The Sequence Listing XML submitted as a file named “UHK_01282_PCT_ST26.xml”, and having a size of 323,020 bytes is hereby incorporated by reference pursuant to 37 C.F.R. § 1.834(c)(1).
The invention is generally in the field of genetic engineering and specifically in the area of CRISPR/Cas based genome editing using guide RNAs designed for enhanced stability and specificity.
CRISPR-Cas9 systems hold great promise for applying genome editing to biomedicine. CRISPR-Cas9 is a programmable gene-editing system that can be used to knock out genes and correct genetic mutations in human cells (Anzalone, et al., Nat Biotechnol. 2020, 38, (7), 824-844). This system utilizes a single guide RNA (sgRNA) that directs the Cas9 protein to the target genomic site for editing. Existing CRISPR/Cas9 toolkits exhibit varying efficiencies across loci, limiting their applicability for therapeutic genome editing. Optimization of such systems is in great need.
Applying genome editing technologies for applications in humans requires tools that are robust, reliable and specific, and a great deal of work has focused on enhancing the specificity of CRISPR/Cas9. Two main approaches have been taken to optimize CRISPR/Cas9 system activity: 1) by modification of the Cas9 protein and 2) by optimization of the sgRNA. Approaches involving Cas9 protein engineering have primarily focused on improving its specificity and targeting scope via directed evolution and targeted mutagenesis (Kleinstiver, et al., Nature 2016, 529, (7587), 490-5; Slaymaker, et al., Science 2016, 351, (6268), 84-8; Hu, et al., Nature 2018, 556, (7699), 57-63; Nishimasu, et al., Science 2018, 361, (6408), 1259-1262; Kleinstiver, et al., Nature 2015, 523, (7561), 481-5; Casini, et al., Nat Biotechnol 2018; Chen, et al., Nature 2017, 550, (7676), 407-410; Choi, et al., Nat Methods 2019, 16, (8), 722-730; Lee, et al., Nat Commun 2018, 9, (1), 3048; and Vakulskas, et al., Nat Med 2018, 24, (8), 1216-1224).
The other approach focuses on optimizing the sgRNAs used. The protospacer sequence of sgRNA is responsible for target site recognition, whereas its scaffold sequence binds to Cas9, which results in the conformational change of Cas9 for its activation. Many studies have been done on elucidating the determinants in the protospacer sequence for sgRNAs to exhibit high on-target and low off-target activities (Hanna, et al., Nat Biotechnol 2020, 38, (7), 813-823). However, specific loci, including therapeutically relevant ones, may have limited choices of protospacer sequences for targeting, and many protospacer sequences result in only a moderate or even low percentage of editing.
The scaffold sequence of sgRNA can be engineered to alter the overall editing activity by increasing its stability and assembly with the Cas9 protein. The “E+F” scaffold variant was engineered with a 5-nucleotide-extended tetraloop that could strengthen the scaffold's interaction with SpCas9 and an A-U base-pair flip in the lower stem that removes a putative polymerase-III terminator sequence (Chen, et al., Cell 2013, 155, (7), 1479-91). The E+F scaffold sequence was further mutated with different substitutions, and specific regions were identified to be more tolerant of mutations without compromising the sgRNA's activity (Jost, et al., Nat Biotechnol 2020, 38, (3), 355-364). Six scaffold variants, three of them containing additional U61C+A66G mutations besides those in the E+F scaffold, were reported to generate more edits. Apart from these efforts, there has been limited success in enhancing SpCas9's activity. Existing engineered guide RNA scaffolds that increase on-target editing of the widely used Streptococcus pyogenes Cas9 (SpCas9) nuclease greatly compromise its on-to-off targeting specificity. No guide RNA scaffold variant with both enhanced efficiency and high genome-wide accuracy has been described for SpCas9. No SpCas9 variant reported to date has exhibited enhanced activity. Also, whether these engineered scaffolds increase off-target edits, which is an important concern for applications of genome editing, has not been evaluated.
Therefore, it is an object of the invention to provide enhanced reagents and methods for CRISPR-Cas9 genomic engineering with enhanced on-site activity and greater specificity than existing reagents.
It is also an object of the invention to provide compositions and methods for genome editing with enhanced on-site activity and minimal off-targeting.
It is a further object of the invention to provide CRISPR-Cas9 editors that generate more edits to attain functional outcomes at loci associated with modest editing using wild type editors.
Variant guide RNA scaffolds that impart enhanced editing activity and high genome-wide targeting specificity in human cells have been developed. The engineered variant guide RNA scaffolds implement activity-enhancing mutations that enhance their editing activities as compared with wild-type guide RNA scaffolds and pre-existing variants.
Variant single guide RNA (sgRNA) including substitution and/or addition of one or more nucleic acid residues that strengthens the interaction of the sgRNA with a Cas enzyme are provided. Typically, the strengthened interaction imparts increased on-target editing and/or increased on-off target specificity relative to a wild type sgRNA that lacks the substitution and/or addition of one or more nucleic acid residues. In some forms, the substitution and/or addition of one or more nucleic acid residues that strengthens the interaction of the sgRNA with a Cas enzyme includes substitution and/or addition of one or more nucleic acid residues within the stem-loop 2 region of the sgRNA. Typically, the Cas enzyme is a Cas9 enzyme, such as the Cas9 enzyme derived from Streptococcus pyogenes (SpCas9). In some forms, the substitution and/or addition of one or more nucleic acid residues strengthens the sgRNAs interaction with residue His721 and/or the PI domain of SpCas9.
In some forms, the variant sgRNA includes a framework region of a wild-type sgRNA having the nucleic acid sequence:
whereby “—X—” represents a hairpin region of stem-loop 2 including between 12 and 24 nucleic acid residues, inclusive. In some forms, the stem-loop 2 region includes the nucleic acid sequence of any one of SEQ ID NOS: 1-312. In particular forms, the stem-loop 2 region includes the nucleic acid sequence GCGGGGUGCCGC (SEQ ID NO:48), or a nucleic acid sequence having at least 75%, up to 99% identity to SEQ ID NO:48. In some forms, the variant sgRNA includes a hairpin region of stem-loop 2 having a nucleic acid sequence having at least 75% sequence identity to GCGGGGUGCCGC (SEQ ID NO:48). For example, in some forms, the variant sgRNA includes a hairpin region of stem-loop 2 having a nucleic acid sequence at least 80%, at least 85%, at least 90%, or at least 95% identical to GCGGGGUGCCGC (SEQ ID NO:48). In other forms, the stem-loop 2 region includes the nucleic acid sequence GGGCCGGGGUGCCGGCCC (SEQ ID NO:240), or a nucleic acid sequence having at least 75%, up to 99% identity to SEQ ID NO:240. In some forms, the variant sgRNA includes a hairpin region of stem-loop 2 having a nucleic acid sequence having at least 75% sequence identity to GGGCCGGGGUGCCGGCCC (SEQ ID NO:240). For example, in some forms, the variant sgRNA includes a hairpin region of stem-loop 2 having a nucleic acid sequence at least 80%, at least 85%, at least 90%, or at least 95% identical to GGGCCGGGGUGCCGGCCC (SEQ ID NO:240). In one form, a variant sgRNAs that imparts increased on-target editing and/or increased on-off target specificity relative to a wild type sgRNA includes the nucleic acid sequence:
In some forms, the variant sgRNA includes a nucleic acid sequence having at least 75% sequence identity to SEQ ID NO:352. For example, in some forms, the variant sgRNA includes a nucleic acid sequence at least 80%, at least 85%, at least 90%, or at least 95% identical to SEQ ID NO:352. In another form, a variant sgRNAs that imparts increased on-target editing and/or increased on-off target specificity relative to a wild type sgRNA includes the nucleic acid sequence:
In some forms, the variant sgRNA includes a nucleic acid sequence having at least 75% sequence identity to SEQ ID NO:353. For example, in some forms, the variant sgRNA includes a nucleic acid sequence at least 80%, at least 85%, at least 90%, or at least 95% identical to SEQ ID NO:353.
Ribonucleoprotein complexes including the variant sgRNAs are also described. Typically, the ribonucleoprotein complexes include: (a) a Cas9 enzyme; and (b) a variant sgRNA, whereby the variant sgRNA includes a stem-loop 2 region including the nucleic acid sequence of any one of SEQ ID NOs:1-312, and whereby the ribonucleoprotein complex has increased on-target editing and/or increased on-off target specificity relative to the corresponding complex between a Cas9 enzyme and wild type sgRNA. In some forms, the Cas9 enzyme is derived from Streptococcus pyogenes (SpCas9). Generally, the variant sgRNA includes a framework having the nucleic acid sequence:
whereby “—X—” represents the stem-loop 2 region including the nucleic acid sequence of any one of SEQ ID NOs: 1-312. In some forms, the ribonucleoprotein complex includes a variant sgRNA having the nucleic acid sequence:
In some forms, the variant sgRNA includes a nucleic acid sequence having at least 75% sequence identity to SEQ ID NO:352. For example, in some forms, the variant sgRNA includes a nucleic acid sequence at least 80%, at least 85%, at least 90%, or at least 95% identical to SEQ ID NO:352. In some forms, the ribonucleoprotein complex includes a variant sgRNA having the nucleic acid sequence:
In some forms, the variant sgRNA includes a nucleic acid sequence having at least 75% sequence identity to SEQ ID NO:353. For example, in some forms, the variant sgRNA includes a nucleic acid sequence at least 80%, at least 85%, at least 90%, or at least 95% identical to SEQ ID NO:353. Vectors encoding of expressing the variant sgRNA and/or the ribonucleoprotein complex thereof, and cells including these compositions are also provided.
Methods for CRISPR-based editing of one or more target genes in a cell are also provided. Generally, the methods include administering into and/or expressing within the cell the variant sgRNA and/or the ribonucleoprotein complex thereof, wherein the variant sgRNA is configured to target the one or more target genes. The administering can be in vitro or in vivo.
Kits including the variant sgRNAs are also disclosed. In some forms, the kits include instructions for performing a method of CRISPR-based editing of one or more target genes, and/or a Cas9 enzyme, or vector encoding or expressing the Cas9 enzyme.
The terms “nucleic acid,” “nucleic acid sequence,” “nucleic acid fragment,” “oligonucleotide,” and “polynucleotide” are used interchangeably and are intended to include, but not limited to, a polymeric form of nucleotides that may have various lengths, either deoxyribonucleotides (DNA) or ribonucleotides (RNA), or analogs or modified nucleotides thereof, including, but not limited to locked nucleic acids (LNA), peptide nucleic acids (PNA), and morpholinos. An oligonucleotide is typically composed of a specific sequence of four nucleotide bases: adenine (A); cytosine (C); guanine (G); and thymine (T) (uracil (U) for thymine (T) when the polynucleotide is RNA). Thus, the term “oligonucleotide sequence” is the alphabetical representation of a polynucleotide molecule; alternatively, the term may be applied to the polynucleotide molecule itself. This alphabetical representation can be input into databases in a computer having a central processing unit and used for bioinformatics applications such as functional genomics and homology searching. Oligonucleotides may optionally include one or more non standard nucleotide(s), nucleotide analog(s) and/or modified nucleotides. In some cases nucleotide sequences are provided using character representations recommended by the International Union of Pure and Applied Chemistry (IUPAC) or a subset thereof. IUPAC nucleotide codes used herein include, A=Adenine, C=Cytosine, G=Guanine, T=Thymine, U=Uracil, R=A or G, Y=C or T, S=G or C, W=A or T, K=G or T, M=A or C, B=C or G or T, D=A or G or T, H=A or C or T, V=A or C or G, N=any base, “.” or “-”=gap. In some forms the set of characters is (A, C, G, T, U) for adenosine, cytidine, guanosine, thymidine, and uridine respectively. In some forms the set of characters is (A, C, G, T, U, I, X) for adenosine, cytidine, guanosine, thymidine, uridine, inosine, xanthosine, respectively. The modified sequences, non-natural sequences, or sequences with modified binding, may be in the genomic, the guide or the tracr sequences.
As used herein, the terms “percent (%) sequence identity,” or “% identical to (sequence)” are used interchangeably and are defined as the percentage of nucleotides or amino acids in a candidate sequence that are identical with the nucleotides or amino acids in a reference nucleic acid sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity. Alignment for purposes of determining percent sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN, ALIGN-2 or Megalign (DNASTAR) software. Appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the full-length of the sequences being compared can be determined by known methods.
The terms “protein” “polypeptide” or “peptide” refer to a natural or synthetic molecule including two or more amino acids linked by the carboxyl group of one amino acid to the alpha amino group of another.
The term “polynucleotide” or “nucleic acid” or “nucleic acid sequence” refers to a natural or synthetic molecule including two or more nucleotides linked by a phosphate group at the 3′ position of one nucleotide to the 5′ end of another nucleotide. The polynucleotide is not limited by length, and thus the polynucleotide can include deoxyribonucleic acid (DNA) or ribonucleic acid (RNA).
A cell can be in vitro. Alternatively, a cell can be in vivo and can be found in a subject. A “cell” can be a cell from any organism including, but not limited to, a bacterium.
The terms “editing fidelity” or “editing efficiency” or “targeting accuracy” or “on-target editing” or “on-off target specificity” or “on-target editing efficiency” are understood to mean the percentage of desired mutation achieved and are measured by the precision of the sgRNA variant in altering the DNA construct of the targeted gene with minimal off-target editing. A DNA editing efficiency of 1 (or 100%) indicates that the number of edited cells and/or edited alleles obtained when the sgRNA variant is used is approximately equal or equal to the number of edited cells and/or edited alleles obtained when the wild type or parent sgRNA variant is used. Conversely, a DNA editing efficiency greater than 1 indicates that the number of edited cells obtained when the sgRNA variant used is greater than the number of edited cells obtained when the parent sgRNA variant is used. In this case, the sgRNA variant has improved properties, for example improved editing efficiency when compared to the parent sgRNA.
The terms “single guide RNA” or “sgRNA” refer to the polynucleotide sequence comprising the guide sequence, tracr sequence and the tracr mate sequence. “Guide sequence” refers to the around 20 base pair (bp) sequence within the guide RNA that specifies the target site and may be used interchangeably with the terms “guide” or “spacer.”
The term “stem-loop 2 region” refers to the polynucleotide sequence of the second hairpin structure of the sgRNA and the flanking sequence.”
The terms “genome editing,” “genome engineering” or “genome mutagenesis” refer to selective and specific changes to one or more targeted genes or DNA sequences within a recipient cell through programming of the CRISPR-Cas system within the cell. The editing or changing of a targeted gene or genome can include one or more of a deletion, knock-in, point mutation, substitution mutation or any combination thereof in one or more genes of the recipient cell.
The terms “vector” or “expression vector” refer to a system suitable for delivering and expressing a desired nucleotide or protein sequence. Some vectors may be expression vectors, cloning vectors, transfer vectors etc.
The term “variant” or “mutant,” as used herein refer to an artificial outcome that has a pattern that deviates from what occurs in nature. In the context of the disclosed sgRNA variants, “variant” refers to a sgRNA that has one or more nucleic acid changes in the scaffold region relative to wildtype sgRNA scaffold region (e.g., SEQ ID NO:345), or relative to a starting, base, or reference sgRNA, such as “E+F” (SEQ ID NO:346); “U61C/A66G” (SEQ ID NO:347); “U61C/A66G” (SEQ ID NO:348); “E+F G62A/A64G” (SEQ ID NO:349); “G62A/A64G” (SEQ ID NO:350); and “5E” (SEQ ID NO:351). Note that the disclosed sgRNA variants have one or nucleic amino acid changes relative to a reference, base, or starting sgRNA (such as, e.g., wildtype sgRNA or “E+F”; “U61C/A66G”; “U61C/A66G”; “E+F G62A/A64G”; “G62A/A64G”; and “5E”. While some such reference, base, or starting sgRNAs (such as, e.g., G62A/A64G) are themselves a “variant” of another or other sgRNA, these reference, base, or starting sgRNAs are not a disclosed variant as described herein, and reference herein to such reference, base, or starting sgRNAs as a “variant” sgRNA is not intended to, and does not, indicate that such reference, base, or starting sgRNAs are a disclosed variant that impart enhanced editing, as described herein.
The terms “Protospacer adjacent motif” or “PAM sequence” or “PAM interaction region” refer to short pieces of genetic code that flag editable sections of DNA and serve as a binding signal for specific CRISPR-Cas nucleases. The PAM interaction region in the wild-type SaCas9 or its variants contains amino acid residues 910-1053 (Nishimasu, et al. Cell, 162, 1113-1126, doi: 10.1016/j.cell.2015.08.007 (2016)) and includes a conserved 13-amino acid region spanning positions 982 to 994 which plays a role in binding to the 4th and 5th bases of the PAM (Ma, et al. Nature Communications, 10, 560, doi: 10.1038/s41467-019-08395-8 (2019)).
The terms “Cas9,” “Cas9 protein,” or “Cas9 nuclease” refer to a RNA-guided endonuclease that is a Cas9 protein that catalyzes the site-specific cleavage of double stranded DNA. Also, referred to as “Cas nuclease” or “CRISPR-associated nuclease.”
The term “mutation” refers to a substitution of a residue within a sequence, e.g., a nucleic acid or amino acid sequence, with another residue, or a deletion or insertion of one or more residues within a sequence. Mutations are described by identifying the original residue followed by the position of the residue within the sequence and by the identity of the change in residue. For the purposes of this disclosure, amino acid positions are identified using the amino acid positions shown in SpCas9 sequence UniProtKB/Swiss-Prot No. Q99ZW2 (PDB ID NO:600Y), with the numbering beginning at the initial methionine residue. Various methods for making the mutations in the amino acids provided herein are well known in the art, and are provided by, for example, Green and Sambrook, Molecular Cloning: A Laboratory Manual 4th Edition, Cold Spring Harbor Laboratory Press, (2012).
The use of the terms “a,” “an,” “the,” and similar referents in the context of describing the present disclosure (especially in the context of the claims) are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context.
Recitation of ranges of values herein are merely intended to serve as a shorthand method of referring individually to each separate value falling within the range, unless otherwise indicated herein, and each separate value is incorporated into the specification as if it were individually recited herein.
Use of the term “about” is intended to describe values either above or below the stated value in a range of approx. +/−10%; in other forms the values may range in value either above or below the stated value in a range of approx. +/−5%; in other forms the values may range in value either above or below the stated value in a range of approx. +/−2%; in other forms the values may range in value either above or below the stated value in a range of approx. +/−1%. The preceding ranges are intended to be made clear by context, and no further limitation is implied. All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g., “such as”) provided herein, is intended merely to better illuminate the disclosure and does not pose a limitation on the scope of the disclosure unless otherwise claimed. No language in the specification should be construed as indicating any non-claimed element as essential to the practice of the disclosure.
Disclosed are materials, compositions, and components that can be used for, can be used in conjunction with, can be used in preparation for, or are products of the disclosed method and compositions. These and other materials are disclosed herein, and it is understood that when combinations, subsets, interactions, groups, etc. of these materials are disclosed that while specific reference of each various individual and collective combinations and permutation of these compounds may not be explicitly disclosed, each is specifically contemplated and described herein. For example, if a ligand is disclosed and discussed and a number of modifications that can be made to a number of molecules including the ligand are discussed, each and every combination and permutation of ligand and the modifications that are possible are specifically contemplated unless specifically indicated to the contrary. Thus, if a class of molecules A, B, and C are disclosed as well as a class of molecules D, E, and F and an example of a combination molecule, A-D is disclosed, then even if each is not individually recited, each is individually and collectively contemplated. Thus, in this example, each of the combinations A-E, A-F, B-D, B-E, B-F, C-D, C-E, and C-F are specifically contemplated and should be considered disclosed from disclosure of A, B, and C; D, E, and F; and the example combination A-D. Likewise, any subset or combination of these is also specifically contemplated and disclosed. Thus, for example, the sub-group of A-E, B-F, and C-E are specifically contemplated and should be considered disclosed from disclosure of A, B, and C; D, E, and F; and the example combination A-D. Further, each of the materials, compositions, components, etc. contemplated and disclosed as above can also be specifically and independently included or excluded from any group, subgroup, list, set, etc. of such materials.
These concepts apply to all aspects of this application including, but not limited to, steps in methods of making and using the disclosed compositions. Thus, if there are a variety of additional steps that can be performed it is understood that each of these additional steps can be performed with any specific form or combination of forms of the disclosed methods, and that each such combination is specifically contemplated and should be considered disclosed.
All methods described herein can be performed in any suitable order unless otherwise indicated or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g., “such as”) provided herein, is intended merely to better illuminate the forms and does not pose a limitation on the scope of the forms unless otherwise claimed. No language in the specification should be construed as indicating any non-claimed element as essential to the practice of the disclosure.
Variant guide RNA scaffolds that impart enhanced editing activity and high genome-wide targeting specificity in human cells have been developed. The engineered variant guide RNA scaffolds implement activity-enhancing mutations that enhance their editing activities as compared with wild-type guide RNA scaffolds and pre-existing variants. An advantage of the CRISPR-Cas system is that a single Cas protein can be programmed by guide molecules to recognize a specific nucleic acid target. In other words the CRISPR-Cas protein can be recruited to a specific nucleic acid target locus of interest using said guide molecule.
The term “CRISPR” (Clustered Regularly Interspaced Short Palindromic Repeats) is an acronym for DNA loci that contain multiple, short, direct repetitions of base sequences. The prokaryotic CRISPR/Cas system has been adapted for use as gene editing (silencing, enhancing or changing specific genes) for use in eukaryotes (see, for example, Cong, Science, 15:339(6121):819-823 (2013) and Jinek, et al., Science, 337(6096):816-21 (2012)). Methods of preparing compositions for use in genome editing using the CRISPR/Cas systems are described in detail in WO 2013/176772 and WO 2014/018423, which are specifically incorporated by reference herein in their entireties.
In general, the term “CRISPR system” refers collectively to transcripts and other elements involved in the expression of or directing the activity of CRISPR-associated (“Cas”) genes, including sequences encoding a Cas gene, a tracr (trans-activating CRISPR) sequence (e.g., tracrRNA or an active partial tracrRNA), a tracr-mate sequence (encompassing a “direct repeat” and a tracrRNA-processed partial direct repeat in the context of an endogenous CRISPR system), a guide sequence (also referred to as a “spacer” in the context of an endogenous CRISPR system), or other sequences and transcripts from a CRISPR locus. One or more tracr mate sequences operably linked to a guide sequence (e.g., direct repeat-spacer-direct repeat) can also be referred to as pre-crRNA (pre-CRISPR RNA) before processing or crRNA after processing by a nuclease. Typically, a CRISPR-Cas9 system includes a guide RNA (gRNA) and Cas9 nuclease, which together form a ribonucleoprotein (RNP) complex. The presence of a specific protospacer adjacent motif (PAM) in the genomic DNA is required for the gRNA to bind to the target sequence. The Cas9 nuclease then makes a double-strand break in the DNA. Endogenous repair mechanisms triggered by the double-strand break may result in gene knockout via a frameshift mutation or knock-in of a desired sequence if a DNA template is present.
In some forms, a tracrRNA and crRNA are linked and form a chimeric crRNA-tracrRNA hybrid where a mature crRNA is fused to a partial tracrRNA via a synthetic stem loop to mimic the natural crRNA:tracrRNA duplex as described in Cong, Science, 15:339(6121):819-823 (2013) and Jinek, et al., Science, 337(6096):816-21 (2012)). A single fused crRNA-tracrRNA construct can also be referred to as a guide RNA or gRNA (or single-guide RNA (sgRNA)). Within an sgRNA, the crRNA portion can be identified as the ‘target sequence’ and the tracrRNA is often referred to as the ‘scaffold’.
CRSIPR systems having enhanced editing activity and high genome-wide targeting specificity typically include two components: (1) a single guide RNA configured for enhanced editing activity; and (2) a Cas enzyme.
It has been established that engineering the activity of an enzyme and its working component (in this case the sgRNA scaffold for Cas9 enzyme) by introducing modifications to the component typically increases or decreases both the on-target and the off-target activities simultaneously. However, it has been established that the described sgRNA scaffold variants decrease undesired off-target activity while also increasing on-target activity at targeted genomic loci (e.g., HBG loci, as indicated in the Examples). Therefore, the described variants achieve accurate and efficient genome editing at any user-defined target.
In some forms, a variant single guide RNA (sgRNA) includes substitution and/or addition of one or more nucleic acid residues that strengthens the interaction of the sgRNA with a Cas enzyme,
In some forms, a variant single guide RNA (sgRNA) includes substitution and/or addition of one or more nucleic acid residues that strengthens the interaction of the sgRNA with a Cas enzyme, whereby the strengthened interaction imparts decreased off-target activity while also increasing on-target activity at a targeted genomic locus relative to a wild type sgRNA that lacks the substitution and/or addition of one or more nucleic acid residues.
A. Single Guide RNA (sgRNA)
The single guide RNA is a specific RNA sequence that recognizes the target DNA region of interest and directs the Cas nuclease there for editing. The gRNA is made up of two parts: CRISPR RNA (crRNA), a 17-20 nucleotide spacer sequence complementary to the target DNA and a conserved repeat fragment (“handle” or “tag”) region that pairs with the tracr RNA, and a tracr RNA, which serves as a binding scaffold for the Cas nuclease. The crRNA component imparts specificity of CRISPR-directed nuclease activity and is the customizable component that directs specific editing.
sgRNA is an abbreviation for “single guide RNA.” sgRNA is a single RNA molecule that contains both the custom-designed short crRNA sequence fused to the scaffold tracrRNA sequence. sgRNA is synthetically generated or made in vitro or in vivo from a DNA template.
While crRNAs and tracrRNAs exist as two separate RNA molecules in nature, sgRNAs include both a crRNA component and a scaffold component fused as a single molecule. The nucleic acid sequence of the scaffold of a wildtype sgRNA appended with corresponding structural features is presented in
In some forms, the nucleic acid sequence of a wild-type sgRNA scaffold sequence is:
In the complete sgRNA, the guide sequence immediately precedes the first nucleotide of the tracr sequence. In some forms, the different regions of an sgRNA scaffold sequence are defined by the secondary structural elements formed within the sequence of scaffold RNA. For example, in some forms, the sgRNA scaffold sequence includes the structural elements set forth in
As described herein, the sgRNA scaffold stem-loop 2 region includes a hairpin region, as well as flanking regions. The flanking regions includes 6 nucleotides (i.e., at positions 44-48 of SEQ ID NO:345 and at position 61 of SEQ ID NO:345) and all other residues within the stem-loop 2 region form the “hairpin region”. For example, in the wild-type sgRNA scaffold having a sequence of SEQ ID NO:345, the flanking region includes nucleotides in positions 44-48 and 61, and the “hairpin region of stem-loop 2” includes 12 nucleotides in positions 49-60.
In some forms, the sgRNA scaffold sequence includes all components of a wild-type sgRNA directly preceding the stem-loop 2 region, and having the nucleic acid sequence:
In some forms, the sgRNA scaffold sequence includes all components of a wild-type sgRNA directly preceding the hairpin region of stem-loop 2, and having the nucleic acid sequence:
In some forms, the sgRNA scaffold sequence includes all components of a wild-type sgRNA directly following the stem-loop 2 region, having the nucleic acid sequence: GCACCGAGUCGGUGCU (SEQ ID NO:358).
In some forms, the sgRNA scaffold sequence includes all components of the wild type sgRNA, but with the hairpin region of stem-loop 2 substituted. For example, in some forms, a sgRNA scaffold includes the sequence:
whereby “—X—” represents between 12 and 24 nucleic acid residues corresponding to a hairpin region of stem-loop 2.
An exemplary stem-loop 2 region of wild-type sgRNA scaffold is:
whereby the hairpin region of stem-loop 2 includes the 12-nucleotide sequence:
1. Variant sgRNAs (sgRNA)
Multiple variant sgRNAs are known in the art to alter or otherwise mediate the editing activity of CRISPR/Cas relative to the Wt sgRNA. Exemplary variant sgRNAs that are known in the art include:
“E+F”, having a nucleic acid sequence of:
“(CR772) E+F U61C/A66G”, having a nucleic acid sequence of:
“U61C/A66G”, having a nucleic acid sequence of:
“E+F G62A/A64G”, having a nucleic acid sequence of:
“G62A/A64G”, having a nucleic acid sequence of:
and
“5E”, having a nucleic acid sequence of:
2. Variant sgRNAs Enhancing Editing
Variant sgRNAs that enhance the specificity and activity of the editing activity of CRISPR/Cas relative to the Wt sgRNA have been developed. In some forms, the variant sgRNAs enhance the specificity and activity of the editing activity of CRISPR/Cas relative to the Wt sgRNA by increasing the stability of the interaction with the Cas enzyme. Therefore, compositions of variant sgRNAs that have increased stability of the interaction with the Cas enzyme relative to the Wt sgRNA and which have enhanced specificity and activity of on-target editing activity of CRISPR/Cas relative to the Wt are described. Exemplary variant sgRNAs which have enhanced specificity and activity of on-target editing activity of CRISPR/Cas relative to the Wt include variants of the stem-loop 2 region.
Exemplary variant sgRNA scaffolds with enhanced specificity and activity of on-target editing activity of CRISPR/Cas relative to the Wt having variants of the hairpin region of stem-loop 2 are set forth in Table 4. Therefore, in some forms, the variant sgRNA scaffold with enhanced specificity and activity of on-target editing activity of CRISPR/Cas relative to the Wt includes a hairpin region of stem-loop 2 having a sequence of nucleic acids of any one of the sequences in Table 4. For example, in some forms, the variant sgRNA scaffold with enhanced specificity and activity of on-target editing activity of CRISPR/Cas relative to the Wt includes a hairpin region of stem-loop 2 having a sequence of nucleic acids of any one of SEQ ID NOs:1-312.
In some forms, the variant strengthens the scaffold's interaction with SpCas9 via His721 and the PI domain of SpCas9. For example, in some forms, the variant has a hairpin region of stem-loop 2 including the nucleic acid sequence:
In some forms, the variant has a hairpin region of stem-loop 2 including the nucleic acid sequence:
In some forms, the variant includes all or part of a “framework” sgRNA, such as that of the wild type sgRNA scaffold (residues corresponding to the stem-loop 2 region are in boldface):
UAUCAACUUGAAAAAGUGGCACCGAGUCGGUGCU.
In some forms, the variant of the sgRNA includes the entire wild type sgRNA scaffold, but with the hairpin region of stem-loop 2 substituted. Therefore, an exemplary sgRNA includes the sequence:
whereby “—X—” represents between 12 and 24 nucleic acid residues corresponding to a hairpin region of stem-loop 2. For example, in some forms, the variant of the sgRNA including the entire wild type sgRNA scaffold with the hairpin region of stem-loop 2 substituted includes the sequence:
whereby each “—N—” independently represents either “A”, “U”, “C” or “G”, respectively.
In some forms, the sgRNA includes SEQ ID NO:354, whereby “—X—” represents any one of SEQ ID NOs: 1-312.
In some forms, the variant sgRNA includes a hairpin region of stem-loop 2 corresponding to SEQ ID NO:48. Therefore, in some forms, the variant sgRNA has a nucleic acid sequence of:
In some forms, the variant sgRNA includes a hairpin region of stem-loop 2 corresponding to SEQ ID NO:240. Therefore, in some forms, the variant sgRNA has a nucleic acid sequence of:
In other forms, the variant sgRNA includes a hairpin region of stem-loop 2 corresponding to a variant having at least 75%, up to 99% identity to SEQ ID NO:48 or SEQ ID NO:240.
The term “identity,” as used herein, can be readily calculated by known methods, including, but not limited to, those described in Computational Molecular Biology, Lesk, A. M., Ed., Oxford University Press, New York, 1988; Biocomputing: Informatics and Genome Projects, Smith, D. W., Ed., Academic Press, New York, 1993; Computer Analysis of Sequence Data, Part I, Griffin, A. M., and Griffin, H. G., Eds., Humana Press, New Jersey, 1994; Sequence Analysis in Molecular Biology, von Heinje, G., Academic Press, 1987; and Sequence Analysis Primer, Gribskov, M. and Devereux, J., Eds., M Stockton Press, New York, 1991; and Carillo, H., and Lipman, D., SIAM J Applied Math., 48: 1073 (1988). Preferred methods to determine identity are designed to give the largest match between the sequences tested. Methods to determine identity and similarity are codified in publicly available computer programs. The percent identity between two sequences can be determined by using analysis software (i.e., Sequence Analysis Software Package of the Genetics Computer Group, Madison Wis.) that incorporates the Needelman and Wunsch, (J. Mol. Biol., 48: 443-453, 1970) algorithm (e.g., NBLAST, and XBLAST). In some forms, the default parameters can be used to determine the identity for the polynucleotides of the present disclosure. In some forms, the % sequence identity of a given nucleic acid sequence “C” to, with, or against a given nucleic acid or amino acid sequence “D” (which can alternatively be phrased as a given sequence C that has or includes a certain % sequence identity to, with, or against a given sequence D) is calculated as follows:
100 times the fraction W/Z,
where W is the number of nucleotides or amino acids scored as identical matches by the sequence alignment program in that program's alignment of C and D, and where Z is the total number of nucleotides in D. It will be appreciated that where the length of sequence C is not equal to the length of sequence D, the % sequence identity of C to D will not equal the % sequence identity of D to C.
In other forms, the variant sgRNA includes a hairpin region of stem-loop 2 corresponding to a variant having at least 75%, up to 99% identity to GCGGGGUGCCGC (“SV48”; SEQ ID NO:48). For example in some forms, the variant has a hairpin region of stem-loop 2 corresponding to a variant having at least about 75%, 80%, 85%, 90%, or 95% identity to SEQ ID NO:48. Therefore, in some forms, the variant sgRNA has a hairpin region of stem-loop 2 with a nucleic acid sequence that has one or more nucleotides different to SEQ ID NO:48, such as one or more substitutions, deletions or additions at any one of the nucleotide positions of SEQ ID NO:48.
In some forms, the variant sgRNA includes a hairpin region of stem-loop 2 having one, two, three, four, or five residues that are substituted, deleted, or added relative to the 12 nucleotide sequence “GCGGGGUGCCGC” (“SV48”; SEQ ID NO:48). Therefore, a variant sequence having a substitution, deletion, or addition at any one of positions 1-12 will result in a variant having approximately 92% sequence identity to SEQ ID NO:48; a variant sequence having two mutations will result in a variant having approximately 83% sequence identity; a variant sequence having three mutations will result in a variant having approximately 75% sequence identity; a variant sequence having four mutations will result in a variant having approximately 66% sequence identity; and a variant sequence having five mutations will result in a variant having approximately 57% sequence identity to SEQ ID NO:48, respectively. Therefore, in some forms, the variant sgRNA includes a hairpin region of stem-loop 2 having at least about 56%, at least about 65%, at least about 74%, at least about 82%, or at least about 91% sequence identity to SEQ ID NO:48.
In other forms, the variant sgRNA includes a hairpin region of stem-loop 2 corresponding to a variant having at least 75%, up to 99% identity to
For example in some forms, the variant has a hairpin region of stem-loop 2 corresponding to a variant having at least 75%, 80%, 85%, 90%, 95% or 99% identity to SEQ ID NO:240.
Therefore, in some forms, the variant sgRNA has a hairpin region of stem-loop 2 nucleic acid sequence that has one or more nucleotides different to SEQ ID NO:240, such as one or more substitutions, deletions, or additions at any one of the nucleotide positions of SEQ ID NO:240. In some forms, the variant sgRNA includes a hairpin region of stem-loop 2 having one, two, three, four, five, or six residues that are substituted, deleted, or added relative to the 18 nucleotide sequence GGGCCGGGGUGCCGGCCC (“SV240”; SEQ ID NO:240). A variant sequence having a mutation (i.e., substitution, deletion, addition) of a single nucleotide at any one position (1-18) will result in a variant having approximately 94% sequence identity to SEQ ID NO:240; a variant sequence having two mutations will result in a variant having approximately 89% sequence identity; a variant sequence having three mutations will result in a variant having approximately 83% sequence identity; a variant sequence having four mutations will result in a variant having approximately 78% sequence identity; a variant sequence having five mutations will result in a variant having approximately 72% sequence identity; and a variant sequence having six mutations will result in a variant having approximately 66% sequence identity to SEQ ID NO:240, respectively. Therefore, in some forms, the variant sgRNA includes a hairpin region of stem-loop 2 having at least about 65%, at least about 71%, at least about 77%, at least about 82%, at least about 88%, or at least 94% sequence identity to SEQ ID NO:240.
In other forms, the framework region of the sgRNA scaffold is not that of the Wt sgRNA scaffold. For example, in some forms, the framework region of the sgRNA scaffold is derived from a variant sgRNA. Exemplary variant sgRNAs are known in the art, for example, including “E+F” (SEQ ID NO:346); “(CR772) E+F U61C/A66G” (SEQ ID NO:347); “U61C/A66G” (SEQ ID NO:348); “E+F G62A/A64G” (SEQ ID NO:349); “G62A/A64G” (SEQ ID NO:350); and “5E” (SEQ ID NO:351).
In some forms, the editing activity and specificity of the described variant sgRNAs including one or more mutations of the stem-loop 2 region is enhanced compared to that of the “Wild Type” (WT) sgRNA that does not include the mutations of the stem-loop 2 region. For example, in some forms, the described variant sgRNAs have increased on-target specificity compared to that of the “Wild Type” (WT) sgRNA that does not include the mutations of the stem-loop 2 region. Typically, when the described variant sgRNAs including one or more mutations of the stem-loop 2 region have increased specificity and editing activity of CRISPR/Cas as compared to that of the “Wild Type” (WT) sgRNA that does not include the mutations of the stem-loop 2 region, the described variant sgRNAs do not have increased off-target activity. In some forms, the described variant sgRNAs have decreased off-target activity compared to that of the “Wild Type” (WT) sgRNA that does not include the mutations of the stem-loop 2 region. In some forms, the described variant sgRNAs have increased on-target specificity and decreased off-target activity compared to that of the “Wild Type” (WT) sgRNA that does not include the mutations of the stem-loop 2 region.
In some forms, the described variant sgRNAs have increased on-target specificity of between about 1% and about 100%, inclusive, compared to that of the “Wild Type” (WT) sgRNA that does not include the mutations of the stem-loop 2 region. For example, in some forms, the described variant sgRNAs have increased on-target specificity of about 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99% or about 100%, or more, as compared to that of the “Wild Type” (WT) sgRNA that does not include the mutations of the stem-loop 2 region. In some forms, the described variant sgRNAs have decreased off-target activity that is between about 1% and about 99% inclusive of that of the “Wild Type” (WT) sgRNA that does not include the mutations of the stem-loop 2 region. For example, in some forms, the described variant sgRNAs have decreased off-target activity that is only about 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, up to about 99%, of that of the “Wild Type” (WT) sgRNA that does not include the mutations of the stem-loop 2 region.
In some forms, the described variant sgRNAs have increased on-target specificity of between about 1% and about 100% inclusive compared to that of the “Wild Type” (WT) sgRNA that does not include the mutations of the stem-loop 2 region, and decreased off-target activity that is between about 1% and about 99% inclusive of that of the “Wild Type” (WT) sgRNA that does not include the mutations of the stem-loop 2 region. For example, in some forms, the described variant sgRNAs have increased on-target specificity of about 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99% or about 100%, or more, as compared to that of the “Wild Type” (WT) sgRNA that does not include the mutations of the stem-loop 2 region, and have decreased off-target activity that is only about 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, up to about 99%, of that of the “Wild Type” (WT) sgRNA that does not include the mutations of the stem-loop 2 region.
Systems including Cas enzymes are provided. The CRISPR-associated Cas nuclease protein is a non-specific endonuclease. It is directed to the specific DNA locus by a gRNA, where it makes a double-strand break. There are several versions of Cas nucleases isolated from different bacteria. The most commonly used one is the Cas9 nuclease from Streptococcus pyogenes (SpCas9).
As used herein, the term “Cas” generally refers to an effector protein of a CRISPR Cas system or complex. The term “Cas” may be used interchangeably with the terms “CRISPR” protein, “CRISPR Cas protein,” “CRISPR effector,” CRISPR Cas effector,” “CRISPR enzyme,” “CRISPR Cas enzyme” and the like, unless otherwise apparent. The Crispr-Cas effector protein may be without limitation a type II, type V, or type VI Cas effector protein. Non-limiting examples of Crispr-Cas effector proteins include Cas1, Cas1B, Cas2, Cas3, Cas4, Cas5, Cas6, Cas7, Cas8, Cas9 (also known as Csn1 and Csx12), Cas1O, Csy1, Csy2, Csy3, Cse1, Cse2, Csc1, Csc2, Csa5, Csn2, Csm2, Csm3, Csm4, Csm5, Csm6, Cmr1, Cmr3, Cmr4, Cmr5, Cmr6, Csb1, Csb2, Csb3, Csx17, Csx14, Csx1O, Csx16, CsaX, Csx3, Csx1, Csx15, Csf1, Csf2, Csf3, Csf4, homologues thereof, or modified versions thereof. In some forms, the CRISPR enzyme has DNA cleavage activity.
In some forms, the Type II CRISPR enzyme is a Cas9 enzyme. The signature Cas9 effector proteins are large multi-domain RNA-dependent endonucleases that locate, bind, and cleave the double-stranded DNA (dsDNA) targets which are complementary to their guide RNAs. For recognition and binding to target DNA, Cas9 requires the protospacer adjacent motif (PAM), as a short conserved sequence located just downstream of the non-complementary strand of the target dsDNA. Recognition of the PAM (5′NGG3′) triggers dsDNA melting, enabling crRNA strand invasion and base pairing. The dsDNA cleavage mediation happens via the activity of separate HNH and RuvC nuclease domains. Also, Cas9 is a member of a small subset of Cas effectors that need a second trans-acting crRNA (tracrRNA) for gRNA processing and DNA cleavage.
Exemplary Cas9 enzymes are disclosed in International Patent Application Publication No. WO/2014/093595. In some forms, the Cas9 enzyme is S. pneumoniae, S. pyogenes or S. thermophilus Cas9, and may include mutated Cas9 derived from these organisms. The enzyme may be a Cas9 homolog or ortholog. Additional orthologs include, for example, Cas9 enzymes from Corynebacter diptheriae, Eubacterium ventriosum, Streptococcus pasteurianus, Lactobacillus farciminis, Sphaerochaeta globus, Azospirillum B510, Gluconacetobacter diazotrophicus, Neisseria cinereal, Roseburia intestinalis, Parvibaculum lavamentivorans, Staphylococcus aureus, Nitratifractor salsuginis DSM 16511, Camplyobacter lari CF89 12, and Streptococcus thermophilus LMD 9.
In some forms, the Cas9 effector protein and orthologs thereof may be modified for enhanced function. For example, improved target specificity of a CRISPR Cas9 system may be accomplished by approaches that include, but are not limited to, designing and preparing guide RNAs having optimal activity, selecting Cas9 enzymes of a specific length, truncating the Cas9 enzyme making it smaller in length than the corresponding wild-type Cas9 enzyme by truncating the nucleic acid molecules coding therefor and generating chimeric Cas9 enzymes wherein different parts of the enzyme are swapped or exchanged between different orthologs to arrive at chimeric enzymes having tailored specificity.
A Cas9 enzyme may include one or more mutations and may be used as a generic DNA binding protein with or without fusion to or being operably linked to a functional domain. The mutations may be artificially introduced mutations and may include but are not limited to one or more mutations in a catalytic domain. Examples of catalytic domains with reference to a Cas9 enzyme may include but are not limited to RuvC I, RuvC II, RuvC III and HNH domains. Preferred examples of suitable mutations are the catalytic residue(s) in the N term RuvC I domain of Cas9 or the catalytic residue(s) in the internal HNH domain.
Generally, the Cas9 is (or is derived from) the Streptococcus pyogenes Cas9 (SpCas9). In such forms, preferred mutations are at any or all of positions 10, 762, 840, 854, 863 and/or 986 of SpCas9 or corresponding positions in other Cas9 orthologs with reference to the position numbering of SpCas9 (which may be ascertained for instance by standard sequence comparison tools, e.g. ClustalW or MegAlign by Lasergene 10 suite). In particular, any or all of the following mutations are preferred in SpCas9: D10A, E762A, H840A, N854A, N863A and/or D986A; as well as conservative substitution for any of the replacement amino acids is also envisaged. The same mutations (or conservative substitutions of these mutations) at corresponding positions with reference to the position numbering of SpCas9 in other Cas9 orthologs are also preferred. Particularly preferred are D10 and H840 in SpCas9. However, in other Cas9s, residues corresponding to SpCas9 D10 and H840 are also preferred. These are advantageous as when singly mutated they provide nickase activity and when both mutations are present the Cas9 is converted into a catalytically null mutant which is useful for generic DNA binding.
In some forms, chimeric Cas9 proteins are used. Chimeric Cas9 proteins are proteins that include fragments that originate from different Cas9 orthologs. For instance, the N terminal of a first Cas9 ortholog may be fused with the C terminal of a second Cas9 ortholog to generate a resultant Cas9 chimeric protein. These chimeric Cas9 proteins may have a higher specificity or a higher efficiency than the original specificity or efficiency of either of the individual Cas9 enzymes from which the chimeric protein was generated. These chimeric proteins may also include one or more mutations or may be linked to one or more functional domains. Also suitable are Cas9 proteins that have different PAM specificities. Typically, Cas9 proteins, such as Cas9 from S. pyogenes (spCas9), require a canonical NGG PAM sequence to bind a particular nucleic acid region.
Cas9 nuclease sequences and structures are known to those of skill in the art (Ferretti, et al. Proc Natl Acad Sci U.S.A, 98, 4658-4863, doi: 10.1073/pnas.071559398 (2001); Deltcheva, et al. Nature, 471, 602-607, doi: 10.1038/nature09886 (2011)). Cas9 orthologs have been described in several species of bacteria, including but not limited to Streptococcus pyogenes and Streptococcus thermophilus, Campylobacter jejuni and Neisseria meningitidis. (Slaymaker, et al. Science, 351, 84-88 doi: 10.1126/science.aad5227 (2016); Kleinstiver, et al. Nature, 529, 490-495, doi: 10.1038/nature 16526 (2016); Chen, et al. Nature, 550, 407-410, doi: 10.1038/nature24268 (2017); Casini, et al. Nat Biotechnol, 6, 265-271, doi: 10.1038/nbt.4066 (2018); Lee, et al. Nat Commun, 9, 3048, doi: 10.1038/s41467-018-05477-x (2018); Vakulskas, et al. Nat Med, 24, 1216-1224, doi: 1.1038/s41591-018-0137-0 (2018); Choi, et al. Nat Methods, 16, 722-730, doi: 10.1038/s41592-019-0473-0 (2019); Kim, et al. Nat Commun, 8, 14500, doi: 10.1038/ncomms14500 (2017); (Edraki, et al. Mol Cell, 73, 714-726, doi: (2019)).
Enhanced ribonucleoprotein complexes including a Cas enzyme and one of the described variant sgRNAs are also provided. Typically, the enhanced ribonucleoprotein complexes have enhanced specificity and activity of on-target editing activity of CRISPR/Cas relative to the ribonucleoprotein complex formed by association of the same Cas enzyme with a Wt sgRNA. In some forms, an enhanced ribonucleoprotein complex includes:
In some forms, the ribonucleoprotein complex includes a variant sgRNA including a stem-loop 2 region set forth in Table 4. Therefore, in some forms, the ribonucleoprotein complex with enhanced specificity and activity of on-target editing activity of CRISPR/Cas relative to the Wt includes a variant sgRNA a stem-loop 2 region having a sequence of nucleic acids of any one of the sequences in Table 4. For example, in some forms, the ribonucleoprotein complex with enhanced specificity and activity of on-target editing activity of CRISPR/Cas relative to the Wt includes a variant sgRNA having a stem-loop 2 region formed from a sequence of nucleic acids of any one of SEQ ID NOs: 1-312.
In some forms, the variant strengthens the scaffold's interaction with SpCas9 via His721 and the PI domain of SpCas9. For example, in some forms, the ribonucleoprotein complex with enhanced specificity and activity of on-target editing activity of CRISPR/Cas relative to the Wt includes a variant sgRNA stem-loop 2 region having a nucleic acid sequence: GCGGGGUGCCGC (“SV48”; SEQ ID NO:48). In other forms, the ribonucleoprotein complex with enhanced specificity and activity of on-target editing activity of CRISPR/Cas relative to the Wt includes a variant sgRNA stem-loop 2 region having a nucleic acid sequence: GGGCCGGGGUGCCGGCCC (“SV240”; SEQ ID NO:240).
In some forms, the ribonucleoprotein complex with enhanced specificity and activity of on-target editing activity of CRISPR/Cas relative to the Wt includes a Cas9 enzyme and a variant sgRNA having a nucleic acid sequence of:
In other forms, the ribonucleoprotein complex with enhanced specificity and activity of on-target editing activity of CRISPR/Cas relative to the Wt includes a Cas9 enzyme and a variant sgRNA having a nucleic acid sequence of:
Methods for using the described compositions for enhanced gene editing are described. The described variant sgRNAs and ribonucleoprotein complexes thereof can be used for any suitable purpose and in any suitable method for CRISPR-based editing of DNA.
Generally, the disclosed variants can be used to cleave target DNA of interest. Such cleavage is preferably used in a method of editing the target DNA of interest. For example, the disclosed variants can be used for and in any known methods of DNA editing, including in vitro and in vivo DNA editing. sgRNAs, of which the disclosed variants are new forms, can be and have been used for various DNA cleavage and editing methods and the disclosed variants can be used as the RNA-guided endonuclease in any of these methods uses. For example, the disclosed variants can be used for altering the genome of a cell. Various methods for selectively altering the genome of a cell using RNA-guided endonucleases are described in the following exemplary U.S. Patent documents: U.S. Pat. Nos. 8,993,233, 9,023,649, and 8,697,359 and U.S. Patent Application Publication Nos. 20140186958, 20160024529, 20160024524, 20160024523, 20160024510, 20160017366, 20160017301, 20150376652, 20150356239, 20150315576, 20150291965, 20150252358, 20150247150, 20150232883, 20150232882, 20150203872, 20150191744, 20150184139, 20150176064, 20150167000, 20150166969, 20150159175, 20150159174, 20150093473, 20150079681, 20150067922, 20150056629, 20150044772, 20150024500, 20150024499, 20150020223, 20140356867, 20140295557, 20140273235, 20140273226, 20140273037, 20140189896, 20140113376, 20140093941, 20130330778, 20130288251, 20120088676, 20110300538, 20110236530, 20110217739, 20110002889, 20100076057, 20110189776, 20110223638, 20130130248, 20150050699, 20150071899, 20150050699, 20150045546, 20150031134, 20150024500, 20140377868, 20140357530, 20140349400, 20140335620, 20140335063, 20140315985, 20140310830, 20140310828, 20140309487, 20140304853, 20140298547, 20140295556, 20140294773, 20140287938, 20140273234, 20140273232, 20140273231, 20140273230, 20140271987, 20140256046, 20140248702, 20140242702, 20140242700, 20140242699, 20140242664, 20140234972, 20140227787, 20140212869, 20140201857, 20140199767, 20140189896, 20140186958, 20140186919, 20140186843, 20140179770, 20140179006, 20140170753, and 20150071899, each of which is incorporated by reference herein, and in particular for their description of the uses of RNA-guided endonucleases.
Various methods for selectively altering the genome of a cell using RNA-guided endonucleases are described in the following exemplary publications: WO 2014/099744; WO 2014/089290; WO 2014/144592; WO 2014/004288; WO 2014/204578; WO 2014/152432; WO 2015/099850; WO 2008/108989; WO 2010/054108; WO 2012/164565; WO 2013/098244; WO 2013/176772; Makarova et al., “Evolution and classification of the CRISPR-Cas systems” 9(6) Nature Reviews Microbiology 467-477 (1-23) (June 2011); Wiedenheft et al., “RNA-guided genetic silencing systems in bacteria and archaea” 482 Nature 331-338 (Feb. 16, 2012); Gasiunas et al., “Cas9-crRNA ribonucleoprotein complex mediates specific DNA cleavage for adaptive immunity in bacteria” 109(39) Proceedings of the National Academy of Sciences USA E2579-E2586 (Sep. 4, 2012); Jinek et al., “A Programmable Dual-RNA-Guided DNA Endonuclease in Adaptive Bacterial Immunity” 337 Science 816-821 (Aug. 17, 2012); Carroll, “A CRISPR Approach to Gene Targeting” 20(9) Molecular Therapy 1658-1660 (September 2012); U.S. Appl. No. 61/652,086, filed May 25, 2012; Al-Attar et al., Clustered Regularly Interspaced Short Palindromic Repeats (CRISPRs): The Hallmark of an Ingenious Antiviral Defense Mechanism in Prokaryotes, Biol Chem. (2011) vol. 392, Issue 4, pp. 277-289; Hale et al., Essential Features and Rational Design of CRISPR RNAs That Function With the Cas RAMP Module Complex to Cleave RNAs, Molecular Cell, (2012) vol. 45, Issue 3, 292-302.
Disclosed are methods of editing a sequence of interest. In some forms, the method includes contacting a disclosed construct with the host of interest, where the host of interest harbors the sequence of interest and where the cell expresses a construct to produce the variant sgRNA and a Cas9 enzyme. In some forms, the method includes contacting a disclosed construct with the host of interest, where the host of interest harbors a sequence of interest and where the cell expresses the construct to produce the variant. In some forms, the method includes contacting the sequence of interest with a disclosed mixture, whereby the variant edits the sequence of interest targeted by the sgRNA.
In some forms, the method can further includes causing a variant sgRNA targeting the sequence of interest to be present in the host of interest with the produced variant, whereby the produced variant edits the sequence of interest targeted by the sgRNA.
The description can be further understood by reference to the following numbered paragraphs:
1. A variant single guide RNA (sgRNA) including substitution and/or addition of one or more nucleic acid residues that strengthens the interaction of the sgRNA with a Cas enzyme,
2. The variant sgRNA of paragraph 1, wherein the substitution and/or addition of one or more nucleic acid residues that strengthens the interaction of the sgRNA with a Cas enzyme includes substitution and/or addition of one or more nucleic acid residues within the hairpin region of the stem-loop 2 of the sgRNA.
3. The variant sgRNA of paragraph 1 or 2, wherein the Cas enzyme is a Cas9 enzyme.
4. The variant sgRNA of paragraph 3, wherein the Cas9 enzyme is derived from Streptococcus pyogenes (spCas9).
5. The variant sgRNA of paragraph 4, wherein the substitution and/or addition of one or more nucleic acid residues strengthens the sgRNAs interaction with residue His721 and/or the PI domain of SpCas9.
6. The variant sgRNA of any one of paragraphs 2-5, including the nucleic acid sequence:
7. The variant sgRNA of any one of paragraphs 2-6, wherein the hairpin region of stem-loop 2 includes the nucleic acid sequence of any one of SEQ ID NOS: 1-312.
8. The variant sgRNA of any one of paragraphs 2-7, wherein the hairpin region of stem-loop 2 includes the nucleic acid sequence GCGGGGUGCCGC (SEQ ID NO:48), or a nucleic acid sequence having at least about 74% identity to SEQ ID NO:48.
9. The variant sgRNA of paragraph 8, wherein the hairpin region of stem-loop 2 includes a nucleic acid sequence having at least 82%, or at least 91% sequence identity to GCGGGGUGCCGC (SEQ ID NO:48).
10. The variant sgRNA of any one of paragraphs 2-7, wherein the hairpin region of stem-loop 2 includes the nucleic acid sequence GGGCCGGGGUGCCGGCCC (SEQ ID NO:240), or a nucleic acid sequence having at least about 75% identity to SEQ ID NO:240.
11. The variant sgRNA of paragraph 10, wherein the hairpin region of stem-loop 2 includes a nucleic acid sequence having at least 77%, at least 82%, at least 88%, or at least 94% sequence identity to GGGCCGGGGUGCCGGCCC (SEQ ID NO:240).
12. A variant sgRNA including a nucleic acid sequence of GUUUUAGAGCUAGAAAUAGCAAGUUAAAAUAAGGCUAGUCCGUUAUCAGCGG GGUGCCGCGGCACCGAGUCGGUGCU (SEQ ID NO:352), or a nucleic acid sequence having at least 75% identity to SEQ ID NO:352.
13. The variant sgRNA of paragraph 12, wherein the sgRNA includes a nucleic acid sequence having at least 80%, at least 85%, at least 90%, or at least 95% sequence identity to SEQ ID NO:352.
14. A variant sgRNA including a nucleic acid sequence of GUUUUAGAGCUAGAAAUAGCAAGUUAAAAUAAGGCUAGUCCGUUAUCAGGGC CGGGGUGCCGGCCCGGCACCGAGUCGGUGCU (SEQ ID NO:353), or a nucleic acid sequence having at least 75% identity to SEQ ID NO:353.
15. The variant sgRNA of paragraph 14, wherein the sgRNA includes a nucleic acid sequence having at least 80%, at least 85%, at least 90%, or at least 95% sequence identity to SEQ ID NO:353.
16. A ribonucleoprotein complex including:
17. The ribonucleoprotein complex of paragraph 16, wherein the Cas9 enzyme is derived from Streptococcus pyogenes (spCas9).
18. The ribonucleoprotein complex of paragraph 16 or 17, wherein the variant sgRNA includes the nucleic acid sequence:
19. The ribonucleoprotein complex of paragraph 18 including the sgRNA of any one of paragraphs 12 to 15.
20. A vector encoding of expressing the sgRNA of any one of paragraphs 1 to 15.
21. A cell including the sgRNA of any one of paragraphs 1 to 15, or the ribonucleoprotein complex of any one of paragraphs 16-19.
22. A method for CRISPR editing of one or more target genes in a cell, the method including administering into and/or expressing within the cell the ribonucleoprotein complex of any one of paragraphs 16-19,
23. The method of paragraph 22, wherein the administering is in vivo.
24. A kit including
The on- and off-target editing activities for SpCas9 nuclease using two published engineered sgRNA scaffold variants, E+F scaffold and cr772 (Chen, et al., Cell 2013, 155, (7), 1479-91; and Jost, et al., Nat Biotechnol 2020, 38, (3), 355-364) were evaluated.
PDB 600Y was used as the template for molecular modelling to simulate the likely consequences of stem-loop 2 lengthening. Variant sequences were submitted with 2-6 bp lengthening at the upper stem-loop 2 region to the ModeRNA server (available on the world wide web at “//iimcb.genesilico.pl/modernaserver/”) to generate threading models of the sgRNA scaffold and examined the sgRNA-SpCas9 protein interactions using UCSF Chimera v 1.14. ModeRNA was also used to generate sgRNA models containing the beneficial mutations previously reported (Jost, et al., Nat Biotechnol 2020, 38, (3), 355-364)) on nucleotides base-pair stack 58-69, 60-67, 61-66, to evaluate whether these mutations brought about fundamental structural changes in the protein models; no detrimental alterations generated by those mutations in the sgRNA scaffolds were identified. To generate a library of sgRNA scaffold variants focusing on the stem-loop 2 regions, RNA designer was used (available on the world wide web at “masoft.ca/cgi-bin/RNAsoft/RNAdesigner/rnadesign.pl”) with parameters: temperature: 37° ° C., target GC %: 50%, and allowing 10 designs. Only stem-loop 2 (position 54-70) was input to reduce computing time; the top design(s) with the minimum free energy was selected. Designs that fit with the U61G-A66C beneficial mutations were also filtered. In other words, the base pairing closest to the “AGAG” tetraloop was fixed to be G-C or C-G. Two versions of the stem-loop 2 lengthening scheme, the proximal (inserted at 61-66 base pair) and the distal (inserted at 58-69 base-pair) to the tetraloop were tested. Stem-length combinations for stem-loop2 (2-6 bp) extend between 61-66, and “GAAA” tetraloop, showing only the base pair 5′-3′ after 61 bp:
Stem-length combinations for stem-loop2 (2-6 bps), extend between 58-69 and 59-68 base-pair, showing only the base pair sequence 5′-3′ after 58 bp:
The DNA vectors used (Table 1) were generated by standard molecular cloning strategies, including PCR, restriction enzyme digestion, oligo annealing and 5′ end phosphorylation, and ligation. Custom oligonucleotides were purchased from Genewiz. Vectors were transformed into E. coli strain DH5α competent cells and selected with ampicillin (100 mg/ml, USB) or carbenicillin (50 mg/ml, Teknova). Plasmid DNA was extracted and purified by Plasmid Mini (Takara) or Midi preparation (QIAGEN) kits. Sequences of the vectors were verified by Sanger sequencing.
sgRNA scaffold E+F in vector pJHp3 was generated by overlapping PCR of primers SY1, SY2, J15, and J16. The same strategy was used to obtain 5E in pJHp13, with the primers SY3, SY4, J17, and J18. The sequences of G62A/A64G in pJHp5 and E+F G62A/A64G in pJHp11 were PCR amplified by primers SA82 and J13 from pAWp28 and pJHp3, respectively. Similarly, the sequences of U61C/A66G in pJHp6 and cr772 in pJHp12 were PCR amplified by primers SA82 and J14 from pA Wp28 and pJHp3, respectively. All the PCR products were digested by XhoI and BamHI and inserted between the same sites in pAWp28 (Addgene, 73850) to generate the vectors. To construct the reporter vector pPZp257 for gene knockout and base editing, the mutant hU6 promoter was firstly PCR-amplified by primer pair Z350 and Z352 from pAWp28 and inserted between the SbfI and BamHI sites in pAWp9 (Addgene, 73851). Then an artificial reporter sequence (5′-3′):
was synthesized and placed downstream of the hU6 promoter, leaving two Esp3I sites in between for sgRNA and scaffold insertion. A nicking sgRNA expression cassette, mutant mU6-dummysg2-v2 sgRNA scaffold, from pPZp138-3-4D (Guschin, et al., Methods Mol Biol 2010, 649, 247-56) was inserted downstream of the reporter. To generate the sgRNA scaffold variants library vector pPZp284a, a sgRNA targeting the reporter region and a truncated scaffold with two Esp3I sites for library insertion were annealed by two pairs of oligos Z518/Z519 and Z520/Z521 and ligated into the Esp3I sites in pPZp257. An array of oligo pairs containing 312 unique sgRNA scaffold sequences was synthesized. The oligo pairs were annealed and then cloned into the vector in pooled fashion. Sixty-fold representation of the library size was achieved in the cloning to ensure the coverage. pJF60b was generated by inserting a PCR amplified fragment from pCMV_AncBE4max_P2A_GFP (Addgene, 112100) into a lentiviral vector. Sequences of the primers and sgRNA spacer sequences used are listed in Table 2 and Table 3.
HEK293T cells were obtained from American Type Culture Collection (ATCC), and OVCAR8-ADR cells were a gift from T. Ochiya (Japanese National Cancer Center Rescarch Institute, Japan). A cell line authentication test (Genetica DNA Laboratories) was performed to confirm the identity of the OVCAR8-ADR cells. OVCAR8-ADR cells that stably express SpCas9 and AncBE4max were generated by transducing pAWp30 (Addgene, 73857) and pJF60b, followed by zeocin selection (Life Technologies) and cell sorting, respectively. Opti-SpCas9 from pAWp63-clone32 (Addgene, 131736), a high-fidelity SpCas9 that has comparable activity to wild-type (Choi, et al., Nat Methods 2019, 16, (8), 722-730), was used in the experiments shown in
For each lentivirus preparation, HEK293T cells were transfected by FuGene HD transfection reagent (Promega) according to the manufacturer's instructions in a 6-well plate, with 0.5 μg of pCMV-VSV-G, 1 μg of pCMV-dR8.2-dvpr, and 0.5 μg of the respective lentiviral vector per well. The virus-containing supernatants collected from 48 and 72 hr post-transfection were combined and filtered by 0.45 mm polyethersulfone membrane (Pall). For routine transduction, 300 μL of the filtered supernatant was applied to one well of a 12-well plate in the presence of 8 mg/ml polybrene (Sigma), with cell confluence at about 30%. For library transduction, cells were transduced by the lentiviruses at a multiplicity of infection (MOI) of <0.3 to ensure most cells were infected with just one virion. Enough cells were transduced to achieve 500-fold representation of the library size.
Cells for flow cytometry analysis were trypsinized and resuspended in FACS buffer (PBS with 2% FBS). BD LSR Fortessa analyser (Becton Dickinson) was used to detect the signal of TurboRFP by 561 nm yellow-green laser (610/20 nm). Data were analysed by FlowJo software (v10.5.3, Becton Dickinson). For cell sorting, samples were prepared similarly as for FACS analysis with the sorting buffer (PBS with 2% FBS and 2× antibiotic-antimycotic). BD Influx cell sorter (Becton Dickinson) equipped with a 100-mm nozzle (24 psi with a frequency of 39.2 kHz) was used. To isolate lentivirus-infected cells, fluorescent protein-positive cells were sorted using 1.0 Drop Pure mode. For cells being infected with the screening libraries, the 1%-2% cells that had the strongest fluorescent protein-positive signals were not collected to minimize the chance of acquiring cells that were infected with more than a single virion. At least 100-fold more cells than the library size were collected.
The on-target activity of scaffold variants was measured using a reporter system as described, in which a sgRNA spacer sequence (i.e., RFPsg5-ON) completely matched with the RFP target site. In contrast, off-target activity was measured using a reporter system in which the RFP target site contained a synonymous mutation (i.e., RFPsg5-OFF5-2). SpCas9-expressing OVCAR8-ADR cells containing the reporter system were transduced with the RFP-targeting sgRNAs containing the different scaffold variants. The fluorescent intensity was measured by flow cytometry.
SpCas9-expressing OVCAR8-ADR cells were transduced with sgRNAs containing different scaffold variants, targeting endogenous loci. Genomic DNA from cells after genome editing was prepared using QuickExtract DNA extraction solution (Epicentre) or the DNeasy Blood and Tissue kit (Qiagen). The targeted loci with flanking regions were amplified by PCR and purified using PCRCleanDX (Aline Biosciences). About 300 ng of the amplicons were denatured, self-annealed, and incubated with 4 U of T7 endonuclease I (New England Biolabs) at 37° C. for 30 min. The reaction products were resolved by 2% agarose gel electrophoresis. Quantification was based on relative band intensities measured using ImageJ. Indel percentage was estimated by the formula
as previously described (Guschin, et al., Methods Mol Biol 2010, 649, 247-56), where a is the integrated intensity of the uncleaved PCR product, and b and c are the integrated intensities of each cleavage product.
GUIDE-seq was performed and analysed as described. Briefly, 1 million SpCas9-expressing OVCAR8-ADR cells were transduced with sgRNA lentiviral vectors containing different scaffold variants at an MOI of ˜3 in a 6-well plate and then electroporated with 1,000 pmol dsODN at the parameters of 1,300V, 10 ms, and 3 pulses using 100-μL NEON tips (Thermo Fisher Scientific). Genomic DNAs were harvested by the DNeasy Blood and Tissue kit (Qiagen) 72 h post-electroporation and subjected to library preparation and sequencing.
Deep sequencing was carried out as previously described (Wong, et al., Proc Natl Acad Sci USA 2016, 113, (9), 2544-9). For validations in gene knockout and cytosine base editing settings, OVCAR8-ADR cells stably expressing SpCas9 or AncBE4max were transduced with lentiviruses of sgRNAs containing different scaffold sequences and collected on day 7 post-transduction with biological triplicates. The targeted loci were amplified from the genomic DNAs and indexed with unique barcodes by PCR. More than 0.8 million reads were obtained through NovaSeq 6000 (Illumina), evaluating editing outcomes from more than 10,000 cells for each sample. For sgRNA scaffold library screening, HEK293T cells containing the scaffold library were sorted out for pCMV_AncBE4max_P2A_GFP (Addgene, 112100) transfection and collected on day 3 post-transfection for deep sequencing. The sgRNA scaffold library-transduced OVCAR8-ADR-SpCas9 cells were collected on day 7 post-transduction. The region containing both the sgRNA scaffold variant and its targeted loci were amplified, indexed and sent for deep sequencing. CRISPresso2 (Clement, et al., Nat Biotechnol 2019, 37, (3), 224-226) was used to analyze all the deep sequencing data in NHEJ and CBE mode with default parameters. To evaluate the editing efficiency of each scaffold in the pooled library, Crispresso2 was run and surveyed the edited alleles around sgRNA from the Crispresso2 results. Alleles possessing at least 0.05% of reads and that were within the top 20 most frequently observed alleles in a sample We focused on to rule out potential defects from PCR and/or sequencing errors. Read 2s that matched with the selected alleles were then extracted and examined the sgRNA scaffold stem-loop 2 sequences at read 1s. The editing frequency of each of the sgRNA scaffold variants were counted using read Is that matched perfectly with the design sequences in the library. In the validation experiments of individual scaffolds, CRISPresso2 was run to survey their editing efficiency based on the percentage of modified reads.
PDB 600Y was used as the template for molecular modelling. To generate the model for sgRNA 5E, SV48, and SV240, the stem-loop 2 regions of the variants was first reconstructed using RNA composer (available on the world wide web at “//rnacomposer.cs.put.poznan.pl/”) with a pre-defined secondary structure of the intended design. Then we grafted the reconstructed stem-loop 2 to the sgRNA scaffold in the template (600Y chain B) using Rosetta (v 2019.35) RNA tools. The sgRNA variants in the reconstructed model were examined using UCSF Chimera v 1.14.
Using red fluorescent protein (RFP) disruption assay, it was confirmed that using either E+F scaffold or cr772 increased SpCas9-mediated editing to 91.7% and 93.6%, respectively, compared to 65.1% when wild-type scaffold was used (
To improve SpCas9's editing activity while maintaining specificity, various regions of the sgRNA scaffold were modified. Previous studies have shown that the upper stem-loop 2 of the scaffold is positioned close to the SpCas9 (Nishimasu, et al., Cell 2014, 156, (5), 935-49) and is highly tolerant to mutations. Whether extending the upper stem-loop 2 of the scaffold could increase editing activity was investigated. A 5-nucleotide-extension was previously added to the upper stem-loop 2 in the E+F scaffold and this scaffold was shown to increase SpCas9's on-target activity (Grevet, et al., Science 2018, 361, (6399), 285-290). The scaffold variant 5E that carries only the 5-nucleotide-extension at the upper stem-loop 2 but not the other modifications present in the E+F scaffold (
In parallel to testing scaffold variant 5E, the functional impact of introducing other modifications to the upper stem-loop 2 region of the sgRNA scaffold on modulating the activity of SpCas9 editors was also explored. Pooled screens were performed with a library of 312 scaffolds containing:
It was realized that some of the above modifications would strengthen the scaffold's interaction with SpCas9. The stem-loop 2 extension was designed using the RNAdesigner webserver (webpage rnasoft.ca/cgi-) and the recommended stable sequences were selected based on minimum free energy calculated by Vienna fold at temperature of 37 degrees Celsius and 50% GC content at stem regions. The library of scaffold variant-bearing sgRNAs tandemly linked to a sgRNA-targeted reporter sequence was delivered into human cells and expressed SpCas9 or its derived base editor AncBE4max to initiate editing (
Using SV48 and SV240 generated more base edits at the five endogenous loci tested than using a wild-type scaffold (
Guide RNA engineering strategies should improve CRISPR's on-target activity while minimizing off-target edits. Intriguingly, it was found that the previously reported sgRNA scaffold variants increase off-target editing more than on-target activity. sgRNA scaffold variants that augment on-target CRISPR editing while achieving high on-to-off targeting specificity have been engineered. Although the exact mechanism on how extending the upper stem-loop 2 alone in these new scaffolds may give such an advantage remains to be understood, molecular modelling hints that it is related to the increase in the scaffold's interaction with His721 and the PI domain of SpCas9. These interactions are distant from where the extended tetraloop in the previously engineered E+F scaffold interacts with SpCas9 (Nishimasu, et al. Cell 2014, 156, (5), 935-49), suggesting that the described scaffolds modulate SpCas9's editing activity via a different mechanism. Strengthened sgRNA:SpCas9 binding via His721 and PI domain interactions with the scaffold may further favor sgRNA loading over competitor intracellular RNA binding (Mekler, et al., Nucleic Acids Res 2016, 44, (6), 2837-45), thus stabilizing Cas9-sgRNA complex formation and enhancing editing activity. At the same time, it remains to be revealed whether it may also render the neighboring RuvC domain less energetically favorable to form a reorganized loop to stabilize target DNA substrate with mismatches (Bravo, et al., Nature 2022, 603, (7900), 343-347) or act through other mechanisms to minimize off-target editing. The data presented above also revealed that the same stem-loop 2-engineered scaffolds could be useful for enhancing the activities of base editors derived from SpCas9. Some scaffolds may adopt different sgRNA design rules. Indeed, the engineering of sgRNA scaffolds is still in its infancy, particularly for those effectors, including prime editor and Cas12f (Nelson, et al., Nat Biotechnol 2022, 40, (3), 402-410; Kim, et al., Nat Biotechnol 2022, 40, (1), 94-102; and Xu, et al., Mol Cell 2021, 81, (20), 4333-4345 e4), that were shown to require more extensive modifications.
In summary, the data have uncovered an engineering route to create new stem-loop 2-modified sgRNA scaffolds for increasing the editing activity of both SpCas9 nuclease and base editor. This work demonstrates the feasibility of engineering sgRNA scaffold variants for SpCas9 to achieve both high efficiency and specificity, highlighting applications for applying high-throughput sgRNA scaffold engineering approaches to enhance the CRISPR-Cas systems for genome editing applications.
Those skilled in the art will recognize or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the method and compositions described herein. Such equivalents are intended to be encompassed by the following claims.
This application claims benefit of and priority to U.S. Patent Application No. 63/484,902, filed on Feb. 14, 2023, the contents of which is hereby incorporated by reference herein in its entirety.
Number | Date | Country | |
---|---|---|---|
63484902 | Feb 2023 | US |