein comprises an amino acid substitution of H246A relative to SEQ ID NO: 624
  • the effector protein encoded by a nucleic acid sequence that is at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 912.
  • the effector protein comprises an amino acid sequence that is at least 90%, at least 95%, at least 97%, at least 98%, or at least 99% identical to SEQ ID NO: 713, wherein the effector protein comprises an amino acid substitution of H244A relative to SEQ ID NO: 713
  • the effector protein comprises an amino acid sequence that is at least 90%, at least 95%, at least 97%, at least 98%, or at least 99% identical to SEQ ID NO: 856, wherein the effector protein comprises an amino acid substitution of H248A relative to SEQ ID NO: 856.
  • the amino acid substitution is a non-conservative amino acid substitution.
  • the amino acid substitution replaces a catalytic residue of the domain(s).
  • a) the effector protein comprises an amino acid sequence at least 90-100% identical to SEQ ID NO: 856 and recognizes a TAM of ATAANNN when complexed with a guide nucleic acid; b) the effector protein comprises an amino acid sequence at least 90-100% identical to SEQ ID NO: 624 and recognizes a TAM of ARRRGNN when complexed with a guide nucleic acid; c) the effector protein comprises an amino acid sequence at least 90-100% identical to SEQ ID NO: 713 and recognizes a TAM of GNAAANN when complexed with a guide nucleic acid; d) the effector protein comprises an amino acid sequence at least 90-100% identical to SEQ ID NO: 833 and recognizes a TAM of ATAANNN when complexed with a guide nucleic acid; e) the effector protein comprises an amino acid sequence at least 90-100% identical to SEQ ID NO:
  • the effector protein has nickase activity. In some embodiments, the effector protein is linked to a nuclear localization signal. In some embodiments, the system or composition further comprises a donor nucleic acid. In some embodiments, the system or composition further comprises an effector partner protein linked to the effector protein. In some embodiments, the effector partner comprises a polypeptide selected from a deaminase, a reverse transcriptase, a transcriptional activator, a transcriptional repressor, or a functional domain thereof.
  • the effector protein comprises at least one mutation that reduces its nuclease activity relative to the effector protein without the at least one mutation as measured in a cleavage assay, optionally wherein the effector protein is a catalytically inactive nuclease.
  • the system or composition further comprises a lipid nanoparticle containing the effector protein or the nucleic acid encoding the effector protein, the engineered guide nucleic acid, or a combination thereof.
  • the effector protein or the nucleic acid encoding the effector protein, and the engineered guide nucleic acid or the nucleic acid encoding the guide nucleic acid are provided in separate compositions.
  • the nucleic acid encoding the effector protein, the nucleic acid encoding the guide nucleic acid, or a combination thereof is an expression vector.
  • the expression vector is a viral vector, optionally wherein the viral vector is an adeno- associated viral (AAV) vector.
  • the present disclosure provides systems or compositions comprising an engineered effector protein, wherein at least one domain of a parent sequence is replaced by one or more corresponding domains from another protein, and wherein the parent sequence is the parent sequence of TABLE 2.
  • the at least one domain of the parent sequence is selected from an HNH domain, a PLMP domain, a BH domain, and a TID domain.
  • the corresponding domain from another protein is any one of the sequences selected from TABLE 2.
  • the system or composition comprises an engineered guide nucleic acid, or a nucleic acid encoding the guide nucleic acid, wherein the engineered guide nucleic acid comprises a protein binding sequence and a spacer sequence.
  • the protein binding sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 858.
  • the protein binding sequence comprises a handle sequence comprising a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to a sequence selected from TABLE 7.
  • the spacer sequence hybridizes to a target sequence that is adjacent to a TAM of 5’-NWRRNA-3’, wherein W is A or T, N is any nucleotide, and R is A or G. In some embodiments, the spacer sequence hybridizes to a target sequence that is adjacent to a TAM of 5’-ATAANNN-3’, wherein N is any nucleotide.
  • the present disclosure provides pharmaceutical compositions comprising the system or composition described herein, and a pharmaceutically acceptable excipient.
  • the present disclosure provides methods of modifying a target nucleic acid, the method comprising contacting the target nucleic acid with the system or composition described herein or the pharmaceutical composition described herein, thereby modifying the target nucleic acid.
  • the present disclosure provides cells comprising the composition described herein.
  • the present disclosure provides cells modified by the system or composition described herein, the pharmaceutical composition described herein, or the method described herein.
  • the present disclosure provides cells comprising a modified target nucleic acid, wherein the modified target nucleic acid is a target nucleic acid modified according to the method described herein.
  • the present disclosure provides methods of treating a disease comprising administering to a subject in need thereof any one of: the system or composition described herein; the pharmaceutical composition described herein; or the cell described herein.
  • the present disclosure provides systems or compositions comprising: a) an effector protein or a nucleic acid encoding the same, wherein the effector protein comprises an amino acid sequence that is at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, identical to SEQ ID NO: 855; b) a reverse transcriptase or a nucleic acid encoding the same; c) a guide nucleic acid or a nucleic acid encoding the same; and d) a retRNA or a nucleic acid encoding the same.
  • the reverse transcriptase comprises a peptide that is capable of binding a secondary structure present in at least one of the guide nucleic acid and the retRNA.
  • the effector protein or nucleic acid encoding the same is covalently linked to the reverse transcriptase or nucleic acid encoding the same.
  • the system or composition comprises the nucleic acid encoding the effector protein, the nucleic acid encoding the reverse transcriptase, the nucleic acid encoding the guide nucleic acid, and the nucleic acid encoding the retRNA, wherein all of the nucleic acids are present in a single expression vector, optionally wherein the expression vector is an AAV vector.
  • the present disclosure provides methods of modifying a target nucleic acid, the method comprising contacting the target nucleic acid with the system or composition described herein. In some embodiments, the method comprises contacting a cell with the system or composition.
  • the present disclosure provides systems or compositions comprising: a) an IscB protein; b) a reverse transcriptase or a nucleic acid encoding the same; c) a guide nucleic acid or a nucleic acid encoding the same; and d) a retRNA or a nucleic acid encoding the same.
  • the present disclosure provides methods of editing a target nucleic acid, the method comprising contacting a target nucleic acid with the system or composition described herein.
  • FIG. 1 depicts the domains, defined by amino acid positions, of the parent sequence that was the basis for engineering.
  • FIG. 2 depicts an exemplary target nucleic acid and exemplary guide nucleic acid to be used with effector proteins described herein for the purposes of showing the orientation of the target adjacent motif (TAM) in relationship to where the guide nucleic acid hybridizes to a target nucleic acid.
  • NTS non-target strand
  • TS target strand.
  • a protein binding sequence is not provided, but labeled for the purpose of showing the orientation of the protein binding sequence with respect to the spacer sequence (the spacer sequence is 5’ of the protein binding sequence).
  • the figure should not be interpreted to indicate that the protein binding sequence or any portion thereof necessarily binds to the target nucleic acid. Also, for the avoidance of doubt, this figure should not suggest that the NTS and TS are paired when the guide nucleic acid hybridizes to the target nucleic acid.
  • FIG. 3 shows an example of a split protein/RNA system for precise editing with an effector (e.g., DIS dual), wherein the retRNA is circularized.
  • an effector e.g., DIS dual
  • FIG. 4A shows precision editing levels of DIS dual (H247A) RT editing is comparable to Cas9 RT editing in a CRISPR+ dual fluorescent reporter assay.
  • FIG. 4B shows precision editing levels of DI S dual (H247A) RT editing is comparable to Cas9 RT editing in a DI S dual fluorescent reporter assay. Bars above each RT tested, from left to right, represent SpCas9, SpCas9 (H840A), DIS dual, and DIS dual (H247A).
  • FIG. 5 shows the position frequency matrix (PFM) from WebLogos revealing the presence of enriched 5’ TAM consensus sequences for various effector protein-guide RNA complexes.
  • FIG. 6 shows that an engineered effector protein represented by SEQ ID NO: 179 (engineered from parental sequence SEQ ID NO: 275) provides increased modifications (indels) in a target nucleic acid in mammalian cells relative to an effector protein represented by SEQ ID NO: 275
  • FIG. 7 shows that an engineered effector proteins represented by SEQ ID NOS: 889- 910 (engineered from parental sequence SEQ ID NO: 275) provides increased modifications (indels) in a target nucleic acid in mammalian cells relative to an effector protein represented by SEQ ID NO: 275
  • FIG. 8A shows an engineered variant of effector protein 3745646 can produce precise edits in RT editing system.
  • FIG. 8B shows precise editing with an engineered variant of effector protein 3745646 can be enhanced with a DNA mismatch repair inhibitor.
  • % identical refers to the percent of residues that are identical between respective positions of two sequences when the two sequences are aligned for maximum sequence identity.
  • the % identity is calculated by dividing the total number of the aligned residues by the number of the residues that are identical between the respective positions of the at least two sequences and multiplying by 100.
  • computer programs can be employed for such calculations. Illustrative programs that compare and align pairs of sequences, include ALIGN (Myers and Miller, Comput Appl Biosci.
  • % complementary refers to the percent of nucleotides in two nucleotide sequences in said nucleic acid molecules of equal length that can undergo cumulative base pairing at two or more individual corresponding positions in an antiparallel orientation. Accordingly, the terms include nucleic acid sequences that are not completely complementary over their entire length, which indicates that the two or more nucleic acid molecules include one or more mismatches. A “mismatch” is present at any position in the two opposed nucleotides that are not complementary.
  • the % complementary is calculated by dividing the total number of the complementary residues by the total number of the nucleotides in one of the equal length sequences, and multiplying by 100.
  • Complete or total complementarity describes nucleotide sequences in 100% of the residues of a nucleotide sequence are complementary to residues in a reference nucleotide sequence.
  • Partially complementarity describes nucleotide sequences in which at least 20%, but less than 100%, of the residues of a nucleotide sequence are complementary to residues in a reference nucleotide sequence. In some instances, at least 50%, but less than 100%, of the residues of a nucleotide sequence are complementary to residues in a reference nucleotide sequence.
  • At least 70%, 80%, 90% or 95%, but less than 100%, of the residues of a nucleotide sequence are complementary to residues in a reference nucleotide sequence.
  • “Noncomplementary” describes nucleotide sequences in which less than 20% of the residues of a nucleotide sequence are complementary to residues in a reference nucleotide sequence.
  • percent similarity refers to a value that is calculated by dividing a similarity score by the length of the alignment.
  • the similarity of two amino acid sequences can be calculated by using a BLOSUM62 similarity matrix (Henikoff and Henikoff, Proc. Natl. Acad. Sci. USA., 89: 10915-10919 (1992)) that is transformed so that any value > 1 is replaced with +1 and any value ⁇ 0 is replaced with 0.
  • BLOSUM62 similarity matrix Henikoff and Henikoff, Proc. Natl. Acad. Sci. USA., 89: 10915-10919 (1992)
  • an He (I) to Leu (L) substitution is scored at +2.0 by the BLOSUM62 similarity matrix, which in the transformed matrix is scored at +1.
  • the proteins when comparing two full protein sequences, the proteins can be aligned using pairwise MUSCLE alignment. Then, the % similarity can be scored at each residue and divided by the length of the alignment. For determining % similarity over a protein domain or motif, a multilevel consensus sequence (or PROSITE motif sequence) can be used to identify how strongly each domain or motif is conserved. In calculating the similarity of a domain or motif, the second and third levels of the multilevel sequence are treated as equivalent to the top level. Additionally, if a substitution could be treated as conservative with any of the amino acids in that position of the multilevel consensus sequence, +1 point is assigned.
  • a multilevel consensus sequence or PROSITE motif sequence
  • the test sequence QIQ would receive three points. This is because in the transformed BLOSUM62 matrix, each combination is scored as: Q-R: +1; Q-Y: +0; I-L: +1; I-C: +0; Q-G: +0; Q-K: +1. For each position, the highest score is used when calculating similarity.
  • bind refers to a non-covalent interaction between macromolecules (e.g., between two polypeptides, between a polypeptide and a nucleic acid; between a polypeptide/guide nucleic acid complex and a target nucleic acid; and the like). While in a state of noncovalent interaction, the macromolecules are said to be “associated” or “interacting” or “binding” (e.g., when a molecule X is said to interact with a molecule Y, it is meant the molecule X binds to molecule Y in a non-covalent manner).
  • Non-limiting examples of non-covalent interactions are ionic bonds, hydrogen bonds, van der Waals and hydrophobic interactions. Not all components of a binding interaction need be sequence-specific (e.g., contacts with phosphate residues in a DNA backbone), but some portions of a binding interaction may be sequence-specific.
  • base editor refers to a polypeptide or fusion protein comprising a base editing activity.
  • the polypeptide with base editing activity may be referred to as an effector partner.
  • the base editor can differ from a naturally occurring base editing enzyme. It is understood that any reference to a base editor herein also refers to a base editing enzyme variant.
  • the base editor is functional when the effector protein is coupled to a guide nucleic acid.
  • the guide nucleic acid imparts sequence specific activity to the base editor.
  • the effector protein may comprise a catalytically inactive effector protein (e.g., a catalytically inactive variant of an effector protein described herein).
  • the base editing enzyme may comprise deaminase activity.
  • catalytically inactive effector protein refers to an effector protein that is modified relative to a naturally-occurring effector protein to have a reduced or eliminated catalytic activity relative to that of the naturally-occurring effector protein, but retains its ability to interact with a guide nucleic acid.
  • the catalytic activity that is reduced or eliminated is often a nuclease activity.
  • the naturally-occurring effector protein may be a wildtype protein.
  • the catalytically inactive effector protein is referred to as a catalytically inactive variant of an effector protein.
  • cleavage refers to cleavage (hydrolysis of a phosphodiester bond) of a target nucleic acid by a complex of an effector protein and a guide nucleic acid (e.g., an RNP complex), wherein at least a portion of the guide nucleic acid is hybridized to at least a portion of the target nucleic acid. Cleavage may occur within or directly adjacent to the portion of the target nucleic acid that is hybridized to the portion of the guide nucleic acid.
  • codon optimized refers to a mutation of a nucleotide sequence encoding a polypeptide, such as a nucleotide sequence encoding an effector protein, to mimic the codon preferences of the intended host organism or cell while encoding the same polypeptide. Thus, the codons can be changed, but the encoded polypeptide remains unchanged. For example, if the intended target cell was a human cell, a human codon-optimized nucleotide sequence encoding an effector protein could be used. As another non-limiting example, if the intended host cell were a mouse cell, then a mouse codon-optimized nucleotide sequence encoding an effector protein could be generated.
  • a eukaryote codon-optimized nucleotide sequence encoding an effector protein could be generated.
  • a prokaryotic cell then a prokaryote codon-optimized nucleotide sequence encoding an effector protein could be generated. Codon usage tables are readily available, for example, at the “Codon Usage Database” available at www.kazusa.or.jp/codon.
  • nucleic acid molecule or nucleotide sequence refer to the characteristic of a polynucleotide having nucleotides that can undergo cumulative base pairing with their Watson-Crick counterparts (C with G; or A with T) in a reference nucleic acid in antiparallel orientation. For example, when every nucleotide in a polynucleotide or a specified portion thereof forms a base pair with every nucleotide in an equal length sequence of a reference nucleic acid, that polynucleotide is said to be 100% complementary to the sequence of the reference nucleic acid.
  • the upper (sense) strand sequence is, in general, understood as going in the direction from its 5'- to 3 '-end, and the complementary sequence is thus understood as the sequence of the lower (antisense) strand in the same direction as the upper strand.
  • the reverse sequence is understood as the sequence of the upper strand in the direction from its 3'- to its 5 '-end, while the “reverse complement” sequence or the “reverse complementary” sequence is understood as the sequence of the lower strand in the direction of its 5 '- to its 3 '-end.
  • Each nucleotide in a double stranded DNA or RNA molecule that is paired with its Watson-Crick counterpart can be referred to as its complementary nucleotide.
  • the complementarity of modified or artificial base pairs can be based on other types of hydrogen bonding and/or hydrophobicity of bases and/or shape complementarity between bases.
  • cleavage assay refers to an assay designed to visualize, quantitate or identify cleavage of a nucleic acid.
  • the cleavage activity may be cis cleavage activity.
  • Example 1 and Example 2 provide non-limiting examples of cleavage assays.
  • cleave in the context of a nucleic acid molecule or nuclease activity of an effector protein, refer to the hydrolysis of a phosphodiester bond of a nucleic acid molecule that results in breakage of that bond.
  • the result of this breakage can be a nick (hydrolysis of a single phosphodiester bond on one side of a double-stranded molecule), single strand break (hydrolysis of a single phosphodiester bond on a single-stranded molecule) or double strand break (hydrolysis of two phosphodiester bonds on both sides of a double-stranded molecule) depending upon whether the nucleic acid molecule is single-stranded (e.g., ssDNA or ssRNA) or double-stranded (e.g., dsDNA) and the type of nuclease activity being catalyzed by the effector protein.
  • a nick hydrolysis of a single phosphodiester bond on one side of a double-stranded molecule
  • single strand break hydrolysis of a single phosphodiester bond on a single-stranded molecule
  • double strand break hydrolysis of two phosphodiester bonds on both sides of a double-stranded molecule
  • CRISPR clustered regularly interspaced short palindromic repeats
  • Genetically encoded amino acids can be divided into four families having related side chains: (1) acidic (negatively charged): Asp (D), Glu (E); (2) basic (positively charged): Lys (K), Arg (R), His (H); (3) non-polar (hydrophobic): Cys (C), Ala (A), Vai (V), Leu (L), He (I), Pro (P), Phe (F), Met (M), Trp (W), Gly (G), Tyr (Y), with non-polar also being subdivided into: (i) strongly hydrophobic: Ala (A), Vai (V), Leu (L), He (I), Met (M), Phe (F); and (ii) moderately hydrophobic: Gly (G), Pro (P), Cys (C), Tyr (Y), Trp (W); and (4) uncharged polar: Asn (N), Gin (Q), Ser (S), Thr (T).
  • Amino acids may be related by aliphatic side chains: Gly (G), Ala (A), Vai (V), Leu (L), He (I), Ser (S), Thr (T), with Ser (S) and Thr (T) optionally being grouped separately as aliphatic-hydroxyl; Amino acids may be related by aromatic side chains: Phe (F), Tyr (Y), Trp (W). Amino acids may be related by amide side chains: Asn (N), Gin (Q). Amino acids may be related by sulfur-containing side chains: Cys (C) and Met (M).
  • the term, “edited target nucleic acid,” as used herein, refers to a target nucleic acid, wherein the target nucleic acid has undergone an editing, for example, after contact with an effector protein.
  • the editing is an alteration in the sequence of the target nucleic acid.
  • the edited target nucleic acid comprises an insertion, deletion, or substitution of one or more nucleotides compared to the unedited target nucleic acid.
  • effector protein refers to a protein, polypeptide, or peptide that is capable of interacting with a nucleic acid, such as a guide nucleic acid, to form a complex (e.g., a RNP complex), wherein the complex interacts with a target nucleic acid.
  • effector partner refers to a protein, polypeptide or peptide that can, in combination with an effector protein, impart some function or activity that can be used to effectuate modification(s) of a target nucleic acid described herein and/or change expression of the target nucleic acid or other nucleic acids associated with the target nucleic acid, when used in connection with compositions, systems and methods described herein.
  • engineered modification refers to a structural change of one or more nucleic acid residues of a nucleotide sequence or one or more amino acid residue of an amino acid sequence.
  • the engineered modifications of a nucleotide sequence can include chemical modification of one or more nucleobases, or a chemical change to the phosphate backbone, a nucleotide, a nucleobase or a nucleoside.
  • the engineered modifications can be made to an effector protein amino acid sequence or guide nucleic acid nucleotide sequence, or any sequence disclosed herein (e.g., a nucleic acid encoding an effector protein or a nucleic acid that encodes a guide nucleic acid).
  • nucleic acids provided herein can be prepared according to any available technique including, but not limited to chemical synthesis, enzymatic synthesis, which is generally termed in vitro-transcription, cloning, enzymatic, or chemical cleavage, etc. In some instances, the nucleic acids provided herein are not uniformly modified along the entire length of the molecule. Different nucleotide modifications and/or backbone structures can exist at various positions within the nucleic acid.
  • the term, “functional domain,” as used herein, refers to a region of one or more amino acids in a protein that is required for an activity of the protein, or the full extent of that activity, as measured in an in vitro assay. Activities include, but are not limited to nucleic acid binding, nucleic acid editing, nucleic acid modifying, nucleic acid cleaving, protein binding. The absence of the functional domain, including mutations of the functional domain, would abolish or reduce activity.
  • the term, “functional fragment,” as used herein, refers to a fragment of a protein that retains some function relative to the entire protein.
  • functions are nucleic acid binding, nucleic acid editing, protein binding, nuclease activity, nickase activity, deaminase activity, demethylase activity, or acetylation activity.
  • a functional fragment may be a recognized functional domain, e.g., a catalytic domain. In some instances, the catalytic domain comprises a RuvC domain.
  • the term, “functional protein,” as used herein, refers to protein that retains at least some if not all activity relative to the wildtype protein.
  • a functional protein can also include a protein having enhanced activity relative to the wildtype protein. Assays are known and available for detecting and quantifying protein activity, e.g., colorimetric and fluorescent assays. In some instances, a functional protein is a wildtype protein. In some instances, a functional protein is a functional portion of a wildtype protein.
  • covalent bond e.g., an amide bond or a phophodiester bond
  • linker e.g., a linker that bonds to at least two sequences that are connected together, such as by a covalent bond (e.g., an amide bond or a phophodiester bond) or by a linker.
  • the covalent bond can be formed by conjugation (e.g., chemical conjugation or enzymatic conjugation) reaction.
  • fusion protein refers to a protein comprising at least two polypeptides.
  • the fusion protein may comprise one or more effector proteins and effector partners. In some instances, an effector protein and effector partner are not found connected to one another as a native protein or complex that occurs together in nature.
  • genetic disease refers to a disease, disorder, condition, or syndrome associated with or caused by one or more mutations in the DNA of an organism having the genetic disease.
  • guide nucleic acid refers to a nucleic acid that, when in a complex with one or more polypeptides described herein (e.g., an RNP complex) can impart sequence selectivity to the complex when the complex interacts with a target nucleic acid.
  • a guide nucleic acid may be referred to interchangeably as a guide RNA, however it is understood that guide nucleic acids may comprise deoxyribonucleotides (DNA), ribonucleotides (RNA), a combination thereof (e.g., RNA with a thymine base), biochemically or chemically modified nucleobases (e.g., one or more engineered modifications described herein), or combinations thereof.
  • CRISPR RNA or “crRNA,” as used herein, refer to a type of guide nucleic acid, wherein the nucleic acid is RNA comprising a first sequence, often referred to herein as a spacer sequence, that hybridizes to a target sequence of a target nucleic acid, and a second sequence, often referred to herein as a repeat sequence or guide sequence, that interacts with an effector protein.
  • the second sequence is bound by the effector protein.
  • the second sequence hybridizes to a portion of a tracrRNA, wherein the tracrRNA forms a complex with the effector protein.
  • tracrRNA trans-activating RNA
  • tracrRNA refers to a nucleic acid that comprises a first sequence that is capable of being non-covalently bound by an effector protein, and a second sequence that hybridizes to a portion of a crRNA, which may be referred to as a repeat hybridization sequence.
  • handle sequence refers to a sequence that binds non- covalently with an effector protein.
  • a handle sequence may also be referred to herein as a “scaffold sequence”.
  • the handle sequence comprises all, or a portion of, a repeat sequence.
  • a single guide nucleic acid also referred to as a single guide RNA (sgRNA)
  • sgRNA single guide RNA
  • the nucleotide sequence of a handle sequence may contain a portion of a tracrRNA, but generally does not comprise a sequence that hybridizes to a repeat sequence, also referred to as a repeat hybridization sequence.
  • template RNA refers to a nucleic acid comprising: a primer binding sequence and a template sequence. It is understood that template RNAs may comprise DNA, RNA, or a combination thereof (e.g., RNA with a thymine base). In some instances, the template RNA is linked to a guide RNA via a linker sequence to form an rtgRNA.
  • template sequence refers to a portion of a retRNA that contains a desired nucleotide modification relative to a target sequence or portion thereof.
  • the desired edit may comprise one or more nucleotide insertions, deletions or substitutions relative to a target sequence or portion thereof. In some embodiments, it is identical to, complementary to, or reverse complementary to a target sequence or portion thereof.
  • the template sequence is complementary to a sequence of the target nucleic acid that is adjacent to a nick site of a target site to be edited, with the exception that it includes a desired edit.
  • the template sequence (also referred in some instances as the RT template (RTT)) can be complementary to at least a portion of the target sequence with the exception of at least one nucleotide.
  • primer binding sequence refers to a portion of a retRNA and serves to bind to a primer sequence of the target nucleic acid.
  • the primer binding sequence binds to a primer sequence in the target nucleic acid that is formed after the target nucleic acid is cleaved by an effector protein.
  • the primer binding sequence is linked to the 3’ end of an retRNA.
  • the primer binding sequence is located at the 5’ end of a retRNA.
  • Primary sequence refers to a portion of the target nucleic acid that is capable of hybridizing with the primer binding sequence portion of an retRNA that is generated after cleavage of the target nucleic acid by an effector protein described herein.
  • heterologous refers to at least two different polypeptide sequences that are not found similarly connected to one another in a native nucleic acid or protein.
  • a protein that is heterologous to the effector protein is a protein that is not covalently linked by an amide bond to the effector protein in nature. In some instances, a protein is heterologous when the protein is not encoded by a species that encodes the effector protein.
  • a guide nucleic acid may comprise “heterologous” sequences, which means that it includes a first sequence and a second sequence, wherein the first sequence and the second sequence are not found covalently linked by a phosphodiester bond in nature.
  • the first sequence is considered to be heterologous with the second sequence, and the guide nucleic acid may be referred to as a heterologous guide nucleic acid.
  • a heterologous system comprises at least one component that is not naturally occurring together with remaining components of the heterologous system.
  • hybridize refers to a nucleotide sequence that is able to noncovalently interact, i.e. form Watson-Crick base pairs and/or G/U base pairs, or anneal, to another nucleotide sequence in a sequence-specific, antiparallel, manner (i.e., a nucleotide sequence specifically interacts to a complementary nucleotide sequence) under the appropriate in vitro and/or in vivo conditions of temperature and solution ionic strength.
  • Standard Watson-Crick base-pairing includes: adenine (A) pairing with thymidine (T), adenine (A) pairing with uracil (U), and guanine (G) pairing with cytosine (C) for both DNA and RNA.
  • adenine (A) pairing with thymidine (T)
  • A adenine
  • U uracil
  • G guanine
  • C cytosine
  • RNA molecules e.g., dsRNA
  • guanine (G) can also base pair with uracil (U).
  • G/U base-pairing is at least partially responsible for the degeneracy (i.e., redundancy) of the genetic code in the context of tRNA anti-codon base-pairing with codons in mRNA.
  • a guanine (G) can be considered complementary to both an uracil (U) and to an adenine (A).
  • G/U base-pair can be made at a given nucleotide position, the position is not considered to be non-complementary, but is instead considered to be complementary. While hybridization typically occurs between two nucleotide sequences that are complementary, mismatches between bases are possible.
  • nucleotide sequences need not be 100% complementary to be specifically hybridizable, hybridizable, partially hybridizable, or for hybridization to occur.
  • a nucleotide sequence may hybridize over one or more segments such that intervening or adjacent segments are not involved in the hybridization event (e.g., a bulge, a loop structure or hairpin structure, etc.).
  • the conditions appropriate for hybridization between two nucleotide sequences depend on the length of the sequence and the degree of complementarity, variables which are well known in the art. For hybridizations between nucleic acids with short stretches of complementarity (e.g.
  • the position of mismatches may become important (see Sambrook et al., supra, 11.7-11.8).
  • the length for a hybridizable nucleic acid is 8 nucleotides or more (e.g., 10 nucleotides or more, 12 nucleotides or more, 15 nucleotides or more, 20 nucleotides or more, 22 nucleotides or more, 25 nucleotides or more, or 30 nucleotides or more). Any suitable in vitro assay may be utilized to assess whether two sequences “hybridize”.
  • One such assay is a melting point analysis where the greater the degree of complementarity between two nucleotide sequences, the greater the value of the melting temperature (Tm) for hybrids of nucleic acids having those sequences.
  • Tm melting temperature
  • the conditions of temperature and ionic strength determine the “stringency” of the hybridization. Temperature, wash solution salt concentration, and other conditions may be adjusted as necessary according to factors such as length of the region of complementation and the degree of complementation.
  • Hybridization and washing conditions are well known and exemplified in Sambrook, J. and Russell, W., Molecular Cloning: A Laboratory Manual, Third Edition, Cold Spring Harbor Laboratory Press, Cold Spring Harbor (2001); and in Green, M. and Sambrook, J., Molecular Cloning: A Laboratory Manual, Fourth Edition, Cold Spring Harbor Laboratory Press, Cold Spring Harbor (2012).
  • indel refers to an insertion-deletion or indel mutation, which is a type of genetic mutation that results from the insertion and/or deletion of one or more nucleotide in a target nucleic acid.
  • An indel can vary in length (e.g., 1 to 1,000 nucleotides in length) and be detected by any suitable method, including sequencing.
  • the term, “indel percentage,” as used herein, refers to a percentage of sequencing reads that show at least one nucleotide has been edited from the insertion and/or deletion of nucleotides regardless of the size of insertion or deletion, or number of nucleotides edited. For example, if there is at least one nucleotide deletion detected in a given target nucleic acid, it counts towards the percent indel value. As another example, if one copy of the target nucleic acid has one nucleotide deleted, and another copy of the target nucleic acid has 10 nucleotides deleted, they are counted the same. This number reflects the percentage of target nucleic acids that are edited by a given effector protein.
  • in vitro refers to describing something outside an organism.
  • An in vitro system, composition or method may take place in a container for holding laboratory reagents such that it is separated from the biological source from which a material in the container is obtained.
  • In vitro assays can encompass cell-based assays in which living or dead cells are employed.
  • In vitro assays can also encompass a cell-free assay in which no intact cells are employed.
  • the term “in vivo” is used to describe an event that takes place within an organism.
  • ex vivo is used to describe an event that takes place in a cell that has been obtained from an organism. An ex vivo assay is not performed on a subject. Rather, it is performed upon a sample separate from a subject.
  • length and “linked” as used herein refer to a nucleic acid (polynucleotide) or polypeptide, may be expressed as “kilobases” (kb) or “base pairs (bp),”. Thus, a length of 1 kb refers to a length of 1000 linked nucleotides, and a length of 500 bp refers to a length of 500 linked nucleotides. Similarly, a protein having a length of 500 linked amino acids may also be simply described as having a length of 500 amino acids.
  • linker refers to a molecule that links a first polypeptide to a second polypeptide (e.g., by an amide bond) or a first nucleic acid to a second nucleic acid (e.g., by a phosphodiester bond).
  • mutation refers to an alteration that changes an amino acid residue or a nucleotide as described herein. Such an alteration can include, for example, deletions, insertions, and/or substitutions.
  • the mutation can refer to a change in structure of an amino acid residue or nucleotide relative to the starting or reference residue or nucleotide.
  • a mutation of an amino acid residue includes, for example, deletions, insertions and substituting one amino acid residue for a structurally different amino acid residue.
  • substitutions can be a conservative substitution, a non-conservative substitution, a substitution to a specific subclass of amino acids, or a combination thereof as described herein.
  • a mutation of a nucleotide includes, for example, changing one naturally occurring base for a different naturally occurring base, such as changing an adenine to a thymine or a guanine to a cytosine or an adenine to a cytosine or a guanine to a thymine.
  • a mutation of a nucleotide base may result in a structural and/or functional alteration of the encoding peptide, polypeptide or protein by changing the encoded amino acid residue of the peptide, polypeptide or protein.
  • a mutation of a nucleotide base may not result in an alteration of the amino acid sequence or function of encoded peptide, polypeptide or protein, also known as a silent mutation. Methods of mutating an amino acid residue or a nucleotide are well known.
  • mutation associated with a disease and “mutation associated with a genetic disorder,” as used herein, refer to the co-occurrence of a mutation and the phenotype of a disease.
  • the mutation may occur in a gene, wherein transcription or translation products from the gene occur at a significantly abnormal level or in an abnormal form in a cell or subject harboring the mutation as compared to a non-disease control subject not having the mutation.
  • nickase refers to an enzyme that possess catalytic activity for single stranded nucleic acid cleavage of a double stranded nucleic acid.
  • nickase activity refers to catalytic activity that results in single stranded nucleic acid cleavage of a double stranded nucleic acid.
  • nucleic acid, nucleotide, protein, polypeptide, peptide or amino acid refers to a molecule, such as but not limited to, a nucleic acid, nucleotide, protein, polypeptide, peptide or amino acid refers to a modification of that molecule (e.g., chemical modification, nucleotide sequence, or amino acid sequence) that is not present in the naturally molecule.
  • a composition or system described herein refer to a composition or system having at least one component that is not naturally associated with the other components of the composition or system.
  • a composition may include an effector protein and a guide nucleic acid that do not naturally occur together.
  • an effector protein or guide nucleic acid that is “natural,” “naturally-occurring,” or “found in nature” includes an effector protein and a guide nucleic acid from a cell or organism that have not been genetically modified by the hand of man.
  • nuclease and “endonuclease” as used herein, refer to an enzyme which possesses catalytic activity for nucleic acid cleavage.
  • nuclease activity refers to catalytic activity that results in nucleic acid cleavage (e.g., ribonuclease activity (ribonucleic acid cleavage), or deoxyribonuclease activity (deoxyribonucleic acid cleavage), etc.).
  • nucleic acid refers to a polymer of nucleotides.
  • a nucleic acid may comprise ribonucleotides, deoxyribonucleotides, combinations thereof, and modified versions of the same.
  • a nucleic acid may be single- stranded or double-stranded, unless specified.
  • Non-limiting examples of nucleic acids are double stranded DNA (dsDNA), single stranded (ssDNA), messenger RNA, genomic DNA, cDNA, DNA-RNA hybrids, and a polymer comprising purine and pyrimidine bases or other natural, chemically or biochemically modified, non-natural, or derivatized nucleotide bases. Accordingly, nucleic acids as described herein may comprise one or more mutations, one or more engineered modifications, or both.
  • nucleic acid expression vector refers to a plasmid that can be used to express a nucleic acid of interest.
  • nuclear localization signal refers to an entity (e.g., peptide) that facilitates localization of a nucleic acid, protein, or small molecule to the nucleus, when present in a cell that contains a nuclear compartment.
  • nucleotide(s) and nucleoside(s) in the context of a nucleic acid molecule having multiple residues, refer to describing the sugar and base of the residue contained in the nucleic acid molecule.
  • nucleosides as used in the context of a nucleic acid having multiple linked residues, are interchangeable and describe linked sugars and bases of residues contained in a nucleic acid molecule.
  • nucleobase(s) or linked nucleobase, as used in the context of a nucleic acid molecule, it can be understood as describing the base of the residue contained in the nucleic acid molecule, for example, the base of a nucleotide, nucleosides, or linked nucleotides or linked nucleosides.
  • nucleotides, nucleosides, and/or nucleobases would also understand the differences between RNA and DNA (generally the exchange of uridine for thymidine or vice versa) and the presence of nucleoside analogs, such as modified uridines, do not contribute to differences in identity or complementarity among polynucleotides as long as the relevant nucleotides (such as thymidine, uridine, or modified uridine) have the same complement (e.g., adenosine for all of thymidine, uridine, or modified uridine; another example is cytosine and 5- methylcytosine, both of which have guanosine or modified guanosine as a complement).
  • nucleoside analogs such as modified uridines
  • sequence 5'-AXG where X is any modified uridine, such as pseudouridine, Nl-methyl pseudouridine, or 5-methoxyuridine is considered 100% identical to AUG in that both are perfectly complementary to the same sequence (5' -CAU).
  • pharmaceutically acceptable excipient, carrier or diluent refers to any substance formulated alongside the active ingredient of a pharmaceutical composition that allows the active ingredient to retain biological activity and is non-reactive with the subject's immune system. Such a substance can be included for the purpose of longterm stabilization, bulking up solid formulations that contain potent active ingredients in small amounts, or to confer a therapeutic enhancement on the active ingredient in the final dosage form, such as facilitating absorption, reducing viscosity, or enhancing solubility. The selection of appropriate substance can depend upon the route of administration and the dosage form, as well as the active ingredient and other factors.
  • compositions having such substances can be formulated by suitable methods (see, e.g., Remington's Pharmaceutical Sciences, 18th edition, A. Gennaro, ed., Mack Publishing Co., Easton, Pa., 1990; and Remington, The Science and Practice of Pharmacy 21st Ed. Mack Publishing, 2005).
  • polypeptide and protein refer to a polymeric form of amino acids.
  • a polypeptide may include coded and non-coded amino acids, chemically or biochemically modified or derivatized amino acids, and polypeptides having modified peptide backbones. Accordingly, polypeptides as described herein may comprise one or more mutations, one or more engineered modifications, or both.
  • coding sequences of polypeptides described herein do not necessarily require a codon encoding an N-terminal Methionine (M) or a Valine (V) as described for the effector proteins described herein.
  • a start codon could be replaced or substituted with a start codon that encodes for an amino acid residue sufficient for initiating translation in a host cell.
  • a heterologous peptide such as an effector partner, protein tag or NLS
  • a start codon for the heterologous peptide serves as a start codon for the effector protein as well.
  • the natural start codon encoding an amino acid residue sufficient for initiating translation e.g., Methionine (M) or a Valine (V)
  • the natural start codon encoding an amino acid residue sufficient for initiating translation e.g., Methionine (M) or a Valine (V)
  • the natural start codon encoding an amino acid residue sufficient for initiating translation e.g., Methionine (
  • RT editing enzyme refers to a protein, polypeptide, or fragment thereof that is capable of catalyzing the editing (insertion, deletion, or base-to-base conversion) of a target nucleotide or nucleotide sequence in a nucleic acid.
  • target adjacent motif and “TAM,” as used herein, refer to a nucleotide sequence found in a target nucleic acid that directs an effector protein to edit the target nucleic acid at a specific location.
  • a TAM is required for a complex of an effector protein and a guide nucleic acid (e.g., an RNP complex) to hybridize to and edit the target nucleic acid.
  • the complex does not require a TAM to edit the target nucleic acid.
  • ribonucleotide protein complex and “RNP” as used herein, refer to a complex of one or more nucleic acids and one or more polypeptides described herein. While the term utilizes “ribonucleotides” it is understood that the one or more nucleic acid may comprise deoxyribonucleotides (DNA), ribonucleotides (RNA), a combination thereof (e.g., RNA with a thymine base), biochemically or chemically modified nucleobases (e.g., one or more engineered modifications described herein), or combinations thereof.
  • DNA deoxyribonucleotides
  • RNA ribonucleotides
  • a combination thereof e.g., RNA with a thymine base
  • biochemically or chemically modified nucleobases e.g., one or more engineered modifications described herein
  • RuvC and RuvC domain refer to a region of an effector protein that is capable of cleaving a target nucleic acid, and in certain instances, of processing a pre-crRNA. In some instances, the RuvC domain is located near the C-terminus of the effector protein.
  • a single RuvC domain may comprise RuvC subdomains, for example a RuvCI subdomain, a RuvCII subdomain and a RuvCIII subdomain.
  • the term “RuvC” domain can also refer to a “RuvC-like” domain.
  • RuvC-like domains are known in the art and are easily identified using online tools such as InterPro (https://www.ebi.ac.uk/interpro/).
  • a RuvC-like domain may be a domain which shares homology with a region of TnpB proteins of the IS605 and other related families of transposons.
  • spacer sequence refers to a nucleotide sequence in a guide nucleic acid that is capable of, at least partially, hybridizing to an equal length portion of a sequence (e.g., a target sequence) of a target nucleic acid.
  • subject refers to an animal.
  • the subject may be a mammal.
  • the subject may be a human.
  • the subject may be diagnosed or at risk for a disease.
  • target nucleic acid refers to a nucleic acid that is selected as the nucleic acid for editing, binding, hybridization or any other activity of or interaction with a nucleic acid, protein, polypeptide, or peptide described herein.
  • a target nucleic acid may comprise RNA, DNA, or a combination thereof.
  • a target nucleic acid may be single-stranded (e.g., single-stranded RNA or single-stranded DNA) or double-stranded (e.g., double-stranded DNA).
  • target sequence in the context of a target nucleic acid, refers to a nucleotide sequence found within a target strand of a target nucleic acid. Such a nucleotide sequence can, for example, hybridize to a respective length portion of a guide nucleic acid.
  • target strand (TS) and “non-target strand (NTS)” are used herein to differentiate between the strands of a double stranded DNA molecule to which a guide nucleic acid does or does not hybridize.
  • target strand (TS), refers to a strand of a double stranded DNA molecule that comprises a target sequence, to which at least a portion of a guide nucleic acid (e.g., a spacer sequence) hybridizes.
  • non-target strand refers to a strand of a double stranded DNA molecule to which a guide nucleic acid does not hybridize. Reference may be made to a target sequence of the TS and a target sequence of the NTS.
  • a guide nucleic acid spacer sequence is generally complementary to the target sequence of the TS and generally identical to the target sequence of the NTS.
  • trans cleavage in the context of cleavage (e.g., hydrolysis of a phosphodiester bond) of one or more target nucleic acids or non-target nucleic acids, or both, by an effector protein that is complexed with a guide nucleic acid and the target nucleic acid.
  • Trans cleavage activity may be triggered by the hybridization of a guide nucleic acid to a target nucleic acid.
  • Trans cleavage of the target nucleic acid may occur away from (e.g., not within or directly adjacent to) the portion of the target nucleic acid that is hybridized to the portion of the guide nucleic acid.
  • transcriptional activator refers to a polypeptide or a fragment thereof that can activate or increase transcription of a target nucleic acid molecule.
  • transcriptional repressor refers to a polypeptide or a fragment thereof that is capable of arresting, preventing, or reducing transcription of a target nucleic acid.
  • transgene refers to a nucleotide sequence that is inserted into a cell for expression of said nucleotide sequence in the cell.
  • a transgene is meant to include (1) a nucleotide sequence that is not naturally found in the cell (e.g., a heterologous nucleotide sequence); (2) a nucleotide sequence that is a mutant form of a nucleotide sequence naturally found in the cell into which it has been introduced; (3) a nucleotide sequence that serves to add additional copies of the same (e.g., exogenous or homologous) or a similar nucleotide sequence naturally occurring in the cell into which it has been introduced; or (4) a silent naturally occurring or homologous nucleotide sequence whose expression is induced in the cell into which it has been introduced.
  • the cell in which transgene expression occurs can be a target cell, such as a host cell.
  • treatment refers to a pharmaceutical or other intervention regimen for obtaining beneficial or desired results in the recipient.
  • beneficial or desired results include but are not limited to a therapeutic benefit and/or a prophylactic benefit.
  • a therapeutic benefit may refer to eradication or amelioration of symptoms or of an underlying disorder being treated.
  • a therapeutic benefit can be achieved with the eradication or amelioration of one or more of the physiological symptoms associated with the underlying disorder such that an improvement is observed in the subject, notwithstanding that the subject may still be afflicted with the underlying disorder.
  • a prophylactic effect includes delaying, preventing, or eliminating the appearance of a disease or condition, delaying, or eliminating the onset of symptoms of a disease or condition, slowing, halting, or reversing the progression of a disease or condition, or any combination thereof.
  • a subject at risk of developing a particular disease, or to a subject reporting one or more of the physiological symptoms of a disease may undergo treatment, even though a diagnosis of this disease may not have been made.
  • variant refers to a form or version of a protein that differs from the wild-type protein. A variant may have a different function or activity relative to the wild-type protein.
  • viral vector refers to a nucleic acid to be delivered into a host cell by a recombinantly produced virus or viral particle.
  • compositions, systems, and methods comprising at least one of: (1) an effector protein or a nucleic acid encoding the effector protein; and (2) an engineered guide nucleic acid or a nucleic acid encoding the engineered guide nucleic acid.
  • the term, “system,” may be used herein for embodiments in which the effector protein or a nucleic acid encoding the effector protein; and the engineered guide nucleic acid or a nucleic acid encoding the engineered guide nucleic acid are not provided in the same container or solution. In some embodiments, these system components are packaged separately, e.g., to be administered separately.
  • Effector proteins described herein may bind and cleave (e.g., nick) nucleic acids in a sequence-specific manner. Effector proteins described herein may also cleave (e.g., nick) the target nucleic acid within a target sequence or at a position adjacent to the target sequence.
  • An effector protein may be similar to a CRISPR-associated (Cas) protein in that it may bind a guide nucleic acid that imparts activity or sequence selectivity to the polypeptide.
  • Cas CRISPR-associated
  • effector proteins described herein are engineered and not necessarily found near a CRISPR array, which includes direct repeats flanking short spacer sequences. Typically, Cas proteins are found within ⁇ 5kb of such an array. In contrast, effector proteins described herein may not have a CRISPR array within 5kb of their corresponding genomic locus. Instead, native effector proteins described herein may be associated in the genome with a non-coding RNA that is capable of secondary structure(s).
  • compositions, systems, and methods comprising guide nucleic acids comprise a first region or sequence, at least a portion of which interacts with a polypeptide.
  • compositions, systems, and methods comprising guide nucleic acids comprise a second sequence that is at least partially complementary to a target nucleic acid, and which may be referred to as a spacer sequence.
  • Effector proteins disclosed herein may bind and cleave (e.g., nick) nucleic acids, including double stranded RNA (dsRNA), single-stranded RNA (ssRNA), double stranded DNA (dsDNA), and single-stranded DNA (ssDNA).
  • Polypeptides disclosed herein may provide cis cleavage activity, trans cleavage activity, binding activity, nickase activity, or a combination thereof.
  • the compositions, systems, and methods described herein are non-naturally occurring.
  • compositions, systems, and methods comprise an engineered guide nucleic acid (also referred to herein as a guide nucleic acid) or a use thereof.
  • compositions, systems, and methods comprise an engineered protein or a use thereof. In some embodiments, compositions, systems, and methods comprise an isolated polypeptide or a use thereof. In general, compositions, methods, and systems described herein are not found in nature. In some embodiments, compositions, methods, and systems described herein comprise at least one non-naturally occurring component.
  • disclosed compositions, methods, and systems may comprise a guide nucleic acid, wherein the sequence of the guide nucleic acid is different or modified from that of a naturally-occurring guide nucleic acid.
  • compositions, systems, and methods comprise at least two components that do not naturally occur together.
  • disclosed compositions, systems, and methods may comprise a guide nucleic acid comprising a first region, at least a portion of which, interacts with a polypeptide, and a second region that is at least partially complementary to a target sequence in a target nucleic acid, wherein the first region and second region do not naturally occur together and/or are heterologous to each other.
  • disclosed compositions, systems, and methods may comprise a guide nucleic acid and an effector protein that do not naturally occur together.
  • compositions, systems, and methods may comprise a ribonucleotide-protein (RNP) complex comprising an effector protein and a guide nucleic acid that do not occur together in nature.
  • RNP ribonucleotide-protein
  • an effector protein or guide nucleic acid that is “natural,” “naturally-occurring,” or “found in nature” includes effector proteins and guide nucleic acids from cells or organisms that have not been genetically modified by a human or machine.
  • the guide nucleic acid comprises a non-natural nucleotide sequence.
  • the non-natural nucleotide sequence is a nucleotide sequence that is not found in nature.
  • the non-natural nucleotide sequence may comprise a portion of a naturally-occurring sequence, wherein the portion of the naturally-occurring sequence is not present in nature absent the remainder of the naturally-occurring sequence.
  • the guide nucleic acid comprises two naturally-occurring sequences arranged in an order or proximity that is not observed in nature.
  • compositions and systems comprise a ribonucleotide complex comprising an effector protein and a guide nucleic acid that do not occur together in nature.
  • compositions and systems comprise at least two components that do not occur together in nature, wherein the at least two components comprise at least one of an effector protein, an effector partner and a guide nucleic acid.
  • Guide nucleic acids may comprise a first sequence and a second sequence that do not occur naturally together.
  • a guide nucleic acid may comprise a first sequence that occurs in a first organism and a second sequence that occurs in a second organism, wherein the first organism and the second organism are different.
  • the guide nucleic acid may comprise a third sequence disposed at a 3’ or 5’ end of the guide nucleic acid, or between the first and second sequences of the guide nucleic acid.
  • the guide nucleic acid comprises two heterologous sequences arranged in an order or proximity that is not observed in nature. Therefore, compositions and systems described herein are not naturally occurring.
  • compositions, systems, and methods described herein comprise a polypeptide (e.g., an effector protein, an effector partner, a fusion protein, or a combination thereof) that is similar to a naturally occurring polypeptide.
  • the polypeptide may lack a portion of the naturally occurring polypeptide.
  • the polypeptide may comprise a mutation relative to the naturally-occurring polypeptide, wherein the mutation is not found in nature.
  • the polypeptide may also comprise at least one additional amino acid relative to the naturally- occurring polypeptide.
  • the polypeptide may comprise a heterologous peptide.
  • the polypeptide may comprise an addition of a nuclear localization signal relative to the natural occurring polypeptide.
  • a nucleotide sequence encoding the polypeptide is codon optimized (e.g., for expression in a eukaryotic cell) relative to the naturally occurring sequence.
  • compositions that comprise one or more effector proteins or a nucleic acid encoding the same, and uses thereof.
  • compositions that comprise a nucleic acid wherein the nucleic acid encodes any of one the effector proteins described herein.
  • the nucleic acid may be an mRNA.
  • the nucleic acid may be a nucleic acid expression vector.
  • the nucleic acid expression vector may be a viral vector, such as an AAV vector.
  • the effector protein comprises a DIS dual protein.
  • a DIS dual protein is a compact effector protein comprising two nuclease domains.
  • the two nuclease domains are a RuvC domain and an HNH domain.
  • the length of a compact effector protein is less than 900, less than 800, less than 700, less than 600, less than 500, or less than 400 linked amino acids, and greater than 300 linked amino acids.
  • the DIS dual protein comprises an IscB protein or engineered variant thereof.
  • the engineered variant is at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, or at least 99% identical to an IscB protein.
  • An effector protein provided herein interacts with a guide nucleic acid to form a complex.
  • the complex interacts with a target nucleic acid, a non-target nucleic acid, or both.
  • an interaction between the complex and a target nucleic acid, a non-target nucleic acid, or both comprises one or more of: recognition of a target adjacent motif (TAM) sequence within the target nucleic acid by the effector protein, hybridization of the guide nucleic acid to the target nucleic acid, modification of the target nucleic acid and/or the non-target nucleic acid by the effector protein, or combinations thereof. See, e.g. FIG. 2.
  • recognition of a TAM sequence within a target nucleic acid may direct the modification activity of an effector protein.
  • the effector protein described herein may bind and, optionally, modify nucleic acids in a sequence-specific manner. Effector proteins described herein may also modify the target nucleic acid within a target sequence or at a position adjacent to the target sequence. In some embodiments, an effector protein is activated when it binds a certain sequence of a nucleic acid described herein, allowing the effector protein to modify a region of a target nucleic acid that is near, but not adjacent to the target sequence. Effector proteins may modify a nucleic acid by cis cleavage or trans cleavage.
  • effector proteins described herein modify a non-target nucleic acid by trans cleavage on the non-target nucleic acid.
  • effector proteins may have nickase activity or nuclease activity.
  • Effector proteins disclosed herein may modify nucleic acids, including single stranded RNA (ssRNA), double stranded DNA (dsDNA), and single-stranded DNA (ssDNA).
  • the modification of the target nucleic acid generated by an effector protein may result in modulation of the expression of the nucleic acid (e.g., increasing or decreasing expression of the nucleic acid) or modulation of the activity of a translation product of the target nucleic acid (e.g., inactivation of a protein binding to an RNA molecule or hybridization).
  • effector proteins described herein comprise modification activities.
  • the modification activity of the polypeptide described herein may be nickase activity for a double stranded nucleic acid, binding activity, insertion activity, substitution activity, chemical modification activity, or a combination thereof.
  • the modification activity of the polypeptide may result in: cleavage of at least one strand of a target nucleic acid, deletion of one or more nucleotides of a target nucleic acid, insertion of one or more nucleotides into a target nucleic acid, substitution of one or more nucleotides of a target nucleic acid with an alternative nucleotide, chemical modification of one or more nucleotides of a target nucleic acid to an alternative nucleotide, or combinations thereof.
  • the cleavage activity is a nicking activity.
  • An effector protein may be an engineered effector protein having increased modification activity (e.g., catalytic activity) and/or increased substrate binding activity (e.g., substrate selectivity, specificity, and/or affinity) relative to a native effector protein.
  • An effector protein may be an engineered effector protein having reduced modification activity (e.g., a catalytically defective effector protein) or no modification activity (e.g., a catalytically inactive effector protein) relative to a native effector protein.
  • effector proteins described herein comprise one or more functional domains.
  • Effector protein functional domains can include a target adjacent motif (TAM)-interacting domain, an oligonucleotide-interacting domain, one or more recognition domains, a non-target strand interacting domain, an HNH domain, a RuvC domain, or a combination thereof.
  • a TAM interacting domain can be a target strand TAM interacting domain (TPID) or a non-target strand TAM interacting domain (NTPID).
  • TPID target strand TAM interacting domain
  • NTPID non-target strand TAM interacting domain
  • a TAM interacting domain, such as a TPID or a NTPID, on an effector protein describes a region of an effector protein that interacts with target nucleic acid.
  • effector proteins comprise one or more recognition domain (REC domain) with a binding affinity for a guide nucleic acid or for a guide nucleic acid-target nucleic acid heteroduplex.
  • FIG. 1 shows the domains and positions thereof for the parent sequence provided in TABLE 2.
  • the amino terminal PLMP domain is named for its PLMP amino acid motif.
  • the TI domain may form a five-stranded antiparallel 13-barrel, with its carboxy-terminal 136 strand interacting with the 136 strand of the RuvC domain to form a seven-stranded 13-sheet.
  • the PI domain may contain a core 13-barrel.
  • the 132- 133 hairpin in the TID may be inserted into the major groove of the TAM duplex.
  • Publicly available tools, such as InterProScan may be used for detection of functional domains and sequence patterns matching previously described functional domains.
  • effector proteins comprise an amino-terminal PLMP domain, RuvC-like nuclease domains containing three conserved catalytic motifs (RuvC-I-III), with an inserted Arg-rich segment known as the bridge helix (BH), and the HNH nuclease domain.
  • the RuvC-I and RuvC-II motifs may be connected via the BH and a P- hairpin-containing linker (referred to as the REC linker), as well as the Wedge (WED) and TAM-interacting (TI) domains located similarly to the corresponding domains in Cas9.
  • WED Wedge
  • TI TAM-interacting
  • the effector protein comprises an HNH domain. In some embodiments, the effector protein comprises an HNH domain and a RuvC domain. In some embodiments, the effector protein may have a mutation in a nuclease domain. In some embodiments, the nuclease domain is a RuvC domain. In some embodiments, the nuclease domain is an HNH domain.
  • An HNH domain may be characterized as comprising two antiparallel P-strands connected with a loop of varying length, and flanked by an a-helix, with a metal (divalent cation) binding site between the two P-strands.
  • a RuvC domain may be characterized by a six-stranded beta sheet surrounded by four alpha helices, with three conserved subdomains contributing catalytic to the activity of the RuvC domain.
  • a RuvC domain comprises with substrate binding activity, catalytic activity, or both.
  • the RuvC domain may be defined by a single, contiguous sequence, or a set of RuvC subdomains that are not contiguous with respect to the primary amino acid sequence of the protein.
  • An effector protein of the present disclosure may include multiple RuvC subdomains, which may combine to generate a RuvC domain with substrate binding or catalytic activity.
  • an effector protein may include three RuvC subdomains (RuvC-I, RuvC-II, and RuvC-III) that are not contiguous with respect to the primary amino acid sequence of the effector protein but form a RuvC domain once the protein is produced and folds.
  • the effector protein cleaves within the target sequence.
  • the RuvC domain is responsible for cleavage.
  • the HNH domain is responsible for cleavage.
  • such a change or changes can include, for example, deletions, insertions, and/or substitutions.
  • the mutation can refer to a change in structure of an amino acid residue or nucleotide relative to the starting or reference residue or nucleotide.
  • a mutation of an amino acid residue includes, for example, deletions, insertions and substituting one amino acid residue for a structurally different amino acid residue.
  • substitutions can be a conservative substitution, a non-conservative substitution, a substitution to a specific sub-class of amino acids, or a combination thereof as described herein.
  • a mutation of a nucleotide includes, for example, changing one naturally occurring base for a different naturally occurring base, such as changing an adenine to a thymine or a guanine to a cytosine or an adenine to a cytosine or a guanine to a thymine.
  • a mutation of a nucleotide base may result in a structural and/or functional alteration of the encoding peptide, polypeptide or protein by changing the encoded amino acid residue of the peptide, polypeptide or protein.
  • a mutation of a nucleotide base may not result in an alteration of the amino acid sequence or function of encoded peptide, polypeptide or protein, also known as a silent mutation.
  • TABLE 1 and TABLE 8 provide illustrative amino acid sequences of an engineered effector protein for use in the compositions, systems and methods described herein.
  • TABLE 6 provides exemplary effector proteins that may be engineered.
  • systems, compositions, and methods described herein comprise an effector protein in TABLE 6 or a use thereof.
  • systems, compositions, and methods described herein comprise an engineered variant of an effector protein in TABLE 6 or a use thereof.
  • compositions, systems and methods described herein comprise an effector protein, or a nucleic acid encoding the effector protein, wherein the amino acid sequence of the effector protein comprises at least 200 contiguous amino acids or more of the amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8, wherein the amino acid sequence of the effector protein is not identical to any one of the amino acid sequences selected from TABLE 1, TABLE 6, and TABLE 8.
  • the amino acid sequence of an effector protein provided herein comprises at least 200, at least 220, at least 240, at least 260, at least 280, at least 300, at least 320, at least 340, at least 360, at least 380, at least 400 contiguous amino acids, at least 420 contiguous amino acids, at least 440 contiguous amino acids, at least 460 contiguous amino acids, at least 480 contiguous amino acids, at least 500 contiguous amino acids of a sequence selected from TABLE 1, TABLE 6, and TABLE 8.
  • the length of the effector protein is about 480 to about 560 amino acids.
  • the amino acid sequence of an effector protein provided herein comprises about 100, about 120, about 140, about 160, about 180, about 200, about 220, about 240, about 260, about 280, about 300, about 320, about 340, about 360, about 380, about 400, about 420, about 440, about 460, about 480, about 500, about 520, about 540 contiguous amino acids of a sequence selected from TABLE 1, TABLE 6, and TABLE 8.
  • the length of the effector protein is less than 600, less than 580, less than 560, less than 540, less than 520, less than 500, less than 480, less than 460, less than 440, less than 420, or less than 400 contiguous amino acids; and at least 300, at least 320, at least 340, at least 360, at least 380 contiguous amino acids of a sequence selected from TABLE 1, TABLE 6, and TABLE 8 [0121]
  • compositions, systems and methods described herein comprise an effector protein or a nucleic acid encoding the effector protein, wherein the effector protein comprises a portion of the amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8.
  • the effector protein comprises a portion of the amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8, wherein the portion does not comprise at least the first 10 amino acids, at least the first 20 amino acids, at least the first 40 amino acids, at least the first 60 amino acids, at least the first 80 amino acids, at least the first 100 amino acids, at least the first 120 amino acids, at least the first 140 amino acids, at least the first 160 amino acids, at least the first 180 amino acids, or at least the first 200 amino acids of the amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8.
  • the effector protein comprises a portion of the amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8, wherein the portion does not comprise the last 10 amino acids, the last 20 amino acids, the last 40 amino acids, the last 60 amino acids, the last 80 amino acids, the last 100 amino acids, the last 120 amino acids, the last 140 amino acids, the last 160 amino acids, the last 180 amino acids, or the last 200 amino acids of the amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8.
  • compositions, systems, and methods described herein comprise an effector protein, or a nucleic acid encoding the effector protein, wherein the effector protein comprises an amino acid sequence that is at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, but less than 100% identical to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8.
  • an effector protein provided herein comprises an amino acid sequence that is at least 65% but less than 100% identical to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8.
  • an effector protein provided herein comprises an amino acid sequence that is at least 70% but less than 100% identical to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, an effector protein provided herein comprises an amino acid sequence that is at least 75% but less than 100% identical to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, an effector protein provided herein comprises an amino acid sequence that is at least 80% but less than 100% identical to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, an effector protein provided herein comprises an amino acid sequence that is at least 85% but less than 100% identical to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8.
  • an effector protein provided herein comprises an amino acid sequence that is at least 90% but less than 100% identical to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, an effector protein provided herein comprises an amino acid sequence that is at least 95% but less than 100% identical to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, an effector protein provided herein comprises an amino acid sequence that is at least 97% but less than 100% identical to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, an effector protein provided herein comprises an amino acid sequence that is at least 98% but less than 100% identical to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8.
  • an effector protein provided herein comprises an amino acid sequence that is at least 99% but less than 100% identical to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, an effector protein provided herein comprises an amino acid sequence that is less than 100% identical to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8
  • compositions, systems, and methods described herein comprise an effector protein, or a nucleic acid encoding the effector protein, wherein the effector protein comprises an amino acid sequence that is at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% similar, but not the same, to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8.
  • an effector protein provided herein comprises an amino acid sequence that is at least 80% similar, but not the same, to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8.
  • an effector protein provided herein comprises an amino acid sequence that is at least 85% similar, but not the same, to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, an effector protein provided herein comprises an amino acid sequence that is at least 90% similar, but not the same, to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, an effector protein provided herein comprises an amino acid sequence that is at least 95% similar, but not the same, to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, an effector protein provided herein comprises an amino acid sequence that is at least 97% similar, but not the same, to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8.
  • an effector protein provided herein comprises an amino acid sequence that is at least 98% similar, but not the same, to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, an effector protein provided herein comprises an amino acid sequence that is at least 99% similar, but not the same, to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, an effector protein provided herein comprises an amino acid sequence that is 100% similar, but not the same, to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8
  • compositions, systems, and methods described herein comprise an effector protein, or a nucleic acid encoding the effector protein, wherein the effector protein comprises one or more amino acid alterations relative to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8.
  • the one or more alterations comprises at least one, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least twelve, at least sixteen, at least twenty, or more amino acid alterations relative to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8.
  • the one or more alterations comprises one to twenty, one to sixteen, one to twelve, one to eight, one to four, four to twenty, four to sixteen, four to twelve, four to eight, eight to twenty, eight to sixteen, eight to twelve, twelve to twenty, twelve to sixteen, sixteen to twenty, or more amino acid alterations relative to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8.
  • the one or more amino acid alterations comprises substitutions (e.g., conservative substitutions, nonconservative substitutions), deletions, or combinations thereof.
  • an effector protein or a nucleic acid encoding the effector protein comprises 1 amino acid alteration, 2 amino acid alterations, 3 amino acid alterations, 4 amino acid alterations, 5 amino acid alterations, 6 amino acid alterations, 7 amino acid alterations, 8 amino acid alterations, 9 amino acid alterations, 10 amino acid alterations or more relative to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8.
  • compositions, systems, and methods described herein comprise an effector protein, or a nucleic acid encoding the effector protein, wherein the effector protein comprises one or more substitutions relative to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8.
  • the one or more substitutions comprises at least one, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least twelve, at least sixteen, at least twenty, or more substitutions relative to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8.
  • the one or more substitutions comprises one to twenty, one to sixteen, one to twelve, one to eight, one to four, four to twenty, four to sixteen, four to twelve, four to eight, eight to twenty, eight to sixteen, eight to twelve, twelve to twenty, twelve to sixteen, sixteen to twenty, or more substitutions relative to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8.
  • the one or more substitutions comprises 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more amino acid substitutions relative to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 86.
  • the one or more amino acid substitutions comprise one, two, three, four, five, six, seven, eight, nine, ten or more substitutions relative to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8.
  • the one or more amino acid substitutions comprise one or more substitutions with a positively charged amino acid residues.
  • the positively charged amino acid residue is independently selected from Lys (K), Arg (R), or His (H).
  • the one or more substitutions comprise one or more conservative substitutions, one or more nonconservative substitutions, or combinations thereof.
  • compositions, systems, and methods described herein comprise an effector protein, or a nucleic acid encoding the effector protein, wherein the effector protein comprises one or more conservative substitutions relative to any one of the sequences selected from TABLE 1, TABLE 6, and TABLE 8.
  • the one or more conservative substitutions comprises at least one, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least twelve, at least sixteen, at least twenty, or more conservative substitutions relative to any one of the sequences selected from TABLE 1, TABLE 6, and TABLE 8.
  • the one or more conservative substitutions comprises one to twenty, one to sixteen, one to twelve, one to eight, one to four, four to twenty, four to sixteen, four to twelve, four to eight, eight to twenty, eight to sixteen, eight to twelve, twelve to twenty, twelve to sixteen, or sixteen to twenty conservative substitutions relative to any one of the sequences selected from TABLE 1, TABLE 6, and TABLE 8.
  • the one or more conservative substitutions comprise one, two, three, four, five, six, seven, eight, nine, ten or more conservative substitutions relative to any one of the sequences selected from TABLE 1, TABLE 6, and TABLE 8
  • compositions, systems, and methods described herein comprise an effector protein, or a nucleic acid encoding the effector protein, wherein the effector protein comprises one or more non-conservative substitutions relative to any one of the sequences selected from TABLE 1, TABLE 6, and TABLE 8.
  • the one or more conservative substitutions comprises not more than one, two, three, four, five, six, seven, eight, nine, or ten substitutions in the amino acid sequence relative to any one of the sequences selected from TABLE 1, TABLE 6, and TABLE 8.
  • the one or more non-conservative substitutions are in a RuvC domain, HNH domain, or both.
  • the non-conservative substitution(s) replaces a catalytic residue(s) of the domain(s).
  • compositions, systems, and methods described herein comprise an effector protein, or a nucleic acid encoding the effector protein, wherein the effector protein comprises an amino acid sequence that is at least 90%, at least 95%, at least 97%, at least 98%, or at least 99% identical to SEQ ID NO: 624, wherein the effector protein comprises an amino acid substitution of H246A relative to SEQ ID NO: 624
  • the H246A amino acid substitution is believed to be located in an HNH domain of effector protein of SEQ ID NO: 624, and thereby provides the engineered variant with nickase activity.
  • compositions, systems, and methods described herein comprise an effector protein, or a nucleic acid encoding the effector protein, wherein the effector protein comprises an amino acid sequence that is at least 90%, at least 95%, at least 97%, at least 98%, or at least 99% identical to SEQ ID NO: 713, wherein the effector protein comprises an amino acid substitution of H244A relative to SEQ ID NO: 713.
  • compositions, systems, and methods described herein comprise an effector protein, or a nucleic acid encoding the effector protein, wherein the effector protein comprises an amino acid sequence that is at least 90%, at least 95%, at least 97%, at least 98%, or at least 99% identical to SEQ ID NO: 856, wherein the effector protein comprises an amino acid substitution of H248A relative to SEQ ID NO: 856.
  • effector proteins are engineered from the parent sequence provided in TABLE 2. Any one of the effector proteins in TABLE 6 could be a parent sequence that is similarly engineered. In some embodiments, any one of the sequences replaced (described by positions replaced in TABLE 2) could be replaced with a similar domain from another protein. For example, where HNH domains were swapped as described in TABLE 2, HNH domains from other proteins could similar be inserted in those positions. In some instances, the sequences inserted as described in TABLE 2 are nearly identical to said sequences.
  • sequences inserted could be at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99% to the sequence provided in TABLE 2.
  • the sequences inserted are similar to the sequences described in TABLE 2.
  • sequences inserted are at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99% to the sequence provided in TABLE 2.
  • effector proteins comprise a combination of sequences replaced, as described in TABLE 2.
  • an effector protein may comprise the parent sequence with the exception of an HNH domain replacement described in TABLE 2 and a TID domain replacement described in TABLE 2.
  • At least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99% or 100% of a domain in the parent sequence of TABLE 2 is replaced by an inserted amino acid sequence.
  • additional amino acids around a domain are deleted or replaced.
  • less than 10, less than 50, less than 100, less than 150, or less than 200 amino acids around a domain are deleted or replaced.
  • effector proteins comprise one or more amino acid alterations relative to the sequences provided in TABLE 1, TABLE 6, and TABLE 8.
  • the one or more amino acid alterations may result in a change in activity of the effector protein relative to a naturally-occurring counterpart.
  • the one or more amino acid alterations does not result in a change in activity of the effector protein relative to a naturally-occurring counterpart.
  • the one or more amino acid alteration increases or decreases catalytic activity of the effector protein relative to a naturally-occurring counterpart.
  • the one or more amino acid alteration increases or decreases binding activity of the effector protein relative to a naturally- occurring counterpart.
  • an effector protein that has decreased catalytic activity may be referred to as catalytically or enzymatically inactive, catalytically or enzymatically dead, as a dead protein or a dCas protein.
  • a protein may comprise an enzymatically inactive domain (e.g. inactive nuclease domain).
  • a nuclease domain e.g., RuvC domain or HNH domain
  • an effector protein may be deleted or mutated relative to a wildtype counterpart so that it is no longer functional or comprises reduced nuclease activity.
  • a catalytically inactive effector protein may bind to a guide nucleic acid and/or a target nucleic acid but does not cleave the target nucleic acid.
  • a catalytically inactive effector protein may associate with a guide nucleic acid to activate or repress transcription of a target nucleic acid.
  • a catalytically inactive effector protein is fused to an effector partner that confers an alternative activity to an effector protein activity. Such fusion proteins are described herein and throughout.
  • effector proteins of the present disclosure may exhibit activity or enhanced activity in the presence of a co-factor.
  • the co-factor allows the effector proteins to perform a function.
  • the function is pre-crRNA processing and/or target nucleic acid cleavage.
  • Cas9 uses divalent metal ions as co-factors. The suitability of a divalent metal ion as a cofactor can easily be assessed, such as by methods based on those described by Sundaresan et al. (Cell Rep. 2017 Dec 26; 21(13): 3728-3739).
  • the co-factor is a divalent metal ion.
  • Non-limiting exemplary divalent metal ions include: Mg2+, Mn2+, Zn2+, Ca2+, and Cu2+.
  • the effector protein forms a complex with a divalent metal ion.
  • the effector protein forms a complex with Mg2+, Mn2+, Zn2+, Ca2+, or Cu2+.
  • Effector proteins of the present disclosure may be synthesized, using any suitable method.
  • the effector proteins may be produced in vitro or by eukaryotic cells or by prokaryotic cells.
  • the effector proteins may be further processed by unfolding (e.g. heat denaturation, dithiothreitol reduction, etc.) and may be further refolded, using any suitable method.
  • the nucleic acid(s) encoding the effector proteins described herein, the recombinant nucleic acid(s) described herein, the vectors described herein may be produced in vitro or in vivo by eukaryotic cells or by prokaryotic cells.
  • effector proteins described herein are isolated from cell lysate.
  • the compositions described herein may comprise 20% or more by weight, 75% or more by weight, 95% or more by weight, or 99.5% or more by weight of an effector protein, related to the method of preparation of compositions described herein and its purification thereof, wherein percentages may be upon total protein content in relation to contaminants.
  • the effector protein is at least 80% pure, at least 85% pure, at least 90% pure, at least 95% pure, at least 98% pure, or at least 99% pure (e.g., free of contaminants, non-engineered proteins or other macromolecules, etc.).
  • TAM Target Adjacent Motif
  • Effector proteins of the present disclosure may cleave or nick a target nucleic acid within or near a target adjacent motif (TAM) sequence of the target nucleic acid.
  • the TAM comprises 4-8 nucleotides.
  • the length of the TAM is 4 nucleotides.
  • the length of the TAM is 5 nucleotides.
  • the length of the TAM is 6 nucleotides.
  • the length of the TAM is 7 nucleotides.
  • the length of the TAM is 8 nucleotides.
  • the target nucleic acid is a double stranded nucleic acid comprising a target strand and a non-target strand.
  • cleavage occurs within 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, or 50 nucleotides of a 5’ or 3’ terminus of a TAM sequence.
  • effector proteins described herein recognize a TAM sequence.
  • recognizing a TAM sequence comprises interacting with a sequence adjacent to the TAM.
  • a target nucleic acid comprises a target sequence that is adjacent to a TAM sequence.
  • the effector protein does not require a TAM to bind and/or cleave a target nucleic acid.
  • a target nucleic acid is a double stranded nucleic acid comprising a target strand and a non-target strand, wherein the target strand comprises a target sequence.
  • the TAM sequence is located on the target strand.
  • the TAM sequence is located on the non-target strand.
  • the TAM sequence described herein is directly adjacent to the target sequence on the non-target strand.
  • the TAM sequence described herein is adjacent (e.g., within 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, or 50 nucleotides) to the target sequence on the non-target strand.
  • the TAM sequence is located 3’ of the target sequence on the non-target strand. In some embodiments, such a TAM described herein is directly adjacent to the target sequence on the target strand or the non-target strand.
  • an RNP cleaves the target strand or the non-target strand. In some embodiments, the RNP cleaves both, the target strand and the non-target strand.
  • an RNP recognizes the TAM sequence, and hybridizes to a target sequence of the target nucleic acid. In some embodiments, the RNP cleaves the target nucleic acid, wherein the RNP has recognized the TAM sequence and is hybridized to the target sequence. In some embodiments, at least a portion of the guide nucleic acid (e.g., the spacer sequence) hybridizes to the target sequence of the target strand.
  • an effector protein described herein, or a multimeric complex thereof recognizes a TAM on a target nucleic acid.
  • multiple effector proteins of the multimeric complex recognize a TAM on a target nucleic acid.
  • at least two of the multiple effector proteins recognize the same TAM sequence.
  • at least two of the multiple effector proteins recognize different TAM sequences.
  • only one effector protein of the multimeric complex recognizes a TAM on a target nucleic acid.
  • An effector protein of the present disclosure may cleave or nick a target nucleic acid within or near a target adjacent motif (TAM) sequence of the target nucleic acid.
  • TAM target adjacent motif
  • cleavage occurs within 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 nucleotides of a 5’ or 3’ terminus of a TAM sequence.
  • Engineered proteins in TABLE 1, TABLE 6 and TABLE 8 may recognize a target adjacent motif (TAM) of 5’-NWRRNA-3’, wherein N is any nucleotide, W is A or T, and R is A or G.
  • TAM target adjacent motif
  • engineered proteins in TABLE 1, TABLE 6 and TABLE 8 may recognize a target sequence that is adjacent to a TAM of 5’-ATAANNN-3’, wherein N is any nucleotide.
  • an effector protein described herein comprises an amino acid sequence at least 90-100% identical to SEQ ID NO: 856 and recognizes a TAM of ATAANNN when complexed with a guide nucleic acid, wherein N is any nucleotide.
  • an effector protein described herein comprises an amino acid sequence at least 90-100% identical to SEQ ID NO: 624 and recognizes a TAM of ARRRGNN when complexed with a guide nucleic acid, wherein N is any nucleotide and R is A or G.
  • an effector protein described herein comprises an amino acid sequence at least 90-100% identical to SEQ ID NO: 713 and recognizes a TAM of GNAAANN when complexed with a guide nucleic acid, wherein N is any nucleotide. In some embodiments, an effector protein described herein comprises an amino acid sequence at least 90-100% identical to SEQ ID NO: 833 and recognizes a TAM of ATAANNN when complexed with a guide nucleic acid, wherein N is any nucleotide.
  • an effector protein described herein comprises an amino acid sequence at least 90-100% identical to SEQ ID NO: 691 and recognizes a TAM of GYARRNN when complexed with a guide nucleic acid, wherein N is any nucleotide, Y is C or T, and R is A or G.
  • an effector protein described herein comprises an amino acid sequence at least 90-100% identical to SEQ ID NO: 774 and recognizes a TAM of RTANNNN when complexed with a guide nucleic acid, wherein N is any nucleotide and R is A or G.
  • compositions, systems, and methods comprising one or more effector partners or uses thereof.
  • the effector partner is a heterologous protein an effector protein described herein.
  • the effector partner is not an effector protein as described herein.
  • the effector partner is capable of imparting a function or activity that is not provided by an effector protein as described herein.
  • the effector partner comprises a second effector protein or a multimeric form thereof.
  • an effector partner imparts a function or activity to a fusion protein comprising an effector protein that is not provided by the effector protein, including but not limited to nuclease activity, methyltransferase activity, demethylase activity, DNA repair activity, DNA damage activity, deamination activity, dismutase activity, alkylation activity, depurination activity, oxidation activity, dimer forming activity (e.g., pyrimidine dimer forming activity), integrase activity, transposase activity, recombinase activity, polymerase activity, ligase activity, helicase activity, photolyase activity, glycosylase activity, acetyltransferase activity, deacetylase activity, kinase activity, phosphatase activity, ubiquitin ligase activity, deubiquitinating activity, adenylation activity, deadenylation activity, SUMOylating activity, deSUMOyl
  • the effector partner is fused or linked to an effector protein described herein.
  • the amino terminus of the effector partner is linked to the carboxy terminus of the effector protein directly or by a linker.
  • the carboxy terminus of the effector partner is linked to the amino terminus of the effector protein directly or by a linker.
  • the effector partner may be functional when the effector protein is coupled to a guide nucleic acid.
  • the effector partner may be functional when the effector protein is coupled to a target nucleic acid.
  • the guide nucleic acid imparts sequence specific activity to the effector partner.
  • the effector protein may comprise a catalytically inactive effector protein (e.g., a catalytically inactive variant of an effector protein described herein) when fused or linked to an effector partner.
  • the effector partner may directly or indirectly edit a target nucleic acid. Edits can be of a nucleobase, nucleotide, or nucleotide sequence of a target nucleic acid.
  • the effector partner may interact with additional proteins, or functional fragments thereof, to make modifications to a target nucleic acid.
  • the effector partner may modify proteins associated with a target nucleic acid.
  • an effector partner may modulate transcription (e.g., inhibits transcription, increases transcription) of a target nucleic acid.
  • an effector partner may directly or indirectly inhibit, reduce, activate or increase expression of a target nucleic acid.
  • RT Reverse Transcriptase
  • systems and methods comprise components or uses of an RT editing system to modify a target nucleic acid.
  • RT editing may also be referred to as precision editing or precise nucleobase editing.
  • an RT editing system comprises an effector protein and an effector partner comprising an RT editing enzyme.
  • an RT editing enzyme comprises a polymerase.
  • an RT editing enzyme comprises a reverse transcriptase.
  • a non-limiting example of a reverse transcriptase is an M-MLV RT enzyme and variants thereof having polymerase activity.
  • the M-MLV RT enzyme comprises at least one mutation selected from D200N, L603W, T330P, T306K, and W313F relative to wildtype M-MLV RT enzyme.
  • systems and methods comprise an RT editing enzyme, wherein the RT editing enzyme is not fused or linked to the effector protein. See, e.g. FIG. 3.
  • the RT editing enzyme comprises a recruiting moiety that recruits the RT editing enzyme to the target nucleic acid.
  • the RT editing enzyme may comprise a peptide that binds an aptamer, wherein the aptamer is located on a guide RNA, template RNA, or combination thereof.
  • the RT editing enzyme may be linked to a protein that binds to (or is bound by) the effector protein or a protein linked/fused to the effector protein.
  • the RT editing enzyme may be linked to an MS2 coat protein that binds an MS2 aptamer, wherein the aptamer is located on a guide RNA, template RNA, or combination thereof. See, e.g. FIG. 3.
  • an RT editing enzyme may require an RT editing guide RNA (referred to herein as a “pegRNA” or “rtgRNA”) to catalyze editing.
  • Such a pegRNA may be capable of identifying a target nucleotide or target sequence in a target nucleic acid to be edited and encoding a new genetic information that replaces the target nucleotide or target sequence in the target nucleic acid.
  • An RT editing enzyme may require a pegRNA and a guide RNA, such as a single guide RNA, to catalyze the editing.
  • the RT editing system comprises a template RNA comprising a primer binding sequence that hybridizes to a primer sequence of the dsDNA molecule that is formed when target nucleic acid is cleaved, and a template sequence that is complementary to at least a portion of the target sequence of the dsDNA molecule except for at least one nucleotide.
  • template RNA refers to a nucleic acid comprising: a primer binding sequence and a template sequence.
  • the primer biding sequence and template binding sequence are linked to an MS2 aptamer.
  • the RT editing enzyme is linked to an MS2 coat protein that is capable of binding the MS2 aptamer, thereby localizing the RT editing enzyme to the nicked DNA.
  • template RNAs may comprise DNA, RNA, or a combination thereof (e.g., RNA with a thymine base).
  • the template RNA is linked to a guide RNA via a linker sequence to form an rtgRNA.
  • the template RNA is covalently linked to a guide RNA (referred to herein as an extended guide RNA or rtgRNA).
  • extended guide RNA rtgRNA
  • rtgRNA extended guide RNA
  • rtgRNA extended guide RNA
  • the orientation of the rtgRNA from 5’ to 3’ is: guide nucleic acid, optional linker, and template RNA.
  • the orientation of the rtgRNA from 5’ to 3’ is: template RNA, linker, and guide RNA.
  • extended guide RNAs may comprise DNA, RNA, or a combination thereof (e.g., RNA with a thymine base).
  • rtgRNA and pegRNA are used interchangeably herein.
  • the template RNA is not covalently linked to a guide RNA.
  • a spacer sequence, a repeat or handle sequence, a primer binding sequence, and a template sequence are comprised in two polynucleotides - the spacer and repeat/handle sequence comprised in a first polynucleotide and the primer binding sequence and the template sequence comprised in a second polynucleotide, referred to herein as a split RNA.
  • a split RNA See e.g., FIG. 3.
  • System components shown in FIG. 3 are exemplary and non-limiting.
  • the guide RNA and/or retRNA may comprise additional nucleotides beyond those labeled as spacer, repeat/handle, linker, PBS and RT template (RTT).
  • the first polynucleotide comprises a spacer sequence and a handle sequence (also referred to herein as a scaffold sequence).
  • the spacer sequence precedes the handle sequence in a 5’ to 3’ direction. See e.g., FIG. 3.
  • the spacer(s) and scaffold sequence(s) are linked directly to one another.
  • a linker is present between the spacer(s) and scaffold sequence(s). Linkers may be any suitable linker.
  • the first polynucleotide comprises a spacer sequence and a handle sequence.
  • the first polynucleotide is a guide RNA as described herein.
  • the second polynucleotide comprises a primer binding sequence and a template sequence (e.g., an retRNA).
  • the second polynucleotide further comprises an aptamer that is recognized by a biological tether protein linked to an RT editing enzyme described herein.
  • the aptamer is an MS2 aptamer (See Said et al (November 2009). "In vivo expression and purification of aptamer-tagged small RNA regulators". Nucleic Acids Research. 37 (20): el33; and Johansson et al (1997). "RNA recognition by the MS2 phage coat protein". Seminars in Virology. 8 (3): 176-185).
  • the second polynucleotide comprises, from 5’ to 3’, an aptamer sequence, a template sequence, and a primer binding sequence. See FIG. 3.
  • the MS2 aptamer is an exemplary sequence and an RNA sequence with the appropriate secondary structure may also be used instead of the aptamer.
  • the second polynucleotide is circularized. See FIG. 3.
  • the present disclosure provides a split gRNA system, comprising a first polynucleotide comprising a spacer sequence and a handle sequence (e.g., a gRNA) and a second polynucleotide comprising a primer binding sequence and a template sequence (e.g., an retRNA), and an effector protein (e.g., effector proteins provided in TABLE 1, TABLE 6, and TABLE 8), and an RT editing enzyme (e.g., M-MLV RT enzyme), or nucleic acids encoding the same.
  • a split gRNA system comprising a first polynucleotide comprising a spacer sequence and a handle sequence (e.g., a gRNA) and a second polynucleotide comprising a primer binding sequence and a template sequence (e.g., an retRNA), and an effector protein (e.g., effector proteins provided in TABLE 1, TABLE 6, and TABLE 8), and an RT editing enzyme
  • the template RNA hybridizes to the target nucleic acid.
  • the target nucleic acid is a dsDNA molecule.
  • at least a portion of the template RNA hybridizes to a first strand of the target nucleic acid and at least a portion of the guide RNA hybridizes to a second strand of the target nucleic acid. See e.g., FIG. 3.
  • the pegRNA comprises: a guide RNA comprising a second region that is bound by the effector protein, and a first region comprising a spacer sequence that is complementary to a target sequence of the dsDNA molecule; and a template RNA comprising a primer binding sequence that hybridizes to a primer sequence of the dsDNA molecule that is formed when target nucleic acid is cleaved, and a template sequence that is complementary to at least a portion of the target sequence of the dsDNA molecule with the exception of at least one nucleotide.
  • At least one nucleotide is incorporated into the target nucleic acid by activity of the RT editing enzyme, thereby modifying the target nucleic acid.
  • the spacer sequence is complementary to the target sequence on a target strand of the dsDNA molecule.
  • the spacer sequence is complementary to the target sequence on a non-target strand of the dsDNA molecule.
  • the primer binding sequence hybridizes to a primer sequence on the non-target strand of the dsDNA molecule.
  • the primer binding sequence hybridizes to a primer sequence on the target strand of the dsDNA molecule.
  • the target strand is cleaved.
  • the non-target strand is cleaved.
  • effector partners have enzymatic activity that modifies a nucleic acid, such as a target nucleic acid.
  • the target nucleic acid may comprise or consist of a ssRNA, dsRNA, ssDNA, or a dsDNA.
  • nuclease activity which comprises the enzymatic activity of an enzyme which allows the enzyme to cleave the phosphodiester bonds between the nucleotide subunits of nucleic acids, such as that provided by a restriction enzyme, or a nuclease (e.g., FokI nuclease); methyltransferase activity such as that provided by a methyltransferase (e.g., Hhal DNA m5c-methyltransferase (M.Hhal), DNA methyltransferase 1 (DNMT1), DNA methyltransferase 3 a (DNMT3a), DNA methyltransferase 3b (DNMT3b), METI, DRM3 (plants), ZMET2, CMT1, CMT2 (plants)); demethylase activity such as that provided by a demethylase (e.g., Ten-Eleven Translocation
  • effector partners target a ssRNA, dsRNA, ssDNA, or a dsDNA.
  • effector partners target ssRNA.
  • Non-limiting examples of effector partners for targeting ssRNA include, but are not limited to, splicing factors (e.g.
  • RS domains protein translation components (e.g., translation initiation, elongation, and/or release factors; e.g., eIF4G); RNA methylases; RNA editing enzymes (e.g., RNA deaminases, e.g., adenosine deaminase acting on RNA (ADAR), including A to I and/or C to U editing enzymes); helicases; and RNA-binding proteins.
  • RNA editing enzymes e.g., RNA deaminases, e.g., adenosine deaminase acting on RNA (ADAR), including A to I and/or C to U editing enzymes
  • helicases e.g., adenosine deaminase acting on RNA (ADAR), including A to I and/or C to U editing enzymes
  • helicases e.g., adenosine deaminase acting on RNA (ADA
  • an effector partner may include an entire protein, or in some embodiments, may include a fragment of the protein (e.g., a functional domain).
  • the functional domain binds or interacts with a nucleic acid, such as ssRNA, including intramolecular and/or intermolecular secondary structures thereof (e.g., hairpins, stem -loops, etc.).
  • the functional domain may interact transiently or irreversibly, directly, or indirectly.
  • a functional domain comprises a region of one or more amino acids in a protein that is required for an activity of the protein, or the full extent of that activity, as measured in an in vitro assay.
  • Activities include but are not limited to nucleic acid binding, nucleic acid editing, nucleic acid mutating, nucleic acid modifying, nucleic acid cleaving, protein binding or combinations thereof.
  • effector partners may comprise a protein or domain thereof selected from: endonucleases (e.g., RNase III, the CRR22 DYW domain, Dicer, and PIN (PilT N-terminus); SMG5 and SMG6; domains responsible for stimulating RNA cleavage (e.g., CPSF, CstF, CFIm and CFIIm); exonucleases such as XRN-1 or Exonuclease T; deadenylases such as HNT3; protein domains responsible for nonsense mediated RNA decay (e.g., UPF1, UPF2, UPF3, UPF3b, RNP SI, Y14, DEK, REF2, and SRml60); protein domains responsible for stabilizing RNA (e.g., PABP); proteins and protein domains responsible for polyadenylation of RNA (e.g., PAP1, GLD-2, and Star- PAP); proteins and protein domains responsible for polyuridinylation of
  • effector partner may comprise a chromatin-modifying enzyme.
  • the effector partner chemically modifies a target nucleic acid, for example by methylating, demethylating, or acetylating the target nucleic acid in a sequence specific or non-specific manner.
  • effector partners edit a nucleobase of a target nucleic acid.
  • Such a effector partner may be referred to as a base editing enzyme.
  • a base editing enzyme variant that differs from a naturally occurring base editing enzyme, but it is understood that any reference to a base editing enzyme herein also refers to a base editing enzyme variant.
  • a base editor is a system comprising an effector protein and a base editing enzyme.
  • the base editor comprises a base editing enzyme and an effector protein as independent components.
  • the base editor comprises a fusion protein comprising a base editing enzyme fused or linked to an effector protein.
  • the amino terminus of the effector partner is linked to the carboxy terminus of the effector protein by the linker.
  • the carboxy terminus of the effector partner is linked to the amino terminus of the effector protein by the linker.
  • the base editor may be functional when the effector protein is coupled to a guide nucleic acid.
  • the base editor may be functional when the effector protein is coupled to a target nucleic acid.
  • the guide nucleic acid imparts sequence specific activity to the base editor.
  • the effector protein may comprise a catalytically inactive effector protein (e.g., a catalytically inactive variant of an effector protein described herein).
  • the base editing enzyme may comprise deaminase activity. Additional base editors are described herein.
  • base editing enzymes are capable of catalyzing editing (e.g., a chemical modification) of a nucleobase of a nucleic acid molecule, such as DNA or RNA (single stranded or double stranded).
  • a base editing enzyme and therefore a base editor, is capable of converting an existing nucleobase to a different nucleobase, such as: an adenine (A) to guanine (G); cytosine (C) to thymine (T); cytosine (C) to guanine (G); uracil (U) to cytosine (C); guanine (G) to adenine (A); hydrolytic deamination of an adenine or adenosine, or methylation of cytosine (e.g., CpG, CpA, CpT or CpC).
  • base editing enzymes edit a nucleobase on a ssDNA.
  • base editing enzymes edit a nucleobase on both strands of dsDNA.
  • base editing enzymes edit a nucleobase of an RNA.
  • a base editing enzyme itself may or may not bind to the nucleic acid molecule containing the nucleobase.
  • a base editing enzyme upon binding to its target locus in the target nucleic acid (e.g., a DNA molecule), base pairing between the guide nucleic acid and target strand leads to displacement of a small segment of ssDNA in an “R-loop”.
  • DNA bases within the R-loop are edited by the base editing enzyme having the deaminase enzyme activity.
  • base editing systems for improved efficiency in eukaryotic cells comprise a base editing enzyme, and a catalytically inactive effector protein that may generate a nick in the non-edited strand and induce repair of the nonedited strand using the edited strand as a template.
  • a base editing enzyme comprises a deaminase enzyme.
  • exemplary deaminases are described in US20210198330, WO2021041945,
  • deaminase domains are described WO 2018027078 and WO20 17070632, and each are hereby incorporated in its entirety by reference.
  • additional exemplary deaminase domains are described in Komor et al., Nature, 533, 420-424 (2016); Gaudelli et al., Nature, 551, 464-471 (2017); Komor et al., Science Advances, 3:eaao4774 (2017), and Rees et al., Nat Rev Genet. 2018 Dec;19(12):770-788.
  • the deaminase functions as a monomer. In some embodiments, the deaminase functions as heterodimer with an additional protein.
  • base editing enzymes comprise a DNA glycosylase inhibitor (e.g. , an uracil glycosylase inhibitor (UGI) or uracil N-glycosylase (UNG)).
  • the effector partner is a deaminase, e.g., ADAR1/2, ADAR-2, AID, or any functional variant thereof.
  • the base editor is a cytosine base editor (CBE), wherein the base editing enzyme is a cytosine base editing enzyme.
  • the cytosine base editing enzyme and therefore CBE, may convert a cytosine to a thymine.
  • a cytosine base editing enzyme may accept ssDNA as a substrate but may not be capable of cleaving dsDNA, wherein the CBE comprises a catalytically inactive effector protein.
  • the catalytically inactive effector protein of the CBE when bound to its cognate DNA, may perform local denaturation of the DNA duplex to generate an R-loop in which the DNA strand not paired with a guide nucleic acid exists as a disordered single-stranded bubble.
  • the catalytically inactive effector protein generated ssDNA R-loop may enable the CBE to perform efficient and localized cytosine deamination in vitro. In some embodiments, deamination activity is exhibited in a window of about 4 to about 10 base pairs.
  • the catalytically inactive effector protein presents a target site to the cytosine base editing enzyme in high effective molarity, which may enable the CBE to deaminate cytosines located in a variety of different sequence motifs, with differing efficacies.
  • the CBE is capable of mediating RNA-programmed deamination of target cytosines in vitro or in vivo.
  • the cytosine base editing enzyme is a cytidine deaminase.
  • the cytosine base editing enzyme is a cytosine base editing enzyme described by Koblan et al. (2016) Nature Biotechnology 36:848-846; Komor et al.
  • the effector partner comprises a uracil glycosylase inhibitor (UGI).
  • the CBE described herein comprises UGI.
  • Base excision repair (BER) of U «G in DNA is initiated by a uracil N-glycosylase (UNG), which recognizes a U»G mismatch and cleaves the glycosidic bond between a uracil and a deoxyribose backbone of DNA.
  • UNG uracil N-glycosylase
  • the UNG may be inhibited by fusion of a UGI to the effector protein.
  • the UGI is a small protein from bacteriophage PBS. In some embodiments, the UGI is a DNA mimic that potently inhibits both human and bacterial UNG. In some embodiments, the UGI inhibitor is any protein or polypeptide that inhibits UNG.
  • the CBE described herein may mediate efficient base editing in bacterial cells and moderately efficient editing in mammalian cells, enabling conversion of a OG base pair to a T»A base pair through a U»G intermediate.
  • the CBE is modified to increase base editing efficiency while editing more than one strand of DNA.
  • the CBE described herein nicks a non-edited DNA strand.
  • the non-edited DNA strand nicked by the CBE biases cellular repair of a U»G mismatch to favor a U»A outcome, elevating base editing efficiency.
  • a base editor described herein comprising one or more base editing enzymes (e.g., APOBEC1, UGI) that efficiently edits in mammalian cells, while minimizing frequency of non-target indels.
  • base editors do not comprise a functional fragment of the base editing enzyme.
  • base editors do not comprise a function fragment of a UGI, where such a fragment may be capable of excising a uracil residue from DNA by cleaving an N-glycosidic bond.
  • the effector partner comprises a non-protein uracil-DNA glycosylase inhibitor (npUGI).
  • npUGI is selected from a group of small molecule inhibitors of uracil-DNA glycosylase (UDG), or a nucleic acid inhibitor of UDG.
  • the npUGI is a small molecule derived from uracil. Examples of small molecule non-protein uracil-DNA glycosylase inhibitors, fusion proteins, and Cas- CRISPR systems comprising base editing activity are described in WO2021087246, which is incorporated by reference in its entirety.
  • the base editor is a cytosine base editor, wherein the based editing enzyme is a cytosine base editing enzyme.
  • the cytosine base editing enzyme is a cytidine deaminase.
  • the base editor comprising the cytidine deaminase is generated by ancestral sequence reconstruction as described in WO2019226953, which is hereby incorporated by reference in its entirety.
  • Non-limiting exemplary cytidine deaminases suitable for use with effector proteins described herein include: APOBEC1, APOBEC2, APOBEC3C, APOBEC3D, APOBEC3F, APOBEC3G, APOBEC3H, AP0BEC4, AP0BEC3A, BE1 (AP0BECl-XTEN-dCas9), BE2 (AP0BECl-XTEN-dCas9- UGI), BE3 (APOBECl-XTEN-dCas9(A840H)-UGI), BE3-Gam, saBE3, saBE4-Gam, BE4, BE4-Gam, saBE4, and saBE4-Gam as described in WO2021163587, WO2021087246, WO2021062227, and WO2020123887, which are incorporated herein by reference in their entirety.
  • a base editor is a cytosine to guanine base editor (CGBE), wherein the base editing enzyme is a cytosine to guanine base editing enzyme.
  • the cytosine to guanine base editing enzyme, and therefore the CGBE may convert a cytosine to a guanine.
  • a base editor is an adenine base editor (ABE), wherein the base editing enzyme is an adenine base editing enzyme.
  • the adenine base editing enzyme, and therefore the ABE may convert an adenine to a guanine.
  • the adenine base editing enzyme converts an A»T base pair to a G»C base pair.
  • the adenine base editing enzyme converts a target A»T base pair to G»C in vivo or in vitro.
  • the adenine base editing enzymes provided herein reverse spontaneous cytosine deamination, which has been linked to pathogenic point mutations.
  • the adenine base editing enzymes provided herein enable correction of pathogenic SNPs (-47% of disease-associated point mutations).
  • the adenine comprises exocyclic amine that has been deaminated (e.g., resulting in altering its base pairing preferences).
  • deamination of adenosine yields inosine.
  • inosine exhibits the base-pairing preference of guanine in the context of a polymerase active site, although inosine in the third position of a tRNA anticodon is capable of pairing with A, U, or C in mRNA during translation.
  • Non-limiting exemplary adenine base editing enzymes suitable for use with effector proteins described herein include: ABE8e, ABE8.20m, APOBEC3A, Anc APOBEC (a.k.a. AncBE4Max), and BtAPOBEC2.
  • Non-limiting exemplary ABEs suitable for use herein include: ABE7, ABE8.1m, ABE8.2m, ABE8.3m, ABE8.4m, ABE8.5m, ABE8.6m, ABE8.7m, ABE8.8m, ABE8.9m, ABE8.10m, ABE8.11m, ABE8.12m, ABE8.13m, ABE8.14m, ABE8.15m, ABE8.16m, ABE8.17m, ABE8.18m, ABE8.19m, ABE8.20m, ABE8.21m, ABE8.22m, ABE8.23m, ABE8.24m, ABE8.1d, ABE8.2d, ABE8.3d, ABE8.4d, ABE8.5d, ABE8.6d, ABE8.7d, ABE8.8d, ABE8.9d, ABE8.10d, ABE8.11d, ABE8.12d, ABE8.13
  • the adenine base editing enzyme is an adenine base editing enzyme described in Chu et al., (2021) The CRISPR Journal 4:2: 169-177, incorporated herein by reference.
  • the adenine deaminase is an adenine deaminase described by Koblan et al. (2016) Nature Biotechnology 36:848-846, incorporated herein by reference.
  • the adenine base editing enzyme is an adenine base editing enzyme described by Tran et al. (2020) Nature Communications 11 :4871.
  • the ABE described herein is capable of targeting polyA signals, splice site acceptors, and start codons. In some embodiments, the ABE cannot create stop codons for knock-down.
  • an adenine base editing enzyme is an adenosine deaminase.
  • Non-limiting exemplary adenosine base editors suitable for use herein include ABE9.
  • the ABE comprises an engineered adenosine deaminase enzyme capable of acting on ssDNA.
  • the engineered adenosine deaminase enzyme may be an adenosine deaminase variant that differs from a naturally occurring deaminase.
  • the adenosine deaminase variant may comprise one or more amino acid alteration, including a V82S alteration, a T166R alteration, a Y147T alteration, a Y147R alteration, a Q154S alteration, a Y123H alteration, a Q154R alteration, or a combination thereof.
  • the base editor comprises an adenine deaminase (e.g., TadA).
  • the adenosine deaminase is a TadA monomer (e.g., Tad*7.10, TadA*8 or TadA*9).
  • the adenosine deaminase is a TadA*8 variant (e.g., any one of TadA*8.1, TadA*8.2, TadA*8.3, TadA*8.4, TadA*8.5, TadA*8.6, TadA*8.7, TadA*8.8, TadA*8.9, TadA*8.10, TadA*8.11, TadA*8.12, TadA*8.13, TadA*8.14, TadA*8.15, TadA*8.16, TadA*8.17, TadA*8.18, TadA*8.19, TadA*8.20, TadA*8.21, TadA*8.22, TadA*8.23, or TadA*8.24 as described in WO2021163587 and W02021050571, which are each hereby incorporated by reference in its entirety).
  • the base editor comprises TadA.
  • a base editing enzyme is a deaminase dimer.
  • the ABE comprises the effector protein, the adenine base editing enzyme and the deaminase dimer.
  • the deaminase dimer comprises an adenosine deaminase.
  • the deaminase dimer comprises TadA and a suitable adenine base editing enzyme including an: ABE8e, ABE8.20m, APOBEC3A, Anc APOBEC (a.k.a. AncBE4Max), BtAPOBEC2, and variants thereof.
  • the adenine base editing enzyme is fused to amino-terminus or the carboxy -terminus of TadA.
  • a base editor is an RNA base editor, wherein the base editing enzyme is an RNA base editing enzyme.
  • the RNA base editing enzyme comprises an adenosine deaminase.
  • ADAR proteins bind to RNAs and alter their sequence by changing an adenosine into an inosine.
  • RNA base editors comprise an effector protein that is activated by or binds RNA.
  • base editing enzymes, and therefore base editors are used for treating a subject having or a subject suspected of having a disease related to a gene of interest.
  • base editing enzymes, and therefore base editors are useful for treating a disease or a disorder caused by a point mutation in a gene of interest.
  • compositions, systems, and methods described herein comprise a base editor and a guide nucleic acid, wherein the base editor comprises an effector protein and a base editing enzyme, and wherein the guide nucleic acid directs the base editor to a sequence in a target gene.
  • an effector partner provides enzymatic activity that modifies a protein associated with a target nucleic acid.
  • the protein may be a histone, an RNA binding protein, or a DNA binding protein.
  • protein modification activities include: methyltransferase activity, such as that provided by a histone methyltransferase (HMT) (e.g., suppressor of variegation 3-9 homolog 1 (SUV39H1, also known as KMT1A), Vietnamese histone lysine methyltransferase 2 (G9A, also known as KMT1C and EHMT2), SUV39H2, ESET/SETDB1, SET1A, SET1B, MLL1 to 5, ASH1, SYMD2, NSD1, DOT1L, Pr-SET7/8, SUV4-20H1, EZH2, RIZ1); demethylase activity such as that provided by a histone demethylase (e.g., Lysine Demethylase 1
  • HMT histone methyltrans
  • effector partners include, but are not limited to, a protein that directly and/or indirectly provides for increased or decreased transcription and/or translation of a target nucleic acid (e.g., a transcription activator or a fragment thereof, a protein or fragment thereof that recruits a transcription activator, a small molecule/drug-responsive transcription and/or translation regulator, a translation-regulating protein, etc.).
  • effector partners that increase or decrease transcription include a transcription activator domain or a transcription repressor domain, respectively.
  • effector partners activate or increase expression of a target nucleic acid.
  • effector partners increase expression of the target nucleic acid relative to its expression in the absence of the effector partners.
  • Relative expression including transcription and RNA levels, may be assessed, quantified, and compared, e.g., by RT-qPCR.
  • effector partners comprise a transcriptional activator.
  • the transcriptional activators may promote transcription by: recruitment of other transcription factor proteins; modification of target DNA such as demethylation; recruitment of a DNA modifier; modulation of histones associated with target DNA; recruitment of a histone modifier such as those that modify acetylation and/or methylation of histones; or a combination thereof.
  • Non-limiting examples of effector partners that promote or increase transcription include: transcriptional activators such as VP 16, VP64, VP48, VP 160, p65 subdomain (e.g., from NFkB), and activation domain of EDLL and/or TAL activation domain (e.g., for activity in plants); histone lysine methyltransferases such as SET1A, SET1B, MLL1 to 5, ASH1, SYMD2, NSD1; histone lysine demethylases such as JHDM2a/b, UTX, JMJD3; histone acetyltransferases such as GCN5, PCAF, CBP, p300, TAF1, TIP60/PLIP, MOZ/MYST3, MORF/MYST4, SRC1, ACTR, Pl 60, CLOCK; and DNA demethylases such as Ten-Eleven Translocation (TET) dioxygenase 1 (TET1CD), TET1, DME, DML
  • suitable effector partners include: proteins and protein domains responsible for stimulating translation (e.g., Staufen); proteins and protein domains responsible for (e.g., capable of) modulating translation (e.g., translation factors such as initiation factors, elongation factors, release factors, etc., e.g., eIF4G); proteins and protein domains responsible for stimulation of RNA splicing (e.g., Serine/ Arginine-rich (SR) domains); and proteins and protein domains responsible for stimulating transcription (e.g., CDK7 and HIV Tat).
  • proteins and protein domains responsible for stimulating translation e.g., Staufen
  • proteins and protein domains responsible for modulating translation e.g., translation factors such as initiation factors, elongation factors, release factors, etc., e.g., eIF4G
  • proteins and protein domains responsible for stimulation of RNA splicing e.g., Serine/ Arginine-rich (SR) domains
  • effector partners inhibit or reduce expression of a target nucleic acid.
  • effector partners reduce expression of the target nucleic acid relative to its expression in the absence of the effector partners.
  • Relative expression including transcription and RNA levels, may be assessed, quantified, and compared, e.g., by RT-qPCR.
  • effector partners may comprise a transcriptional repressor.
  • the transcriptional repressors may inhibit transcription by: recruitment of other transcription factor proteins; modification of target DNA such as methylation; recruitment of a DNA modifier; modulation of histones associated with target DNA; recruitment of a histone modifier such as those that modify acetylation and/or methylation of histones; or a combination thereof.
  • Non-limiting examples of effector partners that decrease or inhibit transcription include: transcriptional repressors such as the Kriippel associated box (KRAB or SKD); K0X1 repression domain; the Mad mSIN3 interaction domain (SID); the ERF repressor domain (ERD), the SRDX repression domain (e.g., for repression in plants); histone lysine methyltransferases such as Pr-SET7/8, SUV4-20H1, RIZ1, and the like; histone lysine demethylases such as JMJD2A/JHDM3A, JMJD2B, JMJD2C/GASC1, JMJD2D, JARID1A/RBP2, JARID1B/PLU-1, JARID1C/SMCX, JARID1D/SMCY; histone lysine deacetylases such as HDAC1, HDAC2, HDAC3, HDAC8, HDAC4, HDAC5, HDAC7, HDAC9, SI
  • suitable effector partners include: proteins and protein domains responsible for repressing translation (e.g., Ago2 and Ago4); proteins and protein domains responsible for repression of RNA splicing (e.g., PTB, Sam68, and hnRNP Al); proteins and protein domains responsible for reducing the efficiency of transcription e.g., FUS (TLS)).
  • proteins and protein domains responsible for repressing translation e.g., Ago2 and Ago4
  • proteins and protein domains responsible for repression of RNA splicing e.g., PTB, Sam68, and hnRNP Al
  • proteins and protein domains responsible for reducing the efficiency of transcription e.g., FUS (TLS)
  • fusion proteins activate or increase expression of a target nucleic acid.
  • fusion proteins inhibit or reduce expression of a target nucleic acid.
  • fusion proteins are targeted by a guide nucleic acid (e.g, guide RNA) to a specific location in a target nucleic acid and exert locus-specific regulation such as blocking RNA polymerase binding to a promoter (which selectively inhibits transcription activator function), and/or changes a local chromatin status (e.g, when a fusion sequence is used that edits the target nucleic acid or modifies a protein associated with the target nucleic acid).
  • the modifications are transient (e.g., transcription repression or activation).
  • the modifications are inheritable.
  • epigenetic modifications made to a target nucleic acid, or to proteins associated with the target nucleic acid, e.g., nucleosomal histones, in a cell can be observed in a successive generation.
  • effector partner comprises an RNA splicing factor.
  • the RNA splicing factor may be used (in whole or as fragments thereof) for modular organization, with separate sequence-specific RNA binding modules and splicing effector domains.
  • the RNA splicing factors comprise members of the Serine/ Arginine-rich (SR) protein family containing N-terminal RNA recognition motifs (RRMs) that bind to exonic splicing enhancers (ESEs) in pre-mRNAs and C-terminal RS domains that promote exon inclusion.
  • RRMs N-terminal RNA recognition motifs
  • ESEs exonic splicing enhancers
  • a hnRNP protein hnRNP Al binds to exonic splicing silencers (ESSs) through its RRM domains and inhibits exon inclusion through a C-terminal Glycine- rich domain.
  • the RNA splicing factors may regulate alternative use of splice site (ss) by binding to regulatory sequences between two alternative sites.
  • ASF/SF2 may recognize ESEs and promote the use of intron proximal sites, whereas hnRNP Al may bind to ESSs and shift splicing towards the use of intron distal sites.
  • One application for such factors is to generate ESFs that modulate alternative splicing of endogenous genes, particularly disease associated genes.
  • Bcl-x pre-mRNA produces two splicing isoforms with two alternative 5' splice sites to encode proteins of opposite functions.
  • Long splicing isoform Bcl-xL is a potent apoptosis inhibitor expressed in long-lived postmitotic cells and is up-regulated in many cancer cells, protecting cells against apoptotic signals.
  • Short isoform Bcl-xS is a pro-apoptotic isoform and expressed at high levels in cells with a high turnover rate (e.g., developing lymphocytes).
  • a ratio of the two Bcl-x splicing isoforms is regulated by multiple coo-elements that are located in either core exon region or exon extension region (z.e., between the two alternative 5' splice sites). For more examples, see W02010075303, which is hereby incorporated by reference in its entirety.
  • effector partners comprise a recombinase.
  • a recombinase system comprising effector proteins described herein and the recombinase.
  • the effector proteins have reduced nuclease activity or no nuclease activity.
  • the recombinase is a site-specific recombinase.
  • the recombinase system comprises a catalytically inactive effector protein, wherein the recombinase can be a site-specific recombinase.
  • a catalytically inactive effector protein wherein the recombinase can be a site-specific recombinase.
  • Such systems can be used for site-directed transgene insertion.
  • Non-limiting examples of site-specific recombinases include a tyrosine recombinase (e.g., Cre, Flp or lambda integrase), a serine recombinase (e.g., gamma-delta resolvase, Tn3 resolvase, Sin resolvase, Gin invertase, Hin invertase, Tn5044 resolvase, IS607 transposase and integrase), or mutants or variants thereof.
  • the recombinase is a serine recombinase.
  • Non-limiting examples of serine recombinases include gamma-delta resolvase, Tn3 resolvase, Sin resolvase, Gin invertase, Hin invertase, Tn5044 resolvase, IS607 transposase, and IS607 integrase.
  • the site-specific recombinase is an integrase.
  • integrases include:Bxbl, wBeta, BL3, phiR4, Al 18, TGI, MR11, phi370, SPBc, TP901-1, phiRV, FC1, K38, phiBTl, and phiC31.
  • the fusion protein comprises a linker that links the recombinase to a Cas-CRISPR domain of the effector protein.
  • the linker is Thr-Ser.
  • a linker comprises a bond or molecule that links a first polypeptide to a second polypeptide. Accordingly, in some embodiments, effector proteins, effector partners, or combinations thereof are connected by one or more linkers.
  • the linker may comprise or consist of a covalent bond.
  • the linker may comprise or consist of a chemical group.
  • the linker comprises an amino acid.
  • a peptide linker comprises at least two amino acids linked by an amide bond.
  • the linker connects a terminus of the effector protein to a terminus of the effector partner.
  • carboxy terminus of the effector protein is linked to the amino terminus of the effector partner.
  • carboxy terminus of the effector partner is linked to the amino terminus of the effector protein.
  • the effector protein and the effector partner are directly linked by a covalent bond.
  • linkers comprise one or more amino acids.
  • linker is a protein.
  • a terminus of the effector protein is linked to a terminus of the effector partner through an amide bond.
  • a terminus of the effector protein is linked to a terminus of the effector partner through a peptide bond.
  • linkers comprise an amino acid.
  • linkers comprise a peptide.
  • an effector protein is coupled to a effector partner by a linker protein.
  • the linker may have any of a variety of amino acid sequences.
  • the linker may comprise a region of rigidity (e.g., beta sheet, alpha helix), a region of flexibility, or any combination thereof.
  • the linker comprises small amino acids, such as glycine and alanine, that impart high degrees of flexibility.
  • design of a peptide conjugated to any desired element may include linkers that are all or partially flexible, such that the linker may include a flexible linker as well as one or more portions that confer less flexible structure.
  • Suitable linkers include proteins of 4 linked amino acids to 40 linked amino acids in length, or between 4 linked amino acids and 25 linked amino acids in length.
  • linked amino acids described herein comprise at least two amino acids linked by an amide bond.
  • Linkers may be produced by using synthetic, linker-encoding oligonucleotides to couple proteins, or may be encoded by a nucleic acid sequence encoding a fusion protein (e.g., an effector protein coupled to an effector partner).
  • the linker is from 1 to 100 amino acids in length. In some embodiments, the linker is more 100 amino acids in length. In some embodiments, the linker is from 10 to 27 amino acids in length.
  • linker proteins include glycine polymers (G)n, glycine-serine polymers (including, for example, (GS)n, GSGGSn, GGSGGSn, and GGGSn, where n is an integer of at least one), glycine-alanine polymers, and alanine-serine polymers.
  • linkers may comprise amino acid sequences including, but not limited to, GGSG, GGSGG, GSGSG, GSGGG, GGGSG, and GSSSG.
  • the linker comprises one or more repeats a tri-peptide GGS.
  • the linker is an XTEN linker.
  • the XTEN linker is an XTEN80 linker. In some embodiments, the XTEN linker is an XTEN20 linker. In some embodiments, the XTEN20 linker has an amino acid sequence of GSGGSPAGSPTSTEEGTSESATPGSG (SEQ ID NO: 916).
  • linkers do not comprise an amino acid. In some embodiments, linkers do not comprise a peptide. In some embodiments, linkers comprise a nucleotide, a polynucleotide, a polymer, or a lipid.
  • linker may be a polyethylene glycol (PEG), polypropylene glycol (PPG), co-poly(ethylene/propylene) glycol, polyoxyethylene (POE), polyurethane, polyphosphazene, polysaccharides, dextran, polyvinyl alcohol, polyvinylpyrrolidones, polyvinyl ethyl ether, polyacrylamide, polyacrylate, polycyanoacrylates, lipid polymers, chitins, hyaluronic acid, heparin, or an alkyl linker.
  • PEG polyethylene glycol
  • PPG polypropylene glycol
  • POE polyoxyethylene
  • polyurethane polyphosphazene
  • polysaccharides dextran
  • polyvinyl alcohol polyvinylpyrrolidones
  • polyvinyl ethyl ether polyacrylamide
  • polyacrylate polycyanoacrylates
  • lipid polymers chitins, hyaluronic acid, he
  • a linker is recognized and cleaved by a protein described herein.
  • a linker comprises a recognition sequence that may be recognized and cleaved by the protein.
  • a guide nucleic acid comprises an aptamer, which may serve a similar function as a linker, bringing an effector protein and an effector partner protein into proximity.
  • the aptamer can functionally connect two proteins (e.g., effector protein, effector partner) by interacting non-covalently with both, thereby bringing both proteins into proximity of the guide nucleic acid.
  • the first protein and/or the second protein comprise or is covalently linked to an aptamer binding moiety.
  • the aptamer is a short single stranded DNA (ssDNA) or RNA (ssRNA) molecule capable of being bound be the aptamer binding moiety.
  • the aptamer is a molecule that is capable of mimicking antibody binding activity and may be classified as a chemical antibody.
  • the aptamer described herein refers to artificial oligonucleotides that bind one or more specific molecules.
  • aptamers exhibit a range of affinities (KD in the pM to pM range) with little or no off-target binding.
  • compositions, systems, and methods comprise a fusion protein or uses thereof.
  • a fusion protein generally comprises an effector protein and an effector partner.
  • the effector partner comprises a polypeptide or peptide that is fused or linked to the effector protein.
  • the effector partner is fused to the N- terminus of the effector protein.
  • the effector partner is fused to the C- terminus of the effector protein.
  • the effector partner is a heterologous protein capable of imparting some function or activity that is not provided by an effector protein.
  • the effector partner is capable of cleaving or modifying the target nucleic acid, a non-target nucleic acid, or both.
  • the fusion protein disclosed herein may provide cleavage activity, such as cis cleavage activity, trans cleavage activity, nickase activity, nuclease activity, other activity, or a combination thereof. Fusion proteins disclosed herein may cleave nucleic acids, including single stranded RNA (ssRNA), double stranded DNA (dsDNA), and single-stranded DNA (ssDNA). In some embodiments, fusion proteins cleave the target nucleic acid at the target sequence or adjacent to the target sequence. [In some embodiments, fusion proteins cleave the non-target nucleic acid.
  • the fusion protein complexes with a guide nucleic acid and the complex interacts with the target nucleic acid, a non-target nucleic acid, or both.
  • the interaction comprises one or more of: recognition of a target adjacent motif (TAM) sequence within the target nucleic acid by the effector protein, hybridization of the guide nucleic acid to the target nucleic acid, modification of the target nucleic acid and/or the non-target nucleic acid by the fusion protein, or combinations thereof.
  • recognition of a TAM sequence within a target nucleic acid may direct the modification activity of a fusion protein.
  • Modification activity of a fusion protein described herein may be cleavage activity, binding activity, insertion activity, substitution activity, and the like. Modification activity of an effector protein may result in: cleavage of at least one strand of a target nucleic acid, deletion of one or more nucleotides of a target nucleic acid, insertion of one or more nucleotides into a target nucleic acid, substitution of one or more nucleotides of a target nucleic acid with an alternative nucleotide, more than one of the foregoing, or any combination thereof.
  • an ability of a fusion protein to edit a target nucleic acid may depend upon the effector protein being complexed with a guide nucleic acid, the guide nucleic acid being hybridized to a target sequence of the target nucleic acid, the distance between the target sequence and a TAM sequence, or combinations thereof.
  • compositions, systems, and methods of the present disclosure may comprise a guide nucleic acid or a use thereof.
  • compositions, systems and methods comprising guide nucleic acids or uses thereof, as described herein and throughout include DNA molecules, such as expression vectors, that encode a guide nucleic acid.
  • compositions, systems, and methods of the present disclosure comprise a guide nucleic acid or a nucleotide sequence encoding the guide nucleic acid.
  • Guide nucleic acids are also referred to herein as “guide RNA.”
  • the guide RNA comprises a protein binding sequence and a targeting sequence (also may be referred to as a spacer sequence).
  • effector proteins bind the protein binding sequence and the targeting sequence hybridizes to a target nucleic acid, thereby bringing an effector protein within the proximity of the target nucleic acid.
  • effector proteins and guide RNAs complex to form a ribonucleoprotein complex (RNP) that recognizes a motif (e.g., a TAM) proximal to a target sequence of the target nucleic acid.
  • RNP ribonucleoprotein complex
  • TAM a motif
  • a guide nucleic acid may comprise one or more deoxyribonucleotides, ribonucleotides, biochemically or chemically modified nucleotides (e.g., one or more engineered modifications as described herein), or any combinations thereof.
  • nucleotide sequences described herein may be described as a nucleotide sequence of either DNA or RNA, however, no matter the form the sequence is described, it is readily understood that such nucleotide sequences can be revised to be RNA or DNA, as needed, for describing a sequence within a guide nucleic acid itself or the sequence that encodes a guide nucleic acid, such as a nucleotide sequence described herein for a vector. Due to WIPO Standard ST.26, the Us are being represented as Ts in RNA in the Sequence Listing provided herein.
  • nucleotide sequences described herein also discloses the complementary nucleotide sequence, the reverse nucleotide sequence, and the reverse complement nucleotide sequence, any one of which can be a nucleotide sequence for use in a guide nucleic acid as described herein.
  • a guide nucleic acid sequence(s) comprises one or more nucleotide alterations at one or more positions in any one of the sequences described herein.
  • Alternative nucleotides can be any one or more of A, C, G, T or U, or a deletion, or an insertion.
  • a guide nucleic acid may comprise a naturally occurring sequence.
  • a guide nucleic acid may comprise a non-naturally occurring sequence, wherein the sequence of the guide nucleic acid, or any portion thereof, may be different from the sequence of a naturally occurring guide nucleic acid.
  • a guide nucleic acid of the present disclosure comprises one or more of the following: a) a single nucleic acid molecule; b) a DNA base; c) an RNA base; d) a modified base; e) a modified sugar; f) a modified backbone; and the like. Modifications are described herein and throughout the present disclosure (e.g., in the section entitled “Engineered Modifications”).
  • a guide nucleic acid may be chemically synthesized or recombinantly produced by any suitable methods. Guide nucleic acids and portions thereof may be found in or identified from a sequence present in the genome of a host organism or cell.
  • compositions, systems and methods described herein comprise a single nucleic acid system comprising a guide nucleic acid or a nucleotide sequence encoding the guide nucleic acid, and one or more effector proteins or a nucleotide sequence encoding the one or more effector proteins.
  • the guide nucleic acid of a single nucleic acid system may be referred to as a single guide RNA (sgRNA).
  • the effector protein comprises an IscB protein or engineered variant thereof, and the sgRNA is referred to as an omega RNA.
  • an omega RNA may comprise a hairpin structure and its coding sequence in the genome is located upstream or downstream of its respective IscB open reading frame (ORF).
  • an omega RNA coding sequence may be located within Ikb upstream or downstream of the IscB ORF.
  • compositions, systems, and methods of the present disclosure comprise two or more guide nucleic acids (e.g., 2, 3, 4, 5, 6, 7, 9, 10 or more guide nucleic acids), and/or uses thereof.
  • Multiple guide nucleic acids may target an effector protein to different locations in the target nucleic acid by hybridizing to different target sequences.
  • a first guide nucleic acid may hybridize within a location of the target nucleic acid that is different from where a second guide nucleic acid may hybridize the target nucleic acid.
  • the first loci and the second loci of the target nucleic acid may be located at least 1, at least 10, at least 20, at least 30, at least 40, at least 50, at least 60, at least 70, at least 80, at least 90 or at least 100 nucleotides apart. In some embodiments, the first loci and the second loci of the target nucleic acid may be located between 100 and 200, 200 and 300, 300 and 400, 400 and 500, 500 and 600, 600 and 700, 700 and 800, 800 and 900 or 900 and 1000 nucleotides apart. In some embodiments, the first loci and/or the second loci of the target nucleic acid are located in an intron of a gene.
  • the first loci and/or the second loci of the target nucleic acid are located in an exon of a gene. In some embodiments, the first loci and/or the second loci of the target nucleic acid span an exon-intron junction of a gene. In some embodiments, the first portion and/or the second portion of the target nucleic acid are located on either side of an exon and cutting at both sites results in deletion of the exon.
  • composition, systems, and methods comprise a donor nucleic acid that may be inserted in replacement of a deleted or cleaved sequence of the target nucleic acid. In some embodiments, compositions, systems, and methods comprising multiple guide nucleic acids or uses thereof comprise multiple effector proteins, wherein the effector proteins may be identical, non-identical, or combinations thereof.
  • a guide nucleic acid comprises at least 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 contiguous nucleotides that are complementary to a eukaryotic sequence.
  • a eukaryotic sequence is a nucleotide sequence that is present in a host eukaryotic cell.
  • Such a nucleotide sequence is distinguished from nucleotide sequences present in other host cells, such as prokaryotic cells, or viruses.
  • Said sequences present in a eukaryotic cell can be located in a gene, an exon, an intron, a non-coding (e.g., promoter or enhancer) region, a selectable marker, tag, signal, and the like.
  • a target sequence is a eukaryotic sequence.
  • the guide nucleic acid comprises 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 linked nucleosides.
  • a guide nucleic acid comprises at least linked nucleosides.
  • a guide nucleic acid comprises at least 25 linked nucleosides.
  • a guide nucleic acid may comprise 10 to 50 linked nucleosides.
  • the guide nucleic acid comprises or consists essentially of about 12 to about 80 linked nucleosides, about 12 to about 50, about 12 to about 45, about 12 to about 40, about 12 to about 35, about 12 to about 30, about 12 to about 25, from about 12 to about 20, about 12 to about 19 , about 19 to about 20, about 19 to about 25, about 19 to about 30, about 19 to about 35, about 19 to about 40, about 19 to about 45, about 19 to about 50, about 19 to about 60, about 20 to about 25, about 20 to about 30, about 20 to about 35, about 20 to about 40, about 20 to about 45, about 20 to about 50, or about 20 to about 60 linked nucleosides.
  • the guide nucleic acid has about 10 to about 60, about 20 to about 50, or about 30 to about 40 linked nucleosides.
  • a length of a guide nucleic acid is about 30 to about 120 linked nucleotides. In some embodiments, the length of a guide nucleic acid is about 40 to about 100, about 40 to about 90, about 40 to about 80, about 40 to about 70, about 40 to about 60, about 40 to about 50, about 50 to about 90, about 50 to about 80, about 50 to about 70, or about 50 to about 60 linked nucleotides. In some embodiments, the length of a guide nucleic acid is about 40, about 45, about 50, about 55, about 60, about 65, about 70 or about 75 linked nucleotides.
  • the length of a guide nucleic acid is greater than about 15, about 20, about 25, about 30, about 35, about 40, about 45, about 50, about 55, about 60, about 65, about 70 or about 75 linked nucleotides. In some embodiments, the length of a guide nucleic acid is not greater than about 40, about 45, about 50, about 55, about 60, about 65, about 70, about 75, about 80, about 85, about 90, about 95, about 100, about 105, about 110, about 115, about 120, or about 125 linked nucleotides.
  • guide nucleic acids comprise additional elements that contribute additional functionality (e.g., stability, heat resistance, etc.) to the guide nucleic acid.
  • additional elements may be one or more nucleotide alterations, nucleotide sequences, intermolecular secondary structures, or intramolecular secondary structures (e.g., one or more hair pin regions, one or more bulges, etc.).
  • guide nucleic acids comprise one or more linkers connecting different nucleotide sequences as described herein.
  • a linker may comprise 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more nucleotides.
  • a linker may be any suitable linker, examples of which are described herein.
  • a guide nucleic acid may comprise one or more modified nucleic acids.
  • Modifications can include changing of nucleic acids described herein (e.g., engineered guide nucleic acids) to provide the nucleic acid with a new or enhanced feature, such as improved stability.
  • Such modifications of a nucleic acid include a nucleobase base modification, a backbone modification, a sugar modification, a phosphorothioate internucleotide linkage, or combinations thereof.
  • the modifications can be of one or more nucleotides, nucleosides, or nucleobases in a nucleic acid.
  • uridines can be exchanged for pseudouridines (e.g., IN-Methyl-Pseudouridine). In some embodiments, all uridines can be exchanged for IN-Methyl-Pseudouridine. In this application, U can represent uracil or IN-Methyl-Pseudouridine.
  • pseudouridines e.g., IN-Methyl-Pseudouridine
  • all uridines can be exchanged for IN-Methyl-Pseudouridine.
  • U can represent uracil or IN-Methyl-Pseudouridine.
  • the guide nucleic acid comprises a protein binding sequence, wherein an effector protein described herein binds the protein binding sequence or at least a portion thereof.
  • a protein binding sequence comprises or consists of a handle sequence (also referred to as a scaffold sequence), wherein the handle sequence comprises at least one secondary structure that the effector protein can bind to.
  • the secondary structure may comprise a hairpin structure, also referred to as a stemloop structure.
  • Engineered proteins in TABLE 1 may bind a protein binding sequence represented by 5’-
  • the protein binding sequence is at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, or at least 98%, at least 99%, or 100% identical to an equal length portion of the sequence of SEQ ID NO: 858.
  • the protein binding sequence comprises at least 50, at least 60, at least 70, at least 80, at least 90, at least 100, at least 110, at least 120, at least 130, at least 140, at least 150, at least 160, at least 1700, at least 180, or at least 190 contiguous nucleotides of the sequence of SEQ ID NO: 858.
  • a handle sequence provided herein comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to a sequence selected from TABLE 7.
  • Effector proteins provided in TABLE 1, TABLE 6 and TABLE 8 may bind a handle sequence provided in TABLE 7.
  • an effector protein selected from any of effector protein 3107961, 3089183, 3092005, 3099541, 3782262, 3745646, 3756102, 370125, and 3778464 in TABLE 6 may bind a handle sequence provided in TABLE 7.
  • the effector protein comprises an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%. , at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 856, and the handle sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 859 or 860.
  • the effector protein comprises an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 624
  • the handle sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 884.
  • the effector protein comprises an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 713
  • the handle sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 885.
  • the effector protein comprises an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 833
  • the handle sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 886.
  • the effector protein comprises an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 691
  • the handle sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 884.
  • the effector protein comprises an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 774
  • the handle sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 888.
  • the effector protein comprises an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 179 and 889-910
  • the protein binding sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 858.
  • a guide nucleic acid described herein comprises a spacer sequence and a handle sequence, wherein the spacer sequence precedes the handle sequence in a 5’ to 3’ direction. See e.g., FIG. 3.
  • Guide nucleic acids described herein may comprise one or more spacer sequences.
  • spacer sequence may be used interchangeably with “targeting sequence.” While the term, “spacer sequence” may technically belong to CRISPR systems, the term, “spacer sequence,” in the instant disclosure may be used to a guiding or targeting sequence of a guide nucleic acid that is useful with any RNA guided nuclease, including but not limited to, Cas proteins, TnpB proteins, and IscB proteins.
  • a spacer sequence is capable of hybridizing to a target sequence of a target nucleic acid. Exemplary hybridization conditions are described herein.
  • the spacer sequence may function to direct an RNP complex comprising the guide nucleic acid to the target nucleic acid for detection and/or modification.
  • the spacer sequence may function to direct a RNP to the target nucleic acid for detection and/or modification.
  • a spacer sequence may be complementary to a target sequence that is adjacent to a TAM that is recognizable by an effector protein described herein.
  • the guide nucleic acid comprises a spacer sequence that is at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100% complementary to the target sequence.
  • the guide nucleic acid comprises at least 10 contiguous nucleotides that are complementary to the target sequence in the target nucleic acid.
  • guide nucleic acid comprises a spacer sequence that is at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100% complementary to the target sequence.
  • the target sequence is a eukaryotic sequence.
  • the spacer sequence may comprise complementarity with (e.g., hybridize to) a target sequence of a target nucleic acid.
  • a spacer sequence comprises at least 5 to about 50, at least 5 to about 25, at least about 10 to at least about 25, or at least about 15 to about 25 linked nucleotides.
  • the spacer sequence is 15-28 linked nucleosides in length.
  • the spacer sequence is 15-26, 15-24, 15-22, 15-20, 15-18, 16-28, 16- 26, 16-24, 16-22, 16-20, 16-18, 17-26, 17-24, 17-22, 17-20, 17-18, 18-26, 18-24, or 18-22 linked nucleosides in length.
  • the spacer sequence is 18-24 linked nucleosides in length. In some cases, the spacer sequence is at least 15 linked nucleosides in length. In some cases, the spacer sequence is at least 16, 18, 20, or 22 linked nucleosides in length. In some cases, the spacer sequence comprises at least 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides. In some cases, the spacer sequence is at least 17 linked nucleosides in length. In some cases, the spacer sequence is at least 18 linked nucleosides in length. In some cases, the spacer sequence is at least 20 linked nucleosides in length.
  • a spacer sequence comprises at least 5 to about 50 contiguous nucleotides that are complementary to a target sequence in a target nucleic acid. In some cases, the spacer sequence comprises at least 15 contiguous nucleobases that are complementary to the target nucleic acid.
  • the spacer sequence is located 5’ of the protein binding sequence. In some embodiments, the spacer sequence is located at the 5’ end of the guide nucleic acid. In some embodiments, the spacer and protein binding sequences are linked directly to one another. In some embodiments, a linker is present between the spacer and protein binding sequences. Linkers may be any suitable linker. In some embodiments, the spacer sequence and the protein binding sequence of the guide nucleic acid are present in separate molecules, which are joined to one another by base pairing interactions.
  • a spacer sequence may comprise a nucleotide sequence that may have partial complementarity with (e.g., hybridize to) an equal length of a target sequence of a target nucleic acid.
  • the guide nucleic acid may comprise at least one uracil between nucleic acid residues 5 to 20 of the spacer sequence that is not complementary to the corresponding nucleoside of the target sequence.
  • the guide nucleic acid may comprise at least one uracil between nucleic acid residues 5 to 9, 10 to 14, or 15 to 20 of the spacer sequence that is not complementary to the corresponding nucleoside of the target sequence.
  • the region of the target nucleic acid that is complementary to the spacer sequence comprises an epigenetic modification or a post-transcriptional modification.
  • the epigenetic modification comprises acetylation, methylation, or thiol modification.
  • a guide nucleic acid for use with compositions, systems, and methods described herein comprises one or more linkers, or a nucleic acid encoding one or more linkers.
  • the guide nucleic acid comprises a linker connecting a protein binding sequence and a spacer sequence.
  • a linker comprises one to ten, one to seven, one to five, one to three, two to ten, two to eight, two to six, two to four, three to ten, three to seven, three to five, four to ten, four to eight, four to six, five to ten, five to seven, six to ten, six to eight, seven to ten, or eight to ten linked nucleotides.
  • the linker comprises one, two, three, four, five, six, seven, eight, nine, or ten linked nucleotides. In some embodiments, a linker comprises a nucleotide sequence of 5’-GAAA-3’.
  • Guide nucleic acids may comprise chemical modifications.
  • guide nucleic acids are chemically synthesized (as opposed to transcribed). Modifications may provide the nucleic acid with a new or enhanced feature, such as improved stability.
  • modifications of a nucleic acid include a base modification, a backbone modification, a sugar modification, or combinations thereof, of one or more nucleotides, nucleosides, or nucleobases in a nucleic acid.
  • nucleic acids e.g., engineered guide nucleic acids
  • nucleic acids comprise one or more modifications comprising: 2’0-methyl modified nucleotides, 2’ Fluoro modified nucleotides; locked nucleic acid (LNA) modified nucleotides; peptide nucleic acid (PNA) modified nucleotides; nucleotides with phosphorothioate linkages; a 5’ cap (e.g., a 7-methylguanylate cap (m7G)), phosphorothioates, chiral phosphorothioates, phosphorodithioates, phosphotriesters, aminoalkylphosphotriesters, methyl and other alkyl phosphonates including 3'-alkylene phosphonates, 5'-alkylene phosphonates and chiral phosphonates, phosphinates, phosphoramidates including 3 '-amino phosphoramidate and aminoalkyl phosphoramidates
  • compositions and systems provided herein comprise a vector system, wherein the vector system comprises one or more vectors.
  • a vector can be used as a vehicle to introduce one or more molecules of interest into a host cell.
  • a molecule of interest may comprise a polypeptide (e.g., an effector protein), a guide nucleic acid, a donor nucleic acid, a nucleic acid encoding a polypeptide, a nucleic acid encoding an engineered guide or a component thereof.
  • the vector may be part of a vector system, wherein a vector system comprises a library of vectors each encoding one or more component of a composition or system described herein.
  • components described herein are encoded by the same vector.
  • components described herein e.g., an effector protein, a guide nucleic acid, and/or a target nucleic acid
  • components described herein are each encoded by different vectors of the system.
  • vector systems described herein can comprise one or more vectors comprising a polypeptide (e.g., an effector protein), an engineered guide nucleic acid, or a nucleic acid or nucleic acids encoding a polypeptide, engineered guide, a donor nucleic acid, or any combination thereof.
  • compositions and systems provided herein comprise a vector system comprising a polypeptide (e.g., an effector protein, effector partner, or fusion protein) described herein.
  • compositions and systems provided herein comprise a vector system comprising a guide nucleic acid described herein.
  • compositions and systems provided herein comprise a vector system comprising a donor nucleic acid described herein.
  • compositions and systems provided herein comprise a vector system encoding a polypeptide (e.g., an effector protein, effector partner, or fusion protein) described herein.
  • compositions and systems provided herein comprise a vector system encoding a guide nucleic acid described herein.
  • compositions and systems provided herein comprise a multi-vector system encoding an effector protein and a guide nucleic acid described herein, wherein the guide nucleic acid and the effector protein are encoded by the same or different vectors.
  • the guide nucleic acid and the effector protein are encoded by different vectors of the system.
  • a nucleic acid encoding a polypeptide comprises an expression vector.
  • a nucleic acid encoding a polypeptide is a messenger RNA.
  • an expression vector comprises or encodes an engineered guide nucleic acid.
  • a vector comprises one or more donor nucleic acids as described herein.
  • the one or more donor nucleic acids comprise at least two donor nucleic acids.
  • the at least two donor nucleic acids are the same.
  • the at least two donor nucleic acids are different from each other.
  • a vector can comprise or encode one or more regulatory elements. Regulatory elements can refer to transcriptional and translational control sequences, such as promoters, enhancers, polyadenylation signals, terminators, protein degradation signals, and the like, that provide for and/or regulate transcription of a non-coding sequence or a coding sequence and/or regulate translation of an encoded polypeptide.
  • a vector can comprise or encode for one or more additional elements, such as, replication origins, antibiotic resistance (or a nucleic acid encoding the same), a tag (or a nucleic acid encoding the same), selectable markers, and the like.
  • Vectors described herein can encode a promoter - a regulatory region on a nucleic acid, such as a DNA sequence, capable of initiating transcription of a downstream (3' direction) coding or non-coding sequence.
  • a promoter can be bound at its 3' terminus to a nucleic acid the expression or transcription of which is desired, and extends upstream (5' direction) to include bases or elements necessary to initiate transcription or induce expression, which could be measured at a detectable level.
  • a promoter can comprise a nucleotide sequence, referred to herein as a “promoter sequence”.
  • a promoter sequence can include a transcription initiation site, and one or more protein binding domains responsible for the binding of transcription machinery, such as RNA polymerase.
  • promoters When eukaryotic promoters are used, such promoters can contain “TATA” boxes and “CAT” boxes.
  • Various promoters, including inducible promoters, may be used to drive expression, z.e., transcriptional activation, of the nucleic acid of interest. Accordingly, in some embodiments, the nucleic acid of interest can be operably linked to a promoter.
  • Promotors can be any suitable type of promoter envisioned for the compositions, systems, and methods described herein. Examples include constitutively active promoters (e.g., CMV promoter), inducible promoters (e.g., heat shock promoter, tetracycline-regulated promoter, steroid-regulated promoter, metal-regulated promoter, estrogen receptor-regulated promoter, etc.), spatially restricted and/or temporally restricted promoters (e.g., a tissue specific promoter, a cell type specific promoter, etc.), etc.
  • constitutively active promoters e.g., CMV promoter
  • inducible promoters e.g., heat shock promoter, tetracycline-regulated promoter, steroid-regulated promoter, metal-regulated promoter, estrogen receptor-regulated promoter, etc.
  • spatially restricted and/or temporally restricted promoters e.g., a tissue specific promoter, a cell type specific promoter, etc.
  • Suitable promoters include, but are not limited to: SV40 early promoter, mouse mammary tumor virus long terminal repeat (LTR) promoter; adenovirus major late promoter (Ad MLP); a herpes simplex virus (HSV) promoter, a cytomegalovirus (CMV) promoter such as the CMV immediate early promoter region (CMVIE), a rous sarcoma virus (RSV) promoter, a human U6 small nuclear promoter (U6), an enhanced U6 promoter, and a human Hl promoter (Hl).
  • SV40 early promoter mouse mammary tumor virus long terminal repeat (LTR) promoter
  • Ad MLP adenovirus major late promoter
  • HSV herpes simplex virus
  • CMV cytomegalovirus
  • CMVIE CMV immediate early promoter region
  • RSV rous sarcoma virus
  • U6 small nuclear promoter U6 small nuclear promoter
  • Hl human Hl promoter
  • vectors used for providing a nucleic acid encoding an engineered guide nucleic acid and/or an effector protein to a cell may include nucleic acid sequences that encode for selectable markers in the target cells, so as to identify cells that have taken up the engineered guide nucleic acid and/or an effector protein.
  • plasmids and vectors described herein comprise at least one promoter.
  • the length of the promoter is less than about 500, less than about 400, or less than about 300 linked nucleotides. In some embodiments, the length of the promoter is at least 100 linked nucleotides.
  • the promoters are constitutive promoters. In other embodiments, the promoters are inducible promoters. In some embodiments, the promoter is an inducible promoter that only drives expression of its corresponding gene when a signal is present, e.g., a hormone, a small molecule, a peptide.
  • Non-limiting examples of inducible promoters are the T7 RNA polymerase promoter, the T3 RNA polymerase promoter, the Isopropyl-beta-D-thiogalactopyranoside (IPTG)-regulated promoter, a lactose induced promoter, a heat shock promoter, a tetracycline-regulated promoter (tetracycline-inducible or tetracycline-repressible), a steroid regulated promoter, a metal-regulated promoter, and an estrogen receptor-regulated promoter.
  • IPTG Isopropyl-beta-D-thiogalactopyranoside
  • the promoter is an activationinducible promoter, such as a CD69 promoter, as described further in Kulemzin et al., (2019), BMC Med Genomics, 12:44.
  • the promoters are prokaryotic promoters (e.g., drive expression of a gene in a prokaryotic cell).
  • the promoters are eukaryotic promoters, (e.g., drive expression of a gene in a eukaryotic cell).
  • Exemplary promoters include, but are not limited to, CMV, 7SK, EFla, RPBSA, hPGK, EFS, SV40, PGK1, Ubc, human beta actin, CAG, TRE, UAS, Ac5, polyhedron, CaMKIIa, GAL1- 10, TEF1, GDS, ADH1, CaMV35S, Ubi, Hl, U6, CaMV35S, MNDU3, MSCV and HSV TK promoter.
  • the promoter is CMV.
  • the promoter is EFla.
  • the promoter is ubiquitin.
  • vectors are bicistronic or polycistronic vector (e.g., having or involving two or more loci responsible for generating a protein) having an internal ribosome entry site (IRES) is for translation initiation in a cap-independent manner.
  • IRS internal ribosome entry site
  • a vector described herein is a delivery vector.
  • the delivery vector may be a eukaryotic vector, a prokaryotic vector (e.g., a bacterial vector) a viral vector, or any combination thereof.
  • the delivery vehicle may be a non-viral vector.
  • the delivery vehicle may be a plasmid.
  • the plasmid comprises DNA.
  • the plasmid comprises RNA.
  • the plasmid comprises circular double-stranded DNA.
  • the plasmid may be linear.
  • the plasmid comprises one or more genes of interest and one or more regulatory elements.
  • the plasmid comprises a bacterial backbone containing an origin of replication and an antibiotic resistance gene or other selectable marker for plasmid amplification in bacteria.
  • the plasmid may be a minicircle plasmid.
  • the plasmid contains one or more genes that provide a selective marker to induce a target cell to retain the plasmid.
  • the plasmid may be formulated for delivery through injection by a needle carrying syringe.
  • the plasmid may be formulated for delivery via electroporation.
  • the plasmids may be engineered through synthetic or other suitable means known in the art.
  • the genetic elements may be assembled by restriction digest of the desired genetic sequence from a donor plasmid or organism to produce ends of the DNA which may then be readily ligated to another genetic sequence.
  • vectors comprise an enhancer.
  • Enhancers are nucleotide sequences that have the effect of enhancing promoter activity.
  • enhancers augment transcription regardless of the orientation of their sequence.
  • enhancers activate transcription from a distance of several kilo basepairs.
  • enhancers are located optionally upstream or downstream of a gene region to be transcribed, and/or located within the gene, to activate the transcription.
  • Exemplary enhancers include, but are not limited to, WPRE; CMV enhancers; the R-U5' segment in LTR of HTLV- I (Mol. Cell. Biol., Vol. 8(1), p.
  • an effector protein or a nucleic acid encoding same
  • an engineered guide nucleic acid or a nucleic acid encoding same
  • Co-administration can be contact with a target nucleic acid, administered to a cell, such as a host cell, or administered as method of nucleic acid detection, editing, and/or treatment as described herein, in a single vehicle, such as a single expression vector.
  • a target nucleic acid administered to a cell, such as a host cell
  • a single vehicle such as a single expression vector.
  • an effector protein (or a nucleic acid encoding same) and/or an engineered guide nucleic acid (or a nucleic acid encoding same) are not co-administered with donor nucleic acid in a single vehicle.
  • an effector protein or a nucleic acid encoding same
  • an engineered guide nucleic acid or a nucleic acid encoding same
  • donor nucleic acid are administered in one or more or two or more vehicles, such as one or more, or two or more expression vectors.
  • the vector is a non-viral vector, and a physical method or a chemical method is employed for delivery into the somatic cell.
  • exemplary physical methods include electroporation, gene gun, sonoporation, magnetofection, or hydrodynamic delivery.
  • Exemplary chemical methods include delivery of the recombinant polynucleotide via liposomes such as, cationic lipids or neutral lipids; dendrimers; nanoparticles; or cellpenetrating peptides.
  • a vector is administered as part of a method of nucleic acid detection, editing, and/or treatment as described herein.
  • a vector is administered in a single vehicle, such as a single expression vector.
  • at least two of the three components, a nucleic acid encoding one or more effector proteins, one or more donor nucleic acids, and one or more guide nucleic acids or a nucleic acid encoding the one or more guide nucleic acid are provided in the single expression vector.
  • components, such as a guide nucleic acid and an effector protein are encoded by the same vector.
  • a polypeptide e.g., an effector protein, effector partner, or fusion protein
  • a nucleic acid encoding same and/or an engineered guide nucleic acid (or a nucleic acid that, when transcribed, produces same) are not co-administered with donor nucleic acid in a single vehicle.
  • an effector protein (or a nucleic acid encoding same), an engineered guide nucleic acid (or a nucleic acid that, when transcribed, produces same), and/or donor nucleic acid are administered in one or more or two or more vehicles, such as one or more, or two or more expression vectors.
  • a vector may be part of a vector system.
  • the vector system comprises a library of vectors each encoding one or more components of a composition or system described herein.
  • a vector system is administered as part of a method of nucleic acid detection, editing, and/or treatment as described herein, wherein at least two vectors are co-administered.
  • the at least two vectors comprise different components.
  • the at least two vectors comprise the same component having different sequences.
  • At least one of the three components, a nucleic acid encoding one or more effector proteins, one or more donor nucleic acids, and one or more guide nucleic acids or a nucleic acid encoding the one or more guide nucleic acids, or a variant thereof is provided in a different vector.
  • the nucleic acid encoding the effector protein, and a guide nucleic acid or a nucleic acid encoding the guide nucleic acid are provided in different vectors.
  • the donor nucleic acid is encoded by a different vector than the vector encoding the effector protein and the guide nucleic acid.
  • compositions and systems provided herein comprise a lipid particle.
  • a lipid particle is a lipid nanoparticle (LNP).
  • LNP lipid nanoparticle
  • a lipid or a lipid nanoparticle can encapsulate an expression vector.
  • a lipid or a lipid nanoparticle can encapsulate the effector protein, the guide nucleic acid, the nucleic acid encoding the effector protein and/or the DNA molecule encoding the guide nucleic acid.
  • LNPs are a non-viral delivery system for gene therapy. LNPs are effective for delivery of nucleic acids.
  • a method can comprise contacting a cell with an expression vector.
  • contacting can comprise electroporation, lipofection, or lipid nanoparticle (LNP) delivery of an expression vector.
  • a nucleic acid expression vector is a non-viral vector.
  • compositions and methods comprise a lipid, polymer, nanoparticle, or a combination thereof, or use thereof, to introduce an effector protein, guide nucleic acid, donor template or any combination thereof to a cell.
  • lipids and polymers are cationic polymers, cationic lipids, or bio-responsive polymers.
  • the bio-responsive polymer exploits chemi cal -physical properties of the endosomal environment (e.g., pH) to preferentially release the genetic material in the intracellular space.
  • compositions comprise an LNP, an mRNA encoding an effector protein, and optionally a guide RNA.
  • the mRNA and optionally, the guide RNA are encapsulated in an LNP.
  • the guide RNA may comprise chemical modifications.
  • a LNP comprises an outer shell and an inner core.
  • the outer shell comprises lipids.
  • the lipids comprise modified lipids.
  • the modified lipids comprise pegylated lipids.
  • the lipids comprise one or more of cationic lipids, anionic lipids, ionizable lipids, and non-ionic lipids.
  • the LNP comprises one or more of N1,N3,N5- tris(3-(didodecylamino)propyl)benzene-l,3,5-tricarboxamide (TT3), 2-dioleoyl-sn-glycero-3- phosphoethanolamine (DOPE), l-palmitoyl-2-oleoylsn-glycero-3-phosphoethanolamine (POPE), l,2-distearoyl-sn-glycero-3-phosphocholine (DSPC), cholesterol (Choi), 1,2- dimyristoyl-sn-glycerol, and methoxypolyethylene glycol (DMG-PEChooo), derivatives, analogs, or variants thereof.
  • DOPE 2-dioleoyl-sn-glycero-3- phosphoethanolamine
  • POPE l-palmitoyl-2-oleoylsn-glycero-3-phosphoethanolamine
  • DSPC l,2-d
  • the LNP has a negative net overall charge prior to complexation with one or more of a guide nucleic acid, a nucleic acid encoding the one or more guide nucleic acid, a nucleic acid encoding the polypeptide (e.g., effector protein, effector partner, fusion protein, or combinations thereof), and/or a donor nucleic acid.
  • the inner core is a hydrophobic core.
  • the one or more of a guide nucleic acid, the one or more nucleic acid encoding the one or more guide nucleic acid, one or more nucleic acid encoding one or more polypeptides, and/or the one or more donor nucleic acid forms a complex with one or more of the cationic lipids and the ionizable lipids.
  • the nucleic acid encoding the polypeptide or the nucleic acid encoding the guide nucleic acid is self-replicating.
  • a LNP comprises a lipid composition targeting to a specific organ.
  • the lipid composition comprises lipids having a specific alkyl chain length that controls accumulation of the LNP in the specific organ (e.g., liver or spleen).
  • the lipid composition comprises a biomimetic lipid that controls accumulation of the LNP in the specific organ (e.g., brain).
  • the lipid composition comprises lipid derivatives (e.g., cholesterol derivatives) that controls accumulation of the LNP in a specific cell (e.g., liver endothelial cells, Kupffer cells, hepatocytes).
  • An expression vector can be a viral vector.
  • a viral vector comprises a nucleic acid to be delivered into a host cell via a recombinantly produced virus or viral particle.
  • the nucleic acid may be single-stranded or double stranded, linear or circular, segmented or non-segmented.
  • the nucleic acid may comprise DNA, RNA, or a combination thereof.
  • the expression vector is an adeno-associated viral vector.
  • viral vectors that are associated with various types of viruses, including but not limited to retroviruses (e.g., lentiviruses and y -retroviruses), adenoviruses, arenaviruses, alphaviruses, adeno-associated viruses (AAVs), baculoviruses, vaccinia viruses, herpes simplex viruses and poxviruses.
  • retroviruses e.g., lentiviruses and y -retroviruses
  • AAVs adeno-associated viruses
  • baculoviruses baculoviruses
  • vaccinia viruses herpes simplex viruses and poxviruses.
  • the virus may be a lentivirus.
  • the virus may be an adenovirus.
  • the virus may be a non-replicating virus.
  • the virus may be an adeno-associated virus (AAV).
  • the viral vector may be a retroviral vector.
  • Retroviral vectors may include gamma-retroviral vectors such as vectors derived from the Moloney Murine Leukemia Virus (MoMLV, MMLV, MuLV, or MLV) or the Murine Stem cell Virus (MSCV) genome. Retroviral vectors may include lentiviral vectors such as those derived from the human immunodeficiency virus (HIV) genome.
  • the viral vector is a chimeric viral vector, comprising viral portions from two or more viruses.
  • the viral vector is a recombinant viral vector.
  • the viral vector is a chimeric viral vector.
  • the chimeric viral vector comprises viral portions from two or more viruses.
  • the viral vector corresponds to a virus of a specific serotype.
  • the viral vector is an AAV.
  • the AAV may be any AAV known in the art.
  • a viral particle that delivers a viral vector described herein is an AAV.
  • the viral vector corresponds to a virus of a specific serotype.
  • the serotype is selected from an AAV1 serotype, an AAV2 serotype, AAV3 serotype, an AAV4 serotype, AAV5 serotype, an AAV6 serotype, AAV7 serotype, an AAV8 serotype, an AAV9 serotype, an AAV 10 serotype, an AAV-rhlO serotype, an AAV11 serotype, and an AAV12 serotype.
  • the AAV vector is a recombinant vector, a hybrid AAV vector, a chimeric AAV vector, a self-complementary AAV (scAAV) vector, a single-stranded AAV or any combination thereof.
  • scAAV genomes are generally known in the art and contain both DNA strands which can anneal together to form double-stranded DNA.
  • the AAV vector may be a chimeric AAV vector.
  • the chimeric AAV vector comprises an exogenous amino acid or an amino acid substitution, or capsid proteins from two or more serotypes.
  • a chimeric AAV vector may be genetically engineered to increase transduction efficiency, selectivity, or a combination thereof.
  • the viral vector is a replication-defective viral vector, comprising an insertion of a therapeutic gene inserted in genes essential to the lytic cycle, preventing the virus from replicating and exerting cytotoxic effects.
  • a viral vector provided herein can be derived from or based on any such virus.
  • the viral vectors provided herein are an adeno-associated viral vector (AAV vector).
  • AAV vector has two inverted terminal repeats (ITRs).
  • the viral vector provided herein comprises two inverted terminal repeats of AAV.
  • the DNA sequence in between the ITRs of an AAV vector provided herein may be referred to herein as the sequence encoding the genome editing tools.
  • genome editing tools can include, but are not limited to, an effector protein, effector protein modifications (e.g., nuclear localization signal (NLS), polyA tail), guide nucleic acid(s), respective promoter(s), and a donor nucleic acid, or combinations thereof.
  • viral vectors provided herein comprise at least one promotor or a combination of promoters driving expression or transcription of one or more genome editing tools described herein.
  • the coding region of the AAV vector forms an intramolecular double-stranded DNA template thereby generating an AAV vector that is a self-complementary AAV (scAAV) vector.
  • the sequence encoding the genome editing tools of an scAAV vector has a length of about 2 kb to about 3 kb.
  • the scAAV vector can comprise nucleotide sequences encoding an effector protein, providing guide nucleic acids described herein, and a donor nucleic acid described herein.
  • the AAV vector provided herein is a self-inactivating AAV vector.
  • an AAV vector provided herein comprises a modification, such as an insertion, deletion, chemical alteration, or synthetic modification, relative to a wild-type AAV vector.
  • a fusion protein as described herein is inserted into a vector.
  • the vector comprises one or more promoters, enhancers, ribosome binding sites, RNA splice sites, polyadenylation sites, a replication origin, and/or transcriptional terminator sequences.
  • methods of producing delivery vectors herein comprise packaging a nucleic acid encoding a polypeptide (e.g., an effector protein, effector partner, fusion protein, or combination thereof) and a guide nucleic acid, or a combination thereof, into an AAV vector.
  • a polypeptide e.g., an effector protein, effector partner, fusion protein, or combination thereof
  • methods of producing the delivery vector comprises, (a) contacting a cell with at least one nucleic acid encoding: (i) a guide nucleic acid; (ii) a Replication (Rep) gene; and (iii) a Capsid (Cap) gene that encodes an AAV capsid protein; (b) expressing the AAV capsid protein in the cell; (c) assembling an AAV particle; and (d) packaging a Cas effector encoding nucleic acid into the AAV particle, thereby generating an AAV delivery vector.
  • promoters, stuffer sequences, and any combination thereof may be packaged in the AAV vector.
  • the AAV vector can package 1, 2, 3, 4, or 5 guide nucleic acids or copies thereof.
  • the AAV vector comprises inverted terminal repeats, e.g., a 5’ inverted terminal repeat and a 3’ inverted terminal repeat.
  • the AAV vector comprises a mutated inverted terminal repeat that lacks a terminal resolution site.
  • a hybrid AAV vector is produced by transcapsidation, e.g., packaging an inverted terminal repeat (ITR) from a first serotype into a capsid of a second serotype, wherein the first and second serotypes may be not the same.
  • ITR inverted terminal repeat
  • the Rep gene and ITR from a first AAV serotype e.g., AAN2'
  • a second AAV serotype e.g., AAV9
  • a hybrid AAV serotype comprising the AAV2 ITRs and AAV9 capsid protein may be indicated AAV2/9.
  • the hybrid AAV delivery vector comprises an AAV2/1, AAV2/2, AAV 2/4, AAV2/5, AAV2/8, or AAV2/9 vector.
  • the AAV particles described herein can be referred to as recombinant AAV (rAAV).
  • rAAV particles are generated by transfecting AAV producing cells with an AAV- containing plasmid carrying the sequence encoding the genome editing tools, a plasmid that carries viral encoding regions, i.e., Rep and Cap gene regions; and a plasmid that provides the helper genes such as E1A, E1B, E2A, E4ORF6 and VA.
  • the AAV producing cells are mammalian cells.
  • host cells for rAAV viral particle production are mammalian cells.
  • a mammalian cell for rAAV viral particle production is a COS cell, a HEK293T cell, a HeLa cell, a KB cell, a derivative thereof, or a combination thereof.
  • rAAV virus particles can be produced in the mammalian cell culture system by providing the rAAV plasmid to the mammalian cell.
  • producing rAAV virus particles in a mammalian cell can comprise transfecting vectors that express the rep protein, the capsid protein, and the gene-of-interest expression construct flanked by the ITR sequence on the 5’ and 3’ ends.
  • rAAV is produced in a non-mammalian cell.
  • rAAV is produced in an insect cell.
  • an insect cell for producing rAAV viral particles comprises a Sf9 cell.
  • production of rAAV virus particles in insect cells can comprise baculovirus.
  • production of rAAV virus particles in insect cells can comprise infecting the insect cells with three recombinant baculoviruses, one carrying the cap gene, one carrying the rep gene, and one carrying the gene-of-interest expression construct enclosed by an ITR on both the 5’ and 3’ end.
  • rAAV virus particles are produced by the One Bac system.
  • rAAV virus particles can be produced by the Two Bac system.
  • the rep gene and the cap gene of the AAV is integrated into one baculovirus virus genome, and the ITR sequence and the gene-of-interest expression construct is integrated into another baculovirus virus genome.
  • an insect cell line that expresses both the rep protein and the capsid protein is established and infected with a baculovirus virus integrated with the ITR sequence and the gene-of-interest expression construct. Details of such processes are provided in, for example, Smith et. al., (1983), Mol. Cell.
  • the viral particle that delivers the viral vector described herein is an AAV.
  • AAVs are characterized by their serotype.
  • Non-limiting examples of AAV serotypes are AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, scAAV, AAV-rhlO, chimeric or hybrid AAV, or any combination, derivative, or variant thereof.
  • the target nucleic acid is a double stranded nucleic acid.
  • the target nucleic acid is a single stranded nucleic acid.
  • the target nucleic acid is a double stranded nucleic acid that is prepared into single stranded nucleic acids before or upon contacting a reagent or sample.
  • the target nucleic acid comprises DNA.
  • the target nucleic acid comprises RNA.
  • the target nucleic acids include but are not limited to mRNA, rRNA, tRNA, non-coding RNA, long non-coding RNA, and microRNA (miRNA).
  • the target nucleic acid is complementary DNA (cDNA) synthesized from a single-stranded RNA template in a reaction catalyzed by a reverse transcriptase.
  • the target nucleic acid is single-stranded RNA (ssRNA) or mRNA.
  • the target nucleic acid is a double stranded nucleic acid comprising a target strand and a non-target strand, wherein the target strand comprises a target sequence.
  • a target strand comprises a target sequence
  • at least a portion of the engineered guide nucleic acid is complementary to the target sequence on the target strand.
  • the target nucleic acid is a double stranded nucleic acid comprising a target strand and a non-target strand
  • the target strand comprises a target sequence
  • at least a portion of the engineered guide nucleic acid is complementary to the target sequence on the target strand.
  • a target nucleic acid comprises a TAM as described herein that is located on the non-target strand.
  • Such a TAM described herein is adjacent (e.g., within 1, 2, 3, 4 or 5 nucleotides) to the 3’ end of the target sequence on the non-target strand of the double stranded DNA molecule.
  • such a TAM described herein is directly adjacent to the 3’ end of a target sequence on the non-target strand of the double stranded DNA molecule, n some embodiments, the target nucleic acid as described in the methods herein does not initially comprise a TAM sequence.
  • any target nucleic acid of interest may be generated using the methods described herein to comprise a TAM sequence, and thus be a TAM target nucleic acid.
  • a TAM target nucleic acid refers to a target nucleic acid that has been amplified to insert a TAM sequence that is recognized by an effector protein system.
  • target nucleic acids comprise a mutation.
  • a composition, system or method described herein can be used to modify a target nucleic acid comprising a mutation such that the mutation is modified to be a wild-type nucleotide or nucleotide sequence.
  • a composition, system or method described herein can be used to detect a target nucleic acid comprising a mutation.
  • the mutation may be a mutation of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more nucleotides.
  • the mutation can be a deletion of about 5, about 10, about 15, about 20, about 25, about 30, about 35, about 40, about 45, about 50, about 55, about 60, about 65, about 70, about 75, about 80, about 85, about 90, about 95, about 100, about 200, about 300, about 400, about 500, about 600, about 700, about 800, about 900, or about 1000 nucleotides.
  • the mutation can be a deletion of 1 to 5, 5 to 10, 10 to 15, 15 to 20, 20 to 25, 25 to 30, 30 to 35, 35 to 40, 40 to 45, 45 to 50, 50 to 55, 55 to 60, 60 to 65, 65 to 70, 70 to 75, 75 to 80, 80 to 85, 85 to 90, 90 to 95, 95 to 100, 100 to 200, 200 to 300, 300 to 400, 400 to 500, 500 to 600, 600 to 700, 700 to 800, 800 to 900, 900 to 1000, 1 to 50, 1 to 100, 25 to 50, 25 to 100, 50 to 100, 100 to 500, 100 to 1000, or 500 to 1000 nucleotides.
  • the mutation can be in the open reading frame of a target nucleic acid that results in the insertion of at least one amino acid in a protein encoded by the target nucleic acid.
  • the mutation can also be in the open reading frame of a target nucleic acid that results in the deletion of at least one amino acid in a protein encoded by the target nucleic acid.
  • the mutation can be in the open reading frame of a target nucleic acid that results in the substitution of at least one amino acid in a protein encoded by the target nucleic acid.
  • a mutation that results in the deletion, insertion, or substitution of one or more amino acids of a protein encoded by the target nucleic acid can result in misfolding of the protein.
  • the mutation can result in a premature stop codon.
  • the mutation can result in a truncation of the protein.
  • At least a portion of a guide nucleic acid of a composition described herein hybridizes to a region of the target nucleic acid comprising the mutation. In some embodiments, at least a portion of a guide nucleic acid of a composition described herein hybridizes to a region of the target nucleic acid that is within 10 nucleotides, within 50 nucleotides, within 100 nucleotides, or within 200 nucleotides of the mutation. The mutation may be located in a non-coding region or a coding region of a gene.
  • compositions, systems, and methods described herein comprise a target nucleic acid may be responsible for a disease, contain a mutation (e.g., single strand polymorphism, point mutation, insertion, or deletion), be contained in an amplicon, or be uniquely identifiable from the surrounding nucleic acids (e.g., contain a unique sequence of nucleotides).
  • the target nucleic acid has undergone a modification (e.g., an editing) after contacting with an RNP.
  • the editing is a change in the sequence of the target nucleic acid.
  • the change comprises an insertion, deletion, or substitution of one or more nucleotides compared to the target nucleic acid that has not undergone any modification.
  • the mutation is an autosomal dominant mutation. In some embodiments, the mutation is a dominant negative mutation. In some embodiments, the mutation is a loss of function mutation. In some embodiments, the mutation is a single nucleotide polymorphism (SNP). In some embodiments, the SNP is associated with a phenotype of the sample or a phenotype of the organism from which the sample was taken. The SNP, in some cases, is associated with altered phenotype from wild type phenotype.
  • the SNP may be a synonymous substitution or a nonsynonymous substitution. The nonsynonymous substitution may be a missense substitution, or a nonsense point mutation. The synonymous substitution may be a silent substitution.
  • the mutation may be a deletion of one or more nucleotides.
  • the single nucleotide mutation, SNP, or deletion is associated with a disease such as cancer or a genetic disorder.
  • the mutation such as a single nucleotide mutation, a SNP, or a deletion, may be encoded in the sequence of a target nucleic acid from the germline of an organism or may be encoded in a target nucleic acid from a diseased cell, such as a cancer cell.
  • the target nucleic acid comprises a mutation associated with a disease.
  • a mutation associated with a disease refers to a mutation whose presence in a subject indicates that the subject is susceptible to, or suffers from, a disease, disorder, condition, or syndrome.
  • a mutation associated with a disease refers to a mutation which causes, contributes to the development of, or indicates the existence of the disease, disorder, condition, or syndrome.
  • a mutation associated with a disease can also refer to any mutation which generates transcription or translation products at an abnormal level, or in an abnormal form, in cells affected by a disease relative to a control without the disease. In some embodiments, the mutation causes the disease.
  • the target nucleic acid is from a gene with a mutation associated with a genetic disorder, from a gene whose overexpression is associated with a genetic disorder, from a gene associated with abnormal cellular growth resulting in a genetic disorder, or from a gene associated with abnormal cellular metabolism resulting in a genetic disorder.
  • diseases associated with genetic mutations are recited in TABLE 4.
  • the disease may comprise, at least in part, a cancer, an inherited disorder, an ophthalmological disorder, a neurological disorder, a neurodegenerative disease, a blood disorder, a metabolic disorder, a genetic disorder, an infection, or a combination thereof.
  • the cancer is a solid cancer (i.e., a tumor).
  • the cancer is a blood cell cancer, including a leukemia or lymphoma.
  • the cancer is colon cancer, rectal cancer, renal -cell carcinoma, liver cancer, bladder cancer, cancer of the kidney or ureter, lung cancer, cancer of the small intestine, esophageal cancer, melanoma, bone cancer, pancreatic cancer, skin cancer, brain cancer (e.g., glioblastoma), cancer of the head or neck, melanoma, uterine cancer, ovarian cancer, breast cancer, testicular cancer, cervical cancer, stomach cancer, Hodgkin's Disease, non-Hodgkin's lymphoma, thyroid cancer.
  • brain cancer e.g., glioblastoma
  • melanoma uterine cancer
  • ovarian cancer breast cancer
  • testicular cancer cervical cancer
  • stomach cancer Hodgkin's Disease
  • non-Hodgkin's lymphoma thyroid cancer.
  • the cancer may be a leukemia, such as, by way of non-limiting example, acute myeloid (or myelogenous) leukemia (AML), chronic myeloid (or myelogenous) leukemia (CML), acute lymphocytic (or lymphoblastic) leukemia (ALL), and chronic lymphocytic leukemia (CLL).
  • AML acute myeloid (or myelogenous) leukemia
  • CML chronic myeloid (or myelogenous) leukemia
  • ALL acute lymphocytic leukemia
  • CLL chronic lymphocytic leukemia
  • the target nucleic acid comprises a portion of a gene comprising a mutation associated with cancer, a gene whose overexpression is associated with cancer, a tumor suppressor gene, an oncogene, a checkpoint inhibitor gene, a gene associated with cellular growth, a gene associated with cellular metabolism, or a gene associated with cell cycle.
  • the target nucleic acid encodes a cancer biomarker, such as a prostate cancer biomarker or non-small cell lung cancer.
  • the assay may be used to detect “hotspots” in target nucleic acids that may be predictive of lung cancer.
  • the target nucleic acid comprises a portion of a nucleic acid that is associated with a hemorrhagic fever.
  • the target nucleic acid is a portion of a nucleic acid from a genomic locus, any DNA amplicon of, a reverse transcribed mRNA, or a cDNA from a locus of at least one of the genes recited in TABLE 5.
  • the target nucleic acid is a portion of a nucleic acid from a genomic locus, any DNA amplicon of, a reverse transcribed mRNA, or a cDNA from a locus of at least one of: ABL, ACE, AF4/HRX, AKT-2, ALK, ALK/NPM, AML1, AML1/MTG8, APC, ATM, AXLN2, AXL, BAP1, BARD1, BCL-2, BCL-3, BCL- 6, BCR/ ABL, BLM, BMPR1A, BRCA1, BRCA2, BRIP1, c-MYC, CASR, CCR5, CDC73, CDH1, CDK4, CDKN1B, CDKN1C, CDKN2A, CEBPA, CHEK2, CREBBP, CTNNA1, DBL, DEK/CAN, DICER1, DIS3L2, E2A/PBX1, EGFR, ENL/HRX, EPCAM, ERG/
  • Non-limiting examples of oncogenes are KRAS, NRAS, BRAF, MYC, CTNNB1, and EGFR.
  • the oncogene is a gene that encodes a cyclin dependent kinase (CDK).
  • CDKs are Cdkl, Cdk4, Cdk5, Cdk7, Cdk8, Cdk9, Cdkll and CDK20.
  • tumor suppressor genes are TP53, RBI, and PTEN. Any region of the aforementioned gene loci may be probed for a mutation or deletion using the compositions and methods disclosed herein.
  • the compositions and methods for detection disclosed herein may be used to detect a single nucleotide polymorphism or a deletion.
  • the target nucleic acid comprises a portion of a nucleic acid from a genomic locus, any DNA amplicon of, a reverse transcribed mRNA, or a cDNA from a locus of at least one of: TRAC, B2M, PD1, PCSK9, DNMT1, HPRT1, RPL32P3, CCR5, FANCF, GRIN2B, EMX1, AAVS1, ALKBH5, CLTA, CDK11, CTNNB1, AXIN1, LRP6, TBK1, BAP1, TLES, PPM1A, BCL2L2, SUFU, RICTOR, VPS35, TOPI, SIRT1, PTEN, MMD, PAQR8, H2AX, P0U5F1, 0CT4, SYS1, ARFRP1, TSPAN14, EMC2, EMC3, SEL1L, DERL2, UBE2G2, UBE2J1, an HRDl.
  • the method for treating a disease comprises modifying at least one gene associated with the disease or modifying expression of the at least one gene such that the disease is treated.
  • the disease is Alzheimer’s disease and the gene is selected from APP, BACE-1, PSD95, MAPT, PSEN1, PSEN2, and APOEe4.
  • the disease is congenital muscular dystrophy 1A (MDC1A) and the gene is LAMA1 or LAMA2.
  • the disease is Ullrich Congenital Muscular Dystrophy (UCMD) and the gene is selected from COL6A1, COL6A2 and COL6A3.
  • the disease is Limb Girdle Muscular Dystrophies (LGMD1B, LGMD2A, LGMD2B) and the gene is selected from EMNA, DYSF, and CAPN3.
  • the disease is Nemaline Myopathy and the gene is selected from ACTA1, NEB, TPM2, TPM3, TNNT1, TNNT3, TNNI2 and EM0D3.
  • the disease is Parkinson’s disease and the gene is selected from SNCA, GDNF, and LRRK2. In some embodiments, the disease comprises Centronuclear myopathy and the gene is DNM2. In some embodiments, the disease is Huntington's disease and the gene is HTT. In some embodiments, the disease is Alpha-1 antitrypsin deficiency (AATD) and the gene is SEBPINAE In some embodiments, the disease is amyotrophic lateral sclerosis (ALS) and the gene is selected from SOD1, FUS, C9ORF72, ATXN2, TARDBP, and CHCHD10. In some embodiments, the disease comprises Alexander Disease and the gene is GFAP.
  • AATD Alpha-1 antitrypsin deficiency
  • ALS amyotrophic lateral sclerosis
  • the disease comprises anaplastic large cell lymphoma and the gene is CD30.
  • the disease comprises Angelman Syndrome and the gene is UBE3A.
  • the disease comprises Calcific Aortic Stenosis and the gene is Apo(a).
  • the disease comprises CD3Z-associated primary T-cell immunodeficiency and the gene is CD3Z or CD247.
  • the disease comprises CD18 deficiency and the gene is ITGB2.
  • the disease comprises CD40L deficiency and the gene is CD40L.
  • the disease comprises CNS trauma and the gene is VEGF.
  • the disease comprises coronary heart disease and the gene is selected from FGA, FGB, and FGG.
  • the disease comprises MECP2 Duplication syndrome and Rett syndrome and the gene is MECP2.
  • the disease comprises a bleeding disorder (coagulation) and the gene is FXI.
  • the disease comprises fragile X syndrome and the gene is FMRP
  • the disease comprises Fuchs Corneal Dystrophy and the gene is selected from ZEB1, SLC4A11, and LOXHD1.
  • the disease comprises GM2-Gangliosidoses (e.g., Tay Sachs Disease, Sandhoff disease) and the gene is selected from HEXA and HEXB.
  • the disease comprises Hearing loss disorders and the gene is DFNA36.
  • the disease is Pompe disease, including infantile onset Pompe Disease (IOPD) and late onset Pompe Disease (LOPD) and the gene is GAA.
  • the disease is Retinitis pigmentosa and the gene is selected from PDE6B, RHO, RP1, RP2, RPGR, PRPH2, IMPDH1, PRPF31, CRBI, PRPF8, TULP1, CA4, HPRPF3, ABCA4, EYS, CERKL, FSCN2, TOPORS, SNRNP200, PRCD, NR2E3, MERTK, USH2A, PR0M1, KLHL7, CNGBI, TTC8, ARL6, DHDDS, BESTI, LRAT, SPARA7, CRX, CLRN1, RPE65, and WDR19.
  • the disease comprises Leber Congenital Amaurosis Type 10 and the gene is CEP290.
  • the disease is cardiovascular disease and/or lipodystrophies and the gene is selected from ABCG5, ABCG8, AGP, ANGPTL3, APOCIH, APOA1, APOL1, ARH, CDKN2B, CFB, CXCLI2, FXI, FXH, GATA-4, MIAS, MKL2, MTHFD1L, MYH7, NKX2-5, NOTCH1, PKK, PCSK9, PSRC1, SMAD3, and TTR.
  • the disease is cardiovascular disease and/or lipodystrophies and the gene ANGPTL3.
  • the disease is cardiovascular disease and/or lipodystrophies and the gene is PCSK9. In some embodiments, the disease is cardiovascular disease and/or lipodystrophies and the gene is TTR. In some embodiments, the disease is severe hypertriglyceridemia (SHTG) and the gene is APOCHI or ANGPTL4. In some embodiments, the disease comprises acromegaly and the gene is GHR. In some embodiments, the disease comprises acute myeloid leukemia and the gene is CD22. In some embodiments, the disease is diabetes and the gene is GCGR. In some embodiments, the disease is NAFLD/NASH and the gene is selected from HSD17B13, PSD3, GPAM, CIDEB, DGAT2 and PNPLA3.
  • the disease is NASH/cirrhosis and the gene is MARC1.
  • the disease is cancer and the gene is selected from STAT3, YAP1, FOXP3, AR (Prostate cancer), and IRF4 (multiple myeloma).
  • the disease is cystic fibrosis and the gene is CFTR.
  • the disease is Duchenne Muscular Dystrophy and the gene is DMD.
  • the disease is ornithine transcarbamylase deficiency (OTCD) and the gene is OTC.
  • the disease is congenital adrenal hyperplasia (CAH) and the gene is CYP21 A2.
  • the disease is atherosclerotic cardiovascular disease (ASCVD) and the gene is LPA.
  • the disease is hepatitis B virus infection (CHB) and the gene is HBV covalently closed circular DNA (cccDNA).
  • the disease is citrullinemia type I and the gene is ASS1.
  • the disease is citrullinemia type I and the gene is SLC25A13.
  • the disease is citrullinemia type I and the gene is ASS1.
  • the disease is arginase-1 deficiency and the gene is ARG1.
  • the disease is carbamoyl phosphate synthetase I deficiency and the gene is CPS1.
  • the disease is argininosuccinic aciduria and the gene is ASL.
  • the disease comprises angioedema and the gene is PKK.
  • the disease comprises thalassemia and the gene is TMPRSS6.
  • the disease comprises achondroplasia and the gene is FGFR3.
  • the disease comprises Cri du chat syndrome and the gene is selected from CTNND2.
  • the disease comprises sickle cell anemia and the gene is Beta globin gene.
  • the disease comprises Alagille Syndrome and the gene is selected from JAG1 and N0TCH2. In some embodiments, the disease comprises Charcot Marie Tooth Disease and the gene is selected from PMP22 and MFN2. In some embodiments, the disease comprises Crouzon syndrome and the gene is selected from FGFR2, FGFR3, and FGFR3. In some embodiments, the disease comprises Dravet Syndrome and the gene is selected from SCN1A and SCN2A. In some embodiments, the disease comprises Emery-Dreifuss syndrome and the gene is selected from EMD, LMNA, SYNE1, SYNE2, FHL1, and TMEM43. In some embodiments, the disease comprises Factor V Leiden Thrombophilia and the gene is F5.
  • the disease is fabry disease and the gene is GLA.
  • the disease is facioscapulohumeral muscular dystrophy (FSHD) and the gene is FSHDE
  • the disease comprises Fanconi anemia and the gene is selected from FANCA, FANCB, FANCC, FANCD1, FANCD2, FANCE, FANCF, FANCG, FANCI, FANCJ, FANCY, FANCM, FANCN, FANCP, FANCS, RAD51C, and XPF.
  • the disease comprises Familial Creutzfeld-Jakob Disease and the gene is PRNP.
  • the disease comprises Familial Mediterranean Fever and the gene isMEFV.
  • the disease comprises Friedreich's ataxia and the gene is FXN.
  • the disease comprises Gaucher disease and the gene is GBA.
  • the disease comprises human papilloma virus (HPV) infection and the gene is HPVE7.
  • the disease comprises Hemochromatosis and the gene G HFE, optionally comprising a C282Y mutation.
  • the disease comprises Hemophilia A and the gene is FVIII.
  • the disease is hereditary angioedema and the gene is SERPING1 or KLKB1.
  • the disease comprises histiocytosis and the gene is CD1.
  • the disease comprises immunodeficiency 17 and the gene is CD3D. In some embodiments, the disease comprises immunodeficiency 13 and the gene is CD4. In some embodiments, the disease comprises Common Variable Immunodeficiency and the gene is selected from CD19 and CD81. In some embodiments, the disease comprises Joubert syndrome and the gene is selected from INPP5E, TMEM216, AHI1, NPHP1, CEP290, TMEM67, RPGRIP1L, ARE13B, CC2D2A, 0FD1, TMEM138, TCTN3, ZNF423, and AMRC9. In some embodiments, the disease comprises leukocyte adhesion deficiency and the gene is CD18.
  • the disease comprises Li-Fraumeni syndrome and the gene is TP53.
  • the disease comprises lymphoproliferative syndrome and the gene is CD27.
  • the disease comprises Lynch syndrome and the gene is selected from MSH2, MLH1, MSH6, PMS2, PMS1, TGFBR2, and MLH3.
  • the disease comprises mantle cell lymphoma and the gene is CD5.
  • the disease comprises Marfan syndrome and the gene is FBNE
  • the disease comprises mastocytosis and the gene is CD2.
  • the disease comprises methylmalonic acidemia and the gene is selected from MMAA, MMAB, and MUT.
  • the disease is mycosis fungoides and the gene is CD7. In some embodiments, the disease is myotonic dystrophy and the gene is selected from CNBP and DMPK. In some embodiments, the disease comprises neurofibromatosis and the gene is selected from NF1, and NF2. In some embodiments, the disease comprises osteogenesis imperfecta and the gene is selected from COL1A1, COL1A2, and IFITM5. In some embodiments, the disease is non-small cell lung cancer and the gene is selected from KRAS, EGFR, ALK, METexl4, BRAF V600E, ROS1, RET, an NTRK.
  • the disease comprises Peutz-Jeghers syndrome and the gene is STK1P
  • the disease comprises polycystic kidney disease and the gene is selected from PKD1 and PKD2.
  • the disease comprises Severe Combined Immune Deficiency and the gene is selected from IL7R, RAG1, JAK3.
  • the disease comprises PRKAG2 cardiac syndrome and the gene is PRKAG2.
  • the disease comprises Spinocerebellar ataxia and the gene is selected from ATXN1, ATXN2, ATXN3, PLEKHG4, SPTBN2, CACNA1A, ATXN7, ATXN8OS, ATXN10, TTBK2, PPP2R2B, KCNC3, PRKCG, ITPR1, TBP, KCND3, and FGF14.
  • the disease is thrombophilia due to antithrombin III deficiency and the gene is SERPINCL
  • the disease is spinal muscular atrophy and the gene is SMN1.
  • the disease comprises Usher Syndrome and the gene is selected from MY07A, USH1C, CDH23, PCDH15, USH1G, USH2A, GPR98, DFNB31, and CLRN1.
  • the disease comprises von Willebrand disease and the gene is VWF.
  • the disease comprises Waardenburg syndrome and the gene is selected from PAX3, MITF, WS2B, WS2C, SNAI2, EDNRB, EDN3, and SOXIO.
  • the disease comprises Wiskott-Aldrich Syndrome and the gene is WAS.
  • the disease comprises von Hippel-Lindau disease and the gene is VHL.
  • the disease comprises Wilson disease and the gene is ATP7B.
  • the disease comprises Zellweger syndrome and the gene is selected from PEX1, PEX2, PEX3, PEX5, PEX6, PEX10, PEX12, PEX13, PEX14, PEX16, PEX19, and PEX26.
  • the disease comprises infantile myofibromatosis and the gene is CD34.
  • the disease comprises platelet glycoprotein IV deficiency and the gene is CD36.
  • the disease comprises immunodeficiency with hyper-IgM type 3 and the gene is CD40.
  • the disease comprises hemolytic uremic syndrome and the gene is CD46.
  • the disease comprises complement hyperactivation, angiopathic thrombosis, or protein-losing enteropathy and the gene is CD55.
  • the disease comprises hemolytic anemia and the gene is CD59.
  • the disease comprises calcification of joints and arteries and the gene is CD73.
  • the disease comprises immunoglobulin alpha deficiency and the gene is CD79A.
  • the disease comprises C syndrome and the gene is CD96.
  • the disease comprises hairy cell leukemia and the gene is CD123.
  • the disease comprises histiocytic sarcoma and the gene is CD163.
  • the disease comprises autosomal dominant deafness and the gene is CD164. In some embodiments, the disease comprises immunodeficiency 25 and the gene is CD247. In some embodiments, the disease comprises methymalonic acidemia due to transcobalamin receptor defect and the gene is CD320.
  • treatment of a disease comprises administration of a gene therapy.
  • a gene therapy comprises use of a vector to introduce a functional gene or transgene.
  • vectors comprise nonviral vectors, including cationic polymers, cationic lipids, or bio-responsive polymers.
  • the bio-responsive polymer exploits chemi cal -physical properties of the endosomal environment (e.g., pH) to preferentially release the genetic material in the intracellular space.
  • vectors comprise viral vectors, including retroviruses, adenoviruses, adeno-associated viruses, and herpes simplex viruses.
  • the vector comprises a replication-defective viral vector, comprising an insertion of a therapeutic gene inserted in genes essential to the lytic cycle, preventing the virus from replicating and exerting cytotoxic effects.
  • a replication-defective viral vector comprising an insertion of a therapeutic gene inserted in genes essential to the lytic cycle, preventing the virus from replicating and exerting cytotoxic effects.
  • Methods of gene therapy are described in more detail in Ingusci et al., “Gene Therapy Tools for Brain Diseases". Front. Pharmacol. 10:724 (2019) which is hereby incorporated by reference in its entirety.
  • the target nucleic acid may be from any organism, including, but not limited to, a bacterium, a virus, a parasite, a protozoon, a fungus, a mammal, a plant, and an insect.
  • the target nucleic acid may be responsible for a disease, contain a mutation (e.g., single strand polymorphism, point mutation, insertion, or deletion), be contained in an amplicon, or be uniquely identifiable from the surrounding nucleic acids (e.g., contain a unique sequence of nucleotides).
  • the target nucleic acid is from a bacteria.
  • the bacteria is Acholeplasma laidlctw ii.
  • Brucella abortus Chlamydia psittaci, Chlamydia trachomatis, Cryptococcus neoformans, Escherichia coli, Legionella pneumophila, Lyme disease spirochetes, methicillin-resistant Staphylococcus aureus, Mycobacterium leprae, Mycobacterium tuberculosis, Mycoplasma arginini, Mycoplasma arthritidis, Mycoplasma genitalium, Mycoplasma hyorhinis, Mycoplasma or ale, Mycoplasma pneumoniae, Mycoplasma salivarium, Neisseria gonorrhoeae, Neisseria meningitidis, Pneumococcus, Pseudomonas aeruginosa, Streptococcus agalactiae, Streptococcus pyogenes, Treponema pallidum, or any combination thereof.
  • the target nucleic acid is from a virus.
  • the virus is adenovirus, blue tongue virus, chikungunya, coronavirus (e.g. SARS-CoV-2), cytomegalovirus, Dengue virus, Ebola, Epstein-Barr virus, feline leukemia virus, Hemophilus influenzae B, Hepatitis Virus A, Hepatitis Virus B, Hepatitis Virus C, herpes simplex virus I, herpes simplex virus II, human papillomavirus (HPV) including HP VI 6 and HP VI 8, human serum parvo-like virus, human T-cell leukemia viruses, immunodeficiency virus (e.g HIV), influenza virus, lymphocytic choriomeningitis virus, measles virus, mouse mammary tumor virus, mumps virus, murine leukemia virus, polio virus, rabies virus, Reovirus, respiratory syncytial virus (RS)
  • SARS-CoV-2 coron
  • the target nucleic acid is from a parasite.
  • the parasite is a helminth, an annelid, a platyhelminth, a nematode, or a thorny-headed worms.
  • the parasite is Babesia bovis, Echinococcus granulosus, Eimeria tenella, Leishmania tropica, Mesocestoides corti, Onchocerca volvulus, Plasmodium falciparum, Plasmodium vivax, Schistosoma japonicum, Schistosoma mansoni, Schistosoma spp., Taenia hydatigena, Taenia ovis, Taenia saginata, Theileria parva, Toxoplasma gondii, Toxoplasma spp., Trichinella spiralis, Trichomonas vaginalis, Trypanosoma brucei, Trypanosoma cruzi, Trypanosoma rangeli, Trypanosoma rhodesiense, Balantidium coli, Entamoeba histolytica, Giardia spp., Isospora spp., Trichomonas
  • the target nucleic acid comprises a nucleic acid sequence from a pathogen responsible for a disease.
  • pathogens are bacteria, a virus and a fungus.
  • the target nucleic acid in some embodiments, is a portion of a nucleic acid from a sexually transmitted infection or a contagious disease.
  • the target nucleic acid is a portion of a nucleic acid from a genomic locus, or any DNA amplicon, such as a reverse transcribed mRNA or a cDNA from a gene locus, a transcribed mRNA, or a reverse transcribed cDNA from a gene locus in at least one of: human immunodeficiency virus (HIV), human papillomavirus (HPV), chlamydia, gonorrhea, syphilis, trichomoniasis, sexually transmitted infection, malaria, Dengue fever, Ebola, chikungunya, and leishmaniasis.
  • HCV human immunodeficiency virus
  • HPV human papillomavirus
  • chlamydia gonorrhea
  • syphilis syphilis
  • trichomoniasis sexually transmitted infection
  • malaria Dengue fever
  • Ebola chikungunya
  • leishmaniasis
  • Pathogens include viruses, fungi, helminths, protozoa, malarial parasites, Plasmodium parasites, Toxoplasma parasites, and Schistosoma parasites.
  • Helminths include roundworms, heartworms, and phytophagous nematodes, flukes, Acanthocephala, and tapeworms.
  • Protozoan infections include infections from Giardia spp., Trichomonas spp., African trypanosomiasis, amoebic dysentery, babesiosis, balantidial dysentery, Chaga's disease, coccidiosis, malaria and toxoplasmosis.
  • pathogens such as parasitic/protozoan pathogens include, but are not limited to: Plasmodium falciparum, P. vivax, Trypanosoma cruzi and Toxoplasma gondii .
  • Fungal pathogens include, but are not limited to Cryptococcus neoformans, Histoplasma capsulatum, Coccidioides immitis, Blastomyces dermatitidis, Chlamydia trachomatis, and Candida albicans.
  • Pathogenic viruses include but are not limited to coronavirus (e.g., SARS- CoV-2); immunodeficiency virus (e.g., HIV); influenza virus; dengue; West Nile virus; herpes virus; yellow fever virus; Hepatitis Virus C; Hepatitis Virus A; Hepatitis Virus B; papillomavirus; and the like.
  • coronavirus e.g., SARS- CoV-2
  • immunodeficiency virus e.g., HIV
  • influenza virus e.g., dengue; West Nile virus; herpes virus; yellow fever virus; Hepatitis Virus C; Hepatitis Virus A; Hepatitis Virus B; papillomavirus; and the like.
  • Pathogens include, e.g., HIV virus, Mycobacterium tuberculosis, Streptococcus agalactiae, methicillin-resistant Staphylococcus aureus, Legionella pneumophila, Streptococcus pyogenes, Escherichia coli, Neisseria gonorrhoeae, Neisseria meningitidis, Pneumococcus, Cryptococcus neoformans, Histoplasma capsulatum, Hemophilus influenzae B, Treponema pallidum, Lyme disease spirochetes, Pseudomonas aeruginosa, Mycobacterium leprae, Brucella abortus, rabies virus, influenza virus, cytomegalovirus, herpes simplex virus I, herpes simplex virus II, human serum parvo-like virus, respiratory syncytial virus (RSV), M.
  • HIV virus e.g.
  • the target sequence is a portion of a nucleic acid from a genomic locus, a transcribed mRNA, or a reverse transcribed cDNA from a gene locus of bacterium or other agents responsible for a disease in the sample comprising a mutation that confers resistance to a treatment, such as a single nucleotide mutation that confers resistance to antibiotic treatment.
  • the target nucleic acid comprises a nucleic acid sequence of a virus, a bacterium, or other pathogen responsible for a disease in a plant (e.g., a crop).
  • Methods and compositions of the disclosure may be used to treat or detect a disease in a plant.
  • the methods of the disclosure may be used to target a viral nucleic acid sequence in a plant.
  • An effector protein of the disclosure may cleave the viral nucleic acid.
  • the target nucleic acid comprises a nucleic acid sequence of a virus or a bacterium or other agents (e.g., any pathogen) responsible for a disease in the plant (e.g., a crop).
  • the target nucleic acid comprises RNA.
  • the target nucleic acid in some embodiments, is a portion of a nucleic acid from a virus or a bacterium or other agents responsible for a disease in the plant (e.g., a crop).
  • the target nucleic acid is a portion of a nucleic acid from a genomic locus, or any NA amplicon, such as a reverse transcribed mRNA or a cDNA from a gene locus, a transcribed mRNA, or a reverse transcribed cDNA from a gene locus in at a virus or a bacterium or other agents (e.g., any pathogen) responsible for a disease in the plant (e.g., a crop).
  • a virus infecting the plant may be an RNA virus.
  • a virus infecting the plant may be a DNA virus.
  • TMV Tobacco mosaic virus
  • TSWV Tomato spotted wilt virus
  • CMV Cucumber mosaic virus
  • PVY Potato virus Y
  • PMV Cauliflower mosaic virus
  • PV Plum pox virus
  • BMV Brome mosaic virus
  • PVX Potato virus X
  • the target nucleic acid is any one of: a naturally occurring eukaryotic sequence, a naturally occurring prokaryotic sequence, a naturally occurring viral sequence, a naturally occurring bacterial sequence, a naturally occurring fungal sequence, an engineered eukaryotic sequence, an engineered prokaryotic sequence, an engineered viral sequence, an engineered bacterial sequence, an engineered fungal sequence, a fragment of a naturally occurring sequence, a fragment of an engineered sequence, and combinations thereof.
  • the target nucleic acid is isolated from any one of: a naturally occurring cell, a eukaryotic cell, a prokaryotic cell, a plant cell, a fungal cell, an animal cell, cell of an invertebrate, a fly cell, a cell of a vertebrate, a mammalian cell, a primate cell, a nonhuman primate cell, a human cell, a living cell, a non-living cell, a modified cell, a derived cell, and a non-naturally occurring cell.
  • Nucleic acids such as DNA and pre-mRNA, described herein can contain at least one intron and at least one exon, wherein as read in the 5’ to the 3’ direction of a nucleic acid strand, the 3’ end of an intron can be adjacent to the 5’ end of an exon, and wherein said intron and exon correspond for transcription purposes. If a nucleic acid strand contains more than one intron and exon, the 5’ end of the second intron is adjacent to the 3’ end of the first exon, and 5’ end of the second exon is adjacent to the 3’ end of the second intron.
  • nucleic acids can contain one or more elements that act as a signal during transcription, splicing, and/or translation.
  • signaling elements include a 5’SS, a 3’SS, a premature stop codon, U1 and/or U2 binding sequences, and cis acting elements such as branch site (BS), polypyridine tract (PYT), exonic and intronic splicing enhancers (ESEs and ISEs) or silencers (ESSs and ISSs).
  • nucleic acids may also comprise a untranslated region (UTR), such as a 5’ UTR or a 3’ UTR.
  • UTR untranslated region
  • the start of an exon or intron is referred to interchangeably herein as the 5’ end of an exon or intron, respectively.
  • the end of an exon or intron is referred to interchangeably herein as the 3’ end of an exon or intron, respectively.
  • At least a portion of at least one target sequence is within about 1, about 5 or more, about 10 or more, about 15 or more, about 20 or more, about 25 or more, about 30 or more, about 35 or more, about 40 or more, about 45 or more, about 50 or more, about 55 or more, about 60 or more, about 65 or more, about 70 or more, about 75 or more, about 80 or more, about 85 or more, about 90 or more, about 95 or more, about 100 or more, about 105 or more, about 110 or more, about 115 or more, about 120 or more, about 125 or more, about 130 or more, about 135 or more, about 140 or more, about 145 or more, or about 150 to about 300 nucleotides adjacent to: the 5’ end of an exon; the 3’ end of an exon; the 5’ end of an intron; the 3’ end of an intron; one or more signaling element comprising a 5’SS, a 3’SS, a premature stop codon, U1 binding sequence
  • the target nucleic acid comprises a target locus. In some embodiments, the target nucleic acid comprises more than one target loci. In some embodiments, the target nucleic acid comprises two target loci. Accordingly, in some embodiments, the target nucleic acid can comprise one or more target sequences.
  • compositions, systems, and methods described herein comprise an edited target nucleic acid which can describe a target nucleic acid wherein the target nucleic acid has undergone a change, for example, after contact with an effector protein.
  • the editing is an alteration in the sequence of the target nucleic acid.
  • the edited target nucleic acid comprises an insertion, deletion, or replacement of one or more nucleotides compared to the unedited target nucleic acid.
  • the editing is a mutation.
  • target nucleic acids described herein comprise a mutation.
  • a composition, system or method described herein can be used to edit a target nucleic acid comprising a mutation such that the mutation is edited to be the wild-type nucleotide or nucleotide sequence.
  • a composition, system or method described herein can be used to detect a target nucleic acid comprising a mutation.
  • a mutation may result in the insertion of at least one amino acid in a protein encoded by the target nucleic acid.
  • a mutation may result in the deletion of at least one amino acid in a protein encoded by the target nucleic acid.
  • a mutation may result in the substitution of at least one amino acid in a protein encoded by the target nucleic acid.
  • a mutation that results in the deletion, insertion, or substitution of one or more amino acids of a protein encoded by the target nucleic acid may result in misfolding of a protein encoded by the target nucleic acid.
  • a mutation may result in a premature stop codon, thereby resulting in a truncation of the encoded protein.
  • Non-limiting examples of mutations are insertion-deletion (indel), a point mutation, single nucleotide polymorphism (SNP), a chromosomal mutation, a copy number mutation or variation, and frameshift mutations.
  • an indel mutation is an insertion or deletion of one or more nucleotides.
  • a point mutation comprises a substitution, insertion, or deletion.
  • a frameshift mutation occurs when the number of nucleotides in the insertion/deletion is not divisible by three, and it occurs in a protein coding region.
  • a chromosomal mutation can comprise an inversion, a deletion, a duplication, or a translocation of one or more nucleotides.
  • a copy number variation can comprise a gene amplification or an expanding trinucleotide repeat.
  • an SNP is associated with a phenotype of the sample or a phenotype of the organism from which the sample was taken.
  • an SNP is associated with altered phenotype from wild type phenotype.
  • the SNP is a synonymous substitution or a nonsynonymous substitution.
  • the nonsynonymous substitution is a missense substitution or a nonsense point mutation.
  • the synonymous substitution is a silent substitution.
  • a target nucleic acid described herein comprises a mutation of one or more nucleotides.
  • the one or more nucleotides comprises 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more nucleotides.
  • the mutation comprises a deletion, insertion, and/or substitution of about 5, about 10, about 15, about 20, about 25, about 30, about 35, about 40, about 45, about 50, about 55, about 60, about 65, about 70, about 75, about 80, about 85, about 90, about 95, about 100, about 200, about 300, about 400, about 500, about 600, about 700, about 800, about 900, or about 1000 nucleotides.
  • the mutation comprises a deletion, insertion, and/or substitution of 1 to 5, 5 to 10, 10 to 15, 15 to 20, 20 to 25, 25 to 30, 30 to 35, 35 to 40, 40 to 45, 45 to 50, 50 to 55, 55 to 60, 60 to 65, 65 to 70, 70 to 75, 75 to 80, 80 to 85, 85 to 90, 90 to 95, 95 to 100, 100 to 200, 200 to 300, 300 to 400, 400 to 500, 500 to 600, 600 to 700, 700 to 800, 800 to 900, 900 to 1000, 1 to 50, 1 to 100, 25 to 50, 25 to 100, 50 to 100, 100 to 500, 100 to 1000, or 500 to 1000 nucleotides.
  • the mutation may be located in a non-coding region or a coding region of a gene, wherein the gene is a target nucleic acid.
  • a mutation may be in an open reading frame of a target nucleic acid.
  • guide nucleic acids described herein hybridize to a portion of the target nucleic acid comprising or adjacent to the mutation.
  • target nucleic acids comprise a mutation, wherein the mutation is a SNP.
  • the single nucleotide mutation or SNP is associated with a phenotype of the sample or a phenotype of the organism from which the sample was taken.
  • the SNP is associated with altered phenotype from wild type phenotype.
  • a single nucleotide mutation, SNP, or deletion described herein is associated with a disease, such as a genetic disease.
  • the SNP is a synonymous substitution or a nonsynonymous substitution.
  • the nonsynonymous substitution is a missense substitution or a nonsense point mutation.
  • the synonymous substitution is a silent substitution.
  • the mutation is a deletion of one or more nucleotides.
  • the single nucleotide mutation, SNP, or deletion is associated with a disease such as a genetic disorder.
  • the mutation, such as a single nucleotide mutation, a SNP, or a deletion may be encoded in the sequence of a target nucleic acid from the germline of an organism or may be encoded in a target nucleic acid from a diseased cell.
  • the mutation is associated with a disease, such as a genetic disorder.
  • the mutation may be encoded in the sequence of a target nucleic acid from the germline of an organism or may be encoded in a target nucleic acid from a diseased cell.
  • a target nucleic acid described herein comprises a mutation associated with a disease.
  • a mutation associated with a disease refers to a mutation whose presence in a subject indicates that the subject is susceptible to or suffers from, a disease, disorder, condition, or syndrome.
  • a mutation associated with a disease refers to a mutation which causes, contributes to the development of, or indicates the existence of the disease, disorder, condition, or syndrome.
  • a mutation associated with a disease may also refer to any mutation which generates transcription or translation products at an abnormal level, or in an abnormal form, in cells affected by a disease relative to a control without the disease.
  • a mutation associated with a disease comprises the co-occurrence of a mutation and the phenotype of a disease. The mutation may occur in a gene, wherein transcription or translation products from the gene occur at a significantly abnormal level or in an abnormal form in a cell or subject harboring the mutation as compared to a non-disease control subject not having the mutation.
  • a target nucleic acid described herein comprises a mutation associated with a disease described herein.
  • a target nucleic acid is in a cell.
  • the cell is a single-cell eukaryotic organism; a plant cell an algal cell; a fungal cell; an animal cell; a cell of an invertebrate animal; a cell of a vertebrate animal such as fish, amphibian, reptile, bird, and mammal; or a cell of a mammal such as a human, a non-human primate, an ungulate, a feline, a bovine, an ovine, and a caprine.
  • the cell is a eukaryotic cell.
  • the cell is a mammalian cell, a human cell, or a plant cell. In some embodiments, the cell is a human cell. In some embodiments, the human cell is a: muscle cell, liver cell, lung cell, cardiac cell, visceral cell, cardiac muscle cell, smooth muscle cell, cardiomyocyte, nodal cardiac muscle cell, smooth muscle cell, visceral muscle cell, skeletal muscle cell, myocyte, red (or slow) skeletal muscle cell, white (fast) skeletal muscle cell, intermediate skeletal muscle, muscle satellite cell, muscle stem cell, myoblast, muscle progenitor cell, induced pluripotent stem cell (iPS), or a cell derived from an iPS cell, modified to have its gene edited and differentiated into myoblasts, muscle progenitor cells, muscle satellite cells, muscle stem cells, skeletal muscle cells, cardiac muscle cells or smooth muscle cells.
  • iPS induced pluripotent stem cell
  • an effector protein-guide nucleic acid complex may comprise high selectivity for a target sequence.
  • an RNP comprise a selectivity of at least 200: 1, 100: 1, 50: 1, 20: 1, 10: 1, or 5: 1 for a target nucleic acid over a single nucleotide variant of the target nucleic acid.
  • an RNP may comprise a selectivity of at least 5: 1 for a target nucleic acid over a single nucleotide variant of the target nucleic acid.
  • some methods described herein may detect a target nucleic acid present in the sample in various concentrations or amounts as a target nucleic acid population.
  • the method detects at least 2 target nucleic acid populations.
  • the method detects at least 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 40, or 50 target nucleic acid populations.
  • the method detects 3 to 50, 5 to 40, or 10 to 25 target nucleic acid populations.
  • the method detects at least 2 individual target nucleic acids.
  • the method detects at least 3, 5, 10, 20, 30, 40, 50, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 2000, 3000, 4000, 5000, 6000, 7000, 8000, 9000, or 10000 individual target nucleic acids. In some embodiments, the method detects 1 to 10,000, 100 to 8000, 400 to 6000, 500 to 5000, 1000 to 4000, or 2000 to 3000 individual target nucleic acids.
  • the method detects target nucleic acid present at least at one copy per 10 non-target nucleic acids, 10 2 non-target nucleic acids, 10 3 non-target nucleic acids, 10 4 non-target nucleic acids, 10 5 non-target nucleic acids, 10 6 non-target nucleic acids, 10 7 non-target nucleic acids, 10 8 non-target nucleic acids, 10 9 non-target nucleic acids, or 10 10 non-target nucleic acids.
  • compositions described herein exhibit indiscriminate transcleavage of a nucleic acid (e.g., ssRNA or ssDNA), enabling their use for detection of a nucleic acid (e.g, RNA or DNA, respectively) in samples.
  • target nucleic acids are generated from many nucleic acid templates (e.g., RNA) in order to achieve cleavage of a reporter (e.g., a FQ reporter) in a device (e.g., a DETECTR platform).
  • effector proteins may be activated by a nucleic acid (e.g., ssDNA or ssRNA), upon which they may exhibit trans-cleavage of the nucleic acid (e.g., ssDNA or ssRNA) and may, thereby, be used to cleave reporter molecules (e.g., ssDNA or ssRNA FQ reporter molecules) in a device (e.g., a DETECTR).
  • a nucleic acid e.g., ssDNA or ssRNA
  • a DETECTR e.g., a DETECTR
  • reagents comprising a single stranded reporter nucleic acid comprising a detection moiety, wherein the reporter nucleic acid (e.g., a ssDNA-FQ reporter described herein) is capable of being cleaved by the effector protein, upon generation (e.g., cDNA) and amplification of nucleic acids from a nucleic acid template (e.g., ssRNA) using the methods disclosed herein, thereby generating a first detectable signal.
  • the reporter nucleic acid e.g., a ssDNA-FQ reporter described herein
  • the reporter nucleic acid e.g., a ssDNA-FQ reporter described herein
  • the reporter nucleic acid e.g., a ssDNA-FQ reporter described herein
  • the reporter nucleic acid e.g., a ssDNA-FQ reporter described herein
  • the reporter nucleic acid e.g., a ss
  • a target nucleic acid is an amplified nucleic acid of interest.
  • the nucleic acid of interest is any nucleic acid disclosed herein or from any sample as disclosed herein.
  • the nucleic acid of interest is an RNA that is reverse transcribed before amplification.
  • the nucleic acid of interest is amplified then the amplicons is transcribed into RNA.
  • target nucleic acids may activate an effector protein to initiate sequence-independent cleavage of a nucleic acid-based reporter (e.g., a reporter comprising an RNA sequence, or a reporter comprising DNA and RNA).
  • a nucleic acid-based reporter e.g., a reporter comprising an RNA sequence, or a reporter comprising DNA and RNA.
  • an effector protein of the present disclosure is activated by a target nucleic acid to cleave reporters having an RNA (also referred to herein as an “RNA reporter”).
  • an effector protein of the present disclosure is activated by a target nucleic acid to cleave reporters having an RNA.
  • an effector protein of the present disclosure is activated by a target RNA to cleave reporters having an RNA (also referred to herein as a “RNA reporter”).
  • the RNA reporter may comprise a single-stranded RNA labelled with a detection moiety or may be any RNA reporter as disclosed herein.
  • compositions for modifying a target nucleic acid in a cell or a subject comprising any one of the effector proteins, engineered effector proteins, or fusion proteins described herein.
  • pharmaceutical compositions comprising a nucleic acid encoding any one of the effector proteins, engineered effector proteins, or fusion proteins described herein.
  • pharmaceutical compositions comprise a guide nucleic acid.
  • pharmaceutical compositions comprise a plurality of guide nucleic acids.
  • Pharmaceutical compositions may be used to modify a target nucleic acid or the expression thereof in a cell in vitro, in vivo or ex vivo.
  • the pharmaceutically acceptable excipient, carrier or diluent allows the active ingredient to retain biological activity.
  • the pharmaceutically acceptable excipient, carrier or diluent is non-reactive with the subject's immune system.
  • the pharmaceutically acceptable excipient, carrier or diluent provides for long-term stabilization of the composition.
  • the pharmaceutically acceptable excipient, carrier or diluent is provided as a bulking agent in solid formulations that contain potent active ingredients in small amounts.
  • the pharmaceutically acceptable excipient, carrier or diluent confers a therapeutic enhancement on the active ingredient in the final dosage form.
  • the pharmaceutically acceptable excipient, carrier or diluent facilitates absorption, reduces viscosity, or enhances solubility.
  • the pharmaceutically acceptable excipient, carrier or diluent is selected based upon the route of administration, dosage form, active ingredient, other factors, or any combination thereof.
  • the pharmaceutically acceptable excipient, carrier or diluent can be formulated by well-known conventional methods (see, e.g., Remington's Pharmaceutical Sciences, 18th edition, A. Gennaro, ed., Mack Publishing Co., Easton, Pa., 1990; and Remington, The Science and Practice of Pharmacy 21st Ed. Mack Publishing, 2005).
  • compositions comprise one or more nucleic acids encoding an effector protein, effector partner, fusion protein, a guide nucleic acid, or a combination thereof; and a pharmaceutically acceptable carrier or diluent.
  • the effector protein, effector partner, fusion protein, or combination thereof may be any one of those described herein.
  • the one or more nucleic acids may comprise a plasmid.
  • the one or more nucleic acids may comprise a nucleic acid expression vector.
  • the one or more nucleic acids may comprise a viral vector.
  • the viral vector is a lentiviral vector.
  • the vector is an adeno-associated viral (AAV) vector.
  • AAV adeno-associated viral
  • compositions comprising a viral vector encoding a fusion protein and a guide nucleic acid, wherein at least a portion of the guide nucleic acid binds to the effector protein of the fusion protein.
  • pharmaceutical compositions comprise a virus comprising a viral vector encoding a fusion protein, an effector protein, an effector partner, a guide nucleic acid, or a combination thereof; and a pharmaceutically acceptable carrier or diluent.
  • compositions described herein may comprise a salt.
  • the salt is a sodium salt.
  • the salt is a potassium salt.
  • the salt is a magnesium salt.
  • the salt is NaCl.
  • the salt is KNO3.
  • the salt is Mg 2 + SO4 2 .
  • Non-limiting examples of pharmaceutically acceptable carriers and diluents suitable for the pharmaceutical compositions disclosed herein include buffers (e.g., neutral buffered saline, phosphate buffered saline); carbohydrates (e.g., glucose, mannose, sucrose, dextran, mannitol); polypeptides or amino acids (e.g., glycine); antioxidants; chelating agents (e.g., EDTA, glutathione); adjuvants (e.g., aluminum hydroxide); surfactants (Polysorbate 80, Polysorbate 20, or Pluronic F68); glycerol; sorbitol; mannitol; polyethyleneglycol; and preservatives.
  • buffers e.g., neutral buffered saline, phosphate buffered saline
  • carbohydrates e.g., glucose, mannose, sucrose, dextran, mannitol
  • polypeptides or amino acids e.g.
  • the vector is formulated for delivery through injection by a needle carrying syringe.
  • the composition is formulated for delivery by electroporation.
  • the composition is formulated for delivery by chemical method.
  • the pharmaceutical compositions comprise a virus vector or a non-viral vector.
  • pharmaceutical compositions are in the form of a solution (e.g., a liquid). The solution may be formulated for injection, e.g., intravenous or subcutaneous injection.
  • the pH of the solution is about 7, about 7.1, about 7.2, about 7.3, about 7.4, about 7.5, about 7.6, about 7.7, about 7.8, about 7.9, about 8, about 8.1, about 8.2, about 8.3, about 8.4, about 8.5, about 8.6, about 8.7, about 8.8, about 8.9, or about 9.
  • the pH is 7 to 7.5, 7.5 to 8, 8 to 8.5, 8.5 to 9, or 7 to 8.5.
  • the pH of the solution is less than 7.
  • the pH is greater than 7. VIII.
  • methods of modifying target nucleic acids or the expression thereof comprise editing a target nucleic acid.
  • editing refers to modifying the nucleobase sequence of a target nucleic acid.
  • methods of modulating the expression of a target nucleic acid Fusion proteins and systems described herein may be used for such methods.
  • Methods of editing a target nucleic acid may comprise one or more of cleaving the target nucleic acid, deleting one or more nucleotides of the target nucleic acid, inserting one or more nucleotides into the target nucleic acid, modifying one or more nucleotides of the target nucleic acid.
  • Methods of modulating expression of target nucleic acids may comprise modifying the target nucleic acid or a protein associated with the target nucleic acid, e.g., a histone.
  • methods comprise contacting a target nucleic acid with a composition described herein. In some embodiments, methods comprise contacting a target nucleic acid with an effector protein described herein. In some embodiments, methods comprise contacting a target nucleic acid with a fusion protein described herein.
  • the effector protein may be an effector protein provided in TABLE 1, TABLE 6, or TABLE 8, or a catalytically inactive variant thereof.
  • the effector protein may comprise an amino acid sequence that is at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99% or 100% identical to a sequence described in TABLE 1, TABLE 6, or TABLE 8.
  • the amino acid sequence of the effector protein is at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99% or 100% identical to a sequence described in TABLE 1, TABLE 6, or TABLE 8
  • methods comprise base editing.
  • base editing comprises contacting a target nucleic acid with a fusion protein comprising an effector protein fused to a base editing enzyme, such as a deaminase, thereby changing a nucleobase of the target nucleic acid to an alternative nucleobase.
  • the nucleobase of the target nucleic acid is adenine (A) and the method comprises changing A to guanine (G).
  • the nucleobase of the target nucleic acid is cytosine (C) and the method comprises changing C to thymine (T).
  • the nucleobase of the target nucleic acid is C and the method comprises changing C to G.
  • the nucleobase of the target nucleic acid is A and the method comprises changing A to G.
  • methods introduce a nucleobase change in a target nucleic acid relative to a corresponding wildtype or mutant nucleobase sequence.
  • methods remove or correct a disease-causing mutation in a nucleic acid sequence, e.g., to produce a corresponding wildtype nucleobase sequence.
  • methods remove/correct point mutations, deletions, null mutations, or tissue-specific mutations in a target nucleic acid.
  • methods generate gene knock-out, gene knock-in, gene editing, gene tagging, or a combination thereof. Methods of the disclosure may be targeted to a locus in a genome of a cell.
  • Modifying at least one gene using the compositions and methods described herein can, in some embodiments, induce a reduction or increase in expression of the one or more genes.
  • the at least one modified gene results in a reduction in expression, also referred to as gene silencing.
  • the gene silencing reduces expression of one or more genes by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or at least 95%.
  • gene silencing is accomplished by transcriptional silencing, post-transcriptional silencing, or meiotic silencing.
  • transcriptional silencing is by genomic imprinting, paramutation, transposon silencing, position effect, or RNA-directed DNA methylation.
  • post-transcriptional silencing is by RNA interference, RNA silencing, or nonsense mediated decay.
  • meiotic silencing is by transfection or meiotic silencing of unpaired DNA.
  • the at least one modified gene results in removing all expression, also referred to as the gene being knocked out (KO).
  • the compositions, methods or systems increase expression of one or more genes by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or at least 100%.
  • methods of editing a target nucleic acid or modulating the expression of a target nucleic acid are performed in vivo.
  • methods of editing a target nucleic acid or modulating the expression of a target nucleic acid are performed in vitro.
  • a plasmid may be modified in vitro using a composition described herein and introduced into a cell or organism.
  • methods of editing a target nucleic acid or modulating the expression of a target nucleic acid are performed ex vivo.
  • methods may comprise obtaining a cell from a subject, modifying a target nucleic acid in the cell with methods and compositions described herein, and returning the cell to the subject.
  • Methods of editing performed ex vivo may be particularly advantageous to produce CAR T-cells.
  • methods comprise editing a target nucleic acid or modulating the expression of the target nucleic acid in a cell or a subject.
  • the cell may be a dividing cell.
  • the cell may be a terminally differentiated cell.
  • the target nucleic acid is a gene.
  • the cell may be a prokaryotic cell.
  • the cell may be an archaeal cell.
  • the cell may be a eukaryotic cell.
  • the cell may be a mammalian cell.
  • the cell may be a human cell.
  • the cell may be a T cell.
  • the cell may be a hematopoietic stem cell.
  • the cell may be a bone marrow derived cell, a white blood cell, a blood cell progenitor, or a combination thereof.
  • Generating a genetically modified cell may comprise contacting a target cell with an effector protein or a fusion protein described herein and a guide nucleic acid. Contacting may comprise electroporation, acoustic poration, optoporation, viral vector-based delivery, iTOP, nanoparticle delivery (e.g., lipid or gold nanoparticle delivery), cell -penetrating peptide (CPP) delivery, DNA nanostructure delivery, or any combination thereof.
  • the nanoparticle delivery comprises lipid nanoparticle delivery or gold nanoparticle delivery.
  • the nanoparticle delivery comprises lipid nanoparticle delivery.
  • the nanoparticle delivery comprises gold nanoparticle delivery.
  • the compositions, methods or systems comprise a nucleic acid expression vector, or use thereof, to introduce an effector protein, guide nucleic acid, donor template or any combination thereof to a cell.
  • the nucleic acid expression vector is a viral vector.
  • Viral vectors include, but are not limited to, retroviruses, adenoviruses, adeno-associated viruses, and herpes simplex viruses.
  • the viral vector is a replication-defective viral vector, comprising an insertion of a therapeutic gene inserted in genes essential to the lytic cycle, preventing the virus from replicating and exerting cytotoxic effects.
  • the viral vector is an adeno associated viral (AAV) vector.
  • AAV adeno associated viral
  • the nucleic acid expression vector is a non-viral vector.
  • compositions and methods comprise a lipid, polymer, nanoparticle, or a combination thereof, or use thereof, to introduce a Cas protein, guide nucleic acid, donor template or any combination thereof to a cell.
  • lipids and polymers are cationic polymers, cationic lipids, or bio-responsive polymers.
  • the bio-responsive polymer exploits chemical-physical properties of the endosomal environment (e.g., pH) to preferentially release the genetic material in the intracellular space.
  • Methods of modifying may comprise contacting a target nucleic acid with one or more components, compositions or systems described herein.
  • a method of modifying comprises contacting a target nucleic acid with at least one of: a) one or more effector proteins, or one or more nucleic acids encoding one or more effector proteins; or b) one or more guide nucleic acids, or one or more nucleic acids encoding one or more guide nucleic acids.
  • a method of modifying comprises contacting a target nucleic acid with a system described herein wherein the system comprises components comprising at least one of: a) one or more effector proteins, or one or more nucleic acids encoding one or more effector proteins; or b) one or more guide nucleic acids, or one or more nucleic acids encoding one or more guide nucleic acids.
  • a method of modifying comprises contacting a target nucleic acid with a composition described herein comprising at least one of: a) one or more effector proteins, or one or more nucleic acids encoding one or more effector proteins; or b) one or more guide nucleic acids, or one or more nucleic acids encoding one or more guide nucleic acids; in a composition.
  • a method of modifying as described herein produces a modified target nucleic acid.
  • Editing a target nucleic acid sequence may introduce a mutation (e.g., point mutations, deletions) in a target nucleic acid relative to a corresponding wildtype nucleotide sequence. Editing may remove or correct a disease-causing mutation in a nucleic acid sequence to produce a corresponding wildtype nucleotide sequence. Editing a target nucleic acid sequence may remove/correct point mutations, deletions, null mutations, or tissue-specific mutations in a target nucleic acid. Editing a target nucleic acid sequence may be used to generate gene knock-out, gene knock-in, gene editing, gene tagging, or a combination thereof. Methods of the disclosure may be targeted to any locus in a genome of a cell.
  • a mutation e.g., point mutations, deletions
  • Modifying may comprise single stranded cleavage, double stranded cleavage, donor nucleic acid insertion, epigenetic modification (e.g., methylation, demethylation, acetylation, or deacetylation), or a combination thereof.
  • cleavage is site-specific, meaning cleavage occurs at a specific site in the target nucleic acid, often within the region of the target nucleic acid that hybridizes with the guide nucleic acid spacer sequence.
  • the effector proteins introduce a singlestranded break in a target nucleic acid to produce a cleaved nucleic acid.
  • the effector protein is capable of introducing a break in a single stranded RNA (ssRNA).
  • the effector protein may be coupled to a guide nucleic acid that targets a particular region of interest in the ssRNA.
  • the target nucleic acid, and the resulting cleaved nucleic acid is contacted with a nucleic acid for homologous recombination (e.g., homology directed repair (HDR)) or non -homologous end joining (NHEJ).
  • HDR homology directed repair
  • NHEJ non -homologous end joining
  • a doublestranded break in the target nucleic acid may be repaired (e.g., by NHEJ or HDR) without insertion of a donor template, such that the repair results in an indel in the target nucleic acid at or near the site of the double-stranded break.
  • an indel sometimes referred to as an insertion-deletion or indel mutation, is a type of genetic mutation that results from the insertion and/or deletion of one or more nucleotide in a target nucleic acid.
  • An indel may vary in length (e.g., 1 to 1,000 nucleotides in length) and be detected using methods well known in the art, including sequencing.
  • Indel percentage is the percentage of sequencing reads that show at least one nucleotide has been mutation that results from the insertion and/or deletion of nucleotides regardless of the size of insertion or deletion, or number of nucleotides mutated. For example, if there is at least one nucleotide deletion detected in a given target nucleic acid, it counts towards the percent indel value.
  • the target nucleic acid As another example, if one copy of the target nucleic acid has one nucleotide deleted, and another copy of the target nucleic acid has 10 nucleotides deleted, they are counted the same. This number reflects the percentage of target nucleic acids that are edited by a given effector protein.
  • methods of modifying described herein cleave a target nucleic acid at one or more locations to generate a cleaved target nucleic acid.
  • the cleaved target nucleic acid undergoes recombination (e.g., NHEJ or HDR).
  • cleavage in the target nucleic acid may be repaired (e.g., by NHEJ or HDR) without insertion of a donor nucleic acid, such that the repair results in an indel in the target nucleic acid at or near the site of the cleavage site.
  • cleavage in the target nucleic acid may be repaired (e.g., by NHEJ or HDR) with insertion of a donor nucleic acid, such that the repair results in an indel in the target nucleic acid at or near the site of the cleavage site.
  • compositions, systems, and methods of the present disclosure comprise an additional guide nucleic acid or a use thereof
  • dual-guided compositions, systems, and methods described herein may modify the target nucleic acid in two locations.
  • dual-guided modifying may comprise cleavage of the target nucleic acid in the two locations targeted by the guide nucleic acids.
  • the wild-type reading frame upon removal of the sequence between the guide nucleic acids, is restored.
  • a wild-type reading frame may be a reading frame that produces at least a partially, or fully, functional protein.
  • a non-wild-type reading frame may be a reading frame that produces a non-functional or partially non-functional protein.
  • compositions, systems, and methods described herein may edit 1 to 1,000 nucleotides or any integer in between, in a target nucleic acid.
  • 1 to 1,000, 2 to 900, 3 to 800, 4 to 700, 5 to 600, 6 to 500, 7 to 400, 8 to 300, 9 to 200, or 10 to 100 nucleotides, or any integer in between may be edited by the compositions, systems, and methods described herein.
  • 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more nucleotides may be edited by the compositions, systems, and methods described herein.
  • 10, 20, 30, 40, 50, 60, 70, 80 90, 100 or more nucleotides, or any integer in between may be edited by the compositions, systems, and methods described herein.
  • 100, 200, 300, 400, 500, 600, 700, 800, 900 or more nucleotides, or any integer in between may be edited by the compositions, systems, and methods described herein.
  • Methods may comprise use of two or more effector proteins.
  • An illustrative method for introducing a break in a target nucleic acid comprises contacting the target nucleic acid with: (a) a first engineered guide nucleic acid comprising a region that binds to a first effector protein described herein; and (b) a second engineered guide nucleic acid comprising a region that binds to a second effector protein described herein, wherein the first engineered guide nucleic acid comprises an additional region that hybridizes to the target nucleic acid and wherein the second engineered guide nucleic acid comprises an additional region that hybridizes to the target nucleic acid.
  • the first and second effector protein are identical. In some embodiments, the first and second effector protein are not identical.
  • editing a target nucleic acid comprises genome editing.
  • Genome editing may comprise editing a genome, chromosome, plasmid, or other genetic material of a cell or organism.
  • the genome, chromosome, plasmid, or other genetic material of the cell or organism is modified in vivo.
  • the genome, chromosome, plasmid, or other genetic material of the cell or organism is modified in a cell.
  • the genome, chromosome, plasmid, or other genetic material of the cell or organism is modified in vitro.
  • a plasmid may be edited in vitro using a composition described herein and introduced into a cell or organism.
  • editing a target nucleic acid may comprise deleting a sequence from a target nucleic acid.
  • a mutated sequence or a sequence associated with a disease may be removed from a target nucleic acid.
  • editing a target nucleic acid may comprise replacing a sequence in a target nucleic acid with a second sequence.
  • a mutated sequence or a sequence associated with a disease may be replaced with a second sequence lacking the mutation or that is not associated with the disease.
  • editing a target nucleic acid may comprise deleting or replacing a sequence comprising markers associated with a disease or disorder.
  • editing a target nucleic acid may comprise introducing a sequence into a target nucleic acid.
  • a beneficial sequence or a sequence that may reduce or eliminate a disease may be inserted into the target nucleic acid.
  • methods comprise inserting a donor nucleic acid into a cleaved target nucleic acid.
  • the donor nucleic acid may be inserted at a specified (e.g., effector protein targeted) point within the target nucleic acid.
  • the cleaved target nucleic acid is cleaved at a single location.
  • the methods comprise contacting a target nucleic acid with an effector protein described herein, thereby introducing a singlestranded break in the target nucleic acid; and contacting the target nucleic acid with a donor nucleic acid for homologous recombination, optionally by HDR or NHEJ, thereby introducing a new sequence into the target nucleic acid (e.g., at a cleavage site).
  • the cleaved target nucleic acid is cleaved at two locations.
  • the methods comprise contacting a target nucleic acid with an effector protein described herein, thereby introducing a single-stranded break in the target nucleic acid; contacting the target nucleic acid with a second effector protein described herein, to generate a second cleavage site in the target nucleic acid, ligating the regions flanking the first and second cleavage site, optionally through NHEJ or single-strand annealing, thereby resulting in the excision of a portion of the target nucleic acid between the first and second cleavage sites from the target nucleic acid; and contacting the target nucleic acid with a donor nucleic acid for homologous recombination, optionally by HDR or NHEJ, thereby introducing a new sequence into the target nucleic acid (e.g., in between two cleavage sites).
  • methods comprise editing a target nucleic acid with two or more effector proteins.
  • Editing a target nucleic acid may comprise introducing a two or more singlestranded breaks in a target nucleic acid.
  • a break may be introduced by contacting a target nucleic acid with an effector protein and a guide nucleic acid.
  • the guide nucleic acid may bind to the effector protein and hybridize to a region of the target nucleic acid, thereby recruiting the effector protein to the region of the target nucleic acid.
  • binding of the effector protein to the guide nucleic acid and the region of the target nucleic acid may activate the effector protein, and the effector protein may introduce a break (e.g., a single stranded break) in the region of the target nucleic acid.
  • editing a target nucleic acid may comprise introducing a first break in a first region of the target nucleic acid and a second break in a second region of the target nucleic acid.
  • editing a target nucleic acid may comprise contacting a target nucleic acid with a first guide nucleic acid that binds to a first effector protein and hybridizes to a first region of the target nucleic acid and a second guide nucleic acid that binds to a second programmable nickase and hybridizes to a second region of the target nucleic acid.
  • the first effector protein may introduce a first break in a first strand at the first region of the target nucleic acid
  • the second effector protein may introduce a second break in a second strand at the second region of the target nucleic acid.
  • a segment of the target nucleic acid between the first break and the second break may be removed, thereby editing the target nucleic acid.
  • a segment of the target nucleic acid between the first break and the second break may be replaced (e.g., with donor nucleic acid), thereby editing the target nucleic acid.
  • Methods, systems and compositions described herein may edit a target nucleic acid wherein such editing may effect one or more indels.
  • the impact on the transcription and/or translation of the target nucleic acid may be predicted depending on: 1) the amount of indels generated; and 2) the location of the indel on the target nucleic acid.
  • the edit or mutation may be a frameshift mutation.
  • a frameshift mutation may not be effected, but a splicing disruption mutation and/or sequence skip mutation may be effected, such as an exon skip mutation. In some embodiments, if the amount of indels is not evenly divisible by three, then a frameshift mutation may be affected.
  • Methods, systems and compositions described herein may edit a target nucleic acid wherein such editing may be measured by indel activity.
  • Indel activity measures the amount of change in a target nucleic acid (e.g., nucleotide deletion(s) and/or insertion(s)) compared to a target nucleic acid that has not been contacted by a polypeptide described in compositions, systems, and methods described herein.
  • indel activity may be detected by next generation sequencing of one or more target loci of a target nucleic acid where indel percentage is calculated as the fraction of sequencing reads containing insertions or deletions relative to an unedited reference sequence.
  • methods, systems, and compositions comprising an effector protein and guide nucleic acid described herein may exhibit about 0.0001% to about 65% or more indel activity upon contact to a target nucleic acid compared to a target nucleic acid non-contacted with compositions, systems, or by methods described herein.
  • methods, systems, and compositions comprising an effector protein and guide nucleic acid described herein may exhibit about 0.0001%, about 0.001%, about 0.01%, about 0.1%, about 1%, about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65% or more indel activity.
  • editing of a target nucleic acid as described herein effects one or more mutations comprising splicing disruption mutations, frameshift mutations (e.g., 1+ or 2+ frameshift mutation), sequence deletion, sequence skipping, sequence reframing, sequence knock-in, or any combination thereof.
  • the splicing disruption can be an editing that disrupts a splicing of a target nucleic acid or a splicing of a sequence that is transcribed from a target nucleic acid relative to a target nucleic acid without the splicing disruption.
  • the frameshift mutation can be an editing that alters the reading frame of a target nucleic acid relative to a target nucleic acid without the frameshift mutation.
  • the frameshift mutation can be a +2 frameshift mutation, wherein a reading frame is edited by 2 bases.
  • the frameshift mutation can be a +1 frameshift mutation, wherein a reading frame is edited by 1 base.
  • the frameshift mutation is an editing that alters the number of bases in a target nucleic acid so that it is not divisible by three.
  • the frameshift mutation can be an editing that is not a splicing disruption.
  • a sequence as described in reference to the sequence deletion, sequence skipping, sequence reframing, and sequence knock-in can be a DNA sequence, a RNA sequence, an edited DNA or RNA sequence, a mutated sequence, a wild-type sequence, a coding sequence, a non-coding sequence, an exonic sequence (exon), an intronic sequence (intron), or any combination thereof.
  • the sequence deletion is an editing where one or more sequences in a target nucleic acid are deleted relative to a target nucleic acid without the sequence deletion.
  • the sequence deletion can result in or effect a splicing disruption or a frameshift mutation.
  • the sequence deletion result in or effect a splicing disruption.
  • the sequence skipping is an editing where one or more sequences in a target nucleic acid are skipped upon transcription or translation of the target nucleic acid relative to a target nucleic acid without the sequence skipping.
  • the sequence skipping can result in or effect a splicing disruption or a frameshift mutation.
  • the sequence skipping can result in or effect a splicing disruption.
  • the sequence reframing is an editing where one or more bases in a target are edited so that the reading frame of the sequence is reframed relative to a target nucleic acid without the sequence reframing.
  • the sequence reframing can result in or effect a splicing disruption or a frameshift mutation.
  • the sequence reframing can result in or effect a frameshift mutation.
  • the sequence knock-in is an editing where one or more sequences is inserted into a target nucleic acid relative to a target nucleic acid without the sequence knock-in.
  • the sequence knock-in can result in or effect a splicing disruption or a frameshift mutation.
  • the sequence knock-in can result in or effect a splicing disruption.
  • editing of a target nucleic acid can be locus specific, wherein compositions, systems, and methods described herein can edit a target nucleic acid at one or more specific loci to effect one or more specific mutations comprising splicing disruption mutations, frameshift mutations, sequence deletion, sequence skipping, sequence reframing, sequence knock-in, or any combination thereof.
  • editing of a specific locus can affect any one of a splicing disruption, frameshift (e.g., 1+ or 2+ frameshift), sequence deletion, sequence skipping, sequence reframing, sequence knock-in, or any combination thereof.
  • editing of a target nucleic acid can be locus specific, modification specific, or both.
  • editing of a target nucleic acid can be locus specific, modification specific, or both, wherein compositions, systems, and methods described herein comprise an effector protein described herein and a guide nucleic acid described herein.
  • Methods of editing a target nucleic acid or modulating the expression of a target nucleic acid may be performed in vivo. Methods of editing a target nucleic acid or modulating the expression of a target nucleic acid may be performed in vitro. For example, a plasmid may be edited in vitro using a composition described herein and introduced into a cell or organism. Methods of editing a target nucleic acid or modulating the expression of a target nucleic acid may be performed ex vivo. For example, methods may comprise obtaining a cell from a subject, editing a target nucleic acid in the cell with methods described herein, and returning the cell to the subject.
  • methods of modifying described herein comprise contacting a target nucleic acid with one or more components, compositions or systems described herein.
  • the one or more components, compositions or systems described herein comprise at least one of: a) one or more effector proteins, or one or more nucleic acids encoding one or more effector proteins; and b) one or more guide nucleic acids, or one or more nucleic acids encoding one or more guide nucleic acids.
  • the one or more effector proteins introduce a single-stranded break or a double-stranded break in the target nucleic acid.
  • methods of modifying described herein comprise using one or more guide nucleic acids or uses thereof, wherein the methods modify a target nucleic acid at a single location.
  • the methods comprise contacting an RNP comprising an effector protein and a guide nucleic acid to the target nucleic acid.
  • the methods introduce a mutation (e.g., point mutations, deletions) in the target nucleic acid relative to a corresponding wildtype nucleotide sequence.
  • the methods remove or correct a disease-causing mutation in a nucleic acid sequence to produce a corresponding wildtype nucleotide sequence.
  • the methods remove/correct point mutations, deletions, null mutations, or tissue-specific mutations in a target nucleic acid.
  • the methods introduce a single stranded cleavage, a nick, a deletion of one or two nucleotides, an insertion of one or two nucleotides, a substitution of one or two nucleotides, an epigenetic modification (e.g., methylation, demethylation, acetylation, or deacetylation), or a combination thereof to the target nucleic acid.
  • the methods comprise using an effector protein and two guide nucleic acids, wherein two RNPs cleave the target nucleic acid at the same location, wherein a first RNP comprises the effector protein and a first guide nucleic acid, and wherein a second RNP comprises the effector protein and a second guide nucleic acid.
  • methods comprising using two effector protein and two guide nucleic acids, wherein both RNPs cleave the target nucleic acid at the same location, wherein a first RNP comprises a first effector protein and a first target nucleic acid, and wherein a second RNP comprises a second effector protein and a second target nucleic acid.
  • methods of modifying described herein comprise using one or more guide nucleic acids or uses thereof, wherein the methods modify a target nucleic acid at two different locations.
  • the methods introduce two cleavage sites in the target nucleic acid, wherein a first cleavage site and a second cleavage site comprise one or more nucleotides therebetween.
  • the methods cause deletion of the one or more nucleotides.
  • the deletion restores a wild-type reading frame.
  • the wild-type reading frame produces at least a partially functional protein.
  • the deletion causes a non-wild-type reading frame.
  • a non-wild-type reading frame produces a partially functional protein or nonfunctional protein.
  • the at least partially functional protein has at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%, at least 110%, at least 120%, at least 130%, at least 140%, at least 150%, at least 180%, at least 200%, at least 300%, at least 400% activity compared to a corresponding wildtype protein.
  • the methods comprise using an effector protein and two guide nucleic acids, wherein two RNPs cleave the target nucleic acid at different locations, wherein a first RNP comprises the effector protein and a first guide nucleic acid, and wherein a second RNP comprises the effector protein and a second guide nucleic acid.
  • methods comprising using two effector protein and two guide nucleic acids, wherein both RNPs cleave the target nucleic acid at the same location, wherein a first RNP comprises a first effector protein and a first target nucleic acid, and wherein a second RNP comprises a second effector protein and a second target nucleic acid.
  • methods of editing described herein comprise inserting a donor nucleic acid into a cleaved target nucleic acid.
  • the cleaved target nucleic acid formed by introducing a single-stranded break into a target nucleic acid.
  • the donor nucleic acid may be inserted at a specified (e.g., effector protein targeted) point within the target nucleic acid.
  • the cleaved target nucleic acid is cleaved at a single location.
  • the methods comprise contacting a target nucleic acid with an effector protein described herein, thereby introducing a single-stranded break in the target nucleic acid; and contacting the target nucleic acid with a donor nucleic acid for homologous recombination, optionally by HDR or NHEJ, thereby introducing a new sequence into the target nucleic acid (e.g., at a cleavage site).
  • the cleaved target nucleic acid is cleaved at two locations.
  • the methods comprise contacting a target nucleic acid with an effector protein described herein, thereby introducing a single-stranded break in the target nucleic acid; contacting the target nucleic acid with a second effector protein described herein, to generate a second cleavage site in the target nucleic acid, ligating the regions flanking the first and second cleavage site, optionally through NHEJ or single-strand annealing, thereby resulting in the excision of a portion of the target nucleic acid between the first and second cleavage sites from the target nucleic acid; and contacting the target nucleic acid with a donor nucleic acid for homologous recombination, optionally by HDR or NHEJ, thereby introducing a new sequence into the target nucleic acid (e.g., in between two cleavage sites).
  • methods comprise contacting a target nucleic acid with a donor nucleic acid.
  • composition described herein comprise a donor nucleic acid.
  • a donor nucleic acid comprises a sequence that is derived from a plant, bacteria, fungi, virus, or an animal.
  • the animal is a non-human animal, such as, by way of non-limiting example, a mouse, rat, hamster, rabbit, pig, bovine, deer, sheep, goat, chicken, cat, dog, ferret, a bird, non-human primate (e.g., marmoset, rhesus monkey).
  • the non-human animal is a domesticated mammal or an agricultural mammal.
  • the animal is a human.
  • the sequence comprises a human wild-type (WT) gene or a portion thereof.
  • the donor nucleic acid comprises single-stranded DNA or linear double-stranded DNA.
  • the donor nucleic acid comprises a nucleotide sequence encoding a functional polypeptide and/or wherein the donor nucleic acid comprises a wildtype sequence.
  • the donor nucleic acid comprises a protein coding sequence, a gene, a gene fragment, an exon, an intron, an exon fragment, an intron fragment, a gene regulatory fragment, a gene regulatory region fragment, coding sequences thereof, or combinations thereof.
  • the donor nucleic acid comprises a naturally occurring sequence. In some embodiments, the naturally occurring sequence does not contain a mutation.
  • the donor nucleic acid comprises a gene fragment, an exon fragment, an intron fragment, a gene regulatory region fragment, or combinations thereof.
  • the fragment is at least 10, at least 20, at least 30, at least 40, at least 50, at least 60, at least 70, or at least 80 contiguous nucleotides.
  • a donor nucleic acid of any suitable size is integrated into a target nucleic acid or a genome.
  • the donor nucleic acid integrated into the target nucleic acid or the genome is less than 3, about 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, 9, 9.5, 10, 10.5, 11, 11.5, 12, 12.5, 13, 13.5, 14, 14.5, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, 100, 150, 200, 250, 300, 350, 400, 450, 500 kilobases in length.
  • the donor nucleic acid is more than 500 kilobases (kb) in length.
  • Methods may comprise contacting a target nucleic acid, including but not limited to a cell comprising the target nucleic acid, with such compositions.
  • the donor nucleic acid is inserted at a site that has been cleaved by a composition disclosed herein, for example, an effector protein, resulting in a nick or double strand break.
  • the term donor nucleic acid refers to a sequence of nucleotides that will be or has been introduced into a cell following transfection of the viral vector.
  • the donor nucleic acid may be introduced into the cell by any mechanism of the transfecting viral vector, including, but not limited to, integration into the genome of the cell or introduction of an episomal plasmid or viral genome.
  • an effector protein as described herein facilitates insertion of a donor nucleic acid at a site of cleavage or between two cleavage sites by cleaving (hydrolysis of a phosphodiester bond) of a nucleic acid resulting in a nick or double strand break - nuclease activity.
  • the donor nucleic acid comprises a sequence that serves as a template in the process of homologous recombination. The sequence may carry one or more nucleobase modifications that are to be introduced into the target nucleic acid. By using this donor nucleic acid as a template, the genetic information, including the modification(s), is copied into the target nucleic acid by way of homologous recombination.
  • the cell may be a eukaryotic cell (e.g., a mammalian cell) or a prokaryotic cell (e.g., an archaeal cell).
  • the cell may be derived from a multicellular organism and cultured as a unicellular entity.
  • the cell may comprise a heritable genetic modification, such that progeny cells derived therefrom comprise the heritable genetic mutation.
  • the cell may be progeny of a genetically modified cell comprising a genetic modification of the genetically modified parent cell.
  • a genetically modified cell may comprise a deletion, insertion, mutation, or non-native sequence relative to a wild-type version of the cell or the organism from which the cell was derived.
  • modified cells or populations of modified cells wherein the modified cell comprises an effector protein described herein, a nucleic acid encoding an effector protein described herein, or a combination thereof.
  • the modified cell comprises a fusion protein described herein, a nucleic acid encoding an effector protein described herein, or a combination thereof.
  • the modified cell is a modified prokaryotic cell.
  • the modified cell is a modified eukaryotic cell.
  • a modified cell may be a modified fungal cell.
  • the modified cell is a modified vertebrate cell.
  • the modified cell is a modified invertebrate cell.
  • the modified cell is a modified mammalian cell. In some embodiments, the modified cell is a modified human cell. In some embodiments, the modified cell is in a subject.
  • a modified cell may be in vitro.
  • a modified cell may be in vivo.
  • a modified cell may be ex vivo.
  • a modified cell may be a cell in a cell culture.
  • a modified cell may be a cell obtained from a biological fluid, organ, or tissue of a subject and modified with a composition and/or method described herein. Non-limiting examples of biological fluids are blood, plasma, serum, and cerebrospinal fluid.
  • Non-limiting examples of tissues and organs are bone marrow, adipose tissue, skeletal muscle, smooth muscle, spleen, thymus, brain, lymph node, adrenal gland, prostate gland, intestine, colon, liver, kidney, pancreas, heart, lung, bladder, ovary, uterus, breast, and testes.
  • Non-limiting examples of cells that may be obtained from a subject are hepatocytes, epithelial cells, endothelial cells, neurons, cardiomyocytes, muscle cells and adipocytes.
  • a cell may be in vitro.
  • a cell may be in vivo.
  • a cell may be ex vivo.
  • a cell may be an isolated cell.
  • a cell may be a cell inside of an organism.
  • a cell may be an organism.
  • a cell may be a cell in a cell culture.
  • a cell may be one of a collection of cells.
  • a cell may be a mammalian cell or derived from a mammalian cell.
  • a cell may be a rodent cell or derived from a rodent cell.
  • a cell may be a human cell or derived from a human cell.
  • a cell may be a prokaryotic cell or derived from a prokaryotic cell.
  • a cell may be a bacterial cell or may be derived from a bacterial cell.
  • a cell may be an archaeal cell or derived from an archaeal cell.
  • a cell may be a eukaryotic cell or derived from a eukaryotic cell.
  • a cell may be a plant cell or derived from a plant cell.
  • a cell may be an animal cell or derived from an animal cell.
  • a cell may be an invertebrate cell or derived from an invertebrate cell.
  • a cell may be a vertebrate cell or derived from a vertebrate cell.
  • a cell may be a microbe cell or derived from a microbe cell.
  • a cell may be a fungi cell or derived from a fungi cell.
  • a cell may be from a specific organ or tissue.
  • the cell is a progenitor cell or derived therefrom.
  • the cell is a pluripotent stem cell or derived therefrom.
  • the cell is from a specific organ or tissue.
  • the cell is a hepatocyte.
  • the tissue is a subject’s blood, bone marrow, or cord blood.
  • the tissue is a heterologous donor blood, cord blood, or bone marrow.
  • the tissue is an allogenic blood, cord blood, or bone marrow.
  • the tissue may be muscle.
  • the muscle may be a skeletal muscle.
  • Non-limiting examples of cells that may be engineered or modified with compositions and methods described herein include stem cells, such as human stem cells, animal stem cells, stem cells that are not derived from human embryonic stem cells, embryonic stem cells, mesenchymal stem cells, pluripotent stem cells, induced pluripotent stem cells (iPS), somatic stem cells, adult stem cells, hematopoietic stem cells, tissue-specific stem cells.
  • stem cells such as human stem cells, animal stem cells, stem cells that are not derived from human embryonic stem cells, embryonic stem cells, mesenchymal stem cells, pluripotent stem cells, induced pluripotent stem cells (iPS), somatic stem cells, adult stem cells, hematopoietic stem cells, tissue-specific stem cells.
  • a cell may be a pluripotent cell.
  • Non-limiting examples of cells that may be engineered or modified with compositions and methods described herein include immune cells, such as CART, T-cells, B-cells, NK cells, granulocytes, basophils, eosinophils, neutrophils, mast cells, monocytes, macrophages, dendritic cells, antigen-presenting cells (APC), or adaptive cells.
  • immune cells such as CART, T-cells, B-cells, NK cells, granulocytes, basophils, eosinophils, neutrophils, mast cells, monocytes, macrophages, dendritic cells, antigen-presenting cells (APC), or adaptive cells.
  • Non-limiting examples of cells that may be engineered or modified with compositions and methods described herein include plant cells, such as parenchyma, sclerenchyma, collenchyma, xylem, phloem, germline (e.g., pollen). Cells from lycophytes, ferns, gymnosperms, angiosperms, bryophytes, charophytes, chiorophytes, rhodophytes, or glaucophytes.
  • compositions of the disclosure may be administered to a subject.
  • a subject may be a human.
  • a subject may be a mammal (e.g., rat, mouse, cow, dog, pig, sheep, horse).
  • a subject may be a vertebrate or an invertebrate.
  • a subject may be a laboratory animal.
  • a subject may be a patient.
  • a subject may be suffering from a disease.
  • a subject may display symptoms of a disease.
  • a subject may at risk of developing a disease.
  • a subject may not display symptoms of a disease, but still have a disease.
  • a subject may be under medical care of a caregiver (e.g., the subject is hospitalized and is treated by a physician).
  • the subject may have a mutation associated with a gene described herein. In some embodiments, the subject may display symptoms associated with a mutation of a gene described herein. Methods of the disclosure may be performed in a plant, bacteria, or a fungus.
  • compositions and methods of the disclosure may be used for agricultural engineering.
  • compositions and methods of the disclosure may be used to confer desired traits on a plant.
  • a plant may be engineered for the desired physiological and agronomic characteristic using the present disclosure.
  • the target nucleic acid sequence comprises a nucleic acid sequence of a plant.
  • the target nucleic acid sequence comprises a genomic nucleic acid sequence of a plant cell.
  • the target nucleic acid sequence comprises a nucleic acid sequence of an organelle of a plant cell.
  • the target nucleic acid sequence comprises a nucleic acid sequence of a chloroplast of a plant cell.
  • the plant may be a dicotyledonous plant.
  • orders of dicotyledonous plants include Magniolales, Illiciales, Laurales, Piperales, Aristochiales, Nymphaeales, Ranunculales, Papeverales, Sarraceniaceae, Trochodendrales, Hamamelidales, Eucomiales, Leitneriales, Myricales, Fagales, Casuarinales, Caryophyllales, Batales, Polygonales, Plumbaginales, Dilleniales, Theales, Malvales, Urticales, Lecythidales, Violates, Salicales, Capparales, Ericales, Diapensales, Ebenales, Primulales, Rosales, Fabales, Podostemales, Haloragales, Myrtales, Comales, Proteales, San tales, Rafftesiales, Celastrales, Euphorbiales, Rhamnales, Sapindales, Juglandales, Gerania
  • the plant may be a monocotyledonous plant.
  • orders of monocotyledonous plants include Alismatales, Hydrocharitales, Najadales, Triuridales, Commelinales, Eriocaulales, Restionales, Poales, Juncales, Cyperales, Typhales, Bromeliales, Zingiberales, Arecales, Cyclanthales, Pandanales, Arales, Lilliales, and Orchid ales.
  • a plant may belong to the order, for example, Gymnospermae, Pinales, Ginkgoales, Cycadales, Araucariales, Cupressales and Gnetales.
  • Non-limiting examples of plants include plant crops, fruits, vegetables, grains, soy bean, corn, maize, wheat, seeds, tomatoes, rice, cassava, sugarcane, pumpkin, hay, potatoes, cotton, cannabis, tobacco, flowering plants, conifers, gymnosperms, ferns, clubmosses, hornworts, liverworts, mosses, wheat, maize, rice, millet, barley, tomato, apple, pear, strawberry, orange, acacia, carrot, potato, sugar beets, yam, lettuce, spinach, sunflower, rape seed, Arabidopsis, alfalfa, amaranth, apple, apricot, artichoke, ash tree, asparagus, avocado, banana, barley, beans, beet, birch, beech, blackberry, blueberry, broccoli, Brussel's sprouts, cabbage, canola, cantaloupe, carrot, cassava, cauliflower, cedar, a cereal, celery, chestnut, cherry, Chinese cabbage
  • effector proteins and guide RNAs are screened by an in vitro enrichment (IVE) assay to determine TAM recognition by each effector protein-guide RNA complex. Briefly, effector proteins are complexed with corresponding guide RNAs for 15 minutes at 37 °C. The complexes are added to an IVE reaction mix.
  • IVE in vitro enrichment
  • TAM screening reactions use 10 pl of RNP in 100 pl reactions with 1,000 ng of a plasmid library containing a randomized TAM sequence (5’-NNNNN-3’, where N is any of A, C, G, T) in lx Cutsmart buffer and are carried out for 15 minutes at 25 °C, 45 minutes at 37 °C, and 15 minutes at 45 °C. Reactions are terminated with 1 pl of proteinase K and 5 pl of 500 mM EDTA for 30 minutes at 37 °C. Any target plasmid that is successfully cleaved has an adapter ligated to the cut end, enabling PCR amplification.
  • Amplification products revealed by gel electrophoresis indicate effector protein-sgRNA complexes that were capable of cz.s-cleavage.
  • An EcoRI site is included near the spacer and is used as a positive control.
  • Next generation sequencing (NGS) is performed on cut sequences to identify enriched TAMs.
  • Effector proteins are tested for their ability to produce indels in a mammalian cell line (e.g., HEK293T cells). Briefly, a plasmid encoding the effector proteins and a guide RNA are delivered by lipofection to the mammalian cells. This is performed with a variety of guide RNAs targeting several loci adjacent to biochemically determined TAM sequences. Indels in the loci are detected by next generation sequencing of PCR amplicons at the targeted loci and indel percentage is calculated as the fraction of sequencing reads containing insertions or deletions relative to an unedited reference sequence. “No plasmid” and Cas9 are included as negative and positive controls, respectively.
  • a mammalian cell line e.g., HEK293T cells.
  • a nucleic acid vector encoding a fusion protein is constructed for base editing.
  • the fusion protein comprises a catalytically inactive variant of an effector protein fused to a deaminase.
  • the fusion protein and at least one guide nucleic acid is tested for its ability to edit a target sequence in eukaryotic cells.
  • Cells are transfected with the nucleic acid vector and guide nucleic acid. After sufficient incubation, DNA is extracted from the transfected cells.
  • Target sequences are PCR amplified and sequenced by NGS and MiSeq. The presence of base modifications are analyzed from sequencing data. Results are recorded as a change in % base call relative to the negative control.
  • a single stranded reporter nucleic acid encoding a fluorescent protein (e.g., enhanced green fluorescent protein (EGFP)) and a eukaryotic promoter is generated with a target sequence that is known to be recognized by complexes of effector proteins disclosed herein and corresponding guide nucleic acids.
  • a nucleic acid vector encoding the Cas effector fused to a transcriptional activator; a guide nucleic acid; and the single stranded reporter nucleic acid encoding EGFP are introduced to eukaryotic cells via lipofection and EGFP expression is quantified by flow cytometry. Relative amounts of RNA, indicative of relative gene expression, are quantified with RT-qPCR.
  • a single stranded reporter nucleic acid encoding a fluorescent protein (e.g., enhanced green fluorescent protein (EGFP)) and a pSV40 promoter that drives constitutive expression of EGFP is generated with a target sequence that is known to be recognized by complexes of effector proteins disclosed herein and corresponding guide nucleic acids.
  • a nucleic acid vector encoding the Cas effector fused to a transcriptional repressor (e.g., DNMT); a guide nucleic acid; and the single stranded reporter nucleic acid encoding EGFP are introduced to eukaryotic cells via lipofection and EGFP expression is quantified by flow cytometry. Relative amounts of RNA, indicative of relative gene expression, are quantified with RT-qPCR.
  • any one of the effector proteins of TABLE 1 to perform enhanced gene editing of a target nucleic acid is assayed as follows.
  • Components of the system include: an effector protein; a reverse transcriptase optionally fused to a MS2 coat protein; a first guide nucleic acid; a second guide nucleic acid; and a template RNA (retRNA) comprising an RT template, a primer binding sequence, and optionally an MS2 aptamer.
  • retRNA template RNA
  • the effector protein may be fused to the reverse transcriptase.
  • the effector protein comprises at least one amino acid substitution relative to the respective sequence of TABLE 1, wherein the amino acid substitution is located in a RuvC domain and/or HNH domain of the effector protein, and wherein the amino acid substitution reduces the catalytic activity of the domain, thereby providing the effector protein with nickase activity.
  • the two guide nucleic acids are designed to bind opposite strands, a target strand and a non-target strand, of the target nucleic acid, wherein the second site is downstream to the first site.
  • HEK293T cells are transiently transfected with these components. This will generate nicking of the target strand at a first site and the non-target strand at a second site downstream of the first site.
  • NGS sequencing is used for assessing gene editing of a target nucleic acid with this system.
  • a system will advantageously enhance editing signal relative to a system that uses only one guide nucleic acid because in resolving whether to retain the edited or unedited strand of the target nucleic acid, a nick on the unedited strand signals to the cell’s repair system that it's damaged and therefore leads to preferential removal.
  • Example 7 DIS Dual systems obtain precise edit levels comparable to Cas9 RT editing
  • DIS dual H247A
  • SEQ ID NO: 855 An exemplary effector protein, referred to as DIS dual (H247A) (SEQ ID NO: 855), was tested in a precision editing context against Cas9.
  • DIS dual H247A
  • H247A may be characterized as an engineered IscB protein.
  • the first plasmid encoded the nuclease (SpCas9 (H840A) or DIS dual (H247A)) and a guide nucleic acid with an MS2 aptamer.
  • the second plasmid encoded a reverse transcriptase (MMLV or TIF1 fused to MS2 coat protein) and a retRNA.
  • the third plasmid encoded a dual fluorescent reporter (CRISPR+ or DIS Dual editing).
  • CRISPR+ a stop codon is edited to allow mRhubarb expression.
  • DIS Dual editing assay 2 nt insertion is needed to reframe mRhubarb and reframe a TGA codon that needs to be corrected to allow the reporter expression.
  • Cells were incubated for 48 hours as fluorescence read by fluorescence spectroscopy. As shown in FIGS. 4 A and 4B, these two different reporters confirmed DI S dual (H247A) obtains similar levels of precision editing as a Cas9 RT editing system. Future experiments will be performed similarly with the effector protein covalently linked to a reverse transcriptase.
  • Effector protein 3107961 (SEQ ID NO: 856) and a guide RNA having a handle sequence of SEQ ID NO: 859 were screened by an in vitro enrichment (IVE) assay to determine TAM recognition by each effector protein-guide RNA complex. Briefly, effector protein 3107961 was complexed with corresponding guide RNAs for 15 minutes at 37°C. The complexes were added to an IVE reaction mix. IVE reactions were carried out in lx Cutsmart® buffer (New England Biolabs), using 10 pl of RNP in 100 pl reactions with 1,000 ng of a plasmid library containing a 7N TAM sequence 5’ of the target (protospacer) sequence.
  • IVE in vitro enrichment
  • Plasmid transfections in HEK293T cells were carried out as follows. 75ng of effector protein expressing plasmid (3745646 (SEQ ID NO: 624), 3756102 (SEQ ID NO: 713), 3782262 (SEQ ID NO: 833), 370125 (SEQ ID NO: 691), and 3778464 (SEQ ID NO: 774)) and 75ng of guide expressing plasmids were delivered by lipofection to HEK293T cells, seeded at 10,000 cells in 200 pL, in 96 well plates.
  • effector protein expressing plasmid 3745646 (SEQ ID NO: 624), 3756102 (SEQ ID NO: 713), 3782262 (SEQ ID NO: 833), 370125 (SEQ ID NO: 691), and 3778464 (SEQ ID NO: 774)
  • 75ng of guide expressing plasmids were delivered by lipofection to HEK293T cells, seeded at 10,000 cells in 200
  • TransIT-293 (Minis Bio) reagent was diluted with warmed up OPTIMEM and mixed with the plasmid DNA at the ratio of 2: 1 lipid:DNA. Lipid:DNA mixture were incubated for 15 minutes at room temperature before adding 20 pL of the lipid:DNA optimem mixture to each well. Cells were incubated for 3 days before being lysed and subjected to PCR amplification. Indels were detected by next generation sequencing of PCR amplicons at the targeted loci and indel percentage was calculated as the fraction of sequencing reads containing insertions or deletions relative to an unedited reference sequence. Results are shown in TABLE 10.
  • Engineered effector proteins were evaluated for their ability to induce indels in HEK293T cells at a target locus.
  • HEK293T cells seeded at 30k cells per well, were cotransfected with 150ng of vector expressing an engineered effector protein and 150ng of the guide nucleic acid expressing plasmid with TransIT-293.
  • the engineered effector proteins are represented by SEQ ID NOS: 179, and 889-910.
  • SEQ ID NOS: 889-910 are engineered variants of SEQ ID NO: 275, having multiple amino acid substitutions relative to SEQ ID NO: 275, as described in TABLE 8.
  • the guide nucleic acid was comprised of a spacer sequence having the sequence of 5’- AAUGGUGGAAACACAG-3’ (SEQ ID NO: 911) and a protein binding sequence of SEQ ID NO: 858, wherein the spacer sequence was located 5’ of the protein binding sequence.
  • the cells were lysed using QuickExtractTM DNA Extraction Solution.
  • Targeted loci were amplified using AccuPrime Taq polymerase and sequenced by next generation sequencing. The indel percentage was calculated as the fraction of sequencing reads containing insertions or deletions relative to an unedited reference sequence. Results are shown in FIG. 6 (SEQ ID NO: 179) and FIG. 7 (SEQ ID NOS: 889- 910)
  • HEK293T cells were transfected with plasmids encoding an engineered variant of effector protein 3745646 (SEQ ID NO: 624), MMLV-RT, a guide nucleic acid, and a retRNA to introduce precise edits in the human gene, B2M.
  • the engineered variant of effector protein 3745646 has an amino acid substitution of H246A relative to SEQ ID NO: 624. Without being bound by theory, the H246A amino acid substitution is believed to be located in an HNH domain of effector protein 3745646, and thereby provides the engineered variant with nickase activity. Nucleotide sequences encoding these gene editing components are provided in TABLE 11.
  • Precise editing as well as unintended byproduct edits (indels) were quantified using amplicon NGS.
  • Results are provided in FIG. 8A.
  • the engineered variant of effector protein 3745646 achieved 0.8% precise edits.
  • This experiment was repeated twice (Exp 1 and Exp 2) in both the absence (Control) and presence (Enhancer) of a hHMLHl variant, amino acid sequence is provided in TABLE 11. Without being bound by theory, the hHMLHl variant is considered an inhibitor of DNA mismatch repair (MMR).
  • MMR DNA mismatch repair
  • the hHMLHl variant increased precise edits from 0.8% to 2%.
  • Results are provided in FIG. 8B.
  • hHMLHl may be considered an enhancer of precise editing systems employing engineered variants of effector protein 3745646 (SEQ ID NO: 624).
  • Data points shown in FIG. 8B come from systems wherein the effector protein was and was not fused to the RT (levels were comparable for both conditions in these experiments).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • Organic Chemistry (AREA)
  • Zoology (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • Physics & Mathematics (AREA)
  • Biophysics (AREA)
  • Plant Pathology (AREA)
  • Medicinal Chemistry (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Mycology (AREA)
  • Immunology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Provided herein are compositions, systems, and methods comprising engineered effector proteins and uses thereof. In general, the engineered effector proteins are capable of forming a complex with a guide RNA that localizes the complex to a nucleic acid of interest. Compositions, systems, and methods of the present disclosure may be useful to modify and detect nucleic acids of interest.

Description

EFFECTOR PROTEINS AND USES THEREOF
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] The present application claims priority to U.S. Provisional Application 63/597,942, filed November 10, 2023; U.S. Provisional Application 63/606,462, filed December 5, 2023; U.S. Provisional Application 63/615,462, filed December 28, 2023; U.S. Provisional Application 63/626,307, filed January 29, 2024; U.S. Provisional Application 63/627,204, filed January 31, 2024; and U.S. Provisional Application 63/569,797, filed March 26, 2024, the contents of each of which are incorporated herein by reference in their entireties.
REFERENCE TO AN ELECTRONIC SEQUENCE LISTING
[0002] The contents of the electronic sequence listing (MABI_049_06WO_SeqList_ST26.xml; Size: 1,123,360 bytes; and Date of Creation: November 6, 2024) are herein incorporated by reference in their entirety.
FIELD
[0003] The present disclosure relates generally to polypeptides, such as effector proteins, compositions of such polypeptides and guide nucleic acids, systems, and methods of using such polypeptides and compositions, including detecting and editing target nucleic acids.
SUMMARY
[0004] The present disclosure provides for polypeptides, such as effector proteins, compositions, systems, and methods comprising the same, and uses thereof. In some instances, effector proteins are engineered. TABLE 1 provides exemplary engineered effector proteins and TABLE 2 shows the parent sequence that was the basis for engineering as well as the engineering strategy corresponding to each effector protein in TABLE 1. TABLE 6 provides exemplary effector proteins that may be engineered. In some embodiments, a domain of an effector protein in TABLE 6 or a portion thereof may be replaced with a sequence described in TABLE 2. TABLE 8 also provides exemplary engineered effector proteins. In some instances, compositions, systems, and methods comprise guide nucleic acids or uses thereof. In some embodiments, a guide nucleic acid comprises a handle sequence provided in TABLE 7. Compositions, systems, and methods disclosed herein may leverage nucleic acid modification activities, such as nucleic acid editing. Nucleic acid editing may comprise: insertion, deletion, substitution, or a combination thereof of one or more nucleotides in a target nucleic acid. Nucleic acid editing may comprise cleavage activity, such as cis cleavage activity, nickase activity, and/or nuclease activity. Modification activities may also include epigenetic modifications of nucleic acids. In some instances, compositions, systems and methods are useful for the treatment of a disease or disorder. The disease or disorder may be associated with a target nucleic acid. The disease or disorder may be associated with one or more mutations in the target nucleic acid.
[0005] In some aspects, the present disclosure provides systems or compositions comprising: an effector protein, or a nucleic acid encoding the effector protein, comprising an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to a sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, the system or composition comprises an engineered guide nucleic acid, or a nucleic acid encoding the guide nucleic acid, wherein the engineered guide nucleic acid comprises a protein binding sequence and a spacer sequence. In some embodiments, the protein binding sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 858. In some embodiments, the protein binding sequence comprises a handle sequence comprising a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to a sequence selected from TABLE 7. In some embodiments, the effector protein comprises an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%. , at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 856, and the handle sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 859 or 860. In some embodiments, the effector protein comprises an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 624, and the handle sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 884. In some embodiments, the effector protein comprises an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 713, and the handle sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 885. In some embodiments, the effector protein comprises an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 833, and the handle sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 886. In some embodiments, the effector protein comprises an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 691, and the handle sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 884. In some embodiments, the effector protein comprises an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 774, and the handle sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 888. In some embodiments, the effector protein comprises an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 179 and 889-910, and the protein binding sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 858. In some embodiments, the spacer sequence is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical, complementary, or reverse complementary to a eukaryotic sequence. In some embodiments, the spacer sequence is linked to a 5’ end of the protein binding sequence. In some embodiments, the effector protein comprises about 100, about 120, about 140, about 160, about 180, about 200, about 220, about 240, about 260, about 280, about 300, about 320, about 340, about 360, about 380, about 400, about 420, about 440, about 460, about 480, about 500, or about 520 contiguous amino acids of a sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, the length of the effector protein is less than 600, less than 580, less than 560, less than 540, less than 520, less than 500, less than 480, less than 460, less than 440, less than 420, or less than 400 contiguous amino acids; and at least 300, at least 320, at least 340, at least 360, at least 380 contiguous amino acids. In some embodiments, the amino acid sequence is less than 100% identical to the sequence selected from TABLE 1, TABLE 6, and TABLE 8, and wherein not more than 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 amino acid substitutions in the amino acid sequence are non-conservative amino acid substitutions relative to the respective sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, the effector protein comprises at least one amino acid substitution in a RuvC domain, an HNH domain, or a combination thereof. In some embodiments, the effector protein comprises an amino acid sequence that is at least 90%, at least 95%, at least 97%, at least 98%, or at least 99% identical to SEQ ID NO: 624, wherein the effector protein comprises an amino acid substitution of H246A relative to SEQ ID NO: 624 In some embodiments, the effector protein encoded by a nucleic acid sequence that is at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 912. In some embodiments, the effector protein comprises an amino acid sequence that is at least 90%, at least 95%, at least 97%, at least 98%, or at least 99% identical to SEQ ID NO: 713, wherein the effector protein comprises an amino acid substitution of H244A relative to SEQ ID NO: 713 In some embodiments, the effector protein comprises an amino acid sequence that is at least 90%, at least 95%, at least 97%, at least 98%, or at least 99% identical to SEQ ID NO: 856, wherein the effector protein comprises an amino acid substitution of H248A relative to SEQ ID NO: 856. In some embodiments, the amino acid substitution is a non-conservative amino acid substitution. In some embodiments, the amino acid substitution replaces a catalytic residue of the domain(s). In some embodiments, a) the effector protein comprises an amino acid sequence at least 90-100% identical to SEQ ID NO: 856 and recognizes a TAM of ATAANNN when complexed with a guide nucleic acid; b) the effector protein comprises an amino acid sequence at least 90-100% identical to SEQ ID NO: 624 and recognizes a TAM of ARRRGNN when complexed with a guide nucleic acid; c) the effector protein comprises an amino acid sequence at least 90-100% identical to SEQ ID NO: 713 and recognizes a TAM of GNAAANN when complexed with a guide nucleic acid; d) the effector protein comprises an amino acid sequence at least 90-100% identical to SEQ ID NO: 833 and recognizes a TAM of ATAANNN when complexed with a guide nucleic acid; e) the effector protein comprises an amino acid sequence at least 90-100% identical to SEQ ID NO: 691 and recognizes a TAM of GYARRNN when complexed with a guide nucleic acid; or f) the effector protein comprises an amino acid sequence at least 90-100% identical to SEQ ID NO: 774 and recognizes a TAM of RTANNNN when complexed with a guide nucleic acid; wherein N is any nucleotide, Y is C or T, and R is A or G. In some embodiments, the effector protein has nickase activity. In some embodiments, the effector protein is linked to a nuclear localization signal. In some embodiments, the system or composition further comprises a donor nucleic acid. In some embodiments, the system or composition further comprises an effector partner protein linked to the effector protein. In some embodiments, the effector partner comprises a polypeptide selected from a deaminase, a reverse transcriptase, a transcriptional activator, a transcriptional repressor, or a functional domain thereof. In some embodiments, the effector protein comprises at least one mutation that reduces its nuclease activity relative to the effector protein without the at least one mutation as measured in a cleavage assay, optionally wherein the effector protein is a catalytically inactive nuclease. In some embodiments, the system or composition further comprises a lipid nanoparticle containing the effector protein or the nucleic acid encoding the effector protein, the engineered guide nucleic acid, or a combination thereof. In some embodiments, the effector protein or the nucleic acid encoding the effector protein, and the engineered guide nucleic acid or the nucleic acid encoding the guide nucleic acid are provided in separate compositions. In some embodiments, the nucleic acid encoding the effector protein, the nucleic acid encoding the guide nucleic acid, or a combination thereof is an expression vector. In some embodiments, the expression vector is a viral vector, optionally wherein the viral vector is an adeno- associated viral (AAV) vector.
[0006] In some aspects, the present disclosure provides systems or compositions comprising an engineered effector protein, wherein at least one domain of a parent sequence is replaced by one or more corresponding domains from another protein, and wherein the parent sequence is the parent sequence of TABLE 2. In some embodiments, the at least one domain of the parent sequence is selected from an HNH domain, a PLMP domain, a BH domain, and a TID domain. In some embodiments, the corresponding domain from another protein is any one of the sequences selected from TABLE 2. In some embodiments, the system or composition comprises an engineered guide nucleic acid, or a nucleic acid encoding the guide nucleic acid, wherein the engineered guide nucleic acid comprises a protein binding sequence and a spacer sequence. In some embodiments, the protein binding sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 858. In some embodiments, the protein binding sequence comprises a handle sequence comprising a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to a sequence selected from TABLE 7. In some embodiments, the spacer sequence hybridizes to a target sequence that is adjacent to a TAM of 5’-NWRRNA-3’, wherein W is A or T, N is any nucleotide, and R is A or G. In some embodiments, the spacer sequence hybridizes to a target sequence that is adjacent to a TAM of 5’-ATAANNN-3’, wherein N is any nucleotide. [0007] In some aspects, the present disclosure provides pharmaceutical compositions comprising the system or composition described herein, and a pharmaceutically acceptable excipient.
[0008] In some aspects, the present disclosure provides methods of modifying a target nucleic acid, the method comprising contacting the target nucleic acid with the system or composition described herein or the pharmaceutical composition described herein, thereby modifying the target nucleic acid.
[0009] In some aspects, the present disclosure provides cells comprising the composition described herein.
[0010] In some aspects, the present disclosure provides cells modified by the system or composition described herein, the pharmaceutical composition described herein, or the method described herein.
[0011] In some aspects, the present disclosure provides cells comprising a modified target nucleic acid, wherein the modified target nucleic acid is a target nucleic acid modified according to the method described herein.
[0012] In some aspects, the present disclosure provides methods of treating a disease comprising administering to a subject in need thereof any one of: the system or composition described herein; the pharmaceutical composition described herein; or the cell described herein.
[0013] In some aspects, the present disclosure provides systems or compositions comprising: a) an effector protein or a nucleic acid encoding the same, wherein the effector protein comprises an amino acid sequence that is at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, identical to SEQ ID NO: 855; b) a reverse transcriptase or a nucleic acid encoding the same; c) a guide nucleic acid or a nucleic acid encoding the same; and d) a retRNA or a nucleic acid encoding the same. In some embodiments, the reverse transcriptase comprises a peptide that is capable of binding a secondary structure present in at least one of the guide nucleic acid and the retRNA. In some embodiments, the effector protein or nucleic acid encoding the same is covalently linked to the reverse transcriptase or nucleic acid encoding the same. In some embodiments, the system or composition comprises the nucleic acid encoding the effector protein, the nucleic acid encoding the reverse transcriptase, the nucleic acid encoding the guide nucleic acid, and the nucleic acid encoding the retRNA, wherein all of the nucleic acids are present in a single expression vector, optionally wherein the expression vector is an AAV vector. [0014] In some aspects, the present disclosure provides methods of modifying a target nucleic acid, the method comprising contacting the target nucleic acid with the system or composition described herein. In some embodiments, the method comprises contacting a cell with the system or composition.
[0015] In some aspects, the present disclosure provides systems or compositions comprising: a) an IscB protein; b) a reverse transcriptase or a nucleic acid encoding the same; c) a guide nucleic acid or a nucleic acid encoding the same; and d) a retRNA or a nucleic acid encoding the same. In some embodiments, the present disclosure provides methods of editing a target nucleic acid, the method comprising contacting a target nucleic acid with the system or composition described herein.
INCORPORATION BY REFERENCE
[0016] All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference in their entirety for any purpose and to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference.
BRIEF DESCRIPTION OF DRAWINGS
[0017] FIG. 1 depicts the domains, defined by amino acid positions, of the parent sequence that was the basis for engineering.
[0018] FIG. 2 depicts an exemplary target nucleic acid and exemplary guide nucleic acid to be used with effector proteins described herein for the purposes of showing the orientation of the target adjacent motif (TAM) in relationship to where the guide nucleic acid hybridizes to a target nucleic acid. NTS = non-target strand and TS = target strand. A protein binding sequence is not provided, but labeled for the purpose of showing the orientation of the protein binding sequence with respect to the spacer sequence (the spacer sequence is 5’ of the protein binding sequence). For the avoidance of doubt, the figure should not be interpreted to indicate that the protein binding sequence or any portion thereof necessarily binds to the target nucleic acid. Also, for the avoidance of doubt, this figure should not suggest that the NTS and TS are paired when the guide nucleic acid hybridizes to the target nucleic acid.
[0019] FIG. 3 shows an example of a split protein/RNA system for precise editing with an effector (e.g., DIS dual), wherein the retRNA is circularized.
[0020] FIG. 4A shows precision editing levels of DIS dual (H247A) RT editing is comparable to Cas9 RT editing in a CRISPR+ dual fluorescent reporter assay. FIG. 4B shows precision editing levels of DI S dual (H247A) RT editing is comparable to Cas9 RT editing in a DI S dual fluorescent reporter assay. Bars above each RT tested, from left to right, represent SpCas9, SpCas9 (H840A), DIS dual, and DIS dual (H247A).
[0021] FIG. 5 shows the position frequency matrix (PFM) from WebLogos revealing the presence of enriched 5’ TAM consensus sequences for various effector protein-guide RNA complexes.
[0022] FIG. 6 shows that an engineered effector protein represented by SEQ ID NO: 179 (engineered from parental sequence SEQ ID NO: 275) provides increased modifications (indels) in a target nucleic acid in mammalian cells relative to an effector protein represented by SEQ ID NO: 275
[0023] FIG. 7 shows that an engineered effector proteins represented by SEQ ID NOS: 889- 910 (engineered from parental sequence SEQ ID NO: 275) provides increased modifications (indels) in a target nucleic acid in mammalian cells relative to an effector protein represented by SEQ ID NO: 275
[0024] FIG. 8A shows an engineered variant of effector protein 3745646 can produce precise edits in RT editing system. FIG. 8B shows precise editing with an engineered variant of effector protein 3745646 can be enhanced with a DNA mismatch repair inhibitor.
DETAILED DESCRIPTION OF THE INVENTION
[0025] It is to be understood that both the foregoing general description and the following detailed description are exemplary, and explanatory only, and are not restrictive of the disclosure.
[0026] The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described.
[0027] All documents, or portions of documents, cited in this application, including, but not limited to, patents, patent applications, articles, books, and treatises, are hereby expressly incorporated by reference in their entirety for any purpose.
Definitions
[0028] Unless otherwise indicated, all technical terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Unless otherwise indicated or obvious from context, the following terms have the following meanings: [0029] The terms, “a,” “an,” and “the,” as used herein, include plural references unless the context clearly dictates otherwise.
[0030] The terms, “or” and “and/or,” as used herein, include any and all combinations of one or more of the associated listed items. [0031] The terms, “including,” “includes,” “included,” and other forms, are not limiting.
[0032] The terms, “comprise” and its grammatical equivalents, as used herein, specify the presence of stated features, integers, steps, operations, elements, and/or components, but do not preclude the presence or addition of one or more other features, integers, steps, operations, elements, components, and/or groups thereof.
[0033] The term, “about,” as used herein in reference to a number or range of numbers, is understood to mean the stated number and numbers +/- 10% thereof, or 10% below the lower listed limit and 10% above the higher listed limit for the values listed for a range.
[0034] The terms, “% identical,” “% identity,” “percent identity,” and grammatical equivalents thereof, as used herein, in the context of an amino acid sequence or nucleotide sequence, refer to the percent of residues that are identical between respective positions of two sequences when the two sequences are aligned for maximum sequence identity. The % identity is calculated by dividing the total number of the aligned residues by the number of the residues that are identical between the respective positions of the at least two sequences and multiplying by 100. Generally, computer programs can be employed for such calculations. Illustrative programs that compare and align pairs of sequences, include ALIGN (Myers and Miller, Comput Appl Biosci. 1988 Mar;4(l): l 1-7), FASTA (Pearson and Lipman, Proc Natl Acad Sci U S A. 1988 Apr;85(8):2444-8; Pearson, Methods Enzymol. 1990;183:63-98) and gapped BLAST (Altschul et al., Nucleic Acids Res. 1997 Sep l;25(17):3389-40), BLASTP, BLASTN, or GCG (Devereux et al., Nucleic Acids Res. 1984 Jan 11;12(1 Pt l):387-95). For the purposes of calculating % identity and % complementarity, thymine (T) may be considered the same as uracil (U).
[0035] The terms, “% complementary”, “% complementarity”, “percent complementary”, “percent complementarity” and grammatical equivalents thereof, as used interchangeably herein, in the context of two or more nucleic acid molecules, refer to the percent of nucleotides in two nucleotide sequences in said nucleic acid molecules of equal length that can undergo cumulative base pairing at two or more individual corresponding positions in an antiparallel orientation. Accordingly, the terms include nucleic acid sequences that are not completely complementary over their entire length, which indicates that the two or more nucleic acid molecules include one or more mismatches. A “mismatch” is present at any position in the two opposed nucleotides that are not complementary. The % complementary is calculated by dividing the total number of the complementary residues by the total number of the nucleotides in one of the equal length sequences, and multiplying by 100. Complete or total complementarity describes nucleotide sequences in 100% of the residues of a nucleotide sequence are complementary to residues in a reference nucleotide sequence. “Partially complementarity” describes nucleotide sequences in which at least 20%, but less than 100%, of the residues of a nucleotide sequence are complementary to residues in a reference nucleotide sequence. In some instances, at least 50%, but less than 100%, of the residues of a nucleotide sequence are complementary to residues in a reference nucleotide sequence. In some instances, at least 70%, 80%, 90% or 95%, but less than 100%, of the residues of a nucleotide sequence are complementary to residues in a reference nucleotide sequence. “Noncomplementary” describes nucleotide sequences in which less than 20% of the residues of a nucleotide sequence are complementary to residues in a reference nucleotide sequence.
[0036] The term, “percent similarity,” or “% similarity,” as used herein, in the context of an amino acid sequence, refers to a value that is calculated by dividing a similarity score by the length of the alignment. The similarity of two amino acid sequences can be calculated by using a BLOSUM62 similarity matrix (Henikoff and Henikoff, Proc. Natl. Acad. Sci. USA., 89: 10915-10919 (1992)) that is transformed so that any value > 1 is replaced with +1 and any value < 0 is replaced with 0. For example, an He (I) to Leu (L) substitution is scored at +2.0 by the BLOSUM62 similarity matrix, which in the transformed matrix is scored at +1. This transformation allows the calculation of percent similarity, rather than a similarity score. Alternately, when comparing two full protein sequences, the proteins can be aligned using pairwise MUSCLE alignment. Then, the % similarity can be scored at each residue and divided by the length of the alignment. For determining % similarity over a protein domain or motif, a multilevel consensus sequence (or PROSITE motif sequence) can be used to identify how strongly each domain or motif is conserved. In calculating the similarity of a domain or motif, the second and third levels of the multilevel sequence are treated as equivalent to the top level. Additionally, if a substitution could be treated as conservative with any of the amino acids in that position of the multilevel consensus sequence, +1 point is assigned. For example, given the multilevel consensus sequence: RLG and YCK, the test sequence QIQ would receive three points. This is because in the transformed BLOSUM62 matrix, each combination is scored as: Q-R: +1; Q-Y: +0; I-L: +1; I-C: +0; Q-G: +0; Q-K: +1. For each position, the highest score is used when calculating similarity. The % similarity can also be calculated using commercially available programs, such as the Geneious Prime software given the parameters matrix = BLOSUM62 and threshold > 1.
[0037] The terms, “bind,” “binding,” “interact” and “interacting,” as used herein, refer to a non-covalent interaction between macromolecules (e.g., between two polypeptides, between a polypeptide and a nucleic acid; between a polypeptide/guide nucleic acid complex and a target nucleic acid; and the like). While in a state of noncovalent interaction, the macromolecules are said to be “associated” or “interacting” or “binding” (e.g., when a molecule X is said to interact with a molecule Y, it is meant the molecule X binds to molecule Y in a non-covalent manner). Non-limiting examples of non-covalent interactions are ionic bonds, hydrogen bonds, van der Waals and hydrophobic interactions. Not all components of a binding interaction need be sequence-specific (e.g., contacts with phosphate residues in a DNA backbone), but some portions of a binding interaction may be sequence-specific.
[0038] The term, “base editor,” as used herein, refers to a polypeptide or fusion protein comprising a base editing activity. The polypeptide with base editing activity may be referred to as an effector partner. The base editor can differ from a naturally occurring base editing enzyme. It is understood that any reference to a base editor herein also refers to a base editing enzyme variant. The base editor is functional when the effector protein is coupled to a guide nucleic acid. The guide nucleic acid imparts sequence specific activity to the base editor. By way of non-limiting example, the effector protein may comprise a catalytically inactive effector protein (e.g., a catalytically inactive variant of an effector protein described herein). Also, by way of non-limiting example, the base editing enzyme may comprise deaminase activity.
[0039] The term, “catalytically inactive effector protein,” as used herein, refers to an effector protein that is modified relative to a naturally-occurring effector protein to have a reduced or eliminated catalytic activity relative to that of the naturally-occurring effector protein, but retains its ability to interact with a guide nucleic acid. The catalytic activity that is reduced or eliminated is often a nuclease activity. The naturally-occurring effector protein may be a wildtype protein. In some instances, the catalytically inactive effector protein is referred to as a catalytically inactive variant of an effector protein.
[0040] The term, “cis cleavage,” as used herein, refers to cleavage (hydrolysis of a phosphodiester bond) of a target nucleic acid by a complex of an effector protein and a guide nucleic acid (e.g., an RNP complex), wherein at least a portion of the guide nucleic acid is hybridized to at least a portion of the target nucleic acid. Cleavage may occur within or directly adjacent to the portion of the target nucleic acid that is hybridized to the portion of the guide nucleic acid.
[0041] The term, “codon optimized,” as used herein, refers to a mutation of a nucleotide sequence encoding a polypeptide, such as a nucleotide sequence encoding an effector protein, to mimic the codon preferences of the intended host organism or cell while encoding the same polypeptide. Thus, the codons can be changed, but the encoded polypeptide remains unchanged. For example, if the intended target cell was a human cell, a human codon-optimized nucleotide sequence encoding an effector protein could be used. As another non-limiting example, if the intended host cell were a mouse cell, then a mouse codon-optimized nucleotide sequence encoding an effector protein could be generated. As another non-limiting example, if the intended host cell were a eukaryotic cell, then a eukaryote codon-optimized nucleotide sequence encoding an effector protein could be generated. As another non-limiting example, if the intended host cell were a prokaryotic cell, then a prokaryote codon-optimized nucleotide sequence encoding an effector protein could be generated. Codon usage tables are readily available, for example, at the “Codon Usage Database” available at www.kazusa.or.jp/codon. [0042] The terms, “complementary” and “complementarity,” as used herein, in the context of a nucleic acid molecule or nucleotide sequence, refer to the characteristic of a polynucleotide having nucleotides that can undergo cumulative base pairing with their Watson-Crick counterparts (C with G; or A with T) in a reference nucleic acid in antiparallel orientation. For example, when every nucleotide in a polynucleotide or a specified portion thereof forms a base pair with every nucleotide in an equal length sequence of a reference nucleic acid, that polynucleotide is said to be 100% complementary to the sequence of the reference nucleic acid. In a double stranded DNA or RNA sequence, the upper (sense) strand sequence is, in general, understood as going in the direction from its 5'- to 3 '-end, and the complementary sequence is thus understood as the sequence of the lower (antisense) strand in the same direction as the upper strand. Following the same logic, the reverse sequence is understood as the sequence of the upper strand in the direction from its 3'- to its 5 '-end, while the “reverse complement” sequence or the “reverse complementary” sequence is understood as the sequence of the lower strand in the direction of its 5 '- to its 3 '-end. Each nucleotide in a double stranded DNA or RNA molecule that is paired with its Watson-Crick counterpart can be referred to as its complementary nucleotide. The complementarity of modified or artificial base pairs can be based on other types of hydrogen bonding and/or hydrophobicity of bases and/or shape complementarity between bases.
[0043] The term, “cleavage assay,” as used herein, refers to an assay designed to visualize, quantitate or identify cleavage of a nucleic acid. In some instances, the cleavage activity may be cis cleavage activity. Example 1 and Example 2 provide non-limiting examples of cleavage assays.
[0044] The terms, “cleave,” “cleaving” and “cleavage,” as used herein, in the context of a nucleic acid molecule or nuclease activity of an effector protein, refer to the hydrolysis of a phosphodiester bond of a nucleic acid molecule that results in breakage of that bond. The result of this breakage can be a nick (hydrolysis of a single phosphodiester bond on one side of a double-stranded molecule), single strand break (hydrolysis of a single phosphodiester bond on a single-stranded molecule) or double strand break (hydrolysis of two phosphodiester bonds on both sides of a double-stranded molecule) depending upon whether the nucleic acid molecule is single-stranded (e.g., ssDNA or ssRNA) or double-stranded (e.g., dsDNA) and the type of nuclease activity being catalyzed by the effector protein.
[0045] The term, “clustered regularly interspaced short palindromic repeats (CRISPR),” as used herein, refers to a segment of DNA found in the genomes of certain prokaryotic organisms, including some bacteria and archaea, that includes repeated short sequences of nucleotides interspersed at regular intervals between unique sequences of nucleotides derived from another organism.
[0046] The term, “conservative substitution,” as used herein, refers to the replacement of one amino acid for another such that the replacement takes place within a family of amino acids that are related in their side chains. Conversely, the term “non-conservative substitution” as used herein refers to the replacement of one amino acid residue for another that does not have a related side chain. Genetically encoded amino acids can be divided into four families having related side chains: (1) acidic (negatively charged): Asp (D), Glu (E); (2) basic (positively charged): Lys (K), Arg (R), His (H); (3) non-polar (hydrophobic): Cys (C), Ala (A), Vai (V), Leu (L), He (I), Pro (P), Phe (F), Met (M), Trp (W), Gly (G), Tyr (Y), with non-polar also being subdivided into: (i) strongly hydrophobic: Ala (A), Vai (V), Leu (L), He (I), Met (M), Phe (F); and (ii) moderately hydrophobic: Gly (G), Pro (P), Cys (C), Tyr (Y), Trp (W); and (4) uncharged polar: Asn (N), Gin (Q), Ser (S), Thr (T). Amino acids may be related by aliphatic side chains: Gly (G), Ala (A), Vai (V), Leu (L), He (I), Ser (S), Thr (T), with Ser (S) and Thr (T) optionally being grouped separately as aliphatic-hydroxyl; Amino acids may be related by aromatic side chains: Phe (F), Tyr (Y), Trp (W). Amino acids may be related by amide side chains: Asn (N), Gin (Q). Amino acids may be related by sulfur-containing side chains: Cys (C) and Met (M).
[0047] The term, “edited target nucleic acid,” as used herein, refers to a target nucleic acid, wherein the target nucleic acid has undergone an editing, for example, after contact with an effector protein. In some instances, the editing is an alteration in the sequence of the target nucleic acid. In some instances, the edited target nucleic acid comprises an insertion, deletion, or substitution of one or more nucleotides compared to the unedited target nucleic acid.
[0048] The term, “effector protein,” as used herein, refers to a protein, polypeptide, or peptide that is capable of interacting with a nucleic acid, such as a guide nucleic acid, to form a complex (e.g., a RNP complex), wherein the complex interacts with a target nucleic acid. [0049] The term, “effector partner,” as used herein, refers to a protein, polypeptide or peptide that can, in combination with an effector protein, impart some function or activity that can be used to effectuate modification(s) of a target nucleic acid described herein and/or change expression of the target nucleic acid or other nucleic acids associated with the target nucleic acid, when used in connection with compositions, systems and methods described herein.
[0050] The term, “engineered modification,” as used herein, refers to a structural change of one or more nucleic acid residues of a nucleotide sequence or one or more amino acid residue of an amino acid sequence. The engineered modifications of a nucleotide sequence can include chemical modification of one or more nucleobases, or a chemical change to the phosphate backbone, a nucleotide, a nucleobase or a nucleoside. The engineered modifications can be made to an effector protein amino acid sequence or guide nucleic acid nucleotide sequence, or any sequence disclosed herein (e.g., a nucleic acid encoding an effector protein or a nucleic acid that encodes a guide nucleic acid). Methods of modifying a nucleic acid or amino acid sequence are known. One of ordinary skill in the art will appreciate that the engineered modification(s) may be located at any position(s) of a nucleic acid such that the function of the nucleic acid, protein, composition or system is not substantially decreased. Nucleic acids provided herein can be prepared according to any available technique including, but not limited to chemical synthesis, enzymatic synthesis, which is generally termed in vitro-transcription, cloning, enzymatic, or chemical cleavage, etc. In some instances, the nucleic acids provided herein are not uniformly modified along the entire length of the molecule. Different nucleotide modifications and/or backbone structures can exist at various positions within the nucleic acid. [0051] The term, “functional domain,” as used herein, refers to a region of one or more amino acids in a protein that is required for an activity of the protein, or the full extent of that activity, as measured in an in vitro assay. Activities include, but are not limited to nucleic acid binding, nucleic acid editing, nucleic acid modifying, nucleic acid cleaving, protein binding. The absence of the functional domain, including mutations of the functional domain, would abolish or reduce activity.
[0052] The term, “functional fragment,” as used herein, refers to a fragment of a protein that retains some function relative to the entire protein. Non-limiting examples of functions are nucleic acid binding, nucleic acid editing, protein binding, nuclease activity, nickase activity, deaminase activity, demethylase activity, or acetylation activity. A functional fragment may be a recognized functional domain, e.g., a catalytic domain. In some instances, the catalytic domain comprises a RuvC domain. [0053] The term, “functional protein,” as used herein, refers to protein that retains at least some if not all activity relative to the wildtype protein. A functional protein can also include a protein having enhanced activity relative to the wildtype protein. Assays are known and available for detecting and quantifying protein activity, e.g., colorimetric and fluorescent assays. In some instances, a functional protein is a wildtype protein. In some instances, a functional protein is a functional portion of a wildtype protein.
[0054] The terms, “fused” and “linked,” are interchangeable and as used herein, refers to at least two sequences that are connected together, such as by a covalent bond (e.g., an amide bond or a phophodiester bond) or by a linker. The covalent bond can be formed by conjugation (e.g., chemical conjugation or enzymatic conjugation) reaction.
[0055] The term, “fusion protein,” as used herein, refers to a protein comprising at least two polypeptides. The fusion protein may comprise one or more effector proteins and effector partners. In some instances, an effector protein and effector partner are not found connected to one another as a native protein or complex that occurs together in nature.
[0056] The term, “genetic disease,” as used herein, refers to a disease, disorder, condition, or syndrome associated with or caused by one or more mutations in the DNA of an organism having the genetic disease.
[0057] The term, “guide nucleic acid,” as used herein, refers to a nucleic acid that, when in a complex with one or more polypeptides described herein (e.g., an RNP complex) can impart sequence selectivity to the complex when the complex interacts with a target nucleic acid. A guide nucleic acid may be referred to interchangeably as a guide RNA, however it is understood that guide nucleic acids may comprise deoxyribonucleotides (DNA), ribonucleotides (RNA), a combination thereof (e.g., RNA with a thymine base), biochemically or chemically modified nucleobases (e.g., one or more engineered modifications described herein), or combinations thereof.
[0058] The terms, “CRISPR RNA” or “crRNA,” as used herein, refer to a type of guide nucleic acid, wherein the nucleic acid is RNA comprising a first sequence, often referred to herein as a spacer sequence, that hybridizes to a target sequence of a target nucleic acid, and a second sequence, often referred to herein as a repeat sequence or guide sequence, that interacts with an effector protein. In some instances, the second sequence is bound by the effector protein. In some instances, the second sequence hybridizes to a portion of a tracrRNA, wherein the tracrRNA forms a complex with the effector protein.
[0059] The term “trans-activating RNA (tracrRNA),” as used herein, refers to a nucleic acid that comprises a first sequence that is capable of being non-covalently bound by an effector protein, and a second sequence that hybridizes to a portion of a crRNA, which may be referred to as a repeat hybridization sequence.
[0060] The term “handle sequence,” as used herein, refers to a sequence that binds non- covalently with an effector protein. A handle sequence may also be referred to herein as a “scaffold sequence”. In some instances, the handle sequence comprises all, or a portion of, a repeat sequence. In general, a single guide nucleic acid, also referred to as a single guide RNA (sgRNA), comprises a handle sequence that is capable of being non-covalently bound by an effector protein. The nucleotide sequence of a handle sequence may contain a portion of a tracrRNA, but generally does not comprise a sequence that hybridizes to a repeat sequence, also referred to as a repeat hybridization sequence.
[0061] The term “template RNA (retRNA)” as used herein, refers to a nucleic acid comprising: a primer binding sequence and a template sequence. It is understood that template RNAs may comprise DNA, RNA, or a combination thereof (e.g., RNA with a thymine base). In some instances, the template RNA is linked to a guide RNA via a linker sequence to form an rtgRNA. [0062] The term, “template sequence,” as used herein, refers to a portion of a retRNA that contains a desired nucleotide modification relative to a target sequence or portion thereof. By way of non-limiting example, the desired edit may comprise one or more nucleotide insertions, deletions or substitutions relative to a target sequence or portion thereof. In some embodiments, it is identical to, complementary to, or reverse complementary to a target sequence or portion thereof. In some embodiments, the template sequence is complementary to a sequence of the target nucleic acid that is adjacent to a nick site of a target site to be edited, with the exception that it includes a desired edit. The template sequence (also referred in some instances as the RT template (RTT)) can be complementary to at least a portion of the target sequence with the exception of at least one nucleotide.
[0063] The terms, “primer binding sequence (PBS),” as used herein, refer to a portion of a retRNA and serves to bind to a primer sequence of the target nucleic acid. In some embodiments, the primer binding sequence binds to a primer sequence in the target nucleic acid that is formed after the target nucleic acid is cleaved by an effector protein. In some embodiments, the primer binding sequence is linked to the 3’ end of an retRNA. In some embodiments, the primer binding sequence is located at the 5’ end of a retRNA.
[0064] “Primer sequence” as used herein refers to a portion of the target nucleic acid that is capable of hybridizing with the primer binding sequence portion of an retRNA that is generated after cleavage of the target nucleic acid by an effector protein described herein. [0065] The term, “heterologous,” as used herein, refers to at least two different polypeptide sequences that are not found similarly connected to one another in a native nucleic acid or protein. A protein that is heterologous to the effector protein is a protein that is not covalently linked by an amide bond to the effector protein in nature. In some instances, a protein is heterologous when the protein is not encoded by a species that encodes the effector protein. A guide nucleic acid may comprise “heterologous” sequences, which means that it includes a first sequence and a second sequence, wherein the first sequence and the second sequence are not found covalently linked by a phosphodiester bond in nature. Thus, the first sequence is considered to be heterologous with the second sequence, and the guide nucleic acid may be referred to as a heterologous guide nucleic acid. A heterologous system comprises at least one component that is not naturally occurring together with remaining components of the heterologous system.
[0066] The terms, “hybridize,” “hybridizable” and grammatical equivalents thereof, refer to a nucleotide sequence that is able to noncovalently interact, i.e. form Watson-Crick base pairs and/or G/U base pairs, or anneal, to another nucleotide sequence in a sequence-specific, antiparallel, manner (i.e., a nucleotide sequence specifically interacts to a complementary nucleotide sequence) under the appropriate in vitro and/or in vivo conditions of temperature and solution ionic strength. Standard Watson-Crick base-pairing includes: adenine (A) pairing with thymidine (T), adenine (A) pairing with uracil (U), and guanine (G) pairing with cytosine (C) for both DNA and RNA. In addition, for hybridization between two RNA molecules (e.g., dsRNA), and for hybridization of a DNA molecule with an RNA molecule (e.g., when a DNA target nucleic acid base pairs with a guide RNA, etc.): guanine (G) can also base pair with uracil (U). For example, G/U base-pairing is at least partially responsible for the degeneracy (i.e., redundancy) of the genetic code in the context of tRNA anti-codon base-pairing with codons in mRNA. Thus, a guanine (G) can be considered complementary to both an uracil (U) and to an adenine (A). Accordingly, when a G/U base-pair can be made at a given nucleotide position, the position is not considered to be non-complementary, but is instead considered to be complementary. While hybridization typically occurs between two nucleotide sequences that are complementary, mismatches between bases are possible. It is understood that two nucleotide sequences need not be 100% complementary to be specifically hybridizable, hybridizable, partially hybridizable, or for hybridization to occur. Moreover, a nucleotide sequence may hybridize over one or more segments such that intervening or adjacent segments are not involved in the hybridization event (e.g., a bulge, a loop structure or hairpin structure, etc.). The conditions appropriate for hybridization between two nucleotide sequences depend on the length of the sequence and the degree of complementarity, variables which are well known in the art. For hybridizations between nucleic acids with short stretches of complementarity (e.g. complementarity over 35 or less, 30 or less, 25 or less, 22 or less, 20 or less, or 18 or less nucleotides) the position of mismatches may become important (see Sambrook et al., supra, 11.7-11.8). Typically, the length for a hybridizable nucleic acid is 8 nucleotides or more (e.g., 10 nucleotides or more, 12 nucleotides or more, 15 nucleotides or more, 20 nucleotides or more, 22 nucleotides or more, 25 nucleotides or more, or 30 nucleotides or more). Any suitable in vitro assay may be utilized to assess whether two sequences “hybridize”. One such assay is a melting point analysis where the greater the degree of complementarity between two nucleotide sequences, the greater the value of the melting temperature (Tm) for hybrids of nucleic acids having those sequences. The conditions of temperature and ionic strength determine the “stringency” of the hybridization. Temperature, wash solution salt concentration, and other conditions may be adjusted as necessary according to factors such as length of the region of complementation and the degree of complementation. Hybridization and washing conditions are well known and exemplified in Sambrook, J. and Russell, W., Molecular Cloning: A Laboratory Manual, Third Edition, Cold Spring Harbor Laboratory Press, Cold Spring Harbor (2001); and in Green, M. and Sambrook, J., Molecular Cloning: A Laboratory Manual, Fourth Edition, Cold Spring Harbor Laboratory Press, Cold Spring Harbor (2012).
[0067] The term, “indel,” as used herein, refers to an insertion-deletion or indel mutation, which is a type of genetic mutation that results from the insertion and/or deletion of one or more nucleotide in a target nucleic acid. An indel can vary in length (e.g., 1 to 1,000 nucleotides in length) and be detected by any suitable method, including sequencing.
[0068] The term, “indel percentage,” as used herein, refers to a percentage of sequencing reads that show at least one nucleotide has been edited from the insertion and/or deletion of nucleotides regardless of the size of insertion or deletion, or number of nucleotides edited. For example, if there is at least one nucleotide deletion detected in a given target nucleic acid, it counts towards the percent indel value. As another example, if one copy of the target nucleic acid has one nucleotide deleted, and another copy of the target nucleic acid has 10 nucleotides deleted, they are counted the same. This number reflects the percentage of target nucleic acids that are edited by a given effector protein.
[0069] The term, “in vitro,” as used herein, refers to describing something outside an organism. An in vitro system, composition or method may take place in a container for holding laboratory reagents such that it is separated from the biological source from which a material in the container is obtained. In vitro assays can encompass cell-based assays in which living or dead cells are employed. In vitro assays can also encompass a cell-free assay in which no intact cells are employed. The term “in vivo” is used to describe an event that takes place within an organism. The term “ex vivo” is used to describe an event that takes place in a cell that has been obtained from an organism. An ex vivo assay is not performed on a subject. Rather, it is performed upon a sample separate from a subject.
[0070] The terms, “length” and “linked” as used herein, refer to a nucleic acid (polynucleotide) or polypeptide, may be expressed as “kilobases” (kb) or “base pairs (bp),”. Thus, a length of 1 kb refers to a length of 1000 linked nucleotides, and a length of 500 bp refers to a length of 500 linked nucleotides. Similarly, a protein having a length of 500 linked amino acids may also be simply described as having a length of 500 amino acids.
[0071] The term, “linker,” as used herein, refers to a molecule that links a first polypeptide to a second polypeptide (e.g., by an amide bond) or a first nucleic acid to a second nucleic acid (e.g., by a phosphodiester bond).
[0072] The term, “mutation,” as used herein, refers to an alteration that changes an amino acid residue or a nucleotide as described herein. Such an alteration can include, for example, deletions, insertions, and/or substitutions. The mutation can refer to a change in structure of an amino acid residue or nucleotide relative to the starting or reference residue or nucleotide. A mutation of an amino acid residue includes, for example, deletions, insertions and substituting one amino acid residue for a structurally different amino acid residue. Such substitutions can be a conservative substitution, a non-conservative substitution, a substitution to a specific subclass of amino acids, or a combination thereof as described herein. A mutation of a nucleotide includes, for example, changing one naturally occurring base for a different naturally occurring base, such as changing an adenine to a thymine or a guanine to a cytosine or an adenine to a cytosine or a guanine to a thymine. A mutation of a nucleotide base may result in a structural and/or functional alteration of the encoding peptide, polypeptide or protein by changing the encoded amino acid residue of the peptide, polypeptide or protein. A mutation of a nucleotide base may not result in an alteration of the amino acid sequence or function of encoded peptide, polypeptide or protein, also known as a silent mutation. Methods of mutating an amino acid residue or a nucleotide are well known.
[0073] The terms, “mutation associated with a disease” and “mutation associated with a genetic disorder,” as used herein, refer to the co-occurrence of a mutation and the phenotype of a disease. The mutation may occur in a gene, wherein transcription or translation products from the gene occur at a significantly abnormal level or in an abnormal form in a cell or subject harboring the mutation as compared to a non-disease control subject not having the mutation. [0074] The term, “nickase,” as used herein, refers to an enzyme that possess catalytic activity for single stranded nucleic acid cleavage of a double stranded nucleic acid.
[0075] The term, “nickase activity,” as used herein, refers to catalytic activity that results in single stranded nucleic acid cleavage of a double stranded nucleic acid.
[0076] The terms, “non-naturally occurring” and “engineered,” as used herein, refer to indicate involvement of the hand of man. The terms, when referring to a nucleic acid, nucleotide, protein, polypeptide, peptide or amino acid, refer to a molecule, such as but not limited to, a nucleic acid, nucleotide, protein, polypeptide, peptide or amino acid refers to a modification of that molecule (e.g., chemical modification, nucleotide sequence, or amino acid sequence) that is not present in the naturally molecule. The terms, when referring to a composition or system described herein, refer to a composition or system having at least one component that is not naturally associated with the other components of the composition or system. By way of a nonlimiting example, a composition may include an effector protein and a guide nucleic acid that do not naturally occur together. Conversely, and as a non-limiting further clarifying example, an effector protein or guide nucleic acid that is “natural,” “naturally-occurring,” or “found in nature” includes an effector protein and a guide nucleic acid from a cell or organism that have not been genetically modified by the hand of man.
[0077] The terms, “nuclease” and “endonuclease” as used herein, refer to an enzyme which possesses catalytic activity for nucleic acid cleavage.
[0078] The term, “nuclease activity,” as used herein, refers to catalytic activity that results in nucleic acid cleavage (e.g., ribonuclease activity (ribonucleic acid cleavage), or deoxyribonuclease activity (deoxyribonucleic acid cleavage), etc.).
[0079] The term, “nucleic acid,” as used herein, refers to a polymer of nucleotides. A nucleic acid may comprise ribonucleotides, deoxyribonucleotides, combinations thereof, and modified versions of the same. A nucleic acid may be single- stranded or double-stranded, unless specified. Non-limiting examples of nucleic acids are double stranded DNA (dsDNA), single stranded (ssDNA), messenger RNA, genomic DNA, cDNA, DNA-RNA hybrids, and a polymer comprising purine and pyrimidine bases or other natural, chemically or biochemically modified, non-natural, or derivatized nucleotide bases. Accordingly, nucleic acids as described herein may comprise one or more mutations, one or more engineered modifications, or both.
[0080] The term, “nucleic acid expression vector,” as used herein, refers to a plasmid that can be used to express a nucleic acid of interest. [0081] The term, “nuclear localization signal (NLS),” as used herein, refers to an entity (e.g., peptide) that facilitates localization of a nucleic acid, protein, or small molecule to the nucleus, when present in a cell that contains a nuclear compartment.
[0082] The terms, “nucleotide(s)” and “nucleoside(s)”, as used herein, in the context of a nucleic acid molecule having multiple residues, refer to describing the sugar and base of the residue contained in the nucleic acid molecule. Similarly, a skilled artisan could understand that linked nucleotides and/or linked nucleosides, as used in the context of a nucleic acid having multiple linked residues, are interchangeable and describe linked sugars and bases of residues contained in a nucleic acid molecule. When referring to a “nucleobase(s)”, or linked nucleobase, as used in the context of a nucleic acid molecule, it can be understood as describing the base of the residue contained in the nucleic acid molecule, for example, the base of a nucleotide, nucleosides, or linked nucleotides or linked nucleosides. A person of ordinary skill in the art when referring to nucleotides, nucleosides, and/or nucleobases would also understand the differences between RNA and DNA (generally the exchange of uridine for thymidine or vice versa) and the presence of nucleoside analogs, such as modified uridines, do not contribute to differences in identity or complementarity among polynucleotides as long as the relevant nucleotides (such as thymidine, uridine, or modified uridine) have the same complement (e.g., adenosine for all of thymidine, uridine, or modified uridine; another example is cytosine and 5- methylcytosine, both of which have guanosine or modified guanosine as a complement). Thus, for example, the sequence 5'-AXG where X is any modified uridine, such as pseudouridine, Nl-methyl pseudouridine, or 5-methoxyuridine, is considered 100% identical to AUG in that both are perfectly complementary to the same sequence (5' -CAU).
[0083] The term, “pharmaceutically acceptable excipient, carrier or diluent,” as used herein, refers to any substance formulated alongside the active ingredient of a pharmaceutical composition that allows the active ingredient to retain biological activity and is non-reactive with the subject's immune system. Such a substance can be included for the purpose of longterm stabilization, bulking up solid formulations that contain potent active ingredients in small amounts, or to confer a therapeutic enhancement on the active ingredient in the final dosage form, such as facilitating absorption, reducing viscosity, or enhancing solubility. The selection of appropriate substance can depend upon the route of administration and the dosage form, as well as the active ingredient and other factors. Compositions having such substances can be formulated by suitable methods (see, e.g., Remington's Pharmaceutical Sciences, 18th edition, A. Gennaro, ed., Mack Publishing Co., Easton, Pa., 1990; and Remington, The Science and Practice of Pharmacy 21st Ed. Mack Publishing, 2005). [0084] The terms, “polypeptide” and “protein,” as used herein, refer to a polymeric form of amino acids. A polypeptide may include coded and non-coded amino acids, chemically or biochemically modified or derivatized amino acids, and polypeptides having modified peptide backbones. Accordingly, polypeptides as described herein may comprise one or more mutations, one or more engineered modifications, or both. It is understood that when describing coding sequences of polypeptides described herein, said coding sequences do not necessarily require a codon encoding an N-terminal Methionine (M) or a Valine (V) as described for the effector proteins described herein. One skilled in the art would understand that a start codon could be replaced or substituted with a start codon that encodes for an amino acid residue sufficient for initiating translation in a host cell. In some instances, when a heterologous peptide, such as an effector partner, protein tag or NLS, is located at the N terminus of the effector protein, a start codon for the heterologous peptide serves as a start codon for the effector protein as well. Thus, the natural start codon encoding an amino acid residue sufficient for initiating translation (e.g., Methionine (M) or a Valine (V)) of the effector protein may be removed or absent.
[0085] The term, “RT editing enzyme”, as used herein, refers to a protein, polypeptide, or fragment thereof that is capable of catalyzing the editing (insertion, deletion, or base-to-base conversion) of a target nucleotide or nucleotide sequence in a nucleic acid.
[0086] The terms, “target adjacent motif’ and “TAM,” as used herein, refer to a nucleotide sequence found in a target nucleic acid that directs an effector protein to edit the target nucleic acid at a specific location. In some instances, a TAM is required for a complex of an effector protein and a guide nucleic acid (e.g., an RNP complex) to hybridize to and edit the target nucleic acid. In some instances, the complex does not require a TAM to edit the target nucleic acid.
[0087] The terms, “ribonucleotide protein complex” and “RNP” as used herein, refer to a complex of one or more nucleic acids and one or more polypeptides described herein. While the term utilizes “ribonucleotides” it is understood that the one or more nucleic acid may comprise deoxyribonucleotides (DNA), ribonucleotides (RNA), a combination thereof (e.g., RNA with a thymine base), biochemically or chemically modified nucleobases (e.g., one or more engineered modifications described herein), or combinations thereof.
[0088] The terms, “RuvC” and “RuvC domain,” as used herein, refer to a region of an effector protein that is capable of cleaving a target nucleic acid, and in certain instances, of processing a pre-crRNA. In some instances, the RuvC domain is located near the C-terminus of the effector protein. A single RuvC domain may comprise RuvC subdomains, for example a RuvCI subdomain, a RuvCII subdomain and a RuvCIII subdomain. The term “RuvC” domain can also refer to a “RuvC-like” domain. Various RuvC-like domains are known in the art and are easily identified using online tools such as InterPro (https://www.ebi.ac.uk/interpro/). For example, a RuvC-like domain may be a domain which shares homology with a region of TnpB proteins of the IS605 and other related families of transposons.
[0089] The term, “spacer sequence,” as used herein, refers to a nucleotide sequence in a guide nucleic acid that is capable of, at least partially, hybridizing to an equal length portion of a sequence (e.g., a target sequence) of a target nucleic acid.
[0090] The term, “subject,” as used herein, refers to an animal. The subject may be a mammal. The subject may be a human. The subject may be diagnosed or at risk for a disease.
[0091] The term, “target nucleic acid,” as used herein, refers to a nucleic acid that is selected as the nucleic acid for editing, binding, hybridization or any other activity of or interaction with a nucleic acid, protein, polypeptide, or peptide described herein. A target nucleic acid may comprise RNA, DNA, or a combination thereof. A target nucleic acid may be single-stranded (e.g., single-stranded RNA or single-stranded DNA) or double-stranded (e.g., double-stranded DNA).
[0092] The term, “target sequence,” as used herein, in the context of a target nucleic acid, refers to a nucleotide sequence found within a target strand of a target nucleic acid. Such a nucleotide sequence can, for example, hybridize to a respective length portion of a guide nucleic acid.
[0093] The terms, “target strand (TS)” and “non-target strand (NTS)” are used herein to differentiate between the strands of a double stranded DNA molecule to which a guide nucleic acid does or does not hybridize. The term, “target strand (TS),” as used herein, in the context of a target nucleic acid, refers to a strand of a double stranded DNA molecule that comprises a target sequence, to which at least a portion of a guide nucleic acid (e.g., a spacer sequence) hybridizes. The term, “non-target strand (NTS),” as used herein, in the context of a target nucleic acid, refers to a strand of a double stranded DNA molecule to which a guide nucleic acid does not hybridize. Reference may be made to a target sequence of the TS and a target sequence of the NTS. A guide nucleic acid spacer sequence is generally complementary to the target sequence of the TS and generally identical to the target sequence of the NTS.
[0094] The term, “trans cleavage,” as used herein, in the context of cleavage (e.g., hydrolysis of a phosphodiester bond) of one or more target nucleic acids or non-target nucleic acids, or both, by an effector protein that is complexed with a guide nucleic acid and the target nucleic acid. Trans cleavage activity may be triggered by the hybridization of a guide nucleic acid to a target nucleic acid. Trans cleavage of the target nucleic acid may occur away from (e.g., not within or directly adjacent to) the portion of the target nucleic acid that is hybridized to the portion of the guide nucleic acid.
[0095] The term, “transcriptional activator,” as used herein, refers to a polypeptide or a fragment thereof that can activate or increase transcription of a target nucleic acid molecule. [0096] The term, “transcriptional repressor,” as used herein, refers to a polypeptide or a fragment thereof that is capable of arresting, preventing, or reducing transcription of a target nucleic acid.
[0097] The term, “transgene,” as used herein, refers to a nucleotide sequence that is inserted into a cell for expression of said nucleotide sequence in the cell. A transgene is meant to include (1) a nucleotide sequence that is not naturally found in the cell (e.g., a heterologous nucleotide sequence); (2) a nucleotide sequence that is a mutant form of a nucleotide sequence naturally found in the cell into which it has been introduced; (3) a nucleotide sequence that serves to add additional copies of the same (e.g., exogenous or homologous) or a similar nucleotide sequence naturally occurring in the cell into which it has been introduced; or (4) a silent naturally occurring or homologous nucleotide sequence whose expression is induced in the cell into which it has been introduced. The cell in which transgene expression occurs can be a target cell, such as a host cell.
[0098] The terms, “treatment” and “treating,” as used herein, refer to a pharmaceutical or other intervention regimen for obtaining beneficial or desired results in the recipient. Beneficial or desired results include but are not limited to a therapeutic benefit and/or a prophylactic benefit. A therapeutic benefit may refer to eradication or amelioration of symptoms or of an underlying disorder being treated. Also, a therapeutic benefit can be achieved with the eradication or amelioration of one or more of the physiological symptoms associated with the underlying disorder such that an improvement is observed in the subject, notwithstanding that the subject may still be afflicted with the underlying disorder. A prophylactic effect includes delaying, preventing, or eliminating the appearance of a disease or condition, delaying, or eliminating the onset of symptoms of a disease or condition, slowing, halting, or reversing the progression of a disease or condition, or any combination thereof. For prophylactic benefit, a subject at risk of developing a particular disease, or to a subject reporting one or more of the physiological symptoms of a disease may undergo treatment, even though a diagnosis of this disease may not have been made.
[0099] The term, “variant,” as used herein, refers to a form or version of a protein that differs from the wild-type protein. A variant may have a different function or activity relative to the wild-type protein. [0100] The term, “viral vector,” as used herein, refers to a nucleic acid to be delivered into a host cell by a recombinantly produced virus or viral particle.
Introduction
[0101] Disclosed herein are compositions, systems, and methods comprising at least one of: (1) an effector protein or a nucleic acid encoding the effector protein; and (2) an engineered guide nucleic acid or a nucleic acid encoding the engineered guide nucleic acid. The term, “system,” may be used herein for embodiments in which the effector protein or a nucleic acid encoding the effector protein; and the engineered guide nucleic acid or a nucleic acid encoding the engineered guide nucleic acid are not provided in the same container or solution. In some embodiments, these system components are packaged separately, e.g., to be administered separately.
[0102] Effector proteins described herein may bind and cleave (e.g., nick) nucleic acids in a sequence-specific manner. Effector proteins described herein may also cleave (e.g., nick) the target nucleic acid within a target sequence or at a position adjacent to the target sequence. An effector protein may be similar to a CRISPR-associated (Cas) protein in that it may bind a guide nucleic acid that imparts activity or sequence selectivity to the polypeptide. However, unlike Cas proteins, effector proteins described herein are engineered and not necessarily found near a CRISPR array, which includes direct repeats flanking short spacer sequences. Typically, Cas proteins are found within ~5kb of such an array. In contrast, effector proteins described herein may not have a CRISPR array within 5kb of their corresponding genomic locus. Instead, native effector proteins described herein may be associated in the genome with a non-coding RNA that is capable of secondary structure(s).
[0103] In some embodiments, compositions, systems, and methods comprising guide nucleic acids comprise a first region or sequence, at least a portion of which interacts with a polypeptide. In some embodiments, compositions, systems, and methods comprising guide nucleic acids comprise a second sequence that is at least partially complementary to a target nucleic acid, and which may be referred to as a spacer sequence.
[0104] Effector proteins disclosed herein may bind and cleave (e.g., nick) nucleic acids, including double stranded RNA (dsRNA), single-stranded RNA (ssRNA), double stranded DNA (dsDNA), and single-stranded DNA (ssDNA). Polypeptides disclosed herein may provide cis cleavage activity, trans cleavage activity, binding activity, nickase activity, or a combination thereof. [0105] The compositions, systems, and methods described herein are non-naturally occurring. In some embodiments, compositions, systems, and methods comprise an engineered guide nucleic acid (also referred to herein as a guide nucleic acid) or a use thereof. In some embodiments, compositions, systems, and methods comprise an engineered protein or a use thereof. In some embodiments, compositions, systems, and methods comprise an isolated polypeptide or a use thereof. In general, compositions, methods, and systems described herein are not found in nature. In some embodiments, compositions, methods, and systems described herein comprise at least one non-naturally occurring component. For example, disclosed compositions, methods, and systems may comprise a guide nucleic acid, wherein the sequence of the guide nucleic acid is different or modified from that of a naturally-occurring guide nucleic acid.
[0106] In some embodiments, compositions, systems, and methods comprise at least two components that do not naturally occur together. For example, disclosed compositions, systems, and methods may comprise a guide nucleic acid comprising a first region, at least a portion of which, interacts with a polypeptide, and a second region that is at least partially complementary to a target sequence in a target nucleic acid, wherein the first region and second region do not naturally occur together and/or are heterologous to each other. Also, by way of non-limiting example, disclosed compositions, systems, and methods may comprise a guide nucleic acid and an effector protein that do not naturally occur together. Likewise, by way of non-limiting example, disclosed compositions, systems, and methods may comprise a ribonucleotide-protein (RNP) complex comprising an effector protein and a guide nucleic acid that do not occur together in nature. Conversely, and for clarity, an effector protein or guide nucleic acid that is “natural,” “naturally-occurring,” or “found in nature” includes effector proteins and guide nucleic acids from cells or organisms that have not been genetically modified by a human or machine.
[0107] In some embodiments, the guide nucleic acid comprises a non-natural nucleotide sequence. In some embodiments, the non-natural nucleotide sequence is a nucleotide sequence that is not found in nature. The non-natural nucleotide sequence may comprise a portion of a naturally-occurring sequence, wherein the portion of the naturally-occurring sequence is not present in nature absent the remainder of the naturally-occurring sequence. In some embodiments, the guide nucleic acid comprises two naturally-occurring sequences arranged in an order or proximity that is not observed in nature. In some embodiments, compositions and systems comprise a ribonucleotide complex comprising an effector protein and a guide nucleic acid that do not occur together in nature. In some embodiments, compositions and systems comprise at least two components that do not occur together in nature, wherein the at least two components comprise at least one of an effector protein, an effector partner and a guide nucleic acid. Guide nucleic acids may comprise a first sequence and a second sequence that do not occur naturally together. A guide nucleic acid may comprise a first sequence that occurs in a first organism and a second sequence that occurs in a second organism, wherein the first organism and the second organism are different. The guide nucleic acid may comprise a third sequence disposed at a 3’ or 5’ end of the guide nucleic acid, or between the first and second sequences of the guide nucleic acid. In some embodiments, the guide nucleic acid comprises two heterologous sequences arranged in an order or proximity that is not observed in nature. Therefore, compositions and systems described herein are not naturally occurring.
[0108] In some embodiments, compositions, systems, and methods described herein comprise a polypeptide (e.g., an effector protein, an effector partner, a fusion protein, or a combination thereof) that is similar to a naturally occurring polypeptide. The polypeptide may lack a portion of the naturally occurring polypeptide. The polypeptide may comprise a mutation relative to the naturally-occurring polypeptide, wherein the mutation is not found in nature. The polypeptide may also comprise at least one additional amino acid relative to the naturally- occurring polypeptide. In some embodiments, the polypeptide may comprise a heterologous peptide. For example, the polypeptide may comprise an addition of a nuclear localization signal relative to the natural occurring polypeptide. In some embodiments, a nucleotide sequence encoding the polypeptide is codon optimized (e.g., for expression in a eukaryotic cell) relative to the naturally occurring sequence.
I. Effector Proteins
[0109] Provided herein, in certain embodiments, are compositions that comprise one or more effector proteins or a nucleic acid encoding the same, and uses thereof. Also provided herein are compositions that comprise a nucleic acid, wherein the nucleic acid encodes any of one the effector proteins described herein. The nucleic acid may be an mRNA. The nucleic acid may be a nucleic acid expression vector. By way of non-limiting example, the nucleic acid expression vector may be a viral vector, such as an AAV vector. In some embodiments, the effector protein comprises a DIS dual protein. In general, a DIS dual protein is a compact effector protein comprising two nuclease domains. In general, the two nuclease domains are a RuvC domain and an HNH domain. In some embodiments, the length of a compact effector protein is less than 900, less than 800, less than 700, less than 600, less than 500, or less than 400 linked amino acids, and greater than 300 linked amino acids. In some embodiments, the DIS dual protein comprises an IscB protein or engineered variant thereof. In some embodiments, the engineered variant is at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, or at least 99% identical to an IscB protein.
[0110] An effector protein provided herein interacts with a guide nucleic acid to form a complex. In some embodiments, the complex interacts with a target nucleic acid, a non-target nucleic acid, or both. In some embodiments, an interaction between the complex and a target nucleic acid, a non-target nucleic acid, or both comprises one or more of: recognition of a target adjacent motif (TAM) sequence within the target nucleic acid by the effector protein, hybridization of the guide nucleic acid to the target nucleic acid, modification of the target nucleic acid and/or the non-target nucleic acid by the effector protein, or combinations thereof. See, e.g. FIG. 2. In some embodiments, recognition of a TAM sequence within a target nucleic acid may direct the modification activity of an effector protein.
[OHl] In some embodiments, the effector protein described herein may bind and, optionally, modify nucleic acids in a sequence-specific manner. Effector proteins described herein may also modify the target nucleic acid within a target sequence or at a position adjacent to the target sequence. In some embodiments, an effector protein is activated when it binds a certain sequence of a nucleic acid described herein, allowing the effector protein to modify a region of a target nucleic acid that is near, but not adjacent to the target sequence. Effector proteins may modify a nucleic acid by cis cleavage or trans cleavage. Alternatively or additionally, effector proteins described herein modify a non-target nucleic acid by trans cleavage on the non-target nucleic acid. In some embodiments, effector proteins may have nickase activity or nuclease activity. Effector proteins disclosed herein may modify nucleic acids, including single stranded RNA (ssRNA), double stranded DNA (dsDNA), and single-stranded DNA (ssDNA). The modification of the target nucleic acid generated by an effector protein, as a non-limiting example, may result in modulation of the expression of the nucleic acid (e.g., increasing or decreasing expression of the nucleic acid) or modulation of the activity of a translation product of the target nucleic acid (e.g., inactivation of a protein binding to an RNA molecule or hybridization).
[0112] In some embodiments, effector proteins described herein comprise modification activities. In some embodiments, the modification activity of the polypeptide described herein may be nickase activity for a double stranded nucleic acid, binding activity, insertion activity, substitution activity, chemical modification activity, or a combination thereof. In some embodiments, the modification activity of the polypeptide may result in: cleavage of at least one strand of a target nucleic acid, deletion of one or more nucleotides of a target nucleic acid, insertion of one or more nucleotides into a target nucleic acid, substitution of one or more nucleotides of a target nucleic acid with an alternative nucleotide, chemical modification of one or more nucleotides of a target nucleic acid to an alternative nucleotide, or combinations thereof. In some embodiments, the cleavage activity is a nicking activity.
[0113] An effector protein may be an engineered effector protein having increased modification activity (e.g., catalytic activity) and/or increased substrate binding activity (e.g., substrate selectivity, specificity, and/or affinity) relative to a native effector protein. An effector protein may be an engineered effector protein having reduced modification activity (e.g., a catalytically defective effector protein) or no modification activity (e.g., a catalytically inactive effector protein) relative to a native effector protein.
[0114] In some embodiments, effector proteins described herein comprise one or more functional domains. Effector protein functional domains can include a target adjacent motif (TAM)-interacting domain, an oligonucleotide-interacting domain, one or more recognition domains, a non-target strand interacting domain, an HNH domain, a RuvC domain, or a combination thereof. A TAM interacting domain can be a target strand TAM interacting domain (TPID) or a non-target strand TAM interacting domain (NTPID). In some embodiments, a TAM interacting domain, such as a TPID or a NTPID, on an effector protein describes a region of an effector protein that interacts with target nucleic acid. In some embodiments, effector proteins comprise one or more recognition domain (REC domain) with a binding affinity for a guide nucleic acid or for a guide nucleic acid-target nucleic acid heteroduplex. An effector protein may comprise a zinc finger domain.
[0115] FIG. 1 shows the domains and positions thereof for the parent sequence provided in TABLE 2. The amino terminal PLMP domain is named for its PLMP amino acid motif. The TI domain (TID) may form a five-stranded antiparallel 13-barrel, with its carboxy-terminal 136 strand interacting with the 136 strand of the RuvC domain to form a seven-stranded 13-sheet. The PI domain (PID) may contain a core 13-barrel. The 132- 133 hairpin in the TID may be inserted into the major groove of the TAM duplex. Publicly available tools, such as InterProScan, may be used for detection of functional domains and sequence patterns matching previously described functional domains. In some instances, effector proteins comprise an amino-terminal PLMP domain, RuvC-like nuclease domains containing three conserved catalytic motifs (RuvC-I-III), with an inserted Arg-rich segment known as the bridge helix (BH), and the HNH nuclease domain. The RuvC-I and RuvC-II motifs may be connected via the BH and a P- hairpin-containing linker (referred to as the REC linker), as well as the Wedge (WED) and TAM-interacting (TI) domains located similarly to the corresponding domains in Cas9. Without being bound by theory, effector proteins provided in TABLE 1, TABLE 6, and TABLE 8 are expected to have a similar structure.
[0116] In some embodiments, the effector protein comprises an HNH domain. In some embodiments, the effector protein comprises an HNH domain and a RuvC domain. In some embodiments, the effector protein may have a mutation in a nuclease domain. In some embodiments, the nuclease domain is a RuvC domain. In some embodiments, the nuclease domain is an HNH domain. An HNH domain may be characterized as comprising two antiparallel P-strands connected with a loop of varying length, and flanked by an a-helix, with a metal (divalent cation) binding site between the two P-strands. A RuvC domain may be characterized by a six-stranded beta sheet surrounded by four alpha helices, with three conserved subdomains contributing catalytic to the activity of the RuvC domain. In some embodiments, a RuvC domain comprises with substrate binding activity, catalytic activity, or both. In some embodiments, the RuvC domain may be defined by a single, contiguous sequence, or a set of RuvC subdomains that are not contiguous with respect to the primary amino acid sequence of the protein. An effector protein of the present disclosure may include multiple RuvC subdomains, which may combine to generate a RuvC domain with substrate binding or catalytic activity. For example, an effector protein may include three RuvC subdomains (RuvC-I, RuvC-II, and RuvC-III) that are not contiguous with respect to the primary amino acid sequence of the effector protein but form a RuvC domain once the protein is produced and folds. In some embodiments, the effector protein cleaves within the target sequence. In some instances, the RuvC domain is responsible for cleavage. In some instances, the HNH domain is responsible for cleavage.
[0117] When describing a mutation that changes an amino acid residue or a nucleotide as described herein, such a change or changes can include, for example, deletions, insertions, and/or substitutions. The mutation can refer to a change in structure of an amino acid residue or nucleotide relative to the starting or reference residue or nucleotide. A mutation of an amino acid residue includes, for example, deletions, insertions and substituting one amino acid residue for a structurally different amino acid residue. Such substitutions can be a conservative substitution, a non-conservative substitution, a substitution to a specific sub-class of amino acids, or a combination thereof as described herein. A mutation of a nucleotide includes, for example, changing one naturally occurring base for a different naturally occurring base, such as changing an adenine to a thymine or a guanine to a cytosine or an adenine to a cytosine or a guanine to a thymine. A mutation of a nucleotide base may result in a structural and/or functional alteration of the encoding peptide, polypeptide or protein by changing the encoded amino acid residue of the peptide, polypeptide or protein. A mutation of a nucleotide base may not result in an alteration of the amino acid sequence or function of encoded peptide, polypeptide or protein, also known as a silent mutation.
[0118] TABLE 1 and TABLE 8 provide illustrative amino acid sequences of an engineered effector protein for use in the compositions, systems and methods described herein. TABLE 6 provides exemplary effector proteins that may be engineered. In some embodiments, systems, compositions, and methods described herein comprise an effector protein in TABLE 6 or a use thereof. In some embodiments, systems, compositions, and methods described herein comprise an engineered variant of an effector protein in TABLE 6 or a use thereof.
[0119] In some embodiments, compositions, systems and methods described herein comprise an effector protein, or a nucleic acid encoding the effector protein, wherein the amino acid sequence of the effector protein comprises at least 200 contiguous amino acids or more of the amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8, wherein the amino acid sequence of the effector protein is not identical to any one of the amino acid sequences selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, the amino acid sequence of an effector protein provided herein comprises at least 200, at least 220, at least 240, at least 260, at least 280, at least 300, at least 320, at least 340, at least 360, at least 380, at least 400 contiguous amino acids, at least 420 contiguous amino acids, at least 440 contiguous amino acids, at least 460 contiguous amino acids, at least 480 contiguous amino acids, at least 500 contiguous amino acids of a sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, the length of the effector protein is about 480 to about 560 amino acids.
[0120] In some embodiments, the amino acid sequence of an effector protein provided herein comprises about 100, about 120, about 140, about 160, about 180, about 200, about 220, about 240, about 260, about 280, about 300, about 320, about 340, about 360, about 380, about 400, about 420, about 440, about 460, about 480, about 500, about 520, about 540 contiguous amino acids of a sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, the length of the effector protein is less than 600, less than 580, less than 560, less than 540, less than 520, less than 500, less than 480, less than 460, less than 440, less than 420, or less than 400 contiguous amino acids; and at least 300, at least 320, at least 340, at least 360, at least 380 contiguous amino acids of a sequence selected from TABLE 1, TABLE 6, and TABLE 8 [0121] In some embodiments, compositions, systems and methods described herein comprise an effector protein or a nucleic acid encoding the effector protein, wherein the effector protein comprises a portion of the amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, the effector protein comprises a portion of the amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8, wherein the portion does not comprise at least the first 10 amino acids, at least the first 20 amino acids, at least the first 40 amino acids, at least the first 60 amino acids, at least the first 80 amino acids, at least the first 100 amino acids, at least the first 120 amino acids, at least the first 140 amino acids, at least the first 160 amino acids, at least the first 180 amino acids, or at least the first 200 amino acids of the amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, the effector protein comprises a portion of the amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8, wherein the portion does not comprise the last 10 amino acids, the last 20 amino acids, the last 40 amino acids, the last 60 amino acids, the last 80 amino acids, the last 100 amino acids, the last 120 amino acids, the last 140 amino acids, the last 160 amino acids, the last 180 amino acids, or the last 200 amino acids of the amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8.
[0122] In some embodiments, compositions, systems, and methods described herein comprise an effector protein, or a nucleic acid encoding the effector protein, wherein the effector protein comprises an amino acid sequence that is at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, but less than 100% identical to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, an effector protein provided herein comprises an amino acid sequence that is at least 65% but less than 100% identical to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, an effector protein provided herein comprises an amino acid sequence that is at least 70% but less than 100% identical to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, an effector protein provided herein comprises an amino acid sequence that is at least 75% but less than 100% identical to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, an effector protein provided herein comprises an amino acid sequence that is at least 80% but less than 100% identical to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, an effector protein provided herein comprises an amino acid sequence that is at least 85% but less than 100% identical to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, an effector protein provided herein comprises an amino acid sequence that is at least 90% but less than 100% identical to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, an effector protein provided herein comprises an amino acid sequence that is at least 95% but less than 100% identical to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, an effector protein provided herein comprises an amino acid sequence that is at least 97% but less than 100% identical to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, an effector protein provided herein comprises an amino acid sequence that is at least 98% but less than 100% identical to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, an effector protein provided herein comprises an amino acid sequence that is at least 99% but less than 100% identical to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, an effector protein provided herein comprises an amino acid sequence that is less than 100% identical to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8
[0123] In some embodiments, compositions, systems, and methods described herein comprise an effector protein, or a nucleic acid encoding the effector protein, wherein the effector protein comprises an amino acid sequence that is at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% similar, but not the same, to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, an effector protein provided herein comprises an amino acid sequence that is at least 80% similar, but not the same, to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, an effector protein provided herein comprises an amino acid sequence that is at least 85% similar, but not the same, to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, an effector protein provided herein comprises an amino acid sequence that is at least 90% similar, but not the same, to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, an effector protein provided herein comprises an amino acid sequence that is at least 95% similar, but not the same, to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, an effector protein provided herein comprises an amino acid sequence that is at least 97% similar, but not the same, to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, an effector protein provided herein comprises an amino acid sequence that is at least 98% similar, but not the same, to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, an effector protein provided herein comprises an amino acid sequence that is at least 99% similar, but not the same, to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, an effector protein provided herein comprises an amino acid sequence that is 100% similar, but not the same, to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8
[0124] In some embodiments, compositions, systems, and methods described herein comprise an effector protein, or a nucleic acid encoding the effector protein, wherein the effector protein comprises one or more amino acid alterations relative to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, the one or more alterations comprises at least one, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least twelve, at least sixteen, at least twenty, or more amino acid alterations relative to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, the one or more alterations comprises one to twenty, one to sixteen, one to twelve, one to eight, one to four, four to twenty, four to sixteen, four to twelve, four to eight, eight to twenty, eight to sixteen, eight to twelve, twelve to twenty, twelve to sixteen, sixteen to twenty, or more amino acid alterations relative to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, the one or more amino acid alterations comprises substitutions (e.g., conservative substitutions, nonconservative substitutions), deletions, or combinations thereof. In some embodiments, an effector protein or a nucleic acid encoding the effector protein comprises 1 amino acid alteration, 2 amino acid alterations, 3 amino acid alterations, 4 amino acid alterations, 5 amino acid alterations, 6 amino acid alterations, 7 amino acid alterations, 8 amino acid alterations, 9 amino acid alterations, 10 amino acid alterations or more relative to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8.
[0125] In some embodiments, compositions, systems, and methods described herein comprise an effector protein, or a nucleic acid encoding the effector protein, wherein the effector protein comprises one or more substitutions relative to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, the one or more substitutions comprises at least one, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least twelve, at least sixteen, at least twenty, or more substitutions relative to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, the one or more substitutions comprises one to twenty, one to sixteen, one to twelve, one to eight, one to four, four to twenty, four to sixteen, four to twelve, four to eight, eight to twenty, eight to sixteen, eight to twelve, twelve to twenty, twelve to sixteen, sixteen to twenty, or more substitutions relative to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, the one or more substitutions comprises 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more amino acid substitutions relative to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 86. In some embodiments, the one or more amino acid substitutions comprise one, two, three, four, five, six, seven, eight, nine, ten or more substitutions relative to an amino acid sequence selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, the one or more amino acid substitutions comprise one or more substitutions with a positively charged amino acid residues. In some embodiments, the positively charged amino acid residue is independently selected from Lys (K), Arg (R), or His (H). In some embodiments, the one or more substitutions comprise one or more conservative substitutions, one or more nonconservative substitutions, or combinations thereof.
[0126] In some embodiments, compositions, systems, and methods described herein comprise an effector protein, or a nucleic acid encoding the effector protein, wherein the effector protein comprises one or more conservative substitutions relative to any one of the sequences selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, the one or more conservative substitutions comprises at least one, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least twelve, at least sixteen, at least twenty, or more conservative substitutions relative to any one of the sequences selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, the one or more conservative substitutions comprises one to twenty, one to sixteen, one to twelve, one to eight, one to four, four to twenty, four to sixteen, four to twelve, four to eight, eight to twenty, eight to sixteen, eight to twelve, twelve to twenty, twelve to sixteen, or sixteen to twenty conservative substitutions relative to any one of the sequences selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, the one or more conservative substitutions comprise one, two, three, four, five, six, seven, eight, nine, ten or more conservative substitutions relative to any one of the sequences selected from TABLE 1, TABLE 6, and TABLE 8
[0127] In some embodiments, compositions, systems, and methods described herein comprise an effector protein, or a nucleic acid encoding the effector protein, wherein the effector protein comprises one or more non-conservative substitutions relative to any one of the sequences selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, the one or more conservative substitutions comprises not more than one, two, three, four, five, six, seven, eight, nine, or ten substitutions in the amino acid sequence relative to any one of the sequences selected from TABLE 1, TABLE 6, and TABLE 8. In some embodiments, the one or more non-conservative substitutions are in a RuvC domain, HNH domain, or both. In some embodiments, the non-conservative substitution(s) replaces a catalytic residue(s) of the domain(s).
[0128] In some embodiments, compositions, systems, and methods described herein comprise an effector protein, or a nucleic acid encoding the effector protein, wherein the effector protein comprises an amino acid sequence that is at least 90%, at least 95%, at least 97%, at least 98%, or at least 99% identical to SEQ ID NO: 624, wherein the effector protein comprises an amino acid substitution of H246A relative to SEQ ID NO: 624 Without being bound by theory, the H246A amino acid substitution is believed to be located in an HNH domain of effector protein of SEQ ID NO: 624, and thereby provides the engineered variant with nickase activity. In some embodiments, the effector protein encoded by a nucleic acid sequence that is at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 912. In some embodiments, compositions, systems, and methods described herein comprise an effector protein, or a nucleic acid encoding the effector protein, wherein the effector protein comprises an amino acid sequence that is at least 90%, at least 95%, at least 97%, at least 98%, or at least 99% identical to SEQ ID NO: 713, wherein the effector protein comprises an amino acid substitution of H244A relative to SEQ ID NO: 713. In some embodiments, compositions, systems, and methods described herein comprise an effector protein, or a nucleic acid encoding the effector protein, wherein the effector protein comprises an amino acid sequence that is at least 90%, at least 95%, at least 97%, at least 98%, or at least 99% identical to SEQ ID NO: 856, wherein the effector protein comprises an amino acid substitution of H248A relative to SEQ ID NO: 856.
[0129] In some embodiments, effector proteins are engineered from the parent sequence provided in TABLE 2. Any one of the effector proteins in TABLE 6 could be a parent sequence that is similarly engineered. In some embodiments, any one of the sequences replaced (described by positions replaced in TABLE 2) could be replaced with a similar domain from another protein. For example, where HNH domains were swapped as described in TABLE 2, HNH domains from other proteins could similar be inserted in those positions. In some instances, the sequences inserted as described in TABLE 2 are nearly identical to said sequences. For example, sequences inserted could be at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99% to the sequence provided in TABLE 2. In some instances, the sequences inserted are similar to the sequences described in TABLE 2. In some instances, sequences inserted are at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99% to the sequence provided in TABLE 2.
[0130] In some embodiments, effector proteins comprise a combination of sequences replaced, as described in TABLE 2. By way of non-limiting example, an effector protein may comprise the parent sequence with the exception of an HNH domain replacement described in TABLE 2 and a TID domain replacement described in TABLE 2.
[0131] In some embodiments, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99% or 100% of a domain in the parent sequence of TABLE 2 is replaced by an inserted amino acid sequence. In some embodiments, additional amino acids around a domain are deleted or replaced. In some embodiments, less than 10, less than 50, less than 100, less than 150, or less than 200 amino acids around a domain are deleted or replaced.
[0132] In some embodiments, effector proteins comprise one or more amino acid alterations relative to the sequences provided in TABLE 1, TABLE 6, and TABLE 8. In some embodiments, the one or more amino acid alterations may result in a change in activity of the effector protein relative to a naturally-occurring counterpart. In some embodiments, the one or more amino acid alterations does not result in a change in activity of the effector protein relative to a naturally-occurring counterpart. For example, and as described in further detail below, the one or more amino acid alteration increases or decreases catalytic activity of the effector protein relative to a naturally-occurring counterpart. In another example, the one or more amino acid alteration increases or decreases binding activity of the effector protein relative to a naturally- occurring counterpart.
[0133] An effector protein that has decreased catalytic activity may be referred to as catalytically or enzymatically inactive, catalytically or enzymatically dead, as a dead protein or a dCas protein. In some embodiments, such a protein may comprise an enzymatically inactive domain (e.g. inactive nuclease domain). For example, a nuclease domain (e.g., RuvC domain or HNH domain) of an effector protein may be deleted or mutated relative to a wildtype counterpart so that it is no longer functional or comprises reduced nuclease activity. In some embodiments, a catalytically inactive effector protein may bind to a guide nucleic acid and/or a target nucleic acid but does not cleave the target nucleic acid. In some embodiments, a catalytically inactive effector protein may associate with a guide nucleic acid to activate or repress transcription of a target nucleic acid. In some embodiments, a catalytically inactive effector protein is fused to an effector partner that confers an alternative activity to an effector protein activity. Such fusion proteins are described herein and throughout. [0134] In some embodiments, effector proteins of the present disclosure may exhibit activity or enhanced activity in the presence of a co-factor. In some embodiments, the co-factor allows the effector proteins to perform a function. In some embodiments, the function is pre-crRNA processing and/or target nucleic acid cleavage. As discussed in Jiang F. and Doudna J.A. (Annu. Rev. Biophys. 2017. 46:505-29), Cas9 uses divalent metal ions as co-factors. The suitability of a divalent metal ion as a cofactor can easily be assessed, such as by methods based on those described by Sundaresan et al. (Cell Rep. 2017 Dec 26; 21(13): 3728-3739). In some embodiments, the co-factor is a divalent metal ion. Non-limiting exemplary divalent metal ions include: Mg2+, Mn2+, Zn2+, Ca2+, and Cu2+. In some embodiments, the effector protein forms a complex with a divalent metal ion. In some embodiments, the effector protein forms a complex with Mg2+, Mn2+, Zn2+, Ca2+, or Cu2+.
[0135] Effector proteins of the present disclosure may be synthesized, using any suitable method. In some embodiments, the effector proteins may be produced in vitro or by eukaryotic cells or by prokaryotic cells. In some embodiments, the effector proteins may be further processed by unfolding (e.g. heat denaturation, dithiothreitol reduction, etc.) and may be further refolded, using any suitable method. In some embodiments, the nucleic acid(s) encoding the effector proteins described herein, the recombinant nucleic acid(s) described herein, the vectors described herein may be produced in vitro or in vivo by eukaryotic cells or by prokaryotic cells. [0136] In some embodiments, effector proteins described herein are isolated from cell lysate. In some embodiments, the compositions described herein may comprise 20% or more by weight, 75% or more by weight, 95% or more by weight, or 99.5% or more by weight of an effector protein, related to the method of preparation of compositions described herein and its purification thereof, wherein percentages may be upon total protein content in relation to contaminants. Thus, in some embodiments, the effector protein is at least 80% pure, at least 85% pure, at least 90% pure, at least 95% pure, at least 98% pure, or at least 99% pure (e.g., free of contaminants, non-engineered proteins or other macromolecules, etc.).
II. Target Adjacent Motif (TAM) Sequences
[0137] Effector proteins of the present disclosure may cleave or nick a target nucleic acid within or near a target adjacent motif (TAM) sequence of the target nucleic acid. In some instances, the TAM comprises 4-8 nucleotides. In some instances, the length of the TAM is 4 nucleotides. In some instances, the length of the TAM is 5 nucleotides. In some instances, the length of the TAM is 6 nucleotides. In some instances, the length of the TAM is 7 nucleotides. In some instances, the length of the TAM is 8 nucleotides. [0138] In some embodiments, the target nucleic acid is a double stranded nucleic acid comprising a target strand and a non-target strand. In some embodiments, cleavage occurs within 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, or 50 nucleotides of a 5’ or 3’ terminus of a TAM sequence. In some embodiments, effector proteins described herein recognize a TAM sequence. In some embodiments, recognizing a TAM sequence comprises interacting with a sequence adjacent to the TAM. In some embodiments, a target nucleic acid comprises a target sequence that is adjacent to a TAM sequence. In some embodiments, the effector protein does not require a TAM to bind and/or cleave a target nucleic acid.
[0139] In some embodiments, a target nucleic acid is a double stranded nucleic acid comprising a target strand and a non-target strand, wherein the target strand comprises a target sequence. In some embodiments, the TAM sequence is located on the target strand. In some embodiments, the TAM sequence is located on the non-target strand. In some embodiments, the TAM sequence described herein is directly adjacent to the target sequence on the non-target strand. In some embodiments, the TAM sequence described herein is adjacent (e.g., within 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, or 50 nucleotides) to the target sequence on the non-target strand. In some embodiments, the TAM sequence is located 3’ of the target sequence on the non-target strand. In some embodiments, such a TAM described herein is directly adjacent to the target sequence on the target strand or the non-target strand. In some embodiments, an RNP cleaves the target strand or the non-target strand. In some embodiments, the RNP cleaves both, the target strand and the non-target strand. In some embodiments, an RNP recognizes the TAM sequence, and hybridizes to a target sequence of the target nucleic acid. In some embodiments, the RNP cleaves the target nucleic acid, wherein the RNP has recognized the TAM sequence and is hybridized to the target sequence. In some embodiments, at least a portion of the guide nucleic acid (e.g., the spacer sequence) hybridizes to the target sequence of the target strand.
[0140] In some embodiments, an effector protein described herein, or a multimeric complex thereof, recognizes a TAM on a target nucleic acid. In some embodiments, multiple effector proteins of the multimeric complex recognize a TAM on a target nucleic acid. In some embodiments, at least two of the multiple effector proteins recognize the same TAM sequence. In some embodiments, at least two of the multiple effector proteins recognize different TAM sequences. In some embodiments, only one effector protein of the multimeric complex recognizes a TAM on a target nucleic acid.
[0141] An effector protein of the present disclosure, or a multimeric complex thereof, may cleave or nick a target nucleic acid within or near a target adjacent motif (TAM) sequence of the target nucleic acid. In some embodiments, cleavage occurs within 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 nucleotides of a 5’ or 3’ terminus of a TAM sequence.
[0142] Engineered proteins in TABLE 1, TABLE 6 and TABLE 8 may recognize a target adjacent motif (TAM) of 5’-NWRRNA-3’, wherein N is any nucleotide, W is A or T, and R is A or G. In some embodiments, engineered proteins in TABLE 1, TABLE 6 and TABLE 8 may recognize a target sequence that is adjacent to a TAM of 5’-ATAANNN-3’, wherein N is any nucleotide.
[0143] In some embodiments, an effector protein described herein comprises an amino acid sequence at least 90-100% identical to SEQ ID NO: 856 and recognizes a TAM of ATAANNN when complexed with a guide nucleic acid, wherein N is any nucleotide. In some embodiments, an effector protein described herein comprises an amino acid sequence at least 90-100% identical to SEQ ID NO: 624 and recognizes a TAM of ARRRGNN when complexed with a guide nucleic acid, wherein N is any nucleotide and R is A or G. In some embodiments, an effector protein described herein comprises an amino acid sequence at least 90-100% identical to SEQ ID NO: 713 and recognizes a TAM of GNAAANN when complexed with a guide nucleic acid, wherein N is any nucleotide. In some embodiments, an effector protein described herein comprises an amino acid sequence at least 90-100% identical to SEQ ID NO: 833 and recognizes a TAM of ATAANNN when complexed with a guide nucleic acid, wherein N is any nucleotide. In some embodiments, an effector protein described herein comprises an amino acid sequence at least 90-100% identical to SEQ ID NO: 691 and recognizes a TAM of GYARRNN when complexed with a guide nucleic acid, wherein N is any nucleotide, Y is C or T, and R is A or G. In some embodiments, an effector protein described herein comprises an amino acid sequence at least 90-100% identical to SEQ ID NO: 774 and recognizes a TAM of RTANNNN when complexed with a guide nucleic acid, wherein N is any nucleotide and R is A or G.
III. Effector Partners
[0144] Provided herein are compositions, systems, and methods comprising one or more effector partners or uses thereof. In some embodiments, the effector partner is a heterologous protein an effector protein described herein. In some embodiments, the effector partner is not an effector protein as described herein. In some embodiments, the effector partner is capable of imparting a function or activity that is not provided by an effector protein as described herein. In some embodiments, the effector partner comprises a second effector protein or a multimeric form thereof. [0145] In some embodiments, an effector partner imparts a function or activity to a fusion protein comprising an effector protein that is not provided by the effector protein, including but not limited to nuclease activity, methyltransferase activity, demethylase activity, DNA repair activity, DNA damage activity, deamination activity, dismutase activity, alkylation activity, depurination activity, oxidation activity, dimer forming activity (e.g., pyrimidine dimer forming activity), integrase activity, transposase activity, recombinase activity, polymerase activity, ligase activity, helicase activity, photolyase activity, glycosylase activity, acetyltransferase activity, deacetylase activity, kinase activity, phosphatase activity, ubiquitin ligase activity, deubiquitinating activity, adenylation activity, deadenylation activity, SUMOylating activity, deSUMOylating activity, ribosylation activity, deribosylation activity, myristoylation activity or demyristoylation activity, modification of a polypeptide associated with target nucleic acid (e.g., a histone), and/or signaling activity.
[0146] In some embodiments, the effector partner is fused or linked to an effector protein described herein. In some embodiments, the amino terminus of the effector partner is linked to the carboxy terminus of the effector protein directly or by a linker. In some embodiments, the carboxy terminus of the effector partner is linked to the amino terminus of the effector protein directly or by a linker. In some embodiments, the effector partner may be functional when the effector protein is coupled to a guide nucleic acid. In some embodiments, the effector partner may be functional when the effector protein is coupled to a target nucleic acid. In some embodiments, the guide nucleic acid imparts sequence specific activity to the effector partner. By way of non-limiting example, the effector protein may comprise a catalytically inactive effector protein (e.g., a catalytically inactive variant of an effector protein described herein) when fused or linked to an effector partner.
[0147] In some embodiments, the effector partner may directly or indirectly edit a target nucleic acid. Edits can be of a nucleobase, nucleotide, or nucleotide sequence of a target nucleic acid. In some embodiments, the effector partner may interact with additional proteins, or functional fragments thereof, to make modifications to a target nucleic acid. In other embodiments, the effector partner may modify proteins associated with a target nucleic acid. In some embodiments, an effector partner may modulate transcription (e.g., inhibits transcription, increases transcription) of a target nucleic acid. In yet another example, an effector partner may directly or indirectly inhibit, reduce, activate or increase expression of a target nucleic acid. Reverse Transcriptase (RT) Editing Systems
[0148] In some embodiments, systems and methods comprise components or uses of an RT editing system to modify a target nucleic acid. RT editing may also be referred to as precision editing or precise nucleobase editing. In some embodiments, an RT editing system comprises an effector protein and an effector partner comprising an RT editing enzyme. In some embodiments, the effector protein that is linked to the RT editing enzyme. In some embodiments, an RT editing enzyme comprises a polymerase. In some embodiments, an RT editing enzyme comprises a reverse transcriptase. A non-limiting example of a reverse transcriptase is an M-MLV RT enzyme and variants thereof having polymerase activity. In some embodiments, the M-MLV RT enzyme comprises at least one mutation selected from D200N, L603W, T330P, T306K, and W313F relative to wildtype M-MLV RT enzyme. In some embodiments, systems and methods comprise an RT editing enzyme, wherein the RT editing enzyme is not fused or linked to the effector protein. See, e.g. FIG. 3. In some embodiments, the RT editing enzyme comprises a recruiting moiety that recruits the RT editing enzyme to the target nucleic acid. By way of non-limiting example, the RT editing enzyme may comprise a peptide that binds an aptamer, wherein the aptamer is located on a guide RNA, template RNA, or combination thereof. Also, by way of non-limiting example, the RT editing enzyme may be linked to a protein that binds to (or is bound by) the effector protein or a protein linked/fused to the effector protein. By way of non-limiting example, the RT editing enzyme may be linked to an MS2 coat protein that binds an MS2 aptamer, wherein the aptamer is located on a guide RNA, template RNA, or combination thereof. See, e.g. FIG. 3. In some embodiments, an RT editing enzyme may require an RT editing guide RNA (referred to herein as a “pegRNA” or “rtgRNA”) to catalyze editing. Such a pegRNA may be capable of identifying a target nucleotide or target sequence in a target nucleic acid to be edited and encoding a new genetic information that replaces the target nucleotide or target sequence in the target nucleic acid. An RT editing enzyme may require a pegRNA and a guide RNA, such as a single guide RNA, to catalyze the editing. In some embodiments, the RT editing system comprises a template RNA comprising a primer binding sequence that hybridizes to a primer sequence of the dsDNA molecule that is formed when target nucleic acid is cleaved, and a template sequence that is complementary to at least a portion of the target sequence of the dsDNA molecule except for at least one nucleotide. The term “template RNA (retRNA)” as used herein, refers to a nucleic acid comprising: a primer binding sequence and a template sequence. In some embodiments, the primer biding sequence and template binding sequence are linked to an MS2 aptamer. The RT editing enzyme is linked to an MS2 coat protein that is capable of binding the MS2 aptamer, thereby localizing the RT editing enzyme to the nicked DNA. See, e.g., FIG. 3. It is understood that template RNAs may comprise DNA, RNA, or a combination thereof (e.g., RNA with a thymine base). In some instances, the template RNA is linked to a guide RNA via a linker sequence to form an rtgRNA.
[0149] In some embodiments, the template RNA is covalently linked to a guide RNA (referred to herein as an extended guide RNA or rtgRNA). The term, “extended guide RNA (rtgRNA),” as used herein refers to a single nucleic acid molecule comprising (not necessarily in the following order) (1) a guide RNA comprising (a) a protein binding sequence and (b) a spacer sequence; (2) optionally, a linker; and (3) a template RNA (retRNA) comprising (a) a primer binding sequence and (b) a template sequence. In some embodiments, the orientation of the rtgRNA from 5’ to 3’ is: guide nucleic acid, optional linker, and template RNA. In some embodiments, the orientation of the rtgRNA from 5’ to 3’ is: template RNA, linker, and guide RNA. It is understood that extended guide RNAs may comprise DNA, RNA, or a combination thereof (e.g., RNA with a thymine base). The terms rtgRNA and pegRNA are used interchangeably herein. In some embodiments, the template RNA is not covalently linked to a guide RNA.
[0150] In some embodiments, a spacer sequence, a repeat or handle sequence, a primer binding sequence, and a template sequence are comprised in two polynucleotides - the spacer and repeat/handle sequence comprised in a first polynucleotide and the primer binding sequence and the template sequence comprised in a second polynucleotide, referred to herein as a split RNA. See e.g., FIG. 3. System components shown in FIG. 3 are exemplary and non-limiting. For instance, the guide RNA and/or retRNA may comprise additional nucleotides beyond those labeled as spacer, repeat/handle, linker, PBS and RT template (RTT).
[0151] In some embodiments, the first polynucleotide comprises a spacer sequence and a handle sequence (also referred to herein as a scaffold sequence). In some embodiments, the spacer sequence precedes the handle sequence in a 5’ to 3’ direction. See e.g., FIG. 3. In some embodiments, the spacer(s) and scaffold sequence(s) are linked directly to one another. In some embodiments, a linker is present between the spacer(s) and scaffold sequence(s). Linkers may be any suitable linker. In some embodiments, the first polynucleotide comprises a spacer sequence and a handle sequence. In some embodiments, the first polynucleotide is a guide RNA as described herein.
[0152] In some embodiments, the second polynucleotide comprises a primer binding sequence and a template sequence (e.g., an retRNA). In some embodiments, the second polynucleotide further comprises an aptamer that is recognized by a biological tether protein linked to an RT editing enzyme described herein. In some embodiments, the aptamer is an MS2 aptamer (See Said et al (November 2009). "In vivo expression and purification of aptamer-tagged small RNA regulators". Nucleic Acids Research. 37 (20): el33; and Johansson et al (1997). "RNA recognition by the MS2 phage coat protein". Seminars in Virology. 8 (3): 176-185). In some embodiments, the second polynucleotide comprises, from 5’ to 3’, an aptamer sequence, a template sequence, and a primer binding sequence. See FIG. 3. The MS2 aptamer is an exemplary sequence and an RNA sequence with the appropriate secondary structure may also be used instead of the aptamer. In some embodiments, the second polynucleotide is circularized. See FIG. 3.
[0153] In some embodiments, the present disclosure provides a split gRNA system, comprising a first polynucleotide comprising a spacer sequence and a handle sequence (e.g., a gRNA) and a second polynucleotide comprising a primer binding sequence and a template sequence (e.g., an retRNA), and an effector protein (e.g., effector proteins provided in TABLE 1, TABLE 6, and TABLE 8), and an RT editing enzyme (e.g., M-MLV RT enzyme), or nucleic acids encoding the same.
[0154] In some embodiments, at least a portion of the template RNA hybridizes to the target nucleic acid. In some embodiments, the target nucleic acid is a dsDNA molecule. In some embodiments, at least a portion of the template RNA hybridizes to a first strand of the target nucleic acid and at least a portion of the guide RNA hybridizes to a second strand of the target nucleic acid. See e.g., FIG. 3. In some embodiments, the pegRNA comprises: a guide RNA comprising a second region that is bound by the effector protein, and a first region comprising a spacer sequence that is complementary to a target sequence of the dsDNA molecule; and a template RNA comprising a primer binding sequence that hybridizes to a primer sequence of the dsDNA molecule that is formed when target nucleic acid is cleaved, and a template sequence that is complementary to at least a portion of the target sequence of the dsDNA molecule with the exception of at least one nucleotide.
[0155] In some embodiments, at least one nucleotide is incorporated into the target nucleic acid by activity of the RT editing enzyme, thereby modifying the target nucleic acid. In some embodiments, the spacer sequence is complementary to the target sequence on a target strand of the dsDNA molecule. In some embodiments, the spacer sequence is complementary to the target sequence on a non-target strand of the dsDNA molecule. In some embodiments, the primer binding sequence hybridizes to a primer sequence on the non-target strand of the dsDNA molecule. In some embodiments, the primer binding sequence hybridizes to a primer sequence on the target strand of the dsDNA molecule. In some embodiments, the target strand is cleaved. In some embodiments, the non-target strand is cleaved.
Epigenetic Modifying Systems
[0156] In some embodiments, effector partners have enzymatic activity that modifies a nucleic acid, such as a target nucleic acid. In some embodiments, the target nucleic acid may comprise or consist of a ssRNA, dsRNA, ssDNA, or a dsDNA. Examples of enzymatic activity that modifies the target nucleic acid include, but are not limited to: nuclease activity, which comprises the enzymatic activity of an enzyme which allows the enzyme to cleave the phosphodiester bonds between the nucleotide subunits of nucleic acids, such as that provided by a restriction enzyme, or a nuclease (e.g., FokI nuclease); methyltransferase activity such as that provided by a methyltransferase (e.g., Hhal DNA m5c-methyltransferase (M.Hhal), DNA methyltransferase 1 (DNMT1), DNA methyltransferase 3 a (DNMT3a), DNA methyltransferase 3b (DNMT3b), METI, DRM3 (plants), ZMET2, CMT1, CMT2 (plants)); demethylase activity such as that provided by a demethylase (e.g., Ten-Eleven Translocation (TET) dioxygenase 1 (TET1CD), TET1, DME, DML1, DML2, ROS1); DNA repair activity; DNA damage (e.g., oxygenation) activity; deamination activity such as that provided by a deaminase (e.g., a cytosine deaminase enzyme such as rat APOB EC 1); dismutase activity; alkylation activity; depurination activity; oxidation activity; pyrimidine dimer forming activity; integrase activity such as that provided by an integrase and/or resolvase (e.g., Gin invertase such as the hyperactive mutant of the Gin invertase, GinH106Y, human immunodeficiency virus type 1 integrase (IN), Tn3 resolvase); transposase activity; recombinase activity such as that provided by a recombinase (e.g., catalytic domain of Gin recombinase); polymerase activity; ligase activity; helicase activity; photolyase activity; and glycosylase activity.
[0157] In some embodiments, effector partners target a ssRNA, dsRNA, ssDNA, or a dsDNA. In some embodiments, effector partners target ssRNA. Non-limiting examples of effector partners for targeting ssRNA include, but are not limited to, splicing factors (e.g. , RS domains); protein translation components (e.g., translation initiation, elongation, and/or release factors; e.g., eIF4G); RNA methylases; RNA editing enzymes (e.g., RNA deaminases, e.g., adenosine deaminase acting on RNA (ADAR), including A to I and/or C to U editing enzymes); helicases; and RNA-binding proteins.
[0158] It is understood that an effector partner may include an entire protein, or in some embodiments, may include a fragment of the protein (e.g., a functional domain). In some embodiments, the functional domain binds or interacts with a nucleic acid, such as ssRNA, including intramolecular and/or intermolecular secondary structures thereof (e.g., hairpins, stem -loops, etc.). The functional domain may interact transiently or irreversibly, directly, or indirectly. In some embodiments, a functional domain comprises a region of one or more amino acids in a protein that is required for an activity of the protein, or the full extent of that activity, as measured in an in vitro assay. Activities include but are not limited to nucleic acid binding, nucleic acid editing, nucleic acid mutating, nucleic acid modifying, nucleic acid cleaving, protein binding or combinations thereof. The absence of the functional domain, including mutations of the functional domain, would abolish or reduce activity.
[0159] Accordingly, effector partners may comprise a protein or domain thereof selected from: endonucleases (e.g., RNase III, the CRR22 DYW domain, Dicer, and PIN (PilT N-terminus); SMG5 and SMG6; domains responsible for stimulating RNA cleavage (e.g., CPSF, CstF, CFIm and CFIIm); exonucleases such as XRN-1 or Exonuclease T; deadenylases such as HNT3; protein domains responsible for nonsense mediated RNA decay (e.g., UPF1, UPF2, UPF3, UPF3b, RNP SI, Y14, DEK, REF2, and SRml60); protein domains responsible for stabilizing RNA (e.g., PABP); proteins and protein domains responsible for polyadenylation of RNA (e.g., PAP1, GLD-2, and Star- PAP); proteins and protein domains responsible for polyuridinylation of RNA (e.g., CI DI and terminal uridylate transferase); and other suitable domains that affect nucleic acid modifications.
[0160] In some embodiments, effector partner may comprise a chromatin-modifying enzyme. In some embodiments, the effector partner chemically modifies a target nucleic acid, for example by methylating, demethylating, or acetylating the target nucleic acid in a sequence specific or non-specific manner.
Base Editing Systems
[0161] In some embodiments, effector partners edit a nucleobase of a target nucleic acid. Such a effector partner may be referred to as a base editing enzyme. In some embodiments, a base editing enzyme variant that differs from a naturally occurring base editing enzyme, but it is understood that any reference to a base editing enzyme herein also refers to a base editing enzyme variant.
[0162] In some embodiments, a base editor is a system comprising an effector protein and a base editing enzyme. In some embodiments, the base editor comprises a base editing enzyme and an effector protein as independent components. In some embodiments, the base editor comprises a fusion protein comprising a base editing enzyme fused or linked to an effector protein. In some embodiments, the amino terminus of the effector partner is linked to the carboxy terminus of the effector protein by the linker. In some embodiments, the carboxy terminus of the effector partner is linked to the amino terminus of the effector protein by the linker. The base editor may be functional when the effector protein is coupled to a guide nucleic acid. The base editor may be functional when the effector protein is coupled to a target nucleic acid. The guide nucleic acid imparts sequence specific activity to the base editor. By way of non-limiting example, the effector protein may comprise a catalytically inactive effector protein (e.g., a catalytically inactive variant of an effector protein described herein). Also, by way of non-limiting example, the base editing enzyme may comprise deaminase activity. Additional base editors are described herein.
[0163] In some embodiments, base editing enzymes are capable of catalyzing editing (e.g., a chemical modification) of a nucleobase of a nucleic acid molecule, such as DNA or RNA (single stranded or double stranded). In some embodiments, a base editing enzyme, and therefore a base editor, is capable of converting an existing nucleobase to a different nucleobase, such as: an adenine (A) to guanine (G); cytosine (C) to thymine (T); cytosine (C) to guanine (G); uracil (U) to cytosine (C); guanine (G) to adenine (A); hydrolytic deamination of an adenine or adenosine, or methylation of cytosine (e.g., CpG, CpA, CpT or CpC). In some embodiments, base editing enzymes edit a nucleobase on a ssDNA. In some embodiments, base editing enzymes edit a nucleobase on both strands of dsDNA. In some embodiments, base editing enzymes edit a nucleobase of an RNA.
[0164] In some embodiments, a base editing enzyme itself may or may not bind to the nucleic acid molecule containing the nucleobase. In some embodiments, upon binding to its target locus in the target nucleic acid (e.g., a DNA molecule), base pairing between the guide nucleic acid and target strand leads to displacement of a small segment of ssDNA in an “R-loop”. In some embodiments, DNA bases within the R-loop are edited by the base editing enzyme having the deaminase enzyme activity. In some embodiments, base editing systems for improved efficiency in eukaryotic cells comprise a base editing enzyme, and a catalytically inactive effector protein that may generate a nick in the non-edited strand and induce repair of the nonedited strand using the edited strand as a template.
[0165] In some embodiments, a base editing enzyme comprises a deaminase enzyme. Exemplary deaminases are described in US20210198330, WO2021041945,
W02021050571 Al, and WO2020123887, all of which are incorporated herein by reference in their entirety. Exemplary deaminase domains are described WO 2018027078 and WO20 17070632, and each are hereby incorporated in its entirety by reference. Also, additional exemplary deaminase domains are described in Komor et al., Nature, 533, 420-424 (2016); Gaudelli et al., Nature, 551, 464-471 (2017); Komor et al., Science Advances, 3:eaao4774 (2017), and Rees et al., Nat Rev Genet. 2018 Dec;19(12):770-788. doi: 10.1038/s41576-018- 0059-1, which are hereby incorporated by reference in their entirety. In some embodiments, the deaminase functions as a monomer. In some embodiments, the deaminase functions as heterodimer with an additional protein. In some embodiments, base editing enzymes comprise a DNA glycosylase inhibitor (e.g. , an uracil glycosylase inhibitor (UGI) or uracil N-glycosylase (UNG)). In some embodiments, the effector partner is a deaminase, e.g., ADAR1/2, ADAR-2, AID, or any functional variant thereof.
[0166] In some embodiments, the base editor is a cytosine base editor (CBE), wherein the base editing enzyme is a cytosine base editing enzyme. In some embodiments, the cytosine base editing enzyme, and therefore CBE, may convert a cytosine to a thymine. In some embodiments, a cytosine base editing enzyme may accept ssDNA as a substrate but may not be capable of cleaving dsDNA, wherein the CBE comprises a catalytically inactive effector protein. In some embodiments, when bound to its cognate DNA, the catalytically inactive effector protein of the CBE may perform local denaturation of the DNA duplex to generate an R-loop in which the DNA strand not paired with a guide nucleic acid exists as a disordered single-stranded bubble. In some embodiments, the catalytically inactive effector protein generated ssDNA R-loop may enable the CBE to perform efficient and localized cytosine deamination in vitro. In some embodiments, deamination activity is exhibited in a window of about 4 to about 10 base pairs. In some embodiments, the catalytically inactive effector protein presents a target site to the cytosine base editing enzyme in high effective molarity, which may enable the CBE to deaminate cytosines located in a variety of different sequence motifs, with differing efficacies. In some embodiments, the CBE is capable of mediating RNA-programmed deamination of target cytosines in vitro or in vivo. In some embodiments, the cytosine base editing enzyme is a cytidine deaminase. In some embodiments, the cytosine base editing enzyme is a cytosine base editing enzyme described by Koblan et al. (2018) Nature Biotechnology 36:848-846; Komor et al. (2016) Nature 533:420-424; Koblan et al. (2021) “Efficient C»G-to-G»C base editors developed using CRISPRi screens, target-library analysis, and machine learning,” Nature Biotechnology; Kurt et al. (2021) Nature Biotechnology 39:41- 46; Zhao et al. (2021) Nature Biotechnology 39:35-40; and Chen et al. (2021) Nature Communications 12: 1384, all incorporated herein by reference.
[0167] In some embodiments, the effector partner comprises a uracil glycosylase inhibitor (UGI). In some embodiments, the CBE described herein comprises UGI. Base excision repair (BER) of U«G in DNA is initiated by a uracil N-glycosylase (UNG), which recognizes a U»G mismatch and cleaves the glycosidic bond between a uracil and a deoxyribose backbone of DNA. BER results in the reversion of the U»G intermediate created by the cytosine base editing enzyme back to a OG base pair. Accordingly, in some embodiments, the UNG may be inhibited by fusion of a UGI to the effector protein. In some embodiments, the UGI is a small protein from bacteriophage PBS. In some embodiments, the UGI is a DNA mimic that potently inhibits both human and bacterial UNG. In some embodiments, the UGI inhibitor is any protein or polypeptide that inhibits UNG.
[0168] In some embodiments, the CBE described herein may mediate efficient base editing in bacterial cells and moderately efficient editing in mammalian cells, enabling conversion of a OG base pair to a T»A base pair through a U»G intermediate. In some embodiments, the CBE is modified to increase base editing efficiency while editing more than one strand of DNA.
[0169] In some embodiments, the CBE described herein nicks a non-edited DNA strand. In some embodiments, the non-edited DNA strand nicked by the CBE biases cellular repair of a U»G mismatch to favor a U»A outcome, elevating base editing efficiency.
[0170] In some embodiments, a base editor described herein comprising one or more base editing enzymes (e.g., APOBEC1, UGI) that efficiently edits in mammalian cells, while minimizing frequency of non-target indels. In some embodiments, base editors do not comprise a functional fragment of the base editing enzyme. In some embodiments, base editors do not comprise a function fragment of a UGI, where such a fragment may be capable of excising a uracil residue from DNA by cleaving an N-glycosidic bond.
[0171] In some embodiments, the effector partner comprises a non-protein uracil-DNA glycosylase inhibitor (npUGI). In some embodiments, the npUGI is selected from a group of small molecule inhibitors of uracil-DNA glycosylase (UDG), or a nucleic acid inhibitor of UDG. In some embodiments, the npUGI is a small molecule derived from uracil. Examples of small molecule non-protein uracil-DNA glycosylase inhibitors, fusion proteins, and Cas- CRISPR systems comprising base editing activity are described in WO2021087246, which is incorporated by reference in its entirety.
[0172] In some embodiments, the base editor is a cytosine base editor, wherein the based editing enzyme is a cytosine base editing enzyme. In some embodiments, the cytosine base editing enzyme is a cytidine deaminase. In some embodiments, the base editor comprising the cytidine deaminase is generated by ancestral sequence reconstruction as described in WO2019226953, which is hereby incorporated by reference in its entirety. Non-limiting exemplary cytidine deaminases suitable for use with effector proteins described herein include: APOBEC1, APOBEC2, APOBEC3C, APOBEC3D, APOBEC3F, APOBEC3G, APOBEC3H, AP0BEC4, AP0BEC3A, BE1 (AP0BECl-XTEN-dCas9), BE2 (AP0BECl-XTEN-dCas9- UGI), BE3 (APOBECl-XTEN-dCas9(A840H)-UGI), BE3-Gam, saBE3, saBE4-Gam, BE4, BE4-Gam, saBE4, and saBE4-Gam as described in WO2021163587, WO2021087246, WO2021062227, and WO2020123887, which are incorporated herein by reference in their entirety.
[0173] In some embodiments, a base editor is a cytosine to guanine base editor (CGBE), wherein the base editing enzyme is a cytosine to guanine base editing enzyme. In some embodiments, the cytosine to guanine base editing enzyme, and therefore the CGBE, may convert a cytosine to a guanine.
[0174] In some embodiments, a base editor is an adenine base editor (ABE), wherein the base editing enzyme is an adenine base editing enzyme. In some embodiments, the adenine base editing enzyme, and therefore the ABE, may convert an adenine to a guanine. In some embodiments, the adenine base editing enzyme converts an A»T base pair to a G»C base pair. In some embodiments, the adenine base editing enzyme converts a target A»T base pair to G»C in vivo or in vitro. In some embodiments, the adenine base editing enzymes provided herein reverse spontaneous cytosine deamination, which has been linked to pathogenic point mutations. In some embodiments, the adenine base editing enzymes provided herein enable correction of pathogenic SNPs (-47% of disease-associated point mutations). In some embodiments, the adenine comprises exocyclic amine that has been deaminated (e.g., resulting in altering its base pairing preferences). In some embodiments, deamination of adenosine yields inosine. In some embodiments, inosine exhibits the base-pairing preference of guanine in the context of a polymerase active site, although inosine in the third position of a tRNA anticodon is capable of pairing with A, U, or C in mRNA during translation. Non-limiting exemplary adenine base editing enzymes suitable for use with effector proteins described herein include: ABE8e, ABE8.20m, APOBEC3A, Anc APOBEC (a.k.a. AncBE4Max), and BtAPOBEC2. Non-limiting exemplary ABEs suitable for use herein include: ABE7, ABE8.1m, ABE8.2m, ABE8.3m, ABE8.4m, ABE8.5m, ABE8.6m, ABE8.7m, ABE8.8m, ABE8.9m, ABE8.10m, ABE8.11m, ABE8.12m, ABE8.13m, ABE8.14m, ABE8.15m, ABE8.16m, ABE8.17m, ABE8.18m, ABE8.19m, ABE8.20m, ABE8.21m, ABE8.22m, ABE8.23m, ABE8.24m, ABE8.1d, ABE8.2d, ABE8.3d, ABE8.4d, ABE8.5d, ABE8.6d, ABE8.7d, ABE8.8d, ABE8.9d, ABE8.10d, ABE8.11d, ABE8.12d, ABE8.13d, ABE8.14d, ABE8.15d, ABE8.16d, ABE8.17d, ABE8.18d, ABE8.19d, ABE8.20d, ABE8.21d, ABE8.22d, ABE8.23d, and ABE8.24d. In some embodiments, the adenine base editing enzyme is an adenine base editing enzyme described in Chu et al., (2021) The CRISPR Journal 4:2: 169-177, incorporated herein by reference. In some embodiments, the adenine deaminase is an adenine deaminase described by Koblan et al. (2018) Nature Biotechnology 36:848-846, incorporated herein by reference. In some embodiments, the adenine base editing enzyme is an adenine base editing enzyme described by Tran et al. (2020) Nature Communications 11 :4871.
[0175] In some embodiments, the ABE described herein is capable of targeting polyA signals, splice site acceptors, and start codons. In some embodiments, the ABE cannot create stop codons for knock-down.
[0176] In some embodiments, an adenine base editing enzyme is an adenosine deaminase. Non-limiting exemplary adenosine base editors suitable for use herein include ABE9. In some embodiments, the ABE comprises an engineered adenosine deaminase enzyme capable of acting on ssDNA. The engineered adenosine deaminase enzyme may be an adenosine deaminase variant that differs from a naturally occurring deaminase. Relative to the naturally occurring deaminase, the adenosine deaminase variant may comprise one or more amino acid alteration, including a V82S alteration, a T166R alteration, a Y147T alteration, a Y147R alteration, a Q154S alteration, a Y123H alteration, a Q154R alteration, or a combination thereof.
[0177] In some embodiments, the base editor comprises an adenine deaminase (e.g., TadA). In some embodiments, the adenosine deaminase is a TadA monomer (e.g., Tad*7.10, TadA*8 or TadA*9). In some embodiments, the adenosine deaminase is a TadA*8 variant (e.g., any one of TadA*8.1, TadA*8.2, TadA*8.3, TadA*8.4, TadA*8.5, TadA*8.6, TadA*8.7, TadA*8.8, TadA*8.9, TadA*8.10, TadA*8.11, TadA*8.12, TadA*8.13, TadA*8.14, TadA*8.15, TadA*8.16, TadA*8.17, TadA*8.18, TadA*8.19, TadA*8.20, TadA*8.21, TadA*8.22, TadA*8.23, or TadA*8.24 as described in WO2021163587 and W02021050571, which are each hereby incorporated by reference in its entirety). In some embodiments, the base editor comprises TadA.
[0178] In some embodiments, a base editing enzyme is a deaminase dimer. In some embodiments, the ABE comprises the effector protein, the adenine base editing enzyme and the deaminase dimer. In some embodiments, the deaminase dimer comprises an adenosine deaminase. In some embodiments, the deaminase dimer comprises TadA and a suitable adenine base editing enzyme including an: ABE8e, ABE8.20m, APOBEC3A, Anc APOBEC (a.k.a. AncBE4Max), BtAPOBEC2, and variants thereof. In some embodiments, the adenine base editing enzyme is fused to amino-terminus or the carboxy -terminus of TadA.
[0179] In some embodiments, a base editor is an RNA base editor, wherein the base editing enzyme is an RNA base editing enzyme. In some embodiments, the RNA base editing enzyme comprises an adenosine deaminase. In some embodiments, ADAR proteins bind to RNAs and alter their sequence by changing an adenosine into an inosine. In some embodiments, RNA base editors comprise an effector protein that is activated by or binds RNA.
[0180] In some embodiments, base editing enzymes, and therefore base editors, are used for treating a subject having or a subject suspected of having a disease related to a gene of interest. In some embodiments, base editing enzymes, and therefore base editors, are useful for treating a disease or a disorder caused by a point mutation in a gene of interest. In some embodiments, compositions, systems, and methods described herein comprise a base editor and a guide nucleic acid, wherein the base editor comprises an effector protein and a base editing enzyme, and wherein the guide nucleic acid directs the base editor to a sequence in a target gene.
Protein Modifying Systems
[0181] In some embodiments, an effector partner provides enzymatic activity that modifies a protein associated with a target nucleic acid. The protein may be a histone, an RNA binding protein, or a DNA binding protein. Examples of such protein modification activities include: methyltransferase activity, such as that provided by a histone methyltransferase (HMT) (e.g., suppressor of variegation 3-9 homolog 1 (SUV39H1, also known as KMT1A), euchromatic histone lysine methyltransferase 2 (G9A, also known as KMT1C and EHMT2), SUV39H2, ESET/SETDB1, SET1A, SET1B, MLL1 to 5, ASH1, SYMD2, NSD1, DOT1L, Pr-SET7/8, SUV4-20H1, EZH2, RIZ1); demethylase activity such as that provided by a histone demethylase (e.g., Lysine Demethylase 1A (KDM1A also known as LSD1), JHDM2a/b, JMJD2A/JHDM3A, JMJD2B, JMJD2C/GASC1, JMJD2D, JARID1A/RBP2, JARID1B/PLU- 1, JARID1C/SMCX, JARID1D/SMCY, UTX, JMJD3); acetyltransferase activity such as that provided by a histone acetylase transferase (e.g., catalytic core/fragment of the human acetyltransferase p300, GCN5, PCAF, CBP, TAF1, TIP60/PLIP, MOZ/MYST3, MORF/MYST4, HBO1/MYST2, HMOF/MYST1, SRC1, ACTR, Pl 60, CLOCK); deacetylase activity such as that provided by a histone deacetylase (e.g., HDAC1, HDAC2, HDAC3, HDAC8, HDAC4, HDAC5, HDAC7, HDAC9, SIRT1, SIRT2, HDAC11); kinase activity; phosphatase activity; ubiquitin ligase activity; deubiquitinating activity; adenylation activity; deadenylation activity; SUMOylating activity; deSUMOylating activity; ribosylation activity; deribosylation activity; myristoylation activity; and demyristoylation activity.
Gene Expression Modulating Systems
[0182] In some embodiments, effector partners include, but are not limited to, a protein that directly and/or indirectly provides for increased or decreased transcription and/or translation of a target nucleic acid (e.g., a transcription activator or a fragment thereof, a protein or fragment thereof that recruits a transcription activator, a small molecule/drug-responsive transcription and/or translation regulator, a translation-regulating protein, etc.). In some embodiments, effector partners that increase or decrease transcription include a transcription activator domain or a transcription repressor domain, respectively.
[0183] In some embodiments, effector partners activate or increase expression of a target nucleic acid. In some embodiments, effector partners increase expression of the target nucleic acid relative to its expression in the absence of the effector partners. Relative expression, including transcription and RNA levels, may be assessed, quantified, and compared, e.g., by RT-qPCR. In some embodiments, effector partners comprise a transcriptional activator. In some embodiments, the transcriptional activators may promote transcription by: recruitment of other transcription factor proteins; modification of target DNA such as demethylation; recruitment of a DNA modifier; modulation of histones associated with target DNA; recruitment of a histone modifier such as those that modify acetylation and/or methylation of histones; or a combination thereof.
[0184] Non-limiting examples of effector partners that promote or increase transcription include: transcriptional activators such as VP 16, VP64, VP48, VP 160, p65 subdomain (e.g., from NFkB), and activation domain of EDLL and/or TAL activation domain (e.g., for activity in plants); histone lysine methyltransferases such as SET1A, SET1B, MLL1 to 5, ASH1, SYMD2, NSD1; histone lysine demethylases such as JHDM2a/b, UTX, JMJD3; histone acetyltransferases such as GCN5, PCAF, CBP, p300, TAF1, TIP60/PLIP, MOZ/MYST3, MORF/MYST4, SRC1, ACTR, Pl 60, CLOCK; and DNA demethylases such as Ten-Eleven Translocation (TET) dioxygenase 1 (TET1CD), TET1, DME, DML1, DML2, and ROS1; and functional domains thereof. Other non-limiting examples of suitable effector partners include: proteins and protein domains responsible for stimulating translation (e.g., Staufen); proteins and protein domains responsible for (e.g., capable of) modulating translation (e.g., translation factors such as initiation factors, elongation factors, release factors, etc., e.g., eIF4G); proteins and protein domains responsible for stimulation of RNA splicing (e.g., Serine/ Arginine-rich (SR) domains); and proteins and protein domains responsible for stimulating transcription (e.g., CDK7 and HIV Tat).
[0185] In some embodiments, effector partners inhibit or reduce expression of a target nucleic acid. In some embodiments, effector partners reduce expression of the target nucleic acid relative to its expression in the absence of the effector partners. Relative expression, including transcription and RNA levels, may be assessed, quantified, and compared, e.g., by RT-qPCR. In some embodiments, effector partners may comprise a transcriptional repressor. In some embodiments, the transcriptional repressors may inhibit transcription by: recruitment of other transcription factor proteins; modification of target DNA such as methylation; recruitment of a DNA modifier; modulation of histones associated with target DNA; recruitment of a histone modifier such as those that modify acetylation and/or methylation of histones; or a combination thereof.
[0186] Non-limiting examples of effector partners that decrease or inhibit transcription include: transcriptional repressors such as the Kriippel associated box (KRAB or SKD); K0X1 repression domain; the Mad mSIN3 interaction domain (SID); the ERF repressor domain (ERD), the SRDX repression domain (e.g., for repression in plants); histone lysine methyltransferases such as Pr-SET7/8, SUV4-20H1, RIZ1, and the like; histone lysine demethylases such as JMJD2A/JHDM3A, JMJD2B, JMJD2C/GASC1, JMJD2D, JARID1A/RBP2, JARID1B/PLU-1, JARID1C/SMCX, JARID1D/SMCY; histone lysine deacetylases such as HDAC1, HDAC2, HDAC3, HDAC8, HDAC4, HDAC5, HDAC7, HDAC9, SIRT1, SIRT2, HDAC11; DNA methylases such as Hhal DNA m5c- m ethyltransferase (M.Hhal), DNA methyltransferase 1 (DNMT1), DNA methyltransferase 3 a (DNMT3a), DNA methyltransferase 3b (DNMT3b), METI, DRM3 (plants), ZMET2, CMT1, CMT2 (plants); and periphery recruitment elements such as Lamin A, and Lamin B; and functional domains thereof. Other non-limiting examples of suitable effector partners include: proteins and protein domains responsible for repressing translation (e.g., Ago2 and Ago4); proteins and protein domains responsible for repression of RNA splicing (e.g., PTB, Sam68, and hnRNP Al); proteins and protein domains responsible for reducing the efficiency of transcription e.g., FUS (TLS)).
[0187] In some embodiments, fusion proteins activate or increase expression of a target nucleic acid. In some embodiments, fusion proteins inhibit or reduce expression of a target nucleic acid. In some embodiments, fusion proteins are targeted by a guide nucleic acid (e.g, guide RNA) to a specific location in a target nucleic acid and exert locus-specific regulation such as blocking RNA polymerase binding to a promoter (which selectively inhibits transcription activator function), and/or changes a local chromatin status (e.g, when a fusion sequence is used that edits the target nucleic acid or modifies a protein associated with the target nucleic acid). In some embodiments, the modifications are transient (e.g., transcription repression or activation). In some embodiments, the modifications are inheritable. For example, epigenetic modifications made to a target nucleic acid, or to proteins associated with the target nucleic acid, e.g., nucleosomal histones, in a cell, can be observed in a successive generation. [0188] In some embodiments, effector partner comprises an RNA splicing factor. The RNA splicing factor may be used (in whole or as fragments thereof) for modular organization, with separate sequence-specific RNA binding modules and splicing effector domains. In some embodiments, the RNA splicing factors comprise members of the Serine/ Arginine-rich (SR) protein family containing N-terminal RNA recognition motifs (RRMs) that bind to exonic splicing enhancers (ESEs) in pre-mRNAs and C-terminal RS domains that promote exon inclusion. In some embodiments, a hnRNP protein hnRNP Al binds to exonic splicing silencers (ESSs) through its RRM domains and inhibits exon inclusion through a C-terminal Glycine- rich domain. In some embodiments, the RNA splicing factors may regulate alternative use of splice site (ss) by binding to regulatory sequences between two alternative sites. For example, in some embodiments, ASF/SF2 may recognize ESEs and promote the use of intron proximal sites, whereas hnRNP Al may bind to ESSs and shift splicing towards the use of intron distal sites. One application for such factors is to generate ESFs that modulate alternative splicing of endogenous genes, particularly disease associated genes. For example, Bcl-x pre-mRNA produces two splicing isoforms with two alternative 5' splice sites to encode proteins of opposite functions. Long splicing isoform Bcl-xL is a potent apoptosis inhibitor expressed in long-lived postmitotic cells and is up-regulated in many cancer cells, protecting cells against apoptotic signals. Short isoform Bcl-xS is a pro-apoptotic isoform and expressed at high levels in cells with a high turnover rate (e.g., developing lymphocytes). A ratio of the two Bcl-x splicing isoforms is regulated by multiple coo-elements that are located in either core exon region or exon extension region (z.e., between the two alternative 5' splice sites). For more examples, see W02010075303, which is hereby incorporated by reference in its entirety.
Recombinases
[0189] In some embodiments, effector partners comprise a recombinase. In some embodiments, provided herein is a recombinase system comprising effector proteins described herein and the recombinase. In some embodiments, the effector proteins have reduced nuclease activity or no nuclease activity. In some embodiments, the recombinase is a site-specific recombinase.
[0190] In some embodiments, the recombinase system comprises a catalytically inactive effector protein, wherein the recombinase can be a site-specific recombinase. Such systems can be used for site-directed transgene insertion. Non-limiting examples of site-specific recombinases include a tyrosine recombinase (e.g., Cre, Flp or lambda integrase), a serine recombinase (e.g., gamma-delta resolvase, Tn3 resolvase, Sin resolvase, Gin invertase, Hin invertase, Tn5044 resolvase, IS607 transposase and integrase), or mutants or variants thereof. In some embodiments, the recombinase is a serine recombinase. Non-limiting examples of serine recombinases include gamma-delta resolvase, Tn3 resolvase, Sin resolvase, Gin invertase, Hin invertase, Tn5044 resolvase, IS607 transposase, and IS607 integrase. In some embodiments, the site-specific recombinase is an integrase. Non-limiting examples of integrases include:Bxbl, wBeta, BL3, phiR4, Al 18, TGI, MR11, phi370, SPBc, TP901-1, phiRV, FC1, K38, phiBTl, and phiC31. Further discussion and examples of suitable recombinase effector partners are described in US 10,975,392, which is incorporated herein by reference in its entirety. In some embodiments, the fusion protein comprises a linker that links the recombinase to a Cas-CRISPR domain of the effector protein. In some embodiments, the linker is Thr-Ser.
Linkers
[0191] In some embodiments, a linker comprises a bond or molecule that links a first polypeptide to a second polypeptide. Accordingly, in some embodiments, effector proteins, effector partners, or combinations thereof are connected by one or more linkers. The linker may comprise or consist of a covalent bond. The linker may comprise or consist of a chemical group. In some embodiments, the linker comprises an amino acid. In some embodiments, a peptide linker comprises at least two amino acids linked by an amide bond. In general, the linker connects a terminus of the effector protein to a terminus of the effector partner. In some embodiments, carboxy terminus of the effector protein is linked to the amino terminus of the effector partner. In some embodiments, carboxy terminus of the effector partner is linked to the amino terminus of the effector protein. In some embodiments, the effector protein and the effector partner are directly linked by a covalent bond.
[0192] In some embodiments, linkers comprise one or more amino acids. In some embodiments, linker is a protein. In some embodiments, a terminus of the effector protein is linked to a terminus of the effector partner through an amide bond. In some embodiments, a terminus of the effector protein is linked to a terminus of the effector partner through a peptide bond. In some embodiments, linkers comprise an amino acid. In some embodiments, linkers comprise a peptide. In some embodiments, an effector protein is coupled to a effector partner by a linker protein. In some embodiments, the linker may have any of a variety of amino acid sequences. In some embodiments, the linker may comprise a region of rigidity (e.g., beta sheet, alpha helix), a region of flexibility, or any combination thereof. In some embodiments, the linker comprises small amino acids, such as glycine and alanine, that impart high degrees of flexibility. The ordinarily skilled artisan will recognize that design of a peptide conjugated to any desired element may include linkers that are all or partially flexible, such that the linker may include a flexible linker as well as one or more portions that confer less flexible structure. Suitable linkers include proteins of 4 linked amino acids to 40 linked amino acids in length, or between 4 linked amino acids and 25 linked amino acids in length. In some embodiments, linked amino acids described herein comprise at least two amino acids linked by an amide bond.
[0193] Linkers may be produced by using synthetic, linker-encoding oligonucleotides to couple proteins, or may be encoded by a nucleic acid sequence encoding a fusion protein (e.g., an effector protein coupled to an effector partner). In some embodiments, the linker is from 1 to 100 amino acids in length. In some embodiments, the linker is more 100 amino acids in length. In some embodiments, the linker is from 10 to 27 amino acids in length. In some embodiments, linker proteins include glycine polymers (G)n, glycine-serine polymers (including, for example, (GS)n, GSGGSn, GGSGGSn, and GGGSn, where n is an integer of at least one), glycine-alanine polymers, and alanine-serine polymers. In some embodiments, linkers may comprise amino acid sequences including, but not limited to, GGSG, GGSGG, GSGSG, GSGGG, GGGSG, and GSSSG. In some embodiments, the linker comprises one or more repeats a tri-peptide GGS. In some embodiments, the linker is an XTEN linker. In some embodiments, the XTEN linker is an XTEN80 linker. In some embodiments, the XTEN linker is an XTEN20 linker. In some embodiments, the XTEN20 linker has an amino acid sequence of GSGGSPAGSPTSTEEGTSESATPGSG (SEQ ID NO: 916).
[0194] In some embodiments, linkers do not comprise an amino acid. In some embodiments, linkers do not comprise a peptide. In some embodiments, linkers comprise a nucleotide, a polynucleotide, a polymer, or a lipid. In some embodiments, linker may be a polyethylene glycol (PEG), polypropylene glycol (PPG), co-poly(ethylene/propylene) glycol, polyoxyethylene (POE), polyurethane, polyphosphazene, polysaccharides, dextran, polyvinyl alcohol, polyvinylpyrrolidones, polyvinyl ethyl ether, polyacrylamide, polyacrylate, polycyanoacrylates, lipid polymers, chitins, hyaluronic acid, heparin, or an alkyl linker.
[0195] In some embodiments, a linker is recognized and cleaved by a protein described herein. In some embodiments, a linker comprises a recognition sequence that may be recognized and cleaved by the protein. In some embodiments, a guide nucleic acid comprises an aptamer, which may serve a similar function as a linker, bringing an effector protein and an effector partner protein into proximity. The aptamer can functionally connect two proteins (e.g., effector protein, effector partner) by interacting non-covalently with both, thereby bringing both proteins into proximity of the guide nucleic acid. In some embodiments, the first protein and/or the second protein comprise or is covalently linked to an aptamer binding moiety. In some embodiments, the aptamer is a short single stranded DNA (ssDNA) or RNA (ssRNA) molecule capable of being bound be the aptamer binding moiety. In some embodiments, the aptamer is a molecule that is capable of mimicking antibody binding activity and may be classified as a chemical antibody. In some instances, the aptamer described herein refers to artificial oligonucleotides that bind one or more specific molecules. In some embodiments, aptamers exhibit a range of affinities (KD in the pM to pM range) with little or no off-target binding.
Fusion Proteins
[0196] In some embodiments, compositions, systems, and methods comprise a fusion protein or uses thereof. A fusion protein generally comprises an effector protein and an effector partner. In some embodiments, the effector partner comprises a polypeptide or peptide that is fused or linked to the effector protein. In some embodiments, the effector partner is fused to the N- terminus of the effector protein. In some embodiments, the effector partner is fused to the C- terminus of the effector protein.
[0197] In some embodiments, the effector partner is a heterologous protein capable of imparting some function or activity that is not provided by an effector protein. In some embodiments, the effector partner is capable of cleaving or modifying the target nucleic acid, a non-target nucleic acid, or both. In some embodiments, the fusion protein disclosed herein may provide cleavage activity, such as cis cleavage activity, trans cleavage activity, nickase activity, nuclease activity, other activity, or a combination thereof. Fusion proteins disclosed herein may cleave nucleic acids, including single stranded RNA (ssRNA), double stranded DNA (dsDNA), and single-stranded DNA (ssDNA). In some embodiments, fusion proteins cleave the target nucleic acid at the target sequence or adjacent to the target sequence. [In some embodiments, fusion proteins cleave the non-target nucleic acid.
[0198] In some embodiments, the fusion protein complexes with a guide nucleic acid and the complex interacts with the target nucleic acid, a non-target nucleic acid, or both. In some embodiments, the interaction comprises one or more of: recognition of a target adjacent motif (TAM) sequence within the target nucleic acid by the effector protein, hybridization of the guide nucleic acid to the target nucleic acid, modification of the target nucleic acid and/or the non-target nucleic acid by the fusion protein, or combinations thereof. In some embodiments, recognition of a TAM sequence within a target nucleic acid may direct the modification activity of a fusion protein.
[0199] Modification activity of a fusion protein described herein may be cleavage activity, binding activity, insertion activity, substitution activity, and the like. Modification activity of an effector protein may result in: cleavage of at least one strand of a target nucleic acid, deletion of one or more nucleotides of a target nucleic acid, insertion of one or more nucleotides into a target nucleic acid, substitution of one or more nucleotides of a target nucleic acid with an alternative nucleotide, more than one of the foregoing, or any combination thereof. In some embodiments, an ability of a fusion protein to edit a target nucleic acid may depend upon the effector protein being complexed with a guide nucleic acid, the guide nucleic acid being hybridized to a target sequence of the target nucleic acid, the distance between the target sequence and a TAM sequence, or combinations thereof.
IV. Guide Nucleic Acids
[0200] The compositions, systems, and methods of the present disclosure may comprise a guide nucleic acid or a use thereof. Unless otherwise indicated, compositions, systems and methods comprising guide nucleic acids or uses thereof, as described herein and throughout, include DNA molecules, such as expression vectors, that encode a guide nucleic acid. Accordingly, compositions, systems, and methods of the present disclosure comprise a guide nucleic acid or a nucleotide sequence encoding the guide nucleic acid. Guide nucleic acids are also referred to herein as “guide RNA.” In general, the guide RNA comprises a protein binding sequence and a targeting sequence (also may be referred to as a spacer sequence). In general, effector proteins bind the protein binding sequence and the targeting sequence hybridizes to a target nucleic acid, thereby bringing an effector protein within the proximity of the target nucleic acid. In general, effector proteins and guide RNAs complex to form a ribonucleoprotein complex (RNP) that recognizes a motif (e.g., a TAM) proximal to a target sequence of the target nucleic acid.
[0201] A guide nucleic acid, as well as any components thereof (e.g., spacer sequence, protein binding sequence, linker nucleotide sequence, handle sequence, intermediary sequence etc.) may comprise one or more deoxyribonucleotides, ribonucleotides, biochemically or chemically modified nucleotides (e.g., one or more engineered modifications as described herein), or any combinations thereof. Such nucleotide sequences described herein may be described as a nucleotide sequence of either DNA or RNA, however, no matter the form the sequence is described, it is readily understood that such nucleotide sequences can be revised to be RNA or DNA, as needed, for describing a sequence within a guide nucleic acid itself or the sequence that encodes a guide nucleic acid, such as a nucleotide sequence described herein for a vector. Due to WIPO Standard ST.26, the Us are being represented as Ts in RNA in the Sequence Listing provided herein. Similarly, disclosure of the nucleotide sequences described herein also discloses the complementary nucleotide sequence, the reverse nucleotide sequence, and the reverse complement nucleotide sequence, any one of which can be a nucleotide sequence for use in a guide nucleic acid as described herein. In some embodiments, a guide nucleic acid sequence(s) comprises one or more nucleotide alterations at one or more positions in any one of the sequences described herein. Alternative nucleotides can be any one or more of A, C, G, T or U, or a deletion, or an insertion.
[0202] A guide nucleic acid may comprise a naturally occurring sequence. A guide nucleic acid may comprise a non-naturally occurring sequence, wherein the sequence of the guide nucleic acid, or any portion thereof, may be different from the sequence of a naturally occurring guide nucleic acid. A guide nucleic acid of the present disclosure comprises one or more of the following: a) a single nucleic acid molecule; b) a DNA base; c) an RNA base; d) a modified base; e) a modified sugar; f) a modified backbone; and the like. Modifications are described herein and throughout the present disclosure (e.g., in the section entitled “Engineered Modifications”). A guide nucleic acid may be chemically synthesized or recombinantly produced by any suitable methods. Guide nucleic acids and portions thereof may be found in or identified from a sequence present in the genome of a host organism or cell.
[0203] In some embodiments, compositions, systems and methods described herein comprise a single nucleic acid system comprising a guide nucleic acid or a nucleotide sequence encoding the guide nucleic acid, and one or more effector proteins or a nucleotide sequence encoding the one or more effector proteins. The guide nucleic acid of a single nucleic acid system may be referred to as a single guide RNA (sgRNA). In some embodiments, the effector protein comprises an IscB protein or engineered variant thereof, and the sgRNA is referred to as an omega RNA. Similar to CRISPR systems, an omega RNA may comprise a hairpin structure and its coding sequence in the genome is located upstream or downstream of its respective IscB open reading frame (ORF). By way of non-limiting example, an omega RNA coding sequence may be located within Ikb upstream or downstream of the IscB ORF.
[0204] In some embodiments, the compositions, systems, and methods of the present disclosure comprise two or more guide nucleic acids (e.g., 2, 3, 4, 5, 6, 7, 9, 10 or more guide nucleic acids), and/or uses thereof. Multiple guide nucleic acids may target an effector protein to different locations in the target nucleic acid by hybridizing to different target sequences. In some embodiments, a first guide nucleic acid may hybridize within a location of the target nucleic acid that is different from where a second guide nucleic acid may hybridize the target nucleic acid. In some embodiments, the first loci and the second loci of the target nucleic acid may be located at least 1, at least 10, at least 20, at least 30, at least 40, at least 50, at least 60, at least 70, at least 80, at least 90 or at least 100 nucleotides apart. In some embodiments, the first loci and the second loci of the target nucleic acid may be located between 100 and 200, 200 and 300, 300 and 400, 400 and 500, 500 and 600, 600 and 700, 700 and 800, 800 and 900 or 900 and 1000 nucleotides apart. In some embodiments, the first loci and/or the second loci of the target nucleic acid are located in an intron of a gene. In some embodiments, the first loci and/or the second loci of the target nucleic acid are located in an exon of a gene. In some embodiments, the first loci and/or the second loci of the target nucleic acid span an exon-intron junction of a gene. In some embodiments, the first portion and/or the second portion of the target nucleic acid are located on either side of an exon and cutting at both sites results in deletion of the exon. In some embodiments, composition, systems, and methods comprise a donor nucleic acid that may be inserted in replacement of a deleted or cleaved sequence of the target nucleic acid. In some embodiments, compositions, systems, and methods comprising multiple guide nucleic acids or uses thereof comprise multiple effector proteins, wherein the effector proteins may be identical, non-identical, or combinations thereof.
[0205] In some embodiments, a guide nucleic acid comprises at least 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 contiguous nucleotides that are complementary to a eukaryotic sequence. Such a eukaryotic sequence is a nucleotide sequence that is present in a host eukaryotic cell. Such a nucleotide sequence is distinguished from nucleotide sequences present in other host cells, such as prokaryotic cells, or viruses. Said sequences present in a eukaryotic cell can be located in a gene, an exon, an intron, a non-coding (e.g., promoter or enhancer) region, a selectable marker, tag, signal, and the like. In some embodiments, a target sequence is a eukaryotic sequence.
[0206] In some cases, the guide nucleic acid comprises 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 linked nucleosides. In general, a guide nucleic acid comprises at least linked nucleosides. In some embodiments, a guide nucleic acid comprises at least 25 linked nucleosides. A guide nucleic acid may comprise 10 to 50 linked nucleosides. In some cases, the guide nucleic acid comprises or consists essentially of about 12 to about 80 linked nucleosides, about 12 to about 50, about 12 to about 45, about 12 to about 40, about 12 to about 35, about 12 to about 30, about 12 to about 25, from about 12 to about 20, about 12 to about 19 , about 19 to about 20, about 19 to about 25, about 19 to about 30, about 19 to about 35, about 19 to about 40, about 19 to about 45, about 19 to about 50, about 19 to about 60, about 20 to about 25, about 20 to about 30, about 20 to about 35, about 20 to about 40, about 20 to about 45, about 20 to about 50, or about 20 to about 60 linked nucleosides. In some cases, the guide nucleic acid has about 10 to about 60, about 20 to about 50, or about 30 to about 40 linked nucleosides.
[0207] In some embodiments, a length of a guide nucleic acid is about 30 to about 120 linked nucleotides. In some embodiments, the length of a guide nucleic acid is about 40 to about 100, about 40 to about 90, about 40 to about 80, about 40 to about 70, about 40 to about 60, about 40 to about 50, about 50 to about 90, about 50 to about 80, about 50 to about 70, or about 50 to about 60 linked nucleotides. In some embodiments, the length of a guide nucleic acid is about 40, about 45, about 50, about 55, about 60, about 65, about 70 or about 75 linked nucleotides. In some embodiments, the length of a guide nucleic acid is greater than about 15, about 20, about 25, about 30, about 35, about 40, about 45, about 50, about 55, about 60, about 65, about 70 or about 75 linked nucleotides. In some embodiments, the length of a guide nucleic acid is not greater than about 40, about 45, about 50, about 55, about 60, about 65, about 70, about 75, about 80, about 85, about 90, about 95, about 100, about 105, about 110, about 115, about 120, or about 125 linked nucleotides.
[0208] In some embodiments, guide nucleic acids comprise additional elements that contribute additional functionality (e.g., stability, heat resistance, etc.) to the guide nucleic acid. Such elements may be one or more nucleotide alterations, nucleotide sequences, intermolecular secondary structures, or intramolecular secondary structures (e.g., one or more hair pin regions, one or more bulges, etc.).
[0209] In some embodiments, guide nucleic acids comprise one or more linkers connecting different nucleotide sequences as described herein. A linker may comprise 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more nucleotides. A linker may be any suitable linker, examples of which are described herein.
[0210] In some embodiments, a guide nucleic acid, as well as any components thereof (e.g., spacer sequence, repeat sequence, linker) may comprise one or more modified nucleic acids. Modifications can include changing of nucleic acids described herein (e.g., engineered guide nucleic acids) to provide the nucleic acid with a new or enhanced feature, such as improved stability. Such modifications of a nucleic acid include a nucleobase base modification, a backbone modification, a sugar modification, a phosphorothioate internucleotide linkage, or combinations thereof. In some embodiments, the modifications can be of one or more nucleotides, nucleosides, or nucleobases in a nucleic acid. In some embodiments, uridines can be exchanged for pseudouridines (e.g., IN-Methyl-Pseudouridine). In some embodiments, all uridines can be exchanged for IN-Methyl-Pseudouridine. In this application, U can represent uracil or IN-Methyl-Pseudouridine.
Protein Binding Sequence
[0211] In some embodiments, the guide nucleic acid comprises a protein binding sequence, wherein an effector protein described herein binds the protein binding sequence or at least a portion thereof. In general, a protein binding sequence comprises or consists of a handle sequence (also referred to as a scaffold sequence), wherein the handle sequence comprises at least one secondary structure that the effector protein can bind to. By way of non-limiting example, the secondary structure may comprise a hairpin structure, also referred to as a stemloop structure. Engineered proteins in TABLE 1 may bind a protein binding sequence represented by 5’-
GGCUCGUCCAACUGCGGUUGAACGAGCACAGGCUGAGACAUUCGUAAGGCCGA AAGGCCGGACGCACCCUGGGAUUUCCCCAGUCCCCGGAACUGCAUAGCGGAUG CCAGUUGAUGGAGCAAUCUAUCAGAUAAGCCAGGGGGAACAAUCACCUCUCUG UAUCAGAGAGAGUUUUACAAAAGGAGGAACGG-3’ (SEQ ID NO: 858).
[0212] In some embodiments, the protein binding sequence is at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, or at least 98%, at least 99%, or 100% identical to an equal length portion of the sequence of SEQ ID NO: 858. [0213] In some embodiments, the protein binding sequence comprises at least 50, at least 60, at least 70, at least 80, at least 90, at least 100, at least 110, at least 120, at least 130, at least 140, at least 150, at least 160, at least 1700, at least 180, or at least 190 contiguous nucleotides of the sequence of SEQ ID NO: 858.
[0214] Exemplary handle sequences are provided in TABLE 7. In some embodiments, a handle sequence provided herein comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to a sequence selected from TABLE 7. Effector proteins provided in TABLE 1, TABLE 6 and TABLE 8 may bind a handle sequence provided in TABLE 7. In some embodiments, an effector protein selected from any of effector protein 3107961, 3089183, 3092005, 3099541, 3782262, 3745646, 3756102, 370125, and 3778464 in TABLE 6 may bind a handle sequence provided in TABLE 7. In some embodiments, the effector protein comprises an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%. , at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 856, and the handle sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 859 or 860. In some embodiments, the effector protein comprises an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 624, and the handle sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 884. In some embodiments, the effector protein comprises an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 713, and the handle sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 885. In some embodiments, the effector protein comprises an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 833, and the handle sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 886. In some embodiments, the effector protein comprises an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 691, and the handle sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 884. In some embodiments, the effector protein comprises an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 774, and the handle sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 888. In some embodiments, the effector protein comprises an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 179 and 889-910, and the protein binding sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 858. In some embodiments, a guide nucleic acid described herein comprises a spacer sequence and a handle sequence, wherein the spacer sequence precedes the handle sequence in a 5’ to 3’ direction. See e.g., FIG. 3.
Spacer Sequence
[0215] Guide nucleic acids described herein may comprise one or more spacer sequences. The term, “spacer sequence,” may be used interchangeably with “targeting sequence.” While the term, “spacer sequence” may technically belong to CRISPR systems, the term, “spacer sequence,” in the instant disclosure may be used to a guiding or targeting sequence of a guide nucleic acid that is useful with any RNA guided nuclease, including but not limited to, Cas proteins, TnpB proteins, and IscB proteins. In some embodiments, a spacer sequence is capable of hybridizing to a target sequence of a target nucleic acid. Exemplary hybridization conditions are described herein. In some embodiments, the spacer sequence may function to direct an RNP complex comprising the guide nucleic acid to the target nucleic acid for detection and/or modification. The spacer sequence may function to direct a RNP to the target nucleic acid for detection and/or modification. A spacer sequence may be complementary to a target sequence that is adjacent to a TAM that is recognizable by an effector protein described herein.
[0216] In general, the guide nucleic acid comprises a spacer sequence that is at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100% complementary to the target sequence. In some embodiments, the guide nucleic acid comprises at least 10 contiguous nucleotides that are complementary to the target sequence in the target nucleic acid. In some embodiments, guide nucleic acid comprises a spacer sequence that is at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100% complementary to the target sequence. In some embodiments, the target sequence is a eukaryotic sequence.
[0217] The spacer sequence may comprise complementarity with (e.g., hybridize to) a target sequence of a target nucleic acid. In some embodiments, a spacer sequence comprises at least 5 to about 50, at least 5 to about 25, at least about 10 to at least about 25, or at least about 15 to about 25 linked nucleotides. In some cases, the spacer sequence is 15-28 linked nucleosides in length. In some cases, the spacer sequence is 15-26, 15-24, 15-22, 15-20, 15-18, 16-28, 16- 26, 16-24, 16-22, 16-20, 16-18, 17-26, 17-24, 17-22, 17-20, 17-18, 18-26, 18-24, or 18-22 linked nucleosides in length. In some cases, the spacer sequence is 18-24 linked nucleosides in length. In some cases, the spacer sequence is at least 15 linked nucleosides in length. In some cases, the spacer sequence is at least 16, 18, 20, or 22 linked nucleosides in length. In some cases, the spacer sequence comprises at least 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides. In some cases, the spacer sequence is at least 17 linked nucleosides in length. In some cases, the spacer sequence is at least 18 linked nucleosides in length. In some cases, the spacer sequence is at least 20 linked nucleosides in length. In some embodiments, a spacer sequence comprises at least 5 to about 50 contiguous nucleotides that are complementary to a target sequence in a target nucleic acid. In some cases, the spacer sequence comprises at least 15 contiguous nucleobases that are complementary to the target nucleic acid.
[0218] In some embodiments, the spacer sequence is located 5’ of the protein binding sequence. In some embodiments, the spacer sequence is located at the 5’ end of the guide nucleic acid. In some embodiments, the spacer and protein binding sequences are linked directly to one another. In some embodiments, a linker is present between the spacer and protein binding sequences. Linkers may be any suitable linker. In some embodiments, the spacer sequence and the protein binding sequence of the guide nucleic acid are present in separate molecules, which are joined to one another by base pairing interactions.
[0219] It is understood that the sequence of a spacer sequence need not be 100% complementary to that of a target sequence of a target nucleic acid to hybridize or hybridize specifically to the target sequence. Accordingly, in some embodiments, a spacer sequence may comprise a nucleotide sequence that may have partial complementarity with (e.g., hybridize to) an equal length of a target sequence of a target nucleic acid. The guide nucleic acid may comprise at least one uracil between nucleic acid residues 5 to 20 of the spacer sequence that is not complementary to the corresponding nucleoside of the target sequence. The guide nucleic acid may comprise at least one uracil between nucleic acid residues 5 to 9, 10 to 14, or 15 to 20 of the spacer sequence that is not complementary to the corresponding nucleoside of the target sequence. In some cases, the region of the target nucleic acid that is complementary to the spacer sequence comprises an epigenetic modification or a post-transcriptional modification. In some cases, the epigenetic modification comprises acetylation, methylation, or thiol modification.
Linkers for Nucleic Acids
[0220] In some embodiments, a guide nucleic acid for use with compositions, systems, and methods described herein comprises one or more linkers, or a nucleic acid encoding one or more linkers. In some embodiments, the guide nucleic acid comprises a linker connecting a protein binding sequence and a spacer sequence. [0221] In some embodiments, a linker comprises one to ten, one to seven, one to five, one to three, two to ten, two to eight, two to six, two to four, three to ten, three to seven, three to five, four to ten, four to eight, four to six, five to ten, five to seven, six to ten, six to eight, seven to ten, or eight to ten linked nucleotides. In some embodiments, the linker comprises one, two, three, four, five, six, seven, eight, nine, or ten linked nucleotides. In some embodiments, a linker comprises a nucleotide sequence of 5’-GAAA-3’.
Guide Nucleic Acid Modifications
[0222] Guide nucleic acids (e.g., guide nucleic acids) described herein may comprise chemical modifications. In some embodiments, guide nucleic acids are chemically synthesized (as opposed to transcribed). Modifications may provide the nucleic acid with a new or enhanced feature, such as improved stability. Such modifications of a nucleic acid include a base modification, a backbone modification, a sugar modification, or combinations thereof, of one or more nucleotides, nucleosides, or nucleobases in a nucleic acid.
[0223] In some embodiments, nucleic acids (e.g., engineered guide nucleic acids) described herein comprise one or more modifications comprising: 2’0-methyl modified nucleotides, 2’ Fluoro modified nucleotides; locked nucleic acid (LNA) modified nucleotides; peptide nucleic acid (PNA) modified nucleotides; nucleotides with phosphorothioate linkages; a 5’ cap (e.g., a 7-methylguanylate cap (m7G)), phosphorothioates, chiral phosphorothioates, phosphorodithioates, phosphotriesters, aminoalkylphosphotriesters, methyl and other alkyl phosphonates including 3'-alkylene phosphonates, 5'-alkylene phosphonates and chiral phosphonates, phosphinates, phosphoramidates including 3 '-amino phosphoramidate and aminoalkyl phosphoramidates, phosphorodiamidates, thionophosphor amidates, thionoalkylphosphonates , thionoalkylphosphotriesters, selenophosphates and boranophosphates having normal 3'-5' linkages, 2'-5' linked analogs of these, and those having inverted polarity wherein one or more intemucleotide linkages is a 3' to 3', 5' to 5' or 2' to 2' linkage; phosphorothioate and/or heteroatom intemucleoside linkages, such as -CH2-NH-0- CH2-, -CH2-N(CH3)-O-CH2- (known as a methylene (methylimino) or MMI backbone), - CH2-O-N(CH3)-CH2-, -CH2-N(CH3)- N(CH3)-CH2- and -O-N(CH3)-CH2-CH2- (wherein the native phosphodiester intemucleotide linkage is represented as -O-P(=O)(OH)-O-CH2-); morpholino linkages (formed in part from the sugar portion of a nucleoside); morpholino backbones; phosphorodiamidate or other non-phosphodiester intemucleoside linkages; siloxane backbones; sulfide, sulfoxide and sulfone backbones; formacetyl and thioformacetyl backbones; methylene formacetyl and thioformacetyl backbones; riboacetyl backbones; alkene containing backbones; sulfamate backbones; methyleneimino and methylenehydrazino backbones; sulfonate and sulfonamide backbones; amide backbones; other backbone modifications having mixed N, O, S and CH2 component parts; and combinations thereof.
V. Delivery
Vectors and Multiplexed Expression Vectors
[0224] In some embodiments, compositions and systems provided herein comprise a vector system, wherein the vector system comprises one or more vectors. When a vector is described herein, such a vector can be used as a vehicle to introduce one or more molecules of interest into a host cell. A molecule of interest may comprise a polypeptide (e.g., an effector protein), a guide nucleic acid, a donor nucleic acid, a nucleic acid encoding a polypeptide, a nucleic acid encoding an engineered guide or a component thereof. The vector may be part of a vector system, wherein a vector system comprises a library of vectors each encoding one or more component of a composition or system described herein. In some embodiments, components described herein (e.g., an effector protein, a guide nucleic acid, and/or a target nucleic acid) are encoded by the same vector. In some embodiments, components described herein (e.g., an effector protein, a guide nucleic acid, and/or a target nucleic acid) are each encoded by different vectors of the system. For example, vector systems described herein can comprise one or more vectors comprising a polypeptide (e.g., an effector protein), an engineered guide nucleic acid, or a nucleic acid or nucleic acids encoding a polypeptide, engineered guide, a donor nucleic acid, or any combination thereof.
[0225] In some embodiments, compositions and systems provided herein comprise a vector system comprising a polypeptide (e.g., an effector protein, effector partner, or fusion protein) described herein. In some embodiments, compositions and systems provided herein comprise a vector system comprising a guide nucleic acid described herein. In some embodiments, compositions and systems provided herein comprise a vector system comprising a donor nucleic acid described herein.
[0226] In some embodiments, compositions and systems provided herein comprise a vector system encoding a polypeptide (e.g., an effector protein, effector partner, or fusion protein) described herein. In some embodiments, compositions and systems provided herein comprise a vector system encoding a guide nucleic acid described herein. In some embodiments, compositions and systems provided herein comprise a multi-vector system encoding an effector protein and a guide nucleic acid described herein, wherein the guide nucleic acid and the effector protein are encoded by the same or different vectors. In some embodiments, the guide nucleic acid and the effector protein are encoded by different vectors of the system. In some embodiments, a nucleic acid encoding a polypeptide (e.g., an effector protein, effector partner, or fusion protein) comprises an expression vector. In some embodiments, a nucleic acid encoding a polypeptide is a messenger RNA. In some embodiments, an expression vector comprises or encodes an engineered guide nucleic acid.
[0227] In some embodiments, a vector comprises one or more donor nucleic acids as described herein. In some embodiments, the one or more donor nucleic acids comprise at least two donor nucleic acids. In some embodiments, the at least two donor nucleic acids are the same. In some embodiments, the at least two donor nucleic acids are different from each other. In some embodiments, a vector can comprise or encode one or more regulatory elements. Regulatory elements can refer to transcriptional and translational control sequences, such as promoters, enhancers, polyadenylation signals, terminators, protein degradation signals, and the like, that provide for and/or regulate transcription of a non-coding sequence or a coding sequence and/or regulate translation of an encoded polypeptide. In some embodiments, a vector can comprise or encode for one or more additional elements, such as, replication origins, antibiotic resistance (or a nucleic acid encoding the same), a tag (or a nucleic acid encoding the same), selectable markers, and the like.
[0228] Vectors described herein can encode a promoter - a regulatory region on a nucleic acid, such as a DNA sequence, capable of initiating transcription of a downstream (3' direction) coding or non-coding sequence. As used herein, a promoter can be bound at its 3' terminus to a nucleic acid the expression or transcription of which is desired, and extends upstream (5' direction) to include bases or elements necessary to initiate transcription or induce expression, which could be measured at a detectable level. A promoter can comprise a nucleotide sequence, referred to herein as a “promoter sequence”. A promoter sequence can include a transcription initiation site, and one or more protein binding domains responsible for the binding of transcription machinery, such as RNA polymerase. When eukaryotic promoters are used, such promoters can contain “TATA” boxes and “CAT” boxes. Various promoters, including inducible promoters, may be used to drive expression, z.e., transcriptional activation, of the nucleic acid of interest. Accordingly, in some embodiments, the nucleic acid of interest can be operably linked to a promoter.
[0229] Promotors can be any suitable type of promoter envisioned for the compositions, systems, and methods described herein. Examples include constitutively active promoters (e.g., CMV promoter), inducible promoters (e.g., heat shock promoter, tetracycline-regulated promoter, steroid-regulated promoter, metal-regulated promoter, estrogen receptor-regulated promoter, etc.), spatially restricted and/or temporally restricted promoters (e.g., a tissue specific promoter, a cell type specific promoter, etc.), etc. Suitable promoters include, but are not limited to: SV40 early promoter, mouse mammary tumor virus long terminal repeat (LTR) promoter; adenovirus major late promoter (Ad MLP); a herpes simplex virus (HSV) promoter, a cytomegalovirus (CMV) promoter such as the CMV immediate early promoter region (CMVIE), a rous sarcoma virus (RSV) promoter, a human U6 small nuclear promoter (U6), an enhanced U6 promoter, and a human Hl promoter (Hl). By transcriptional activation, it is intended that transcription will be increased above basal levels in the target cell by 10-fold, by 100-fold, or by 1000-fold, or more. In addition, vectors used for providing a nucleic acid encoding an engineered guide nucleic acid and/or an effector protein to a cell may include nucleic acid sequences that encode for selectable markers in the target cells, so as to identify cells that have taken up the engineered guide nucleic acid and/or an effector protein.
[0230] In general, plasmids and vectors described herein comprise at least one promoter. In some embodiments, the length of the promoter is less than about 500, less than about 400, or less than about 300 linked nucleotides. In some embodiments, the length of the promoter is at least 100 linked nucleotides. In some embodiments, the promoters are constitutive promoters. In other embodiments, the promoters are inducible promoters. In some embodiments, the promoter is an inducible promoter that only drives expression of its corresponding gene when a signal is present, e.g., a hormone, a small molecule, a peptide. Non-limiting examples of inducible promoters are the T7 RNA polymerase promoter, the T3 RNA polymerase promoter, the Isopropyl-beta-D-thiogalactopyranoside (IPTG)-regulated promoter, a lactose induced promoter, a heat shock promoter, a tetracycline-regulated promoter (tetracycline-inducible or tetracycline-repressible), a steroid regulated promoter, a metal-regulated promoter, and an estrogen receptor-regulated promoter. In some embodiments, the promoter is an activationinducible promoter, such as a CD69 promoter, as described further in Kulemzin et al., (2019), BMC Med Genomics, 12:44. In additional embodiments, the promoters are prokaryotic promoters (e.g., drive expression of a gene in a prokaryotic cell). In some embodiments, the promoters are eukaryotic promoters, (e.g., drive expression of a gene in a eukaryotic cell). Exemplary promoters include, but are not limited to, CMV, 7SK, EFla, RPBSA, hPGK, EFS, SV40, PGK1, Ubc, human beta actin, CAG, TRE, UAS, Ac5, polyhedron, CaMKIIa, GAL1- 10, TEF1, GDS, ADH1, CaMV35S, Ubi, Hl, U6, CaMV35S, MNDU3, MSCV and HSV TK promoter. In some embodiments, the promoter is CMV. In some embodiments, the promoter is EFla. In some embodiments, the promoter is ubiquitin. In some embodiments, vectors are bicistronic or polycistronic vector (e.g., having or involving two or more loci responsible for generating a protein) having an internal ribosome entry site (IRES) is for translation initiation in a cap-independent manner.
[0231] In some embodiments, a vector described herein is a delivery vector. In some examples, the delivery vector may be a eukaryotic vector, a prokaryotic vector (e.g., a bacterial vector) a viral vector, or any combination thereof. In some embodiments, the delivery vehicle may be a non-viral vector. In some embodiments, the delivery vehicle may be a plasmid. In some embodiments, the plasmid comprises DNA. In some embodiments, the plasmid comprises RNA. In some examples, the plasmid comprises circular double-stranded DNA. In some examples, the plasmid may be linear. In some examples, the plasmid comprises one or more genes of interest and one or more regulatory elements. In some examples, the plasmid comprises a bacterial backbone containing an origin of replication and an antibiotic resistance gene or other selectable marker for plasmid amplification in bacteria. In some examples, the plasmid may be a minicircle plasmid. In some examples, the plasmid contains one or more genes that provide a selective marker to induce a target cell to retain the plasmid. In some examples, the plasmid may be formulated for delivery through injection by a needle carrying syringe. In some examples, the plasmid may be formulated for delivery via electroporation. In some examples, the plasmids may be engineered through synthetic or other suitable means known in the art. For example, in some cases, the genetic elements may be assembled by restriction digest of the desired genetic sequence from a donor plasmid or organism to produce ends of the DNA which may then be readily ligated to another genetic sequence.
[0232] In some embodiments, vectors comprise an enhancer. Enhancers are nucleotide sequences that have the effect of enhancing promoter activity. In some embodiments, enhancers augment transcription regardless of the orientation of their sequence. In some embodiments, enhancers activate transcription from a distance of several kilo basepairs. Furthermore, enhancers are located optionally upstream or downstream of a gene region to be transcribed, and/or located within the gene, to activate the transcription. Exemplary enhancers include, but are not limited to, WPRE; CMV enhancers; the R-U5' segment in LTR of HTLV- I (Mol. Cell. Biol., Vol. 8(1), p. 466-472, 1988); SV40 enhancer; the intron sequence between exons 2 and 3 of rabbit P-globin (Proc. Natl. Acad. Sci. USA., Vol. 78(3), p. 1527-31, 1981); and the genome region of human growth hormone (J Immunol., Vol. 155(3), p. 1286-95, 1995). [0233] In some embodiments, an effector protein (or a nucleic acid encoding same) and/or an engineered guide nucleic acid (or a nucleic acid encoding same) are co-administered with a donor nucleic acid. Co-administration can be contact with a target nucleic acid, administered to a cell, such as a host cell, or administered as method of nucleic acid detection, editing, and/or treatment as described herein, in a single vehicle, such as a single expression vector. In certain embodiments, an effector protein (or a nucleic acid encoding same) and/or an engineered guide nucleic acid (or a nucleic acid encoding same) are not co-administered with donor nucleic acid in a single vehicle. In certain embodiments, an effector protein (or a nucleic acid encoding same), an engineered guide nucleic acid (or a nucleic acid encoding same), and/or donor nucleic acid are administered in one or more or two or more vehicles, such as one or more, or two or more expression vectors.
Administration of a non-viral vector
[0234] In some embodiments, the vector is a non-viral vector, and a physical method or a chemical method is employed for delivery into the somatic cell. Exemplary physical methods include electroporation, gene gun, sonoporation, magnetofection, or hydrodynamic delivery. Exemplary chemical methods include delivery of the recombinant polynucleotide via liposomes such as, cationic lipids or neutral lipids; dendrimers; nanoparticles; or cellpenetrating peptides.
[0235] In some embodiments, a vector is administered as part of a method of nucleic acid detection, editing, and/or treatment as described herein. In some embodiments, a vector is administered in a single vehicle, such as a single expression vector. In some embodiments, at least two of the three components, a nucleic acid encoding one or more effector proteins, one or more donor nucleic acids, and one or more guide nucleic acids or a nucleic acid encoding the one or more guide nucleic acid, are provided in the single expression vector. In some embodiments, components, such as a guide nucleic acid and an effector protein, are encoded by the same vector. In some embodiments, a polypeptide (e.g., an effector protein, effector partner, or fusion protein) (or a nucleic acid encoding same) and/or an engineered guide nucleic acid (or a nucleic acid that, when transcribed, produces same) are not co-administered with donor nucleic acid in a single vehicle. In some embodiments, an effector protein (or a nucleic acid encoding same), an engineered guide nucleic acid (or a nucleic acid that, when transcribed, produces same), and/or donor nucleic acid are administered in one or more or two or more vehicles, such as one or more, or two or more expression vectors.
[0236] In some embodiments, a vector may be part of a vector system. In some embodiments, the vector system comprises a library of vectors each encoding one or more components of a composition or system described herein. In some embodiments, a vector system is administered as part of a method of nucleic acid detection, editing, and/or treatment as described herein, wherein at least two vectors are co-administered. In some embodiments, the at least two vectors comprise different components. In some embodiments, the at least two vectors comprise the same component having different sequences. In some embodiments, at least one of the three components, a nucleic acid encoding one or more effector proteins, one or more donor nucleic acids, and one or more guide nucleic acids or a nucleic acid encoding the one or more guide nucleic acids, or a variant thereof is provided in a different vector. In some embodiments, the nucleic acid encoding the effector protein, and a guide nucleic acid or a nucleic acid encoding the guide nucleic acid are provided in different vectors. In some embodiments, the donor nucleic acid is encoded by a different vector than the vector encoding the effector protein and the guide nucleic acid.
Lipid Particles and Non-viral Vectors
[0237] In some embodiments, compositions and systems provided herein comprise a lipid particle. In some embodiments, a lipid particle is a lipid nanoparticle (LNP). In some embodiments, a lipid or a lipid nanoparticle can encapsulate an expression vector. In some embodiments, a lipid or a lipid nanoparticle can encapsulate the effector protein, the guide nucleic acid, the nucleic acid encoding the effector protein and/or the DNA molecule encoding the guide nucleic acid. LNPs are a non-viral delivery system for gene therapy. LNPs are effective for delivery of nucleic acids. Beneficial properties of LNP include ease of manufacture, low cytotoxicity and immunogenicity, high efficiency of nucleic acid encapsulation and cell transfection, multi-dosing capabilities and flexibility of design (Kulkarni et al., (2018) Nucleic Acid Therapeutics, 28(3): 146-157). In some embodiments, a method can comprise contacting a cell with an expression vector. In some embodiments, contacting can comprise electroporation, lipofection, or lipid nanoparticle (LNP) delivery of an expression vector. In some embodiments, a nucleic acid expression vector is a non-viral vector. In some embodiments, compositions and methods comprise a lipid, polymer, nanoparticle, or a combination thereof, or use thereof, to introduce an effector protein, guide nucleic acid, donor template or any combination thereof to a cell. Non-limiting examples of lipids and polymers are cationic polymers, cationic lipids, or bio-responsive polymers. In some embodiments, the bio-responsive polymer exploits chemi cal -physical properties of the endosomal environment (e.g., pH) to preferentially release the genetic material in the intracellular space.
[0238] In some embodiments, compositions comprise an LNP, an mRNA encoding an effector protein, and optionally a guide RNA. In some embodiments, the mRNA and optionally, the guide RNA are encapsulated in an LNP. The guide RNA may comprise chemical modifications. [0239] In some embodiments, a LNP comprises an outer shell and an inner core. In some embodiments, the outer shell comprises lipids. In some embodiments, the lipids comprise modified lipids. In some embodiments, the modified lipids comprise pegylated lipids. In some embodiments, the lipids comprise one or more of cationic lipids, anionic lipids, ionizable lipids, and non-ionic lipids. In some embodiments, the LNP comprises one or more of N1,N3,N5- tris(3-(didodecylamino)propyl)benzene-l,3,5-tricarboxamide (TT3), 2-dioleoyl-sn-glycero-3- phosphoethanolamine (DOPE), l-palmitoyl-2-oleoylsn-glycero-3-phosphoethanolamine (POPE), l,2-distearoyl-sn-glycero-3-phosphocholine (DSPC), cholesterol (Choi), 1,2- dimyristoyl-sn-glycerol, and methoxypolyethylene glycol (DMG-PEChooo), derivatives, analogs, or variants thereof. In some embodiments, the LNP has a negative net overall charge prior to complexation with one or more of a guide nucleic acid, a nucleic acid encoding the one or more guide nucleic acid, a nucleic acid encoding the polypeptide (e.g., effector protein, effector partner, fusion protein, or combinations thereof), and/or a donor nucleic acid. In some embodiments, the inner core is a hydrophobic core. In some embodiments, the one or more of a guide nucleic acid, the one or more nucleic acid encoding the one or more guide nucleic acid, one or more nucleic acid encoding one or more polypeptides, and/or the one or more donor nucleic acid forms a complex with one or more of the cationic lipids and the ionizable lipids. In some embodiments, the nucleic acid encoding the polypeptide or the nucleic acid encoding the guide nucleic acid is self-replicating.
[0240] In some embodiments, a LNP comprises a lipid composition targeting to a specific organ. In some embodiments, the lipid composition comprises lipids having a specific alkyl chain length that controls accumulation of the LNP in the specific organ (e.g., liver or spleen). In some embodiments, the lipid composition comprises a biomimetic lipid that controls accumulation of the LNP in the specific organ (e.g., brain). In some embodiments, the lipid composition comprises lipid derivatives (e.g., cholesterol derivatives) that controls accumulation of the LNP in a specific cell (e.g., liver endothelial cells, Kupffer cells, hepatocytes).
Viral Vectors
[0241] An expression vector can be a viral vector. In some embodiments, a viral vector comprises a nucleic acid to be delivered into a host cell via a recombinantly produced virus or viral particle. The nucleic acid may be single-stranded or double stranded, linear or circular, segmented or non-segmented. The nucleic acid may comprise DNA, RNA, or a combination thereof. In some embodiments, the expression vector is an adeno-associated viral vector. There are a variety of viral vectors that are associated with various types of viruses, including but not limited to retroviruses (e.g., lentiviruses and y -retroviruses), adenoviruses, arenaviruses, alphaviruses, adeno-associated viruses (AAVs), baculoviruses, vaccinia viruses, herpes simplex viruses and poxviruses. The virus may be a lentivirus. The virus may be an adenovirus. The virus may be a non-replicating virus. The virus may be an adeno-associated virus (AAV). The viral vector may be a retroviral vector. Retroviral vectors may include gamma-retroviral vectors such as vectors derived from the Moloney Murine Leukemia Virus (MoMLV, MMLV, MuLV, or MLV) or the Murine Stem cell Virus (MSCV) genome. Retroviral vectors may include lentiviral vectors such as those derived from the human immunodeficiency virus (HIV) genome. In some embodiments, the viral vector is a chimeric viral vector, comprising viral portions from two or more viruses. In some embodiments, the viral vector is a recombinant viral vector. In some embodiments, the viral vector is a chimeric viral vector. In some embodiments, the chimeric viral vector comprises viral portions from two or more viruses. In some embodiments, the viral vector corresponds to a virus of a specific serotype.
[0242] In some embodiments, the viral vector is an AAV. The AAV may be any AAV known in the art. In some embodiments, a viral particle that delivers a viral vector described herein is an AAV. In some embodiments, the viral vector corresponds to a virus of a specific serotype. In some examples, the serotype is selected from an AAV1 serotype, an AAV2 serotype, AAV3 serotype, an AAV4 serotype, AAV5 serotype, an AAV6 serotype, AAV7 serotype, an AAV8 serotype, an AAV9 serotype, an AAV 10 serotype, an AAV-rhlO serotype, an AAV11 serotype, and an AAV12 serotype. In some embodiments the AAV vector is a recombinant vector, a hybrid AAV vector, a chimeric AAV vector, a self-complementary AAV (scAAV) vector, a single-stranded AAV or any combination thereof. scAAV genomes are generally known in the art and contain both DNA strands which can anneal together to form double-stranded DNA.
[0243] In some embodiments, the AAV vector may be a chimeric AAV vector. In some embodiments, the chimeric AAV vector comprises an exogenous amino acid or an amino acid substitution, or capsid proteins from two or more serotypes. In some examples, a chimeric AAV vector may be genetically engineered to increase transduction efficiency, selectivity, or a combination thereof.
[0244] In some embodiments, the viral vector is a replication-defective viral vector, comprising an insertion of a therapeutic gene inserted in genes essential to the lytic cycle, preventing the virus from replicating and exerting cytotoxic effects. A viral vector provided herein can be derived from or based on any such virus. Often the viral vectors provided herein are an adeno-associated viral vector (AAV vector). Generally, an AAV vector has two inverted terminal repeats (ITRs). According, in some embodiments, the viral vector provided herein comprises two inverted terminal repeats of AAV. The DNA sequence in between the ITRs of an AAV vector provided herein may be referred to herein as the sequence encoding the genome editing tools. These genome editing tools can include, but are not limited to, an effector protein, effector protein modifications (e.g., nuclear localization signal (NLS), polyA tail), guide nucleic acid(s), respective promoter(s), and a donor nucleic acid, or combinations thereof. In general, viral vectors provided herein comprise at least one promotor or a combination of promoters driving expression or transcription of one or more genome editing tools described herein. In some embodiments, the coding region of the AAV vector forms an intramolecular double-stranded DNA template thereby generating an AAV vector that is a self-complementary AAV (scAAV) vector. In general, the sequence encoding the genome editing tools of an scAAV vector has a length of about 2 kb to about 3 kb. The scAAV vector can comprise nucleotide sequences encoding an effector protein, providing guide nucleic acids described herein, and a donor nucleic acid described herein. In some embodiments, the AAV vector provided herein is a self-inactivating AAV vector. In some embodiments, an AAV vector provided herein comprises a modification, such as an insertion, deletion, chemical alteration, or synthetic modification, relative to a wild-type AAV vector.
[0245] In some embodiments, a fusion protein as described herein is inserted into a vector. In some embodiments, the vector comprises one or more promoters, enhancers, ribosome binding sites, RNA splice sites, polyadenylation sites, a replication origin, and/or transcriptional terminator sequences.
Producing AA V Delivery Vectors
[0246] In some embodiments, methods of producing delivery vectors herein comprise packaging a nucleic acid encoding a polypeptide (e.g., an effector protein, effector partner, fusion protein, or combination thereof) and a guide nucleic acid, or a combination thereof, into an AAV vector. In some embodiments, methods of producing the delivery vector comprises, (a) contacting a cell with at least one nucleic acid encoding: (i) a guide nucleic acid; (ii) a Replication (Rep) gene; and (iii) a Capsid (Cap) gene that encodes an AAV capsid protein; (b) expressing the AAV capsid protein in the cell; (c) assembling an AAV particle; and (d) packaging a Cas effector encoding nucleic acid into the AAV particle, thereby generating an AAV delivery vector. In some embodiments, promoters, stuffer sequences, and any combination thereof may be packaged in the AAV vector. In some examples, the AAV vector can package 1, 2, 3, 4, or 5 guide nucleic acids or copies thereof. In some embodiments, the AAV vector comprises inverted terminal repeats, e.g., a 5’ inverted terminal repeat and a 3’ inverted terminal repeat. In some embodiments, the AAV vector comprises a mutated inverted terminal repeat that lacks a terminal resolution site.
[0247] In some embodiments, a hybrid AAV vector is produced by transcapsidation, e.g., packaging an inverted terminal repeat (ITR) from a first serotype into a capsid of a second serotype, wherein the first and second serotypes may be not the same. In some examples, the Rep gene and ITR from a first AAV serotype (e.g., AAN2') may be used in a capsid from a second AAV serotype (e.g., AAV9), wherein the first and second AAV serotypes may be not the same. As a non-limiting example, a hybrid AAV serotype comprising the AAV2 ITRs and AAV9 capsid protein may be indicated AAV2/9. In some examples, the hybrid AAV delivery vector comprises an AAV2/1, AAV2/2, AAV 2/4, AAV2/5, AAV2/8, or AAV2/9 vector.
Producing AA V Particles
[0248] The AAV particles described herein can be referred to as recombinant AAV (rAAV). Often, rAAV particles are generated by transfecting AAV producing cells with an AAV- containing plasmid carrying the sequence encoding the genome editing tools, a plasmid that carries viral encoding regions, i.e., Rep and Cap gene regions; and a plasmid that provides the helper genes such as E1A, E1B, E2A, E4ORF6 and VA. In some embodiments, the AAV producing cells are mammalian cells. In some embodiments, host cells for rAAV viral particle production are mammalian cells. In some embodiments, a mammalian cell for rAAV viral particle production is a COS cell, a HEK293T cell, a HeLa cell, a KB cell, a derivative thereof, or a combination thereof. In some embodiments, rAAV virus particles can be produced in the mammalian cell culture system by providing the rAAV plasmid to the mammalian cell. In some embodiments, producing rAAV virus particles in a mammalian cell can comprise transfecting vectors that express the rep protein, the capsid protein, and the gene-of-interest expression construct flanked by the ITR sequence on the 5’ and 3’ ends. Methods of such processes are provided in, for example, Naso et al., BioDrugs, 2017 Aug;31(4):317-334 and Benskey et al., (2019), Methods Mol Biol., 1937:3-26, each of which is incorporated by reference in their entireties.
[0249] In some embodiments, rAAV is produced in a non-mammalian cell. In some embodiments, rAAV is produced in an insect cell. In some embodiments, an insect cell for producing rAAV viral particles comprises a Sf9 cell. In some embodiments, production of rAAV virus particles in insect cells can comprise baculovirus. In some embodiments, production of rAAV virus particles in insect cells can comprise infecting the insect cells with three recombinant baculoviruses, one carrying the cap gene, one carrying the rep gene, and one carrying the gene-of-interest expression construct enclosed by an ITR on both the 5’ and 3’ end. In some embodiments, rAAV virus particles are produced by the One Bac system. In some embodiments, rAAV virus particles can be produced by the Two Bac system. In some embodiments, in the Two Bac system, the rep gene and the cap gene of the AAV is integrated into one baculovirus virus genome, and the ITR sequence and the gene-of-interest expression construct is integrated into another baculovirus virus genome. In some embodiments, in the One Bac system, an insect cell line that expresses both the rep protein and the capsid protein is established and infected with a baculovirus virus integrated with the ITR sequence and the gene-of-interest expression construct. Details of such processes are provided in, for example, Smith et. al., (1983), Mol. Cell. Biol., 3(12):2156-65; Urabe et al., (2002), Hum. Gene. Then, 1; 13(16): 1935-43; and Benskey etal., (2019), Methods Mol Biol., 1937:3-26, each of which is incorporated by reference in its entirety.
[0250] In some embodiments, the viral particle that delivers the viral vector described herein is an AAV. AAVs are characterized by their serotype. Non-limiting examples of AAV serotypes are AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, scAAV, AAV-rhlO, chimeric or hybrid AAV, or any combination, derivative, or variant thereof.
VI. T ar get Nucleic Acids
[0251] Disclosed herein are compositions, systems, and methods for modifying and detecting target nucleic acids. In some embodiments, the target nucleic acid is a double stranded nucleic acid. In some embodiments, the target nucleic acid is a single stranded nucleic acid. In some embodiments, the target nucleic acid is a double stranded nucleic acid that is prepared into single stranded nucleic acids before or upon contacting a reagent or sample. In some embodiments, the target nucleic acid comprises DNA. In some embodiments, the target nucleic acid comprises RNA. The target nucleic acids include but are not limited to mRNA, rRNA, tRNA, non-coding RNA, long non-coding RNA, and microRNA (miRNA). In some embodiments, the target nucleic acid is complementary DNA (cDNA) synthesized from a single-stranded RNA template in a reaction catalyzed by a reverse transcriptase. In some cases, the target nucleic acid is single-stranded RNA (ssRNA) or mRNA. In some embodiments, the target nucleic acid is a double stranded nucleic acid comprising a target strand and a non-target strand, wherein the target strand comprises a target sequence. In some embodiments, where a target strand comprises a target sequence, at least a portion of the engineered guide nucleic acid is complementary to the target sequence on the target strand. In some embodiments, where the target nucleic acid is a double stranded nucleic acid comprising a target strand and a non-target strand, and wherein the target strand comprises a target sequence, at least a portion of the engineered guide nucleic acid is complementary to the target sequence on the target strand. In some embodiments, a target nucleic acid comprises a TAM as described herein that is located on the non-target strand. Such a TAM described herein, in some embodiments, is adjacent (e.g., within 1, 2, 3, 4 or 5 nucleotides) to the 3’ end of the target sequence on the non-target strand of the double stranded DNA molecule. In certain embodiments, such a TAM described herein is directly adjacent to the 3’ end of a target sequence on the non-target strand of the double stranded DNA molecule, n some embodiments, the target nucleic acid as described in the methods herein does not initially comprise a TAM sequence. However, any target nucleic acid of interest may be generated using the methods described herein to comprise a TAM sequence, and thus be a TAM target nucleic acid. A TAM target nucleic acid, as used herein, refers to a target nucleic acid that has been amplified to insert a TAM sequence that is recognized by an effector protein system.
[0252] In some embodiments, target nucleic acids comprise a mutation. In some embodiments, a composition, system or method described herein can be used to modify a target nucleic acid comprising a mutation such that the mutation is modified to be a wild-type nucleotide or nucleotide sequence. In some embodiments, a composition, system or method described herein can be used to detect a target nucleic acid comprising a mutation. The mutation may be a mutation of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more nucleotides. The mutation can be a deletion of about 5, about 10, about 15, about 20, about 25, about 30, about 35, about 40, about 45, about 50, about 55, about 60, about 65, about 70, about 75, about 80, about 85, about 90, about 95, about 100, about 200, about 300, about 400, about 500, about 600, about 700, about 800, about 900, or about 1000 nucleotides. The mutation can be a deletion of 1 to 5, 5 to 10, 10 to 15, 15 to 20, 20 to 25, 25 to 30, 30 to 35, 35 to 40, 40 to 45, 45 to 50, 50 to 55, 55 to 60, 60 to 65, 65 to 70, 70 to 75, 75 to 80, 80 to 85, 85 to 90, 90 to 95, 95 to 100, 100 to 200, 200 to 300, 300 to 400, 400 to 500, 500 to 600, 600 to 700, 700 to 800, 800 to 900, 900 to 1000, 1 to 50, 1 to 100, 25 to 50, 25 to 100, 50 to 100, 100 to 500, 100 to 1000, or 500 to 1000 nucleotides. The mutation can be in the open reading frame of a target nucleic acid that results in the insertion of at least one amino acid in a protein encoded by the target nucleic acid. The mutation can also be in the open reading frame of a target nucleic acid that results in the deletion of at least one amino acid in a protein encoded by the target nucleic acid. The mutation can be in the open reading frame of a target nucleic acid that results in the substitution of at least one amino acid in a protein encoded by the target nucleic acid. A mutation that results in the deletion, insertion, or substitution of one or more amino acids of a protein encoded by the target nucleic acid can result in misfolding of the protein. The mutation can result in a premature stop codon. The mutation can result in a truncation of the protein.
[0253] In some embodiments, at least a portion of a guide nucleic acid of a composition described herein hybridizes to a region of the target nucleic acid comprising the mutation. In some embodiments, at least a portion of a guide nucleic acid of a composition described herein hybridizes to a region of the target nucleic acid that is within 10 nucleotides, within 50 nucleotides, within 100 nucleotides, or within 200 nucleotides of the mutation. The mutation may be located in a non-coding region or a coding region of a gene.
[0254] In some embodiments, compositions, systems, and methods described herein comprise a target nucleic acid may be responsible for a disease, contain a mutation (e.g., single strand polymorphism, point mutation, insertion, or deletion), be contained in an amplicon, or be uniquely identifiable from the surrounding nucleic acids (e.g., contain a unique sequence of nucleotides). In some embodiments, the target nucleic acid has undergone a modification (e.g., an editing) after contacting with an RNP. In some embodiments, the editing is a change in the sequence of the target nucleic acid. In some embodiments, the change comprises an insertion, deletion, or substitution of one or more nucleotides compared to the target nucleic acid that has not undergone any modification.
[0255] In some embodiments, the mutation is an autosomal dominant mutation. In some embodiments, the mutation is a dominant negative mutation. In some embodiments, the mutation is a loss of function mutation. In some embodiments, the mutation is a single nucleotide polymorphism (SNP). In some embodiments, the SNP is associated with a phenotype of the sample or a phenotype of the organism from which the sample was taken. The SNP, in some cases, is associated with altered phenotype from wild type phenotype. The SNP may be a synonymous substitution or a nonsynonymous substitution. The nonsynonymous substitution may be a missense substitution, or a nonsense point mutation. The synonymous substitution may be a silent substitution. The mutation may be a deletion of one or more nucleotides. Often, the single nucleotide mutation, SNP, or deletion is associated with a disease such as cancer or a genetic disorder. The mutation, such as a single nucleotide mutation, a SNP, or a deletion, may be encoded in the sequence of a target nucleic acid from the germline of an organism or may be encoded in a target nucleic acid from a diseased cell, such as a cancer cell. [0256] In some embodiments, the target nucleic acid comprises a mutation associated with a disease. In some examples, a mutation associated with a disease refers to a mutation whose presence in a subject indicates that the subject is susceptible to, or suffers from, a disease, disorder, condition, or syndrome. In some examples, a mutation associated with a disease refers to a mutation which causes, contributes to the development of, or indicates the existence of the disease, disorder, condition, or syndrome. A mutation associated with a disease can also refer to any mutation which generates transcription or translation products at an abnormal level, or in an abnormal form, in cells affected by a disease relative to a control without the disease. In some embodiments, the mutation causes the disease.
[0257] In some embodiments, the target nucleic acid is from a gene with a mutation associated with a genetic disorder, from a gene whose overexpression is associated with a genetic disorder, from a gene associated with abnormal cellular growth resulting in a genetic disorder, or from a gene associated with abnormal cellular metabolism resulting in a genetic disorder. Nonlimiting examples of diseases associated with genetic mutations are recited in TABLE 4.
[0258] The disease may comprise, at least in part, a cancer, an inherited disorder, an ophthalmological disorder, a neurological disorder, a neurodegenerative disease, a blood disorder, a metabolic disorder, a genetic disorder, an infection, or a combination thereof. In some embodiments, the cancer is a solid cancer (i.e., a tumor). In some embodiments, the cancer is a blood cell cancer, including a leukemia or lymphoma. In some embodiments, the cancer is colon cancer, rectal cancer, renal -cell carcinoma, liver cancer, bladder cancer, cancer of the kidney or ureter, lung cancer, cancer of the small intestine, esophageal cancer, melanoma, bone cancer, pancreatic cancer, skin cancer, brain cancer (e.g., glioblastoma), cancer of the head or neck, melanoma, uterine cancer, ovarian cancer, breast cancer, testicular cancer, cervical cancer, stomach cancer, Hodgkin's Disease, non-Hodgkin's lymphoma, thyroid cancer. The cancer may be a leukemia, such as, by way of non-limiting example, acute myeloid (or myelogenous) leukemia (AML), chronic myeloid (or myelogenous) leukemia (CML), acute lymphocytic (or lymphoblastic) leukemia (ALL), and chronic lymphocytic leukemia (CLL).
[0259] In some embodiments, the target nucleic acid comprises a portion of a gene comprising a mutation associated with cancer, a gene whose overexpression is associated with cancer, a tumor suppressor gene, an oncogene, a checkpoint inhibitor gene, a gene associated with cellular growth, a gene associated with cellular metabolism, or a gene associated with cell cycle. Sometimes, the target nucleic acid encodes a cancer biomarker, such as a prostate cancer biomarker or non-small cell lung cancer. In some cases, the assay may be used to detect “hotspots” in target nucleic acids that may be predictive of lung cancer. In some cases, the target nucleic acid comprises a portion of a nucleic acid that is associated with a hemorrhagic fever. [0260] In some cases, the target nucleic acid is a portion of a nucleic acid from a genomic locus, any DNA amplicon of, a reverse transcribed mRNA, or a cDNA from a locus of at least one of the genes recited in TABLE 5.
[0261] In some cases, the target nucleic acid is a portion of a nucleic acid from a genomic locus, any DNA amplicon of, a reverse transcribed mRNA, or a cDNA from a locus of at least one of: ABL, ACE, AF4/HRX, AKT-2, ALK, ALK/NPM, AML1, AML1/MTG8, APC, ATM, AXLN2, AXL, BAP1, BARD1, BCL-2, BCL-3, BCL- 6, BCR/ ABL, BLM, BMPR1A, BRCA1, BRCA2, BRIP1, c-MYC, CASR, CCR5, CDC73, CDH1, CDK4, CDKN1B, CDKN1C, CDKN2A, CEBPA, CHEK2, CREBBP, CTNNA1, DBL, DEK/CAN, DICER1, DIS3L2, E2A/PBX1, EGFR, ENL/HRX, EPCAM, ERG/TLS, ERBB, ERBB-2, ETS-I, EWS/FLI-1, FH, FKRP, FLCN, FMS, FOS, FPS, GATA2, GCG, GLI, GPC3, GPGSP, GREM1, HER2/neu, H0X11, H0XB13, HRAS, HST, IL-3, INT-2, JAK1, JUN, KIT, KS3, K-SAM, LBC, LCK, EM01, EM02, L-MYC, LYL-1, LYT-10, LYT-10/Cal, MAS, MAX, MDM-2, MEN1, MET, MITF, MLH1, MLL, MOS, MSH1, MSH2, MSH3, MSH6, MTG8/AML1, MUTYH, MYB, MYH11/CBFB, NBN, NEU, NF1, NF2, N-MYC, NTHL1, OST, PALB2, PAX-5, PBX1/E2A, PCDC1, PDGFRA, PHOX2B, PIM-1, PMS2, POLDI, POLE, POTI, PPARG, PRAD-1, PRKAR1A, PTCHI, PTEN, RAD50, RAD51C, RAD51D, RAF, RAR/PML, RAS-H, RAS-K, RAS-N, RBI, RECQL4, REL/NRG, RET, RH0M1, RH0M2, ROS, RUNX1, SDHA, SDHAF, SDHAF2, SDHB, SDHC, SDHD, SET/CAN, SIS, SKI, SMAD4, SMARCA4, SMARCB1, SMARCE1, SRC, STK11, SUFU, TALI, TAL2, TAN-1, TIAM1, TERC, TERT, TIMP3, TMEM127, INF, TP53, TRAC, TSC1, TSC2, TRK, VHL, WRN, and WT1. Non-limiting examples of oncogenes are KRAS, NRAS, BRAF, MYC, CTNNB1, and EGFR. In some embodiments, the oncogene is a gene that encodes a cyclin dependent kinase (CDK). Nonlimiting examples of CDKs are Cdkl, Cdk4, Cdk5, Cdk7, Cdk8, Cdk9, Cdkll and CDK20. Non-limiting examples of tumor suppressor genes are TP53, RBI, and PTEN. Any region of the aforementioned gene loci may be probed for a mutation or deletion using the compositions and methods disclosed herein. For example, in the EGFR gene locus, the compositions and methods for detection disclosed herein may be used to detect a single nucleotide polymorphism or a deletion.
[0262] In some cases, the target nucleic acid comprises a portion of a nucleic acid from a genomic locus, any DNA amplicon of, a reverse transcribed mRNA, or a cDNA from a locus of at least one of: TRAC, B2M, PD1, PCSK9, DNMT1, HPRT1, RPL32P3, CCR5, FANCF, GRIN2B, EMX1, AAVS1, ALKBH5, CLTA, CDK11, CTNNB1, AXIN1, LRP6, TBK1, BAP1, TLES, PPM1A, BCL2L2, SUFU, RICTOR, VPS35, TOPI, SIRT1, PTEN, MMD, PAQR8, H2AX, P0U5F1, 0CT4, SYS1, ARFRP1, TSPAN14, EMC2, EMC3, SEL1L, DERL2, UBE2G2, UBE2J1, an HRDl.
[0263] In some embodiments, the method for treating a disease comprises modifying at least one gene associated with the disease or modifying expression of the at least one gene such that the disease is treated. In some embodiments, the disease is Alzheimer’s disease and the gene is selected from APP, BACE-1, PSD95, MAPT, PSEN1, PSEN2, and APOEe4. In some embodiments, the disease is congenital muscular dystrophy 1A (MDC1A) and the gene is LAMA1 or LAMA2. In some embodiments, the disease is Ullrich Congenital Muscular Dystrophy (UCMD) and the gene is selected from COL6A1, COL6A2 and COL6A3. In some embodiments, the disease is Limb Girdle Muscular Dystrophies (LGMD1B, LGMD2A, LGMD2B) and the gene is selected from EMNA, DYSF, and CAPN3. In some embodiments, the disease is Nemaline Myopathy and the gene is selected from ACTA1, NEB, TPM2, TPM3, TNNT1, TNNT3, TNNI2 and EM0D3.
[0264] In some embodiments, the disease is Parkinson’s disease and the gene is selected from SNCA, GDNF, and LRRK2. In some embodiments, the disease comprises Centronuclear myopathy and the gene is DNM2. In some embodiments, the disease is Huntington's disease and the gene is HTT. In some embodiments, the disease is Alpha-1 antitrypsin deficiency (AATD) and the gene is SEBPINAE In some embodiments, the disease is amyotrophic lateral sclerosis (ALS) and the gene is selected from SOD1, FUS, C9ORF72, ATXN2, TARDBP, and CHCHD10. In some embodiments, the disease comprises Alexander Disease and the gene is GFAP. In some embodiments, the disease comprises anaplastic large cell lymphoma and the gene is CD30. In some embodiments, the disease comprises Angelman Syndrome and the gene is UBE3A. In some embodiments, the disease comprises Calcific Aortic Stenosis and the gene is Apo(a). In some embodiments, the disease comprises CD3Z-associated primary T-cell immunodeficiency and the gene is CD3Z or CD247. In some embodiments, the disease comprises CD18 deficiency and the gene is ITGB2. In some embodiments, the disease comprises CD40L deficiency and the gene is CD40L. In some embodiments, the disease comprises CNS trauma and the gene is VEGF. In some embodiments, the disease comprises coronary heart disease and the gene is selected from FGA, FGB, and FGG. In some embodiments, the disease comprises MECP2 Duplication syndrome and Rett syndrome and the gene is MECP2. In some embodiments, the disease comprises a bleeding disorder (coagulation) and the gene is FXI. In some embodiments, the disease comprises fragile X syndrome and the gene is FMRP In some embodiments, the disease comprises Fuchs Corneal Dystrophy and the gene is selected from ZEB1, SLC4A11, and LOXHD1. In some embodiments, the disease comprises GM2-Gangliosidoses (e.g., Tay Sachs Disease, Sandhoff disease) and the gene is selected from HEXA and HEXB. In some embodiments, the disease comprises Hearing loss disorders and the gene is DFNA36. In some embodiments, the disease is Pompe disease, including infantile onset Pompe Disease (IOPD) and late onset Pompe Disease (LOPD) and the gene is GAA. In some embodiments, the disease is Retinitis pigmentosa and the gene is selected from PDE6B, RHO, RP1, RP2, RPGR, PRPH2, IMPDH1, PRPF31, CRBI, PRPF8, TULP1, CA4, HPRPF3, ABCA4, EYS, CERKL, FSCN2, TOPORS, SNRNP200, PRCD, NR2E3, MERTK, USH2A, PR0M1, KLHL7, CNGBI, TTC8, ARL6, DHDDS, BESTI, LRAT, SPARA7, CRX, CLRN1, RPE65, and WDR19. In some embodiments, the disease comprises Leber Congenital Amaurosis Type 10 and the gene is CEP290. In some embodiments, the disease is cardiovascular disease and/or lipodystrophies and the gene is selected from ABCG5, ABCG8, AGP, ANGPTL3, APOCIH, APOA1, APOL1, ARH, CDKN2B, CFB, CXCLI2, FXI, FXH, GATA-4, MIAS, MKL2, MTHFD1L, MYH7, NKX2-5, NOTCH1, PKK, PCSK9, PSRC1, SMAD3, and TTR. In some embodiments, the disease is cardiovascular disease and/or lipodystrophies and the gene ANGPTL3. In some embodiments, the disease is cardiovascular disease and/or lipodystrophies and the gene is PCSK9. In some embodiments, the disease is cardiovascular disease and/or lipodystrophies and the gene is TTR. In some embodiments, the disease is severe hypertriglyceridemia (SHTG) and the gene is APOCHI or ANGPTL4. In some embodiments, the disease comprises acromegaly and the gene is GHR. In some embodiments, the disease comprises acute myeloid leukemia and the gene is CD22. In some embodiments, the disease is diabetes and the gene is GCGR. In some embodiments, the disease is NAFLD/NASH and the gene is selected from HSD17B13, PSD3, GPAM, CIDEB, DGAT2 and PNPLA3. In some embodiments, the disease is NASH/cirrhosis and the gene is MARC1. In some embodiments, the disease is cancer and the gene is selected from STAT3, YAP1, FOXP3, AR (Prostate cancer), and IRF4 (multiple myeloma). In some embodiments, the disease is cystic fibrosis and the gene is CFTR. In some embodiments, the disease is Duchenne Muscular Dystrophy and the gene is DMD. In some embodiments, the disease is ornithine transcarbamylase deficiency (OTCD) and the gene is OTC. In some embodiments, the disease is congenital adrenal hyperplasia (CAH) and the gene is CYP21 A2. In some embodiments, the disease is atherosclerotic cardiovascular disease (ASCVD) and the gene is LPA. In some embodiments, the disease is hepatitis B virus infection (CHB) and the gene is HBV covalently closed circular DNA (cccDNA). In some embodiments, the disease is citrullinemia type I and the gene is ASS1. In some embodiments, the disease is citrullinemia type I and the gene is SLC25A13. In some embodiments, the disease is citrullinemia type I and the gene is ASS1. In some embodiments, the disease is arginase-1 deficiency and the gene is ARG1. In some embodiments, the disease is carbamoyl phosphate synthetase I deficiency and the gene is CPS1. In some embodiments, the disease is argininosuccinic aciduria and the gene is ASL. In some embodiments, the disease comprises angioedema and the gene is PKK. In some embodiments, the disease comprises thalassemia and the gene is TMPRSS6. In some embodiments, the disease comprises achondroplasia and the gene is FGFR3. In some embodiments, the disease comprises Cri du chat syndrome and the gene is selected from CTNND2. In some embodiments, the disease comprises sickle cell anemia and the gene is Beta globin gene. In some embodiments, the disease comprises Alagille Syndrome and the gene is selected from JAG1 and N0TCH2. In some embodiments, the disease comprises Charcot Marie Tooth Disease and the gene is selected from PMP22 and MFN2. In some embodiments, the disease comprises Crouzon syndrome and the gene is selected from FGFR2, FGFR3, and FGFR3. In some embodiments, the disease comprises Dravet Syndrome and the gene is selected from SCN1A and SCN2A. In some embodiments, the disease comprises Emery-Dreifuss syndrome and the gene is selected from EMD, LMNA, SYNE1, SYNE2, FHL1, and TMEM43. In some embodiments, the disease comprises Factor V Leiden Thrombophilia and the gene is F5. In some embodiments, the disease is fabry disease and the gene is GLA. In some embodiments, the disease is facioscapulohumeral muscular dystrophy (FSHD) and the gene is FSHDE In some embodiments, the disease comprises Fanconi anemia and the gene is selected from FANCA, FANCB, FANCC, FANCD1, FANCD2, FANCE, FANCF, FANCG, FANCI, FANCJ, FANCY, FANCM, FANCN, FANCP, FANCS, RAD51C, and XPF. In some embodiments, the disease comprises Familial Creutzfeld-Jakob Disease and the gene is PRNP. In some embodiments, the disease comprises Familial Mediterranean Fever and the gene isMEFV. In some embodiments, the disease comprises Friedreich's ataxia and the gene is FXN. In some embodiments, the disease comprises Gaucher disease and the gene is GBA. In some embodiments, the disease comprises human papilloma virus (HPV) infection and the gene is HPVE7. In some embodiments, the disease comprises Hemochromatosis and the gene G HFE, optionally comprising a C282Y mutation. In some embodiments, the disease comprises Hemophilia A and the gene is FVIII. In some embodiments, the disease is hereditary angioedema and the gene is SERPING1 or KLKB1. In some embodiments, the disease comprises histiocytosis and the gene is CD1. In some embodiments, the disease comprises immunodeficiency 17 and the gene is CD3D. In some embodiments, the disease comprises immunodeficiency 13 and the gene is CD4. In some embodiments, the disease comprises Common Variable Immunodeficiency and the gene is selected from CD19 and CD81. In some embodiments, the disease comprises Joubert syndrome and the gene is selected from INPP5E, TMEM216, AHI1, NPHP1, CEP290, TMEM67, RPGRIP1L, ARE13B, CC2D2A, 0FD1, TMEM138, TCTN3, ZNF423, and AMRC9. In some embodiments, the disease comprises leukocyte adhesion deficiency and the gene is CD18. In some embodiments, the disease comprises Li-Fraumeni syndrome and the gene is TP53. In some embodiments, the disease comprises lymphoproliferative syndrome and the gene is CD27. In some embodiments, the disease comprises Lynch syndrome and the gene is selected from MSH2, MLH1, MSH6, PMS2, PMS1, TGFBR2, and MLH3. In some embodiments, the disease comprises mantle cell lymphoma and the gene is CD5. In some embodiments, the disease comprises Marfan syndrome and the gene is FBNE In some embodiments, the disease comprises mastocytosis and the gene is CD2. In some embodiments, the disease comprises methylmalonic acidemia and the gene is selected from MMAA, MMAB, and MUT. In some embodiments, the disease is mycosis fungoides and the gene is CD7. In some embodiments, the disease is myotonic dystrophy and the gene is selected from CNBP and DMPK. In some embodiments, the disease comprises neurofibromatosis and the gene is selected from NF1, and NF2. In some embodiments, the disease comprises osteogenesis imperfecta and the gene is selected from COL1A1, COL1A2, and IFITM5. In some embodiments, the disease is non-small cell lung cancer and the gene is selected from KRAS, EGFR, ALK, METexl4, BRAF V600E, ROS1, RET, an NTRK. In some embodiments, the disease comprises Peutz-Jeghers syndrome and the gene is STK1P In some embodiments, the disease comprises polycystic kidney disease and the gene is selected from PKD1 and PKD2. In some embodiments, the disease comprises Severe Combined Immune Deficiency and the gene is selected from IL7R, RAG1, JAK3. In some embodiments, the disease comprises PRKAG2 cardiac syndrome and the gene is PRKAG2. In some embodiments, the disease comprises Spinocerebellar ataxia and the gene is selected from ATXN1, ATXN2, ATXN3, PLEKHG4, SPTBN2, CACNA1A, ATXN7, ATXN8OS, ATXN10, TTBK2, PPP2R2B, KCNC3, PRKCG, ITPR1, TBP, KCND3, and FGF14. In some embodiments, the disease is thrombophilia due to antithrombin III deficiency and the gene is SERPINCL In some embodiments the disease is spinal muscular atrophy and the gene is SMN1. In some embodiments, the disease comprises Usher Syndrome and the gene is selected from MY07A, USH1C, CDH23, PCDH15, USH1G, USH2A, GPR98, DFNB31, and CLRN1. In some embodiments, the disease comprises von Willebrand disease and the gene is VWF. In some embodiments, the disease comprises Waardenburg syndrome and the gene is selected from PAX3, MITF, WS2B, WS2C, SNAI2, EDNRB, EDN3, and SOXIO. In some embodiments, the disease comprises Wiskott-Aldrich Syndrome and the gene is WAS. In some embodiments, the disease comprises von Hippel-Lindau disease and the gene is VHL. In some embodiments, the disease comprises Wilson disease and the gene is ATP7B. In some embodiments, the disease comprises Zellweger syndrome and the gene is selected from PEX1, PEX2, PEX3, PEX5, PEX6, PEX10, PEX12, PEX13, PEX14, PEX16, PEX19, and PEX26. In some embodiments, the disease comprises infantile myofibromatosis and the gene is CD34. In some embodiments, the disease comprises platelet glycoprotein IV deficiency and the gene is CD36. In some embodiments, the disease comprises immunodeficiency with hyper-IgM type 3 and the gene is CD40. In some embodiments, the disease comprises hemolytic uremic syndrome and the gene is CD46. In some embodiments, the disease comprises complement hyperactivation, angiopathic thrombosis, or protein-losing enteropathy and the gene is CD55. In some embodiments, the disease comprises hemolytic anemia and the gene is CD59. In some embodiments, the disease comprises calcification of joints and arteries and the gene is CD73. In some embodiments, the disease comprises immunoglobulin alpha deficiency and the gene is CD79A. In some embodiments, the disease comprises C syndrome and the gene is CD96. In some embodiments, the disease comprises hairy cell leukemia and the gene is CD123. In some embodiments, the disease comprises histiocytic sarcoma and the gene is CD163. In some embodiments, the disease comprises autosomal dominant deafness and the gene is CD164. In some embodiments, the disease comprises immunodeficiency 25 and the gene is CD247. In some embodiments, the disease comprises methymalonic acidemia due to transcobalamin receptor defect and the gene is CD320.
[0265] In some embodiments, treatment of a disease comprises administration of a gene therapy. In some embodiments, a gene therapy comprises use of a vector to introduce a functional gene or transgene. In some embodiments, vectors comprise nonviral vectors, including cationic polymers, cationic lipids, or bio-responsive polymers. In some embodiments, the bio-responsive polymer exploits chemi cal -physical properties of the endosomal environment (e.g., pH) to preferentially release the genetic material in the intracellular space. In some embodiments, vectors comprise viral vectors, including retroviruses, adenoviruses, adeno-associated viruses, and herpes simplex viruses. In some embodiments, the vector comprises a replication-defective viral vector, comprising an insertion of a therapeutic gene inserted in genes essential to the lytic cycle, preventing the virus from replicating and exerting cytotoxic effects. Methods of gene therapy are described in more detail in Ingusci et al., “Gene Therapy Tools for Brain Diseases". Front. Pharmacol. 10:724 (2019) which is hereby incorporated by reference in its entirety. [0266] The target nucleic acid may be from any organism, including, but not limited to, a bacterium, a virus, a parasite, a protozoon, a fungus, a mammal, a plant, and an insect. As another non-limiting example, the target nucleic acid may be responsible for a disease, contain a mutation (e.g., single strand polymorphism, point mutation, insertion, or deletion), be contained in an amplicon, or be uniquely identifiable from the surrounding nucleic acids (e.g., contain a unique sequence of nucleotides). In some embodiments, the target nucleic acid is from a bacteria. In some embodiments, the bacteria is Acholeplasma laidlctw ii. Brucella abortus, Chlamydia psittaci, Chlamydia trachomatis, Cryptococcus neoformans, Escherichia coli, Legionella pneumophila, Lyme disease spirochetes, methicillin-resistant Staphylococcus aureus, Mycobacterium leprae, Mycobacterium tuberculosis, Mycoplasma arginini, Mycoplasma arthritidis, Mycoplasma genitalium, Mycoplasma hyorhinis, Mycoplasma or ale, Mycoplasma pneumoniae, Mycoplasma salivarium, Neisseria gonorrhoeae, Neisseria meningitidis, Pneumococcus, Pseudomonas aeruginosa, Streptococcus agalactiae, Streptococcus pyogenes, Treponema pallidum, or any combination thereof.
[0267] In some embodiments, the target nucleic acid is from a virus. In some embodiments, the virus is adenovirus, blue tongue virus, chikungunya, coronavirus (e.g. SARS-CoV-2), cytomegalovirus, Dengue virus, Ebola, Epstein-Barr virus, feline leukemia virus, Hemophilus influenzae B, Hepatitis Virus A, Hepatitis Virus B, Hepatitis Virus C, herpes simplex virus I, herpes simplex virus II, human papillomavirus (HPV) including HP VI 6 and HP VI 8, human serum parvo-like virus, human T-cell leukemia viruses, immunodeficiency virus (e.g HIV), influenza virus, lymphocytic choriomeningitis virus, measles virus, mouse mammary tumor virus, mumps virus, murine leukemia virus, polio virus, rabies virus, Reovirus, respiratory syncytial virus (RSV), rubella virus, Sendai virus, simian virus 40, Sindbis virus, varicellazoster virus, vesicular stomatitis virus, wart virus, West Nile virus, yellow fever virus, or any combination thereof. In some cases, the target nucleic acid comprises a portion of a nucleic acid that is associated with a hemorrhagic fever.
[0268] In some embodiments, the target nucleic acid is from a parasite. In some embodiments, the parasite is a helminth, an annelid, a platyhelminth, a nematode, or a thorny-headed worms. In some embodiments, the parasite is Babesia bovis, Echinococcus granulosus, Eimeria tenella, Leishmania tropica, Mesocestoides corti, Onchocerca volvulus, Plasmodium falciparum, Plasmodium vivax, Schistosoma japonicum, Schistosoma mansoni, Schistosoma spp., Taenia hydatigena, Taenia ovis, Taenia saginata, Theileria parva, Toxoplasma gondii, Toxoplasma spp., Trichinella spiralis, Trichomonas vaginalis, Trypanosoma brucei, Trypanosoma cruzi, Trypanosoma rangeli, Trypanosoma rhodesiense, Balantidium coli, Entamoeba histolytica, Giardia spp., Isospora spp., Trichomonas spp., or any combination thereof.
[0269] In some embodiments, the target nucleic acid comprises a nucleic acid sequence from a pathogen responsible for a disease. Non-limiting examples of pathogens are bacteria, a virus and a fungus. The target nucleic acid, in some embodiments, is a portion of a nucleic acid from a sexually transmitted infection or a contagious disease. In some embodiments, the target nucleic acid is a portion of a nucleic acid from a genomic locus, or any DNA amplicon, such as a reverse transcribed mRNA or a cDNA from a gene locus, a transcribed mRNA, or a reverse transcribed cDNA from a gene locus in at least one of: human immunodeficiency virus (HIV), human papillomavirus (HPV), chlamydia, gonorrhea, syphilis, trichomoniasis, sexually transmitted infection, malaria, Dengue fever, Ebola, chikungunya, and leishmaniasis. Pathogens include viruses, fungi, helminths, protozoa, malarial parasites, Plasmodium parasites, Toxoplasma parasites, and Schistosoma parasites. Helminths include roundworms, heartworms, and phytophagous nematodes, flukes, Acanthocephala, and tapeworms. Protozoan infections include infections from Giardia spp., Trichomonas spp., African trypanosomiasis, amoebic dysentery, babesiosis, balantidial dysentery, Chaga's disease, coccidiosis, malaria and toxoplasmosis. Examples of pathogens such as parasitic/protozoan pathogens include, but are not limited to: Plasmodium falciparum, P. vivax, Trypanosoma cruzi and Toxoplasma gondii . Fungal pathogens include, but are not limited to Cryptococcus neoformans, Histoplasma capsulatum, Coccidioides immitis, Blastomyces dermatitidis, Chlamydia trachomatis, and Candida albicans. Pathogenic viruses include but are not limited to coronavirus (e.g., SARS- CoV-2); immunodeficiency virus (e.g., HIV); influenza virus; dengue; West Nile virus; herpes virus; yellow fever virus; Hepatitis Virus C; Hepatitis Virus A; Hepatitis Virus B; papillomavirus; and the like. Pathogens include, e.g., HIV virus, Mycobacterium tuberculosis, Streptococcus agalactiae, methicillin-resistant Staphylococcus aureus, Legionella pneumophila, Streptococcus pyogenes, Escherichia coli, Neisseria gonorrhoeae, Neisseria meningitidis, Pneumococcus, Cryptococcus neoformans, Histoplasma capsulatum, Hemophilus influenzae B, Treponema pallidum, Lyme disease spirochetes, Pseudomonas aeruginosa, Mycobacterium leprae, Brucella abortus, rabies virus, influenza virus, cytomegalovirus, herpes simplex virus I, herpes simplex virus II, human serum parvo-like virus, respiratory syncytial virus (RSV), M. genitalium, T. vaginalis, varicella-zoster virus, hepatitis B virus, hepatitis C virus, measles virus, adenovirus, human T-cell leukemia viruses, Epstein-Barr virus, murine leukemia virus, mumps virus, vesicular stomatitis virus, Sindbis virus, lymphocytic choriomeningitis virus, wart virus, blue tongue virus, Sendai virus, feline leukemia virus, Reovirus, polio virus, simian virus 40, mouse mammary tumor virus, dengue virus, rubella virus, West Nile virus, Plasmodium falciparum, Plasmodium vivax, Toxoplasma gondii, Trypanosoma rangeli, Trypanosoma cruzi, Trypanosoma rhodesiense, Trypanosoma brucei, Schistosoma mansoni, Schistosoma japonicum, Babesia bovis, Eimeria tenella, Onchocerca volvulus, Leishmania tropica, Mycobacterium tuberculosis, Trichinella spiralis, Theileria parva, Taenia hydatigena, Taenia ovis, Taenia saginata, Echinococcus granulosus, Mesocestoides corti, Mycoplasma arthritidis, M. hyorhinis, M. orale, M. arginini, Acholeplasma laidlawii, M. salivarium and M. pneumoniae. In some embodiments, the target sequence is a portion of a nucleic acid from a genomic locus, a transcribed mRNA, or a reverse transcribed cDNA from a gene locus of bacterium or other agents responsible for a disease in the sample comprising a mutation that confers resistance to a treatment, such as a single nucleotide mutation that confers resistance to antibiotic treatment.
[0270] In some embodiments, the target nucleic acid comprises a nucleic acid sequence of a virus, a bacterium, or other pathogen responsible for a disease in a plant (e.g., a crop). Methods and compositions of the disclosure may be used to treat or detect a disease in a plant. For example, the methods of the disclosure may be used to target a viral nucleic acid sequence in a plant. An effector protein of the disclosure may cleave the viral nucleic acid. In some embodiments, the target nucleic acid comprises a nucleic acid sequence of a virus or a bacterium or other agents (e.g., any pathogen) responsible for a disease in the plant (e.g., a crop). In some embodiments, the target nucleic acid comprises RNA. The target nucleic acid, in some embodiments, is a portion of a nucleic acid from a virus or a bacterium or other agents responsible for a disease in the plant (e.g., a crop). In some embodiments, the target nucleic acid is a portion of a nucleic acid from a genomic locus, or any NA amplicon, such as a reverse transcribed mRNA or a cDNA from a gene locus, a transcribed mRNA, or a reverse transcribed cDNA from a gene locus in at a virus or a bacterium or other agents (e.g., any pathogen) responsible for a disease in the plant (e.g., a crop). A virus infecting the plant may be an RNA virus. A virus infecting the plant may be a DNA virus. Non-limiting examples of viruses that may be targeted with the disclosure include Tobacco mosaic virus (TMV), Tomato spotted wilt virus (TSWV), Cucumber mosaic virus (CMV), Potato virus Y (PVY), Cauliflower mosaic virus (CaMV) (RT virus), Plum pox virus (PPV), Brome mosaic virus (BMV) and Potato virus X (PVX).
[0271] In some embodiments, the target nucleic acid is any one of: a naturally occurring eukaryotic sequence, a naturally occurring prokaryotic sequence, a naturally occurring viral sequence, a naturally occurring bacterial sequence, a naturally occurring fungal sequence, an engineered eukaryotic sequence, an engineered prokaryotic sequence, an engineered viral sequence, an engineered bacterial sequence, an engineered fungal sequence, a fragment of a naturally occurring sequence, a fragment of an engineered sequence, and combinations thereof. [0272] In some embodiments, the target nucleic acid is isolated from any one of: a naturally occurring cell, a eukaryotic cell, a prokaryotic cell, a plant cell, a fungal cell, an animal cell, cell of an invertebrate, a fly cell, a cell of a vertebrate, a mammalian cell, a primate cell, a nonhuman primate cell, a human cell, a living cell, a non-living cell, a modified cell, a derived cell, and a non-naturally occurring cell.
[0273] Nucleic acids, such as DNA and pre-mRNA, described herein can contain at least one intron and at least one exon, wherein as read in the 5’ to the 3’ direction of a nucleic acid strand, the 3’ end of an intron can be adjacent to the 5’ end of an exon, and wherein said intron and exon correspond for transcription purposes. If a nucleic acid strand contains more than one intron and exon, the 5’ end of the second intron is adjacent to the 3’ end of the first exon, and 5’ end of the second exon is adjacent to the 3’ end of the second intron. The junction between an intron and an exon can be referred to herein as a splice junction, wherein a 5’ splice site (SS) can refer to the +1/+2 position at the 5’ end of intron and a 3’SS can refer to the last two positions at the 3’ end of an intron. Alternatively, a 5’ SS can refer to the 5’ end of an exon and a 3’SS can refer to the 3’ end of an exon. In some embodiments, nucleic acids can contain one or more elements that act as a signal during transcription, splicing, and/or translation. In some embodiments, signaling elements include a 5’SS, a 3’SS, a premature stop codon, U1 and/or U2 binding sequences, and cis acting elements such as branch site (BS), polypyridine tract (PYT), exonic and intronic splicing enhancers (ESEs and ISEs) or silencers (ESSs and ISSs). In some embodiments, nucleic acids may also comprise a untranslated region (UTR), such as a 5’ UTR or a 3’ UTR. In some embodiments, the start of an exon or intron is referred to interchangeably herein as the 5’ end of an exon or intron, respectively. Likewise, in some embodiments, the end of an exon or intron is referred to interchangeably herein as the 3’ end of an exon or intron, respectively.
[0274] In some embodiments, at least a portion of at least one target sequence is within about 1, about 5 or more, about 10 or more, about 15 or more, about 20 or more, about 25 or more, about 30 or more, about 35 or more, about 40 or more, about 45 or more, about 50 or more, about 55 or more, about 60 or more, about 65 or more, about 70 or more, about 75 or more, about 80 or more, about 85 or more, about 90 or more, about 95 or more, about 100 or more, about 105 or more, about 110 or more, about 115 or more, about 120 or more, about 125 or more, about 130 or more, about 135 or more, about 140 or more, about 145 or more, or about 150 to about 300 nucleotides adjacent to: the 5’ end of an exon; the 3’ end of an exon; the 5’ end of an intron; the 3’ end of an intron; one or more signaling element comprising a 5’SS, a 3’SS, a premature stop codon, U1 binding sequence, U2 binding sequence, a BS, a PYT, ESE, an ISE, an ESS, an ISS; a 5’ UTR; a 3’ UTR; more than one of the foregoing, or any combination thereof. In some embodiments, the target nucleic acid comprises a target locus. In some embodiments, the target nucleic acid comprises more than one target loci. In some embodiments, the target nucleic acid comprises two target loci. Accordingly, in some embodiments, the target nucleic acid can comprise one or more target sequences.
[0275] In some embodiments, compositions, systems, and methods described herein comprise an edited target nucleic acid which can describe a target nucleic acid wherein the target nucleic acid has undergone a change, for example, after contact with an effector protein. In some embodiments, the editing is an alteration in the sequence of the target nucleic acid. In some embodiments, the edited target nucleic acid comprises an insertion, deletion, or replacement of one or more nucleotides compared to the unedited target nucleic acid. In some embodiments, the editing is a mutation.
Mutations
[0276] In some embodiments, target nucleic acids described herein comprise a mutation. In some embodiments, a composition, system or method described herein can be used to edit a target nucleic acid comprising a mutation such that the mutation is edited to be the wild-type nucleotide or nucleotide sequence. In some embodiments, a composition, system or method described herein can be used to detect a target nucleic acid comprising a mutation. A mutation may result in the insertion of at least one amino acid in a protein encoded by the target nucleic acid. A mutation may result in the deletion of at least one amino acid in a protein encoded by the target nucleic acid. A mutation may result in the substitution of at least one amino acid in a protein encoded by the target nucleic acid. A mutation that results in the deletion, insertion, or substitution of one or more amino acids of a protein encoded by the target nucleic acid may result in misfolding of a protein encoded by the target nucleic acid. A mutation may result in a premature stop codon, thereby resulting in a truncation of the encoded protein.
[0277] Non-limiting examples of mutations are insertion-deletion (indel), a point mutation, single nucleotide polymorphism (SNP), a chromosomal mutation, a copy number mutation or variation, and frameshift mutations. In some embodiments, an indel mutation is an insertion or deletion of one or more nucleotides. In some embodiments, a point mutation comprises a substitution, insertion, or deletion. In some embodiments, a frameshift mutation occurs when the number of nucleotides in the insertion/deletion is not divisible by three, and it occurs in a protein coding region. In some embodiments, a chromosomal mutation can comprise an inversion, a deletion, a duplication, or a translocation of one or more nucleotides. In some embodiments, a copy number variation can comprise a gene amplification or an expanding trinucleotide repeat. In some embodiments, an SNP is associated with a phenotype of the sample or a phenotype of the organism from which the sample was taken. In some embodiments, an SNP is associated with altered phenotype from wild type phenotype. In some embodiments, the SNP is a synonymous substitution or a nonsynonymous substitution. In some embodiments, the nonsynonymous substitution is a missense substitution or a nonsense point mutation. In some embodiments, the synonymous substitution is a silent substitution.
[0278] In some embodiments, a target nucleic acid described herein comprises a mutation of one or more nucleotides. In some embodiments, the one or more nucleotides comprises 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more nucleotides. In some embodiments, the mutation comprises a deletion, insertion, and/or substitution of about 5, about 10, about 15, about 20, about 25, about 30, about 35, about 40, about 45, about 50, about 55, about 60, about 65, about 70, about 75, about 80, about 85, about 90, about 95, about 100, about 200, about 300, about 400, about 500, about 600, about 700, about 800, about 900, or about 1000 nucleotides. In some embodiments, the mutation comprises a deletion, insertion, and/or substitution of 1 to 5, 5 to 10, 10 to 15, 15 to 20, 20 to 25, 25 to 30, 30 to 35, 35 to 40, 40 to 45, 45 to 50, 50 to 55, 55 to 60, 60 to 65, 65 to 70, 70 to 75, 75 to 80, 80 to 85, 85 to 90, 90 to 95, 95 to 100, 100 to 200, 200 to 300, 300 to 400, 400 to 500, 500 to 600, 600 to 700, 700 to 800, 800 to 900, 900 to 1000, 1 to 50, 1 to 100, 25 to 50, 25 to 100, 50 to 100, 100 to 500, 100 to 1000, or 500 to 1000 nucleotides. The mutation may be located in a non-coding region or a coding region of a gene, wherein the gene is a target nucleic acid. A mutation may be in an open reading frame of a target nucleic acid. In some embodiments, guide nucleic acids described herein hybridize to a portion of the target nucleic acid comprising or adjacent to the mutation.
[0279] In some embodiments, target nucleic acids comprise a mutation, wherein the mutation is a SNP. In some embodiments, the single nucleotide mutation or SNP is associated with a phenotype of the sample or a phenotype of the organism from which the sample was taken. In some embodiments, the SNP is associated with altered phenotype from wild type phenotype. In some embodiments, a single nucleotide mutation, SNP, or deletion described herein is associated with a disease, such as a genetic disease. In some embodiments, the SNP is a synonymous substitution or a nonsynonymous substitution. In some embodiments, the nonsynonymous substitution is a missense substitution or a nonsense point mutation. In some embodiments, the synonymous substitution is a silent substitution. In some embodiments, the mutation is a deletion of one or more nucleotides. In some embodiments, the single nucleotide mutation, SNP, or deletion is associated with a disease such as a genetic disorder. In some embodiments, the mutation, such as a single nucleotide mutation, a SNP, or a deletion, may be encoded in the sequence of a target nucleic acid from the germline of an organism or may be encoded in a target nucleic acid from a diseased cell.
[0280] In some embodiments, the mutation is associated with a disease, such as a genetic disorder. In some embodiments, the mutation may be encoded in the sequence of a target nucleic acid from the germline of an organism or may be encoded in a target nucleic acid from a diseased cell. In some embodiments, a target nucleic acid described herein comprises a mutation associated with a disease. In some examples, a mutation associated with a disease refers to a mutation whose presence in a subject indicates that the subject is susceptible to or suffers from, a disease, disorder, condition, or syndrome. In some examples, a mutation associated with a disease refers to a mutation which causes, contributes to the development of, or indicates the existence of the disease, disorder, condition, or syndrome. A mutation associated with a disease may also refer to any mutation which generates transcription or translation products at an abnormal level, or in an abnormal form, in cells affected by a disease relative to a control without the disease. In some embodiments, a mutation associated with a disease, comprises the co-occurrence of a mutation and the phenotype of a disease. The mutation may occur in a gene, wherein transcription or translation products from the gene occur at a significantly abnormal level or in an abnormal form in a cell or subject harboring the mutation as compared to a non-disease control subject not having the mutation. In some embodiments, a target nucleic acid described herein comprises a mutation associated with a disease described herein.
[0281] In some embodiments, a target nucleic acid is in a cell. In some embodiments, the cell is a single-cell eukaryotic organism; a plant cell an algal cell; a fungal cell; an animal cell; a cell of an invertebrate animal; a cell of a vertebrate animal such as fish, amphibian, reptile, bird, and mammal; or a cell of a mammal such as a human, a non-human primate, an ungulate, a feline, a bovine, an ovine, and a caprine. In some embodiments, the cell is a eukaryotic cell. In some embodiments, the cell is a mammalian cell, a human cell, or a plant cell. In some embodiments, the cell is a human cell. In some embodiments, the human cell is a: muscle cell, liver cell, lung cell, cardiac cell, visceral cell, cardiac muscle cell, smooth muscle cell, cardiomyocyte, nodal cardiac muscle cell, smooth muscle cell, visceral muscle cell, skeletal muscle cell, myocyte, red (or slow) skeletal muscle cell, white (fast) skeletal muscle cell, intermediate skeletal muscle, muscle satellite cell, muscle stem cell, myoblast, muscle progenitor cell, induced pluripotent stem cell (iPS), or a cell derived from an iPS cell, modified to have its gene edited and differentiated into myoblasts, muscle progenitor cells, muscle satellite cells, muscle stem cells, skeletal muscle cells, cardiac muscle cells or smooth muscle cells.
[0282] In some embodiments, an effector protein-guide nucleic acid complex may comprise high selectivity for a target sequence. In some embodiments, an RNP comprise a selectivity of at least 200: 1, 100: 1, 50: 1, 20: 1, 10: 1, or 5: 1 for a target nucleic acid over a single nucleotide variant of the target nucleic acid. In some embodiments, an RNP may comprise a selectivity of at least 5: 1 for a target nucleic acid over a single nucleotide variant of the target nucleic acid.
[0283] By leveraging such effector protein selectivity, some methods described herein may detect a target nucleic acid present in the sample in various concentrations or amounts as a target nucleic acid population. In some embodiments, the method detects at least 2 target nucleic acid populations. In some embodiments, the method detects at least 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 40, or 50 target nucleic acid populations. In some embodiments, the method detects 3 to 50, 5 to 40, or 10 to 25 target nucleic acid populations. In some embodiments, the method detects at least 2 individual target nucleic acids. In some embodiments, the method detects at least 3, 5, 10, 20, 30, 40, 50, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 2000, 3000, 4000, 5000, 6000, 7000, 8000, 9000, or 10000 individual target nucleic acids. In some embodiments, the method detects 1 to 10,000, 100 to 8000, 400 to 6000, 500 to 5000, 1000 to 4000, or 2000 to 3000 individual target nucleic acids. In some embodiments, the method detects target nucleic acid present at least at one copy per 10 non-target nucleic acids, 102 non-target nucleic acids, 103 non-target nucleic acids, 104 non-target nucleic acids, 105 non-target nucleic acids, 106 non-target nucleic acids, 107 non-target nucleic acids, 108 non-target nucleic acids, 109 non-target nucleic acids, or 1010 non-target nucleic acids.
[0284] In some embodiments, compositions described herein exhibit indiscriminate transcleavage of a nucleic acid (e.g., ssRNA or ssDNA), enabling their use for detection of a nucleic acid (e.g, RNA or DNA, respectively) in samples. In some embodiments, target nucleic acids are generated from many nucleic acid templates (e.g., RNA) in order to achieve cleavage of a reporter (e.g., a FQ reporter) in a device (e.g., a DETECTR platform). Certain effector proteins may be activated by a nucleic acid (e.g., ssDNA or ssRNA), upon which they may exhibit trans-cleavage of the nucleic acid (e.g., ssDNA or ssRNA) and may, thereby, be used to cleave reporter molecules (e.g., ssDNA or ssRNA FQ reporter molecules) in a device (e.g., a DETECTR). These effector proteins may target nucleic acids present in the sample or nucleic acids generated and/or amplified from any number of nucleic acid templates (e.g., RNA). Described herein are reagents comprising a single stranded reporter nucleic acid comprising a detection moiety, wherein the reporter nucleic acid (e.g., a ssDNA-FQ reporter described herein) is capable of being cleaved by the effector protein, upon generation (e.g., cDNA) and amplification of nucleic acids from a nucleic acid template (e.g., ssRNA) using the methods disclosed herein, thereby generating a first detectable signal. While DNA and RNA are used as an exemplary reporter in the foregoing, any suitable reporter may be used.
[0285] In some embodiments, a target nucleic acid is an amplified nucleic acid of interest. In some embodiments, the nucleic acid of interest is any nucleic acid disclosed herein or from any sample as disclosed herein. In some embodiments, the nucleic acid of interest is an RNA that is reverse transcribed before amplification. In some embodiments, the nucleic acid of interest is amplified then the amplicons is transcribed into RNA.
[0286] In some embodiments, target nucleic acids may activate an effector protein to initiate sequence-independent cleavage of a nucleic acid-based reporter (e.g., a reporter comprising an RNA sequence, or a reporter comprising DNA and RNA). For example, an effector protein of the present disclosure is activated by a target nucleic acid to cleave reporters having an RNA (also referred to herein as an “RNA reporter”). Alternatively, an effector protein of the present disclosure is activated by a target nucleic acid to cleave reporters having an RNA. Alternatively, an effector protein of the present disclosure is activated by a target RNA to cleave reporters having an RNA (also referred to herein as a “RNA reporter”). The RNA reporter may comprise a single-stranded RNA labelled with a detection moiety or may be any RNA reporter as disclosed herein.
[0287] Further description of editing or detecting a target nucleic acid in a gene of interest can be found in more detail in Kim et al., “Enhancement of target specificity of CRISPR-Casl2a by using a chimeric DNA-RNA guide”, Nucleic Acids Res. 2020 Sep 4;48(15):8601-8616; Wang etal., “Specificity profiling of CRISPR system reveals greatly enhanced off-target gene editing”, Scientific Reports volume 10, Article number: 2269 (2020); Tuladhar et al., “CRISPR-Cas9-based mutagenesis frequently provokes on-target mRNA misregulation”, Nature Communications volume 10, Article number: 4056 (2019); Dong etal., “Genome-Wide Off-Target Analysis in CRISPR-Cas9 Modified Mice and Their Offspring”, G3, Volume 9, Issue 11, 1 November 2019, Pages 3645-3651; Winter et al., “Genome-wide CRISPR screen reveals novel host factors required for Staphylococcus aureus a-hemolysin-mediated toxicity”, Scientific Reports volume 6, Article number: 24242 (2016); and Ma et al., “A CRISPR-Based Screen Identifies Genes Essential for West-Nile- Virus-Induced Cell Death”, Cell Rep. 2015 Jul 28;12(4):673-83, which are hereby incorporated by reference in their entirety.
VII. Pharmaceutical Compositions and Modes of Administration
[0288] Disclosed herein, in some aspects, are pharmaceutical compositions for modifying a target nucleic acid in a cell or a subject, comprising any one of the effector proteins, engineered effector proteins, or fusion proteins described herein. Also disclosed herein, in some aspects, are pharmaceutical compositions comprising a nucleic acid encoding any one of the effector proteins, engineered effector proteins, or fusion proteins described herein. In some embodiments, pharmaceutical compositions comprise a guide nucleic acid. In some embodiments, pharmaceutical compositions comprise a plurality of guide nucleic acids. Pharmaceutical compositions may be used to modify a target nucleic acid or the expression thereof in a cell in vitro, in vivo or ex vivo.
[0289] In some embodiments, the pharmaceutically acceptable excipient, carrier or diluent allows the active ingredient to retain biological activity. In some embodiments, the pharmaceutically acceptable excipient, carrier or diluent is non-reactive with the subject's immune system. In some embodiments, the pharmaceutically acceptable excipient, carrier or diluent provides for long-term stabilization of the composition. In some embodiments, the pharmaceutically acceptable excipient, carrier or diluent is provided as a bulking agent in solid formulations that contain potent active ingredients in small amounts. In some embodiments, the pharmaceutically acceptable excipient, carrier or diluent confers a therapeutic enhancement on the active ingredient in the final dosage form. In some embodiments, the pharmaceutically acceptable excipient, carrier or diluent facilitates absorption, reduces viscosity, or enhances solubility. In some embodiments, the pharmaceutically acceptable excipient, carrier or diluent is selected based upon the route of administration, dosage form, active ingredient, other factors, or any combination thereof. In some embodiments, the pharmaceutically acceptable excipient, carrier or diluent can be formulated by well-known conventional methods (see, e.g., Remington's Pharmaceutical Sciences, 18th edition, A. Gennaro, ed., Mack Publishing Co., Easton, Pa., 1990; and Remington, The Science and Practice of Pharmacy 21st Ed. Mack Publishing, 2005).
[0290] In some embodiments, pharmaceutical compositions comprise one or more nucleic acids encoding an effector protein, effector partner, fusion protein, a guide nucleic acid, or a combination thereof; and a pharmaceutically acceptable carrier or diluent. The effector protein, effector partner, fusion protein, or combination thereof may be any one of those described herein. The one or more nucleic acids may comprise a plasmid. The one or more nucleic acids may comprise a nucleic acid expression vector. The one or more nucleic acids may comprise a viral vector. In some embodiments, the viral vector is a lentiviral vector. In some embodiments, the vector is an adeno-associated viral (AAV) vector. In some embodiments, compositions, including pharmaceutical compositions, comprise a viral vector encoding a fusion protein and a guide nucleic acid, wherein at least a portion of the guide nucleic acid binds to the effector protein of the fusion protein. In some embodiments, pharmaceutical compositions comprise a virus comprising a viral vector encoding a fusion protein, an effector protein, an effector partner, a guide nucleic acid, or a combination thereof; and a pharmaceutically acceptable carrier or diluent.
[0291] Pharmaceutical compositions described herein may comprise a salt. In some embodiments, the salt is a sodium salt. In some embodiments, the salt is a potassium salt. In some embodiments, the salt is a magnesium salt. In some embodiments, the salt is NaCl. In some embodiments, the salt is KNO3. In some embodiments, the salt is Mg2+ SO42 .
[0292] Non-limiting examples of pharmaceutically acceptable carriers and diluents suitable for the pharmaceutical compositions disclosed herein include buffers (e.g., neutral buffered saline, phosphate buffered saline); carbohydrates (e.g., glucose, mannose, sucrose, dextran, mannitol); polypeptides or amino acids (e.g., glycine); antioxidants; chelating agents (e.g., EDTA, glutathione); adjuvants (e.g., aluminum hydroxide); surfactants (Polysorbate 80, Polysorbate 20, or Pluronic F68); glycerol; sorbitol; mannitol; polyethyleneglycol; and preservatives. In some embodiments, the vector is formulated for delivery through injection by a needle carrying syringe. In some embodiments, the composition is formulated for delivery by electroporation. In some embodiments, the composition is formulated for delivery by chemical method. In some embodiments, the pharmaceutical compositions comprise a virus vector or a non-viral vector. [0293] In some embodiments, pharmaceutical compositions are in the form of a solution (e.g., a liquid). The solution may be formulated for injection, e.g., intravenous or subcutaneous injection. In some embodiments, the pH of the solution is about 7, about 7.1, about 7.2, about 7.3, about 7.4, about 7.5, about 7.6, about 7.7, about 7.8, about 7.9, about 8, about 8.1, about 8.2, about 8.3, about 8.4, about 8.5, about 8.6, about 8.7, about 8.8, about 8.9, or about 9. In some embodiments, the pH is 7 to 7.5, 7.5 to 8, 8 to 8.5, 8.5 to 9, or 7 to 8.5. In some cases, the pH of the solution is less than 7. In some cases, the pH is greater than 7. VIII. Methods of Modifying Nucleic Acids
[0294] Provided herein are methods of modifying target nucleic acids or the expression thereof. In some embodiments, methods comprise editing a target nucleic acid. In general, editing refers to modifying the nucleobase sequence of a target nucleic acid. Also provided herein are methods of modulating the expression of a target nucleic acid. Fusion proteins and systems described herein may be used for such methods. Methods of editing a target nucleic acid may comprise one or more of cleaving the target nucleic acid, deleting one or more nucleotides of the target nucleic acid, inserting one or more nucleotides into the target nucleic acid, modifying one or more nucleotides of the target nucleic acid. Methods of modulating expression of target nucleic acids may comprise modifying the target nucleic acid or a protein associated with the target nucleic acid, e.g., a histone.
[0295] In some embodiments, methods comprise contacting a target nucleic acid with a composition described herein. In some embodiments, methods comprise contacting a target nucleic acid with an effector protein described herein. In some embodiments, methods comprise contacting a target nucleic acid with a fusion protein described herein. The effector protein may be an effector protein provided in TABLE 1, TABLE 6, or TABLE 8, or a catalytically inactive variant thereof. The effector protein may comprise an amino acid sequence that is at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99% or 100% identical to a sequence described in TABLE 1, TABLE 6, or TABLE 8. In some embodiments, the amino acid sequence of the effector protein is at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99% or 100% identical to a sequence described in TABLE 1, TABLE 6, or TABLE 8
[0296] In some embodiments, methods comprise base editing. In some embodiments, base editing comprises contacting a target nucleic acid with a fusion protein comprising an effector protein fused to a base editing enzyme, such as a deaminase, thereby changing a nucleobase of the target nucleic acid to an alternative nucleobase. In some embodiments, the nucleobase of the target nucleic acid is adenine (A) and the method comprises changing A to guanine (G). In some embodiments, the nucleobase of the target nucleic acid is cytosine (C) and the method comprises changing C to thymine (T). In some embodiments, the nucleobase of the target nucleic acid is C and the method comprises changing C to G. In some embodiments, the nucleobase of the target nucleic acid is A and the method comprises changing A to G.
[0297] In some embodiments, methods introduce a nucleobase change in a target nucleic acid relative to a corresponding wildtype or mutant nucleobase sequence. In some embodiments, methods remove or correct a disease-causing mutation in a nucleic acid sequence, e.g., to produce a corresponding wildtype nucleobase sequence. In some embodiments, methods remove/correct point mutations, deletions, null mutations, or tissue-specific mutations in a target nucleic acid. In some embodiments, methods generate gene knock-out, gene knock-in, gene editing, gene tagging, or a combination thereof. Methods of the disclosure may be targeted to a locus in a genome of a cell.
[0298] Modifying at least one gene using the compositions and methods described herein can, in some embodiments, induce a reduction or increase in expression of the one or more genes. In some embodiments, the at least one modified gene results in a reduction in expression, also referred to as gene silencing. In some embodiments, the gene silencing reduces expression of one or more genes by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or at least 95%. In some embodiments, gene silencing is accomplished by transcriptional silencing, post-transcriptional silencing, or meiotic silencing. In some embodiments, transcriptional silencing is by genomic imprinting, paramutation, transposon silencing, position effect, or RNA-directed DNA methylation. In some embodiments, post-transcriptional silencing is by RNA interference, RNA silencing, or nonsense mediated decay. In some embodiments, meiotic silencing is by transfection or meiotic silencing of unpaired DNA. In some embodiments, the at least one modified gene results in removing all expression, also referred to as the gene being knocked out (KO). In some embodiments, the compositions, methods or systems increase expression of one or more genes by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or at least 100%.
[0299] In some embodiments, methods of editing a target nucleic acid or modulating the expression of a target nucleic acid are performed in vivo. In some embodiments, methods of editing a target nucleic acid or modulating the expression of a target nucleic acid are performed in vitro. For example, a plasmid may be modified in vitro using a composition described herein and introduced into a cell or organism. In some embodiments, methods of editing a target nucleic acid or modulating the expression of a target nucleic acid are performed ex vivo. For example, methods may comprise obtaining a cell from a subject, modifying a target nucleic acid in the cell with methods and compositions described herein, and returning the cell to the subject. Methods of editing performed ex vivo may be particularly advantageous to produce CAR T-cells. In some embodiments, methods comprise editing a target nucleic acid or modulating the expression of the target nucleic acid in a cell or a subject. The cell may be a dividing cell. The cell may be a terminally differentiated cell. In some embodiments, the target nucleic acid is a gene.
[0300] Methods of editing a target nucleic acid or modulating the expression of a target nucleic acid described herein may be employed to generate a genetically modified cell. The cell may be a prokaryotic cell. The cell may be an archaeal cell. The cell may be a eukaryotic cell. The cell may be a mammalian cell. The cell may be a human cell. The cell may be a T cell. The cell may be a hematopoietic stem cell. The cell may be a bone marrow derived cell, a white blood cell, a blood cell progenitor, or a combination thereof. Generating a genetically modified cell may comprise contacting a target cell with an effector protein or a fusion protein described herein and a guide nucleic acid. Contacting may comprise electroporation, acoustic poration, optoporation, viral vector-based delivery, iTOP, nanoparticle delivery (e.g., lipid or gold nanoparticle delivery), cell -penetrating peptide (CPP) delivery, DNA nanostructure delivery, or any combination thereof. In some cases, the nanoparticle delivery comprises lipid nanoparticle delivery or gold nanoparticle delivery. In some cases, the nanoparticle delivery comprises lipid nanoparticle delivery. In some cases, the nanoparticle delivery comprises gold nanoparticle delivery.
[0301] In some embodiments, the compositions, methods or systems comprise a nucleic acid expression vector, or use thereof, to introduce an effector protein, guide nucleic acid, donor template or any combination thereof to a cell. In some embodiments, the nucleic acid expression vector is a viral vector. Viral vectors include, but are not limited to, retroviruses, adenoviruses, adeno-associated viruses, and herpes simplex viruses. In some embodiments, the viral vector is a replication-defective viral vector, comprising an insertion of a therapeutic gene inserted in genes essential to the lytic cycle, preventing the virus from replicating and exerting cytotoxic effects. In some embodiments, the viral vector is an adeno associated viral (AAV) vector. In some embodiments, the nucleic acid expression vector is a non-viral vector. In some embodiments, compositions and methods comprise a lipid, polymer, nanoparticle, or a combination thereof, or use thereof, to introduce a Cas protein, guide nucleic acid, donor template or any combination thereof to a cell. Non-limiting examples of lipids and polymers are cationic polymers, cationic lipids, or bio-responsive polymers. In some embodiments, the bio-responsive polymer exploits chemical-physical properties of the endosomal environment (e.g., pH) to preferentially release the genetic material in the intracellular space.
[0302] Methods of modifying may comprise contacting a target nucleic acid with one or more components, compositions or systems described herein. In some embodiments, a method of modifying comprises contacting a target nucleic acid with at least one of: a) one or more effector proteins, or one or more nucleic acids encoding one or more effector proteins; or b) one or more guide nucleic acids, or one or more nucleic acids encoding one or more guide nucleic acids. In some embodiments, a method of modifying comprises contacting a target nucleic acid with a system described herein wherein the system comprises components comprising at least one of: a) one or more effector proteins, or one or more nucleic acids encoding one or more effector proteins; or b) one or more guide nucleic acids, or one or more nucleic acids encoding one or more guide nucleic acids. In some embodiments, a method of modifying comprises contacting a target nucleic acid with a composition described herein comprising at least one of: a) one or more effector proteins, or one or more nucleic acids encoding one or more effector proteins; or b) one or more guide nucleic acids, or one or more nucleic acids encoding one or more guide nucleic acids; in a composition. In some embodiments, a method of modifying as described herein produces a modified target nucleic acid.
[0303] Editing a target nucleic acid sequence may introduce a mutation (e.g., point mutations, deletions) in a target nucleic acid relative to a corresponding wildtype nucleotide sequence. Editing may remove or correct a disease-causing mutation in a nucleic acid sequence to produce a corresponding wildtype nucleotide sequence. Editing a target nucleic acid sequence may remove/correct point mutations, deletions, null mutations, or tissue-specific mutations in a target nucleic acid. Editing a target nucleic acid sequence may be used to generate gene knock-out, gene knock-in, gene editing, gene tagging, or a combination thereof. Methods of the disclosure may be targeted to any locus in a genome of a cell.
[0304] Modifying may comprise single stranded cleavage, double stranded cleavage, donor nucleic acid insertion, epigenetic modification (e.g., methylation, demethylation, acetylation, or deacetylation), or a combination thereof. In some embodiments, cleavage (single-stranded or double-stranded) is site-specific, meaning cleavage occurs at a specific site in the target nucleic acid, often within the region of the target nucleic acid that hybridizes with the guide nucleic acid spacer sequence. In some embodiments, the effector proteins introduce a singlestranded break in a target nucleic acid to produce a cleaved nucleic acid. In some embodiments, the effector protein is capable of introducing a break in a single stranded RNA (ssRNA). The effector protein may be coupled to a guide nucleic acid that targets a particular region of interest in the ssRNA. In some embodiments, the target nucleic acid, and the resulting cleaved nucleic acid is contacted with a nucleic acid for homologous recombination (e.g., homology directed repair (HDR)) or non -homologous end joining (NHEJ). In some embodiments, a doublestranded break in the target nucleic acid may be repaired (e.g., by NHEJ or HDR) without insertion of a donor template, such that the repair results in an indel in the target nucleic acid at or near the site of the double-stranded break. In some embodiments, an indel, sometimes referred to as an insertion-deletion or indel mutation, is a type of genetic mutation that results from the insertion and/or deletion of one or more nucleotide in a target nucleic acid. An indel may vary in length (e.g., 1 to 1,000 nucleotides in length) and be detected using methods well known in the art, including sequencing. If the number of nucleotides in the insertion/deletion is not divisible by three, and it occurs in a protein coding region, it is also a frameshift mutation. Indel percentage is the percentage of sequencing reads that show at least one nucleotide has been mutation that results from the insertion and/or deletion of nucleotides regardless of the size of insertion or deletion, or number of nucleotides mutated. For example, if there is at least one nucleotide deletion detected in a given target nucleic acid, it counts towards the percent indel value. As another example, if one copy of the target nucleic acid has one nucleotide deleted, and another copy of the target nucleic acid has 10 nucleotides deleted, they are counted the same. This number reflects the percentage of target nucleic acids that are edited by a given effector protein.
[0305] In some embodiments, methods of modifying described herein cleave a target nucleic acid at one or more locations to generate a cleaved target nucleic acid. In some embodiments, the cleaved target nucleic acid undergoes recombination (e.g., NHEJ or HDR). In some embodiments, cleavage in the target nucleic acid may be repaired (e.g., by NHEJ or HDR) without insertion of a donor nucleic acid, such that the repair results in an indel in the target nucleic acid at or near the site of the cleavage site. In some embodiments, cleavage in the target nucleic acid may be repaired (e.g., by NHEJ or HDR) with insertion of a donor nucleic acid, such that the repair results in an indel in the target nucleic acid at or near the site of the cleavage site.
[0306] In some embodiments, wherein the compositions, systems, and methods of the present disclosure comprise an additional guide nucleic acid or a use thereof, and such dual-guided compositions, systems, and methods described herein may modify the target nucleic acid in two locations. In some embodiments, dual-guided modifying may comprise cleavage of the target nucleic acid in the two locations targeted by the guide nucleic acids. In some embodiments, upon removal of the sequence between the guide nucleic acids, the wild-type reading frame is restored. A wild-type reading frame may be a reading frame that produces at least a partially, or fully, functional protein. A non-wild-type reading frame may be a reading frame that produces a non-functional or partially non-functional protein. [0307] Accordingly, in some embodiments, compositions, systems, and methods described herein may edit 1 to 1,000 nucleotides or any integer in between, in a target nucleic acid. In some embodiments, 1 to 1,000, 2 to 900, 3 to 800, 4 to 700, 5 to 600, 6 to 500, 7 to 400, 8 to 300, 9 to 200, or 10 to 100 nucleotides, or any integer in between, may be edited by the compositions, systems, and methods described herein. In some embodiments, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more nucleotides may be edited by the compositions, systems, and methods described herein. In some embodiments, 10, 20, 30, 40, 50, 60, 70, 80 90, 100 or more nucleotides, or any integer in between, may be edited by the compositions, systems, and methods described herein. In some embodiments, 100, 200, 300, 400, 500, 600, 700, 800, 900 or more nucleotides, or any integer in between, may be edited by the compositions, systems, and methods described herein.
[0308] Methods may comprise use of two or more effector proteins. An illustrative method for introducing a break in a target nucleic acid comprises contacting the target nucleic acid with: (a) a first engineered guide nucleic acid comprising a region that binds to a first effector protein described herein; and (b) a second engineered guide nucleic acid comprising a region that binds to a second effector protein described herein, wherein the first engineered guide nucleic acid comprises an additional region that hybridizes to the target nucleic acid and wherein the second engineered guide nucleic acid comprises an additional region that hybridizes to the target nucleic acid. In some embodiments, the first and second effector protein are identical. In some embodiments, the first and second effector protein are not identical.
[0309] In some embodiments, editing a target nucleic acid comprises genome editing. Genome editing may comprise editing a genome, chromosome, plasmid, or other genetic material of a cell or organism. In some embodiments, the genome, chromosome, plasmid, or other genetic material of the cell or organism is modified in vivo. In some embodiments, the genome, chromosome, plasmid, or other genetic material of the cell or organism is modified in a cell. In some embodiments, the genome, chromosome, plasmid, or other genetic material of the cell or organism is modified in vitro. For example, a plasmid may be edited in vitro using a composition described herein and introduced into a cell or organism.
[0310] In some embodiments, editing a target nucleic acid may comprise deleting a sequence from a target nucleic acid. For example, a mutated sequence or a sequence associated with a disease may be removed from a target nucleic acid. In some embodiments, editing a target nucleic acid may comprise replacing a sequence in a target nucleic acid with a second sequence. For example, a mutated sequence or a sequence associated with a disease may be replaced with a second sequence lacking the mutation or that is not associated with the disease. In some embodiments, editing a target nucleic acid may comprise deleting or replacing a sequence comprising markers associated with a disease or disorder. In some embodiments, editing a target nucleic acid may comprise introducing a sequence into a target nucleic acid. For example, a beneficial sequence or a sequence that may reduce or eliminate a disease may be inserted into the target nucleic acid.
[0311] In some embodiments, methods comprise inserting a donor nucleic acid into a cleaved target nucleic acid. The donor nucleic acid may be inserted at a specified (e.g., effector protein targeted) point within the target nucleic acid. In some embodiments, the cleaved target nucleic acid is cleaved at a single location. In such embodiments, the methods comprise contacting a target nucleic acid with an effector protein described herein, thereby introducing a singlestranded break in the target nucleic acid; and contacting the target nucleic acid with a donor nucleic acid for homologous recombination, optionally by HDR or NHEJ, thereby introducing a new sequence into the target nucleic acid (e.g., at a cleavage site). In some embodiments, the cleaved target nucleic acid is cleaved at two locations. In such embodiments, the methods comprise contacting a target nucleic acid with an effector protein described herein, thereby introducing a single-stranded break in the target nucleic acid; contacting the target nucleic acid with a second effector protein described herein, to generate a second cleavage site in the target nucleic acid, ligating the regions flanking the first and second cleavage site, optionally through NHEJ or single-strand annealing, thereby resulting in the excision of a portion of the target nucleic acid between the first and second cleavage sites from the target nucleic acid; and contacting the target nucleic acid with a donor nucleic acid for homologous recombination, optionally by HDR or NHEJ, thereby introducing a new sequence into the target nucleic acid (e.g., in between two cleavage sites).
[0312] In some embodiments, methods comprise editing a target nucleic acid with two or more effector proteins. Editing a target nucleic acid may comprise introducing a two or more singlestranded breaks in a target nucleic acid. In some embodiments, a break may be introduced by contacting a target nucleic acid with an effector protein and a guide nucleic acid. The guide nucleic acid may bind to the effector protein and hybridize to a region of the target nucleic acid, thereby recruiting the effector protein to the region of the target nucleic acid. Binding of the effector protein to the guide nucleic acid and the region of the target nucleic acid may activate the effector protein, and the effector protein may introduce a break (e.g., a single stranded break) in the region of the target nucleic acid. In some embodiments, editing a target nucleic acid may comprise introducing a first break in a first region of the target nucleic acid and a second break in a second region of the target nucleic acid. For example, editing a target nucleic acid may comprise contacting a target nucleic acid with a first guide nucleic acid that binds to a first effector protein and hybridizes to a first region of the target nucleic acid and a second guide nucleic acid that binds to a second programmable nickase and hybridizes to a second region of the target nucleic acid. The first effector protein may introduce a first break in a first strand at the first region of the target nucleic acid, and the second effector protein may introduce a second break in a second strand at the second region of the target nucleic acid. In some embodiments, a segment of the target nucleic acid between the first break and the second break may be removed, thereby editing the target nucleic acid. In some embodiments, a segment of the target nucleic acid between the first break and the second break may be replaced (e.g., with donor nucleic acid), thereby editing the target nucleic acid.
[0313] Methods, systems and compositions described herein may edit a target nucleic acid wherein such editing may effect one or more indels. In some embodiments, where compositions, systems, and/or methods described herein effect one or more indels, the impact on the transcription and/or translation of the target nucleic acid may be predicted depending on: 1) the amount of indels generated; and 2) the location of the indel on the target nucleic acid. For example, as described herein, in some embodiments, if the amount of indels is not divisible by three, and the indels occur within or along a protein coding region, then the edit or mutation may be a frameshift mutation. In some embodiments, if the amount of indels is divisible by three, then a frameshift mutation may not be effected, but a splicing disruption mutation and/or sequence skip mutation may be effected, such as an exon skip mutation. In some embodiments, if the amount of indels is not evenly divisible by three, then a frameshift mutation may be affected.
[0314] Methods, systems and compositions described herein may edit a target nucleic acid wherein such editing may be measured by indel activity. Indel activity measures the amount of change in a target nucleic acid (e.g., nucleotide deletion(s) and/or insertion(s)) compared to a target nucleic acid that has not been contacted by a polypeptide described in compositions, systems, and methods described herein. For example, indel activity may be detected by next generation sequencing of one or more target loci of a target nucleic acid where indel percentage is calculated as the fraction of sequencing reads containing insertions or deletions relative to an unedited reference sequence. In some embodiments, methods, systems, and compositions comprising an effector protein and guide nucleic acid described herein may exhibit about 0.0001% to about 65% or more indel activity upon contact to a target nucleic acid compared to a target nucleic acid non-contacted with compositions, systems, or by methods described herein. For example, methods, systems, and compositions comprising an effector protein and guide nucleic acid described herein may exhibit about 0.0001%, about 0.001%, about 0.01%, about 0.1%, about 1%, about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65% or more indel activity.
[0315] In some embodiments, editing of a target nucleic acid as described herein effects one or more mutations comprising splicing disruption mutations, frameshift mutations (e.g., 1+ or 2+ frameshift mutation), sequence deletion, sequence skipping, sequence reframing, sequence knock-in, or any combination thereof. In some embodiments, the splicing disruption can be an editing that disrupts a splicing of a target nucleic acid or a splicing of a sequence that is transcribed from a target nucleic acid relative to a target nucleic acid without the splicing disruption. In some embodiments, the frameshift mutation can be an editing that alters the reading frame of a target nucleic acid relative to a target nucleic acid without the frameshift mutation. In some embodiments, the frameshift mutation can be a +2 frameshift mutation, wherein a reading frame is edited by 2 bases. In some embodiments, the frameshift mutation can be a +1 frameshift mutation, wherein a reading frame is edited by 1 base. In some embodiments, the frameshift mutation is an editing that alters the number of bases in a target nucleic acid so that it is not divisible by three. In some embodiments, the frameshift mutation can be an editing that is not a splicing disruption. In some embodiments a sequence as described in reference to the sequence deletion, sequence skipping, sequence reframing, and sequence knock-in can be a DNA sequence, a RNA sequence, an edited DNA or RNA sequence, a mutated sequence, a wild-type sequence, a coding sequence, a non-coding sequence, an exonic sequence (exon), an intronic sequence (intron), or any combination thereof. In some embodiments, the sequence deletion is an editing where one or more sequences in a target nucleic acid are deleted relative to a target nucleic acid without the sequence deletion. In some embodiments, the sequence deletion can result in or effect a splicing disruption or a frameshift mutation. In some embodiments, the sequence deletion result in or effect a splicing disruption. In some embodiments, the sequence skipping is an editing where one or more sequences in a target nucleic acid are skipped upon transcription or translation of the target nucleic acid relative to a target nucleic acid without the sequence skipping. In some embodiments, the sequence skipping can result in or effect a splicing disruption or a frameshift mutation. In some embodiments, the sequence skipping can result in or effect a splicing disruption. In some embodiments, the sequence reframing is an editing where one or more bases in a target are edited so that the reading frame of the sequence is reframed relative to a target nucleic acid without the sequence reframing. In some embodiments, the sequence reframing can result in or effect a splicing disruption or a frameshift mutation. In some embodiments, the sequence reframing can result in or effect a frameshift mutation. In some embodiments, the sequence knock-in is an editing where one or more sequences is inserted into a target nucleic acid relative to a target nucleic acid without the sequence knock-in. In some embodiments, the sequence knock-in can result in or effect a splicing disruption or a frameshift mutation. In some embodiments, the sequence knock-in can result in or effect a splicing disruption.
[0316] In some embodiments, editing of a target nucleic acid can be locus specific, wherein compositions, systems, and methods described herein can edit a target nucleic acid at one or more specific loci to effect one or more specific mutations comprising splicing disruption mutations, frameshift mutations, sequence deletion, sequence skipping, sequence reframing, sequence knock-in, or any combination thereof. For example, editing of a specific locus can affect any one of a splicing disruption, frameshift (e.g., 1+ or 2+ frameshift), sequence deletion, sequence skipping, sequence reframing, sequence knock-in, or any combination thereof. In some embodiments, editing of a target nucleic acid can be locus specific, modification specific, or both. In some embodiments, editing of a target nucleic acid can be locus specific, modification specific, or both, wherein compositions, systems, and methods described herein comprise an effector protein described herein and a guide nucleic acid described herein.
[0317] Methods of editing a target nucleic acid or modulating the expression of a target nucleic acid may be performed in vivo. Methods of editing a target nucleic acid or modulating the expression of a target nucleic acid may be performed in vitro. For example, a plasmid may be edited in vitro using a composition described herein and introduced into a cell or organism. Methods of editing a target nucleic acid or modulating the expression of a target nucleic acid may be performed ex vivo. For example, methods may comprise obtaining a cell from a subject, editing a target nucleic acid in the cell with methods described herein, and returning the cell to the subject.
[0318] In some embodiments, methods of modifying described herein comprise contacting a target nucleic acid with one or more components, compositions or systems described herein. In some embodiments, the one or more components, compositions or systems described herein comprise at least one of: a) one or more effector proteins, or one or more nucleic acids encoding one or more effector proteins; and b) one or more guide nucleic acids, or one or more nucleic acids encoding one or more guide nucleic acids. In some embodiments, the one or more effector proteins introduce a single-stranded break or a double-stranded break in the target nucleic acid. [0319] In some embodiments, methods of modifying described herein comprise using one or more guide nucleic acids or uses thereof, wherein the methods modify a target nucleic acid at a single location. In some embodiments, the methods comprise contacting an RNP comprising an effector protein and a guide nucleic acid to the target nucleic acid. In some embodiments, the methods introduce a mutation (e.g., point mutations, deletions) in the target nucleic acid relative to a corresponding wildtype nucleotide sequence. In some embodiments, the methods remove or correct a disease-causing mutation in a nucleic acid sequence to produce a corresponding wildtype nucleotide sequence. In some embodiments, the methods remove/correct point mutations, deletions, null mutations, or tissue-specific mutations in a target nucleic acid. In some embodiments, the methods introduce a single stranded cleavage, a nick, a deletion of one or two nucleotides, an insertion of one or two nucleotides, a substitution of one or two nucleotides, an epigenetic modification (e.g., methylation, demethylation, acetylation, or deacetylation), or a combination thereof to the target nucleic acid. In some embodiments, the methods comprise using an effector protein and two guide nucleic acids, wherein two RNPs cleave the target nucleic acid at the same location, wherein a first RNP comprises the effector protein and a first guide nucleic acid, and wherein a second RNP comprises the effector protein and a second guide nucleic acid. In some embodiments, methods comprising using two effector protein and two guide nucleic acids, wherein both RNPs cleave the target nucleic acid at the same location, wherein a first RNP comprises a first effector protein and a first target nucleic acid, and wherein a second RNP comprises a second effector protein and a second target nucleic acid.
[0320] In some embodiments, methods of modifying described herein comprise using one or more guide nucleic acids or uses thereof, wherein the methods modify a target nucleic acid at two different locations. In some embodiments, the methods introduce two cleavage sites in the target nucleic acid, wherein a first cleavage site and a second cleavage site comprise one or more nucleotides therebetween. In some embodiments, the methods cause deletion of the one or more nucleotides. In some embodiments, the deletion restores a wild-type reading frame. In some embodiments, the wild-type reading frame produces at least a partially functional protein. In some embodiments, the deletion causes a non-wild-type reading frame. In some embodiments, a non-wild-type reading frame produces a partially functional protein or nonfunctional protein. In some embodiments, the at least partially functional protein has at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%, at least 110%, at least 120%, at least 130%, at least 140%, at least 150%, at least 180%, at least 200%, at least 300%, at least 400% activity compared to a corresponding wildtype protein. In some embodiments, the methods comprise using an effector protein and two guide nucleic acids, wherein two RNPs cleave the target nucleic acid at different locations, wherein a first RNP comprises the effector protein and a first guide nucleic acid, and wherein a second RNP comprises the effector protein and a second guide nucleic acid. In some embodiments, methods comprising using two effector protein and two guide nucleic acids, wherein both RNPs cleave the target nucleic acid at the same location, wherein a first RNP comprises a first effector protein and a first target nucleic acid, and wherein a second RNP comprises a second effector protein and a second target nucleic acid.
[0321] In some embodiments, methods of editing described herein comprise inserting a donor nucleic acid into a cleaved target nucleic acid. In some embodiments, the cleaved target nucleic acid formed by introducing a single-stranded break into a target nucleic acid. The donor nucleic acid may be inserted at a specified (e.g., effector protein targeted) point within the target nucleic acid. In some embodiments, the cleaved target nucleic acid is cleaved at a single location. In such embodiments, the methods comprise contacting a target nucleic acid with an effector protein described herein, thereby introducing a single-stranded break in the target nucleic acid; and contacting the target nucleic acid with a donor nucleic acid for homologous recombination, optionally by HDR or NHEJ, thereby introducing a new sequence into the target nucleic acid (e.g., at a cleavage site). In some embodiments, the cleaved target nucleic acid is cleaved at two locations. In such embodiments, the methods comprise contacting a target nucleic acid with an effector protein described herein, thereby introducing a single-stranded break in the target nucleic acid; contacting the target nucleic acid with a second effector protein described herein, to generate a second cleavage site in the target nucleic acid, ligating the regions flanking the first and second cleavage site, optionally through NHEJ or single-strand annealing, thereby resulting in the excision of a portion of the target nucleic acid between the first and second cleavage sites from the target nucleic acid; and contacting the target nucleic acid with a donor nucleic acid for homologous recombination, optionally by HDR or NHEJ, thereby introducing a new sequence into the target nucleic acid (e.g., in between two cleavage sites).
Donor Nucleic Acids
[0322] In some embodiments, methods comprise contacting a target nucleic acid with a donor nucleic acid. In some embodiments, composition described herein comprise a donor nucleic acid. In some embodiments, a donor nucleic acid comprises a sequence that is derived from a plant, bacteria, fungi, virus, or an animal. In some embodiments, the animal is a non-human animal, such as, by way of non-limiting example, a mouse, rat, hamster, rabbit, pig, bovine, deer, sheep, goat, chicken, cat, dog, ferret, a bird, non-human primate (e.g., marmoset, rhesus monkey). In some embodiments, the non-human animal is a domesticated mammal or an agricultural mammal. In some embodiments, the animal is a human. In some embodiments, the sequence comprises a human wild-type (WT) gene or a portion thereof.
[0323] In some embodiments, the donor nucleic acid comprises single-stranded DNA or linear double-stranded DNA. In some embodiments, the donor nucleic acid comprises a nucleotide sequence encoding a functional polypeptide and/or wherein the donor nucleic acid comprises a wildtype sequence. In some embodiments, the donor nucleic acid comprises a protein coding sequence, a gene, a gene fragment, an exon, an intron, an exon fragment, an intron fragment, a gene regulatory fragment, a gene regulatory region fragment, coding sequences thereof, or combinations thereof. In some embodiments, the donor nucleic acid comprises a naturally occurring sequence. In some embodiments, the naturally occurring sequence does not contain a mutation.
[0324] In some embodiments, the donor nucleic acid comprises a gene fragment, an exon fragment, an intron fragment, a gene regulatory region fragment, or combinations thereof. In some embodiments, the fragment is at least 10, at least 20, at least 30, at least 40, at least 50, at least 60, at least 70, or at least 80 contiguous nucleotides.
[0325] In some embodiments, a donor nucleic acid of any suitable size is integrated into a target nucleic acid or a genome. In some embodiments, the donor nucleic acid integrated into the target nucleic acid or the genome is less than 3, about 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, 9, 9.5, 10, 10.5, 11, 11.5, 12, 12.5, 13, 13.5, 14, 14.5, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, 100, 150, 200, 250, 300, 350, 400, 450, 500 kilobases in length. In some embodiments, the donor nucleic acid is more than 500 kilobases (kb) in length.
[0326] Methods may comprise contacting a target nucleic acid, including but not limited to a cell comprising the target nucleic acid, with such compositions. In some embodiments, the donor nucleic acid is inserted at a site that has been cleaved by a composition disclosed herein, for example, an effector protein, resulting in a nick or double strand break.
[0327] In reference to a viral vector, the term donor nucleic acid refers to a sequence of nucleotides that will be or has been introduced into a cell following transfection of the viral vector. The donor nucleic acid may be introduced into the cell by any mechanism of the transfecting viral vector, including, but not limited to, integration into the genome of the cell or introduction of an episomal plasmid or viral genome.
[0328] In some embodiments, an effector protein as described herein facilitates insertion of a donor nucleic acid at a site of cleavage or between two cleavage sites by cleaving (hydrolysis of a phosphodiester bond) of a nucleic acid resulting in a nick or double strand break - nuclease activity. [0329] In some embodiments, the donor nucleic acid comprises a sequence that serves as a template in the process of homologous recombination. The sequence may carry one or more nucleobase modifications that are to be introduced into the target nucleic acid. By using this donor nucleic acid as a template, the genetic information, including the modification(s), is copied into the target nucleic acid by way of homologous recombination.
Genetically modified cells and organisms
[0330] Methods of editing described herein may be employed to generate a genetically modified cell. The cell may be a eukaryotic cell (e.g., a mammalian cell) or a prokaryotic cell (e.g., an archaeal cell). The cell may be derived from a multicellular organism and cultured as a unicellular entity. The cell may comprise a heritable genetic modification, such that progeny cells derived therefrom comprise the heritable genetic mutation. The cell may be progeny of a genetically modified cell comprising a genetic modification of the genetically modified parent cell. A genetically modified cell may comprise a deletion, insertion, mutation, or non-native sequence relative to a wild-type version of the cell or the organism from which the cell was derived.
[0331] In some aspects, disclosed herein are modified cells or populations of modified cells, wherein the modified cell comprises an effector protein described herein, a nucleic acid encoding an effector protein described herein, or a combination thereof. In some embodiments, the modified cell comprises a fusion protein described herein, a nucleic acid encoding an effector protein described herein, or a combination thereof. In some embodiments, the modified cell is a modified prokaryotic cell. In some embodiments, the modified cell is a modified eukaryotic cell. A modified cell may be a modified fungal cell. In some embodiments, the modified cell is a modified vertebrate cell. In some embodiments, the modified cell is a modified invertebrate cell. In some embodiments, the modified cell is a modified mammalian cell. In some embodiments, the modified cell is a modified human cell. In some embodiments, the modified cell is in a subject. A modified cell may be in vitro. A modified cell may be in vivo. A modified cell may be ex vivo. A modified cell may be a cell in a cell culture. A modified cell may be a cell obtained from a biological fluid, organ, or tissue of a subject and modified with a composition and/or method described herein. Non-limiting examples of biological fluids are blood, plasma, serum, and cerebrospinal fluid. Non-limiting examples of tissues and organs are bone marrow, adipose tissue, skeletal muscle, smooth muscle, spleen, thymus, brain, lymph node, adrenal gland, prostate gland, intestine, colon, liver, kidney, pancreas, heart, lung, bladder, ovary, uterus, breast, and testes. Non-limiting examples of cells that may be obtained from a subject are hepatocytes, epithelial cells, endothelial cells, neurons, cardiomyocytes, muscle cells and adipocytes.
[0332] Methods of the disclosure may be performed in a cell. A cell may be in vitro. A cell may be in vivo. A cell may be ex vivo. A cell may be an isolated cell. A cell may be a cell inside of an organism. A cell may be an organism. A cell may be a cell in a cell culture. A cell may be one of a collection of cells. A cell may be a mammalian cell or derived from a mammalian cell. A cell may be a rodent cell or derived from a rodent cell. A cell may be a human cell or derived from a human cell. A cell may be a prokaryotic cell or derived from a prokaryotic cell. A cell may be a bacterial cell or may be derived from a bacterial cell. A cell may be an archaeal cell or derived from an archaeal cell. A cell may be a eukaryotic cell or derived from a eukaryotic cell. A cell may be a plant cell or derived from a plant cell. A cell may be an animal cell or derived from an animal cell. A cell may be an invertebrate cell or derived from an invertebrate cell. A cell may be a vertebrate cell or derived from a vertebrate cell. A cell may be a microbe cell or derived from a microbe cell. A cell may be a fungi cell or derived from a fungi cell. A cell may be from a specific organ or tissue. In some embodiments, the cell is a progenitor cell or derived therefrom. In some embodiments, the cell is a pluripotent stem cell or derived therefrom. In some embodiments, the cell is from a specific organ or tissue. In some embodiments, the cell is a hepatocyte. In some embodiments, the tissue is a subject’s blood, bone marrow, or cord blood. In some embodiments, the tissue is a heterologous donor blood, cord blood, or bone marrow. In some embodiments, the tissue is an allogenic blood, cord blood, or bone marrow. In some embodiments, the tissue may be muscle. In some embodiments, the muscle may be a skeletal muscle.
[0333] Non-limiting examples of cells that may be engineered or modified with compositions and methods described herein include stem cells, such as human stem cells, animal stem cells, stem cells that are not derived from human embryonic stem cells, embryonic stem cells, mesenchymal stem cells, pluripotent stem cells, induced pluripotent stem cells (iPS), somatic stem cells, adult stem cells, hematopoietic stem cells, tissue-specific stem cells. A cell may be a pluripotent cell.
[0334] Non-limiting examples of cells that may be engineered or modified with compositions and methods described herein include immune cells, such as CART, T-cells, B-cells, NK cells, granulocytes, basophils, eosinophils, neutrophils, mast cells, monocytes, macrophages, dendritic cells, antigen-presenting cells (APC), or adaptive cells.
[0335] Non-limiting examples of cells that may be engineered or modified with compositions and methods described herein include plant cells, such as parenchyma, sclerenchyma, collenchyma, xylem, phloem, germline (e.g., pollen). Cells from lycophytes, ferns, gymnosperms, angiosperms, bryophytes, charophytes, chiorophytes, rhodophytes, or glaucophytes.
[0336] Methods of the disclosure may be performed in a subject. Compositions of the disclosure may be administered to a subject. A subject may be a human. A subject may be a mammal (e.g., rat, mouse, cow, dog, pig, sheep, horse). A subject may be a vertebrate or an invertebrate. A subject may be a laboratory animal. A subject may be a patient. A subject may be suffering from a disease. A subject may display symptoms of a disease. A subject may at risk of developing a disease. A subject may not display symptoms of a disease, but still have a disease. A subject may be under medical care of a caregiver (e.g., the subject is hospitalized and is treated by a physician). In some embodiments, the subject may have a mutation associated with a gene described herein. In some embodiments, the subject may display symptoms associated with a mutation of a gene described herein. Methods of the disclosure may be performed in a plant, bacteria, or a fungus.
Agricultural Engineering
[0337] Compositions and methods of the disclosure may be used for agricultural engineering. For example, compositions and methods of the disclosure may be used to confer desired traits on a plant. A plant may be engineered for the desired physiological and agronomic characteristic using the present disclosure. In some embodiments, the target nucleic acid sequence comprises a nucleic acid sequence of a plant. In some embodiments, the target nucleic acid sequence comprises a genomic nucleic acid sequence of a plant cell. In some embodiments, the target nucleic acid sequence comprises a nucleic acid sequence of an organelle of a plant cell. In some embodiments, the target nucleic acid sequence comprises a nucleic acid sequence of a chloroplast of a plant cell.
[0338] The plant may be a dicotyledonous plant. Non-limiting examples of orders of dicotyledonous plants include Magniolales, Illiciales, Laurales, Piperales, Aristochiales, Nymphaeales, Ranunculales, Papeverales, Sarraceniaceae, Trochodendrales, Hamamelidales, Eucomiales, Leitneriales, Myricales, Fagales, Casuarinales, Caryophyllales, Batales, Polygonales, Plumbaginales, Dilleniales, Theales, Malvales, Urticales, Lecythidales, Violates, Salicales, Capparales, Ericales, Diapensales, Ebenales, Primulales, Rosales, Fabales, Podostemales, Haloragales, Myrtales, Comales, Proteales, San tales, Rafftesiales, Celastrales, Euphorbiales, Rhamnales, Sapindales, Juglandales, Geraniales, Polygalales, Umbellales, Gentianales, Polemoniales, Lamiales, Plantaginales, Scrophulariales, Campanulales, Rubiales, Dipsacales, and Asterales.
[0339] The plant may be a monocotyledonous plant. Non-limiting examples of orders of monocotyledonous plants include Alismatales, Hydrocharitales, Najadales, Triuridales, Commelinales, Eriocaulales, Restionales, Poales, Juncales, Cyperales, Typhales, Bromeliales, Zingiberales, Arecales, Cyclanthales, Pandanales, Arales, Lilliales, and Orchid ales. A plant may belong to the order, for example, Gymnospermae, Pinales, Ginkgoales, Cycadales, Araucariales, Cupressales and Gnetales.
[0340] Non-limiting examples of plants include plant crops, fruits, vegetables, grains, soy bean, corn, maize, wheat, seeds, tomatoes, rice, cassava, sugarcane, pumpkin, hay, potatoes, cotton, cannabis, tobacco, flowering plants, conifers, gymnosperms, ferns, clubmosses, hornworts, liverworts, mosses, wheat, maize, rice, millet, barley, tomato, apple, pear, strawberry, orange, acacia, carrot, potato, sugar beets, yam, lettuce, spinach, sunflower, rape seed, Arabidopsis, alfalfa, amaranth, apple, apricot, artichoke, ash tree, asparagus, avocado, banana, barley, beans, beet, birch, beech, blackberry, blueberry, broccoli, Brussel's sprouts, cabbage, canola, cantaloupe, carrot, cassava, cauliflower, cedar, a cereal, celery, chestnut, cherry, Chinese cabbage, citrus, clementine, clover, coffee, com, cotton, cowpea, cucumber, cypress, eggplant, elm, endive, eucalyptus, fennel, figs, fir, geranium, grape, grapefruit, groundnuts, ground cherry, gum hemlock, hickory, kale, kiwifruit, kohlrabi, larch, lettuce, leek, lemon, lime, locust, pine, maidenhair, maize, mango, maple, melon, millet, mushroom, mustard, nuts, oak, oats, oil palm, okra, onion, orange, an ornamental plant or flower or tree, papaya, palm, parsley, parsnip, pea, peach, peanut, pear, peat, pepper, persimmon, pigeon pea, pine, pineapple, plantain, plum, pomegranate, potato, pumpkin, radicchio, radish, rapeseed, raspberry, rice, rye, sorghum, safflower, sallow, soybean, spinach, spruce, squash, strawberry, sugar beet, sugarcane, sunflower, sweet potato, sweet com, tangerine, tea, tobacco, tomato, trees, triticale, turf grasses, turnips, vine, walnut, watercress, watermelon, wheat, yams, yew, and zucchini. A plant may include algae. SEQUENCES AND TABLES
TABLE 1. Engineered Proteins
Figure imgf000118_0001
TABLE 2. Description of Engineering
Figure imgf000118_0002
Figure imgf000119_0001
Figure imgf000120_0001
Figure imgf000121_0001
Figure imgf000122_0001
Figure imgf000123_0001
Figure imgf000124_0001
Figure imgf000125_0001
Figure imgf000126_0001
Figure imgf000127_0001
Figure imgf000128_0001
TABLE 3. Exemplary Nuclear Localization Signals
Figure imgf000128_0002
Figure imgf000129_0001
TABLE 4. Exemplary disease and syndromes.
Figure imgf000129_0002
Figure imgf000130_0001
Figure imgf000131_0001
Figure imgf000132_0001
Figure imgf000133_0001
TABLE 5. Exemplary target nucleic acids.
Figure imgf000133_0002
Figure imgf000134_0001
Figure imgf000135_0002
TABLE 6. Exemplary Effector Proteins
Figure imgf000135_0001
Figure imgf000136_0001
Figure imgf000136_0002
Figure imgf000137_0001
Figure imgf000137_0002
Figure imgf000138_0001
Figure imgf000138_0002
Figure imgf000139_0001
Figure imgf000139_0002
Figure imgf000140_0001
Figure imgf000140_0002
TABLE 7. Exemplary Handle Sequences
Figure imgf000140_0003
Figure imgf000141_0002
TABLE 8. Exemplary Engineered Effector Proteins- SEQ ID NO: 275 with multiple amino acid substitutions
Figure imgf000141_0001
Figure imgf000142_0001
Figure imgf000143_0001
Figure imgf000144_0001
Figure imgf000145_0001
EXAMPLES
[0341] The following examples are included for illustrative purposes only and are not intended to limit the scope of the invention.
Example 1. TAM Screening for Effector Proteins
[0342] To assess the ability of these protein to carry out cz.s-cleavage, an in vitro enrichment experiment is carried out. Effector proteins and guide RNAs are screened by an in vitro enrichment (IVE) assay to determine TAM recognition by each effector protein-guide RNA complex. Briefly, effector proteins are complexed with corresponding guide RNAs for 15 minutes at 37 °C. The complexes are added to an IVE reaction mix. TAM screening reactions use 10 pl of RNP in 100 pl reactions with 1,000 ng of a plasmid library containing a randomized TAM sequence (5’-NNNNNNN-3’, where N is any of A, C, G, T) in lx Cutsmart buffer and are carried out for 15 minutes at 25 °C, 45 minutes at 37 °C, and 15 minutes at 45 °C. Reactions are terminated with 1 pl of proteinase K and 5 pl of 500 mM EDTA for 30 minutes at 37 °C. Any target plasmid that is successfully cleaved has an adapter ligated to the cut end, enabling PCR amplification. Amplification products revealed by gel electrophoresis indicate effector protein-sgRNA complexes that were capable of cz.s-cleavage. An EcoRI site is included near the spacer and is used as a positive control. Next generation sequencing (NGS) is performed on cut sequences to identify enriched TAMs.
Example 2. Effector proteins edit genomic DNA in mammalian cells
[0343] Effector proteins are tested for their ability to produce indels in a mammalian cell line (e.g., HEK293T cells). Briefly, a plasmid encoding the effector proteins and a guide RNA are delivered by lipofection to the mammalian cells. This is performed with a variety of guide RNAs targeting several loci adjacent to biochemically determined TAM sequences. Indels in the loci are detected by next generation sequencing of PCR amplicons at the targeted loci and indel percentage is calculated as the fraction of sequencing reads containing insertions or deletions relative to an unedited reference sequence. “No plasmid” and Cas9 are included as negative and positive controls, respectively.
Example 3. Base Editing
[0344] A nucleic acid vector encoding a fusion protein is constructed for base editing. The fusion protein comprises a catalytically inactive variant of an effector protein fused to a deaminase. The fusion protein and at least one guide nucleic acid is tested for its ability to edit a target sequence in eukaryotic cells. Cells are transfected with the nucleic acid vector and guide nucleic acid. After sufficient incubation, DNA is extracted from the transfected cells. Target sequences are PCR amplified and sequenced by NGS and MiSeq. The presence of base modifications are analyzed from sequencing data. Results are recorded as a change in % base call relative to the negative control.
Example 4. Activation of Gene Expression with Cas Effector Fusion Polypeptide
[0345] A single stranded reporter nucleic acid encoding a fluorescent protein (e.g., enhanced green fluorescent protein (EGFP)) and a eukaryotic promoter is generated with a target sequence that is known to be recognized by complexes of effector proteins disclosed herein and corresponding guide nucleic acids. A nucleic acid vector encoding the Cas effector fused to a transcriptional activator; a guide nucleic acid; and the single stranded reporter nucleic acid encoding EGFP are introduced to eukaryotic cells via lipofection and EGFP expression is quantified by flow cytometry. Relative amounts of RNA, indicative of relative gene expression, are quantified with RT-qPCR.
Example 5. Reduction of Gene Expression with Cas Effector Fusion Polypeptide
[0346] A single stranded reporter nucleic acid encoding a fluorescent protein (e.g., enhanced green fluorescent protein (EGFP)) and a pSV40 promoter that drives constitutive expression of EGFP is generated with a target sequence that is known to be recognized by complexes of effector proteins disclosed herein and corresponding guide nucleic acids. A nucleic acid vector encoding the Cas effector fused to a transcriptional repressor (e.g., DNMT); a guide nucleic acid; and the single stranded reporter nucleic acid encoding EGFP are introduced to eukaryotic cells via lipofection and EGFP expression is quantified by flow cytometry. Relative amounts of RNA, indicative of relative gene expression, are quantified with RT-qPCR.
Example 6. Reverse transcriptase fusion for enhanced gene editing in HEK293T cells
[0347] The ability of any one of the effector proteins of TABLE 1 to perform enhanced gene editing of a target nucleic acid is assayed as follows. Components of the system include: an effector protein; a reverse transcriptase optionally fused to a MS2 coat protein; a first guide nucleic acid; a second guide nucleic acid; and a template RNA (retRNA) comprising an RT template, a primer binding sequence, and optionally an MS2 aptamer. In the instance where an aptamer and aptamer binding protein (e.g., MS2 and MS2 coat protein) are not employed, the effector protein may be fused to the reverse transcriptase. In some instances, the effector protein comprises at least one amino acid substitution relative to the respective sequence of TABLE 1, wherein the amino acid substitution is located in a RuvC domain and/or HNH domain of the effector protein, and wherein the amino acid substitution reduces the catalytic activity of the domain, thereby providing the effector protein with nickase activity. The two guide nucleic acids are designed to bind opposite strands, a target strand and a non-target strand, of the target nucleic acid, wherein the second site is downstream to the first site. HEK293T cells are transiently transfected with these components. This will generate nicking of the target strand at a first site and the non-target strand at a second site downstream of the first site. NGS sequencing is used for assessing gene editing of a target nucleic acid with this system. Without being bound by theory, such a system will advantageously enhance editing signal relative to a system that uses only one guide nucleic acid because in resolving whether to retain the edited or unedited strand of the target nucleic acid, a nick on the unedited strand signals to the cell’s repair system that it's damaged and therefore leads to preferential removal.
Example 7. DIS Dual systems obtain precise edit levels comparable to Cas9 RT editing
[0348] An exemplary effector protein, referred to as DIS dual (H247A) (SEQ ID NO: 855), was tested in a precision editing context against Cas9. Without being bound by theory, DIS dual (H247A) may be characterized as an engineered IscB protein. Briefly, HEK293T cells were transfected with three plasmids. The first plasmid encoded the nuclease (SpCas9 (H840A) or DIS dual (H247A)) and a guide nucleic acid with an MS2 aptamer. The second plasmid encoded a reverse transcriptase (MMLV or TIF1 fused to MS2 coat protein) and a retRNA. The third plasmid encoded a dual fluorescent reporter (CRISPR+ or DIS Dual editing). In the CRISPR+ assay, a stop codon is edited to allow mRhubarb expression. In the DIS Dual editing assay, 2 nt insertion is needed to reframe mRhubarb and reframe a TGA codon that needs to be corrected to allow the reporter expression. Cells were incubated for 48 hours as fluorescence read by fluorescence spectroscopy. As shown in FIGS. 4 A and 4B, these two different reporters confirmed DI S dual (H247A) obtains similar levels of precision editing as a Cas9 RT editing system. Future experiments will be performed similarly with the effector protein covalently linked to a reverse transcriptase.
TABLE 9. Sequences relevant to Example 7
Figure imgf000148_0001
Figure imgf000149_0001
Figure imgf000150_0001
Example 8. TAM Screening for Effector Protein 3107961
[0349] Effector protein 3107961 (SEQ ID NO: 856) and a guide RNA having a handle sequence of SEQ ID NO: 859 were screened by an in vitro enrichment (IVE) assay to determine TAM recognition by each effector protein-guide RNA complex. Briefly, effector protein 3107961 was complexed with corresponding guide RNAs for 15 minutes at 37°C. The complexes were added to an IVE reaction mix. IVE reactions were carried out in lx Cutsmart® buffer (New England Biolabs), using 10 pl of RNP in 100 pl reactions with 1,000 ng of a plasmid library containing a 7N TAM sequence 5’ of the target (protospacer) sequence. The reactions were carried out for 15 minutes at 25°C, followed by 45 minutes at 37°C and then 15 minutes at 45°C. Reactions were terminated with 1 pl of proteinase K and 5 pl of 500 mM EDTA for 30 minutes at 37°C. Next generation sequencing was performed on cut sequences to identify enriched TAMs. Cis cleavage was observed and examination of the position frequency matrix (PFM) derived WebLogos revealed the presence of enriched 3 TAM consensus sequences of ATAANNN. The WebLogo is shown in FIG. 5.
Example 9. Effector proteins 3745646, 3756102, 3782262, 370125, and 3778464 cleave DNA in mammalian cells
[0350] Plasmid transfections in HEK293T cells (product company) were carried out as follows. 75ng of effector protein expressing plasmid (3745646 (SEQ ID NO: 624), 3756102 (SEQ ID NO: 713), 3782262 (SEQ ID NO: 833), 370125 (SEQ ID NO: 691), and 3778464 (SEQ ID NO: 774)) and 75ng of guide expressing plasmids were delivered by lipofection to HEK293T cells, seeded at 10,000 cells in 200 pL, in 96 well plates. TransIT-293 (Minis Bio) reagent was diluted with warmed up OPTIMEM and mixed with the plasmid DNA at the ratio of 2: 1 lipid:DNA. Lipid:DNA mixture were incubated for 15 minutes at room temperature before adding 20 pL of the lipid:DNA optimem mixture to each well. Cells were incubated for 3 days before being lysed and subjected to PCR amplification. Indels were detected by next generation sequencing of PCR amplicons at the targeted loci and indel percentage was calculated as the fraction of sequencing reads containing insertions or deletions relative to an unedited reference sequence. Results are shown in TABLE 10.
TABLE 10. Sequences relevant to Example 9 and experiment results
Figure imgf000151_0001
Figure imgf000152_0001
Example 10. Engineered effector proteins cleave DNA in mammalian cells
[0351] Engineered effector proteins were evaluated for their ability to induce indels in HEK293T cells at a target locus. HEK293T cells, seeded at 30k cells per well, were cotransfected with 150ng of vector expressing an engineered effector protein and 150ng of the guide nucleic acid expressing plasmid with TransIT-293. The engineered effector proteins are represented by SEQ ID NOS: 179, and 889-910. SEQ ID NOS: 889-910 are engineered variants of SEQ ID NO: 275, having multiple amino acid substitutions relative to SEQ ID NO: 275, as described in TABLE 8. The guide nucleic acid was comprised of a spacer sequence having the sequence of 5’- AAUGGUGGAAACACAG-3’ (SEQ ID NO: 911) and a protein binding sequence of SEQ ID NO: 858, wherein the spacer sequence was located 5’ of the protein binding sequence. After 48 or 72 hours the cells were lysed using QuickExtract™ DNA Extraction Solution. Targeted loci were amplified using AccuPrime Taq polymerase and sequenced by next generation sequencing. The indel percentage was calculated as the fraction of sequencing reads containing insertions or deletions relative to an unedited reference sequence. Results are shown in FIG. 6 (SEQ ID NO: 179) and FIG. 7 (SEQ ID NOS: 889- 910)
Example 11. Precise edits are obtained with engineered variant of effector protein 3745646
[0352] HEK293T cells were transfected with plasmids encoding an engineered variant of effector protein 3745646 (SEQ ID NO: 624), MMLV-RT, a guide nucleic acid, and a retRNA to introduce precise edits in the human gene, B2M. The engineered variant of effector protein 3745646 has an amino acid substitution of H246A relative to SEQ ID NO: 624. Without being bound by theory, the H246A amino acid substitution is believed to be located in an HNH domain of effector protein 3745646, and thereby provides the engineered variant with nickase activity. Nucleotide sequences encoding these gene editing components are provided in TABLE 11. The RT -template RNA encoded a +4 nucleotide insertion. Precise editing as well as unintended byproduct edits (indels) were quantified using amplicon NGS. Results are provided in FIG. 8A. As shown in FIG. 8A, the engineered variant of effector protein 3745646 achieved 0.8% precise edits. This experiment was repeated twice (Exp 1 and Exp 2) in both the absence (Control) and presence (Enhancer) of a hHMLHl variant, amino acid sequence is provided in TABLE 11. Without being bound by theory, the hHMLHl variant is considered an inhibitor of DNA mismatch repair (MMR). The hHMLHl variant increased precise edits from 0.8% to 2%. Results are provided in FIG. 8B. Thus, hHMLHl may be considered an enhancer of precise editing systems employing engineered variants of effector protein 3745646 (SEQ ID NO: 624). Data points shown in FIG. 8B come from systems wherein the effector protein was and was not fused to the RT (levels were comparable for both conditions in these experiments).
TABLE 11. Sequences relevant to Example 10
Figure imgf000153_0001
Figure imgf000154_0001

Claims

1. A system or composition comprising an effector protein, or a nucleic acid encoding the effector protein, comprising an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to a sequence selected from TABLE 1, TABLE 6, and TABLE 8.
2. The system or composition of claim 1, comprising an engineered guide nucleic acid, or a nucleic acid encoding the guide nucleic acid, wherein the engineered guide nucleic acid comprises a protein binding sequence and a spacer sequence.
3. The system or composition of claim 2, wherein the protein binding sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 858.
4. The system or composition of claim 2, wherein the protein binding sequence comprises a handle sequence comprising a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to a sequence selected from TABLE 7.
5. The system or composition of claim 2, wherein the effector protein comprises an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 856, and wherein the handle sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 859 or 860.
6. The system or composition of claim 2, wherein the effector protein comprises an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 624, and wherein the handle sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 884.
7. The system or composition of claim 2, wherein the effector protein comprises an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 713, and wherein the handle sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 885.
8. The system or composition of claim 2, wherein the effector protein comprises an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 833, and wherein the handle sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 886.
9. The system or composition of claim 2, wherein the effector protein comprises an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 691, and wherein the handle sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 884.
10. The system or composition of claim 2, wherein the effector protein comprises an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 774, and wherein the handle sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 888.
11. The system or composition of claim 2, wherein the effector protein comprises an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 179 and 889-910, and wherein the protein binding sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 858.
12. The system or composition of any one of claims 2-11, wherein the spacer sequence is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical, complementary, or reverse complementary to a eukaryotic sequence.
13. The system or composition of any one of claims 2-12, wherein the spacer sequence is linked to a 5’ end of the protein binding sequence.
14. The system or composition of any preceding claim, wherein the effector protein comprises about 100, about 120, about 140, about 160, about 180, about 200, about 220, about 240, about 260, about 280, about 300, about 320, about 340, about 360, about 380, about 400, about 420, about 440, about 460, about 480, about 500, or about 520 contiguous amino acids of a sequence selected from TABLE 1, TABLE 6, and TABLE 8.
15. The system or composition of any preceding claim, wherein the length of the effector protein is less than 600, less than 580, less than 560, less than 540, less than 520, less than 500, less than 480, less than 460, less than 440, less than 420, or less than 400 contiguous amino acids; and at least 300, at least 320, at least 340, at least 360, at least 380 contiguous amino acids.
16. The system or composition of any preceding claim, wherein the amino acid sequence is less than 100% identical to the sequence selected from TABLE 1, TABLE 6, and TABLE 8, and wherein not more than 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 amino acid substitutions in the amino acid sequence are non-conservative amino acid substitutions relative to the respective sequence selected from TABLE 1, TABLE 6, and TABLE 8.
17. The system or composition of claim 16, wherein the effector protein comprises at least one amino acid substitution in a RuvC domain, an HNH domain, or a combination thereof.
18. The system or composition of claim 17, wherein the amino acid substitution is a nonconservative amino acid substitution.
19. The system or composition of claim 17 or 18, wherein the amino acid substitution replaces a catalytic residue of the domain(s).
20. The system or composition of claim 1, wherein the effector protein comprises an amino acid sequence that is at least 90%, at least 95%, at least 97%, at least 98%, or at least 99% identical to SEQ ID NO: 624, wherein the effector protein comprises an amino acid substitution of H246A relative to SEQ ID NO: 624.
21. The system or composition of claim 20, wherein the effector protein encoded by a nucleic acid sequence that is at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 912.
22. The system or composition of claim 1, wherein the effector protein comprises an amino acid sequence that is at least 90%, at least 95%, at least 97%, at least 98%, or at least 99% identical to SEQ ID NO: 713, wherein the effector protein comprises an amino acid substitution of H244A relative to SEQ ID NO: 713.
23. The system or composition of claim 1, wherein the effector protein comprises an amino acid sequence that is at least 90%, at least 95%, at least 97%, at least 98%, or at least 99% identical to SEQ ID NO: 856, wherein the effector protein comprises an amino acid substitution of H248A relative to SEQ ID NO: 856.
24. The system or composition of any preceding claim, wherein: a) the effector protein comprises an amino acid sequence at least 90-100% identical to SEQ ID NO: 856 and recognizes a target adjacent motif (TAM) of ATAANNN when complexed with a guide nucleic acid; b) the effector protein comprises an amino acid sequence at least 90-100% identical to SEQ ID NO: 624 and recognizes a TAM of ARRRGNN when complexed with a guide nucleic acid; c) the effector protein comprises an amino acid sequence at least 90-100% identical to SEQ ID NO: 713 and recognizes a TAM of GNAAANN when complexed with a guide nucleic acid; d) the effector protein comprises an amino acid sequence at least 90-100% identical to SEQ ID NO: 833 and recognizes a TAM of ATAANNN when complexed with a guide nucleic acid; e) the effector protein comprises an amino acid sequence at least 90-100% identical to SEQ ID NO: 691 and recognizes a TAM of GYARRNN when complexed with a guide nucleic acid; or f) the effector protein comprises an amino acid sequence at least 90-100% identical to SEQ ID NO: 774 and recognizes a TAM of RTANNNN when complexed with a guide nucleic acid; wherein N is any nucleotide, Y is C or T, and R is A or G.
25. The system or composition of any preceding claim, wherein the effector protein has nickase activity.
26. The system or composition of any preceding claim, wherein the effector protein is linked to a nuclear localization signal.
27. The system or composition of any preceding claim, further comprising a donor nucleic acid.
28. The system or composition of any preceding claim, further comprising an effector partner protein linked to the effector protein.
29. The system or composition of claim 28, wherein the effector partner comprises a polypeptide selected from a deaminase, a reverse transcriptase, a transcriptional activator, a transcriptional repressor, or a functional domain thereof.
30. The system or composition of any preceding claim, wherein the effector protein comprises at least one mutation that reduces its nuclease activity relative to the effector protein without the at least one mutation as measured in a cleavage assay, optionally wherein the effector protein is a catalytically inactive nuclease.
31. The system or composition of any preceding claim, further comprising a lipid nanoparticle containing the effector protein or the nucleic acid encoding the effector protein, the engineered guide nucleic acid, or a combination thereof.
32. The system or composition of any preceding claims, wherein the effector protein or the nucleic acid encoding the effector protein, and the engineered guide nucleic acid or the nucleic acid encoding the guide nucleic acid are provided in separate compositions.
33. The system or composition of any preceding claim, wherein the nucleic acid encoding the effector protein, the nucleic acid encoding the guide nucleic acid, or a combination thereof is an expression vector.
34. The system or composition of claim 33, wherein the expression vector is a viral vector, optionally wherein the viral vector is an adeno-associated viral (AAV) vector.
35. A system or composition comprising an engineered effector protein, wherein at least one domain of a parent sequence is replaced by one or more corresponding domains from another protein, and wherein the parent sequence is the parent sequence of TABLE 2.
36. The system or composition of claim 35, wherein the at least one domain of the parent sequence is selected from an HNH domain, a PLMP domain, a BH domain, and a TID domain.
37. The system or composition of claim 35 or 36, wherein the corresponding domain from another protein is any one of the sequences inserted described in TABLE 2.
38. The system or composition of any one of claims 35-37, comprising an engineered guide nucleic acid, or a nucleic acid encoding the guide nucleic acid, wherein the engineered guide nucleic acid comprises a protein binding sequence and a spacer sequence.
39. The system or composition of claim 38, wherein the protein binding sequence comprises a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 858
40. The system or composition of claim 38, wherein the protein binding sequence comprises a handle sequence comprising a nucleotide sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100% identical to a sequence selected from TABLE 7.
41. The system or composition of any of claims 38 to 40, wherein the spacer sequence hybridizes to a target sequence that is adjacent to a TAM of 5’-NWRRNA-3’, wherein W is A or T, N is any nucleotide, and R is A or G.
42. The system or composition of any of claims 38 to 40, wherein the spacer sequence hybridizes to a target sequence that is adjacent to a TAM of 5’-ATAANNN-3’, wherein N is any nucleotide.
43. A pharmaceutical composition comprising the system or composition of any one of claims 1-42, and a pharmaceutically acceptable excipient.
44. A method of modifying a target nucleic acid, the method comprising contacting the target nucleic acid with the system or composition of any one of claims 1-42 or the pharmaceutical composition of claim 43, thereby modifying the target nucleic acid.
45. A cell comprising the system or composition of any one of claims 1-42.
46. A cell modified by the system or composition or system of any one of claims 1-42, the pharmaceutical composition of claim 43, or the method of claim 44.
47. A cell comprising a modified target nucleic acid, wherein the modified target nucleic acid is a target nucleic acid modified according to the method of claim 44.
48. A method of treating a disease comprising administering to a subject in need thereof any one of: the system or composition of any one of claims 1-42; the pharmaceutical composition of claim 43; or the cell of any one of claims 45-47.
49. A system or composition comprising: a) an effector protein or a nucleic acid encoding the same, wherein the effector protein comprises an amino acid sequence that is at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, identical to SEQ ID NO: 855; b) a reverse transcriptase or a nucleic acid encoding the same; c) a guide nucleic acid or a nucleic acid encoding the same; and d) a retRNA or a nucleic acid encoding the same.
50. The system or composition of claim 49, wherein the reverse transcriptase comprises a peptide that is capable of binding a secondary structure present in at least one of the guide nucleic acid and the retRNA.
51. The system or composition of claim 49, wherein the effector protein or nucleic acid encoding the same is covalently linked to the reverse transcriptase or nucleic acid encoding the same.
52. The system or composition of claim 49, comprising the nucleic acid encoding the effector protein, the nucleic acid encoding the reverse transcriptase, the nucleic acid encoding the guide nucleic acid, and the nucleic acid encoding the retRNA, wherein all of the nucleic acids are present in a single expression vector, optionally wherein the expression vector is an AAV vector.
53. A method of modifying a target nucleic acid, the method comprising contacting the target nucleic acid with the system or composition of any one of claims 1-42 and 49-52.
54. The method of claim 53, comprising contacting a cell with the system or composition.
55. A system or composition comprising: a) an IscB protein; b) a reverse transcriptase or a nucleic acid encoding the same; c) a guide nucleic acid or a nucleic acid encoding the same; and d) a retRNA or a nucleic acid encoding the same.
56. A method of editing a target nucleic acid, the method comprising contacting a target nucleic acid with the system or composition of claim 55.
PCT/US2024/054826 2023-11-10 2024-11-07 Effector proteins and uses thereof Pending WO2025101679A1 (en)

Applications Claiming Priority (12)

Application Number Priority Date Filing Date Title
US202363597942P 2023-11-10 2023-11-10
US63/597,942 2023-11-10
US202363606462P 2023-12-05 2023-12-05
US63/606,462 2023-12-05
US202363615462P 2023-12-28 2023-12-28
US63/615,462 2023-12-28
US202463626307P 2024-01-29 2024-01-29
US63/626,307 2024-01-29
US202463627204P 2024-01-31 2024-01-31
US63/627,204 2024-01-31
US202463569797P 2024-03-26 2024-03-26
US63/569,797 2024-03-26

Publications (1)

Publication Number Publication Date
WO2025101679A1 true WO2025101679A1 (en) 2025-05-15

Family

ID=95696620

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2024/054826 Pending WO2025101679A1 (en) 2023-11-10 2024-11-07 Effector proteins and uses thereof

Country Status (1)

Country Link
WO (1) WO2025101679A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022087494A1 (en) * 2020-10-23 2022-04-28 The Broad Institute, Inc. Reprogrammable iscb nucleases and uses thereof
WO2023215915A1 (en) * 2022-05-06 2023-11-09 Cornell University Use of iscb in genome editing

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022087494A1 (en) * 2020-10-23 2022-04-28 The Broad Institute, Inc. Reprogrammable iscb nucleases and uses thereof
WO2023215915A1 (en) * 2022-05-06 2023-11-09 Cornell University Use of iscb in genome editing

Similar Documents

Publication Publication Date Title
JP7280312B2 (en) Novel CRISPR enzymes and systems
US11814620B2 (en) Effector proteins and methods of use
WO2023028444A1 (en) Effector proteins and methods of use
JP2023052236A (en) Novel type VI CRISPR orthologues and systems
CN116209755A (en) Programmable nucleases and methods of use
US20240384247A1 (en) Effector proteins and uses thereof
WO2023102329A2 (en) Effector proteins and uses thereof
WO2022221581A1 (en) Programmable nucleases and methods of use
US12077775B2 (en) Effector proteins and methods of use
US20240191280A1 (en) Enhanced guide nucleic acids and methods of use
US20240327812A1 (en) Fusion effector proteins and uses thereof
US20230257739A1 (en) Effector proteins and methods of use
WO2024220911A9 (en) Effector proteins, compositions, systems and methods of use thereof
WO2024138202A2 (en) Effector proteins, compositions, systems and methods of use thereof
WO2025101679A1 (en) Effector proteins and uses thereof
WO2024220715A2 (en) Effector proteins and uses thereof
WO2023092136A1 (en) Effector proteins and uses thereof
WO2024192211A2 (en) Effector proteins and uses thereof
US20250145974A1 (en) Engineered cas-phi proteins and uses thereof
WO2025201481A1 (en) Crispr-cas systems
WO2024006824A2 (en) Effector proteins, compositions, systems and methods of use thereof
WO2024196911A1 (en) Ultracompact precision editing systems and uses thereof
WO2024182440A2 (en) Methods and compositions for modifying nucleotide sequences
WO2024091958A1 (en) Effector proteins, compositions, systems and methods for the modification of serpina1
EP4423263A2 (en) Effector proteins, compositions, systems, devices, kits and methods of use thereof

Legal Events

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

Ref document number: 24889538

Country of ref document: EP

Kind code of ref document: A1