BIPARTITE BASE EDITOR (BBE) ARCHITECTURES AND TYPE-II-C-CAS9 ZINC FINGER EDITING

Provided herein are fusion proteins useful in a bipartite cytosine base editor system comprising: (i) a first fusion protein comprising a non-R-loop-forming programmable DNA binding domain, preferably a transcription-activator-like effector (TALE) or zinc finger array (ZF), fused to a deaminase enzyme, or an active portion thereof, optionally with a linker therebetween, or (ii) a second fusion protein comprising an R-loop-forming Cas9 protein that lacks nuclease activity or is a nickase, but can interact with a guide RNA and target DNA, fused to a Uracil glycosylase inhibitor (UGI), optionally with a linker therebetween. Additionally, provided are methods for targeted deamination of one or more selected cytosines in a nucleic acid.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CLAIM OF PRIORITY

This application claims the benefit of U.S. Provisional Patent Application Ser. No. 62/511,296, filed on May 25, 2017; Ser. No. 62/541,544, filed on Aug. 4, 2017; and Ser. No. 62/622,676, filed on Jan. 26, 2018. The entire contents of the foregoing are hereby incorporated by reference.

FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT

This invention was made with Government support under Grant No. GM118158 awarded by the National Institutes of Health. The Government has certain rights in the invention.

TECHNICAL FIELD

Described herein are methods and compositions for improving the genome-wide specificities of targeted base editing technologies.

BACKGROUND

Base editing (BE) technology is based on the fusion of a DNA binding domain as e.g. catalytically inactive or nicking Cas9 (dCas9 or nCas9) to a cytidine or adenine deaminase and a uracil glycosylase inhibitor (UGI). These fusion proteins allow for R-loop formation and deamination of cytosine or adenine at a specific genomic locus introducing targeted substitutions (C/G→T/A or A/T→G/C)1-4. Precise and efficient correction of single nucleotide variants might yield substantial therapeutic benefits, specifically as the current approach of double-strand break (DSB) based donor template integration via homology directed repair (HDR) is inefficient and not feasible in post-mitotic cells5. Since the deaminases used for cytosine-to-thymine conversion require an ssDNA substrate, nCas9-meditated R-loop formation, which leaves the nCas9 non-target strand single-stranded, is a critical factor in BE targeting success1.

SUMMARY

CRISPR base editor platforms (BE) possess the unique capability to generate precise, user-defined genome-editing events without the need for a donor DNA molecule. Base editors (BEs) that include a single strand nicking CRISPR-Cas9 (nCas9) protein fused to a cytidine deaminase domain and uracil glycosylase inhibitor (UGI) (BE3) efficiently induce cytosine-to-thymine (C-to-T) base transitions in a site-specific manner as determined by the CRISPR guide RNA (gRNA) spacer sequence1. As with all genome editing reagents, it is critical to first determine and then mitigate BE's capacity for generating off-target mutations before it is used for therapeutics so as to limit its potential for creating deleterious and irreversible genetically-encoded side-effects. Herein, we describe bipartite BEs (BBEs) that rely on the use of adjacently-targeting DNA binding domains to properly function. Unlike previously-described BEs that contain machinery to both deaminate and form R-loops in a monomeric unit, we describe a system where these two critical functions are separated onto two proteins that must work together to stimulate robust deamination. To do so, we fused engineered zinc-fingers (ZFs) to deaminase enzymes (ZF-deaminase) and targeted it adjacent to an nCas9-UGI binding site, thereby reconstituting the full functionality of a BE around adjacently-targeting DNA sequences and stimulating robust DNA modification. Since efficient activity of this bipartite enzyme relies on the specific adjacent binding of two separate DNA-binding domains, it should increase the specificity of base editing by decreasing the ability of its constituent pieces to function robustly at any of their off-target binding sites in genomic DNA, so long as adjacent binding of the two constituents does not occur at that site. Spatially separating nCas9 from the deaminase domain should also reduce the synergistically induced indel formation adjacent to and at the site of targeted base editing, thereby enhancing overall product purity and off-target indel formation.

While some Cas9 orthologs and variants may be deficient for CRISPR-mediated DNA-binding and genome- or base editing (such as PAM-interacting deficient (PID) mutants or type-II-C Cas9s14-16), we noted that rescuing their DNA-targeting capabilities by fusing them to an adjacently-targeted engineered DNA binding domain such as a ZF or a TALE may allow them to constitute the basis of BEs with better specificity profiles than those described as yet in the literature. By requiring the adjacent binding of the two DNA binding domains (the ZF or TALE and the deficient Cas9 variant or ortholog, the resulting protein specifies a total DNA target sequence comprising both target sites, increasing the number of total bases specified by the complex and thereby imparting greater overall specificity to the technique. Use of these proteins as the basis of base editors would therefore constitute a more specific base editing platform. Thus, provided herein are fusion proteins useful in a bipartite cytosine base editor system comprising: (i) a first fusion protein comprising a non-R-loop-forming programmable DNA binding domain, preferably a transcription-activator-like effector (TALE) or zinc finger array (ZF), fused to a deaminase enzyme, or an active portion thereof, optionally with a linker therebetween, or (ii) a second fusion protein comprising an R-loop-forming Cas9 protein that lacks nuclease activity or is a nickase, but can interact with a guide RNA and target DNA, fused to a Uracil glycosylase inhibitor (UGI), optionally with a linker therebetween.

In some embodiments, the first fusion protein comprises a cytidine deaminase selected from the group consisting of apolipoprotein B mRNA-editing enzyme, catalytic polypeptide-like 1 (APOBEC1), APOBEC2, APOBEC3A, APOBEC3B, APOBEC3C, APOBEC3D/E, APOBEC3F, APOBEC3G, APOBEC3H, APOBEC4; activation induced cytidine deaminase (AICDA), cytosine deaminase 1 (CDA1), and CDA2, and cytosine deaminase acting on tRNA (CDAT).

In some embodiments, the second fusion protein comprises an SpCas9 nickase or inactive nuclease with a mutation at D10, E762, D839, H983, or D986 and H840 or N863, or an inactive Cpf1 (dCpf1) protein. In some embodiments, the mutation is one or both of D10A or D10N, or H840A/H840N/H840Y.

In some embodiments, the DNA binding domain comprises a zinc finger DNA binding array.

Also described herein are monomeric fusion proteins comprising (i) a first portion comprising a deaminase enzyme, or an active portion thereof, (ii) a second portion comprising a Cas9 protein that lacks nuclease activity or is a nickase and can interact with a guide RNA but does not sufficiently bind target DNA or form R-loops to potentiate genome editing or base events, (iii) a third portion comprising a DNA binding domain selected from zinc fingers and TALEs, and optionally (iv) a fourth portion comprising a Uracil DNA glycosylase inhibitor (UGI), optionally with peptide linkers in between some or all of the first, second, third, or fourth portions, and wherein the domains (i)-(iv) can be in any order.

Further provided herein are fusion proteins comprising (i) a first portion comprising a deaminase enzyme, or an active portion thereof, (ii) a second portion comprising a Cas9 protein that lacks nuclease activity or is a nickase and can interact with a guide RNA but does not sufficiently bind target DNA or form R-loops to potentiate genome editing or base events, such as type-II-C Cas9 orthologs or similarly deficient species, and (iii) a third portion comprising a DNA binding domain selected from zinc fingers and TALEs, optionally with peptide linkers in between some or all of the first, second, or third portions, and wherein the domains (i)-(iii) can be in any order. These fusion proteins can be used in combination with a protein comprising a Uracil DNA glycosylase inhibitor (UGI), e.g., in the methods described herein.

In some embodiments, the first portion comprises a cytidine deaminase selected from the group consisting of apolipoprotein B mRNA-editing enzyme, catalytic polypeptide-like 1 (APOBEC1), APOBEC2, APOBEC3A, APOBEC3B, APOBEC3C, APOBEC3D/E, APOBEC3F, APOBEC3G, APOBEC3H, APOBEC4; activation induced cytidine deaminase (AICDA), cytosine deaminase 1 (CDA1), and CDA2, and cytosine deaminase acting on tRNA (CDAT).

In some embodiments, the second portion comprises an SpCas9 nuclease with a mutation at D10, E762, D839, H983, or D986 and H840 or N863, or a type-II-C Cas9 ortholog. In some embodiments, the mutation is one or both of D10A or D10N, or H840A/H840N/H840Y.

In some embodiments, the second portion comprises PlCas9, ClCas9, or NcCas9; or SpCas9, SaCas9, or CjCas9 with a mutations to a PAM interacting domain (PID), e.g., as shown in Table 2.

In some embodiments, the DNA binding domain comprises a zinc finger DNA binding array.

Also provided herein are compositions comprising a purified fusion protein described herein, optionally further comprising a guide RNA that interacts with the Cas9 portion of the fusion protein. In some embodiments, the composition includes one or more ribonucleoprotein (RNP) complexes.

Further provided herein are nucleic acids encoding the fusion proteins described herein, vectors comprising the nucleic acids, and isolated host cells comprising and/or expressing the fusion proteins.

Also provided are compositions comprising a nucleic acid encoding the fusion proteins described herein.

Additionally, provided are methods for targeted deamination of one or more selected cytosines in a nucleic acid. The methods include contacting the nucleic acid with fusion proteins described herein, and a guide RNA (gRNA) that interacts with the Cas9 portion of the fusion protein and binds to a portion of the nucleic acid comprising or adjacent to (e.g., within 1, 2, 5, 10, 15, or 20 nts of) the selected cytosines. In some embodiments, the nucleic acid is in a cell, preferably a eukaryotic cell.

Also provided herein are methods for targeted deamination of a nucleic acid in a cell. The methods include expressing in the cell, or contacting the cell with, the fusion proteins described herein, and a guide RNA (gRNA) that interacts with the Cas9 portion of the fusion protein and binds to a portion of the nucleic acid comprising or adjacent to (e.g., within 1, 2, 5, 10, 15, or 20 nts of) the selected cytosine.

In some embodiments, the fusion protein is delivered as an RNP, or as an mRNA or plasmid comprising a sequence encoding the fusion protein.

Also provided herein are ribonucleoprotein (RNP) complexes that include a variant SpCas9 protein as described herein and a guide RNA that targets a sequence having a PAM sequence targeted by the bipartite deaminase fusion protein comprising Cas9.

Also provided herein are methods for targeted deamination, or improving specificity of targeted deamination, of a selected cytosine in a nucleic acid, comprising contacting the nucleic acid with one or more of the fusion proteins or base editing systems described herein. In some embodiments, the fusion protein is delivered as an RNP, or as an mRNA or plasmid comprising a sequence encoding a fusion protein described herein.

Also provided herein are methods for deaminating a selected cytosine in a nucleic acid, the method comprising contacting the nucleic acid with a fusion protein or base editing system described herein.

Additionally, provided herein are compositions comprising a purified a fusion protein or base editing system as described herein.

Further, provided herein are nucleic acids encoding a fusion protein or base editing system described herein, as well as vectors comprising the nucleic acids, and host cells comprising the nucleic acids, e.g., stem cells, e.g., hematopoietic stem cells.

Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Methods and materials are described herein for use in the present invention; other, suitable methods and materials known in the art can also be used. The materials, methods, and examples are illustrative only and not intended to be limiting. All publications, patent applications, patents, sequences, database entries, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control.

Other features and advantages of the invention will be apparent from the following detailed description and figures, and from the claims.

DESCRIPTION OF DRAWINGS

FIG. 1. Cas9 PID mutants or Type II-C Cas9s as chimeric BE technologies with ZFs. The 6-finger zinc finger array and UGI are fused to the C-terminus of nCas9 and any of the wild type or engineered deaminase domains is fused to the N-terminus of the nCas9. The ZF binding sequence is directly adjacent to the gRNA target site. In this model, binding of the ZF is a prerequisite for formation of the R-loop by nCas9, greatly reducing the number of possible off-target sites at which R-loops might be formed to those that are adjacent to a ZF binding site. The analogous strategy using TALEs instead of ZFs is also described herein.

FIG. 2. Type II-C Cas9s demonstrate increased binding and nuclease activity at their target sites in human cells when fused to 3-finger ZFs. In this assay, a chromosomally-integrated EGFP gene is used as a reporter for binding (via steric hindrance of transcription) or nuclease activity (via NHEJ-mediated small insertion and deletions in the coding sequence). Transient transfection of plasmids expressing various Cas9 orthologs and gRNAs with 20- or 23-nucleotide homology to the EGFP reporter results in disruption or repression of EGFP, resulting in EGFP-negative cells when assayed by flow cytometry. In all cases, addition of a ZF fusion partner increases the EGFP disruption rate of the Cas9 ortholog tested. Notably, addition of a ZF fusion partner to PlCas9 increases EGFP disruption activity of this ortholog from background levels to 85%.

FIG. 3. Diagram of a bipartite base editing architecture. A nicking Cas9 (nCas9) is targeted to the deamination site by a guide RNA (gRNA). Subsequent R-loop formation produces an ssDNA editing window on the DNA strand that is not bound by nCas9 (deaminated strand). The zinc finger-deaminase complex targets dsDNA near the formed R-loop. Target cytosines in the editing window get deaminated. Nicking Cas9 induces a single strand break (SSB) in the genomic DNA (non-deaminated strand) thus attracting DNA repair proteins, e.g. involving the mismatch repair (MMR) pathway. As the edited target cytosine is converted to a uracil and the deaminated strand serves as a template for the nicked (undeaminated) strand repair is biased towards the intended cytosine to thymine transition.

FIG. 4. Superimposed cytosine to thymine (C-to-T) transitions at gRNA 5 and gRNA 2 target sites in different experimental conditions using hA3A fusions. Conventional hA3A-BEs (hA3A-nCas9-UGI) serve as positive controls. Bipartite base editors (BBEs) with zinc finger-deaminase complex binding 5′ of the Cas9 target sites show potent base editing at the respective target sites. hA3A-ZF-UGI alone or BBEs targeting sites nearby show no or greatly reduced substitution rates compared to targeted BBEs. SEQ ID NOs.:1-2 are shown.

FIG. 5. Superimposed C-to-T transitions at gRNA 2 target site comparing rAPOBEC1-fusions. Conventional rAPOBEC1-BEs (BE3) serve as a positive control. Bipartite base editors (BBEs) with zinc finger-deaminase complex binding 5′ of the Cas9 target sites show potent base editing at the target site. rAPOBEC1-ZF-UGI alone show no base editing activity compared to targeted BBEs. SEQ ID NO:3 is shown.

FIG. 6. Superimposed cytosine to thymine transitions at gRNA 4 target site comparing hA3A-fusions. In contrast to previous experiments, the zinc finger-deaminase complex is oriented anti-parallel to nCas9, binding 3′ of the gRNA 4 target site. Thus the N-terminal deaminase (hA3A) is facing the 3′ end of the non-target strand (ssDNA deamination strand) which leads to a shift of C-to-T editing to the 3′ end and outside of the canonical BE1-4 editing window. The spatial control of the base editing window by differential orientation/positioning of the ZF-deaminase component is shown. SEQ ID NO:6 is shown.

FIGS. 7A-7D. Bipartite base editors may confer enhanced product purity by flanking the editing window with UGIs and spatial separation of the nicking domain and the deaminase. (7A) shows “product impurities” of conventional cytosine to thymine base editing which consist of cytosine to purine (R) byproducts as well as indels. (7B) Compared to conventional deaminase-nCas9-UGI fusions (e.g. BE3 or hA3A-nCas9-UGI), BBEs containing a dual, bipartite UGI show greatly reduced cytosine to purine (R) byproducts across all cytidines of the EGFP gRNA2 target site. (7C) Superimposition of single bases across the gRNA2 site showing relevant deletion frequencies. Both hA3A and rAPOBEC1 bipartite base editors markedly decrease deletion frequencies back to control levels. (7D) Depicting the data from C as a distribution of deletion frequencies across the target site and +/−25 bp flanking sites.

DETAILED DESCRIPTION

In the most efficient BE configuration described to date, a cytidine deaminase (DA) domain and uracil glycosylase inhibitor (UGI; a small bacteriophage protein that inhibits host cell uracil DNA glycosylase (UDG), the enzyme responsible for excising uracil from the genome1,4) are both fused to nCas9 (derived from either Streptococcus pyogenes Cas9 (SpCas9) or Staphylococcus aureus Cas9 (SaCas9). The nCas9 forms an R-loop at a target site specified by its guide RNA (gRNA) and recognition of an adjacent protospacer adjacent motif (PAM), leaving approximately 4-8 nucleotides of the non-target strand exposed as single stranded DNA (ssDNA) near the PAM-distal end of the R-loop. This region of the ssDNA is the template that is able to be deaminated by the ssDNA-specific DA domain to produce a guanosine:uracil (G:U) mismatch and defines the editing window. The nCas9 nicks the non-deaminated strand of DNA, biasing conversion of the G:U mismatch to an adenine:thymine (A:T) base pair by directing the cell to repair the nick lesion using the deaminated strand as a template. To date, the DA domains described in these fusion proteins have been rat APOBEC1 (rAPO1), an activation-induced cytidine deaminase (AID) derived from lamprey termed CDA (PmCDA), human AID (hAID), or a hyperactive form of hAID lacking a nuclear export signal1-2,5-7. BE technology was primarily established using the SpCas9 protein for its nCas9 domain (nSpCas9), but although herein we refer to nCas9, in general any Cas9-like nickase or catalytically inactive Cas9 could be used based on any ortholog of the Cas9 protein (including the related Cpf1 enzyme class), unless specifically indicated.

An important consideration for the use of BE in therapeutic settings will be to assess its genome-wide capacity for off-target mutagenesis and to modify the technology to minimize or, ideally, to eliminate the risks of stimulating deleterious off-target mutations. Herein, we described technological improvements to BEs that can be used to reduce or eliminate potential unwanted BE mutagenesis.

Zinc-Finger/Cas9 Fusion Proteins Limit Off-Target Binding and Deamination

While the fusion of RNA-guided nucleases (e.g., nCas9, see Table 1 for a non-exhaustive list of exemplary possible fusion orthologs) to cytidine or adenine deaminases (BEs) has been proven to be an efficient tool, the main safety concern related to this technology centers around the possibility of unintended deamination of DNA. While unbiased genome-wide detection of Cas9-independent deaminase activity on single strand DNA (ssDNA) is lacking, recently published data suggests off-target base editing takes place at known off-target binding sites of Cas96,7. As this type of off-target is related to the targeting and binding of the nuclease, as opposed to cleavage-related off-targeting, the use of high fidelity Cas9 variants might be of limited use in this context.

Importantly, Cas9:gRNA targeting can often tolerate off-target binding events to various degrees depending on the specific gRNA sequence, which could serve as an important source of off-target BE mutagenesis8. While efforts to engineer S. pyogenes Cas9 (SpCas9) to increase its specificity as a nuclease have been largely successful, the specificity advantages of engineered reagents like high-fidelity SpCas9 (HF-Cas9)9 or eSpCas910 may not translate to contexts where Cas9 is being used mainly as a targeting platform, as with BEs. It is widely thought that the thermodynamic parameters for Cas9 binding and Cas9 cleavage are significantly different, as evidenced by the observation that very short gRNAs (˜12-14 nts) are sufficient to target catalytically-inactive SpCas9-based transcription factor fusions to alter gene expression11, while gRNAs 17 nucleotides or longer are required for DNA cleavage by SpCas912. Therefore, engineering BE's nSpCas9 domain to simply include the engineered HF-SpCas9 mutations may not be the optimal strategy for eliminating the capacity for BE's nSpCas9 domain to stimulate off-target deamination events.

One strategy to reduce off-target deamination events is to restrict nCas9 binding and R-loop formation to gRNA sites that are directly adjacent to a transcription-activator-like effector (TALE) or zinc finger array (ZF) binding site (FIG. 3) whose cognate TALE or ZF has been incorporated into the BE architecture. Several groups have described “traditional” double-strand-break inducing genome-editing reagents that make use of an extra engineered DNA binding domain in addition to what is usually considered necessary to increase overall specificity of the fusion protein13,14. Notably, a fusion between a knocked-down version of SpCas9 and a 6-finger ZF domain was shown to increase specificity of SpCas9-mediated genome editing when the ZF and gRNA are targeted to adjacent genomic sequences14. Described herein is improved BE technology using non-R-loop-forming DNA binding domain such as aTALE or a ZF to limit the number and scope of transient R-loops and thus ssDNA exposed for deamination by BE across the genome. This strategy increases the number of bases specified to include those by the gRNA in addition to the bases specified by a TALE or ZF domain (for example, addition of 3-finger ZF domain would specify ˜9-10 more bases than nCas9 alone).

To accomplish this, TALE domain arrays or ZF arrays are fused to the N- or C-termini of Cas9 proteins that are unable to bind efficiently to the target sequence specified by their gRNAs in human cells (see Table 2 for a list of proteins that can be used in these fusion proteins). Mutations to the PAM interacting domain (PID) of the Type II-A Cas9 proteins SpCas9 or SaCas9 are sufficient to prevent these proteins from functioning efficiently in human cells without a second DNA-binding domain fusion partner14,15 (see Table 2 for a list of PID mutations). Type II-C Cas9 orthologs are an evolutionarily divergent class of Cas9 enzymes that do not possess the ability to unwind double stranded DNA16. Some of these Cas9 orthologs are unable to cleave genomic sites in human cells when co-expressed with their cognate gRNAs; however, we have found that the added affinity of a 3-finger ZF fusion partner allows these Cas9 orthologs to bind and cleave genomic sites with high activity (FIG. 2). In this setup, a UGI is fused either to the opposite terminus of the ZF as the nCas9 or co-expressed with the ZF-Cas9 fusion using either a self-cleaving peptide such as a 2A element or be independently translated from an internal ribosome entry site. This strategy can also be used with TALE DNA binding domains in place of the ZFs. In any configuration, such configurations can be used to enhance BE fidelity by extending the functional DNA binding site specified by the targeting complex.

Bipartite Base Editor (BBE) Architecture Limits Off-Target Deamination, Enhances Product Purity & Enables Spatial Control of the Base Editing Window

A bipartite BE (BBE) platform, which includes a non-R-loop-forming DNA-binding domain (such as a ZF or a TALE) fused to a deaminase domain and an R-loop-forming Cas9 component (either catalytically inactive dCas9 or nicking nCas9 or a similarly inactive Cas9 ortholog) that are targeted to adjacent sequences as shown in FIG. 3, can also be used to enhance BE specificity. In this configuration, binding of the deaminase-containing protein requires adjacent binding of an R-loop-forming protein to function efficiently, since deaminases (in principle, any deaminase domain listed in Table 2, or any other enzymatic domain capable of chemically mutating DNA bases, can be considered) used for base-editing are primarily active on ssDNA, which is exposed during R-loop formation. This configuration can be used to increase specificity by effectively extending the overall number of DNA bases that need to be bound by the complex to mutate DNA, since each off-target binding of either the ZF- or TALE-deaminase or R-loop forming components on their own should have a reduced capacity to mutate DNA. Though it still includes a fully-intact deaminase enzyme, off target binding of a zinc finger-deaminase protein has a significantly reduced capacity to cause mutations compared to standard BEs since the ZF-deaminase lacks the capability to create a stable R-loop to expose ssDNA. At the same time, off-target binding of a dCas9 or nCas9 molecule has no capacity to stimulate unwanted deamination events on its own, since it lacks a deaminase domain, so it can be considered enzymatically inert and can only induce low-level off-target mutations through its ability to create single-stranded nicks in DNA or by exposing ssDNA to other nuclear factors as a by-product of R-loop formation.

Second, while the nCas9 (with or without UGI) component of a BBE can bind to and nick putative off-target sites in this configuration, it cannot stimulate off-target base-editing on its own since it lacks a deaminase domain. Being a highly modular system, on-target deamination can be tightly controlled by co-expression/application of various nicking or catalytically inactive Cas9 variants (see Table 1 for possible orthologs), e.g., enabling different PAM preferences. In summary, the genomic co-localization (an “AND gate”) comes from two different proteins that are independently expressed minimizing off-targeting risk. Each “half” of this bipartite base editing (BBE) system will be necessary to direct deaminase-mediated mutagenesis at a target site of interest, reducing the potential of off-target deamination and resulting mutations at nCas9 off-target sites across the genome by limiting deaminase localization to sites adjacent to nCas9-mediated R-loop formation.

In summary, the BBE platform offers highly optimized product purity at constant or potentially enhanced target cytosine to thymine editing efficiency with the benefit of enhanced editing window control and the prospect of reduced genome-wide Cas9-associated off-target deamination.

DNA Binding Domain

The present fusion proteins can include one, two, or more DNA binding domains such as zinc-finger nucleases, transcription activator effectors (TALEs), and Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) Cas RNA-guided nucleases (RGN).

CRISPR-Cas Nucleases

Although herein we refer to nCas9 and dCas9, in general, for the BBE system any R-loop forming Cas9-like enzyme that is inactive or has only nickase activity could be used, e.g., any ortholog of the Cas9 protein (including the related Cpf1 enzyme class), unless specifically indicated.

TABLE 1 List of Exemplary Cas9 Orthologs UniProt Nickase Accession Mutations/Catalytic Ortholog Number residues S. pyogenes Cas9 (SpCas9) Q99ZW2 D10A, E762A, H840A, N854A, N863A, D986A18 S. aureus Cas9 (SaCas9) J7RUA5 D10A and N5801819 S. thermophilus Cas9 G3ECR1 D31A and N891A20 (St1Cas9) S. pasteurianus Cas9 F5X275 D10, H599* (SpaCas9) C. jejuni Cas9 (CjCas9) Q0P897 D8A, H559A21 F. novicida Cas9 (FnCas9) A0Q5Y3 D11, N99522 P. lavamentivorans Cas9 A7HP89 D8, H601* (PlCas9) C. lari Cas9 (ClCas9) G1UFN3 D7, H567* F. novicida Cpf1 A0Q7Q2 D917, E1006, D125523 (FnCpf1) M. bovoculi Cpf1 Sequence N/A** (MbCpf1) given at end A. sp. BV3L6 (AsCpf1) U2UMQ6 D908, 993E, Q1226, D126324 L. bacterium N2006 A0A182DWE3 D832A25 (LbCpf1) *predicted based on UniRule annotation on the UniProt database. **May be determinable based on sequence alignment with other Cpf1 orthologs

These orthologs, and mutants and variants thereof as known in the art, can be used in any of the fusion proteins described herein. See, e.g., WO 2017/040348 (which describes variants of SaCas9 and SpCas 9 with increased specificity) and WO 2016/141224 (which describes variants of SaCas9 and SpCas9 with altered PAM specificity).

The Cas9 nuclease from S. pyogenes (hereafter simply Cas9) can be guided via simple base pair complementarity between 17-20 nucleotides of an engineered guide RNA (gRNA), e.g., a single guide RNA or crRNA/tracrRNA pair, and the complementary strand of a target genomic DNA sequence of interest that lies next to a protospacer adjacent motif (PAM), e.g., a PAM matching the sequence NGG or NAG (Shen et al., Cell Res (2013); Dicarlo et al., Nucleic Acids Res (2013); Jiang et al., Nat Biotechnol 31, 233-239 (2013); Jinek et al., Elife 2, e00471 (2013); Hwang et al., Nat Biotechnol 31, 227-229 (2013); Cong et al., Science 339, 819-823 (2013); Mali et al., Science 339, 823-826 (2013c); Cho et al., Nat Biotechnol 31, 230-232 (2013); Jinek et al., Science 337, 816-821 (2012)). The engineered CRISPR from Prevotella and Francisella 1 (Cpf1) nuclease can also be used, e.g., as described in Zetsche et al., Cell 163, 759-771 (2015); Schunder et al., Int J Med Microbiol 303, 51-60 (2013); Makarova et al., Nat Rev Microbiol 13, 722-736 (2015); Fagerlund et al., Genome Biol 16, 251 (2015). Unlike SpCas9, Cpf1 requires only a single 42-nt crRNA, which has 23 nt at its 3′ end that are complementary to the protospacer of the target DNA sequence (Zetsche et al., 2015). Furthermore, whereas SpCas9 recognizes an NGG PAM sequence that is 3′ of the protospacer, AsCpf1 and LbCp1 recognize TTTN PAMs that are found 5′ of the protospacer (Id.).

In some embodiments, the present system utilizes a wild type or variant Cas9 protein from S. pyogenes or Staphylococcus aureus, or a wild type Cpf1 protein from Acidaminococcus sp. BV3L6 or Lachnospiraceae bacterium ND2006 either as encoded in bacteria or codon-optimized for expression in mammalian cells and/or modified in its PAM recognition specificity and/or its genome-wide specificity. A number of variants have been described; see, e.g., WO 2016/141224, PCT/US2016/049147, Kleinstiver et al., Nat Biotechnol. 2016 August; 34(8):869-74; Tsai and Joung, Nat Rev Genet. 2016 May; 17(5):300-12; Kleinstiver et al., Nature. 2016 Jan. 28; 529(7587):490-5; Shmakov et al., Mol Cell. 2015 Nov. 5; 60(3):385-97; Kleinstiver et al., Nat Biotechnol. 2015 December; 33(12):1293-1298; Dahlman et al., Nat Biotechnol. 2015 November; 33(11):1159-61; Kleinstiver et al., Nature. 2015 Jul. 23; 523(7561):481-5; Wyvekens et al., Hum Gene Ther. 2015 July; 26(7):425-31; Hwang et al., Methods Mol Biol. 2015; 1311:317-34; Osborn et al., Hum Gene Ther. 2015 February; 26(2):114-26; Konermann et al., Nature. 2015 Jan. 29; 517(7536):583-8; Fu et al., Methods Enzymol. 2014; 546:21-45; and Tsai et al., Nat Biotechnol. 2014 June; 32(6):569-76, inter alia. The guide RNA is expressed or present in the cell together with Cas9 or Cpf1. Either the guide RNA or the nuclease, or both, can be expressed transiently or stably in the cell or introduced as a purified protein or nucleic acid.

Table 2 shows potential Cas9 orthologs that can be used as chimeric proteins with ZFs or TALEs. PlCas9, ClCas9, and NcCas9 are unable to efficiently bind their target sites in eukaryotic cells, while CjCas9 has substantial activity at its target sites, necessitating PAM interacting domain mutations to knock down its ability to efficiently bind its site. Nickase mutation residues represents a position of the enzyme either known to be required for catalytic activity of the conserved RuvC nuclease domain or predicted to be required for this catalytic activity based on sequence alignment to CjCas9 where structural information is lacking

TABLE 2 Cas9 orthologs Nickase Cas9 ortholog Enzyme class PID mutations mutation SpCas9 Type II-A R1333K/S, R1335K/S D10A SaCas9 Type II-A E782K/N968K/R1015H D10A PlCas9* Type II-C D8A ClCas9* Type II-C D7A NcCas9* Type II-C D8A CjCas9 Type II-C R866K/S, T913S, S915T D8A *indicates proteins lacking sufficient structural information. All positional information refers to the wild-type protein sequences acquired from uniprot.org.

In some embodiments, the Cas9 also includes one or mutations that reduce nuclease activity of the Cas9; e.g., for SpCas9, mutations at D10A or H840A (which creates a single-strand nickase).

In some embodiments, the SpCas9 variants also include mutations at one of the following amino acid positions, which destroy the nuclease activity of the Cas9: D10, E762, D839, H983, or D986 and H840 or N863, e.g., D10A/D10N and H840A/H840N/H840Y, to render the nuclease portion of the protein catalytically inactive; substitutions at these positions could be alanine (as they are in Nishimasu al., Cell 156, 935-949 (2014)), or other residues, e.g., glutamine, asparagine, tyrosine, serine, or aspartate, e.g., E762Q, H983N, H983Y, D986N, N863D, N863S, or N863H (see WO 2014/152432).

In some embodiments, the fusion proteins include one or more Uracil glycosylase inhibitor (UGI) protein sequences; an exemplary UGI sequence is as follows: TNLSDIIEKETGKQLVIQESILMLPEEVEEVIGNKPESDILVHTAYDESTDENVM LLTSDAPEYKPWALVIQDSNGENKIKML (SEQ ID NO:7; Uniprot: P14739). Typically, the UGIs are at the C-terminal, but can also be at the N terminal, or between the components of the fusion proteins described herein. Linkers as known in the art can be used.

TAL Effector Repeat Arrays

TAL effectors of plant pathogenic bacteria in the genus Xanthomonas play important roles in disease, or trigger defense, by binding host DNA and activating effector-specific host genes. Specificity depends on an effector-variable number of imperfect, typically ˜33-35 amino acid repeats. Polymorphisms are present primarily at repeat positions 12 and 13, which are referred to herein as the repeat variable-diresidue (RVD). The RVDs of TAL effectors correspond to the nucleotides in their target sites in a direct, linear fashion, one RVD to one nucleotide, with some degeneracy and no apparent context dependence. In some embodiments, the polymorphic region that grants nucleotide specificity may be expressed as a triresidue or triplet.

Each DNA binding repeat can include a RVD that determines recognition of a base pair in the target DNA sequence, wherein each DNA binding repeat is responsible for recognizing one base pair in the target DNA sequence. In some embodiments, the RVD can comprise one or more of: HA for recognizing C; ND for recognizing C; HI for recognizing C; HN for recognizing G; NA for recognizing G; SN for recognizing G or A; YG for recognizing T; and NK for recognizing C and one or more of: HD for recognizing C; NG for recognizing T; NI for recognizing A; NN for recognizing G or A; NS for recognizing A or C or G or T; N* for recognizing C or T, wherein * represents a gap in the second position of the RVD; HG for recognizing T; H* for recognizing T, wherein * represents a gap in the second position of the RVD; and IG for recognizing T.

TALE proteins may be useful in research and biotechnology as targeted chimeric nucleases that can facilitate homologous recombination in genome engineering (e.g., to add or enhance traits useful for biofuels or biorenewables in plants). These proteins also may be useful as, for example, transcription factors, and especially for therapeutic applications requiring a very high level of specificity such as therapeutics against pathogens (e.g., viruses) as non-limiting examples.

Methods for generating engineered TALE arrays are known in the art, see, e.g., the fast ligation-based automatable solid-phase high-throughput (FLASH) system described in U.S. Ser. No. 61/610,212, and Reyon et al., Nature Biotechnology 30, 460-465 (2012); as well as the methods described in Bogdanove & Voytas, Science 333, 1843-1846 (2011); Bogdanove et al., Curr Opin Plant Biol 13, 394-401 (2010); Scholze & Boch, J. Curr Opin Microbiol (2011); Boch et al., Science 326, 1509-1512 (2009); Moscou & Bogdanove, Science 326, 1501 (2009); Miller et al., Nat Biotechnol 29, 143-148 (2011); Morbitzer et al., T. Proc Natl Acad Sci USA 107, 21617-21622 (2010); Morbitzer et al., Nucleic Acids Res 39, 5790-5799 (2011); Zhang et al., Nat Biotechnol 29, 149-153 (2011); Geissler et al., PLoS ONE 6, e19509 (2011); Weber et al., PLoS ONE 6, e19722 (2011); Christian et al., Genetics 186, 757-761 (2010); Li et al., Nucleic Acids Res 39, 359-372 (2011); Mahfouz et al., Proc Natl Acad Sci USA 108, 2623-2628 (2011); Mussolino et al., Nucleic Acids Res (2011); Li et al., Nucleic Acids Res 39, 6315-6325 (2011); Cermak et al., Nucleic Acids Res 39, e82 (2011); Wood et al., Science 333, 307 (2011); Hockemeye et al. Nat Biotechnol 29, 731-734 (2011); Tesson et al., Nat Biotechnol 29, 695-696 (2011); Sander et al., Nat Biotechnol 29, 697-698 (2011); Huang et al., Nat Biotechnol 29, 699-700 (2011); and Zhang et al., Nat Biotechnol 29, 149-153 (2011); all of which are incorporated herein by reference in their entirety.

Also suitable for use in the present methods are MegaTALs, which are a fusion of a meganuclease with a TAL effector; see, e.g., Boissel et al., Nucl. Acids Res. 42(4):2591-2601 (2014); Boissel and Scharenberg, Methods Mol Biol. 2015; 1239:171-96.

Zinc Fingers

Zinc finger proteins are DNA-binding proteins that contain one or more zinc fingers, independently folded zinc-containing mini-domains, the structure of which is well known in the art and defined in, for example, Miller et al., 1985, EMBO J., 4:1609; Berg, 1988, Proc. Natl. Acad. Sci. USA, 85:99; Lee et al., 1989, Science. 245:635; and Klug, 1993, Gene, 135:83. Crystal structures of the zinc finger protein Zif268 and its variants bound to DNA show a semi-conserved pattern of interactions, in which typically three amino acids from the alpha-helix of the zinc finger contact three adjacent base pairs or a “subsite” in the DNA (Pavletich et al., 1991, Science, 252:809; Elrod-Erickson et al., 1998, Structure, 6:451). Thus, the crystal structure of Zif268 suggested that zinc finger DNA-binding domains might function in a modular manner with a one-to-one interaction between a zinc finger and a three-base-pair “subsite” in the DNA sequence. In naturally occurring zinc finger transcription factors, multiple zinc fingers are typically linked together in a tandem array (e.g., of 3, 4, or 6 zinc fingers) to achieve sequence-specific recognition of a contiguous DNA sequence (Klug, 1993, Gene 135:83).

Multiple studies have shown that it is possible to artificially engineer the DNA binding characteristics of individual zinc fingers by randomizing the amino acids at the alpha-helical positions involved in DNA binding and using selection methodologies such as phage display to identify desired variants capable of binding to DNA target sites of interest (Rebar et al., 1994, Science, 263:671; Choo et al., 1994 Proc. Natl. Acad. Sci. USA, 91:11163; Jamieson et al., 1994, Biochemistry 33:5689; Wu et al., 1995 Proc. Natl. Acad. Sci. USA, 92: 344). Such recombinant zinc finger proteins can be fused to functional domains, such as transcriptional activators, transcriptional repressors, methylation domains, and nucleases to regulate gene expression, alter DNA methylation, and introduce targeted alterations into genomes of model organisms, plants, and human cells (Carroll, 2008, Gene Ther., 15:1463-68; Cathomen, 2008, Mol. Ther., 16:1200-07; Wu et al., 2007, Cell. Mol. Life Sci., 64:2933-44).

One existing method for engineering zinc finger arrays, known as “modular assembly,” advocates the simple joining together of pre-selected zinc finger modules into arrays (Segal et al., 2003, Biochemistry, 42:2137-48; Beerli et al., 2002, Nat. Biotechnol., 20:135-141; Mandell et al., 2006, Nucleic Acids Res., 34:W516-523; Carroll et al., 2006, Nat. Protoc. 1:1329-41; Liu et al., 2002, J. Biol. Chem., 277:3850-56; Bae et al., 2003, Nat. Biotechnol., 21:275-280; Wright et al., 2006, Nat. Protoc., 1:1637-52). Although straightforward enough to be practiced by any researcher, recent reports have demonstrated a high failure rate for this method, particularly in the context of zinc finger nucleases (Ramirez et al., 2008, Nat. Methods, 5:374-375; Kim et al., 2009, Genome Res. 19:1279-88), a limitation that typically necessitates the construction and cell-based testing of very large numbers of zinc finger proteins for any given target gene (Kim et al., 2009, Genome Res. 19:1279-88).

Combinatorial selection-based methods that identify zinc finger arrays from randomized libraries have been shown to have higher success rates than modular assembly (Maeder et al., 2008, Mol. Cell, 31:294-301; Joung et al., 2010, Nat. Methods, 7:91-92; Isalan et al., 2001, Nat. Biotechnol., 19:656-660). In preferred embodiments, the zinc finger arrays are described in, or are generated as described in, WO 2011/017293 and WO 2004/099366. Additional suitable zinc finger DBDs are described in U.S. Pat. Nos. 6,511,808, 6,013,453, 6,007,988, and 6,503,717 and U.S. patent application 2002/0160940.

Bipartite Base Editors

In some embodiments, the base editor is a fusion of a DNA-binding domain to a deaminase, or an active portion thereof, that modifies cytosine DNA bases, e.g., a cytidine deaminase from the apolipoprotein B mRNA-editing enzyme, catalytic polypeptide-like (APOBEC) family of deaminases, including APOBEC1, APOBEC2, APOBEC3A, APOBEC3B, APOBEC3C, APOBEC3D/E, APOBEC3F, APOBEC3G, APOBEC3H, APOBEC4 (see, e.g., Yang et al., J Genet Genomics. 2017 Sep. 20; 44(9):423-437); activation-induced cytidine deaminase (AID), e.g., activation induced cytidine deaminase (AICDA), cytosine deaminase 1 (CDA1), and CDA2, and cytosine deaminase acting on tRNA (CDAT). The following Table 2 provides exemplary sequences; other sequences can also be used.

TABLE 2 Gen Bank Accession Nos. Deaminase Nucleic Acid Amino Acid hAID/AICDA NM_020661.3 isoform 1 NP_065712.1 variant 1 NM_020661.3 isoform 2 NP_065712.1 variant 2 rat APOBEC1 NM_012907.2 NP_037039.1 hAPOBEC1 NM_001644.4 isoform a NP_001635.2 variant 1 NM_005889.3 isoform b NP_005880.2 variant 3 hAPOBEC2 NM_006789.3 NP_006780.1 hAPOBEC3A NM_145699.3 isoform a NP_663745.1 variant 1 NM_001270406.1 NP_001257335.1 isoform b variant 2 hAPOBEC3B NM_004900.4 isoform a NP_004891.4 variant 1 NM_001270411.1 NP_001257340.1 isoform b variant 2 hAPOBEC3C NM_014508.2 NP_055323.2 hAPOBEC3D NM_152426.3 NP_689639.2 APOBEC3F NM_145298.5 isoform a NP_660341.2 variant 1 NM_001006666.1 NP_001006667.1 isoform b variant 2 APOBEC3G NM_021822.3 (isoform a) NP_068594.1 (variant 1) APOBEC3H NM_001166003.2 NP_001159475.2 (variant SV-200) APOBEC4 NM_203454.2 NP_982279.1 CDA1* NM_127515.4 NP_179547.1 *from Saccharomyces cerevisiae S288C

Variants

In some embodiments, the components of the fusion proteins are at least 80%, e.g., at least 85%, 90%, 95%, 97%, or 99% identical to the amino acid sequence of a exemplary sequence (e.g., as provided herein), e.g., have differences at up to 5%, 10%, 15%, or 20% of the residues of the exemplary sequence replaced, e.g., with conservative mutations, in addition to the mutations described herein. In preferred embodiments, the variant retains desired activity of the parent, e.g., in the case of a Cas9 variant or fragment, nickase activity or no nuclease activity, and/or the ability to interact with a guide RNA and/or target DNA, or deaminase activity in the case of a deaminase variant or active portion thereof.

To determine the percent identity of two nucleic acid sequences, the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in one or both of a first and a second amino acid or nucleic acid sequence for optimal alignment and non-homologous sequences can be disregarded for comparison purposes). The length of a reference sequence aligned for comparison purposes is at least 80% of the length of the reference sequence, and in some embodiments is at least 90% or 100%. The nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position (as used herein nucleic acid “identity” is equivalent to nucleic acid “homology”). The percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which need to be introduced for optimal alignment of the two sequences. Percent identity between two polypeptides or nucleic acid sequences is determined in various ways that are within the skill in the art, for instance, using publicly available computer software such as Smith Waterman Alignment (Smith, T. F. and M. S. Waterman (1981) J Mol Biol 147:195-7); “BestFit” (Smith and Waterman, Advances in Applied Mathematics, 482-489 (1981)) as incorporated into GeneMatcher Plus™, Schwarz and Dayhof (1979) Atlas of Protein Sequence and Structure, Dayhof, M. O., Ed, pp 353-358; BLAST program (Basic Local Alignment Search Tool; (Altschul, S. F., W. Gish, et al. (1990) J Mol Biol 215: 403-10), BLAST-2, BLAST-P, BLAST-N, BLAST-X, WU-BLAST-2, ALIGN, ALIGN-2, CLUSTAL, or Megalign (DNASTAR) software. In addition, those skilled in the art can determine appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the length of the sequences being compared. In general, for proteins or nucleic acids, the length of comparison can be any length, up to and including full length (e.g., 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 100%). For purposes of the present compositions and methods, at least 80% of the full length of the sequence is aligned.

For purposes of the present disclosure, the comparison of sequences and determination of percent identity between two sequences can be accomplished using a Blossum 62 scoring matrix with a gap penalty of 12, a gap extend penalty of 4, and a frameshift gap penalty of 5.

Conservative substitutions typically include substitutions within the following groups: glycine, alanine; valine, isoleucine, leucine; aspartic acid, glutamic acid, asparagine, glutamine; serine, threonine; lysine, arginine; and phenylalanine, tyrosine.

Also provided herein are isolated nucleic acids encoding the bipartite deaminase fusion proteins, vectors comprising the isolated nucleic acids, optionally operably linked to one or more regulatory domains for expressing the variant proteins, and host cells, e.g., mammalian host cells, comprising the nucleic acids, and optionally expressing the variant proteins. In some embodiments, the host cells are stem cells, e.g., hematopoietic stem cells.

The bipartite deaminase fusion proteins described herein can be used for altering the genome of a cell. The methods generally include expressing or contacting the fusion proteins in the cells; in versions using one or two Cas9s, the methods include using a guide RNA having a region complementary to a selected portion of the genome of the cell. Methods for selectively altering the genome of a cell are known in the art, see, e.g., U.S. Pat. No. 8,993,233; US 20140186958; U.S. Pat. No. 9,023,649; WO/2014/099744; WO 2014/089290; WO2014/144592; WO144288; WO2014/204578; WO2014/152432; WO2115/099850; U.S. Pat. No. 8,697,359; US20160024529; US20160024524; US20160024523; US20160024510; US20160017366; US20160017301; US20150376652; US20150356239; US20150315576; US20150291965; US20150252358; US20150247150; US20150232883; US20150232882; US20150203872; US20150191744; US20150184139; US20150176064; US20150167000; US20150166969; US20150159175; US20150159174; US20150093473; US20150079681; US20150067922; US20150056629; US20150044772; US20150024500; US20150024499; US20150020223; US20140356867; US20140295557; US20140273235; US20140273226; US20140273037; US20140189896; US20140113376; US20140093941; US20130330778; US20130288251; US20120088676; US20110300538; US20110236530; US20110217739; US20110002889; US20100076057; US20110189776; US20110223638; US20130130248; US20150050699; US20150071899; US20150050699; US20150045546; US20150031134; US20150024500; US20140377868; US20140357530; US20140349400; US20140335620; US20140335063; US20140315985; US20140310830; US20140310828; US20140309487; US20140304853; US20140298547; US20140295556; US20140294773; US20140287938; US20140273234; US20140273232; US20140273231; US20140273230; US20140271987; US20140256046; US20140248702; US20140242702; US20140242700; US20140242699; US20140242664; US20140234972; US20140227787; US20140212869; US20140201857; US20140199767; US20140189896; US20140186958; US20140186919; US20140186843; US20140179770; US20140179006; US20140170753; WO/2008/108989; WO/2010/054108; WO/2012/164565; WO/2013/098244; WO/2013/176772; US 20150071899; 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.

In some embodiments, the fusion proteins include a linker between the DNA binding domain (e.g., ZFN, TALE, or nCas9) and the BE domains. Linkers that can be used in these fusion proteins (or between fusion proteins in a concatenated structure) can include any sequence that does not interfere with the function of the fusion proteins. In preferred embodiments, the linkers are short, e.g., 2-20 amino acids, and are typically flexible (i.e., comprising amino acids with a high degree of freedom such as glycine, alanine, and serine). In some embodiments, the linker comprises one or more units consisting of GGGS (SEQ ID NO:8) or GGGGS (SEQ ID NO:9), e.g., two, three, four, or more repeats of the GGGS (SEQ ID NO:8) or GGGGS (SEQ ID NO:9) unit. Other linker sequences can also be used.

In some embodiments, the fusion protein includes a cell-penetrating peptide sequence that facilitates delivery to the intracellular space, e.g., HIV-derived TAT peptide, penetratins, transportans, or hCT derived cell-penetrating peptides, see, e.g., Caron et al., (2001) Mol Ther. 3(3):310-8; Langel, Cell-Penetrating Peptides: Processes and Applications (CRC Press, Boca Raton Fla. 2002); El-Andaloussi et al., (2005) Curr Pharm Des. 11(28):3597-611; and Deshayes et al., (2005) Cell Mol Life Sci. 62(16):1839-49.

Cell penetrating peptides (CPPs) are short peptides that facilitate the movement of a wide range of biomolecules across the cell membrane into the cytoplasm or other organelles, e.g. the mitochondria and the nucleus. Examples of molecules that can be delivered by CPPs include therapeutic drugs, plasmid DNA, oligonucleotides, siRNA, peptide-nucleic acid (PNA), proteins, peptides, nanoparticles, and liposomes. CPPs are generally 30 amino acids or less, are derived from naturally or non-naturally occurring protein or chimeric sequences, and contain either a high relative abundance of positively charged amino acids, e.g. lysine or arginine, or an alternating pattern of polar and non-polar amino acids. CPPs that are commonly used in the art include Tat (Frankel et al., (1988) Cell. 55:1189-1193, Vives et al., (1997) J. Biol. Chem. 272:16010-16017), penetratin (Derossi et al., (1994) J. Biol. Chem. 269:10444-10450), polyarginine peptide sequences (Wender et al., (2000) Proc. Natl. Acad. Sci. USA 97:13003-13008, Futaki et al., (2001) J. Biol. Chem. 276:5836-5840), and transportan (Pooga et al., (1998) Nat. Biotechnol. 16:857-861).

CPPs can be linked with their cargo through covalent or non-covalent strategies. Methods for covalently joining a CPP and its cargo are known in the art, e.g. chemical cross-linking (Stetsenko et al., (2000) J. Org. Chem. 65:4900-4909, Gait et al. (2003) Cell. Mol. Life. Sci. 60:844-853) or cloning a fusion protein (Nagahara et al., (1998) Nat. Med. 4:1449-1453). Non-covalent coupling between the cargo and short amphipathic CPPs comprising polar and non-polar domains is established through electrostatic and hydrophobic interactions.

CPPs have been utilized in the art to deliver potentially therapeutic biomolecules into cells. Examples include cyclosporine linked to polyarginine for immunosuppression (Rothbard et al., (2000) Nature Medicine 6(11): 1253-1257), siRNA against cyclin B1 linked to a CPP called MPG for inhibiting tumorigenesis (Crombez et al., (2007) Biochem Soc. Trans. 35:44-46), tumor suppressor p53 peptides linked to CPPs to reduce cancer cell growth (Takenobu et al., (2002) Mol. Cancer Ther. 1(12):1043-1049, Snyder et al., (2004) PLoS Biol. 2:E36), and dominant negative forms of Ras or phosphoinositol 3 kinase (PI3K) fused to Tat to treat asthma (Myou et al., (2003) J. Immunol. 171:4399-4405).

CPPs have been utilized in the art to transport contrast agents into cells for imaging and biosensing applications. For example, green fluorescent protein (GFP) attached to Tat has been used to label cancer cells (Shokolenko et al., (2005) DNA Repair 4(4):511-518). Tat conjugated to quantum dots have been used to successfully cross the blood-brain barrier for visualization of the rat brain (Santra et al., (2005) Chem. Commun. 3144-3146). CPPs have also been combined with magnetic resonance imaging techniques for cell imaging (Liu et al., (2006) Biochem. and Biophys. Res. Comm. 347(1):133-140). See also Ramsey and Flynn, Pharmacol Ther. 2015 Jul. 22. pii: S0163-7258(15)00141-2.

Alternatively or in addition, the fusion proteins can include a nuclear localization sequence, e.g., SV40 large T antigen NLS (PKKKRRV (SEQ ID NO: 10)) and nucleoplasmin NLS (KRPAATKKAGQAKKKK (SEQ ID NO: 11)). Other NLSs are known in the art; see, e.g., Cokol et al., EMBO Rep. 2000 Nov. 15; 1(5): 411-415; Freitas and Cunha, Curr Genomics. 2009 December; 10(8): 550-557.

In some embodiments, the fusion proteins include a moiety that has a high affinity for a ligand, for example GST, FLAG or hexahistidine sequences. Such affinity tags can facilitate the purification of recombinant fusion proteins.

For methods in which the fusion proteins are delivered to cells, the proteins can be produced using any method known in the art, e.g., by in vitro translation, or expression in a suitable host cell from nucleic acid encoding the fusion protein; a number of methods are known in the art for producing proteins. For example, the proteins can be produced in and purified from yeast, E. coli, insect cell lines, plants, transgenic animals, or cultured mammalian cells; see, e.g., Palomares et al., “Production of Recombinant Proteins: Challenges and Solutions,” Methods Mol Biol. 2004; 267:15-52. In addition, the fusion proteins can be linked to a moiety that facilitates transfer into a cell, e.g., a lipid nanoparticle, optionally with a linker that is cleaved once the protein is inside the cell. See, e.g., LaFountaine et al., Int J Pharm. 2015 Aug. 13; 494(1):180-194.

Expression Systems

To use the fusion proteins described herein, it may be desirable to express them from a nucleic acid that encodes them. This can be performed in a variety of ways. For example, the nucleic acid encoding the fusion can be cloned into an intermediate vector for transformation into prokaryotic or eukaryotic cells for replication and/or expression. Intermediate vectors are typically prokaryote vectors, e.g., plasmids, or shuttle vectors, or insect vectors, for storage or manipulation of the nucleic acid encoding the fusion for production of the fusion protein. The nucleic acid encoding the fusion protein can also be cloned into an expression vector, for administration to a plant cell, animal cell, preferably a mammalian cell or a human cell, fungal cell, bacterial cell, or protozoan cell.

To obtain expression, a sequence encoding a fusion protein is typically subcloned into an expression vector that contains a promoter to direct transcription. Suitable bacterial and eukaryotic promoters are well known in the art and described, e.g., in Sambrook et al., Molecular Cloning, A Laboratory Manual (3d ed. 2001); Kriegler, Gene Transfer and Expression: A Laboratory Manual (1990); and Current Protocols in Molecular Biology (Ausubel et al., eds., 2010). Bacterial expression systems for expressing the engineered protein are available in, e.g., E. coli, Bacillus sp., and Salmonella (Palva et al., 1983, Gene 22:229-235). Kits for such expression systems are commercially available. Eukaryotic expression systems for mammalian cells, yeast, and insect cells are well known in the art and are also commercially available.

The promoter used to direct expression of a nucleic acid depends on the particular application. For example, a strong constitutive promoter is typically used for expression and purification of fusion proteins. In contrast, when the fusion protein is to be administered in vivo for gene regulation, either a constitutive or an inducible promoter can be used, depending on the particular use of the fusion protein. In addition, a preferred promoter for administration of the fusion protein can be a weak promoter, such as HSV TK or a promoter having similar activity. The promoter can also include elements that are responsive to transactivation, e.g., hypoxia response elements, Gal4 response elements, lac repressor response element, and small molecule control systems such as tetracycline-regulated systems and the RU-486 system (see, e.g., Gossen & Bujard, 1992, Proc. Natl. Acad. Sci. USA, 89:5547; Oligino et al., 1998, Gene Ther., 5:491-496; Wang et al., 1997, Gene Ther., 4:432-441; Neering et al., 1996, Blood, 88:1147-55; and Rendahl et al., 1998, Nat. Biotechnol., 16:757-761).

In addition to the promoter, the expression vector typically contains a transcription unit or expression cassette that contains all the additional elements required for the expression of the nucleic acid in host cells, either prokaryotic or eukaryotic. A typical expression cassette thus contains a promoter operably linked, e.g., to the nucleic acid sequence encoding the fusion protein, and any signals required, e.g., for efficient polyadenylation of the transcript, transcriptional termination, ribosome binding sites, or translation termination. Additional elements of the cassette may include, e.g., enhancers, and heterologous spliced intronic signals.

The particular expression vector used to transport the genetic information into the cell is selected with regard to the intended use of the fusion protein, e.g., expression in plants, animals, bacteria, fungus, protozoa, etc. Standard bacterial expression vectors include plasmids such as pBR322 based plasmids, pSKF, pET23D, and commercially available tag-fusion expression systems such as GST and LacZ.

Expression vectors containing regulatory elements from eukaryotic viruses are often used in eukaryotic expression vectors, e.g., SV40 vectors, papilloma virus vectors, and vectors derived from Epstein-Barr virus. Other exemplary eukaryotic vectors include pMSG pAV009/A+, pMTO10/A+, pMAMneo-5, baculovirus pDSVE, and any other vector allowing expression of proteins under the direction of the SV40 early promoter, SV40 late promoter, metallothionein promoter, murine mammary tumor virus promoter, Rous sarcoma virus promoter, polyhedrin promoter, or other promoters shown effective for expression in eukaryotic cells.

The vectors for expressing the fusion protein can include RNA Pol III promoters to drive expression of the guide RNAs, e.g., the HI, U6 or 7SK promoters. These human promoters allow for expression of fusion protein in mammalian cells following plasmid transfection.

Some expression systems have markers for selection of stably transfected cell lines such as thymidine kinase, hygromycin B phosphotransferase, and dihydrofolate reductase. High yield expression systems are also suitable, such as using a baculovirus vector in insect cells, with the gRNA encoding sequence under the direction of the polyhedrin promoter or other strong baculovirus promoters.

The elements that are typically included in expression vectors also include a replicon that functions in E. coli, a gene encoding antibiotic resistance to permit selection of bacteria that harbor recombinant plasmids, and unique restriction sites in nonessential regions of the plasmid to allow insertion of recombinant sequences.

Standard transfection methods are used to produce bacterial, mammalian, yeast or insect cell lines that express large quantities of protein, which are then purified using standard techniques (see, e.g., Colley et al., 1989, J. Biol. Chem., 264:17619-22; Guide to Protein Purification, in Methods in Enzymology, vol. 182 (Deutscher, ed., 1990)). Transformation of eukaryotic and prokaryotic cells are performed according to standard techniques (see, e.g., Morrison, 1977, J. Bacteriol. 132:349-351; Clark-Curtiss & Curtiss, Methods in Enzymology 101:347-362 (Wu et al., eds, 1983).

Any of the known procedures for introducing foreign nucleotide sequences into host cells may be used. These include the use of calcium phosphate transfection, polybrene, protoplast fusion, electroporation, nucleofection, liposomes, microinjection, naked DNA, plasmid vectors, viral vectors, both episomal and integrative, and any of the other well-known methods for introducing cloned genomic DNA, cDNA, synthetic DNA or other foreign genetic material into a host cell (see, e.g., Sambrook et al., supra). It is only necessary that the particular genetic engineering procedure used be capable of successfully introducing at least one gene into the host cell capable of expressing the fusion protein.

Alternatively, the methods can include delivering the fusion protein and guide RNA together, e.g., as a complex. For example, the fusion protein and gRNA can be can be overexpressed in a host cell and purified, then complexed with the guide RNA (e.g., in a test tube) to form a ribonucleoprotein (RNP), and delivered to cells. In some embodiments, the fusion protein can be expressed in and purified from bacteria through the use of bacterial expression plasmids. For example, His-tagged fusion protein can be expressed in bacterial cells and then purified using nickel affinity chromatography. The use of RNPs circumvents the necessity of delivering plasmid DNAs encoding the nuclease or the guide, or encoding the nuclease as an mRNA. RNP delivery may also improve specificity, presumably because the half-life of the RNP is shorter and there's no persistent expression of the nuclease and guide (as you'd get from a plasmid). The RNPs can be delivered to the cells in vivo or in vitro, e.g., using lipid-mediated transfection or electroporation. See, e.g., Liang et al. “Rapid and highly efficient mammalian cell engineering via Cas9 protein transfection.” Journal of biotechnology 208 (2015): 44-53; Zuris, John A., et al. “Cationic lipid-mediated delivery of proteins enables efficient protein-based genome editing in vitro and in vivo.” Nature biotechnology 33.1 (2015): 73-80; Kim et al. “Highly efficient RNA-guided genome editing in human cells via delivery of purified Cas9 ribonucleoproteins.” Genome research 24.6 (2014): 1012-1019.

The present invention also includes the vectors and cells comprising the vectors, as well as kits comprising the proteins and nucleic acids described herein, e.g., for use in a method described herein.

Examples

The invention is further described in the following examples, which do not limit the scope of the invention described in the claims.

Materials and Methods

The following materials and methods were used in the examples herein.

Molecular Cloning

Expression plasmids were constructed by selectively amplifying desired DNA sequences using the PCR method such that they had significant overlapping ends and using isothermal assembly (or “Gibson Assembly”, NEB) to assemble them in the desired order in a pCAG expression vector. PCR was conducted using Q5 or Phusion polymerases (NEB). Cas9 gRNAs were cloned using the pUC 19-based entry vector BPK 1520 (via BsmBI cassette) under control of a U6 promoter.

Guide RNAs and Zinc Finger Target Sites

The following EGFP targeting zinc fingers were used:

ZF 11-1 (223L), target site (SEQ ID NO: 12) gGTCGGGGTAg ZF 13-6 (292L), target site (SEQ ID NO: 13) aGAAGATGGTg Cas9 guide RNA2 protospacer (PAM): (SEQ ID NO: 14) AGCACTGCACGCCGTAGGTC(AGG) Cas9 guide RNA4 protospacer (PAM): (SEQ ID NO: 15) CATGCCCGAAGGCTACGTCC(AGG) Cas9 guide RNA5 protospacer (PAM): (SEQ ID NO: 16) CTGGACGTAGCCTTCGGGCA(TGG)

Cell Culture and Transfections

For the BBE experiments, a HEK293 cell line in which an integrated EGFP reporter gene has been integrated (unpublished) was grown in culture using media consisting of Advanced Dulbeccos Modified Medium (Gibco) supplemented with 10% FBS (Gibco), 1% 10,000 U/ml penicillin-streptomycin solution (Gibco), and 1% Glutamax (Gibco). Cells were passaged every 3-4 days to maintain an actively growing population and avoid anoxic conditions. Transfections containing 800 nanogram of transfection quality DNA (Qiagen Maxi- or Miniprep) encoding desired proteins/gRNAs were conducted by seeding 1×105 cells in 24-well TC-treated plates and using TransIT-293 reagent according to manufacturer's protocol (Mirus Bio). gDNA was harvested 3 days post-transfection using the DNAdvance kit (Agencourt). U2OS.EGFP cells containing a single stably integrated copy of the EGFP-PEST reporter gene and HEK293T cells were cultured in DMEM supplemented with 10% heat-inactivated fetal bovine serum, 2 mM GlutaMax, penicillin and streptomycin at 37° C. with 5% C02.

The media for U2OS.EGFP cells was supplemented with 400 μg ml−1 Geneticin. Cell line identity was validated by STR profiling (ATCC), and cells were tested regularly for mycoplasma contamination. U2OS.EGFP cells were transfected with 750 ng of plasmid expressing BE and 250 ng of plasmid expressing gRNA according to the manufacturer's recommendations using the DN-100 program and SE cell line kit on a Lonza 4-D Nucleofector. Disruption of the EGFP gene was evaluated using flow cytometry 48 hours post-transfection.

High-Throughput Amplicon Sequencing

Target site genomic DNA was amplified using gene-specific DNA primers flanking desired target sequence. Illumina TruSeq adapters were added to the ends of the amplicons either by PCR or NEBNext Ultra II kit (NEB) and molecularly indexed with NEBNext Dual Index Primers (NEB). Samples were combined into libraries and sequenced on the Illumina MiSeq machine using the MiSeq Reagent Micro Kit v2 (Illumina). Sequencing results were analyzed using a batch version of the software CRISPResso (crispresso.rocks).

Relevant Protein Sequences

N-nCas9(D10A)-SGGS linker-UGI-SV40NLS-C (SEQ ID NO: 17) MDKKYSIGLAIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIKKNLIGALLFDSGETAEATRLKRTARRRYTRRKNRICY LQEIFSNEMAKVDDSFFHRLEESFLVEEDKKHERHPIFGNIVDEVAYHEKYPTIYHLRKKLVDSTDKADLRLIYLALAHMI KFRGHFLIEGDLNPDNSDVDKLFIQLVQTYNQLFEENPINASGVDAKAILSARLSKSRRLENLIAQLPGEKKNGLFGNLIA LSLGLTPNFKSNFDLAEDAKLQLSKDTYDDDLDNLLAQIGDQYADLFLAAKNLSDAILLSDILRVNTEITKAPLSASMIKR YDEHHQDLTLLKALVRQQLPEKYKEIFFDQSKNGYAGYIDGGASQEEFYKFIKPILEKMDGTEELLVKLNREDLLRKQRTF DNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKILTFRIPYYVGPLARGNSRFAWMTRKSEETITPWNFEEVVDKGA SAQSFIERMTNFDKNLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRKPAFLSGEQKKAIVDLLFKTNRKVTVKQLKED YFKKIECFDSVEISGVEDRFNASLGTYHDLLKIIKDKDFLDNEENEDILEDIVLTLTLFEDREMIEERLKTYAHLFDDKVM KQLKRRRYTGWGRLSRKLINGIRDKQSGKTILDFLKSDGFANRNFMQLIHDDSLTFKEDIQKAQVSGQGDSLHEHIANLAG SPAIKKGILQTVKVVDELVKVMGRHKPENIVIEMARENQTTQKGQKNSRERMKRIEEGIKELGSQILKEHPVENTQLQNEK LYLYYLQNGRDMYVDQELDINRLSDYDVDHIVPQSFLKDDSIDNKVLTRSDKNRGKSDNVPSEEVVKKMKNYWRQLLNAKL ITQRKFDNLTKAERGGLSELDKAGFIKRQLVETRQITKHVAQILDSRMNTKYDENDKLIREVKVITLKSKLVSDFRKDFQF YKVREINNYHHAHDAYLNAVVGTALIKKYPKLESEFVYGDYKVYDVRKMIAKSEQEIGKATAKYFFYSNIMNFFKTEITLA NGEIRKRPLIETNGETGEIVWDKGRDFATVRKVLSMPQVNIVKKTEVQTGGFSKESILPKRNSDKLIARKKDWDPKKYGGF DSPTVAYSVLVVAKVEKGKSKKLKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKYSLFELENGRKRMLA SAGELQKGNELALPSKYVNFLYLASHYEKLKGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANLDKVLSAYNK HRDKPIREQAENIIHLFTLTNLGAPAAFKYFDTTIDRKRYTSTKEVLDATLIHQSITGLYETRIDLSQLGGDSGGSTNLSD IIEKETGKQLVIQESILMLPEEVEEVIGNKPESDILVHTAYDESTDENVMLLTSDAPEYKPWALVIQDSNGENKIKMLSGG SPKKKRKV N-hAPOBEC3A-8AA linker-ZF-LRGS linker-UGI-SGGSlinker-SV40NLS-C (SEQ ID NO: 18) MEASPASGPRHLMDPHIFTSNFNNGIGREIKTYLCYEVERLDNGTSVKMDQHRGFLHNQAKNLLCGFYGRHAELRFLDLVP SLQLDPAQIYRVTWFISWSPCFSWGCAGEVRAFLQENTHVRLRIFAARIYDYDPLYKEALQMLRDAGAQVSIMTYDEFKHC WDTFVDHQGCPFQPWDGLDEHSQALSGRLRAILQNQGNSGGGLGSTSRPGERPFQCRICMRNFSXXXXLXXHTRTHTGEKP FQCRICMRNFSXXXXLXXHLRTHTGEKPFQCRICMRNFSXXXXLXXHLKTHLRGSTNLSDIIEKETGKQLVIQESILMLPE EVEEVIGNKPESDILVHTAYDESTDENVMLLTSDAPEYKPWALVIQDSNGENKIKMLSGGSPKKKRKV N-rAPOBEC1-XTEN-8AA linker-ZF-LRGS linker-UGI-SGGS linker-SV40NLS-C (SEQ ID NO: 19) MSSETGPVAVDPTLRRRIEPHEFEVFFDPRELRKETCLLYEINWGGRHSIWRHTSQNTNKHVEVNFIEKFTTERYFCPNTR CSITWFLSWSPCGECSRAITEFLSRYPHVTLFIYIARLYHHADPRNRQGLRDLISSGVTIQIMTEQESGYCWRNFVNYSPS NEAHWPRYPHLWVRLYVLELYCIILGLPPCLNILRRKQPQLTFFTIALQSCHYQRLPPHILWATGLKSGSETPGTSESATP ESSGGGLGSTSRPGERPFQCRICMRNFSXXXXLXXHTRTHTGEKPFQCRICMRNFSXXXXLXXHLRTHTGEKPFQCRICMF SXXXXLXXHLKTHLRGSTNLSDIIEKETGKQLVIQESILMLPEEVEEVIGNKPESDILVHTAYDESTDENVMLLTSDAPEY KPWALVIQDSNGENKIKMLSGGSPKKKRKV S. aureus Cas9 (SEQ ID NO: 20) MKRNYILGLDIGITSVGYGIIDYETRDVIDAGVRLFKEANVENNEGRRSKRGARRLKRRRRHRIQRVKKLLFDYNLLTDHS ELSGINPYEARVKGLSQKLSEEEFSAALLHLAKRRGVHNVNEVEEDTGNELSTKEQISRNSKALEEKYVAELQLERLKKDG EVRGSINRFKTSDYVKEAKQLLKVQKAYHQLDQSFIDTYIDLLETRRTYYEGPGEGSPFGWKDIKEWYEMLMGHCTYFPEE LRSVKYAYNADLYNALNDLNNLVITRDENEKLEYYEKFQIIENVFKQKKKPTLKQIAKEILVNEEDIKGYRVTSTGKPEFT NLKVYHDIKDITARKEIIENAELLDQIAKILTIYQSSEDIQEELTNLNSELTQEEIEQISNLKGYTGTHNLSLKAINLILD ELWHTNDNQIAIFNRLKLVPKKVDLSQQKEIPTTLVDDFILSPVVKRSFIQSIKVINAIIKKYGLPNDIIIELAREKNSKD AQKMINEMQKRNRQTNERIEEIIRTTGKENAKYLIEKIKLHDMQEGKCLYSLEAIPLEDLLNNPFNYEVDHIIPRSVSFDN SFNNKVLVKQEENSKKGNRTPFQYLSSSDSKISYETFKKHILNLAKGKGRISKTKKEYLLEERDINRFSVQKDFINRNLVD TRYATRGLMNLLRSYFRVNNLDVKVKSINGGFTSFLRRKWKFKKERNKGYKHHAEDALIIANADFIFKEWKKLDKAKKVME NQMFEEKQAESMPEIETEQEYKEIFITPHQIKHIKDFKDYKYSHRVDKKPNRELINDTLYSTRKDDKGNTLIVNNLNGLYD KDNDKLKKLINKSPEKLLMYHHDPQTYQKLKLIMEQYGDEKNPLYKYYEETGNYLTKYSKKDNGPVIKKIKYYGNKLNAHL DITDDYPNSRNKVVKLSLKPYRFDVYLDNGVYKFVTVKNLDVIKKENYYEVNSKCYEEAKKLKKISNQAEFIASFYNNDLI KINGELYRVIGVNNDLLNRIEVNMIDITYREYLENMNDKRPPRIIKTIASKTQSIKKYSTDILGNLYEVKSKKHPQIIKKG C. jejuni Cas9 (SEQ ID NO: 21) MARILAFDIGISSIGWAFSENDELKDCGVRIFTKVENPKTGESLALPRRLARSARKRLARRKARLNHLKHLIANEFKLNYE DYQSFDESLAKAYKGSLISPYELRFRALNELLSKQDFARVILHIAKRRGYDDIKNSDDKEKGAILKAIKQNEEKLANYQSV GEYLYKEYFQKFKENSKEFTNVRNKKESYERCIAQSFLKDELKLIFKKQREFGFSFSKKFEEEVLSVAFYKRALKDFSHLV GNCSFFTDEKRAPKNSPLAFMFVALTRIINLLNNLKNTEGILYTKDDLNALLNEVLKNGTLTYKQTKKLLGLSDDYEFKGE KGTYFIEFKKYKEFIKALGEHNLSQDDLNEIAKDITLIKDEIKLKKALAKYDLNQNQIDSLSKLEFKDHLNISFKALKLVT PLMLEGKKYDEACNELNLKVAINEDKKDFLPAFNETYYKDEVTNPVVLRAIKEYRKVLNALLKKYGKVHKINIELAREVGK NHSQRAKIEKEQNENYKAKKDAELECEKLGLKINSKNILKLRLFKEQKEFCAYSGEKIKISDLQDEKMLEIDHIYPYSRSF DDSYMNKVLVFTKQNQEKLNQTPFEAFGNDSAKWQKIEVLAKNLPTKKQKRILDKNYKDKEQKNFKDRNLNDTRYIARLVL NYTKDYLDFLPLSDDENTKLNDTQKGSKVHVEAKSGMLTSALRHTWGFSAKDRNNHLHHAIDAVIIAYANNSIVKAFSDFK KEQESNSAELYAKKISELDYKNKRKFFEPFSGFRQKVLDKIDEIFVSKPERKKPSGALHEETFRKEEEFYQSYGGKEGVLK ALELGKIRKVNGKIVKNGDMFRVDIFKHKKTNKFYAVPIYTMDFALKVLPNKAVARSKKGEIKDWILMDENYEFCFSLYKD SLILIQTKDMQEPEFVYYNAFTSSTVSLIVSKHDNKFETLSKNQKILFKNANEKEVIAKSIGIQNLKVFEKYIVSALGEVT KAEFRQREDFKK P. lavamentivorans Cas9 (SEQ ID NO: 22) MERIFGFDIGTTSIGFSVIDYSSTQSAGNIQRLGVRIFPEARDPDGTPLNQQRRQKRMMRRQLRRRRIRRKALNETLHEAG FLPAYGSADWPVVMADEPYELRRRGLEEGLSAYEFGRAIYHLAQHRHFKGRELEESDTPDPDVDDEKEAANERAATLKALK NEQTTLGAWLARRPPSDRKRGIHAHRNVVAEEFERLWEVQSKFHPALKSEEMRARISDTIFAQRPVFWRKNTLGECRFMPG EPLCPKGSWLSQQRRMLEKLNNLAIAGGNARPLDAEERDAILSKLQQQASMSWPGVRSALKALYKQRGEPGAEKSLKFNLE LGGESKLLGNALEAKLADMFGPDWPAHPRKQEIRHAVHERLWAADYGETPDKKRVIILSEKDRKAHREAAANSFVADFGIT GEQAAQLQALKLPTGWEPYSIPALNLFLAELEKGERFGALVNGPDWEGWRRTNFPHRNQPTGEILDKLPSPASKEERERIS QLRNPTVVRTQNELRKVVNNLIGLYGKPDRIRIEVGRDVGKSKREREEIQSGIRRNEKQRKKATEDLIKNGIANPSRDDVE KWILWKEGQERCPYTGDQIGFNALFREGRYEVEHIWPRSRSFDNSPRNKTLCRKDVNIEKGNRMPFEAFGHDEDRWSAIQI RLQGMVSAKGGTGMSPGKVKRFLAKTMPEDFAARQLNDTRYAAKQILAQLKRLWPDMGPEAPVKVEAVTGQVTAQLRKLWT LNNILADDGEKTRADHRHHAIDALTVACTHPGMTNKLSRYWQLRDDPRAEKPALTPPWDTIRADAEKAVSEIVVSHRVRKK VSGPLHKETTYGDTGTDIKTKSGTYRQFVTRKKIESLSKGELDEIRDPRIKEIVAAHVAGRGGDPKKAFPPYPCVSPGGPE IRKVRLTSKQQLNLMAQTGNGYADLGSNHHIAIYRLPDGKADFEIVSLFDASRRLAQRNPIVQRTRADGASFVMSLAAGEA IMIPEGSKKGIWIVQGVWASGQVVLERDTDADHSTTTRPMPNPILKDDAKKVSIDPIGRVRPSND N. cinerea Cas9 (SEQ ID NO: 23) MAAFKPNPMNYILGLDIGIASVGWAIVEIDEEENPIRLIDLGVRVFERAEVPKTGDSLAAARRLARSVRRLTRRRAHRLLR ARRLLKREGVLQAADFDENGLIKSLPNTPWQLRAAALDRKLTPLEWSAVLLHLIKHRGYLSQRKNEGETADKELGALLKGV ADNTHALQTGDFRTPAELALNKFEKESGHIRNQRGDYSHTFNRKDLQAELNLLFEKQKEFGNPHVSDGLKEGIETLLMTQR PALSGDAVQKMLGHCTFEPTEPKAAKNTYTAERFVWLTKLNNLRILEQGSERPLTDTERATLMDEPYRKSKLTYAQARKLL DLDDTAFFKGLRYGKDNAEASTLMEMKAYHAISRALEKEGLKDKKSPLNLSPELQDEIGTAFSLFKTDEDITGRLKDRVQP EILEALLKHISFDKFVQISLKALRRIVPLMEQGNRYDEACTEIYGDHYGKKNTEEKIYLPPIPADEIRNPVVLRALSQARK VINGVVRRYGSPARIHIETAREVGKSFKDRKEIEKRQEENRKDREKSAAKFREYFPNFVGEPKSKDILKLRLYEQQHGKCL YSGKEINLGRLNEKGYVEIDHALPFSRTWDDSFNNKVLALGSENQNKGNQTPYEYFNGKDNSREWQEFKARVETSRFPRSK KQRILLQKFDEDGFKERNLNDTRYINRFLCQFVADHMLLTGKGKRRVFASNGQITNLLRGFWGLRKVRAENDRHHALDAVV VACSTIAMQQKITRFVRYKEMNAFDGKTIDKETGEVLHQKAHFPQPWEFFAQEVMIRVFGKPDGKPEFEEADTPEKLRTLL AEKLSSRPEAVHKYVTPLFISRAPNRKMSGQGHMETVKSAKRLDEGISVLRVPLTQLKLKDLEKMVNREREPKLYEALKAR LEAHKDDPAKAFAEPFYKYDKAGNRTQQVKAVRVEQVQKTGVWVHNHNGIADNATIVRVDVFEKGGKYYLVPIYSWQVAKG ILPDRAVVQGKDEEDWTVMDDSFEFKFVLYANDLIKLTAKKNEFLGYFVSLNRATGAIDIRTHDTDSTKGKNGIFQSVGVK TALSFQKYQIDELGKEIRPCRLKKRPPVR hAID (SEQ ID NO: 24) MDSLLMNRRKFLYQFKNVRWAKGRRETYLCYVVKRRDSATSFSLDFGYLRNKNGCHVELLFLRYISDWDLDPGRCYRVTWF TSWSPCYDCARHVADFLRGNPNLSLRIFTARLYFCEDRKAEPEGLRRLHRAGVQIAIMTFKDYFYCWNTFVENHERTFKAW EGLHENSVRLSRQLRRILLPLYEVDDLRDAFRTLGL hAIDv solubility variant lacking N-terminal RNA-binding region (SEQ ID NO: 25) MDPHIFTSNFNNGIGRHKTYLCYEVERLDSATSFSLDFGYLRNKNGCHVELLFLRYISDWDLDPGRCYRVTWFTSWSPCYD CARHVADFLRGNPNLSLRIFTARLYFCEDRKAEPEGLRRLHRAGVQIAIMTFKDYFYCWNTFVENHERTFKAWEGLHENSV RLSRQLRRILLPLYEVDDLRDAFRTLGL hAIDv solubility variant lacking N-terminal RNA-binding region and the C-terminal poorly structured region (SEQ ID NO: 26) MDPHIFTSNFNNGIGRHKTYLCYEVERLDSATSFSLDFGYLRNKNGCHVELLFLRYISDWDLDPGRCYRVTWFTSWSPCYD CARHVADFLRGNPNLSLRIFTARLYFCEDRKAEPEGLRRLHRAGVQIAIMTFKDYFYCWNTFVENHERTFKAWEGLHENSV RLSRQLRRILLPL rAPOBEC1 (SEQ ID NO: 27) MSSETGPVAVDPTLRRRIEPHEFEVFFDPRELRKETCLLYEINWGGRHSIWRHTSQNTNKHVEVNFIEKFTTERYFCPNTR CSITWFLSWSPCGECSRAITEFLSRYPHVTLFIYIARLYHHADPRNRQGLRDLISSGVTIQIMTEQESGYCWRNFVNYSPS NEAHWPRYPHLWVRLYVLELYCIILGLPPCLNILRRKQPQLTFFTIALQSCHYQRLPPHILWATGLK mAPOBEC3 (SEQ ID NO: 28) MGPFCLGCSHRKCYSPIRNLISQETFKFHFKNLGYAKGRKDTFLCYEVTRKDCDSPVSLHHGVFKNKDNIHAEICFLYWFH DKVLKVLSPREEFKITWYMSWSPCFECAEQIVRFLATHHNLSLDIFSSRLYNVQDPETQQNLCRLVQEGAQVAAMDLYEFK KCWKKFVDNGGRRFRPWKRLLTNFRYQDSKLQEILRRMDPLSEEEFYSQFYNQRVKHLCYYHRMKPYLCYQLEQFNGQAPL KGCLLSEKGKQHAEILFLDKIRSMELSQVTITCYLTWSPCPNCAWQLAAFKRDRPDLILHIYTSRLYFHWKRPFQKGLCSL WQSGILVDVMDLPQFTDCWTNFVNPKRPFRPWKGLEIISRRTQRRLRRIKESWGLQDLVNDFGNLQLGPPMSN mAPOBEC3 catalytic domain (SEQ ID NO: 29) MGPFCLGCSHRKCYSPIRNLISQETFKFHFKNLGYAKGRKDTFLCYEVTRKDCDSPVSLHHGVFKNKDNIHAEICFLYWFH DKVLKVLSPREEFKITWYMSWSPCFECAEQIVRFLATHHNLSLDIFSSRLYNVQDPETQQNLCRLVQEGAQVAAMDLYEFK KCWKKFVDNGGRRFRPWKRLLTNFRYQDSKLQEILRR hAPOBEC3A (SEQ ID NO: 30) MEASPASGPRHLMDPHIFTSNFNNGIGREIKTYLCYEVERLDNGTSVKMDQHRGFLHNQAKNLLCGFYGRHAELRFLDLVP SLQLDPAQIYRVTWFISWSPCFSWGCAGEVRAFLQENTHVRLRIFAARIYDYDPLYKEALQMLRDAGAQVSIMTYDEFKHC WDTFVDHQGCPFQPWDGLDEHSQALSGRLRAILQNQGN hAPOBEC3G (SEQ ID NO: 31) MKPHFRNTVERMYRDTFSYNFYNRPILSRRNTVWLCYEVKTKGPSRPPLDAKIFRGQVYSELKYHPEMRFFHWFSKWRKLH RDQEYEVTWYISWSPCTKCTRDMATFLAEDPKVTLTIFVARLYYFWDPDYQEALRSLCQKRDGPRATMKIMNYDEFQHCWS KFVYSQRELFEPWNNLPKYYILLHIMLGEILRHSMDPPTFTFNFNNEPWVRGRHETYLCYEVERMHNDTWVLLNQRRGFLC NQAPHKHGFLEGRHAELCFLDVIPFWKLDLDQDYRVTCFTSWSPCFSCAQEMAKFISKNKHVSLCIFTARIYDDQGRCQEG LRTLAEAGAKISIMTYSEFKHCWDTFVDHQGCPFQPWDGLDEHSQDLSGRLRAILQNQEN hAPOBEC3G catalytic domain (SEQ ID NO: 32) PPTFTFNFNNEPWVRGRHETYLCYEVERMHNDTWVLLNQRRGFLCNQAPHKHGFLEGRHAELCFLDVIPFWKLDLDQDYRV TCFTSWSPCFSCAQEMAKFISKNKHVSLCIFTARIYDDQGRCQEGLRTLAEAGAKISIMTYSEFKHCWDTFVDHQGCPFQP WDGLDEHSQDLSGRLRAILQNQEN hAPOBEC3H (SEQ ID NO: 33) MALLTAETFRLQFNNKRRLRRPYYPRKALLCYQLTPQNGSTPTRGYFENKKKCHAEICFINEIKSMGLDETQCYQVTCYLT WSPCSSCAWELVDFIKAHDHLNLGIFASRLYYHWCKPQQKGLRLLCGSQVPVEVMGFPKFADCWENFVDHEKPLSFNPYKM LEELDKNSRAIKRRLERIKIPGVRAQGRYMDILCDAEV hAPOBEC3F (SEQ ID NO: 34) MKPHFRNTVERMYRDTFSYNFYNRPILSRRNTVWLCYEVKTKGPSRPRLDAKIFRGQVYSQPEHHAEMCFLSWFCGNQLPA YKCFQITWFVSWTPCPDCVAKLAEFLAEHPNVTLTISAARLYYYWERDYRRALCRLSQAGARVKIMDDEEFAYCWENFVYS EGQPFMPWYKFDDNYAFLHRTLKEILRNPMEAMYPHIFYFHFKNLRKAYGRNESWLCFTMEVVKHHSPVSWKRGVFRNQVD PETHCHAERCFLSWFCDDILSPNTNYEVTWYTSWSPCPECAGEVAEFLARHSNVNLTIFTARLYYFWDTDYQEGLRSLSQE GASVEIMGYKDFKYCWENFVYNDDEPFKPWKGLKYNFLFLDSKLQEILE hAPOBEC3F catalytic domain (SEQ ID NO: 35) KEILRNPMEAMYPHIFYFHFKNLRKAYGRNESWLCFTMEVVKHHSPVSWKRGVFRNQVDPETHCHAERCFLSWFCDDILSP NTNYEVTWYTSWSPCPECAGEVAEFLARHSNVNLTIFTARLYYFWDTDYQEGLRSLSQEGASVEIMGYKDFKYCWENFVYN DDEPFKPWKGLKYNFLFLDSKLQEILE C. lari Cas9 (SEQ ID NO: 36) MRILGFDIGINSIGWAFVENDELKDCGVRIFTKAENPKNKESLALPRRNARSSRRRLKRRKARLIAIKRILAKELKLNYKD YVAADGELPKAYEGSLASVYELRYKALTQNLETKDLARVILHIAKHRGYMNKNEKKSNDAKKGKILSALKNNALKLENYQS VGEYFYKEFFQKYKKNTKNFIKIRNTKDNYNNCVLSSDLEKELKLILEKQKEFGYNYSEDFINEILKVAFFQRPLKDFSHL VGACTFFEEEKRACKNSYSAWEFVALTKIINEIKSLEKISGEIVPTQTINEVLNLILDKGSITYKKFRSCINLHESISFKS LKYDKENAENAKLIDFRKLVEFKKALGVHSLSRQELDQISTHITLIKDNVKLKTVLEKYNLSNEQINNLLEIEFNDYINLS FKALGMILPLMREGKRYDEACEIANLKPKTVDEKKDFLPAFCDSIFAHELSNPVVNRAISEYRKVLNALLKKYGKVHKIHL ELARDVGLSKKAREKIEKEQKENQAVNAWALKECENIGLKASAKNILKLKLWKEQKEICIYSGNKISIEHLKDEKALEVDH IYPYSRSFDDSFINKVLVFTKENQEKLNKTPFEAFGKNIEKWSKIQTLAQNLPYKKKNKILDENFKDKQQEDFISRNLNDT RYIATLIAKYTKEYLNFLLLSENENANLKSGEKGSKIHVQTISGMLTSVLRHTWGFDKKDRNNHLHHALDAIIVAYSTNSI IKAFSDFRKNQELLKARFYAKELTSDNYKHQVKFFEPFKSFREKILSKIDEIFVSKPPRKRARRALHKDTFHSENKIIDKC SYNSKEGLQIALSCGRVRKIGTKYVENDTIVRVDIFKKQNKFYAIPIYAMDFALGILPNKIVITGKDKNNNPKQWQTIDES YEFCFSLYKNDLILLQKKNMQEPEFAYYNDFSISTSSICVEKHDNKFENLTSNQKLLFSNAKEGSVKVESLGIQNLKVFEK YIITPLGDKIKADFQPRENISLKTSKKYGLR

Example 1

We generated BBEs consisting of either a rAPOBEC1(WT)-ZF or hAPOBEC3A(WT)-ZF fusion and an nCas9-Uracil DNA Glycosylase inhibitor (UGI) targeted to adjacent sequences. An nCas9-UGI architecture mimics the standard BE3 architecture that is known to be an important part of a functional BE protein. As there is evidence for the use of a second UGI to enhance product purity we fused a UGI to both the ZF (via LRGS linker) as well as the nCas9 (same linker as in conventional BE3) C-terminus17.

In addition, the physical separation of the deaminase domain from nCas9 and fusion of UGI to both components of the bipartite base editor seemed to yield lower synergistic on-target side effects of conventional base editors such as insertions/deletion (indels) or unwanted substitutions (cytosine to purine, e.g., C-to-G or C-to-A), commonly referred to as “product impurities.” This property may arise for a stronger inhibition of the UDG enzyme by UGI, which is known to aid in product purity of BEs.

When targeted to adjacent sites 6-8 basepairs apart from each other on an integrated EGFP reporter gene in HEK293 cells, both rAPOBEC1 and hAPOBEC3A BBEs are able to stimulate efficient base-editing at rates similar to or greater than monomeric BE counterparts (of the form deaminase-nCas9-UGI). (FIGS. 4-6). This effect is consistent regardless of the orientation of the two components with respect to each other and works using multiple different ZFs or gRNAs. When the ZF-deaminase fusion was targeted upstream (toward the 5′ end) of the nCas9 site, the BBE mutated cytosines in a similar “editing window” as those described for conventional BEs and with a similar distribution (FIGS. 4 and 5). When the ZF-deaminase is positioned 8 basepairs downstream of the nCas9 site, the BBE has a much broader editing window that shifts towards the 3′ end of the gRNA target site, suggesting a possible control of the editing window by programmed proximity of the deaminase-containing bipartite “half”. This editing window flexibility adds spatial control to the bipartite base editing circuitry (FIG. 6). It is conceivable or even likely that as-yet untested BBE spatial orientations and spacings will uncover more interesting and useful properties of BBEs. ZFs used in these experiments were originally constructed using the OPEN method and subsequently described as highly active DNA-binding proteins. Expectedly, using a dual UGI approach reduces the amount of target site cytosine-to-guanine and cytosine-to-adenine editing (FIGS. 7A & 7B).8 Most importantly though, and not necessarily only attributable to the second UGI, we saw a substantial reduction in indel formation using BBEs (FIGS. 7C & 7D), thus indicating that BBEs may represent a base editing methodology with significantly fewer product impurities and improved off-targeting properties.

REFERENCES

  • 1. Komor A C, Kim B, Packer M S, Zuris J A, Liu D R. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature. 2016; 533. doi:10.1038/naturel 7946.
  • 2. Gaudelli N M, Komor A C, Rees H A, et al. Programmable base editing of A•T to G•C in genomic DNA without DNA cleavage. Nature. 2017:1-27. doi: 10.1038/nature24644.
  • 3. Yang L, Briggs A W, Chew W L, et al. Engineering and optimising deaminase fusions for genome editing. Nat Commun. 2016; 7 (May): 13330. doi: 10.1038/ncomms 13330.
  • 4. Nishida K, Arazoe T, Yachie N, et al. Targeted nucleotide editing using hybrid prokaryotic and vertebrate adaptive immune systems. Science (80-). 2016. doi: 10.1126/science.aaf8729.
  • 5. Hess G T, Tycko J, Yao D, Bassik M C. Methods and Applications of CRISPR-Mediated Base Editing in Eukaryotic Genomes. Mol Cell. 2017; 68:26-43. doi:10.1016/j.molcel.2017.09.029.
  • 6. Kim D, Lim K, Kim S-T, et al. Genome-wide target specificities of CRISPR RNA-guided programmable deaminases. Nat Biotechnol. 2017; 35(5):475-480. doi: 10.1038/nbt.3852.
  • 7. Rees H A, Komor A C, Yeh W-H, et al. Improving the DNA specificity and applicability of base editing through protein engineering and protein delivery. Nat Commun. 2017. doi:10.1038/ncomms 15790.
  • 8. Kim, Daesik, Kayeong Lim, Sang-Tae Kim, Sun-Heui Yoon, Kyoungmi Kim, Seuk-Min Ryu, and Jin-Soo Kim. “Genome-wide Target Specificities of CRISPR RNA-guided Programmable Deaminases.” Nature Biotechnology (2017).
  • 9. Kleinstiver, Benjamin P., Vikram Pattanayak, Michelle S. Prew, Shengdar Q. Tsai, Nhu T. Nguyen, Zongli Zheng, and J. Keith Joung. “High-fidelity CRISPR-Cas9 Nucleases with No Detectable Genome-wide Off-target Effects.” Nature 529.7587 (2016): 490-95.
  • 10. Slaymaker, I. M., L. Gao, B. Zetsche, D. A. Scott, W. X. Yan, and F. Zhang. “Rationally Engineered Cas9 Nucleases with Improved Specificity.” Science 351.6268 (2015): 84-88.
  • 11. Dahlman, James E., Omar O. Abudayyeh, Julia Joung, Jonathan S. Gootenberg, Feng Zhang, and Silvana Konermann. “Orthogonal Gene Knockout and Activation with a Catalytically Active Cas9 Nuclease.” Nature Biotechnology 33.11 (2015): 1159-161.
  • 12. Fu, Yanfang, et al. “Improving CRISPR-Cas nuclease specificity using truncated guide RNAs.” Nature biotechnology 32.3 (2014): 279-284.
  • 13. Boissel, S., J. Jarjour, A. Astrakhan, A. Adey, A. Gouble, P. Duchateau, J. Shendure, B. L. Stoddard, M. T. Certo, D. Baker, and A. M. Scharenberg. “MegaTALs: A Rare-cleaving Nuclease Architecture for Therapeutic Genome Engineering.” Nucleic Acids Research 42.4 (2013): 2591-601.
  • 14. Bolukbasi, Mehmet Fatih, Ankit Gupta, Sarah Oikemus, Alan G. Derr, Manuel Garber, Michael H. Brodsky, Lihua Julie Zhu, and Scot A. Wolfe. “DNA-binding-domain Fusions Enhance the Targeting Range and Precision of Cas9.” Nature Methods 12.12 (2015): 1150-156.
  • 15. Kleinstiver, Benjamin P., et al. “Broadening the targeting range of Staphylococcus aureus CRISPR-Cas9 by modifying PAM recognition.” Nature biotechnology (2015).
  • 16. Ma, Enbo, et al. “Single-stranded DNA cleavage by divergent CRISPR-Cas9 enzymes.” Molecular cell 60.3 (2015): 398-407.
  • 17. Komor A C, Zhao K T, Packer M S, et al. Improved base excision repair inhibition and bacteriophage Mu Gam protein yields C: G-to-T: A base editors with higher efficiency and product purity. Sci Adv. 2017; (August): 1-10.
  • 18. CA2915837A1
  • 19. Friedland, Ari E., et al. “Characterization of Staphylococcus Aureus Cas9: a Smaller Cas9 for All-in-One Adeno-Associated Virus Delivery and Paired Nickase Applications.” Genome Biology, vol. 16, no. 1, 2015, doi:10.1186/s13059-015-0817-8.
  • 20. Gasiunas, G., et al. “Cas9-CrRNA Ribonucleoprotein Complex Mediates Specific DNA Cleavage for Adaptive Immunity in Bacteria.” Proceedings of the National Academy of Sciences, vol. 109, no. 39, April 2012, doi: 10.1073/pnas. 1208507109.
  • 21. Yamada, Mari, et al. “Crystal Structure of the Minimal Cas9 from Campylobacter Jejuni Reveals the Molecular Diversity in the CRISPR-Cas9 Systems.” Molecular Cell, vol. 65, no. 6, 2017, doi:10.1016j.molcel.2017.02.007.
  • 22. Zetsche, Bernd, et al. “Cpf1 Is a Single RNA-Guided Endonuclease of a Class 2 CRISPR-Cas System.” Cell, vol. 163, no. 3, 2015, pp. 759-771., doi:10.1016/j.cell.2015.09.038.
  • 23. Hirano, H., et al. “Crystal Structure of Francisella Novicida Cas9 RHA in Complex with GRNA and Target DNA (TGG PAM).” February 2016, doi: 10.2210/pdb5b2q/pdb.
  • 24. Yamano, T., et al. “Crystal Structure of Acidaminococcus Sp. Cpf1 in Complex with CrRNA and Target DNA.” April 2016, doi: 10.2210/pd
  • 25. Tang, Xu, et al. “A CRISPR-Cpf1 System for Efficient Genome Editing and Transcriptional Repression in Plants.” Nature Plants, vol. 3, no. 7, 2017, p. 17103, doi: 10.1038/nplants.2017.103.

Other Embodiments

It is to be understood that while the invention has been described in conjunction with the detailed description thereof, the foregoing description is intended to illustrate and not limit the scope of the invention, which is defined by the scope of the appended claims. Other aspects, advantages, and modifications are within the scope of the following claims.

Claims

1. A fusion protein useful in a bipartite cytosine base editor system comprising:

(i) a first fusion protein comprising a non-R-loop-forming programmable DNA binding domain, preferably a transcription-activator-like effector (TALE) or zinc finger array (ZF), fused to a deaminase enzyme, or an active portion thereof, optionally with a linker therebetween, or
(ii) a second fusion protein comprising an R-loop-forming Cas9 protein that lacks nuclease activity or is a nickase, but can interact with a guide RNA and target DNA, fused to a Uracil glycosylase inhibitor (UGI), optionally with a linker therebetween.

2. The fusion protein of claim 1, wherein the first fusion protein comprises a cytidine deaminase selected from the group consisting of apolipoprotein B mRNA-editing enzyme, catalytic polypeptide-like 1 (APOBEC1), APOBEC2, APOBEC3A, APOBEC3B, APOBEC3C, APOBEC3D/E, APOBEC3F, APOBEC3G APOBEC3H, APOBEC4; activation induced cytidine deaminase (AICDA), cytosine deaminase 1 (CDA1), and CDA2, and cytosine deaminase acting on tRNA (CDAT).

3. The fusion protein of claim 1, wherein the second fusion protein comprises an SpCas9 nickase or inactive nuclease with a mutation at D10, E762, D839, H983, or D986 and H840 or N863, or an inactive Cpf1 (dCpf1) protein.

4. The fusion protein of claim 3, wherein the mutation is one or both of D10A or D10N, or H840A/H840N/H840Y.

5. The fusion protein of claim 1, wherein the DNA binding domain comprises a zinc finger DNA binding array.

6. A composition comprising a purified fusion protein of claims 1-5.

7. The composition of claim 6, further comprising a guide RNA that interacts with the Cas9 portion of the fusion protein.

8. The composition of claim 6, comprising one or more ribonucleoprotein (RNP) complexes.

9. A nucleic acid encoding the fusion protein of claims 1-5.

10. A vector comprising the nucleic acid of claim 9.

11. An isolated host cell expressing the fusion protein of claims 1-5.

12. A composition comprising a nucleic acid encoding the fusion protein of claims 1-5.

13. A method of targeted deamination of one or more selected cytosines in a nucleic acid, the method comprising contacting the nucleic acid with the fusion proteins of claims 1-5, and a guide RNA (gRNA) that interacts with the Cas9 portion of the fusion protein and binds to a portion of the nucleic acid comprising or adjacent to the selected cytosines.

14. The method of claim 13, wherein the nucleic acid is in a cell, preferably a eukaryotic cell.

15. A method of targeted deamination of a nucleic acid in a cell, the method comprising expressing in the cell, or contacting the cell with, the fusion proteins of claims 1-5, and a guide RNA (gRNA) that interacts with the Cas9 portion of the fusion protein and binds to a portion of the nucleic acid comprising or adjacent to the selected cytosine.

16. The method of claim 15, wherein the fusion protein is delivered as an RNP, mRNA, or plasmid.

17. A monomeric fusion protein comprising (i) a first portion comprising a deaminase enzyme, or an active portion thereof, (ii) a second portion comprising a Cas9 protein that lacks nuclease activity or is a nickase and can interact with a guide RNA but does not sufficiently bind target DNA or form R-loops to potentiate genome editing or base events, (iii) a third portion comprising a DNA binding domain selected from zinc fingers and TALEs, and optionally (iv) a fourth portion comprising a Uracil DNA glycosylase inhibitor (UGI), optionally with peptide linkers in between some or all of the first, second, third, or fourth portions, and wherein the domains (i)-(iv) can be in any order.

18. A fusion protein comprising (i) a first portion comprising a deaminase enzyme, or an active portion thereof, (ii) a second portion comprising a Cas9 protein that lacks nuclease activity or is a nickase and can interact with a guide RNA but does not sufficiently bind target DNA or form R-loops to potentiate genome editing or base events, such as type-II-C Cas9 orthologs or similarly deficient species, and (iii) a third portion comprising a DNA binding domain selected from zinc fingers and TALEs, optionally with peptide linkers in between some or all of the first, second, or third portions, and wherein the domains (i)-(iii) can be in any order.

19. The fusion proteins of claim 17 and 18, wherein the first portion comprises a cytidine deaminase selected from the group consisting of apolipoprotein B mRNA-editing enzyme, catalytic polypeptide-like 1 (APOBEC1), APOBEC2, APOBEC3A, APOBEC3B, APOBEC3C, APOBEC3D/E, APOBEC3F, APOBEC3G APOBEC3H, APOBEC4; activation induced cytidine deaminase (AICDA), cytosine deaminase 1 (CDA1), and CDA2, and cytosine deaminase acting on tRNA (CDAT).

20. The fusion proteins of claim 17 or 18, wherein the second portion comprises an SpCas9 nuclease with a mutation at D10, E762, D839, H983, or D986 and H840 or N863, or a type-II-C Cas9 ortholog.

21. The fusion proteins of claim 20, wherein the mutation is one or both of D10A or D10N, or H840A/H840N/H840Y.

22. The fusion protein of claim 17 or 18, wherein the second portion comprises PlCas9, ClCas9, or NcCas9; or SpCas9, SaCas9, or CjCas9 with a mutations to a PAM interacting domain (PID).

23. The fusion proteins of claim 22, wherein the mutation to the PID is shown in Table 2.

24. The fusion proteins of claims 17 or 18, wherein the DNA binding domain comprises a zinc finger DNA binding array.

25. A composition comprising a purified fusion protein of claims 17-24.

26. A composition comprising the purified fusion protein of claim 18, and a protein comprising a Uracil DNA glycosylase inhibitor (UGI),

27. The composition of claims 25 or 26, further comprising a guide RNA that interacts with the Cas9 portion of the fusion protein.

28. The composition of claim 27, comprising one or more ribonucleoprotein (RNP) complexes.

29. A nucleic acid encoding the fusion protein of claims 17-24.

30. A vector comprising the nucleic acid of claim 29.

31. An isolated host cell expressing the fusion protein of claims 17-24.

32. A composition comprising a nucleic acid encoding the fusion protein of claims 17 or 19-24.

33. A method of targeted deamination of a selected cytosine in a nucleic acid, the method comprising contacting the nucleic acid with the fusion proteins of claims 17 or 19-24, and a guide RNA (gRNA) that interacts with the Cas9 portion of the fusion protein and binds to a portion of the nucleic acid comprising or adjacent to the selected cytosine.

34. The method of claim 33, wherein the nucleic acid is in a cell.

35. A method of targeted deamination of a nucleic acid in a cell, the method comprising expressing in the cell, or contacting the cell with, the fusion proteins of claims 17 or 19-24, and a guide RNA (gRNA) that interacts with the Cas9 portion of the fusion protein and binds to a portion of the nucleic acid comprising or adjacent to the selected cytosine.

36. The method of claim 35, wherein the fusion protein is delivered as an RNP, or as an mRNA or plasmid encoding the fusion protein.

37. A method of targeted deamination of a selected cytosine in a nucleic acid, the method comprising contacting the nucleic acid with the fusion protein of claim 18, a UGI protein, and a guide RNA (gRNA) that interacts with the Cas9 portion of the fusion protein and binds to a portion of the nucleic acid comprising or adjacent to the selected cytosine.

38. The method of claim 37, wherein the nucleic acid is in a cell.

39. A method of targeted deamination of a nucleic acid in a cell, the method comprising expressing in the cell, or contacting the cell with, the fusion protein of claim 18, a UGI protein, and a guide RNA (gRNA) that interacts with the Cas9 portion of the fusion protein and binds to a portion of the nucleic acid comprising or adjacent to the selected cytosine.

40. The method of claim 39, wherein the fusion protein is delivered as an RNP, or as an mRNA or plasmid encoding the fusion protein.

Patent History
Publication number: 20200140842
Type: Application
Filed: May 25, 2018
Publication Date: May 7, 2020
Inventors: J. Keith Joung (Winchester, MA), James Angstman (Charlestown, MA), Jason Michael Gehrke (Cambridge, MA), Julian Grunewald (Charlestown, MA)
Application Number: 16/616,014
Classifications
International Classification: C12N 9/78 (20060101); C12N 15/10 (20060101); C12N 15/63 (20060101); C12N 15/90 (20060101); C12N 9/22 (20060101);