EP2526199A2 - Procédés de production de nucléases en doigt de zinc ayant une activité modifiée - Google Patents

Procédés de production de nucléases en doigt de zinc ayant une activité modifiée

Info

Publication number
EP2526199A2
EP2526199A2 EP11735277A EP11735277A EP2526199A2 EP 2526199 A2 EP2526199 A2 EP 2526199A2 EP 11735277 A EP11735277 A EP 11735277A EP 11735277 A EP11735277 A EP 11735277A EP 2526199 A2 EP2526199 A2 EP 2526199A2
Authority
EP
European Patent Office
Prior art keywords
zfn
gene
mutated
cell
zinc finger
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP11735277A
Other languages
German (de)
English (en)
Other versions
EP2526199A4 (fr
Inventor
Carlos F. Barbas Iii
Jing Guo
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Scripps Research Institute
Original Assignee
Scripps Research Institute
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Scripps Research Institute filed Critical Scripps Research Institute
Publication of EP2526199A2 publication Critical patent/EP2526199A2/fr
Publication of EP2526199A4 publication Critical patent/EP2526199A4/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/1034Isolating an individual clone by screening libraries
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • C07K14/4703Inhibitors; Suppressors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/1034Isolating an individual clone by screening libraries
    • C12N15/1086Preparation or screening of expression libraries, e.g. reporter assays
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/16Hydrolases (3) acting on ester bonds (3.1)
    • C12N9/22Ribonucleases RNAses, DNAses
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/914Hydrolases (3)
    • G01N2333/916Hydrolases (3) acting on ester bonds (3.1), e.g. phosphatases (3.1.3), phospholipases C or phospholipases D (3.1.4)
    • G01N2333/922Ribonucleases (RNAses); Deoxyribonucleases (DNAses)

Definitions

  • the invention relates generally to zinc finger nucleases having improved catalytic acti vity and more specifically to methods of generating such nucleases,
  • Zinc finger nucleases are chimeric enzymes made by fusing the nonspecific DNA cleavage domain of the endonuclease Fokl with site-specific DNA binding zinc finger domains; these nucleases are powerful tools for gene editing. Due to the flexible nature of zinc finger proteins (ZFPs), ZFNs can be assembled that induce double strand breaks (DSBs) site- specifically into genomic DNA. ZFNs allow specific gene disruption as during DNA repair, the targeted genes can be disrupted via mutagenic non-homologous end joint (NHEJ) or modified via homologous recombination (FIR) if a closely related DNA template is supplied. This method has been applied in many organisms, including plants, Drosophila, C. elegans, zebrafish and mammalian cells. These chimeric enzymes can also be used in basic molecular research as other endonucleases are, providing diverse choices for molecular cloning.
  • ZFPs zinc finger proteins
  • DNBs double strand
  • ZFPs are coupled with the nonspecific Fokl cleavage domain, their affinity and specificity are major determinants of the activity and toxicity of the resulting ZF s.
  • ZFNs are typically composed of lower affinity three- or four- finger ZFPs, This is partially due to the nature of ZFN target sites.
  • ZF target sites are composed of two ZFP binding sites in a tail-to- tail orientation, separated by 5 to 7 bp.
  • the present invention is based on the design of a directed evolution method to identify ZFNs having enhanced activity, as well as the discovery that ZFNs can enter cells directly without fusion or conjugation to a protein transduction domain. Accordingly, particular embodiments of the invention are directed to methods of identifying DNA cleavage domains having increased catalytic activity, such as a hyperactive Fokl cleavage domain (FCD), that can enhance the performance of ZFNs.
  • FCD hyperactive Fokl cleavage domain
  • FCD variant a FCD variant called Sharkey, which is 4-5 fold more active than the wild- type enzyme.
  • Sharkey When coupled with ZFPs, Sharkey stimulated 3-6 fold more mutagenesis in mammalian cells than did ZFNs constaicted with the wild-type Fokl domain.
  • This novel FCD variant will be useful in future ZFN optimization and applications. Accordingly, the present invention relates to methods of improving the catalytic activity of zinc finger nucleases and methods of use of such nucleases.
  • a DNA cleavage domain (CD) of a zinc linger nuclease (ZFN) having enhanced catalytic activity as compared to a reference ZFN includes expressing a mutated zinc finger nuclease (ZFN) having a DNA. cleavage domain (CD) having one or more mutations, and a DNA binding zinc finger domain (ZFD) in a cell comprising a reporter construct.
  • the reporter construct includes in 5' to 3' order a promoter, a toxic gene, and a zinc finger nuclease cleavage site that is recognized by the ZFN, such that the toxic gene is operativeiy linked to the promoter, and whereby the ZFN cleaves the reporter construct, thereby allowing the reporter construct comprising the toxic gene to be degraded.
  • a survival rate is determined for the cell, wherein survival rate is positively correlated with ca talytic activity of the CD of the ZFN, and wherein a survival rate for a eel 1 expressing the mutated ZFN that is higher than a survival rate of a cell expressing a reference ZFN is indicative of the CD of the mutated ZFN having enhanced catalytic activity.
  • a zinc finger nuclease having enhanced catalytic activity.
  • the method includes subjecting a polynucleotide encoding a DNA cleavage domain (CD) to mutagenesis to produce mutated polynucleotides encoding CDs having one or more mutations; fusing the mutated polynucleotides encoding the CDs having one or more mutations to a polynucleotide encoding a DNA binding zinc finger domain (ZFD), thereby creating a librar of
  • the library is expressed in ceils comprising a reporter construct, wherein the reporter construct comprises in 5' to 3' order a promoter, a toxic gene, and a zinc finger nuclease cleavage site that is recognized by the ZFN, wherein the toxic gene is operatively linked to the promoter, and whereby the ZFN cleaves the reporter construct, thereby allowing the reporter construct comprising the toxic gene to be degraded.
  • Cells are selected that express a mutated ZFN having a survival rate that is higher than a survival rate of a cell expressing a reference ZFN , wherein a higher survival rate is indicative of the mutated ZFN having enhanced catalytic activity.
  • isolated zinc finger nuclease (ZFN) proteins including a zinc finger DNA cleavage domain (CD) having enhanced catalytic activity obtained by a method provided herein, and a DNA binding zinc finger domain (ZFD).
  • the isolated zinc finger nuclease includes a CD having an amino acid sequence selected from the group consisting of SEQ ID NOs:3-6.
  • the ZFD contains three, or four, or more zinc finger proteins.
  • the ZFD contains three zinc finger proteins; in another aspect, the ZFD contains four zinc finger proteins.
  • polynucleotide molecules encoding such ZFNs are also provided.
  • isolated zinc finger nucleases having altered catalytic activity obtained by a method of the invention.
  • the isolated zinc finger nuclease includes the amino acid sequence of SEQ ID NOs: 1 or 2.
  • polynucleotide molecules encoding such ZFNs.
  • methods of introducing a break into a nucleic acid molecule at a site of interest includes contacting a nucleic acid molecule with a ZFN as provided herein or identified by a method provided herein, or containing a CD provided herein or identified by a method provided herein.
  • the ZFN contains a DNA binding zinc finger domain (ZFD) that binds a target site in proximity to the site of interest so that upon binding of the ZFN to the target site, the ZFN cleaves the nucleic acid at the site of interest, thereby introducing a break into the nucleic acid molecule.
  • ZFD DNA binding zinc finger domain
  • methods of treating a subject having a cell proliferative disorder includes inactivating or mutating a gene according by administering a ZFN as provided herein or identified by a method provided herein, or containing a CD provided herein or identified by a method provided herein to the subject, wherein over-expression of the gene is associated the cell prolifera tive disorder, thereby treating the cell proliferative disorder.
  • the method includes mutating the gene of interest in a cell or population of cells by introducing, into the cells, a ZFN as pro vided herein, wherein the ZFN contains a DNA binding zinc finger domain (ZFD) that binds a target site within the gene of interest, such that the ZFN is expressed in the cell, whereby the ZFN binds to the target site and cleaves the gene of interest; and culturing the cells whereby progeny cells in which the gene of interest is muta ted are produced.
  • the cell is transfected with a nucleic acid molecule encoding the ZFN.
  • mutating or knocking out a gene of interest in a cell or population of cells includes mutating the gene of interest in a target cell by contacting the cell with a ZFN protein provided herein or a ZFN containing a CD of native or engineered sequence, wherein the ZFD binds a target site within the cell genome, with the proviso that the ZFN is not fused or conjugated to a protein transduction domain, such that the ZFN binds to the target site and cleaves the gene of interest; and culturing the cell, whereby progeny cells in which the gene of interest is mutated or knocked out are produced.
  • mutating the gene of interest results in activation or restoration of expression of the gene of interest.
  • methods of mutating a gene of interest in a cell or population of cells by mutating the gene of interest in a target cell by contacting the cell with a ZFN protein containing a protein transduction domain.
  • the ZFN is as provided herein or containing a CD of engineered sequence, wherein the ZFD binds a target site within the cell genome, and wherein the ZFN is fused or conjugated to a protein transduction domain, such that the ZFN binds to the target site and cleaves the gene of interest and culturing the ceil, whereby progeny cells in which the gene of interest is mutated or knocked out are produced.
  • the method further includes delivering to the cell, either prior to, simultaneously with or following mutating the gene of interest, a corrective nucleic acid or vector containing the nucleic acid, thereby providing a substitute for the knocked out or mutated gene of interest.
  • Figure 1 shows a schematic of the reporter construct ( Figure l a), the ZFN construct ( Figure lb), and the selection strategy for identifying ZFNs having altered catalytic activity.
  • Figure 2a shows photographs of the colonies resulting from the transformation of a library of ZFNs into selection strain BW25141 and subjected to multiple rounds of evolution.
  • Figure 2b shows a plot of the survival rate curves for wt, R3, R6, and R9.
  • Figure 2c shows a photograph of the extent of linearization of the substrate in cellular extracts.
  • Figure 2d shows a plot of the survi val rate measured for each round of selection at 1 hour.
  • Figure 3a shows a photograph of the in vitro cleavage of target DNA by P3 nuclease with either Sharkey or wtFokl catalytic domain.
  • Figure 3b shows a plot of cleavage rates determined by measuring the initial velocity of pSub ⁇ P3 linearization for Sharkey and wt.
  • Figure 3 c shows a three-dimensional stmcture of full-length Fokl in complex with DN A (PDB ID: 1 FOK).
  • Figure 3d shows a diagram depicting the location of S418P and Q481 H proximal to Asp450, Asp467 and Lys469 in Fokl.
  • Figure 3e shows activity analysis of FCD variants containing the selected mutations 8418P, K441 E, Q481 H, N527D, and S418P::K441E with the P3 ZF domain.
  • Figure 3f shows an activity analysis of FCD variants
  • FIG. 4a shows a schematic overview of the reporter system used to evaluate the efficiency of mutagenesis in mammalian cells (EGFP sense (SEQ ID NO:8) and antisense (SEQ ID NO:9)).
  • Figure 4b shows representative flow cytometry data for reporter cells transfected with CMV controlled wtFokl and Sharkey cleavage domains with 3, 4, 5 and 6- finger zinc finger DNA binding domains.
  • Figure 4c shows a plot of the quantification of EGFP positive reporter cells following transfeetion with ZFN.
  • Figure 4d shows a photograph of the results of the Mlul restriction digest assay of HEK 293 reporter cells transfected with ZFN. 'Cut' indicates the presence of unmodified reporter gene. 'Uncut' indicates the presence of ZFN modified reporter gene.
  • Figure 5 shows plots depicting the efficiencies for ZFN dimerization variants consisting of wild-type and Sharkey cleavage domains.
  • Figure 6 shows the sequences of nuclease constructs (SEQ ID NO'S 1 to 6).
  • Figure 6a sho ws the complete amino acid sequence of the P3.wt construct used in protein evolution and the E6.wt construct used in the mutagenesis assay. The recognition a-helices is underlined,
  • Figure 7 shows a plot of the results of a ⁇ - ⁇ 2 ⁇ based cytotoxicity assay.
  • Figure 8 shows the amino acid sequence of the E4.FN construct (SEQ ID NO:7).
  • Figure 9 shows a schematic overview of the reporter system used to evalua te the efficiency of mutagenesis in mammalian cells in Example 2 (SEQ ID NO'S 8 and 9).
  • Figure 10 shows plots of the %EGFP positive cells by FACS analysis.
  • Figure 1 1 shows a photograph of results of an Mlul digestion assay.
  • Figure 12 shows a schematic of the reporter construct, the ZFN construct, and the selection strategy for identifying ZFNs having altered activity using a negative selection strategy.
  • Figure 13a shows an electrosta tic potential map of the ZFN surface
  • Figure 13b shows an SDS-PADE analysis of purified rZFN consisting of the native Foki (wt) and the Sharkey (Sh) cleavage domain.
  • Figures 13c and 13d show flow cytometry analysis of HEK 293 cells following transduction with fc) medium and fd) rZFN.
  • the present in vention is based on the design of a directed evolution method to identify ZFNs having enhanced activity, as well as the discovery that ZFNs can enter cells directly without fusion or conjugation to a protein transduction domain. Accordingly, particular embodiments of the invention are directed to methods of identifying D A cleavage domains having increased catalytic activity, such as a hyperactive Fokl cleavage domain, that can enhance the performance of ZFNs.
  • Zinc finger nucleases are enzymes having a DNA cleavage domain and a UNA binding zinc finger domain. ZFNs may be made by fusing the nonspecific DNA cleavage domain of an endonuclease with site-specific DNA binding zinc finger domains.
  • nucleases are powerful tools for gene editing and can be assembled to induce double strand breaks (DSBs) site-specificaily into genomic DNA.
  • ZFNs allow specific gene disruption as during DNA repair, the targeted genes can be disrupted via mutagenic nonhomologous end joint (NHEJ) or modified via homologous recombination (HR) if a closely related DNA template is supplied.
  • NHEJ nonhomologous end joint
  • HR homologous recombination
  • the zinc finger nucleases have altered catalytic activity and are obtained by a method of the in vention.
  • the ZFN is ceil permeable, that is, the ZFN is able to cross the cell membrane when contacted with the cell.
  • such cell permeable ZFNs are not fused or conjugated to a protein transduction domain.
  • the ZFN is fused or conjugated to a protein transduction domain.
  • Protein transduction domains (PTDs) as used herein generally refer to polypeptides capable of transducing cargo across the plasma membrane, allowing the proteins to accumulate within the cell.
  • Three exemplary PTDs include the Drosophila homeotic transcription protein antennapedia (Antp), the herpes simplex virus structural protein VP22, and the human immunodeficiency virus 1 (HIV-1) transcriptional activator Tat protein.
  • PTDs are known in the art (e.g., Wadia & Dowdy, Curr Opin Biotech 13:52-6, 2002; Snyder & Dowdy, Expert Opin Drug Deliv 2(1):43-51, 2005) may be fused or conjugated to a ZFN by recombinant or chemical conjugation methods known in the art.
  • the isolated zinc finger nuclease includes the amino acid sequence of SEQ ID NOs: 1 or 2.
  • the ZFN contains a cleavage domain selected from the group consisting of SEQ ID NOs:3 ⁇ 6,
  • the ZFN has increased catalytic activity relative to a reference ZFN .
  • a reference ZFN as used herein is generally a ZFN having known activity.
  • the reference ZFN contains a wild type cleavage domain; in another aspect the reference ZFN is a mutated ZFN, which is further mutated to form successive generations of mutated ZFNs.
  • the reference ZFN may be the immediately- preceding generation of mutated ZFN, when the mutagenesis and selection steps are repeated one or more times.
  • a "DNA cleavage domain” or “cleavage domain” includes one or more polypeptide sequences which possesses catalytic activity for DNA cleavage.
  • a cleavage domain can be contained in a single polypeptide chain or cleavage activity can result from the association of two (or more) polypepti des.
  • a CD can be obtained from any endoniiclease or exonuclease.
  • Exemplary endonucleases from which a cleavage domain can be derived include, but are not limited to, restriction endonucleases and homing endonucleases.
  • Restriction endonucleases are present in many species and are capable of sequence-specific binding to DN A (at a recognition site), and cleaving DNA at or near the site of binding.
  • Certain restriction enzymes e.g., Type I IS
  • Fokl catalyzes double-stranded cleavage of DNA, at 9 nucleotides from its recognition site on one strand and 13 nucleotides from its recognition site on the other. See, for example, U.S. Pat. Nos.
  • the CD may be a variant of a wild type cleavage domain. Such variant CDs may contain 1, 2, 3, 4, 5, 6, or more mutations. Such variant CDs may be generated by the methods provided herein.
  • the CD may be a wild type Fokl cleavage domain (FCD) from endoniiclease Fokl.
  • FCD contains the sequence set forth in SEQ ID NO:3.
  • the CD may be a variant of the FC D, Such variant FCDs may contain 1, 2, 3, 4, 5, 6, or more mutations.
  • the FCD has one or more mutations selected from the group consisting of S4I 8P, F432L, K441E, Q481 H, H523Y, N527D, and K559Q.
  • the CD contains a sequence as set forth in SEQ ID NO:4, 5, or 6.
  • Clea vage refers to the breakage of the covalent backbone of a DNA molecule. Cleavage can be initiated by a variety of methods including, but not limited to, enzymatic or chemical hydrolysis of a phosphodiester bond, Both single-stranded cleavage and double- stranded cleavage are possible, and double-stranded cleavage can occur as a result of two distinct single-stranded cleavage events. DNA cleavage can result in the production of either blunt ends or staggered ends. In certain embodiments, fusion polypeptides are used for targeted double-stranded DNA cleavage.
  • a "DNA binding zinc finger domain” (ZFD) or binding domain is a protein, or a domain within a larger protein, that binds DNA in a sequence-specific-manner through one or more zinc fingers, which are regions of amino acid sequence within the binding domain whose structure is stabilized through coordination of a zinc ion.
  • the term zinc finger DNA binding protein is often abbreviated as zinc finger protein or ZFP.
  • zinc finger protein refers to a polypeptide having nucleic acid, e.g., DNA, binding domains that are stabilized by zinc
  • the individual DNA binding domains are typically referred to as “fingers,” such that a zinc finger protein or polypeptide has at least one finger, more typically two fingers, or three fingers, or even four or five fingers, to at least six or more fingers.
  • the ZFP contains 3 zinc fingers; in another aspect, the ZFP contains 4 zinc fingers.
  • Each finger binds from two to four base pairs of DN A, typically three or four base pairs of DNA.
  • a ZFP binds to a nucleic acid sequence called a target nucleic acid sequence.
  • Each finger usually comprises an approximately 30 amino acids, zinc-chelating, DNA-binding subdomain.
  • a zinc finger protein can have at least two DNA-binding domains, one of which i s a zinc finger polypeptide, linked to the other domain via a flexible linker.
  • the two domains can be identical or different.
  • Both domains can be zinc finger proteins, either identical or different zinc finger proteins.
  • framework (or backbone) derived from a naturally occurring zinc finger protein means that the protein or peptide sequence within the naturally occurring zinc finger protein that is involved in non-sequence specific binding with a target nucleotide sequence is not substantially changed from its natural sequence.
  • framework (or backbone) derived from the naturally occurring zinc finger protein maintains at least 50%, and preferably, 60%, 70%, 80%, 90%, 95°/», 99% or 100% identity compared to its natural sequence in the non-sequence specific binding region.
  • the nucleic acid encoding such framework for backbone) derived from the naturally occurring zinc finger protein can be hybridizable with the nucleic acid encoding the naturally occurring zinc finger protein, either entirely or within the non-sequence specific binding region, under low, medium or high stringency condition.
  • the nucleic acid encoding such framework (or backbone) derived from the naturally occurring zinc finger protein is hybridizable with the nucleic acid encoding the naturally occurring zinc finger protein, either entirely or within the non-sequence specific binding region, under high stringency condition.
  • Zinc finger proteins can be designed and predicted according to the procedures in WO 98/54311 can be used in the present methods.
  • WO 98/543 ! 1 discloses technology which allows the design of zinc finger protein domains that bind specific nucleotide sequences that are unique to a target gene. It has been calculated that a sequence comprising 18 nucleotides is sufficient to specify an unique location in the genome of higher organisms. Typically, therefore, the zinc finger protein domains are hexadactyl, i.e., contain 6 zinc fingers, each with its specifically designed alpha helix for interaction with a particular triplet. However, in some instances, a shorter or longer nucleotide target sequence may be desirable.
  • the zinc finger domains in the proteins may contain at least 3 fingers, or from 2-12 fingers, or 3-8 fingers, or 3-4 fingers, or 5-7 fingers, or even 6 fingers.
  • the ZFP contains 3 zinc fingers; in another aspect, the ZFP contains 4 zinc fingers.
  • a multi-finger protein binds to a polynucleotide duplex, e.g., DNA, R A, PNA or any hybrids thereof, its fingers typically line up along the polynucleotide duplex with a periodicit of about one finger per 3 bases of nucleotide sequence,
  • the binding sites of individual zinc fingers (or subsites) typically span three to four bases, and subsites of adjacent fingers usually overlap by one base,
  • a three-finger zinc finger protein XYZ binds to the 10 base pair site abcdefghij (where these letters indicate one of the duplex DNA) with the subsite of finger X being ghij, finger Y being defg and finger Z being abed.
  • zinc fingers Y and Z would have the same polypeptide sequence as found in the original zinc finger discussed above (perhaps a wild type zinc fingers which bind defg and abed, respectively), Finger X would have a mutated polypeptide sequence.
  • finger X would have mutations at one or more of the base-contacting positions, i.e., finger X would have the same polypeptide sequence as a wild type zinc finger except that at leas t one of the four amino residues at the primary positions would differ.
  • fingers X and Z have the same sequence as the wild type zinc fingers which bind ghij and abed, respectively, while finger Y would have residues at one or more base-coating positions which differ from those in a wild type finger.
  • the present method can employ multi-fingered proteins in which more than one finger differs from a wild type zinc finger.
  • the present method can also employ multi-fingered protein in which the amino acid sequence in all the fingers have been changed, including those designed by combinatorial chemistry or other protein design and binding assays,
  • a zinc finger protein it is also possible to design or select a zinc finger protein to bind to a targeted polynucleotide in which more than four bases have been altered. In this case, more than one finger of the binding protein must be altered. For example, in the 10 base sequence
  • a three-finger binding protein could be designed in which fingers X and Z differ from the corresponding fingers in a wild type zinc finger, while finger Y will have the same polypeptide sequence as the corresponding finger in the wild type fingers which binds to the subsite defg.
  • Binding proteins having more than three fingers can also be designed for base sequences of longer length. For example, a four finger-protein will optimally bind to a 13 base sequence, while a five-finger protein will optimally bind to a 16 base sequence.
  • a multi-finger protein can also be designed in which some of the fingers are no t involved in binding to the selected DNA. Slight variations are also possible in the spacing of the fingers and framework.
  • Methods for designing and identifying a zinc finger protein with desired nucleic acid binding characteristics also include those described in WO98/53060, which reports a method for preparing a nucleic acid binding protein of the Cys2-His2 (SEQ ID NO: 10) zinc finger class capable of binding to a nucleic acid quadruplet in a target nucleic acid sequence.
  • Zinc finger proteins useful in the present method can include at least one zinc finger polypeptide linked via a linker, preferably a flexible linker, to at least a second DNA binding domain, which optionally is a second zinc finger polypeptide.
  • the zinc finger protein may contain more than two DNA-binding domains.
  • the zinc finger polypeptides used in the present methods can be engineered to recognize a selected target site in a gene of choice.
  • a backbone from any suitable C2H2-ZFP, such as SPA, SPIC, or ZIF268, is used as the scaffold for the engineered zinc finger polypeptides (see, e.g., Jacobs, EMBO J. (1992) 11 :4507; and Desjarlais & Berg, Proc.
  • a number of methods can then be used to design and select a zinc finger polypeptide with high affinity for its target.
  • a zinc finger polypeptide can be designed or selected to bind to any suitable target site in the target gene, with high affinity.
  • nucleic acids encoding zinc finger polypeptides e.g., phage display, random mutagenesis
  • Zinc linger proteins and zinc finger nucleases can be made by any recombinant DNA technology method for gene construction.
  • PCR based construction can be used. Ligation of desired fragments can also be performed, using linkers or appropriately complementary restriction sites.
  • linkers or appropriately complementary restriction sites One can also synthesize entire finger domain or parts thereof by any protein synthesis method.
  • Preferred for cost and flexibility is the use of PCR primers that encode a finger sequence or part thereof with known base pair specificity , and that can be reused or recombined to create new combinations of fingers and ZFP sequences,
  • the amino acid linker should be flexible, a beta turn structure is preferred, to allow each three finger domain to independently bind to its target sequence and avoid steric hindrance of each other's binding.
  • Linkers can be designed and empirically tested.
  • the ZFP can be designed to bind to non-contiguous target sequences,
  • a target sequence for a six-finger ZFP can be a nine base pair sequence (recognized by three fingers) with intervening bases (that do not contact the zinc finger nucleic acid binding domain) between a second nine base pair sequence (recognized by a second set of three fingers).
  • the number of intervening bases can vary, such that one can compensate for this intervening distance with an
  • a range of intervening nucleic acid bases in a target binding site is preferably 20 or less bases, more preferably 10 or less, and even more preferably 6 or less bases. It is of course recognized that the linker must maintain the reading frame between the linked parts of ZFP protein,
  • a minimum length of a linker is the length that would allow the two zinc finger domains to be connected without providing steric hindrance to the domains or the linker,
  • a linker that provides more than the minimum length is a "flexible linker.” Determining the length of minimum linkers and flexible linkers can be performed using physical or computer models of DNA-binding proteins bound to their respective target sites as are known in the art.
  • the six-finger zinc finger peptides can use a conventional "TGE P" (SEQ ID NO: 12) linker to connect two three-finger zinc finger peptides or to add additional fingers to a three-finger protein.
  • TGE P SEQ ID NO: 12
  • Other zinc finger peptide linkers both natural and synthetic, are also suitable.
  • a useful zinc finger framework is that of ZIF268 (see WO00/23464 and references cited therein.), however, others are suitable.
  • Examples of known zinc finger nucleotide binding polypeptides that can be truncated, expanded, and/or mutagenized in order to change the function of a nucleotide sequence containing a zinc finger nucleotide binding motif includes TFIIIA and zif268.
  • Other zinc finger nucleotide binding proteins are known to those of skill in the art.
  • the murine CYS2-HiS2 (SEQ ID NO: 10) zinc finger protein Zif268 is structurally well characterized of the zinc finger proteins (Pavletich and Pabo, Science (1991) 252:809-817; Elrod-Erickson et al, Structure (London) (1996) 4:1171-1180; and Swimoff et al., Mol. Cell, Biol, (1995) 15:2275-2287).
  • DNA recognition in each of the three zinc linger domains of this protein is mediated by residues in the N-terminus of the alpha-helix contacting primarily three nucleotides on a single strand of the DNA ,
  • the operator binding site for this three finger protein is 5'-GCGTGGGCG-'3.
  • zinc finger domains appear to specify overlapping 4 bp sites rather than individual 3 bp sites.
  • residues in addition to those found at helix positions -1, 3, and 6 are involved in contacting DNA (Elrod- Erickson et al,, Structure (1996) 4: 1171 -1180).
  • an aspartate in helix position 2 of the middle finger plays several roles in recognition and makes a variety of contacts.
  • This aspartate may also participate in water-mediated contacts with the guanine's complementary cytosine.
  • this carboxylate is observed to make a direct contact to the N4 of the cytosine base on the opposite strand of the 5'-guanine base of the finger 1 binding site. It is this interaction which is the chemical basis for target site overlap.
  • any suitable method of protein purification known to those of skill in the art can be used to purify the zinc finger nucleases of the invention (see Sambrook et al., Molecular Cloning: A Laboratory Manual (2d ed., Cold Spring Harbor Laboratory Press, Plainview, N.Y.) (1989)).
  • any suitable host can be used, e.g., bacterial cells, insect cells, yeast cells, mammalian cells, and the like.
  • vector or plasmid refers to discrete elements that are used to introduce heterologous DNA into cells for either expression or replication thereof. Selection and use of such vehicles are well known within the skill of the artisan.
  • An expression vector includes vec tors capable of expressing DNAs that are operatively linked with regulatory sequences, such as promoter regions, that are capable of effecting expression of such DN A fragments.
  • an expression vector refers to a recombinant DNA or RNA construct, such as a plasmid, a phage, recombinant virus or other vector that, upon introduction into an appropriate host ceil, results in expression of the cloned DNA
  • Appropriate expression vectors are well known to those of skill in the art and include those that are replicable in eukaryotic cells and/or prokaryotic cells and those that remain episomal or those which integrate into the host cell genome.
  • the present methods include reporter constructs or vectors and ZFN expression constructs or vectors. In one aspect these expression constructs are plasmids.
  • a DNA cleavage domain (CD) of a zinc finger nuclease (ZFN) having enhanced catalytic activity as compared to a reference ZFN includes expressing a mutated zinc finger nuclease (ZFN) having a DN A cleavage domain (CD) having one or more mutations, and a DNA binding zinc finger domain (ZFD) in a cell comprising a reporter construct.
  • ZFN mutated zinc finger nuclease
  • CD DNA binding zinc finger domain
  • the reporter construct includes in 5' to 3' order a promoter, a toxic gene, and a zinc finger nuclease cleavage site that is recognized by the ZFN, such that the toxic gene is operatively linked to the promoter, and whereby the ZFN cleaves the reporter construct, thereby allowing the reporter construct comprising the toxic gene to be degraded.
  • a survival rate is determined for the cell, wherein survival rate is positively correlated with ca talytic activity of the CD of the ZFN, and wherein a survival rate for a cell expressing the mutated ZFN that is higher than a survival rate of a cell expressing a reference ZFN is indicative of the CD of the mutated ZFN having enhanced catalytic activity.
  • the selection system is a positive selection system.
  • mutants that can cleave the reporter construct at the ZFN cleavage site will survive on a corresponding solid selection medium
  • the reporter construct contains the toxic gene ccdB and a downstream (or 3') ZFN cleavage site; mutants that cleave at the cleavage sit will survive on solid selection medium (e.g., medium containing 25 ng/mL zeocin and 10 mM arabinose) (see e.g., Figure 1).
  • Recombinant ZFNs are also used to genetically correct mutations in cells.
  • FGFR3 mutations in primary fibroblast cells derived from patients with achondroplasia can be corrected.
  • Current gene deliver methods include plasmid DNA nucleofection, integrase-deficient lentivirai vectors and adenoviral vectors. These strategies, however, require the use of transfection reagents and/or introduction of plasmid DNA that often result in high cellular toxicity.
  • the present invention provides a protocol for the expression, purification and introduction of cell-permeable rZFNs into human cells.
  • PTDs Positively charged protein transduction domains
  • ZFNs may associate with the negatively charged components of the cell membrane in a manner that results in ceil penetration.
  • the inventors engineered ZFNs to stimulate cleavage and disrupt expression of the HIV-1 co-receptor CCR5.
  • ZFNs designed to target the CCR.5 locus were cloned into an expression vector and genetically fused to an N-terminal polyhistidine tag. These enzymes were expressed in E, coli and purified to >90% homogeneity by column chromatography, The conditions have been optimized such that protein re-folding or dialysis is not necessary to restore enzyme activity.
  • rZFNs are cell-permeable and can stimulate genomic cleavage in the context of the human cell, we incubated rZFNs with HEK 293 reporter cells. As described above, these cells have been transformed to contain a
  • ZFN-mediated muta genesis of the target allele results in the restoration of EGFP iluorescence.
  • rZFNs to disrupt CCR5 expression in CD4+ T cells in culture.
  • rZFN entry into cells does not require additional factors such as transfection reagents or viral vectors, thus minimizing toxicity as well as the potential for random integration events.
  • the data demonstrate that recombinant ZFNs may be used to stimulate mutagenesis in the context of the human genome.
  • rZFNs may further expand the utility of ZFNs as reagents for routine stem cell modification.
  • the reporter construct contains the ampicillin resistance gene and a downstream (or 3') ZFN cleavage site; mutants that cleave at the cleavage site will not survive on solid selection medium (e.g., medium containing 25 ng/mL zeocin and 100 ⁇ g/mL carbenicillin) (see e.g., Figure 12).
  • solid selection medium e.g., medium containing 25 ng/mL zeocin and 100 ⁇ g/mL carbenicillin
  • the reporter construct or expression construct encoding the ZFN may include a promoter that tightly controls expression of the protein.
  • the BAD promoter is used.
  • the determining the survival rate of the cell step is performed by plating on selective medium the cel l expressing the mutated ZFN and comparing to the number of colonies produced to the number of colonies produced by plating cells expressing the reference ZFN. Bacterial cells are typically used for these assays. In one aspect E. coli cells are used.
  • the method further includes isolating the expression construct encoding the mutated ZFN; mutating the polynucleotide encoding the mutated ZFN to produce a second mutated ZFN, and repeating the expressing and determining steps with the second mutated ZFN to identify altered catalytic activity in the second mutated ZFN, as compared to the mutated ZFN.
  • the catalytic activity is increased as compared to the prior generation of mutated ZFN. This process be repeated one or more times to produce successive generations of mutated ZFNs having further increased activity.
  • Mutations may be introduced into the polynucleotides encoding the ZFNs by methods known to the skilled artisan. Some of these methods include, random mutagenesis, error-prone PGR., chemical mutagenesis, site-directed mutagenesis, and other methods well known in the art (for a comprehensive listing of current mutagenesis methods, see Maniatis, Molecular Cloning: A Laboratory Manual, ( "' old Spring Harbor Press, Cold Spring, N.Y. (1982)), Random mutagenesis has been the most widely recognized method to date.
  • Random mutagenesis has been the most widely recognized method to date. Typically, this has been carried out either through the use of error-prone PGR (as described in Moore, J., et al, Nature Biotechnology 14:458, (1996), or through the application of randomized synthetic
  • one or more mutations are introduced into a polynucleotide by error-prone amplification (e.g., error-prone PGR) of the polynucleotide.
  • error-prone amplification e.g., error-prone PGR
  • mutation of polynucleotides may be achieved by DNA shuffling.
  • DNA shuffling involves the assembly of two or more DNA segments by
  • DNA shuffling combines the principal of in vitro recombination, along with the method of error-prone PGR. Beginning with a randomly digested pool of small fragments of the polynucleotide, created by Dnase I digestion, random fragments are used in an error-prone PGR assembly reaction. During the PGR reaction, the randomly sized DNA fragments not only hybridize to their cognate strand, but also may hybridize to other DNA fragments corresponding to different regions of the polynucleotide of interest— regions not typically accessibl e via hybridization of the entire polynucleotide.
  • the PGR assembly reaction utilizes error-prone PGR reaction conditions, random mutations are introduced during the DN A synthesis step of the PGR reaction for all of the fragments further diversifying the potential hy bridization sites during the anneal ing step of the reaction.
  • a method of introducing a break into a nucleic acid molecule at a site of interest includes contacting a nucleic acid molecule with a ZFN as provided herein or identified by a method provided herein, or containing a CD provided herein or identified by a method provided herein.
  • the ZFN contains a DNA binding zinc finger domain (ZFD) that binds a target site in proximity to the site of interest so that upon binding of the ZFN to the target site, the ZFN cleaves the nucleic acid at the site of interest, thereby introducing a break into the nucleic acid molecule.
  • ZFD DNA binding zinc finger domain
  • Recombination refers to a process of exchange of genetic information between two polynucleotides.
  • homologous recombination refers to the specialized form of such exchange that takes place, for example, during repair of double-strand breaks in cells. This process requires nucleotide sequence homology, uses a "donor” molecule to template repair of a "target” molecule (i.e., the one that experienced the double-strand break), and is variously known as “non-crossover gene conversion” or “short tract gene con version,” because it leads to the transfer of genetic information from the donor to the target.
  • such transfer can involve mismatch correction of heteroduplex DNA that forms between the broken target and the donor, and/or "synthesis-dependent strand annealing," in which the donor is used to resynthesize genetic information that w ill become part of the target, and/or related processes.
  • Such specialized HR often results in an alteration of the sequence of the target molecule such that part or all of the sequence of the donor polynucleotide is incorporated into the target polynucleotide.
  • the site at which the DNA is cleaved generally lies between the binding sites for the two ZFNs. Double-strand breakage of DNA often results from two single-strand breaks, or "nicks," offset by 1 , 2, 3, 4, 5, 6 or more nucleotides, (for example, cleavage of double- stranded DNA by native Fok I results from single-strand breaks offset by 4 nucleotides). Thus, cleavage does not necessarily occur at exactly opposite sites on each DN A strand.
  • the structure of the ZFNs and the distance between the target sites can influence whether cleavage occurs adjacent a single nucleotide pair, or whether cleavage occurs at several sites. However, for many applications, including targeted recombination and targeted mutagenesis, cleavage within a range of nucleotides is generally sufficient, and cleavage between particular base pairs is not required.
  • a ZFN can be expressed in a cell following the introduction, into the cell, of polypeptides and/or polynucleotides.
  • a ZFN protein may exert its effect on the chromatin contained within a cell by contacting the ceil with the ZFN. in such cases, the ZFN enters the cell and modifies the target gene.
  • ZFNs for use in the present methods include ZFNs having a DNA cleavage domain (CD) with enhanced catalytic activity obtained by a method provided herein, and a DNA binding zinc finger domain (ZFD).
  • the isolated zinc finger nuclease includes a CD having an amino acid sequence selected from the group consisting of SEQ ID NOs:3-6.
  • the ZFD contains three, or four, or more zinc finger proteins.
  • the ZFD contains three zinc finger proteins; in another aspect, the ZFD contains four zinc finger proteins.
  • the zinc finger nucleases (ZFN) having altered catalytic activity may be obtained by a method of the invention.
  • the isolated zinc finger nuclease includes the amino acid sequence of SEQ ID NOs: ! or 2.
  • targeted cleavage in a genomic region by a ZFN results in alteration of the nucleotide sequence of the region, follo wing repair of the cleavage event by non-homologous end joining (NHEJ).
  • NHEJ non-homologous end joining
  • targeted cleavage in a genomic region by a ZFN can also be part of a procedure in which a genomic sequence (e.g., a region of interest in cellular chromatin) is replaced with a homologous non-identical sequence (i.e., by targeted recombination) via homology-dependent mechanisms (e.g., insertion of a donor sequence comprising an exogenous sequence together with one or more sequences that are either identical, or homologous but non-identical, with a predetermined genomic sequence (i.e., a target site)).
  • a genomic sequence e.g., a region of interest in cellular chromatin
  • homologous non-identical sequence i.e., by targeted recombination
  • homology-dependent mechanisms e.g., insertion of a donor sequence comprising an exogenous sequence together with one or more sequences that are either identical, or homologous but non-identical, with a predetermined genomic sequence (i.e., a target site
  • Targeted replacement of a selected genomic sequence requires, in addition to the ZFNs described herein, the introduction of an exogenous (donor) polynucleotide.
  • the donor polynucleotide can be introduced into the cell prior to, concurrently with, or subsequent to, expression of the ZFNs.
  • the donor polynucleotide contains sufficient homology to a genomic sequence to support homologous recombination (or homology-directed repair) between it and the genomic sequence to which it bears homology.
  • Donor polynucleotides can range in length from 10 to 5,000 nucleotides (or any integral value of nucleotides therebetween) or longer.
  • nucleotide sequence of the donor polynucleotide is typically not identical to that of the genomic sequence that it replaces.
  • sequence of the donor polynucleotide can contain one or more substitutions, insertions, deletions, inversions or rearrangements with respect to the genomic sequence, so long as sufficient homology with chromosomal sequences is present.
  • sequence changes can be of any size and can be as small as a single nucleotide pair,
  • a donor polynucleotide can contain a non-homologous sequence (i.e., an exogenous sequence, to be distinguished from an exogenous polynucleotide) flanked by two regions of homology.
  • donor polynucleotides can comprise a vector molecule containing sequences that are not homologous to the region of interest in cellular chromatin.
  • the homologous region(s) of a donor polynucleotide will have at least 50% sequence identity to a genomic sequence with which recombination is desired. In certain embodiments, 60%, 70%, 80%, 90%, 95%, 98%, 99%, or 99.9% sequence identity is present. Any value between 1% and 100% sequence identity can be present, depending upon the length of the donor polynucleotide.
  • a donor molecule can contain several, discontinuous regions of homology to cellular chromatin. For example, for targeted insertion of sequences not normally present in a region of interest, said sequences can be present in a donor nucleic acid molecule and flanked by regions of homology to sequence in the region of interest.
  • the method includes inactivating or mutating a gene according by administering a ZFN as provided herein or identified by a method provided herein, or containing a CD provided herein or identified by a method provided herein to the subject, wherein over-expression of the gene is associated the cell proliferative disorder, thereby treating the cell proliferative disorder.
  • the ZFN is administered as a protein; in other embodiments, the ZFN is administered as an expression construct encoding the ZFN.
  • the cell proliferative disorder is cancer.
  • a cancer can include, but is not limited to, colorectal cancer, esophageal cancer, stomach cancer, leukemia,''! ymphoma, Sung cancer, prostate cancer, uterine cancer, breast cancer, skin cancer, endocrine cancer, urinary cancer, pancreas cancer, other gastrointestinal cancer, ovarian cancer, cervical cancer, head cancer, neck cancer, and adenomas.
  • Zinc finger nucleases are powerful tools for gene therapy and genetic engineering. The high specificity and affinity of these chimeric enzymes are based on custom- designed zinc finger proteins (ZFPs).
  • ZFPs custom- designed zinc finger proteins
  • This example illustrates the development of a method to improve the performance of ZFNs, specifically, an in vivo evolution-based approach to improve the efficacy of the Fokl cleavage domain (FCD).
  • FCD Fokl cleavage domain
  • An in vitro DNA cleavage assay indicated that the catalytic activity of Sharkey was 4-5 fold higher than that of the original cleavage domain.
  • a mammalian cell-based assay showed a 3 ⁇ 6 fold improvement in mutagenesis stimulation for ZFNs containing the Sharkey cleavage domain.
  • Sharkey was compatible with published hetero-dimer architectures.
  • Plasmid construction Plasmid pi 1-LacY was used.
  • the P3.F recognition sequence was PCR amplified using the primers 5'-
  • CTATATTACCCTGTTATCCCTAG-3 ' (SEQ ID NO: 16) to generate pl l-LacY-sE6/P3.
  • an additional expression cassette was PCR amplified from pROLar.A322 and cloned into the Nsil site of l l-LacY- sE6/P3-AXbaI.
  • the gene E6.FDC9-3 (D483R) was then cloned into the ne expression cassette to generate the final reporter plasmid.
  • the expression plasmid pPDAZ was constructed by first removing nucleotides 5-33 from pPRALar.A322 (Clontech Laboratories, Inc.) to generate pPROLar.del.ara. This modification abolished arabinose control over protein expression.
  • the zeocin resistance gene was next PCR amplified from pcDNA3.2/zeo(-) and cloned into pPROLar.del.ara with Aatll and Sad to form pPDAZ.
  • Transformed cells were recovered in SOC at 37°C for 1 hr before plating on solid media containing 25 ng/mL zeocin and 10 rnM arabinose. Following 9 rounds of selection, the start codon ATG was replaced with GTG and the recovery time following electroporation was increased to 3 hr. Subsequent rounds of selection saw a decrease in recovery time.
  • L1POFECT AMINE 2000 transfection reagent (Invitrogen) under conditions specified by the manufacturer.
  • cells were co-transfected with 100 ng of each ZFN expression plasmids and 400 ng pcDNA3.
  • l/Zeo(-) carrier DNA Trans fection efficiencies were measured to be between 70-80%.
  • 3 days post transfection 30,000 cells were analyzed by flow cytometry (FACScan Dual Laser Flow Cytometer, BD Biosciences) to measure the percentage of EGFP positi ve cells. Additionally, the rate of mutagenesis was measured by Mini cleavage.
  • HT1080 cells in 24-weil plates were transfected with 100 ng of ZFN or I Seel expression plasmid or the empty expression vector p VAXl (Invitrogen) plus 500 ng carrier DNA using LIPGFECTAMINE 2000 transfection reagent (Invitrogen). 30 hours post transfection, cells were harvested, stained using the H2A.X phosphorylation assay kit (Miliipore) according to the manufacturer's protocol and analyzed by FAGS. Alternatively, cells were treated with etoposide at indicated concentrations for 60 min 2h before staining. Etoposide and the reported toxic ZF construct GZF3.w ⁇ 28 were used as positive controls, I Seel and pVAXl were negative controls,
  • the DNA cleavage rate of a mutant will influence the rate of step two and, therefore, the time to linearization of the toxic gene.
  • the survival rate (SR) of a mutant the ratio of the number of colonies on an arabinose selection plate to that on a non-selective plate, was positively correlated with its catalytic activity. Mutants with higher catalytic activity linearize all reporter plasmids in an E. co!i cell within a shorter time window and will be enriched during evolution (Figure lc).
  • FIG. 1 Schematic representation of the selection strategy used for isolating novel FCD variants.
  • (a,b) A two-piasmid approach utilizing a reporter consisting of a single ZFN cleavage site downstream of ccdB and a ZFN expression plasrnid under tigh t control of a modified lac promoter can be used to selectively enrich for catalytically improved Fokl cleavage domains,
  • (c) A library of FCD variants can be transformed into the ccdB harboring BW25141 selection strain and enriched following ZFN mediated reporter plasrnid
  • Figure 6 Sequences of nuclease constructs, a) The complete amino acid sequence of the P3.wt construct used in protein evolution and the E6.wt construct used in the mutagenesis assay. The recognition a-helices are underlined, b) The amino acid sequences of the Fokl cleavage domain, Sharkey, Sharkey D483R and Sharkey DAMQS (SEQ ID NG:29). Amino acids 384 to 579 of the full-length Fokl were used as the cleavage domain. Differences between wild-type and other variants are underlined. Differences in Sharkey relative to wt are in red. Mutations unique to heterodimers are in blue.
  • round 9 displayed an SR of over 50% at the 1-hour time point, higher selection stringency was required for further evolution.
  • One way of increasing stringency is to decrease the level of protein expression.
  • the initiation codon of a transcript has a direct impact on its translation efficiency. Changing the start codon from the most frequently used ATG to GTG can reduce protein translation level by five-fold.
  • This strategy was tested on clone L9-3 isolated from the 9 lh round and it was found that the change in the start codon reduced the SR of this clone from -80% to about 8%.
  • a secondary library was then constructed based on the pool of ZF s from the 9 m round via error prone PCR with the initiation codon switched to GTG, IPTG was added during the 10 th round of evolution with this secondary library.
  • FIG. 1 Enhanc ng the Fokl cleavage domain by directed evolution.
  • a library of ZFNs was transformed into selection strain BW25141 and subjected to multiple rounds of evolution,
  • (c) The extent of substrate linearization from Rounds 1-5 was measured from cellular extracts prepared from overnight cultures. 'Sub' indicates supercoiled substrate plasmid pSub-P3. 'Prod' indicates linearized substrate plasmid pSub-P3.
  • FCD mutants For a more direct comparison of FCD mutants, the FCD of the 18 lh round was re-amplified, placed it back into the original framework and screened for optimal performance in vivo.
  • One of the most active catalytic domains termed Sharkey (S418P, I 432! ., K441E, Q481 H, H523Y, N527D, K559Q) was selected for further
  • Rates of DN A cleavage were determined using a constant concentration of ZF s (12 nM) and increasing substrate concentrations (from 4 to 36 nM).
  • Linearized piasmid pSub-P3 DNA with a singl e P3.FN recognition site positioned in the middle of the DNA molecule was used as a substrate. Cleavage of this substrate generates two product DNA molecules of the same size, simplifying the analysis.
  • the progress of each reaction was monitored over time measuring the initial velocity ( Figure 3a).
  • the rate of cleavage for Sharkey was 4-5 fold higher than that of wt o , demonstrating that Sharkey had enhance catalytic activity relative to the wt ZFN.
  • Sharkey was also observed to have a faster turnover rate than wt o/d (Figure 3b).
  • FIG. 3 Sharkey has an enhanced catalytic profile as demonstrated by in vitro DNA cleavage, (a) In vitro cleavage of target DNA by P3 nuclease with either Sharkey or wtFokl catalytic domain. 'Uncut' indicates supercoiled substrate piasmid pSub-P3. 'Cut' indicates linearized substrate piasmid pSub-P3. Cleavage was monitored incrementally over 90 min.
  • the recognition site for E2C nuclease (Table I) was inserted into the gene encoding EGFP and subsequently disabled with a franieshift.
  • the resulting nonfunctional transgene was stably integrated at a single location in the genome of HE 293 cells using the Flp-IN system ( Invitrogen).
  • Certain deletions (e.g., 2, 5, or 8-bp) or insertions (e.g., 1, 4, or 7-bp) caused by NHEJ mediated mutagenesis will restore the frame and hence restore EGFP function (Figure 4).
  • This assay should reflect a small portion of the total mutation e vents (about 1/3), but has advantages of no background, robustness and high throughput with little background.
  • the rate of mutagenesis was measured by MM cleavage assay. If the 6-bp spacer sequence between two ZFP binding sites was that of a Mlul restriction site, any mutations in this spacer region would abolish cleavage by Mlul and could be evaluated with a limited-cycle PCR/restriction digest assay ( Figure 4).
  • FIG. 4 Sharkey increases the rate of mutagenesis in a mammalian model system
  • (a) Schematic overview of the reporter system used to evaluate the efficiency of mutagenesis in mammalian cells.
  • the model system consists of a HEK 293 cel l line containing a modified and disabled EGFP transgene stably integrated in a single locus.
  • An Mlul restriction site flanked by E2C nuclease recognition sites was insetted between EGFP residues 157 and 158.
  • Figure 7 ⁇ - ⁇ 2 ⁇ based cytotoxicity assay.
  • HT1080 cells in 24-well plates were transfected with 100 ng of ZFN or I Seel expression plasmid or the empty expression vector pVAXl (Invitrogen) plus 500 ng carrier DNA using Lipofectamine 2000. 30 hours post transfection, cells were harvested, stained using the H2A.X phosphorylation assay kit
  • FCD9-3 S418P, K448E, H523Y, N527D, R570D
  • S418P, K448E, H523Y, N527D, R570D The best clone from the 9* round, FCD9-3 (S418P, K448E, H523Y, N527D, R570D), was selected as a starting point, as Sharkey had not yet been evolved when these experiments were begun.
  • ZFN E6,FCD9-3(D483R) was cloned into the reporter plasmid. Positions 483 to 487 of the P3.FCD9-3 in the expression plasmid were NNK randomized to generate the library. Following three rounds of selection, aspartic acid was observed to be the consensus residue at position 483. A smaller library with D483 fixed and positions 484 to 487 NNK randomized was then generated.
  • ZFN dimerization variants consisting of wild-type and Sharkey cleavage domains were determined.
  • FCD mutant with enhanced catalytic activity relative to the wild-type domain, as demonstrated by bacterial genetic assays, in vitro DNA cleavage assays, and targeted mammalian genome mutagenesis assays (Fig. 2-4).
  • a 77-bp sequence containing the E4JFN recognition site was inserted in between EGFP residues 157 and 158, with the C-terminal part of the coding sequence out of frame ( Figure 9).
  • Introducing cell permeable ZFN leads to cleavage at the sequence in between two ZFP binding sites.
  • Certain types of NHEJ-mediated insertions or deletions restore the reading frame and EGFP function. Mutations between the two ZFP binding sites will also destroy the M site ( Figure 1 1).
  • ZFN glycerol stock was diluted with DMEM with or without 10% FBS. Reporter cells were treated with E4.FN in concentrations as indicated for 3 hours 3 days before FACS analysis ( Figure 10). We discovered the unexpected finding that E4.FN lacking fusion or conjugation to a protein transduction domain (PTD) was able to enter cells directly and modify the target gene by cleavage.
  • PTD protein transduction domain
  • ZFN E4.FN action in cells restores the reading frame of the EGFP gene resulting in enhanced fluorescence in a significant portion of cells exposed to the E4.FN protein, This result is similar to results obtained by fusion of E4.FN to the well-known TAT or polyarginine PTDs (Snyder & Dowdy, Expert Opin Drug Deliv 2(1):43-51, 2005), Other ZFNs also lacking fusion or conjugation to PTDs were also active in entering living cells and mutating targeted nucleic acids.
  • the unexpected finding of the direct acti vity of ZFNs applied as proteins to enter cells directly upon application without the assistance of a PTD allows for their direct application to cells for targeted nucleic acid modification.
  • ZFNs to stimulate cleavage and disrupt expression of the HTV-1 co-receptor CCR5.
  • ZFNs designed to target the CCR5 locus were cloned into an expression vector and genetically fused to an N-terminai polyhistidine tag. These enzymes were expressed in E. coii and purified to >90% homogeneity by column chromatography (Fig. 4B). Importantly, we have optimized these conditions such that protein re-folding or dialysis is not necessary to restore enzyme activity.
  • rZFNs are cell-permeable and can stimulate genomic cleavage in the context of the human cell, we incubated rZFNs with HEK 293 reporter cells.
  • these cells have been transformed to contain a nonfunctional EGFP transgene disabled with a ZFN target- site derived from the CCR5 gene.
  • ZFN-mediated mutagenesis of the target allele results in the restoration of EGFP fluorescence.
  • Fig, 4C transient transfection
  • recombinant ZFNs may be used to stimulate mutagenesis in the context of the human genome and the FGFR3 locus.
  • rZFNs may further expand the utility of ZFNs as reagents for routine stem ceil modification.
  • FIG. 13 Recombinant ZFNs can efficiently stimulate mutagenesis in the human genome.
  • A Electrostatic potential map of the ZFN surface. Positively charged residues are depicted blue. Negatively charged residues are depicted red.
  • (13) SDS-PADE analysis of purified rZFN consisting of the native Fokl (wt) and the Sharkey (Sh) cleavage domain.
  • C-D Flow cytometry analysis of HEK 293 cells following transduction with (C) medium and (D) rZFN.
  • Effective ZFNs are cloned into an expression vector and genetically fused to an N- terminal poiyhistidine tag. As described above, these ZFNs will be expressed in E. coli and purified to homogeneity by column chromatography. To ensure that rZFNs can stimulate mutagenesis against the FGFR3 locus, we will analyze enzyme activity in vitro using the EGFP reporter assay described in Aim 1. Fibroblast cells derived from an achondroplasia patient will be obtained from the Coriell Cell Repositories (CCR) at the Coriell Institute for Medical Research (Catalog ID: GM0885S). These cells will be heterozygous for the G380R amino acid mutation in the transmembrane domain of FGFR3.
  • CCR Coriell Cell Repositories
  • GM0885S Coriell Institute for Medical Research
  • Fibroblast cells will be cultured in Eagle's Minimum Essential Medium (MEM) in the presence of Earle's salts and nonessential amino acids.
  • MEM Eagle's Minimum Essential Medium
  • rZFN and donor plasmid will be pre -mixed and internalized concurrently.
  • fibroblast cells will be washed to remove surface-bound protein.
  • the efficiency and specificity of ZFN- induced modification of FGFR3 will be analyzed by a variety of methods including limited- cycle PGR.
  • genomic PCR products will be used to quantitatively determine if rZFN-driven gene editing results in modification of sequences other than the intended target.
  • Cellular toxicity resulting from non-specific DSBs will he addressed by monitoring the phosphor lation of ⁇ 2 ⁇ . in the event that donor plasmid is unable to be internalized with rZFN, we will use a biodegradable iianoparticulate polymeric vector to introduce donor plasmid after incubation with rZFN44.
  • the iPSC-based models of human achondroplasia described herein has utility in high-throughput screening applications aimed at identifying novel compounds that can be used to treat skeletal dysplasia.
  • these iPSC-based models will permit analysis of the efficacy of thousands of complex small molecule compounds at various development stages during chondrogenesis and bone development.
  • these methods will allow the generation of iPSC-based models of different types of skeletal dy splasia including
  • hypochondroplasia and thanatophoric dysplasia will provide further insight into the mechanisms controlling chondrocyte differentiation, bone development and disease pathophysiology.

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Biophysics (AREA)
  • Microbiology (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Toxicology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Bioinformatics & Computational Biology (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

La présente invention porte sur des nucléases en doigt de zinc ayant une activité catalytique modifiée et améliorée, et en particulier aride, et sur des procédés de production de telles nucléases. Par conséquent, l'invention porte sur des procédés qui permettent d'identifier une activité catalytique améliorée d'une ZFN par l'expression d'une nucléase en doigt de zinc mutée dans une cellule contenant un produit de construction rapporteur avec un gène toxique et un site de clivage par une nucléase en doigt de zinc qui est reconnu par la ZFN. La survie de la cellule est corrélée positivement avec l'activité catalytique de la ZFN ; ainsi, des bibliothèques de ZFN mutées peuvent être sélectionnées d'après leur activité catalytique modifiée sur la base de taux de survie relatifs. L'invention porte également sur des procédés d'utilisation de ZFN identifiées.
EP11735277.3A 2010-01-22 2011-01-21 Procédés de production de nucléases en doigt de zinc ayant une activité modifiée Withdrawn EP2526199A4 (fr)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US29759810P 2010-01-22 2010-01-22
US32903510P 2010-04-28 2010-04-28
PCT/US2011/022154 WO2011091324A2 (fr) 2010-01-22 2011-01-21 Procédés de production de nucléases en doigt de zinc ayant une activité modifiée

Publications (2)

Publication Number Publication Date
EP2526199A2 true EP2526199A2 (fr) 2012-11-28
EP2526199A4 EP2526199A4 (fr) 2013-08-07

Family

ID=44307625

Family Applications (1)

Application Number Title Priority Date Filing Date
EP11735277.3A Withdrawn EP2526199A4 (fr) 2010-01-22 2011-01-21 Procédés de production de nucléases en doigt de zinc ayant une activité modifiée

Country Status (2)

Country Link
EP (1) EP2526199A4 (fr)
WO (1) WO2011091324A2 (fr)

Families Citing this family (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102660642A (zh) * 2012-04-24 2012-09-12 西北农林科技大学 一种筛选锌指蛋白的筛选***
EP3289104B1 (fr) 2015-04-29 2020-11-04 New York University Procédé pour le traitement de gliomes de haut grade
US20210047633A1 (en) * 2017-03-29 2021-02-18 Editas Medicine, Inc. Selection methods
WO2018227114A1 (fr) 2017-06-09 2018-12-13 Editas Medicine, Inc. Nucléases cas9 modifiées
CN108118059B (zh) * 2017-12-30 2021-03-19 苏州金唯智生物科技有限公司 一种改进的启动子及其组成的载体和应用
US20220403553A1 (en) * 2019-09-12 2022-12-22 Glaxosmithkline Intellectual Property Development Limited Method for screening libraries
AU2021263745A1 (en) 2020-04-28 2022-12-08 Intellia Therapeutics, Inc. Methods of in vitro cell delivery
US20230416709A1 (en) * 2020-11-06 2023-12-28 Editforce, Inc. Foki nuclease domain mutant
MX2023007465A (es) 2020-12-23 2023-08-18 Intellia Therapeutics Inc Composiciones y metodos para modificar geneticamente ciita en una celula.
CR20230320A (es) 2020-12-23 2023-10-23 Intellia Therapeutics Inc Composiciones y métodos para reducir los hla-a en una célula
AU2021411521A1 (en) 2020-12-30 2023-08-03 Intellia Therapeutics, Inc. Engineered t cells
AU2022258733A1 (en) 2021-04-17 2023-11-30 Intellia Therapeutics, Inc. Inhibitors of dna-dependent protein kinase and compositions and uses thereof
EP4322920A1 (fr) 2021-04-17 2024-02-21 Intellia Therapeutics, Inc. Compositions de nanoparticules lipidiques
BR112023021445A2 (pt) 2021-04-17 2024-01-23 Intellia Therapeutics Inc Composições de nanopartículas lipídicas
WO2023205148A1 (fr) 2022-04-19 2023-10-26 Intellia Therapeutics, Inc. Compositions de récepteurs antigéniques chimériques et utilisations
TW202409271A (zh) 2022-06-16 2024-03-01 美商英特利亞醫療公司 用於減少細胞中之mhc 1類之組成物及方法
WO2024006955A1 (fr) 2022-06-29 2024-01-04 Intellia Therapeutics, Inc. Lymphocytes t modifiés

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007139898A2 (fr) * 2006-05-25 2007-12-06 Sangamo Biosciences, Inc. Demi-domaines résultant de clivage par génie génétique

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BRPI0307383B1 (pt) * 2002-01-23 2019-12-31 The Univ Of Utah Research Foundation método de recombinação genética direcionada em célula de planta hospedeira
WO2006121703A2 (fr) * 2005-05-06 2006-11-16 The Board Of Trustees Of The University Of Illinois Cartographie de nouveaux sites pour action antibiotique dans le ribosome
EP2568048A1 (fr) * 2007-06-29 2013-03-13 Pioneer Hi-Bred International, Inc. Procédés de modification du génome d'une cellule de plante monocotylédone

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007139898A2 (fr) * 2006-05-25 2007-12-06 Sangamo Biosciences, Inc. Demi-domaines résultant de clivage par génie génétique

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
"Structure-Based Engineering of the FokI DNA Cleavage Domain in the Context of a Chimeric Endonuclease: Towards Maximally Efficient ''Gene Editing'' Therapy", MOLECULAR THERAPY, NATURE PUBLISHING GROUP, GB, vol. 9, 1 May 2004 (2004-05-01), page 272, XP004635080, ISSN: 1525-0016 *
JING GUO ET AL: "Directed Evolution of an Enhanced and Highly Efficient FokI Cleavage Domain for Zinc Finger Nucleases", JOURNAL OF MOLECULAR BIOLOGY, vol. 400, no. 1, 4 May 2010 (2010-05-04), pages 96-107, XP055065197, ISSN: 0022-2836, DOI: 10.1016/j.jmb.2010.04.060 *
MANI M ET AL: "Design, engineering, and characterization of zinc finger nucleases", BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS, ACADEMIC PRESS INC. ORLANDO, FL, US, vol. 335, no. 2, 23 September 2005 (2005-09-23), pages 447-457, XP027230194, ISSN: 0006-291X, DOI: 10.1016/J.BBRC.2005.07.089 [retrieved on 2005-08-11] *
See also references of WO2011091324A2 *
URNOV F D ET AL: "Highly efficient endogenous human gene correction using designed zinc-finger nucleases", NATURE: INTERNATIONAL WEEKLY JOURNAL OF SCIENCE, NATURE PUBLISHING GROUP, UNITED KINGDOM, vol. 435, no. 7042, 2 June 2005 (2005-06-02), pages 646-651, XP002411069, ISSN: 0028-0836, DOI: 10.1038/NATURE03556 *

Also Published As

Publication number Publication date
WO2011091324A8 (fr) 2012-08-30
EP2526199A4 (fr) 2013-08-07
WO2011091324A3 (fr) 2011-12-22
WO2011091324A2 (fr) 2011-07-28

Similar Documents

Publication Publication Date Title
WO2011091324A2 (fr) Procédés de production de nucléases en doigt de zinc ayant une activité modifiée
US20210403861A1 (en) Nucleotide-specific recognition sequences for designer tal effectors
JP6745391B2 (ja) 遺伝子操作CRISPR−Cas9ヌクレアーゼ
Guo et al. Directed evolution of an enhanced and highly efficient FokI cleavage domain for zinc finger nucleases
AU2006272634B2 (en) Targeted integration and expression of exogenous nucleic acid sequences
AU2005220148B2 (en) Methods and compostions for targeted cleavage and recombination
EP2927318B1 (fr) Procédés et compositions pour clivage et recombinaison ciblés
KR102271292B1 (ko) Rna-안내 게놈 편집의 특이성을 증가시키기 위한 rna-안내 foki 뉴클레아제(rfn)의 용도
US8349810B2 (en) Methods for targeted cleavage and recombination of CCR5
US20120329067A1 (en) Methods of Generating Zinc Finger Nucleases Having Altered Activity
KR20220054434A (ko) 신규한 crispr dna 표적화 효소 및 시스템
AU2012245168B2 (en) Targeted Integration and Expression of Exogenous Nucleic Acid Sequences
AU2007201649B2 (en) Methods and Compositions for Targeted Cleavage and Recombination
Guo Directed Evolution and Engineering of Zinc Finger Nucleases for Targeted Genome Modification

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20120813

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

DAX Request for extension of the european patent (deleted)
A4 Supplementary search report drawn up and despatched

Effective date: 20130710

RIC1 Information provided on ipc code assigned before grant

Ipc: C12N 9/14 20060101ALI20130704BHEP

Ipc: C12Q 1/34 20060101AFI20130704BHEP

Ipc: C12N 15/55 20060101ALI20130704BHEP

Ipc: C07K 14/47 20060101ALN20130704BHEP

Ipc: C12N 15/10 20060101ALN20130704BHEP

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION HAS BEEN WITHDRAWN

18W Application withdrawn

Effective date: 20140917