专利摘要:
target genomic alteration. disclosed herein are methods and compositions for target integration and / or target excision of one or more sequences in a cell, for example, for the expression of one or more polypeptides of interest.
公开号:BR112012018249A2
申请号:R112012018249
申请日:2011-01-24
公开日:2020-02-04
发明作者:Zeiter Bryan;Urnov Fyodor;G Murray Michael;M Ainley William
申请人:Dow Agrosciences Llc;Sangamo Biosciences Inc;
IPC主号:
专利说明:

Invention Patent Descriptive Report for AMENDMENT
TARGET GENOMICS.
Cross Reference to Related Orders
This claim claims the benefit of the US Interim Order
61 / 336,457, filed on January 22, 2010, the disclosure of which is incorporated herein by reference in its entirety.
Declaration of Rights to Inventions Made under Research Sponsored by the Federal Government
Not applicable.
Technical Field
The present disclosure refers to the field of genomic engineering, particularly the target integration and / or target excision of one or more exogenous sequences in the genome of a cell.
Background 15 Biotechnology has emerged as an essential tool in efforts to meet the challenge of increasing global demand for food production. Conventional approaches to improving agricultural productivity, for example, improved yield or projected pest resistance, rely on either reproduction with mutation or the introduction of new genes into the genomes of plant species by transformation. Both processes are inherently non-specific and relatively inefficient. For example, conventional plant transformation methods distribute the exogenous DNA that integrates into the genome at random locations. Thus, in order to identify and isolate transgenic strains with desirable attributes it is necessary to generate thousands of unique random integration events and subsequently to screen for the desired individuals. As a result, conventional plant trace engineering is a laborious, time-consuming, and unpredictable task. In addition, the random nature of these integrations makes it difficult to predict whether pleiotropic effects due to unintended genome disturbance have occurred. As a result, the generation, isolation and characterization of plant strains with projected traits or genes has been an extreme and cost-intensive work process with a low probability of success.
The modification of the target gene overcomes the logistical challenges of conventional practices in plant systems, and as such has been a long-standing but elusive objective, both in research on basic plant biology and agricultural biotechnology. However, with the exception of the selection of positive-negative drugs via the target gene in rice or the use of pre-designed restriction sites, the modification of the target genome in all plant species, both the model and the crops, has until recently if 10 shown very difficult. Terada et al. (2002) Nat Biotechnol 20 (10): 1030; Terada et al. (2007) Plant Physiol 144 (2): 846; D'Halluin et al. (2008) Plant Biotechnology J. 6 (1): 93.
In mammalian cells, stable transgenesis and insertion of the target gene have many potential applications in both gene therapy and cell engineering. However, current strategies are often inefficient and do not specifically insert the transgene into genomic DNA. The inability to control the insertion site of the genome can lead to extremely variable levels of expression of the transgene in the entire population due to the effect of position within the genome. In addition, current methods of stable transgenesis and amplification of transgenes often result in physical loss of the transgene, silencing of the transgene over time, insertional mutagenesis through the integration of a gene and autonomous promoter within or adjacent to an endogenous gene, the creation of chromosomal anomalies and expression of rearranged gene products 25 (composed of endogenous genes, inserted transgenes, or both), and / or the creation of vector-related toxicities or in vivo immunogenicity from genes derived from the vector that are expressed permanently, due to the need for long-term persistence of the vector to provide stable transgene expression.
Recently, methods and compositions for the target cleavage of
Genomic DNA have been described. Such target cleavage events can be used, for example, to induce target mutagenesis, induce deletions3 / 88 targets of cellular DNA sequences, and facilitate target recombination to a predetermined chromosomal locus. See, for example, US Patent Publications 20030232410; 20050208489; 20050026157; 20050064474; and 20060188987, and International Publication WO 2007/014275, the disclosures of which are incorporated by reference in their entirety for all purposes. US Patent Publication 20080182332 describes the use of non-canonical zinc finger nucleases (ZFNs) for specific modification of plant genomes and US Patent Publication 20090205083 describes ZFN-mediated target modification of a plant EPSPS locus. In addition, Moehle et al. (2007) Proc. Natl. Acad, Sei. USA 104 (9): 3055-3060) describe the use of designated ZFNs for adding target gene to a specified locus.
However, there is still a need for compositions and methods for target integration, including target integration in plants for stable establishment, heritable genetic modifications in the plant and its progeny, and for target integration in mammalian cells for gene therapy and for cell line development purposes.
summary
The present disclosure provides methods and compositions for expressing one or more products of an exogenous nucleic acid sequence (i.e., a protein or an RNA molecule) that has been integrated into a multiple insertion site integrated into a cell genome. The cell can be a eukaryotic cell, for example, a plant, yeast or mammalian cell.
The integration of exogenous nucleic acid sequences is facilitated by the integration of the genome of a polynucleotide sequence comprising multiple target sites for one or more nucleases, for example, zinc finger nucleases (ZFNs) in the cell genome. Polynucleotides (also referred to here as a multiple insertion site) allow specific target double-strand divination within the cell genome, whose double-strand divination, in turn, results in the integration of se4 / 88 sequence (s) exogenous (s) through homology-dependent and homology-independent mechanisms.
Thus, in one aspect, nucleic acid molecules, also known as multiple insertion sites, are disclosed herein, comprising one or more target sites for nucleases, such as zinc finger nucleases (ZFNs). In certain embodiments, the target sites are not present in the endogenous genome into which the multiple insertion site is integrated. The multiple insertion site can include one, two, three, four, five, six, seven or more target sites for nucleases. In certain embodiments, dimerization of the dividing half-domains of two binding DNA binding proteins that bind to adjacent target sites (paired target sites) is necessary for divage (for example, a pair of nucleases, a link for each location, it is necessary for divagem). At any of the multiple insertion sites described here, a target site from each pair of target sites can comprise the same sequence. See, for example, Figure 1. In certain embodiments, the target sites of at least one pair are the same. In other embodiments, at least a pair of target sites comprise individual target sequences from different targets (for example, different genes and / or genes from different organisms). In certain embodiments, at least one of the paired target sites comprises a sequence selected from the group consisting of SEQ ID NOs: 1-20. In certain embodiments, the multiple insertion site may include one or more coding sequences, for example, a plant transcription unit (PTU) comprising a phosphinothricin-acetyl transferase (PAT) transferase coding sequence, or a screening marker for use with mammalian cells.
Multiple insertion sites are integrated into the genome of a cell (eg, plant or mammalian cell) to provide genomic targets for nucleases (eg, ZFNs). In certain embodiments, the target sites are located in such a way that one or more pairs of zinc finger nucleases bind and cleave as homodimers. In other modalities, the target sites are located in such a way that one or
5/88 more pairs of zinc finger nucleases bind and cleave as heterodimers.
In another aspect, plants or seeds comprising one or more multiple insertion sites as described herein and / or one or more exogenous sequences integrated into the multiple insertion site are disclosed herein. In certain embodiments, the multiple insertion site and / or the exogenous sequence (s) is (are) integrated into the gametophyte of a corn plant.
In certain respects, modified mammalian cell lines, modified primary cells, modified stem cells and / or transgenic animals are provided here comprising one or more multiple insertion sites, as described herein and / or one or more integrated exogenous sequences at the multiple insertion site.
In another aspect, a method is provided here for the integration of an exogenous sequence for the multiple insertion site integrated into the genome of a cell (for example, plant or mammalian cell), the method comprising: (a) integrating a polynucleotide of multiple insertion site comprising one or more target sites for nucleases in the cell genome, (b) providing and / or expressing one or more nucleases that bind to a first target site on the multiple insertion site polynucleotide, such that the binding of the nuclease (s) to its target sites cleaves the cell's genome, and (c) contacting the cell with a polynucleotide comprising a sequence of exogenous nucleic acids, thus resulting in homology-dependent integration of the exogenous sequence into the cell genome within the multiple insertion site polynucleotide.
In another aspect, a method is provided here for integrating multiple exogenous sequences into the genome of a cell (for example, a plant or mammalian cell), the method comprising:
(a) integrating a first multiple insertion site polynucleotide comprising one or more target sites for nucleases in the cell genome, wherein the first multiple insertion site polynucleotide comprises at least one first gene flanked by target sites for the
6/88 first and second nucleases, and (b) expressing the first or second nuclease in the cell in the presence of a second multiple insertion site polynucleotide comprising at least a second gene flanked by target sites for third and fourth nucleases, thus resulting in integrating the first and second genes into the cell's genome. In certain embodiments, the method further comprises repeating, one or more times, the step of expressing suitable nucleases present at the inserted multiple insertion sites to integrate the additional exogenous sequences, including coding sequences and / or nuclease sites. The nucleases can be heterodimeric ZFNs and there may be a monomer in common between one or more of the nucleases. In some embodiments, the exogenous DNA sequence for insertion may comprise one half of the ZFN target site in such a way that upon integration of the exogenous sequence, a new ZFN target site is created comprising the associated half of the target site with donor DNA, and half a target site associated with genomic DNA. This new ZFN target site can similarly serve as a target site for a new heterodimeric ZFN.
In another aspect, a method for expressing the product of one or more exogenous nucleic acid sequences in a cell (e.g., plant or mammalian cell) is described herein, the method comprising: integrating one or more exogenous nucleic acid sequences according to any of the methods described herein, such that the exogenous sequence is integrated into the cell genome in the integrated nucleic acid molecule and the product of the exogenous sequence is expressed.
Also provided is a method of deleting one or more genes inserted into a cell's genome, the method comprising, integrating a plurality of exogenous sequences by any of the methods described herein and expressing the appropriate nucleases in the cell such that one or more of exogenous sequences are deleted from the genome. In certain embodiments, exogenous sequences are deleted from marker genes. In certain modalities, the deletion of the exogenous sequence and the
7/88 Subsequent rejection of the ends within the genome creates a gene or functional sequence at the genomic site, for example, the creation of an expressable screening marker.
In yet another aspect, a method of providing a genomically altered cell is provided, the method comprising integrating and / or excising one or more sequences of exogenous nucleic acids in a first cell according to any of the methods described herein, allowing the first cell develops into a first sexually mature organism, traversing the organism with a second organism comprising genomic changes in an allelic position to generate a second cell with the first and second organism genomic changes. In certain embodiments, the organism (s) is (are) the plant. In other modalities, the organism (s) is / are transgenic animals.
In any of the methods described herein, the methods can be used in combination with other methods of genomic alteration, including target integration and / or target inactivation to one or more endogenous loci. In addition, in any of the methods described herein, the nuclease may comprise one or more fusion proteins comprising a zinc finger binding domain and a half diving domain, wherein the zinc finger binding domain is designed to connect to a target site on the multiple insertion site. In addition, in any of these methods, the exogenous nucleic acid sequence comprises one or more sequences that are (are) homologous to the sequences at the multiple insertion site and / or endogenous sequences in the region where the multiple insertion site is integrated.
In any of the methods described herein, the one or more multiple insertion sites can be integrated into the genome by any suitable method, for example, by target integration via a nuclease (for example, ZFN) using ZFNs that target the endogenous gene in which the insertion is desired. Alternatively, the one or more multiple insertion sites can be randomly integrated into the cell's genome, using standard techniques.
8/88
The exogenous nucleic acid sequence may comprise a sequence that encodes one or more functional polypeptides (for example, a cDNA), with or without one or more promoters, and / or can produce one or more RNA sequences (for example, through a or more shRNA expression cassettes), which provide desirable traits for the organism. Such traits in plants include, but are not limited to, herbicide resistance or tolerance; insect resistance or tolerance; resistance or tolerance to diseases (viral, bacterial, nematodes, fungal); tolerance and / or resistance to stress, as exemplified by resistance or tolerance to drought, heat, refrigeration, freezing, excessive humidity, saline stress, oxidative stress, increased yields, constitution and food content; physical appearance, male sterility; drydown; verticality; prolificacy; quantity and quality of starch; quantity and quality of oil; protein quality and quantity; amino acid composition, and the like. Naturally, any two or more exogenous nucleic acids of any description, such as those that confer herbicide, insects, disease (viral, bacterial, nematode, fungal) or resistance to dryness, male sterility, gradual and controlled dehydration (drydown), verticality, prolificacy, starch properties, and oil quality and quantity, or those that increase yield or nutritional quality can be employed as desired. In certain embodiments, the exogenous nucleic acid sequence comprises a sequence that encodes an herbicide-resistant protein (for example, the AAD (aryloxyalkanoate dioxigenase) gene) and / or functional fragments thereof. Expression of the integrated sequence can be driven by a promoter operably linked to the integrated sequence. Alternatively, the integrated sequence is without a promoter and transcription is conducted by the endogenous promoter in the insertion region of the multiple insertion site. In other embodiments, imprecise repair and divage of a binding site can inactivate or activate genes of interest. In certain embodiments, the polynucleotide is a plasmid. In other embodiments, the polynucleotide is a linear DNA molecule.
In mammalian cells, the methods and compositions of the invention can be used for the construction of cell lines, for example, for the construction of cell lines that express multimeric polypeptides, such as antibodies. In some embodiments, cell lines can be used for research purposes, for example, to build cell lines that express members of a pathway of interest. In some embodiments, primary cells or stem cells can be used to express the multimeric proteins of interest to cells for therapeutic purposes.
In another aspect, methods for measuring zinc finger nuclease activity are provided here. In certain embodiments, the methods comprise: (a) providing at least one zinc finger nuclease and a nucleic acid molecule as described herein, wherein each paired target site comprises two half finger nuclease target sites of zinc to which the zinc finger nuclease attaches, and a cutting site that is cut by the attached zinc finger nuclease, in which the cutting site is interposed between the target medium means, (b) combining the nuclease zinc finger with nucleic acid such that the zinc finger nuclease cleaves the paired target site at least within the cut site, (c) sequence at least the cut site to generate sequence data, and (d) , compare in the sequence data, the number and length of base pair deletions within the cut site for the number and length of base pair deletions within the cut site in the absence of the zinc finger nuclease so that so measure naked activity zinc finger clease at the paired target sites. In certain embodiments, a deletion of more than one base pair indicates increased activity of the zinc finger nuclease (s).
In still other embodiments, methods are provided here to optimize zinc finger nuclease activity at a paired target site. In certain embodiments, the methods comprise (a) providing at least one zinc finger nuclease and one nucleic acid molecule as described herein, wherein each of the target sites comprises two half zinc finger nuclease target sites to which the finger nuclease
10/88 zinc binds, and a cutting site that is cut by the linked zinc finger nuclease, the cutting site of which is interposed between the target mediums, (b) combining with one or more zinc finger nucleases with nucleic acid such that the zinc finger nuclease cleaves the target site paired by me5 within the cutting site, (c) determining the level of zinc finger nuclease activity at the cutting site; (d) varying the number of base pairs at the cutting site; (e) repeat steps (b) to (d) a plurality of times, and (f) select the cut site for incorporation into the nucleic acid, which comprises the number of base pairs that provide the highest level of activity zinc finger nuclease, thus optimizing zinc finger nuclease activity at the paired target site.
In any of the methods described herein involving zinc finger nucleases, the first and second dividing half-domains are from an IIS type restriction endonuclease, for example, Fokl or 15 Sfsl. In addition, in any of the methods described here, at least one of the fusion proteins may comprise a change in the amino acid sequence of the dimerization interface of the half domain domain, for example, such that the mandatory heterodimers of the half domain domain are formed.
20 In any of the methods described herein, the plant cell may comprise a monocot or dicot plant cell. In certain embodiments, the plant cell is a cultivation plant, for example, corn. In certain embodiments, the cell can comprise a mammalian cell such as a primary cell, a cell line, or a stem cell. In some embodiments, the mammalian cell line can be used to produce polypeptides of interest.
Brief Description of Drawings
Figure 1 is a schematic diagram representing exemplary multiple insertion sites as described herein. Figure 1 shows a multiple insertion site 30 composed of 7 ZFN target sites. The ZFN pairs that bind to the target sites are described as geometric figures. Block 1 ”is an exogenous sequence that is integrated at the insertion site
11/88 multiple, in the presence of the appropriate ZFN pair, while maintaining the target ZFN sites (shaded and checkered triangles). The Figure shows the integration of Block 1 in a multiple insertion site, in the presence of the appropriate ZFN pair in place of the ZFN target sites.
Figure 2 is a schematic diagram representing the exemplary multiple insertion site as shown in Figure 1, in which the Block is an exogenous sequence that is integrated into the multiple insertion site, in the presence of the appropriate ZFN pair.
Figure 3 is a schematic diagram of ZFN-enhanced interalelic recombination. Two inserts from an identical genomic site, but are displaced from each other, can be subjected to homologous recombination or filament exchange after double filament dividing by a ZFN. The ZFN pair (with both ZFN monomers expressed together) can be provided by crossing a plant expressing the ZFN pair with plants comprising both alleles together or by introducing two ZFN monomers from both sides crossing with plants that contain a single allele.
Figure 4 is a schematic diagram representing the use of heterodimeric ZFN on the left and target domains on the right. The top line represents the genome with the ZFN target domains on the left and right (shaded triangle and checkered triangle). When the appropriate ZFN pair is added in the presence of an exogenous molecule including a gene flanked by different heterodimeric pairs, the gene and the flanking nuclease sites are inserted into the genome as shown.
Figure 5 is a schematic diagram that represents the integration and excision of exogenous sequences (represented as genes) on both sides of a genomically integrated sequence. The added genes are flanked by regions of homology to direct the gene cassettes at the appropriate site. The two halves of the ZFN target site used for insertion are recombined by creating two new combinations in the inserted DNA. Excision of a gene cassette is achieved by ligating the appropriate ZFN pairs to cleave at the flanking ZFN target sites. Excision may require a model containing homology arms to avoid deletions of the desired DNA sequence. Each gene can include one or more sequences, for example, one or more coding sequences.
Figure 6 is a schematic diagram representing the excision and recycling of marker genes inserted using heterodimeral ZFNs (represented as triangles with different shading).
Figure 7 is a map of plasmid pDAB105900.
Figure 8 is a map of plasmid pDAB105908.
Figure 9 is a diagram of the zinc finger nuclease homodimer expression cassette.
Figure 10 is a diagram of the zinc finger nuclease heterodimer expression cassette.
Figure 11 shows the dividing activity of eZFN in maize, as determined by the frequency of deletions resulting from junctions of non-homologous ends after diving.
Figure 12 shows the dividing activity of eZFN in tobacco, as determined by the frequency of deletions resulting from the non-homologous end junction, after divating.
Figure 13 is a schematic diagram of two transgenic inserts for the same genetic locus. The top line shows the random sequence marked MIS for the multiple insertion site (also referred to here as a landing pad) containing eZFN binding sites required for homologous recombination at the locus and Block 1 comprising a selectable kanamycin marker gene and a GUS triable marker gene. The midline represents the same multiple insertion site (MIS) as in the upper DNA together with Block2 comprising a selectable marker gene for hygromycin resistance and a triable marker of yellow fluorescence protein. (HPT / YFP). The bottom line represents the locus after recombination.
Figure 14 shows the homologous recombination in a position
13/88 allele by ZFNs and the generation of the two different DNA inserts in the same genetic locus described in Figure 13. A construct including Block1 (which comprises kanamycin and GUC, GUS / NPT markers), a multiple insertion site (MIS or landing pad) and Block2 (comprising hygomycin and yellow fluorescence markers, HPT7YFP) is transformed into Arabidopsis. To generate each block individually together with the multiple insertion site in separate plants, Block2 is excised from the integrated site to generate a configuration of Block 1 only or Blocol is excised from the integrated site to generate a configuration of Block2 only . The removal of gene blocks is accomplished by crossing the plants that contain the original transgenic event with plants that express ZFNs that cleave at eZFN binding sites that flank each of the gene blocks. The recovered single-block plants are crossed to bring the two configurations together into a single plant and the plant is crossed with a plant that expresses a specific meiosis promoter to effect the DNA exchange between the two alleles of Blocol and Block2 .
Figure 15 is a schematic flowchart that represents the steps for obtaining recombination between two DNA sequences located in the same genetic locus by dividing ZFN at an intermediate site between the two sequences. The construct described in Figure 16 is transformed into Arabidopsis. One of the two gene blocks (described in Figure 14) is removed by crossing with plants expressing eZFNs whose block binding sites flank, resulting in plants containing either Blocol or Block2.
Figure 16 is a schematic diagram of the plasmid used for the introduction of the Exchange Locus in Arabidopsis. It contains Blocks 1 and 2, as described in Figure 14 and in the sequence of the multiple insertion site. EZFN binding sites are indicated and Blocks 1 and 2 flank (Blocol: eZFN1 and 8; Block2: eZFNs 3 and 6) or are centrally located at the multiple insertion site (eZFNs 4 and 7) to facilitate homologous recombination.
14/88
Detailed Description
The present disclosure relates to methods and compositions for target integration (TI) in a genome, for example, a crop plant, such as corn or a mammalian cell. A multiple insertion site containing multiple target sites for one or more nucleases (for example, ZFNs) is integrated into the genome. Following the integration of the multiple insertion site into the genome, suitable nucleases are introduced into the cell, together with an exogenous sequence to be inserted.
In certain embodiments, the nuclease (s) comprise one or more ZFNs. ZFNs typically comprise a dividing domain (or a half diving domain) and a zinc finger binding domain and can be introduced as proteins, as polynucleotides that encode these proteins, or as combinations of polypeptides and polynucleotides that encode polypeptides. Zinc finger nucleases typically function as dimeric proteins after dimerization of half dividing domains. Mandatory heterodimeric ZFNs, in which the ZFN monomers bind to the left and right of the recognition domains can associate, to form an active nuclease have been described. See, for example, US Patent Publication 2008/0131962. Thus, given the appropriate target sites, a monomer on the left could form an active ZF nuclease with any monomer on the right. This significantly increases the number of useful nuclease sites based on the proven left and right domains that can be used in various combinations. For example, recombination of the 4 homodimeric ZF nuclease binding sites yields an additional 12 heterodimeric ZF nucleases. Most importantly, this allows for a systematic approach to transgenic models so that each new sequence introduced becomes flanked by a unique ZFN site that can be used to excise the gene back or to target additional genes close to it. In addition, this method can simplify stacking strategies for a single locus that is triggered by ZFN-dependent double filament breaks.
15/88
A zinc finger binding domain can be a canonical zinc finger (C2H2) or a non-canonical zinc finger (for example, C3H). In addition, the zinc finger binding domain can comprise one or more zinc fingers (for example, 2, 3, 4, 5, 6, 7, 8, 9 or more zinc fingers), and can be designed to bind to any sequence within the multiple insertion site. The presence of such a fusion protein (or proteins) in a cell results in the binding of the fusion protein (s) to its binding site (s) and dividing within the multiple insertion site, resulting in integration of the exogenous sequence (s).
General
The practice of the methods, as well as the preparation and use of the compositions disclosed herein employ, unless otherwise indicated, conventional techniques of molecular biology, biochemistry, chromatin structure and analysis, computational chemistry, cell culture, recombinant DNA and related fields such as those within the skill of the person skilled in the art. These techniques are fully explained in the literature. See, for example, Sambrook et al. MOLECULAR CLONING: A LABORATORY MANUAL, Second edition, Cold Spring Harbor Laboratory Press, 1989 and Third edition, 2001; Ausubel et al., CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, New York, 1987 and periodic updates; the series METHODS IN ENZYMOLOGY, Academic Press, San Diego; Wolffe, CHROMATIN STRUCTURE AND FUNCTION, Third edition, Academic Press, San Diego, 1998; METHODS IN ENZYMOLOGY, Vol. 304, Chromatin (P.M. Wassarman and A. P. Wolffe, eds.), Academic Press, San Diego, 1999; and METHODS IN MOLECULAR BIOLOGY, Vol. 119, Chromatin Protocols (P.B. Becker, ed.) Humana Press, Totowa, 1999.
Definitions
The terms nucleic acid, polynucleotide and oligonucleotide are used interchangeably and refer to a deoxyribonucleotide or ribonucleotide polymer, in linear or circular conformation, and either in the form of single or double filament. For the purposes of this description, these terms should not be interpreted as limiting with respect to the
16/88 length of a polymer. The terms can encompass known analogs of natural nucleotides, as well as nucleotides that are modified in the base, sugar, and / or phosphate fractions (for example, phosphorothioate backbones). In general, an analogue of a particular nucleotide has the same base pairing specificity, that is, an analogue of A will form a base pair with T.
The terms polypeptide, peptide and protein are used interchangeably to refer to a polymer of amino acid residues. The term also applies to amino acid polymers in which one or more amino acids are modified derivatives or corresponding naturally occurring chemical amino acid analogues.
Linkage refers to a sequence-specific, non-covalent interaction between macromolecules (for example, between a protein and a nucleic acid). Not all components of a binding interaction need to be sequence specific (for example, contacts with phosphate residues in a DNA skeleton), as long as the interaction as a whole is sequence specific. Such interactions are generally characterized by a dissociation constant (Kd) of 10 6 M ' 1 or less. Affinity refers to the binding strength: increased binding affinity being correlated with a lower Kd.
The binding protein is a protein that is able to bind to another molecule. A binding protein can bind, for example, to a DNA molecule (a DNA binding protein), an RNA molecule (an RNA binding protein) and / or a protein molecule (a binding protein the protein). In the case of a protein-binding protein, it can bind itself (to form homodimers, homotrimers, etc.) and / or can bind to one or more molecules of a different protein or proteins. A binding protein can have more than one type of binding activity. For example, zinc finger proteins have DNA-binding, RNA-binding and protein-binding activity.
A zinc finger DNA binding protein (or binding domain) is a protein, or a domain within a larger protein,
17/88 that binds to DNA in a specific sequence form through one or more zinc fingers, which are regions of amino acid sequences within the binding domain whose structure is stabilized through the coordination of a zinc ion. The term zinc finger DNA binding protein is often abbreviated as zinc finger protein or ZFP.
The zinc finger binding domains can be designed to bind to a predetermined nucleotide sequence. Non-limiting examples of methods for the design of zinc finger proteins are modeling and selection. A projected zinc finger protein and 10 a non-naturally occurring protein whose model / composition results, mainly from rational criteria. Rational criteria for the model include the application of substitution rules and computerized algorithms for processing information in an information store database of existing ZFP models and binding data.
See, for example, US Patents 6,140,081; US 6,453,242; and US 6,534,261; see also WO 98/53058, WO 98/53059, WO 98/53060, WO 02 / 016.536 and WO 03 / 016.496.
A selected zinc finger protein is a protein not found in nature, the production of which results mainly from an empirical process such as phage display, interaction trap or selection of hybrids. See, for example, US 5,789,538; US 5,925,523; US 6,007,988; US 6,013,453; US 6,200,759; WO 95/19431, WO 96/06166, WO 98/53057, WO 98/54311, WO 00/27878, WO 01/60970 WO 01 / 88,197 and WO 02 / 099.084.
25 the word sequence refers to a sequence of nucleotides of any length, which can be DNA or RNA; it can be linear, circular or branched and can be either single or double filament. The donor sequence refers to a sequence of nucleotides that is inserted into a genome. A donor sequence can be of any length, for example, between 2 and 10,000 nucleotides in length (or any integer value in between or above), preferably between about 100 and 1000 nucleotides in length (or any
18/88 integer between them), more preferably between about 200 and 500 nucleotides in length.
A homologous non-identical sequence refers to a first sequence that shares a degree of sequence identity with a second sequence, but whose sequence is not identical to the second sequence. For example, a polynucleotide comprising the wild-type sequence of a mutant gene is homologous and not identical to the sequence of the mutant gene. In certain embodiments, the degree of homology between the two sequences is sufficient to allow homologous recombination between the same sequences, using normal cellular mechanisms. Two homologous non-identical sequences can be of any length and their degree of non-homology can be as small as a single nucleotide (for example, for the correction of a point genomic mutation by target homologous recombination) or as large as 10 or more kilobases (for example, for inserting a gene into a predetermined ectopic site on a chromosome). Two polynucleotides comprising non-identical homologous sequences do not have to be the same length. For example, an exogenous polynucleotide (i.e., donor polynucleotide) of between 20 and 10,000 nucleotides or pairs of nucleotides can be used.
Techniques for determining nucleic acid and amino acid sequence identity are known in the art. Typically, such techniques include determining the nucleotide sequence of the mRNA for a gene and / or determining the amino acid sequence encoded in this way, and comparing these sequences with a second nucleotide or amino acid sequence. Genomic sequences can also be determined and compared in this way. In general, identity refers to an exact nucleotide-to-nucleotide or amino acid-to-amino acid match of two polypeptide or polynucleotide sequences, respectively. Two or more sequences (polynucleotide or amino acid) can be compared by determining their percentage identity. The percentage identity of two sequences, whether nucleic acid or amino acid sequences, is the number of exact matches between two aligned sequences, divided by the length of the shortest sequences and multiplied by 100. An approximate alignment for acid sequences nucleic acids is provided by the local homology of Smith and Waterman, 2'482-489 (1981). This algorithm can be applied to sequences of amino acids using the scoring matrix developed by Dayhoff, ^^ n ^ e ^ dSVy ^. M.O. Dayhoff ed., 5 suppl 3: 353-358, National Biomedical Research Foundation. Washington. D C., USA, and normalized by Gribskov, NyçLAçidsR ^ 14 (6): 6745-6763 (1986). An exemplary implementation of the present aigoritmo for determining the percent identity of a sequence is provided by Genetics Computer Group (Madison Wl) utility application BestFif. Programs suitable for calculating the identity or percent similarity between the sequences are generally known in the art, for example. another BLAST program, used with default parameters. For example, BLASTN and BLASTP can be used with the following standard parameters: genet code, co-pattern; filter = none; filament = both; cut - 60, wait 'BLOSUM62: Descriptions = 50 strings: choose by = HIGH SCORE, Databases = non-redundant, GenBank * EMBL ♦ DDBJ + PD ♦ GenBank CDS translations f Swiss protein f Spupdate ♦ PIR. Details of these programs can be found on the internet. With respect to the sequences described herein, the range of desired degrees of sequence identity is approximately 80% to 100% and any integer value between them. Typically the percent identities between the strings are pek, minus 70-75%, preferably 80-82%. more preferably 85-90%, even more preferably 92%. even more preferably 95%. and most preferably 98% sequence identity.
Alternatively, the degree of sequence similarity between polynucleotides can be determined by pollnucleotide hybridization, under conditions that allow the development of stable duplexes between homologous regions, followed by digestion with specific single filament nuclease (s) and determination of the size of fragments digested. The aedos nu20 / 88 cleicos, or two polypeptide sequences are substantially homologous to each other when the sequences exhibit at least about 70% -75%, preferably 80% -82%, more preferably 85% -90%, even more preferably 92%, even more preferably 95%, and most preferably 98% sequence identity over a defined length of the molecules, as determined using the methods above. As used herein, substantially homologous also refers to sequences that show complete identity for a specified peptide or DNA sequence. DNA sequences that are substantially homologous can be identified in a Southern hybridization experiment, for example, stringent conditions, as defined for that particular system. The definition of suitable hybridization conditions is within the skill of the person skilled in the art. See, for example, Sambrook et al „supra; Nucleic Acid Hybridization: A Practical Approach, 15 B.D. Hames and S.J. Higgins, (1985) Oxford; Washington, DC; IRL
Press).
The selective hybridization of two nucleic acid fragments can be determined as follows. The degree of sequence identity between two nucleic acid molecules affects the efficiency and strength of hybridization events between these molecules. A partially identical nucleic acid sequence will at least partially inhibit the hybridization of a completely identical sequence to a target molecule. Inhibition of completely identical sequence hybridization can be assessed using hybridization assays that are well known in the art (for example, 25 Southern blot (DNA), Northern blot (RNA), solution hybridization, or the like, see Sambrook, et a /., Molecular Cloning: A Laboratory Manual, Second edition, (1989) Cold Spring Harbor, NY). Such tests can be performed using varying degrees of selectivity, for example, using different conditions from low to high rigor. If low stringency conditions are employed, the absence of non-specific binding can be assessed using a secondary probe that does not even have a degree of partial sequence identity (for example, a probe having
21/88 less than about 30% sequence identity with the target molecule), such that, in the absence of non-specific binding events, the secondary probe will not hybridize to the target.
When using a hybridization-based detection system, a nucleic acid probe that is complementary to a reference nucleic acid sequence is chosen, and then by selecting suitable conditions the probe and the reference sequence selectively hybridize, or if bind together to form a duplex molecule. A nucleic acid molecule that is capable of selectively hybridizing to a reference sequence under moderately stringent hybridization conditions typically hybrid under conditions that allow the detection of a target nucleic acid sequence of at least about 10-14 nucleotides in length with at least about 70% sequence identity with the selected nucleic acid probe sequence. Stringent hybridization conditions typically allow detection of nucleic acid target sequences of at least about 10-14 nucleotides in length having a sequence identity of more than about 90-95% with the selected nucleic acid probe sequence. Hybridization conditions useful for the probe / reference sequence hybridization, where the probe and the reference sequence have a specific degree of sequence identity, can be determined as is known in the art (see, for example, Nucleic Acid Hybridization : A Practical Approach, editors BD Hames and SJ Higgins, (1985) Oxford; Washington, DC; IRL Press).
Hybridization conditions are well known to those skilled in the art. Hybridization stringency refers to the degree to which hybridization conditions favor the formation of hybrids that contain incompatible nucleotides, with greater stringency correlated with a lower tolerance for incompatible hybrids. Factors affecting the stringency of hybridization are well known to those skilled in the art and include, but are not limited to, temperature, pH, ionic strength, and concentration of organic solvents, such as, for example, formamide and dimethyl sulfoxide. How is it
22/88 known to those skilled in the art, the stringency of hybridization is increased by higher temperatures, lower ionic strength and lower concentrations of solvents.
With regard to stringency conditions for hybridization, it is well known in the art that numerous equivalent conditions can be used to establish a special stringency varying, for example, the following factors: length and nature of the sequences, basic composition of the various sequences , the concentrations of salts and other components of the hybridization solution, the presence or absence of 10 blocking agents in the hybridization solutions (for example, dextran sulfate, and polyethylene glycol), the reaction hybridization temperature and the time parameters , as well as variation of washing conditions. The selection of a particular set of hybridization conditions is selected according to standard methods in the art (see, for example, Sam15 brook, et al „Molecular Cloning: A Laboratory Manual, Second edition, (1989) Cold Spring Harbor, Ν. Υ.).
Recombination ”refers to a process of exchanging genetic information between two polynucleotides. For the purposes of this disclosure, homologous recombination (HR) refers to the specialized form of exchange that occurs, for example, during the repair of double strand breaks in cells. This process requires nucleotide sequence homology, uses a donor molecule to repair a model of a target molecule (that is, one that experienced double strand breakage), and is also known as non-cross gene conversion or gene conversion from short, because it leads to the transfer of genetic information from the donor to the target. Without wishing to be bound by any particular theory, such transfers may involve correcting heteroduplex DNA incompatibility that forms between the broken target and the donor, and / or annealing the synthesis-dependent strand, in which the donor is used to resynthesize the genetic information that will become part of the target, and / or related processes. These specialized HR often result in a change in the sequence of the target molecule
23/88 such that part or all of the donor polynucleotide sequence is incorporated into the target polynucleotide.
Divage refers to the breaking of the covalent skeleton of a DNA molecule. Divage can be initiated by a variety of methods, including, but not limited to, enzymatic or chemical hydrolysis of a phosphodiester bond. Both single-stranded and double-stranded divage are possible, and double-stranded divage can occur as a result of two distinct single-stranded dividing events. DNA splitting can result in the production of either blunt ends or staggered ends. In certain embodiments, fusion polypeptides are used for dividing double-stranded DNA strands.
A divage domain comprises one or more polypeptide sequences that have catalytic activity for DNA dividing. A divage domain can be contained in a single peptide chain or the divage activity can result from the association of two (or more) polypeptides.
A half-domain domain is a peptide sequence which, together with a second polypeptide (whether identical or different) 20 forms a complex with dividing activity (preferably double-stranded dividing activity).
Chromatin is the structure of the nucleoprotein comprising the cell genome. Cell chromatin comprises nucleic acid, mainly DNA, and protein, including histone and 25 non-histone chromosomal proteins. Most eukaryotic cell chromatins exist in the form of nucleosomes, in which a nucleosome nucleus comprises about 150 base pairs of DNA associated with an octamer comprising two of each of the histones H2A, H2B, H3 and H4; and the ligand DNA (of variable length, depending on the organism) extends between the nucleosome nuclei. A histone H1 molecule is usually associated with DNA binding. For the purposes of this description, the term chromatin is intended to encompass all types of cell nucleoproteins, both prokaryotic and eukaryotic. Cell chromatin includes both chromosomal and episomal chromatin.
A chromosome is a chromatin complex comprising all or a portion of a cell's genome. The genome of a cell is often characterized by its karyotype, which is the collection of all the chromosomes that make up the cell's genome. The genome of a cell can comprise one or more chromosomes.
An episome is a replicating nucleic acid, nucleoprotein complex or other structure comprising a nucleic acid that is not part of a cell's chromosomal karyotype. Examples of episomes include plasmids and certain viral genomes.
An accessible region is a site on cell chromatin in which a target site present in the nucleic acid can be linked to an exogenous molecule that recognizes the target site. Without wishing to be bound by any particular theory, it is believed that an accessible region is one that is not packaged in a nucleosomal structure. The distinct structure of an accessible region can often be detected by its sensitivity to chemical and enzymatic probes, for example, nucleases.
A target site or target sequence is a sequence of nucleic acids that defines a portion of a nucleic acid to which a binding molecule will bind, provided that sufficient conditions for binding exist. For example, the sequence 5'-GAATTC-3 'is a target site for the restriction endonuclease Eco RI.
An exogenous molecule is a molecule that is not normally present in a cell, but can be introduced into a cell by one or more genetic, biochemical or other methods. The normal presence in the cell is determined in relation to the particular stage of development and environmental conditions of the cell. Thus, for example, a molecule that is present only during embryonic muscle development is an exogenous molecule with respect to an adult muscle cell. Likewise, a heat shock-induced molecule is an exogenous molecule with respect to a non-thermal shock cell. A molecule
Exogenous 25/88 may comprise, for example, a coding sequence for any polypeptide or fragment thereof, a functioning version of a malfunctioning endogenous molecule or a malfunctioning version of a normally functioning endogenous molecule. In addition, an exogenous molecule can comprise a coding sequence from another species that is an ortholog of an endogenous gene in the host cell.
An exogenous molecule can be, among other things, a small molecule, such as it is generated by a combative chemistry process, or a macromolecule such as a protein, nucleic acid, carbohydrates, lipids, glycoprotein, lipoprotein, polysaccharide, any modified derivative of the above molecules, or any complex comprising one or more of the above molecules. Nucleic acids include DNA and RNA, can be single or double stranded; They can be linear, branched or circular, and can be of any length. Nucleic acids include those capable of forming duplexes, as well as triplex forming nucleic acids. See, e.g., US Patent Nos 5,176,996 and 5,422,251. Proteins include, but are not limited to, DNA binding proteins, transcription factors, chromatin remodeling factors, methylated DNA binding proteins, polymerases, methylases, demethylases, acetylases, deacetylases, kinases, phosphatases, integrases, recombinases, ligases, topoisomerases, girases and helicases.
An exogenous molecule can be of the same type as a molecule such as an endogenous molecule, for example, an exogenous protein or nucleic acid. For example, an exogenous nucleic acid may comprise an infectious viral genome, a plasmid or episome introduced into a cell, or a chromosome that is not normally present in the cell. Methods for introducing exogenous molecules into cells are known to those skilled in the art and include, but are not limited to, lipid-mediated transfer, (ie, liposomes, including neutral and cationic lipids), electroporation, direct injection, cell fusion , bombard26 / 88 particle growth, calcium phosphate coprecipitation, DEAE-dextran mediated transfer and viral vector mediated transfer.
In contrast, an endogenous molecule is one that is normally present in a particular cell at a certain stage of development under certain environmental conditions. For example, an endogenous nucleic acid may comprise a chromosome, the genome of a mitochondria, chloroplast, or other organelle, or a naturally occurring episomic nucleic acid. Additional endogenous molecules can include proteins, for example, transcription factors and enzymes.
10 As used herein, the term product of an exogenous nucleic acid comprises polynucleotides and polypeptide products, e.g., transcription products (polynucleotide such as RNA), and translation products (polypeptides).
A fusion molecule is a molecule in which two or more 15 subunit molecules are preferably linked covalently. The subunit molecules can be of the same chemical type of molecule, or they can be of different chemical types of molecules. Examples of the first type of fusion molecule include, but are not limited to, fusion proteins (for example, a fusion between a ZFP DNA binding domain and a dividing domain) and fusion nucleic acids (for example, a nucleic acid encoding the fusion protein described above). Examples of the second type of fusion molecule include, but are not limited to, a fusion between a triplex-forming nucleic acid and a polypeptide, and a fusion between a minor groove ligand and a nucleic acid.
The expression of a fusion protein in a cell can result from the distribution of the fusion protein to the cell or by the distribution of a polynucleotide that encodes the fusion protein to a cell, in which the polynucleotide is transcribed and the transcript is translated to generate the fusion protein. Primary transcripts (trans-splicing), polypeptide dividing and polypeptide binding can also be involved in the expression of a protein in a cell. Methods of delivering polynu27 / 88 cleotide and polypeptide to cells are presented in the present disclosure.
A gene, for the purposes of the present description, includes a DNA region that encodes a gene product (see below), as well as all DNA regions that regulate the production of the gene product, whether or not such regulatory sequences are adjacent to the coding and / or transcribed sequences. Thus, a gene includes, but is not necessarily limited to, promoter sequences, terminators, translational regulatory sequences such as ribosome binding sites and entry sites with internal bossomas, enhancers, silencers, isolators, boundary elements, origins of replication, matrix fixation sites and locus control regions.
Gene expression refers to the conversion of information, contained in a gene, into a gene product. A gene product can be the direct product of transcription of a gene (for example, mRNA, tRNA, rRNA, antisense RNA, ribozyme, structural RNA or any other type of RNA) or a protein produced by translating an mRNA. Gene products also include RNAs that are modified, by processes such as capping, polyadenylation, methylation, and editing, and proteins modified, for example, by methylation, acetylation, phosphorylation, ubiquitination, ADPribosylation, myristylation, and glycosylation.
Modulation of gene expression refers to a change in the activity of a gene. Modulation of expression may include, but is not limited to, gene activation and gene suppression.
Plant cells include, but are not limited to, monocotyledonous (monocots) or dicotyledonous (dicots) plant cells. Non-limiting examples of monocots include cereal plants such as corn, rice, barley, oats, wheat, sorghum, rye, cane, pineapple, onion, banana and coconut. Non-limiting examples of dicots include tobacco, soda, sunflower, cotton, beet, potato, lettuce, melon, soy, canola (rapeseed), and alfalfa. Plant cells can be from any part of the plant and / or from any stage of plant development.
28/88
A region of interest is any region of the cell chromatin, such as, for example, a gene or a non-coding sequence within or adjacent to a gene, where it is desirable to bind an exogenous molecule. The ligation may be for the purpose of dividing the target DNA5 and / or target recombination. A region of interest may be present on a chromosome, an episome, an organellar genome (for example, mitochondrial, chloroplast), or an infectious viral genome, for example. A region of interest can be within the coding region of a gene, within transcribed non-coding regions, such as, for example, leader sequences, trailer sequences or introns, or within non-transcribed regions, either upstream or downstream. downstream of the coding region. A region of interest can be as small as a single pair of nucleotides or up to 2000 pairs of nucleotides in length, or any integral value of pairs of nucleotides.
15 The terms operatively linked and operably linked (or operably linked) are used interchangeably with reference to a juxtaposition of two or more components (such as sequence elements), in which the components are arranged in such a way that both components function normally and it allows the possibility that at least one of the components can mediate a function that is exercised over at least one of the other components. By way of illustration, a transcriptional regulatory sequence, such as a promoter, is operably linked to a coding sequence, if the transcriptional regulatory sequence controls the level of transcription of the coding sequence in response to the presence or absence of one or more factors regulatory transcription. A transcriptional regulatory sequence is generally operably cis-linked with a coding sequence, but need not be directly adjacent to it. For example, a potentializer is a transcriptional regulatory sequence that is operably linked to a coding sequence, even though they are not contiguous.
With respect to fusion polypeptides, the term operatively linked can refer to the fact that each of the components performs the
29/88 same function in the connection with the other component as it would be if it were not connected. For example, with respect to a fusion polypeptide in which a DNA-ΖΕΡ binding domain is fused to a dividing domain, the DNA-ΖΕΡ binding domain and dividing domain are operatively linked if, in the polypeptide of fusion, the portion of the DNA-ZFP binding domain is capable of binding its target site and / or its binding site, whereas the dividing domain is capable of cleaving DNA in the vicinity of the target site.
A functional fragment of a nucleic protein, polypeptide or acid10 is a protein, polypeptide or nucleic acid whose sequence is not identical to that of the full-length protein, polypeptide or nucleic acid, yet retains the same function as the protein, polypeptide or nude acid full length. A functional fragment can have more, less or the same number of residues as the corresponding native molecule15, and / or it can contain one or more amino acid or nucleotide substitutions. Methods for determining the function of a nucleic acid (for example, the coding function, the ability to hybridize to another nucleic acid) are well known in the art. Likewise, methods for determining protein function are well known. For example, the DNA binding function of a polypeptide can be determined, for example, by the binding filter, displacement of electrophoretic mobility or immunoprecipitation assays. DNA divage can be assayed by gel electrophoresis. See Ausubel et al., Supra. The ability of a protein to interact with another protein can be determined, for example, by co-immunoprecipitation, two hybrid assays or complementation, both genetic and biochemical. See, for example, Fields et al. (1989) Nature 340: 245-246; US patent 5,585,245 and PCT WO 98/44350.
Multiple Insertion Sites
Multiple insertion sites are described herein, i.e., polynucleotides comprising a plurality of zinc finger nuclease (ZFN) binding sites such that, after binding the appropriate ZFN pair, the multiple insertion site is cleaved between the target sites of the ZFN pair.
30/88
The target sites included in the multiple insertion site are preferably not found in the genome of the cell in which it is integrated. As such, the occurrence of undesirable divination in the genome is reduced or eliminated. Any number of target sites can be included in the multiple insertion site polynucleotide, for example, 1-50 (or any number in between), preferably between 2 and 30 (or any number in between, and even more preferably between 5 and 20 (or any number in between) .For zinc finger nucleases the target sites are typically paired such that the zinc finger nucleases form homo or heterodimers to cleave at the appropriate location.
In addition, as shown in Figure 1, a target site from each pair of the target site (shaded triangle figure 1) can be the same across the entire multiple insertion site. Alternatively, heterodimeric pairs can be different, as between sites.
15 the multiple insertion site can include target sites linked by only homodimers, target sites linked by heterodimers only, or a combination of target sites linked by homo- and heterodimers. Target sites linked by homodimers may be preferred in some cases, for one or more of the following reasons: the distribution of one ZFN may be more efficient than two, homodimerization reduces the emission of unequal stoichiometry due to the uneven expression of ZFNs; toxicity of divage at off-target sites can be reduced, the homodimer is half as likely to be disrupted when using CCHC (non-canonical) zinc finger domains, and / or the total number of single sites that can be targeted can be expanded. Alternatively, heterodimers may be preferred in other cases, as they allow mixing and matching of different target sites and, therefore, a potential increase in sites that can be targeted for ZFN pairs. In addition, heterodimers may allow sequential addition of donors as needed by the practitioner. Heterodimeric combinations can also allow for the specific deletion of any desired sections of a donor through the use of new ZFN pairs
31/88
It will be apparent that a target site is not required to be a multiple of three nucleotides for zinc finger nucleases. For example, in cases where cross-filament interactions occur (see, for example, US Patent 6,453,242 and WO 02 / 077,227), one or more of the individual zinc fingers of a multiple finger binding domain to quadruple overlay subsites. As a result, a three-finger protein can bind to a sequence of 10 nucleotides, where the tenth nucleotide is part of a quadruplet connected by a terminal finger, a four-finger protein can bind to a sequence of 13 nucleotides , in which the thirteenth nucleotide and part of a quadruplet connected by a terminal finger, etc.
The length and nature of amino acid binding sequences between the individual zinc fingers of a multi-finger binding domain also affects the binding to a target sequence. For example, the presence of a so-called non-canonical ligand, long ligand or structured ligand between adjacent zinc fingers in a multi-finger bonding domain can allow the fingers to attach subsites that are not immediately adjacent. Non-limiting examples of such binders are described, for example, in US Patent 6,479,626 and WO 01/53480. Therefore, one or more subsites, at a target site for a zinc finger binding domain, can be separated from each other by 1, 2, 3, 4, 5 or more nucleotides. To provide just one example, a four-finger binding domain can bind to a 13 nucleotide target site that comprises, in sequence, two contiguous 3-nucleotide subsites, an intervention nucleotide, and two contiguous triplet subsites.
Distance between sequences (for example, target sites) refers to the number of nucleotides or pairs of nucleotides that intervene between two sequences, as measured from the edges of the sequences closest to each other.
In certain embodiments where diving depends on the attachment of two zinc finger diving / domain half-domain fusion molecules to separate target sites, the two target sites may be in queue32 / 88 opposite DNA strands. In other embodiments, both target sites are on the same DNA strand.
The multiple insertion site can be integrated into any part of the plant's genome. In certain embodiments, the multiple insertion site 5 is integrated into a Zp15 in a corn genome, which, as described in US Application 12 / 653,735, is a desirable site for the target integration of exogenous sequences.
DNA Binding Domains
Any DNA binding domain can be used in the methods described herein. In certain embodiments, the DNA binding domain comprises a zinc finger protein. A zinc finger binding domain comprises one or more zinc fingers. Miller et al. (1985) EMBO J. 4: 1609-1614; Rhodes (1993) Scientific American Feb. 5,665 US Patent 6,453,242. The zinc finger binding domains described herein generally include 2, 3, 4, 5, 6 or even more zinc fingers.
Typically, a single zinc finger domain is about 30 amino acids in length. Structural studies have shown that each zinc finger domain (motif) contains two beta leaves (held in a beta loop that contains the two invariant cysteine residues) and 20 an alpha helix (containing two invariant histidine residues), which are kept in a particular conformation through the coordination of a zinc atom by the two cysteines and the two histidines.
Zinc fingers include both canonical C 2 H 2 zinc fingers (ie those in which the zinc ion is coordinated by two cysteines and two histidine residues) and non-canonical zinc fingers, such as, for example , C 3 H zinc fingers (those in which the zinc ion is coordinated by three cysteine residues and one histidine residue) and C 4 zinc fingers (those in which the zinc ion is coordinated by four cysteine residues ). See also WO 02/057293 and also Patent Publication 30 US 20080182332 on non-canonical ZFPs for use in plants.
A designed zinc finger binding domain may have a new binding specificity compared to a finger protein
33/88 of naturally occurring zinc. Design methods include, but are not limited to, rational model and various types of selection. The rational model includes, for example, the use of databases comprising triplet (or quadruplet) nucleotide sequences and individual zinc finger ammo acid sequences, where each tnplet or quadruplet nucleotide sequence is associated with one or more sequences of zinc finger ammoacids that bind to the special triplet or quadruplet sequence.
Examples of selection methods, including phage display and 10 two hybrid systems, are described in US Patents 5,789,538;
5,925,523; 6,007,988; 6,013,453; 6,410,248; 6,140,466; 6,200,759, and 6,242,568, as well as WO 98/37186, WO 98/53057; WO 00/27878, WO 01/88197 and GB 2,338,237.
The improvement of binding specificity for zinc finger binding domains has been described, for example, in co-ownership WO 02 / 077.227.
Since an individual zinc finger attaches to a sequence of three nucleotides (ie, triplet) (or a sequence of four nucleotides that can overlap, by one nucleotide, with Hgaçao 20 sites of four nucleotides from one adjacent zinc finger), the length of a sequence to which a zinc finger binding domain is designed to bind (for example, a target sequence) will determine the number of zinc fingers in a zinc finger designed. For example, for ZFPs in which the finger motifs do not bind to overlapping subsets, a target sequence of six nucleotides is linked by a two-finger binding domain; a target sequence of nine nucleotides is linked by a three-finger binding domain, etc. As noted here, the binding sites for the individual zinc fingers (i.e., subsites) at a target site need not be contiguous , but they can be separated by 30 or several nucleotides, depending on the length and nature of the amino acid sequences between the zinc fingers (that is, the interdedo ligands) in a multi-finger binding domain.
34/88
In a multiple zinc finger binding domain, the adjacent zinc fingers can be separated by amino acid ligand sequences of about 5 amino acids (the so-called canonical middled ligands) or, alternatively, by one or more non-canonical ligands.
See, for example, US Patents 6,453,242 and US 6,534,261 to co-ownership. For projected zinc finger binding domains that comprise more than three fingers, the insertion of longer (non-canonical) interdedo ligands between some of the zinc fingers may be desirable in some cases where this may increase affinity and / or specificity of Hgaçao 10 by the binding domain. See, for example, US Patent 6,479,626 and WO 01/53480. Therefore, multiple zinc finger binding domains can also be characterized for the presence and location of non-canonical interdedo ligands. For example, a six-finger zinc finger binding domain comprising three fingers (joined by two canonical interdedo ligands), one! Ongo ligand and three additional fingers (one by two canonical interdedo ligands) is denoted with a 2x3 configuration. Likewise, a link domain comprising two fingers (with a canonical link between them), a long link and two additional fingers (joined by a canonical link) is denoted with a configuration
2x2 A protein comprising three units of two fingers (each of which the two fingers are joined by a canonical ligand), and where each unit of two fingers is joined to the unit of two adjacent fingers by a long ligand, it is referred to as a 3x2 configuration.
The presence of a linker or long interdedos non - canonical en tre two fingers 25 adjacent a zinc binding domain multiple fingers often allows the two fingers bind to subsrtros that are not immediately adjacent on the target sequence. Accordingly, there may be gaps of one or more nucleotides between subsides in a target site, that is, a target site may contain one or more nucleotides that are not contacted by a zinc finger. For example, a 2x2 finger zinc binding domain can bind to two sequences of six nucleotides separated by a nucleotide, i.e.. which binds to a 1335/88 nucleotide target site. See also Moore et al. (2001a) Proo. Nat !. Acad. Know. USA
93: 1432-1436; Moore el al. (2001b) Proc. Natl. Acad. Sci. USA 98: 1437
1441 and WO 01/53480.
As mentioned earlier, a target subsite is a sequence of three or four nucleotides that is linked by a single zinc finger. For certain purposes, a two-finger unit is denoted a binding module. A binding module can be obtained, for example, by selecting two adjacent fingers in the context of a multi-finger protein (usually three fingers) that bind to a particular target sequence of 10 six nucleotides. Alternatively, the modules can be constructed by assembling individual zinc fingers. See also WO 98/53057 and WO
01/53480. .
Alternatively, the DNA binding domain can be derived from a nuclease. For example, ho15 ming endonuclease and meganuclease recognition sequences such as l-Scel, l-Ceul, Pl-Pspl, PlSce l-ScelV l-Csml, l-Panl, l-Scell, l-Ppol, l-Scelll , l-Crel, l-Tevl, l-Tevll and | -Tevlll are known. See also US Patent 5,420,032; US Patent 6,833,252 · Belfort et al. (1997) Nucleic Acids Res. 25: 3379-3388; Dujon et al (1989) Gene 82: 115-118; Perler et al. (1994) Nucleic Acids Res. 22, 20 1125-1127; Jasin (1996) Trends Genet. 12: 224-228; Gimble et al. (1996) J.
Mol Biol. 263: 163-180; Argast et al. (1998) J. Mol. Biol. 280: 345-353 and the New England Biolabs catalog. In addition, the DNA binding specificity of homing endonucleases and meganucleases can be designed to bind unnatural target sites. See, for example, Chevalier et al. (2002) 25 Molec Cell 10: 895-905; Epinat et al. (2003) Nucleic Acids Res. 31: 29522962; Ashworth et al. (2006) Nature 441: 656-659; Paques et al. (2007) Current Gene Therapy 7: 49-66; US Patent Publication 20070117128.
As another alternative, the DNA binding domain can be derived from a leucine zipper protein. Leucine zippers are a class of proteins that are involved in protein-protein interactions in many eukaryotic regulatory proteins that are important transcription factors associated with gene expression. The leucine zipper
36/88 refers to a common structural motif shared by these transcription factors across various kingdoms including animals, plants, yeasts, etc. The leucine zipper is formed by two polypeptides (homodimer or heterodimer) that bind to specific sequences of DNA in a way where the leucine residues are evenly spaced through an helix, such that the leucine residues of two polypeptides end up on the same face of the helix. The DNA binding specificity of leucine zippers can be used in the DNA binding domains described herein.
In some embodiments, the DNA binding domain is a domain designed from a TAL effector derived from Xanthomonas plant pathogens (see, Miller et al. (2010) Nature Biotechnology, Dec 22 [Epub ahead of print]; Boch et al, (2009) Science 29 Oct 2009 (10.1126 / science.117881) and Moscow and Bogdanove, (2009) Science 29 Oct 2009 (10.1126 / science. 1178817).
Cleavage Domains
As noted above, the DNA binding domain can be associated with a cleavage domain (nuclease). For example, hommg endonucleases can be modified in their DNA binding specificity, while maintaining nuclease function. In addition, zinc finger proteins can also be fused to a cleavage domain to form a zinc finger nuclease (ZFN). The fusion protein cleavage domain portion disclosed herein can be obtained from any endonuclease or exonuclease. Examples of endonucleases from which a cleavage domain can be derived include, but are not limited to, restriction endonucleases and homing endonucleases. See, for example, Catalog 2002-2003, New England Biolabs, Beverly, MA, and Belfort et al. (1997) Nucleic Acids Res. 25: 3379-3388. Enzymes that cleave additional DNA are known (e.g., S1 nuclease; mung bean nuclease; pancreatic DNase I; micrococcal nuclease; yeast HO endonuclease, Linn 30 et al. (Eds.) Nucleases, Cold Spring Harbor Laboratory Press, 1993). Non-limiting examples of endonucleases and hommg meganucleases include | -Scel, l-Ceul, Pl-Pspl, Pl-Sce, l-ScelV, l-Csml, l-Panl, l-Scell, l-Ppol, I37 / 88
Scelll, l-Crel, I-Tevl, l-Tevll and I-Tevlll which are known. See also US Patent 5,420,032; US patent 6,833,252; Belfort et al. (1997) Nucleic Acids Res. 25: 3379-3388; Dujon et al. (1989) Gene 82: 115-118; Perleret al. (1994) Nucleic Acids Res. 22, 1125-1127; Jasin (1996) Trends Genet.
12: 224-228; Gimble et al. (1996) J. Mol. Biol. 263: 163-180; Argast et al.
(1998) J. Mol. Biol. 280: 345-353 and the New England Biolabs catalog. One or more of these enzymes (or functional fragments thereof) can be used as a source of cleavage domains and half divination domains.
Restriction endonucleases (restriction enzymes) are present in many species and are capable of DNA sequence specific binding (at a recognition site), and cleavage DNA at or near the binding site. Certain specific restriction enzymes (for example, Type IIS) cleave DNA at sites removed from the recognition site and have separable ligation and cleavage domains. For example, the Type IIS Fold enzyme catalyzes double-stranded DNA cleavage, in 9 nucleotides from its recognition site in one strand and 13 nucleotides from its recognition site in the other. See, for example, US Patents 5,356,802; 5,436,150 and 6,487,994; as well as Li et at. (1992) Proc. Natl
Acad. Sd. USA 89: 4275-4279; Li et al. (1993) Proc. Natl. Acad. Sc /. USA 90-2764-2763; Kim et al (1994a) Proc. Natl. Acad. Sd. USA 91: 883-887; Kim etal. (1994b) J. Biol. Quote. 269: 31.978-31.982. So, in one mode. fusion proteins comprise the cleavage domain (or cleavage half-domain) of at least one T restriction enzyme, by IIS, and one or more finger zinc binding domains, which may or may not be designed.
An exemplary Type IIS restriction enzyme, whose dividing domain is separable from the binding domain, is Fold. This particular enzyme is active as a dimer, Bitinalte et al. (1998) Proc. Natl. Acad. Sd. USA 95: 30 10 570-10 575. Therefore, for the purposes of the present disclosure, the portion of the Fokl enzyme used in the disclosed fusion proteins is considered to be a half-domain of cleavage. Thus, for double-stranded 38/88 aphea and / or target substitution of cell sequences using zinc FoMd fusions, two fusion proteins, each comprising a half, FoM dividing domain. can be used to reconstitute a gift, catalytically active dividing hand. Alternatively, a single, 5-pohpept molecule containing a zinc and finger finger binding domain, half FoM dividing domains can also be used. Parameters for changing target sequence and target cleavage using zinc FoM-finger fusions are provided elsewhere in this disclosure.
A diving domain or half diving domain can be any portion of a protein that retains divar activity, or that retains the ability to multimerize (e.g., dimerize) to form a functional divar domain.
Exemplary US Type restriction enzymes are described in International Co-Ownership Publication WO 2007/014275, incorporated herein by reference in their entirety.
To improve the divage specificity, divage domains can also be modified. In certain embodiments, variants of the half domain divage are employed to minimize or prevent homodimerization of the half domain divage. Non-limiting examples of modified divination of such domino media are described in detail in WO 2007/014275. Incorporated by reference in its entirety here. See also examples. In certain embodiments, the divination domain comprises a half-poetized divination domain (also referred to as dimerization domain mutants) m.mmtza or 25 prevents homodimerization are known to those skilled in the art and described. for example, in Patent Publications US 20050064474 and US 20060188987 incorporated by reference in their entirety here. Amino acid residues at positions 446. 447. 479. 483. 484. 486, 487. 490, 491, 496, 498, 499, 500, 531, 534, 537, and 538 of FoM are all 30 targets to influence the dimerization of FoM dividing domain domains. See, for example. Patent Publications US 20050064474 and US 20060188987; International Patent WO 07 / 139,898; Miller et al. (2007) Nat.
39/88
Biotechnol. 25 (7): 778-785; and Doyon et al (2011) Nature Methods 8 (1): 7479.
The additional Fok projected half divination domains that form mandatory heterodimers can also be used in the ZFNs described herein. In one embodiment, the first divage half domain includes mutations in the amino acid residues at positions 490 and 538 of Fok and the second half divage domain includes mutations in amino acid residues 486 and 499.
In certain embodiments, the divage domain comprises two half divage domains, both of which are part of a single polypeptide comprising a binding domain, a first half divage domain and a second half divage domain. The dividing half domains can have the same amino acid sequence or different amino acid sequences, as long as they work to cleave DNA.
In general, two fusion proteins are required for divage if the fusion proteins comprise half domains of divage. Alternatively, a single protein comprising two half dividing domains can be used. The two dividing half domains can be derived from the same endonuclease (or functional fragments thereof), or each half diving domain can be derived from a different endonuclease (or functional fragments thereof). In addition, the target sites for the two fusion proteins are preferably arranged in relation to each other, such that the binding of the two fusion proteins to their respective target sites places the dividing half domains in a spatial orientation for each another that allows the half diving domains to form a functional diving domain, for example, by dimerization. Thus, in certain embodiments, the edges close to the target sites are separated by 5-8 nucleotides or 15-18 nucleotides. However, any integral number of nucleotides or pairs of nucleotides can intervene between two target sites (for example, from 2 to 50 nucleotides or more). In general, the dividing point is located between the target sites.
40/88
Fusion Proteins
Methods for modeling and constructing fusion proteins (and polynucleotides that encode them) are known to those skilled in the art. For example, methods for the modeling and construction of fusion proteins comprising DNA-binding domains (e.g., zinc finger domains) and regulatory or dividing domains (or half diving domains), and polynucleotides that encode such fusion proteins are described in US patents 6,453,242 and US 6,534,261 co-owned and US Patent Application Publications 2007/0134796 and 2005/0064474; incorporated herein by reference in their entirety. In certain embodiments, polynucleotides that encode fusion proteins are constructed. These polynucleotides can be inserted into a vector and the vector can be introduced into a cell (see below for further disclosure on vectors and methods for introducing polynucleotides into cells).
In certain embodiments of the methods described herein, a zinc finger nuclease comprises a fusion protein comprising a zinc finger binding domain and a half divination domain of the restriction enzyme Fokl, and two fusion proteins are expressed in a cell . The expression of two cell fusion proteins can result from the distribution of the two proteins to the cell; distribution of a protein and a nucleic acid that encodes one of the proteins to the cell; distribution of two nucleic acids, each encoding one of the proteins, to the cell, or by the distribution of a single nucleic acid, encoding both proteins, to the cell. In additional embodiments, a fusion protein comprises a single polypeptide chain comprising two half dividing domains and a zinc finger binding domain. In this case, a single fusion protein is expressed in a cell and, without wishing to be bound by theory, it is believed to cleave DNA as a result of the formation of an intramolecular dimer from the dividing half domains.
In certain embodiments, the components of fusion proteins
41/88 (for example, ZFP-Fo / d fusions) are arranged in such a way that the zinc finger domain is the closest to the amino terminal of the fusion protein and the half diving domain is the closest to the terminal carboxy. This reflects the relative orientation of the divage domain in naturally occurring dimerization cleavage domains such as those derived from the FcM enzyme, where the DNA binding domain is closest to the amino terminus and the middle dividing domain. it is the closest to the carboxy terminal. In these embodiments, dimerization donates half the dividing domains to form a functional nuclease is caused by the attachment of the 10 fusion proteins to sites in the opposite DNA strands, with the 5 end of the binding sites being proximal to each other.
In additional embodiments, the components of the fusion proteins (for example, ZFP-Fo / d fusions) are arranged in such a way that the closest dividing domain medium is the amino terminus of the fusion protein and the finger domain of zinc is the closest to the carboxy terminal. In these embodiments, the dimerization of the dividing half-domains to form a functional nuclease is caused by the binding of the fusion proteins to sites in the opposite DNA strands, with the 3 'ends of the binding sites being proximal to each other.
20 Still in additional embodiments, a first fusion protein contains the half dividing domain closest to the amino terminus of the fusion protein, and the zinc finger domain closest to the carboxy terminus, and a second fusion protein is arranged in such a way. so that the zinc finger domain is closest to the amino terminal of the fusion protein, and the half divage domain is closest to the carboxy terminal. In these embodiments, both fusion proteins bind to the same DNA strand, with the fusion protein binding site containing the first zinc finger domain closest to the carboxy terminus located on the 5 'side of the second protein binding site fusion containing zinc finger domain closest to the amino terminus.
In certain embodiments of the disclosed fusion proteins, the amino acid sequence between the zinc finger domain and the
42/88 divage (or half divage domain) is denoted as a ZC ligand. The ZC ligand must be distinguished from the interdedo ligands discussed above. See, for example, US Patent Publications 20050064474A1 and 20030232410, and International Patent Publication W005 / 084190, for details on obtaining ZC binders that optimize divage.
In one embodiment, the disclosure provides a ZFN comprising a zinc finger protein having one or more of the recognition helix amino acid sequences shown in Table 1. In another embodiment, a ZFP expression vector comprising a sequence of nucleotides encoding a ZFP having one or more recognition helices shown in Table 1.
Target integration
The described methods and compositions can be used to cleave DNA in any cell genome into which a multiple insertion site has been integrated, which facilitates the stable, target integration of an exogenous sequence at the multiple insertion site and / or excision of exogenous sequences in the presence of the appropriate ZFN pairs. See, Figures 1 and 2.
Also described here are the methods in which ZFN insertion sites, as part of an exogenous sequence, are introduced into the cell's genome in series. See, Figures 4 and 5. For example, an exogenous sequence flanked by a different combination of heterodimeric nuclease sites is inserted into the genome. Subsequently, a pair of ZFNs that cleave at one of the appropriate flanking ZFN sites is introduced into the cell in the presence of another exogenous sequence, which again includes different combinations of heterodimeric nuclease sites. The process can be repeated, as desired to insert the exogenous sequences. In addition, in the presence of the appropriate ZFN pairs, one or more exogenous sequences can be excised from the genome.
Figure 6 shows another embodiment in which the exogenous sequence comprises a marker gene and a gene of interest. Both the marker gene and the gene of interest are flanked by different ZFN binding sites (represented as triangles with different shadings), so that the marker gene can be deleted as appropriate, for example, when inserting genes additional. In organisms, such as plants, where there is a limited number of effective selectable markers, this allows for the use of as few as a selectable marker gene, greatly facilitating the potential to stack the genes of interest. In certain embodiments, for example, depending on the homology-directed DNA repair efficiency, a selectable division marker can be used. The correct integration of a donor DNA sequence using a selectable division marker creates an expressible selectable marker gene. Selectable markers can be excised from an integrated DNA sequence and can therefore be recycled. In another embodiment, the exogenous sequence for removal is flanked in the genome by partial sequences from a division marker gene. After excision, the marker gene is reconstructed, resulting in the creation of a functional marker gene. The use of selectable marker excision limits the number of selectable markers required to two or possibly just one.
For target integration at an integrated multiple insertion site as described herein, one or more DNA-binding domains (e.g., ZFPs) are designed to bind to a target site at, or close to, the dividing site predetermined, and a fusion protein comprising the designed DNA binding domain and a dividing domain is expressed in a cell. By attaching the DNA-binding portion (e.g., zinc finger) of the fusion protein to the target site, the DNA is cleaved, preferably through a double strand break, close to the target site by the dividing domain.
The presence of a double filament break at the multiple insertion site facilitates the integration of exogenous sequences through homologous recombination. In certain embodiments, the polynucleotide comprising the exogenous sequence to be inserted into the multiple insertion site will include one or more regions of homology with the multiple insertion site polynucleotide and / or the surrounding genome to facilitate homologous recombination.
44/88
Approximately 25, 50, 100, 200, 500, 750, 1000, 1500, 2,000 nucleotides or more of sequence homology between a donor and a genomic sequence (or any integral value between 10 and 2000 nucleotides, or more) will support recombination homologous between them. In certain embodiments, the homology arms are less than 1,000 base pairs in length. In other embodiments, the homology arms are less than 750 base pairs in length. See also, Provisional Patent Application US 61 / 124,047, which is incorporated herein by reference. A donor molecule (exogenous sequence) can contain several discontinuous regions of homology with cellular chromatin. For example, for target insertion of sequences not normally present in a region of interest, said sequences may be present in a donor nucleic acid molecule and flanked by regions of homology with a gene sequence of the region of interest.
Any sequence of interest (exogenous sequence) can be introduced or excised from a multiple insertion site as described herein. Examples of exogenous sequences include, but are not limited to, any polypeptide coding sequence (for example, cDNAs), promoter, enhancer and other regulatory sequences (for example, interference RNA sequences, Lentivirus expression cassettes, epitope, marker genes, divage enzyme recognition sites and various types of expression constructs, such sequences can be readily obtained using standard molecular biology techniques (synthesis, cloning, etc.) and / or are commercially available. it can be introduced into the cell prior to, concurrently with, or subsequent to, expression of the fusion protein (s).
The donor polynucleotide can be DNA or RNA, single-stranded or double-stranded and can be introduced into the cell in a linear or circular shape. If introduced in linear form, the ends of the donor sequence can be protected (for example, from exonucleolytic degradation) by methods known to those skilled in the art. For example,
45/88 one or more dideoxynucleotide residues are added to the 3'-terminus of a linear molecule and / or self-complementary oligonucleotides are attached to one or both ends. See, for example, Chang et al. (1987) Proc. Natl. Acad. Know. USA 84: 4959-4963; Nehls et al. (1996) Science 272: 886-889. Additional methods for protecting exogenous polynucleotides from degradation include, but are not limited to, addition of amino terminal group (s) and the use of modified internucleotide bonds, such as, for example, phosphorothioates and phosphoramidates, and residues of O-methyl ribose or deoxyribose.
A polynucleotide can be introduced into a cell as part of a vector molecule having additional sequences, such as, for example, the origins of replication, promoters and genes encoding antibiotic resistance. In addition, donor polynucleotides can be introduced as bare nucleic acid, as nucleic acid complexed with an agent such as a nanoparticle, liposome, or poloxamer, or can be delivered to plant cells by bacteria or viruses (for example, Agrobacterium, Rhizobium NGR234 sp, Sinorhizoboium meliloti, Mesorhizobium loti, tobacco mosaic virus, potato X virus, cauliflower mosaic virus and cassava mosaic virus, see, for example, Chung et al. (2006) Trends Plant Sei 11 (1): 1-4.
As detailed above, the binding sites at the multiple insertion site for two fusion proteins (homodimers or heterodimers), each comprising a zinc finger binding domain and a half diving domain, can be located from 5-8 or 15-18 nucleotides apart, as measured from the edge of each binding site closest to another binding site, and dividing occurs between the binding sites. Whether diving occurs at a single site or at multiple sites between the binding sites is immaterial, since the cleaved genomic sequences are replaced by donor sequences. Thus, for efficient alteration of the sequence of a single pair of nucleotides by target recombination, the midpoint of the region between the binding sites is within 10,000 nucleotides of that pair of nucleotides, preferably within 1,000 nu46 / 88 cleotides, or 500 nucleotides, or 200 nucleotides, or 100 nucleotides, or 50 nucleotides, or 20 nucleotides, or 10 nucleotides, or 5 nucleotides, or 2 nucleotides, or one nucleotide, or in the pair of nucleotides of interest.
Methods and compositions are also provided that can enhance target recombination levels, including, but not limited to, use of fusions of additional ZFP functional domains that activate the expression of genes involved in homologous recombination, such as, for example, genes from plants of the epistasis group RAD54 (for example, AtRad54, AtRad51), and genes whose products interact with the products of the genes mentioned above. See, for example, Klutstein et al. Genetics. 2008 Apr; 178 (4): 2389-97.
Similarly, fusions of functional ZFP domains can be used, in combination with the methods and compositions described herein, to suppress the expression of genes involved in the junction of the non-homologous end (for example, Ku70 / 80, XRCC4, poly (ADP ribose) polymerase, DNA ligase 4). See, for example, Riha et al. (2002) EMBO 21: 28192826; Freisner et al. (2003) Plant J. 34: 427-440; Chen et al. (1994) European Journal of Biochemistry 224: 135-142. Methods for activating and suppressing gene expression using fusions between a zinc finger binding domain and a functional domain are described, for example, in US Patent 6,534,261; 6,824,978 and 6,933,113 joint ownership. Additional methods of suppression include the use of antisense oligonucleotides and / or small interfering RNAs (siRNA or RNAi) or target shRNAs for the gene sequence to be repressed.
Additional increases in terms of target recombination efficiency, in cells comprising a zinc nuclease / finger fusion molecule and a donor DNA molecule, are achieved by blocking cells in the G 2 phase of the cell cycle, when repair processes conducted by homology are maximally active. Such retention can be achieved in one of several ways. For example, cells can be treated, for example, with drugs, compounds and / or molecules
47/88 small cells that influence the progression of the cell cycle in order to retain cells in the G 2 phase. Examples of such molecules include, but are not limited to, compounds that affect the polymerization of microtubule compounds (eg vinblastine, Nocodazol, Taxol), which interact with DNA (eg c / s-platinum dichloride (II) diamine, Cisplatin, doxorubicin) and / or compounds that affect DNA synthesis (for example, thymidine, hydroxyurea, L-mimosin, etoposide, 5-fluorouracil). Additional increases in recombination efficiency are achieved through the use of histone deacetylase (HDAC) inhibitors (eg, sodium butyrate, trichostatin A) that alter the chromatin structure to produce more accessible genomic DNA for cell recombination machinery.
Additional methods for cell cycle arrest include overexpression of proteins that inhibit the activity of cell cycle kinases -CDK, for example, by introducing a cDNA that encodes the protein into the cell or by introducing a projected ZFP into the cell that activates the expression of the gene that encodes the protein. Cell cycle retention is also achieved by inhibiting the activity of cyclins and CDKs, for example, using RNAi methods (for example, US Patent 6,506,559) or by introducing a projected ZFP into the cell that suppresses the expression of one or more genes involved in the progression of the cell cycle, such as, for example, CDK and / or cyclin genes. See, for example, US Patent 6,534,261 for co-ownership for methods for the synthesis of zinc finger proteins designed for the regulation of gene expression.
Alternatively, in certain cases, the target cleavage is conducted in the absence of a donor polynucleotide (preferably in S or G 2 phase), and recombination occurs between homologous chromosomes.
Expression Vectors
A nucleic acid that encodes one or more fusion proteins (e.g., ZFNs) as described herein can be cloned into a vector for transformation into prokaryotic or eukaryotic cells for replication and / or expression. Vectors can be prokaryotic vectors, for example,
48/88 plasmids or shuttle vectors, insect vectors, or eukaryotic vectors. A nucleic acid that encodes a fusion protein can also be cloned into an expression vector, for administration to a cell.
To express the fusion proteins (for example, ZFNs), the sequences encoding the fusion proteins are 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, for example, in Sambrook et al, Molecular Cloning, A Laboratory Manual (2nd ed 1989;.. 3rd ed, 2001); Kriegler, Gene Transfer and Expression: A Laboratory Manual (1990); and Current Protocols in Molecular Biology (Ausubel et al., supra. Bacterial expression systems to express ZFP are available from, for example, E. coli, Bacillus sp., and Salmonella (Paiva et al, Gene 22: 229-235 (1983)) Kits for such expression systems are commercially available Eukaryotic expression systems for mammalian cells, yeasts, and insect cells are well known to those skilled in the art and are also commercially available.
The promoter used to direct the expression of a fusion protein-encoding nucleic acid depends on the particular application. For example, a strong constitutive promoter suitable for the host cell is normally used for the expression and purification of fusion proteins.
In contrast, when a fusion protein is administered in vivo for the regulation of a plant gene (see, Nucleic Acid Delivery to Plant Cells below), or a constitutive, regulated promoter (for example, during development, by tissue or type cell, or by the environment) or an inducible promoter is used, depending on the particular use of the fusion protein. Non-limiting examples of plant promoters include promoter sequences derived from A. thaliana ubiquitin-3 (ubi-3) (Callis, et al., 1990, J. Biol. Chem. 265-12486-12493); A. tumifaciens manopino synthase (Amas) (Petolino et al. US Patent 6,730,824); and / or Cassava Rib Mosaic Virus (CsVMV) (Verdaguer et al., 1996, Plant Molecular Biology 31: 1129-1139). See also the Examples.
49/88
In addition to the promoter, the expression vector typically contains a transcription unit or expression cassette that contains all the additional elements necessary for the expression of nucleic acid in host cells, either prokaryotic or eukaryotic. A typical expression cassette thus contains a promoter operably linked, for example, to a nucleic acid sequence encoding the fusion protein, and the necessary signals, for example, for efficient transcript polyadenylation, transcriptional termination, ribosome binding sites , or translation termination. Additional cassette elements may include, for example, enhancers, heterologous splicing signals, and / or a nuclear location signal (NLS).
The particular expression vector used to carry genetic information to the cell is selected with a view to the intended use of fusion proteins, for example, expression in plants, animals, bacteria, fungi, protozoa, etc. (see expression vectors described below). Expression vectors of bacterial and animal patterns are known in the art and are described in detail, for example, in US Patent Publication 20050064474A1 and International Patent Publications W005 / 084190, W005 / 014791 and W003 / 080809.
Standard transfection methods can be used to produce bacterial, yeast, mammalian or insect cell lines that express large amounts of protein, which can then be purified using standard techniques (see, for example, Colley et al., J. Biol. Chem. 264: 17619-17622 (1989); Guide to Protein Purification, in Methods in Enzymology, vol. 182 (Deutscher, ed., 1990)). The transformation of eukaryotic and prokaryotic cells is performed according to standard techniques: (see, for example, Morrison, J. Bact. 132: 349-351 (1977); Clark-Curtiss & Curtiss, Methods in Enzymology 101: 347-362 (Wu eta!., Eds., 1983).
Any of the well-known processes for introducing foreign nucleotide sequences into such host cells can be used. These include the use of calcium phosphate transfection, polybrene, protoplast fusion, electroporation, ultrasonic methods (eg
50/88 example, sonoporation), liposomes, microinjection, naked DNA, plasmid vectors, viral vectors, both episomic and integrative, and any of the other well-known methods for introducing cloned genomic DNA, cDNA, synthetic DNA or other genetic material in a host cell (see, for example, Sambrook et al., supra). It is only necessary that the particular genetic engineering procedure used is capable of successfully introducing at least one gene into the host cell capable of expressing the protein of choice.
Distribution of Nucleic Acids to Plant Cells
As noted above, DNA constructs can be introduced (for example, into the genome of) a desired host plant by a variety of conventional techniques. For reviews of such techniques, see, for example, Weissbach & Weissbach Methods for Plant Molecular Biology (1988, Academic Press, N.Y.) Section VIII, pp. 421-463; and Grierson & Corey, Plant Molecular Biology (1988, 2nd Ed.), Blackie, London, Ch. 7-9.
For example, the DNA construct can be introduced directly into the genomic DNA of the plant cell, using techniques such as electroporation and microinjection of plant cell protoplasts, or the DNA constructs can be introduced directly to plant tissues using biological methods, such as as bombardment of DNA particles (see, for example, Klein et al. (1987) Nature 327: 70-73). Alternatively, the DNA construct can be introduced into the plant cell through transformation of nanoparticles (see, for example, US Patent Application 12 / 245,685, which is incorporated herein by reference in its entirety). Alternatively, the DNA constructs can be combined with suitable T-DNA flanking / margin regions and introduced into a conventional host vector Agrobacterium tumefaciens. Transformation techniques mediated by Agrobacterium tumefaciens, including disarming and use of binary vectors, are well described in the scientific literature. See, for example, Horsch et al. (1984) Science 233: 496-498, and Fraley et al. (1983) Proc. Nat'l. Acad. Know. USA 80: 4803.
In addition, gene transfer can be achieved using 51/88 of non-Agrobacterium bacteria or viruses, such as Rhizobium sp. NGR234, Sinorhizoboium meliloti, Mesorhizobium loti, potato X virus, cauliflower mosaic virus and cassava rib mosaic virus and / or tobacco mosaic virus, See, for example, Chung et al. (2006) Trends Plant 5 Sci. 11 (1): 1-4.
The virulence functions of the host Agrobacterium tumefaciens will direct the insertion of a T-strand containing the construct and adjacent marker in the DNA of plant cells when the cell is infected by bacteria using a binary T DNA vector (Bevan (1984) Nuc. Acid Res.
12: 8711-8721) or coculture production (Horsch et al. (1985) Science 227: 1229-1231). Generally, the Agrobacterium transformation system is used to design dicot plants (Bevan et al. (1982) Ann. Rev. Genet 16: 357-384; Rogers et al. (1986) Methods Enzymol. 118: 627-641). The Agrobacterium transformation system can also be used to transform, as well as transfer, DNA to monocot plants and plant cells. See US Patent 5,591,616; Hernalsteen et al. (1984) EMBO J 3: 3039-3041; Hooykass-Van Slogteren et al. (1984) Nature 311: 763-764; Grimsley et al. (1987) Nature 325: 1677-179; Boulton et al. (1989) Plant Mol. Biol. 12: 31-40; and Gould et al. (1991) Plant Physiol.
95: 426-434.
Alternative gene transfer and transformation methods include, but are not limited to, transformation of protoplasts via calcium, polyethylene glycol (PEG) or electroporation-mediated absorption by naked DNA (see Paszkowski et al. (1984) EMBO J 3: 2717- 2722, Potrykus et al.
(1985) Molec. Gen. Genet. 199: 169-177; Fromm et al. (1985) Proc. Nat.
Acad. Sci. USA 82: 5824-5828; and Shimamoto (1989) Nature 338: 274-276) and electroporation of plant tissues (D'Halluin et al. (1992) Plant Cell 4: 1495-1505). Additional methods for transforming plant cells include microinjection, silicon carbide-mediated DNA absorption (Kaeppler et al. (1990) Plant Cell Reporter 9: 415-418), and bombardment of microprojectiles (see Klein et al. (1988 ) Proc. Nat. Acad. Sci. USA 85: 4305-4309; and Gordon-Kamm et al. (1990) Plant Cell 2: 603-618).
52/88
The described methods and compositions can be used to insert the exogenous sequences at the multiple insertion site that have been inserted into the genome of a plant cell. This is useful in that the expression of a transgene introduced into a plant's genome depends critically on its integration site. Therefore, genes encoding, for example, herbicide tolerance, resistance to insects, nutrients, antibiotics or therapeutic molecules can be inserted, by target recombination, into regions of a plant genome favorable to their expression.
The transformed plant cells that are produced by any of the above transformation techniques can be grown to regenerate an entire plant that has the transformed genotype and, therefore, the desired phenotype. Such regeneration techniques depend on the manipulation of phytohormones determined in a tissue culture growth medium, typically depending on a biocide and / or herbicide marker that has been introduced in conjunction with the desired nucleotide sequences. The regeneration of plants from cultured protoplasts is described in Evans, et a!., Protoplasts Isolation and Culture in Handbook of Plant Cell Culture, pp. 124-176, Macmillian Publishing Company, New York, 1983; and Binding, Regeneration of Plants, Plant Protoplasts, pp. 21-73, CRC Press, Boca Raton, 1985. Regeneration can also be obtained from the callus of plants, explants, organs, pollens, embryos or their parts. Such regeneration techniques are described, in general, in Klee etal. (1987) Ann. Rev. of Plant Phys. 38: 467-486.
Nucleic acids introduced into a plant cell can be used to impart the desired traits to essentially any plant. A wide variety of plants and plant cell systems can be designed for the desired physiological and agronomic characteristics described herein using the nucleic acid constructs of the present disclosure and the transformation methods mentioned above. In preferred embodiments, the target plants and plant cells for the project include, but are not limited to, those monocotyledonous plants and
53/88 dicots, such as crops including grain crops (eg wheat, maize, rice, millet, barley), fruit crops (eg tomato, apple, pear, strawberry, orange), fodder crops (eg alfalfa), vegetable tuber crops (eg carrot, potato, beet, 5 yams), green leafy crops (eg lettuce, spinach), flowering plants (eg petunia, rose, chrysanthemum), conifers and pines (for example, spruce pine, spruce) and plants used in phytoremediation (for example, heavy metal accumulation plants); oilseeds (eg sunflower, rapeseed) and plants used for experimental purposes (eg 10 Arabidopsis). Thus, the methods and compositions described have use over a wide range of plants, including, but not limited to, species of the genus Asparagus, Avena, Brassica, Citrus, Citrullus, Capsicum, Cucurbita, Daucus, Erigeron, Glycine, Gossypium, Hordeum, Lactuca, Lolium, Lycopersicon, Malus, Manihot, Nicotiana, Orychophragmus, Oryza, Persea, Phaseo15 lus, Pisum, Pyrus, Prunus, Raphanus, Secale, Solanum, Sorghum, Triticum, Vitis, Vigna, and Zea.
A person skilled in the art will recognize that after the exogenous sequence is stably incorporated into transgenic plants and confirmed to be operable, it can be introduced to other plants through sexual crossing. Any of a number of standard breeding techniques can be used, depending on the species to be bred.
A transformed plant cell, callus, tissue or plant can be identified and isolated by selecting or screening the plant material designed for traits encoded by the marker genes present in the transformation DNA. For example, selection can be carried out by growing the plant material projected onto media containing an imbibing amount of the antibiotic or herbicide to which the transforming gene construct confers resistance. In addition, transformed plants and plant cells 30 can also be identified by screening for the activities of any visible marker genes (for example, β-glucurmdase, luciferase, B or C1 genes) that may be present in the constructs54 / 88 of recombinant nucleic acids. These selection and screening methodologies are well known to those skilled in the art.
Physical and biochemical methods can also be used to identify transformants of plant cells or plants containing inserted gene constructs. These methods include, but are not limited to: 1) Southern analysis or PCR amplification to detect and determine the structure of the recombinant DNA insert, 2) Northern blot, S1 RNase protection, primer extension or amplification reverse transcriptase by PCR for the detection and examination of RNA transcripts of gene constructs; 3) enzymatic assays for the detection of enzyme or ribozyme activity, where such gene products are encoded by the gene construct, 4) protein gel electrophoresis, Western blot techniques, immunoprecipitation, or enzyme binding immunoassays (ELISA), where the products of gene constructs are proteins. Additional techniques, such as in situ hybridization, enzyme staining, and immunostaining, can also be used to detect the presence or expression of the recombinant construct in specific plant organs and tissues. The methods for doing all of these assays are well known to those skilled in the art.
Effects of gene manipulation using the methods described herein can be observed through, for example, Northern blots of RNA (e.g., mRNA) isolated from tissues of interest. Normally, if mRNA is present or the amount of mRNA has increased, it can be assumed that the corresponding transgene is being expressed. Other methods of measuring encoded polypeptide and / or gene activity can be used. Different types of enzyme assays can be used, depending on the substrate used and the method of detecting the increase or decrease of a reaction product or by-product. In addition, the expressed polypeptide levels can be measured immunochemically, i.e., ELISA, RIA, EIA and other antibody-based assays well known to those skilled in the art, such as by electrophoretic detection assays (either with staining or western blotting). As a non-limiting example, the detection of AAD-1 and PAT proteins using an ELI55 / 88 assay
SA is described in US Patent Application 11 / 587,893 the references of which are incorporated herein by reference in their entirety. The transgene can be selectively expressed in some plant tissues or in some stages of development, or the transgene can be expressed in substantially all plant tissues, substantially throughout its life cycle. However, any form of combinatorial expression is also applicable.
The present disclosure also encompasses the seeds of the transgenic plants described above in which the seed has the transgene or gene construct. The present disclosure further encompasses the progeny, cell lines, clones, or cells of transgenic plants described above wherein said progeny, clone, cell line, or cell has the transgene or gene construct.
Fusion proteins (e.g., ZFNs) and expression vectors that encode fusion proteins can be administered directly to the plant for gene regulation, target cleavage, and / or recombination. In certain embodiments, the plant contains multiple similar target genes. Thus, one or more different fusion proteins or expression vectors encoding fusion proteins can be administered to a plant in order to target one or more of these parallel genes (for example, Zp15, see PCT patent publication WO2010077319 ) in the plant.
The administration of effective amounts is, by any of the routes normally used for the introduction of fusion proteins in final contact with the plant cell to be treated. ZFPs are administered in any appropriate manner, preferably with acceptable carriers. Suitable methods of administering such modulators are available and well known to those skilled in the art and, although more than one route can be used to administer a particular composition, a particular route can often provide a more immediate and more immediate reaction. effective than the other route.
Carriers can also be used and are determined, in part, by the particular composition to be administered, as well as by the
56/88 particular method used to administer the composition. Therefore, there is a wide variety of suitable carrier formulations that are available.
Distribution to Mammalian Cells
The ZFNs described herein can be delivered to a mammalian target cell by any suitable means, including, for example, by injection of ZFN mRNA. See, Hammerschmidt et al. (1999) Methods Cell Biol. 59: 87-115.
Protein delivery methods comprising zinc fingers are described, for example, in US Patents 6,453,242; 6,503,717; 6,534,261; 6,599,692; 6,607,882; 6,689,558; 6,824,978; 6,933,113; 6,979,539; 7,013,219; and 7,163,824, the descriptions of which are incorporated herein by reference in their entirety.
The ZFNs as described herein can also be distributed using vectors containing sequences that encode one or more of the ZFNs. Any vector systems can be used, including, but not limited to, plasmid vectors, retroviral, lentiviral vectors, adenovirus vectors, poxvirus vectors; herpesvirus vectors and adeno-associated virus vectors, etc. See also US Patents 6,534,261; 6,607,882 '6,824,978; 6,933,113; 6,979,539; 7,013,219; and 7,163,824, hereby incorporated by reference in their entirety. In addition, it will be apparent that any of these vectors may comprise one or more ZFN coding sequences. Thus, when one or more pairs of ZFNs are introduced into the cell, the ZFNs can be transported in the same or different vectors. When multiple vectors are used, each vector can comprise a sequence that encodes one or multiple ZFNs.
Conventional methods of transferring viral-based and non-viral genes can be used to introduce nucleic acids encoding projected ZFPs into mammalian cells. Such methods can also be used to deliver nucleic acids encoding ZFPs to mammalian cells in vitro. In certain embodiments, the nucleic acids encoding the ZFPs are administered for use in vivo or ex vivo.
57/88
Non-viral vector delivery systems include electroporation, lipofection, microinjection, biobalistics, virosomes, liposomes, immunoliposomes, polycation or nucleic acid conjugates: lipid, naked DNA, artificial virions, and DNA-enhancing agent. Sleep5 portioning using, for example, the Sonitron 2000 system (Rich-Mar). Can also be used for the distribution of nucleic acids. Viral vector delivery systems include DNA and RNA viruses, which have either episomal or integrated genomes after delivery to the cell. Other exemplary nucleic acid delivery systems include those 10 supplied by Amaxa Biosystems (Cologne, Germany), Maxcyte Inc.
(Rockville, Maryland), Molecular BTX delivery systems (Holliston, MA) and Copernicus Therapeutics Inc, (see, for example, US 6,008,336). Lipofection is described in, for example, US 5,049,386, US 4,946,787; and US 4,897,355) and lipofection reagents are sold commercially (for example, TRANSFECTAM ™ and LIPOFECTIN ™). Cationic and neutral lipids that are suitable for efficient polynucleotide receptor recognition lipofection include those of Feigner, WO 91/17424, WO 91/16024. Distribution can be to cells (ex vivo administration) or target tissues (in vivo administration). The preparation of lipid complexes: nucleic acid, including target liposomes, such as immunolipid complexes, is well known to one skilled in the art (see, for example, Crystal, Science 270: 404-410 (1995); Blaese et al., Cancer Gene Ther. 2: 291-297 (1995); Behr et al., Bioconjugate Chem. 5: 382-389 (1994); Remy et al., Bioconjugate Chem. 5: 647-654 (1994); Gao et al., Gene Therapy 2: 710-722 25 (1995); Ahmad et al., Cancer Res. 52: 4817-4820 (1992); US Patents
4,186,183, 4,217,344, 4,235,871, 4,261,975, 4,485,054, 4,501,728, 4,774,085, 4,837,028, and 4,946,787).
As noted above, the described methods and compositions can be used in any type of mammalian cell. The proteins (for example, ZFPs), polynucleotides that encode them and compositions that comprise the proteins and / or polynucleotides described herein can be distributed to a target cell by any suitable means. The cells58 / 88
Suitable ones include, but are not limited to, eukanotic and prokaryotic cells and / or cell lines. Non-limiting examples of such cells or cell lines generated from such cells include COS, CHO (for example, CHO-S, CHO-K1, CHO-DG44, CHO-DUXB11, CHO-DUKX, 5 CHOK1SV), VERO, MDCK, WI38, V79, B14AF28-G3, BHK, HaK, NSO
SP2 / 0-Ag14, HeLa, HEK293 (e.g., HEK293-F, HEK293-H, HEK293T), and perC6 cells as well as insect cells such as Spodoptera fugiperda (Sf), or fungal cells such as Saccharomyces, Pichia and Schizosaccharomyces. In certain embodiments, the cell line is a 10-line cell of CHO-K1, MDCK or HEK293 cells. Suitable primary cells include peripheral blood mononuclear cells (PBMC), and other subsets of blood cells, such as, but not limited to, CD4 + or CD8 + T cells. Suitable cells also include stem cells, such as, for example, embryonic stem cells, induced pluripotent stem cells, hematopoietic stem cells, neural stem cells and mesenchymal stem cells.
EXAMPLES
Example 1; Plasmid Models
Example 1.1: Binding Sites the eZFN 20 Eight sit 'the ügação zinc finger nuclease (eZFN) designed (CL: AR - SEQ ID NOS: 1, RL: PR - SEQ ID No: 2, AL. PR - SEQ ID NO: 3, PL.AR - SEQ ID NO: 4, CL: RR - SEQ ID NO: 5, RL: CR - SEQ ID NO: 6, CL: PR - SEQ ID NO: : 7, RL: AR - SEQ ID NO: 8) were combined into a single DNA fragment (multi-eZFN binding site), with flanking sites of unique PCR myciators for each of the eZFN binding sites . In addition, other eZFN binding sites have been modeled and shown to cleave at high levels in yeast (see, for example, US Patent Publication 2009/0111119), including: PL: RR - SEQ ID NO: 9, AL: RR SEQ ID NO: 10, AL: CR - SEQ ID NO: 11, PL: CR - SEQ ID NO: 12 and Homodimme 30 ro eZFN's RR: RR - SEQ ID NO: 13, RL: RL - SEQ ID No. 14 PR: PR - SEQ ID NO: 15, PL: PL - SEQ ID No. 16 CL CL - SEQ ID No.: 17, CR: CR - SEQ ID NO: 18, AR : AR - SEQ ID NO: 19, and AL: AL - SEQ ID NO: 20. CL and CR refer to 59/88, respectively, left and right hand zinc finger models for the CCR5 receiver designated 8266 and 8196, which have the sequences and bind to the target sites shown in US Patent Publication 2008 / 0159996. AL and AR refer, respectively, to the left and right zinc finger 5 models for the AAVS1 locus designated 15556 and 15590 and have the helix recognition strings and bind to the target sites shown in US Patent Publication 2008/0299580 . The helix recognition sequences and target sites for the PL and PR models, as well as the RL and RR models are listed below in Tables 10 1 and 2. PL and PR both refer to left and right zinc finger models for ZFNs specific for the human PRMT1 gene, while RL and RR refer to left and right zinc finger models for ZFNs specific for the mouse Rosa26 locus.
None of these target sites is present in the corn genome 15 as measured by bioinformatics analysis. The PCR primer sites were included for evaluation of NHEJ resulting from the double-stranded dividing chromosomally located DNA fragment by eZFNs.
Table 1: ZFN models
ZFN name (gene) F1 F2 F3 F4 F5 F6 ZFN 19353 DRSNLSRRSDALTQ TSGNLTR TSGSLTR TSGHLSR(PRMT) PL (SEQN °: 27) ID (SEQ ID NO: 28) (SEQ IDN °: 29) (SEQ ID NQ30) (SEQ IDN °: 31) AT ZFN 19354 RSANLSVDRANLSR RSDNLRE ERGTLAR TSSNRKT(PRMT) PR (SEQN °: 32) ID (SEQ ID NO: 33) (SEQ IDN °: 34) (SEQ ID NO: 35) (SEQ IDN °: 36) AT ZFN 18473 DRSARTRQSGHLSR RSDDLSK RNDHRKN AT(mRosa26) RL (SEQN °: 37) ID (SEQ ID NO: 38) (SEQ IDN °: 39) (SEQ ID NO: 40) AT FN 18477 (mRosa26) RR SGDLTRSEQN °: 41) D SGSLTRSEQ ID NO: 42) SGHLAR SEQ IDN °: 43) SSDLTRSEQ ID NO: 44) SDNLSE SEQ IDN °: 45) NAHRKT (SEQ IDNo. 46)
60/88
Table 2: Target ZFN binding sites
ZFN name (gene) Target connection site ZFN 19353 (PRMT) PL acGGT GTT GAGcAT GGACtcgtagaaga (SEQ ID N °: 47) ZFN 19354 (PRMT) PR tcTATGCCCGGGACAAGtggctggtgag (SEQ ID N °: 48) ZFN 18473 (mRosa26)RL gaTGGGCGGGAGTCttctgggcaggctt (SEQ ID N °: 49) ZFN 18477 (mRosa26)RR ctAGAAAGACTGGAGTTGCAgatcacga (SEQ ID N °: 50)
Att sites were included in the synthesized DNA fragment and the fragment cloned into a plasmid using TOPO cloning (Invitrogen, Carlsbad, CA). The Gateway LR CLONASE ™ recombination reaction (Invitrogen) was used to transfer this fragment into pDAB101834 and pDAB101849. These vectors contain selectable markers suitable for tobacco and corn, respectively. pDAB101834 is composed of the cassava vein mosaic virus promoter (CsVMV; promoter and 5 'untranslated region derived from the cassava rib mosaic virus; Verdaguer et al., (1996) Plant Molecular Biology, 31 (6) 1129- 1139), the phosphinothricin acetyl transferase gene (PAT; Wohlleben et al., (1988) Gene 70 (1), 25-37) and the AtuORFI 3 'UTR (untranslated region 3' (UTR) comprising the polyadenylation site and Agrobacterium tumefaciens pTi15955 open reading frame transcription terminator 1 (ORF1); Barker et al., (1983) Plant Molecular Biology, 2 (6), 335-50). The corn vector pDAB101849 contains the selectable marker cassette, including the rice 1 actin gene promoter (OsActl; promoter, 5 'untranslated region (UTR) and intron derived from the actin 1 Oryza sativa gene (Act1); McElroy et al., (1990) Plant Cell 2 (2): 163-71) and the 3 'UTR ZmLip (3' untranslated region (UTR) comprising the polyadenylation site and transcription terminator of the Zea mays LIP gene; GenBank access L35913).
The resulting Tobacco vector, pDAB105900 (Figure 7), was transferred to Agrobacterium tumefaciens using electroporation. After validating the restriction enzyme, Agrobacteríum was stored as glycerol stocks until used. The corn vector, pDAB105908 (Figure 8), was added and purified using the Qiagen QIAfilter plasmid Giga kit (Qiagen, Valencia, CA) according to the manufacturer's protocol
Example 1.2: Vectors for the expression of eZFNs
ZFN vectors that express the appropriate recognition helices in either a canonical (C2H2) or non-canonical (C3H) skeleton were prepared essentially as described in US Patent Publication 2008/0182332 and 2008/0159996.
The function of ZFNs was tested at the eZFN multiple insertion site as described in Example 1.1 inserted in a ZFN yeast screening system (see, US Patent Publication 2009 / 0.111.119). All ZFN pairs tested were active in the yeast system.
Eight eZFNs are cloned into vectors that contain the necessary regulatory sequences for expression in plant cells. The cloning strategies deployed for the constructs are essentially as described in US Patent Publications 2009 / 0111188A1 and US 20100199389. Figures 9 and 10 show generalized eZFN expression cassette schemes.
Example 2: Evaluation of eZFNs in Maize
Example 2.1: DNA distribution mediated by WISKERS ™
Cultures of corn embryogenic Hi-ll cells were produced, and were used as a source of live plant cells where integration was demonstrated. One skilled in the art can imagine using cell cultures derived from a variety of plant species, or differentiated plant tissues derived from a variety of plant species, as the source of living plant cells where integration has been demonstrated. .
In this example, a plasmid (pDAB105908) containing a selectable PAT plant marker cassette and the multi-eZFN binding site insertion sequence was used to generate the transgenic events. The transgenic isolates were transformed with eZFNs to assess the double-stranded divage.
62/88
In particular, 12 ml of packed cell volume (PCV) from a previously cryopreserved cell line plus 28 ml of conditioned medium was subcultured into 80 ml of GN6 liquid medium (N6 medium (Chu et al. (1975) Scientia Sin 18: 659-668), 2.0 mg / L 2, 4-D, 30 g / L of sucrose, pH 5.8) in a 500 ml Erlenmeyer flask, and placed on a shaker at 125 rpm at 28 ° Ç. This step was repeated twice using the same cell line so that a total of 36 ml of PCV was distributed between 3 vials. After 24 hours, the GN6 liquid media was removed and replaced with 72 ml of GN6 S / M osmotic medium (N6 medium, 2.0 mg / L 2,4-D, 30 g / L sucrose, 45.5 g / L of sorbitol, 45.5 g / L mannitol, 100 mg / L of myo-inositol, pH 6.0). The flask was incubated in the dark for 30-35 minutes at 28 ° C with moderate agitation (125 rpm). During the incubation period, a 50 mg / ml suspension of silicon carbide capillary crystals (Advanced Composite Materials, LLC, Greer, SC) was prepared by adding 8.1 ml of liquid GN6 S / M to 405 mg sterile capillary crystals of silicon carbide.
After incubation in GN6 S / M osmotic medium, the contents of each flask were collected in a 250 ml centrifuge container. After all the cells in the flask had settled to the bottom, the excess content of about 14 ml of liquid GN6 S / M was removed and collected in a sterile 1 L flask for future use. The pre-moistened suspension of capillary crystals was mixed at maximum speed in a vortex for 60 seconds and then added to the centrifugal vessel.
In this example, 170 pg of fragment purified from plasmid DNA pDAB105908 was added to each container. Once the DNA was added, the container was immediately placed in a modified commercial Red Devil 5400 ink mixer (Red Devil Equipment Co., Plymouth, MN) and stirred for 10 seconds. After shaking, the cocktail of cells, media, hair crystals and DNA was added to the contents of a 1 L flask, along with 125 ml of fresh GN6 liquid medium to reduce osmolarity. The cells were allowed to recover from
63/88 a stirrer set at 125 rpm for two hours. Six mL of a dispersed suspension was filtered over 5.5 cm Whatman # 4 filter paper using a glass cell collection unit connected to a vacuum line in the shelter such that 60 filters were obtained per container.
The filters were placed on 60 x 20 mm plates of solid GN6 medium (the same as GN6 liquid medium, except with 2.5 g / L of gelling agent Gelnte) and cultured at 28 ° C under dark conditions for one week.
Example 2.2. Identification and Isolation of the Putative Transgenic Event 10 One week after DNA distribution, filter papers were transferred to 60X20 mm plates of GN6 (1H) selection medium (N6 medium, 2.0 mg / L 2, 4-D, 30 g / L sucrose, 100 mg / L myo-inositol, 2.5 g / L Gelrite, pH 5.8) containing a selective agent. These selection plates were incubated at 28 ° C for one week, in the dark. After a week of drying in the dark, the tissue was incorporated into fresh media by scraping half the cells from each plate into a tube containing 3.0 ml of GN6 agarose medium maintained at 37-38 ° C (medium N6, 2 , 0 mg / L 2, 4-D, 30 g / L of sucrose, 100 mg / L of myo-inositol, 7 g / L of SeaPlaque agarose, pH 5.8, autoclaved for only 10 minutes at 121 ° C) .
20 The agarose / tissue mixture was broken with a spatula, and subsequently, 3 ml of agarose / tissue mixture was uniformly poured onto the surface of a 100 x 15 mm petri dish containing GN6 medium (1H). This process was repeated for both halves of each plate. Once all the tissue was incorporated, the plates were individually sealed with NESCOFILM® or PARAFILM M®, and grown at 28 ° C in dark conditions for up to 10 weeks.
Putatively transformed isolates growing under these selection conditions were removed from the incorporated plates and transferred to fresh selection medium in 60 x 20 mm plates. If sustained growth was evident after approximately two weeks, an event was considered resistant to the applied herbicide (selective agent) and an aliquot of cells was subsequently harvested for genotype analysis.
64/88
Example 2.3: Extraction of Genomic DNA
Genomic DNA (gDNA) was extracted from isolated corn cells as described in Example 2.2 and used as a template for PCR genotyping experiments. gDNA was extracted from approximately 100-300 µl volume of the packaged cell (PCV) of Hi-ll callus which was isolated as described above according to the manufacturer's protocols detailed in the DNeasy Plant 96 Kit (QIAGEN Inc., Valencia, HERE). Genomic DNA was eluted in 100 μΙ of elution buffer provided by the kit giving final concentrations of 20-200 ng / μΙ and subsequently analyzed using genotyping PCR methods described below.
Example 2.4: Molecular Analysis of Number of Copies
TAQMAN® assays were performed with screening samples of herbicide-resistant callus to identify those that contained single copy integration of the pDAB105908 transgene. Detailed analysis was conducted using specific primers and probes for gene expression cassettes. Single copy events have been identified for further analysis.
Customized TaqMan® assays were developed for analysis of the PAT gene in Hi-ll callus by third generation Technologies (Madison, Wl). The genomic DNA samples were first denatured in 96-well plates formatted by incubation at 95 ° C and then cooled to room temperature. Then, the master mix (containing PAT probe mix and an internal reference gene for adding buffer) was added to each well and the samples were covered with mineral oil. The plates were sealed and incubated in a TETRAD BioRad® thermocycler. The plates were cooled to room temperature before being read in a fluorescence plate reader. All plates contained one copy, two copies and 4 standard copies, as well as wild-type control samples and blank wells that did not contain a sample. The readings were collected and compared for development at level zero (ie, background), each channel was determined for each sample 65/88 by the raw sample signal divided by the raw signal number of the model.
From these data, a standard curve was constructed and the best fit determined by linear regression analysis. Using the parameters identified from this adjustment, the number of apparent PAT copies was then estimated for each sample.
Example 2.5: Initiator model for PCR genotyping
In this example, PCR genotyping was understood to include, but not limited to, polymerase chain reaction amplification10 se of genomic DNA (PCR) derived from isolated corn callus tissue predicted to contain donor DNA embedded in the genome, followed by standard cloning and sequence analysis of PCR amplification products. PCR genotyping methods have been well described (for example, Rios, G. etal. (2002) Plant J. 32: 243-253) and can be applied to genomic DNA derived from any plant species or tissue type, including cell cultures.
One skilled in the art can devise strategies for PCR genotyping that include (but are not limited to) amplifying specific sequences in the plant genome, amplifying 20 multiple specific sequences in the plant genome, amplifying non-specific sequences in the genome of the plant, or combinations thereof. Amplification can be followed by cloning and sequencing, as described in this example, or by analyzing the direct sequence of the amplification products. One skilled in the art can imagine alternative methods for analyzing the amplification products generated in this document. In an embodiment described here, primers specific for the target gene are used in PCR amplifications.
In the examples presented here, a primer oligonucleotide is synthesized, for example, by Integrated DNA Technologies, Inc. (Coralville, Iowa), under standard desalination conditions and diluted with water to a concentration of 100 μΜ. The primer oligonucleotide was modeled for annealing to the flanking regions of the DNA insert. The primers were tested using dilutions of plasmid DNA in the presence of DNA isolated from non-transgenic plants. The pDAB105908 transgene was amplified by PCR from genomic DNA from putative events using the primers. The resulting fragment was cloned into a plasmid vector and sequenced to confirm that the multi-eZFN binding site sequence was fully integrated into the plant's genome during transformation.
Example 2.6: Selection of Transgenic Events with the target DNA
Low copy events (1-2) were screened by PCR for the intact multi-eZFN binding site sequence and for the PAT gene. The copy number was confirmed by Southern analysis using standard methods with a PAT gene probe. The calluses of the intact inserts of transgenic events selected from carriers of a single copy, were kept for later evaluation with transiently expressed eZFNs.
Example 3. Distribution of eZFN DNA in Plant Cells
In order to allow eZFN-mediated double strand dividing, it is understood that the distribution of eZFN-encoding DNA followed by expression of the functional eZFN protein in the plant cell is necessary. One skilled in the art can imagine that the expression of functional ZFN protein can be achieved by several methods, including, but not limited to, transgenesis of the ZFN coding construct, or the transient expression of the ZFN coding construct.
In the examples cited here, methods are described for delivering eZFN encoding DNA to plant cells. One skilled in the art can use any of a variety of DNA delivery methods appropriate for plant cells, including, but not limited to, Agrobacterium-mediated transformation, biobalistic-based DNA distribution or WHISKERS ™ -mediated DNA distribution In one embodiment described here, biobalistic-mediated DNA distribution experiments were performed using various DNA constructs encoding eZFN.
67/88
Example 3.1: Biobalanced Mediated DNA Distribution
As described above, cultures of corn embryogenic Hi-ll cells were produced, and were used as the source of live plant to evaluate eZFN function. One skilled in the art can imagine using cell cultures derived from a variety of plant species, or differentiated plant tissues derived from a variety of plant species, as the source of living plant cells in which the target integration is demonstrated.
Plasmids expressing one of eight eZFNs that bind to a specific target sequence on the multi-eZFN binding site, together with an internal control (IPK-1), were bombarded in a callus pool of 5-10 isolates transgenics.
The transgenic Hi-ll corn callus events were subcultured weekly in GN6 medium (1H). Seven days after culture, approximately 400 mg of cells were dispersed in a 2.5 cm diameter circle over the center of a 100x15 mm petri dish containing GN6 S / M solidified medium with 2.5 g / L of gelrite . The cells were grown under dark conditions for 4 hours. To coat the biological particles with DNA, 3 mg of gold particles of 0.6 micron in diameter were washed once with 100% ethanol, twice with sterile distilled water and resuspended in 50 pl of water in a siliconized Eppendorf tube . A total of 5 pg of plasmid DNA, 20 µl of spermidine (0.1 M) and 50 μΙ of calcium chloride (2.5 M) were added separately to the gold suspension and mixed gently over a vortex. The mixture was incubated at room temperature for 10 min, pelleted at 10,000 rpm in a bench microcentrifuge for 10 seconds, resuspended in 60 μΙ of 100% cold ethanol, and 8-9 μ | were distributed in each macrocarrier.
The bombardment was carried out using the biological PDS1000 / HE ™ system (Bio-Rad Laboratories, Hercules, CA). Plates containing the cells were placed on the middle shelf under conditions of 1100 psi and 27 inches of Hg vacuum, and were bombarded following the manual
68/88 operational. Twenty-four hours after the bombardment, the tissue was transferred in small clusters of solid GN6 medium.
Example 4: Solexa Analysis and Sequencing
Example 4.1: Sample Preparation
Seventy-two hours after bombardment with eZFNs and an IPK1-ZFN control (Shukla et al. (1990) Nature 459, 437-441), the tissue was collected in 2 ml microcentrifuge tubes and lyophilized for at least 48 hours . Genomic DNA was extracted from lyophilized tissue using a QIAGEN® gDNA extraction kit according to the manufacturer's specifications. Finally, the DNA was resuspended in 200 µl of water and the concentration was determined using a Nanodrop spectrophotometer (Thermo Scientific, Wilmington, DE). DNA integrity was estimated by running all samples on 0.8% agarose E-gel (Invitrogen, Carlsbad, CA). All samples were normalized (25 ng / ul) for PCR amplification to generate amplified fragments for Solexa sequencing.
The PCR primers for region amplification encompassing each of the eZFN dividing sites, as well as the IPK1-ZFN target site of the target (treated ZFN) and control samples were acquired from IDT (Integrated DNA Technologies, San Jose, CA). The optimal amplification conditions for these primers were identified by PCR gradient using appropriate 0.2 μΜ primers, the Accuprime pfx Supermix (1.1X, Invitrogen, Carlsbad, CA) and 100 ng of model genomic DNA in a reaction of 23, 5 pL Cycling parameters include an initial denaturation at 95 '(5 min), followed by 35 denaturation cycles (95 ° C, 15 sec), annealing [55-72 ° C, 30 s] extension, (68 ° C, 1 min) and a final extension (72 ° C, 7 min). The amplification products were analyzed in 3.5% TAE agarose gels. After identifying an optimal annealing temperature, preparative PCR reactions were performed to validate each set of PCR primers and to generate the Solexa amplicon. The oligonucleotides used for the amplification of target regions of corn and tobacco eZFN are shown in Table 3 below. The IPK1 target regions were
69/88 using the primers (SEQ ID NO: 27 GCAGTGCATGTTATGAGC (forward primer) and SEQ ID NO: 28 CAGGACATAAATGAACTGAATC (reverse primer)).
Table 3: Starter Sequences Used to Amplify eZFN Cleavage Sites .________
Name ofInitiator Seq ID No.: Sequence Name ofInitiator Seq ID N “: Sequence SP / ALPR SEQ ID No.; 29 GGCACAGAGTAA-GAGGAAAA ASP / ALPR SEQ ID NO: 38 GCAGTGCTCTGTGGGGTC SP / CLAR SEQ ID NO: 30 AGGGACCCAGGTATACATTT ASP / CLAR SEQ ID N °: 39 CCTGGACAGTTGTCAAAATT SP / CLPR SEQ ID N “: 31 CATTCCGCCCTTGCCAGC ASP / CLPR SEQ ID NO: 40 GTGAACTTAT-TATCCATCTGICC SP / CL: RR SEQ ID NO: 33 GACAATGCCT-GACTCCCG ASP / CLRR SEQ ID N “: 41 CACTCAGACAC-CAGGGTTT SP / PLAR SEQ ID NO: 34 CAAGGAATGAAT-GAAACCG ASP / PLAR SEQ ID NO: 42 AGCCGGGAGAT-GAGGAAG SP / RL: AR SEQ ID NO: 35 CTGCAGGAGA-CAGGTGCC ASP / RLAR SEQ ID NO: 43 CCTGGGCTGCTTCACAAC SP / RLCR SEQ ID NO: 36 CAATCCCCACCCA-ACACT ASP / RLCR SEQ ID N ° 44 AGGAGGGT-GATGGTGAGG SP / RL: PR | SEQ ID NO: 37 CCTGGGGAGTAG-CAGTGTT________ ASP / RLPR I SEQIDN °: 45 TGTGATTAC-TACCCTGCCC I
For preparative PCR, 8 individual small-scale PCR reactions were completed for each model using the conditions described above, and the products were pooled and gel purified on 3.5% agarose gels using Qiagen MinElute ™ gel purification kit. The concentrations of the gel-purified amplicons were determined using a Nanodrop spectrophotometer, and Solexa samples were prepared by pooling approximately 100 ng of amplified from target eZFN and corresponding to the wild type controls, as well as the normalization of wild type controls. and IPK-1 target. From the target samples eZFN + IPK-1
70/88 the target IPK-1 sample and wild-type controls, four final Solexa samples comprising amplicons were generated and sequenced. The amplicons were cloned in PCR-Blunt ll-TOPO (Invitrogen) and subjected to sequencing to validate the primers before Solexa sequencing.
Example 4.2: Solexa Analysis and Sequencing
Solexa sequencing resulted in the production of thousands of sequences. The sequences were analyzed using Next Generation Sequence (NGS) DAS analysis scripts. Low quality sequences (sequences with a cut-off quality of <5) were filtered. The sequences were then aligned with the reference sequence and scored for insertions / deletions (Indels) at the ZFN dividing site caused by ZFN-mediated divination and NHEJ-mediated repair, which often causes indels that are indicative of ZFN activity. The editing activity was determined by the number of deletions greater than one bp within the sequence of the gap between the binding sites for the ZFN proteins after subtracting the background activity. The activity for each eZFN in the study was calculated compared to the wild-type and normalized control for ZFN IPK-1 activity. The normalized activities for each eZFN were then compared to classify the eZFNs used in the study. The activity was also evaluated at the level of sequence alignment (reference compared to the production of Solexa) by the presence of indels at the eZFN dividing site.
As shown in Figure 11, seven of the eight eZFNs show activity editing in corn.
Example 5: Evaluation of eZFNs in Tobacco
Example 5.1: Stable Integration of MultieZFN Binding Site Sequence
To make transgenic plant events with an integrated copy of the multi-eZFN binding site sequence described above, leaf discs (1 cm) cut from Petit Havana tobacco plants (for example, event 1585-10 containing a site of binding of ZFN-IL1 previously inte71 / 88 degrees), aseptically grown in MS medium (Phytotechnology Labs, Shawnee Mission, KS) and 30 g / L of sucrose in PhytaTrays (Sigma, St. Louis, MO), were floated in a culture overnight at Agrobacteríum LBA4404 harboring plasmid pDAB105900 grown at ODeoo ~ 1> 2, blotted dry on sterile filter paper and then placed on the same medium with the addition of 1 mg / L of indolacetic acid and 1 mg / L of benzamine purine in 60 x 20 mm plates (5 discs per plate). After 72 hours of co-culture, the leaf discs were transferred to the same medium with 250 mg / L of cefotaxime and 5 mg / L of BASTA®. After 3-4 weeks, the seedlings were transferred to MS medium with 250 mg / L of cephotaxime and 10 mg / L of BASTA® in PhytaTrays for an additional 2-3 weeks before leaf harvest and molecular analysis.
Example 5.2: Number of Copies and PTU Analysis of Multi-eZFN Binding Site Transgenic Sequence Events
DNA isolation. Transgenic tobacco plant tissue was harvested from BASTA® resistant seedlings and lyophilized for at least 2 days in 96-well collection plates. The DNA was then isolated using the 96-well DNeasy ™ extraction kit (Qiagen, Valencia, CA), following the manufacturer's instructions. A Kleco model 2-96A tissue sprayer (Garcia Manufacturing, Visalia CA) was used to break the tissue.
DNA quantification. The resulting genomic DNA was quantified using the QUANT-IT® Pico Green DNA assay kit (Molecular Probes, Invitrogen, Carlsbad, CA). Five pre-quantified DNA standards ranging from 20 ng / pL to 1.25 ng / pL (diluted in series) were used to generate the standard curve. The unknown samples were first diluted in 1:10 or 1:20 dilutions to be within the linear range of the assay. 5 pL of diluted samples and standards were mixed with 100 pL of diluted Pico Green substrate (1: 200) and incubated for ten minutes in the dark. The fluorescence was then recorded using a Synergy2 plate reader (Biotek, Winooski, VT). The concentration of genomic DNA was estimated from the standard curve calculated after background fluorescence corrections. Using TE or water, the DNA was then diluted to a common concentration of 10 ng / pL using a Biorobot3000 automated liquid handler (Qiagen).
Estimated Number of Copies. Putative transgenic events were analyzed for the complexity of DNA integration using multiplexed DNA hydrolysis probe assays that is analogous to the TAQMAN® assay The number of copies of the multisite construct was estimated using sequence specific primers and probes for the PAT transgene and a reference gene for endogenous tobacco, PAL. The assays for both genes were modeled using Realtime LIGHTCYCLER® PCR probe modeling software 2.0 for both genes was evaluated using the LIGHTCYCLER®480 system (Roche Applied Science, Indianapolis, IN). For amplification, a master mix of probes, LIGHTCYCLER®480 was prepared in a final concentration of 1X in a multiplex © reaction of 10 pL volume containing 0.4 μΜ of each primer and 0.2 pM of each probe (Table 4 below ). A two-stage amplification reaction is performed with an extension at 58 ° C for 38 seconds with the acquisition of fluorescence. All samples were processed in triplicate and the average Ct values were used for analysis of each sample. The analysis of PCR data in real time was performed using LIGHTCYCLER® software using the relative quant module and was based on the AACt method. For this, a sample of gDNA from a single copy of the calibrator was included to normalize the results. The single copy calibrator event was identified by Southern analysis and was confirmed to have a single PAT gene insert.
73/88
Table 4: Primers and probes used in hydrolysis probe assays
PAT and PAL
NAME String (5’-3 ’) Type Probe TQPATS (SEQ ID NO: 9) ACAAGAGTGGATTGATGATCTAGAGAGGT Initiator AT TQPATA (SEQ ID NO: 10) CTTTGATGCCTATGTGACACGTAAACAGT Initiator AT TQPATFQ (SEQIDN °: 11) CY5-GGTGTTGTGGCTGGTATTGCTTACGCTGG-BHQ2 Probe Cy5 TQPALS (SEQ ID NO: 12) TACTATGACTTGATGTTGTGTGGTGACTGA Initiator AT TQPALA (SEQIDN °: 13) GAGCGGTCTAAATTCCGACCCTTATTTC Initiator AT TQPALFQ (SEQ ID NO: 14) 6FAM-AAACGATGGCAGGAGTGCCCTTTTTCTATCAAT-BHQ1 Probe 6FAM
PCR. Low copy events (1-2) were subsequently screened by PCR for the intact plant transcriptional unit (PTU) 5 for the PAT gene and an intact multi-eZFN binding site.
Example 6: eZFN Cleavage Test on the Multi-eZFN Binding Site Sequence
To test the ability of eZFNs to facilitate target cleavage in the integrated multi-eZFN binding site sequence, a transient assay 10 was used based on the transient expression of eZFN constructs via Agrobacterium coculture from transgenic tobacco leaf discs. The leaf disc slices (1 cm 2 ) of transgenic events containing a single, full-length copy of the multieZFN binding site sequence containing the construct (as well as a single, full-length copy of a construct of ZFN-IL1), were submerged in an overnight Agrobacterium culture grown at OD 60 o ~ 1.2, blotted dry with sterile filter paper and then placed on the same medium with the addition of 1 mg / L of acid indolacetic and 1 mg / L of benziamine purine. For each eZFN tested, three treatments were used: pDAB1601 (negative control
74/88
- PAT only), pDAB4346 only (positive control - ZFN-IL1 only) and pDAB4346 + pDABeZFN-X (ZFN-IL1 + eZFN to be tested) with twenty leaf discs per treatment.
Example 6.1: Sequence Analysis
Genomic DNA was isolated from disks of transgenic tobacco leaves treated with Agrobacterium, using a Qiagen DNA extraction kit. All treatments were in the genomic and duplicate DNA and all samples were resuspended in 100 pL of water and the concentrations were determined by Nanodrop. Equal amounts of genomic DNA from each replicate for individual treatments were pooled together and were used as a starting model for the generation of Solexa amplicon.
The PCR primers for amplifying regions that span the multi-eZFN binding site and target cleavage site (eZFN-treated) sequence and control samples were Integrated DNA Technologies (Coralville, Iowa) and were purified by HPLC. Optimal amplification conditions were identified by gradient PCR using 0.2 pM of suitable primers, Accuprime pfx Supermix (1.1x, Invitrogen, Carlsbad, CA) and 100 ng of model genomic DNA in a 23.5 pL reaction. The cycling parameters were the initial denaturation at 95 ° C (5 min), followed by 35 denaturation cycles (95 ° C, 15 sec), annealing [55-72 ° C, 30 sec], extension, (68 ° C , 1 min) and a final extension (72 ° C, 7 min). The amplification products were analyzed in 3.5% agarose TAE gels. After identifying an optimal annealing temperature (56.1 ° C), preparative PCR reactions were performed to validate each set of PCR primers and to generate the Solexa amplicon.
For preparative PCR, 8 individual small-scale PCR reactions were performed for each model using the conditions described above, and the products were grouped together and gel purified on 3.5% agarose gels using gel purification kit Qiagen MinElute. The concentrations of the gel-purified amplicons were determined using a Nanodrop spectrophotometer and approximately
75/88
200 ng of each amplicon was pooled together to prepare the final Solexa sequencing sample (800 ng of total sample). The amplicons were also cloned in PCR-Blunt ll-TOPO and subjected to normal sequencing to validate the primers before Solexa sequencing. The analysis of Solexa (Shendure et al. (2008) Nat. Biotechnology, 26: 1135-1145) was performed and the sequences were analyzed.
Example 6.2: Solexa Sequencing and Analysis
Solexa sequencing was performed resulting in the production of thousands of sequences. The sequences were analyzed using DAS NGS analysis scripts. Low quality sequences (sequences with a cut quality score <5) were filtered. The sequences were then aligned with the reference sequence (pDAB105900 containing the multi-eZFN binding site) and scored for insertions / deletions (Indels) at the divage site. The editing activity (NHEJ%) for each untreated eZFN and control was calculated (number of high quality strings with indels / total number of high quality strings x 100) and are shown in Figure 12 below. The activity of 8-eZFNs in two transgenic tobacco events (105900 / # 33 and 105900 / # 45) has been demonstrated (Figure 12). Three of the eight eZFNs were active in the two transgenic tobacco events tested. The activity was also evaluated at the level of sequence alignment (reference vs production of Solexa) by the presence of indels at the eZFN dividing site in samples treated with eZFN.
All combinations of ZFN monomer halves (right and left) were active in the yeast assay. The data described for the maize and tobacco experiments show that some or most of the combinations are active in plants, supporting the possibility of using a significant number of exchanges of the two ZFN monomers from the original four ZFNs selected for the study.
Example 7: Intra-allelic recombination
Intra-allelic recombination allows the development and optimization of two independent blocks of transgenes, which can then be stacked together in one locus, by recombination. To enhance
76/88 the level of recombination between the two blocks, the double-stranded divage initiates DNA exchange by gene conversion or chromatid exchange.
To demonstrate this concept in plants, transgenic inserts illustrated in Figure 13 are made in Arabidopsis thaliana. The constructs include blocks of genes that contain a selectable marker (neomycin phosphotransferase (NPTII) or hygromycin phosphotransferase (HPT) and a marker capable of being scored (β-glucuronidase (GUS) or yellow fluorescent protein (YFP)). genes are in the identical genomic location, but shifted approximately 2 kb apart. The recombination between the two blocks is achieved by combining chromosomes that transport each of the two blocks in a single plant by crossing and then re-forming the plant progeny expressing a ZFN that cleaves in a central location between the two blocks (black bar above MIS in Figure 13). The ZFN are expressed using a specific meiosis / preferred promoter. The landing pad sequences that are used include those described in US Patent Application 61 / 297,641, incorporated herein by reference.
In order to generate two independent blocks at an identical genomic location, a construct was made comprising both blocks in a contiguous arrangement (Figure 14). To create the plants that carry the independent blocks individually, each block is excised at separate crossings using pmodelated ZFNs to cut the DNA from both sides of the respective block at the corresponding ZFN binding sites (red and blue bars). Figure 15 illustrates that the blocks are excised, generating single block inserts, after crossing with appropriate strains (Arabidopsis expressing ZFNs). These strains have the PAT gene as the selectable marker. The recovery of plants with the expected phenotypes (HygR +, KanR-, PAT +, YFP + or KanR +, HygR-, PAT +, GUS +) is confirmed through phenotype screening (resistance to herbicides for the HygR, kanR and PAT genes or marker gene expression capable of scoring GUS and YFP) or by molecular analyzes such as PCR and Southerns. Plants carrying one of the two different blocks are crossed to
77/88 generate progeny of HygR +, KanR +, PAT-, GUS +, YFP +.
After the molecular characterization of the resulting plants, the plants with the confirmed insert are crossed with the strains expressing a ZFN whose binding site is located between the two blocks 5 using a specific meiotic promoter to effect the DNA exchange. This results in the stacking of the two blocks together at a DNA location. The final stacked gene plants carry the HygR +, KanR +, GUS +, YFP + configuration as a single, segregated locus. Alternatively, plants containing one of the blocks are crossed with one of the 10 two monomers comprising the ZFN media and promoter / constructs, the plants homozygous for the two inserts obtained and then crossed together.
Example 7.1: DNA construction
The cloning strategies deployed for constructs 15 of the ZFN constructs were essentially as described in US Patent Publications 2009 / 0111188A1 and 2010/0199389. Figure 9 represents an exemplary eZFN expression cassette. ZFN coding sequences were expressed using the ZmUbil promoter (promoter, 5 'untranslated region (UTR) and intron derived from the Zea mays ubiquitin 1 (Ubi-1) gene;
Christensen et al. (1992) Plant Molec. Biol. 18 (4), 675-89). These were subsequently cloned into a binary GATEWAY ™ target vector containing an actinal rice promoter directing PAT gene expression. The resulting plasmids pDAB105951 (ZFN1; CL.AR), 105954 (ZFN8; RL: AR), 105952 (ZFN3; AL: PR), 105953 (ZFN6; CL: RR) designated as 25 Block Excisor constructs (eZFN1, 8) or Bloco2Excisor (eZFN3, 6), respectively, were transferred to Agrobacterium DA2552recA strain.
The Agrobacterium DA2552 strain was made competent for electroporation by preparing a starter culture, inoculating DA2552 strain from a glycerol stock in 10 ml spectinomycin containing 30 YEP (spec) (100 pg / mL) and erythromycin (ery) (150 pg / ml). The 10 ml culture was incubated overnight at 28 ° C at 200 rpm. Five ml of the starter culture was used to inoculate 500 ml of YEP with the appropriate antibiotics
78/88 in an appropriately marked 1.5 L Erlenmeyer flask. The culture was incubated overnight at 28 ° C at 200 rpm. After overnight incubation, the culture was cooled, placing it in an ice water bath and gently shaking. The cells were maintained at 4 ° C for all other 5 steps. The cells were pelleted by centrifugation at 4000 xg for 10 min. at 4 ° C in a marked sterile centrifuge container in a previously cooled rotor. The supernatant was decanted and discarded, then 5 to 10 ml of sterile bidistilled ice water was added, and the cells were gently pipetted up and down until no 10 pellets remained. The suspension volume was adjusted to about 500 ml with sterile bidistilled ice water. The cells were pelleted by centrifugation at 4000 xg for 10 min. at 4 ° C in a previously cooled rotor. The supernatant was discarded and 5 to 10 ml of sterile bidistilled ice water was added, then a sterile wide-bore pipette was used to gently pipette the cells up and down until no pellets remained. The suspension volume was adjusted to approximately 250 ml with sterile bidistilled ice water and the cells were pelleted again by centrifugation at 4000 x g for 10 min. at 4 ° C in a previously cooled rotor. The supernatant was discarded and 5 to 20 10 ml of sterile bidistilled ice water was added, the pellet was resuspended gently and the final volume was adjusted to 50 ml with sterile bidistilled ice water. The cells were pelleted by centrifugation at 4000 xg for 10 min. at 4 ° C in a previously cooled rotor. The cells were resuspended in a final volume of 5 ml of sterile glycerol cooled with 10% (v / v) ice. The cells were placed in 50 pl aliquots in sterile 0.5 ml microcentrifuge tubes and frozen in liquid nitrogen.
Twenty microliters of competent DA2552 cells were electroporated with 50 ng of plasmid DNA using a 30 GENE PULSER® XCELL® electroporation system (BioRad Hercules, CA.). According to the predefined configurations and manufacturing protocols for electroporation of Agrobacteríum. The cells recovered for two hours in SOC at 28 ° C and
79/88 then plated on YEP spec / ery agar plates and cultured for 48 h at 28 ° C.
Example 7.2: Exchange Locus Construct
The Locus DNA construct was prepared from GATEWAY ™ input 5 vectors including vector 1: AtAct2 promoter (AtAct2 ^ 2 promoter (promoter, untranslated region 5 θ intron of an Arabinopsis thaliana (ACT2) actin gene; An et al. (1996) Plant J. 10, 107-121)) / GUS (Jefferson, (1987) EMBO J. 6, 3901-3907) / AtuORF23 3 'UTR (3' untranslated region (UTR), comprising the transcriptional terminator and polyadenic site 10 open reader structure 23 (ORF23) of Agrobacterium tumefaciens pTi15955; Barker et al., (1983) Plant Molec. Biol. 2 (6): 335-50) :: AtAct2 / NPTII promoter ( Bevan et al. (1983) Nature 304, 184-187) / AtuORF23 3 'UTR, flanked by eZFNs 1 and 8, vector 2: synthetic 2 kb region with eZFN 4 and 7 in the sequence center, and vector 3: promoter CsVMV / HPT (Kaster et al.
(1983) Nucleic Acids Res. 11 (19), 6895-6911 (1983)) / AtuORF23 3 ’promoter
RTU :: AtUbi10 (promoter, 5 'untranslated region and gene intron of Arabidopsis thaliana polyubiquitin 10 (UBQ10); Norris et al. (1993) Plant Molecular Biology 21 (5) .895-906) / PhiYFP (Shagin et al ., (2004) MolecularBiol. Evol. 21: 841-850) / AtuORF23 3 'UTR, flanked by eZFNs 3 and 6. The target vector was prepared by inserting two randomized 1 kb synthetic DNA sequences into one Agrobacterium binary vector skeleton, with restriction sites included to clone a GATEWAY ™ ccc / B negative selectable marker cassette. The input vectors were cloned into the target vector using a Clonase LR reaction. The resulting vector, pDAB100646 (Figure 16) was transferred to Agrobacterium as described above.
Example 7.3: Transformation of Arabidopsis
All Arabidopsis transformations were performed following the methods described by Clough & Bent (1998 Plant J., 16, 735-743). 30 Exciting Strains
The excisor strain construct has the phosphinothricin acetyltransferase (PAT) gene that transmits resistance to glufosinate. Seven, ten and
80/88 thirteen days after planting, the Ti plants were sprayed with a 284 mg / L solution of Liberty herbicide (200 grams of active ingredient per liter (g ai / L) glufosinate, Bayer Crop Sciences, Kansas City, MO) with a spray volume of 10 ml / tray (703 L / ha), using a DeVilbiss compressed air spray tip 5 to deliver an effective rate of 200 g ai / ha of glufosinate per application. Survivors (growing plants) were identified 4-7 days after the final spray and transplanted individually in 3-inch pots prepared with substrates (Metro Mix 360).
The expression of eZFNs in the Exciter events is determined by
Reverse transcriptase PCR (RT PCR) and the number of copies determined by qPCR as described herein from the PAT gene and confirmed by Southern analysis. Three low copy events that express ZFNs at a high level are crossed into Locus switch events.
Exchange Locus Strains 15 Exchange Locus Strains are generated in Arabidopsis following the methods described by Clough & Bent (1998 Plant J., 16, 735743), including selection in the medium containing hygromycin or kanamycin. Example 7.4: Crossing Arabidopsis and Progeny Recovery
The crossing of exchange Locus events with the two 20 sets of Blocol and Block2 Exciter lines is done using standard methods.
The crossing seeds are grown in hygromycin (Blocol deletion) or kanamycin (Block2 deletion) and resistant plants analyzed for GUS expression (Blocol deletion) or YFP 25 expression (Block2 deletion). GUS activity is determined with a histochemistry assay (Jefferson et al. (1987) Mol. Plant. Biol. Rep 5, 387-405) and YFP by fluorescence microscopy. Plants with the desired phenotypes (positive block: GUS +, TNP +, HPT, YFP; positive block 2: GUS-, TNP-, HPT +, YFP +) are analyzed by PCR and Southerns to confirm the desired gene configuration. The leaves of the selected plants are painted with a bialaphos solution to assess which are PAT +.
Plants containing Blocol and Bloco2 gene cassettes
81/88 are crossed and the progeny selected on hygromycin / kanamycin plates. HygR / kanR plants are analyzed for the presence of all genes by PCR and phenotype screening. Fí plants with the desired phenotype are grown and crossed with ZFN / meiosis promoters to achieve the recombination between Blocol and Bloco2. The resulting progenies are grown on hygromycin / kanamycin plates. The plants that survived the selection are selected for GUS and YFP. Confirmation and characterization of the recombinants is done using PCR analysis, Southerns, sequencing and segregation.
Example 8: Gene Stacking on eZFN Sites
The strategies shown in Figures 1, 2, 4, 5 and 6 can be carried out using the following methods.
Construct Model
Various combinations of heterodimeric eZFN sites can be assembled as a concatomer in a plasmid vector suitable for plant transformation. Figure 1, Figure 2, and Figure 4 illustrate various versions of heterodimeric eZFN sites that can be incorporated into a vector and transformed into a plant chromosome. Transformations with WHISKERS ™
Cultures of corn embryogenic Hi-ll cells are produced as described in the US Patent. 7,179,902, and are used as the source of living plant cells in which the target integration is exemplified. DNA fragments containing the eZFN heterodimeric sites linked to a selectable plant marker cassette are used to generate transgenic events. Transgenic events are isolated and characterized.
Twelve mL of packed cell volume (PCV) from a cryopreserved cell line plus 28 mL of conditioned medium are subcultured into 80 mL of liquid GN6 medium (N6 medium (Chu et al., (1975) Sei Sin. 18: 659-668), 2.0 mg / L 2, 4-D, 30 g / L of sucrose, pH 5.8) in a 500 mL Erlenmeyer flask, and placed on a shaker at 125 rpm at 28 ° C. This step is repeated twice using the same cell line, such that a total of 36 mL of PCV is distributed through three vials.
82/88
After 24 hours, the GN6 liquid media is removed and replaced with 72 mL of GN6 S / M osmotic medium (N6 medium, 2.0 mg / L 2,4-D, 30 g / L sucrose, 45.5 g / L of sorbitol, 45.5 g / L of mannitol, 100 mg / L of myoinositol, pH 6.0). The flask is incubated in the dark for 30-35 minutes at 5 28 ° C, with moderate shaking (125 rpm). During the incubation period, a 50 mg / ml (w / v) suspension of silicon carbide capillary crystals (Advanced Composite Materials, LLC, Greer, SC) is prepared by adding 8.1 ml of GN6 S liquid medium / M for 405 mg of capillary crystals of silicon carbide. After incubation in GN6 S / M osmotic medium, the contents of 10 each flask are grouped in a 250 ml centrifuge container. After all cells in the flask settle to the bottom, the excess volume content of approximately 14 mL of GN6 S / M liquid is removed and collected in a sterile 1 L flask for future use. The pre-moistened suspension of capillary crystals is mixed at a maximum speed in 15 vortex for 60 seconds and then added to the centrifuge bottle.
An 85 pg aliquot of purified DNA fragment is added to each centrifuge container. Once the DNA is added, the container is immediately placed in a commercially-modified Red Devil 20 5400 ink mixer (Red Devil Equipment Co., Plymouth, MN), and stirred for 10 seconds. After shaking, the cocktail of cells, media, hair crystals and DNA is added to the contents of a 1 L flask, along with 125 mL of fresh GN6 liquid medium to reduce osmolarity. The cells are allowed to recover on a shaker set at 125 rpm for two hours. 6 mL of a dispersed suspension is filtered over Whatman # 4 paper filter (5.5 cm) using a glass cell collection unit connected to a vacuum line in the shelter such that 60 filters are obtained per flask. The filters are placed on 60 x 20 mm plates of solid GN6 medium (same as liquid GN6 medium except with 2.5 g / L 30 of Gelrite gelling agent) and grown at 28 ° C under dark conditions for one week.
83/88
Identification and isolation of putative transgenic target-interaction events
One week after DNA distribution, filter papers were transferred to 60X20 mm plates of GN6 (1H) selection medium (N6 medium, 5 2.0 mg / L 2, 4-D, 30 g / L of sucrose, 100 mg / L myo-inositol, 2.5 g / L Gelrite, pH 5.8) containing a selective agent. These selection plates were incubated at 28 ° C for one week, in the dark. After a week of selection in the dark, the tissue was incorporated in fresh media by scraping half the cells from each plate into a tube containing 3.0 ml of 10 GN6 agarose medium maintained at 37-38 ° C (N6, 2 medium , 0 mg / L 2, 4-D, 30 g / L of sucrose, 100 mg / L of myo-inositol, 7 g / L of SeaPlaque® agarose, pH 5.8, autoclaved for only 10 minutes at 121 ° C ).
The tissue / agarose mixture is broken with a spatula and subsequently 3 ml of tissue / agarose mixture is evenly poured onto the surface of a 100 mm x 25 plate containing GN6 medium (1H). This process is repeated for the two halves of each plate. Once all of the tissue is incorporated, the plates are grown at 28 ° C in the dark for up to 10 weeks. Putatively transformed isolates that grew under these selection conditions are removed from the embedded plates and transferred to fresh selection medium in 60 x 20 mm plates. If sustained growth is evident after about two weeks, an event is considered resistant to the applied herbicide (selective agent) and an aliquot of cells is subsequently harvested for analysis of the genotype. The stable plant transformation produces members of single copies that 25 are used for stacking experiments.
Molecular Characterization of Events
Genomic DNA (gDNA) is extracted from isolated maize cells described and used as a model for PCR genotyping experiments. gDNA is extracted from approximately 100-300 uL of 30 volume of packaged cell (PCV) of Hi-ll callus which is isolated according to the manufacturer's protocols detailed in the DNEASY® 96 Plant Kit (QIAGEN lnc „Valencia, CA ). Genomic DNA is eluted in 100 pl of tam84 / 88 elution bread provided by the kit, producing final concentrations of 20-200 ng / pL, and subsequently analyzed using PCR-based genotyping methods.
Molecular Analysis of Number of Copies
Hydrolysis probe or INVADER® assays are performed with herbicide resistant callus screening samples to identify those that contain the single copy integration of T-filament DNA. Detailed analysis is conducted using specific primers and probes for expression cassettes of genes. Single copy events are identified for further analysis.
Customized INVADER® assays are developed for analysis of the selectable marker gene in Hi-ll callus by third generation Technologies (Madison, Wl). Genomic DNA samples were amplified using the INVADER® assay kit and readings were collected. From these readings, sometimes above zero (ie, background) for each channel are determined for each sample by the raw sample signal divided by the model's raw signal number. From these data, a standard curve is constructed and the best fit determined by linear regression analysis. Using the parameters identified from this adjustment, the number of apparent selectable marker copies is then estimated for each sample.
Selection of Transgenic Events with target DNA
Low copy events (1-2 copies of the transgene) are screened by PCR, as described above, for an intact plant transcriptional unit (PTU) containing the selectable marker gene cassette and the intact eZFN site. The number of copies is further confirmed by Southern analysis using standard methods with the selectable marker gene. Calluses from selected transgenic events harboring intact single copy inserts are maintained.
Biolistic-mediated DNA distribution in plant cells that contain an eZFN
As described above, bryogenic 85/88 Hi-ll corn cell cultures are produced, and are used as the source of live plant cells in which target integration is demonstrated. Embryogenic corn suspensions are subcultured in liquid GN6 medium approximately 24 hours before experimentation, as described above. The excess liquid medium is removed and approximately 0.4 mL of cell PCV is dispersed within a 2.5 cm diameter circle over the center of a 100x15 mm petri dish containing GN6 S / M solidified medium with 2 , 5 g / L of gelrite.
The cells are grown under dark conditions for 4 hours. To coat the biolistic particles with DNA containing a donor DNA fragment (Blocol in Figure 1, Block 2 in Figure 2, or gene 1 in Figure 4), 3 mg of 1.0 micron diameter gold particles were washed once once with 100% ethanol, twice with sterile distilled water, and resuspended in 50 pl of water in a siliconized Eppendorf tube. A total of 5 μg of plasmid DNA (containing, in a single vector or in separate vectors nucleic acid molecules encoding the DNA fragment and donor eZFN), 20 μΙ of spermidine (0.1 M) and 50 μΙ of chloride of calcium (2.5 M) are added separately to the gold suspension and mixed in a vortex. The mixture is incubated at room temperature for 10 min, pelleted at 10,000 rpm in a bench microcentrifuge for 10 seconds, resuspended in 60 μΙ of 100% cold ethanol, and 8-9 μ | are distributed in each macrocarrier.
Bombardment is carried out using the biological PDS1000 / HE ™ system (Bio-Rad Laboratories, Hercules, CA). Plates containing the cells are placed in the middle of the shelf under conditions of 1,100 psi and 27 inches of Hg vacuum, and are bombarded following the operating manual. Sixteen hours after bombardment, the tissue is transferred in small clusters of GN6 medium (1H) and grown for 2-3 weeks at 28 ° C under dark conditions. Transfers continue every 2-4 weeks until putative transgenic isolates resulting from donor DNA integration begin to appear. Bialaph-resistant colonies are generally analyzed by PCR and Southern blotting using methods
86/88 described above to generate the isolates containing the target sequences. Screening for Target Integration Events via PCR Genotipaqem
PCR reactions are performed to investigate the presence of an intact copy of the donor DNA. Additional reactions focus on the 5 'boundary between the target and the donor and the 3' boundary between the donor and the target. The amplified fragments are excised on gel and purified according to standard protocols. Purified fragments are subsequently cloned into plasmid pCR2.1 using TOPO TA CLONAGEM® kit (with pCR2.1 vector) and chemically competent E. coli cells ONE SHOT® TOP10 (Life Technologies Invitrogen, Carlsbad, CA) according to the manufacturer's protocol .
Individual colonies are selected and confirmed to contain the amplicon by PCR. Double-stranded sequencing reactions of plasmid clones are performed to confirm that the PCR-amplified genomic sequence contains the integrated donor. Events identified to contain the donor fragment represent a target in which the homology-led repair of a ZFN-mediated double strand break and a target integration of a donor DNA into a specific target gene. Specific Gene Stacking Application Using eZFN Sites
Figure 1 shows the variations of multiple insertion sites made of seven (7) target sites of eZFN stably transformed on the chromosome of a plant. EZFN pairs that bind to target sites are described as geometric figures. Blocol is an exogenous polynucleotide sequence that can be integrated into the multiple insertion site of the appropriate eZFN pair when transformed with an eZFN modeled to cleave a specific eZFN site. The cotransformation of the eZFN and Blocol from the donor DNA sequence can be achieved using a biological transformation method, previously described above. The fidelity of several other eZFN sites is maintained since eZFN transformed into the plant cell does not cleave to these other sites. Blocol integrates into the plant's chromosome through homologous recombination, resulting in plant cells that contain the Blocol sequence. The resulting plant cells po87 / 88 can be grown in mature plants and screened for the presence of BlocoT 'using analytical molecular biology methods known in the art, such as Southern blotting Taqman assay, or Invader assay.
Figure 2 illustrates another variation of Figure 1, in which a binding site other than eZFN is the target of a Block 2 polynucleotide donor sequence. The resulting integration of the DNA fragment produces a stable plant containing Block 2 within the chromosome.
Figure 4 illustrates the use of eZFN domains on the left and right. The top line represents the genome of a plant transformed with the left and right eZFN domains (shaded triangle and checkered triangle). When the appropriate eZFN is added in the presence of an exogenous molecule including Gene 1 flanked by new and different heterodimeric eZFN sites, Gene 1 and flanking eZFN sites are inserted into the genome. The resulting progeny containing Gene 1 and the flanking eZFN sites are identified and these plants can later be redirected using new heterodimeric eZFN sites that were not present on the source plant (ie, eZFN sites containing the shaded triangle and triangle checkered).
Figure 5 and Figure 6 illustrate how eZFN sites can be used to stack new transgenes at a chromosomal site. In addition, this strategy allows excision of other gene expression cassettes. In some cases a gene expression cassette can be completely removed (Figure 5), in other scenarios the gene expression cassette can be removed in a specific generation of plants and, eventually, be reintroduced into the progeny of these plants, thus allowing recycling a gene expression cassette. A deleted marker sequence (Figure 6) can be reintroduced via homologous recombination-mediated gene target using the protocol described above. Target gene for heterodimeric eZFN sites is completed using the protocol described above. In this example, eZFN binding sites are used to allow in-plant deletion of any transgene, including selectable marker genes, from a transformed plant. See Pe88 / 88 Provisional Patent Order US 61 / 297,628, filed January 22, 2010, hereby incorporated by reference.
All patents, patent applications and publications mentioned herein are hereby incorporated by reference, in their entirety, for all purposes.
Although disclosure has been provided in some detail by way of illustration and examples for the sake of clarity of understanding, it will be apparent to those skilled in the art that various changes and modifications can be practiced without departing from the spirit or scope of the disclosure10. Therefore, the preceding descriptions and examples should not be construed as limiting.
权利要求:
Claims (16)
[1]
1. Isolated nucleic acid, comprising a plurality of target sites paired to one or more pairs of zinc finger nucleases.
[2]
2. Nucleic acid molecule, as defined in claim 1, wherein a target site from each paired target site comprises the same sequence.
[3]
Nucleic acid molecule, as defined in claim 1 or 2, further comprising one or more coding sequences.
[4]
A cell or cell line, comprising a nucleic acid molecule as defined in any one of claims 1 to 3.
[5]
A cell according to claim 1, wherein the cell is a eukaryotic cell, such as a plant cell or mammalian cell or mammalian cell line.
[6]
A cell according to claim 4 or 5, wherein the nucleic acid molecule is integrated into the cell's genome.
[7]
7. Method for integrating one or more exogenous sequences into the genome of a cell, the method comprising:
(a) providing one or more pairs of zinc finger nucleases to a cell as defined in claim 6, wherein the zinc finger nucleases bind to a target site on the integrated nucleic acid molecule, so that the binding from nucleases to their target sites cleaves the cell's genome; and (b) contacting the cell with a polynucleotide comprising an exogenous sequence, wherein the exogenous sequence is integrated into the cell genome within the integrated nucleic acid.
[8]
8. The method of claim 7, further comprising repeating steps (a) and (b) with additional zinc finger nucleases that cleave additional target sites on the integrated nucleic acid molecule in the presence of additional exogenous sequences, thus inserting additional exogenous sequences into the cell's genome.
[9]
A method according to claim 8, wherein one or more
2/3 more of the exogenous sequences comprise one or more target sites for zinc finger nucleases.
[10]
10. The method of claim 9, wherein the target site is a zinc finger nuclease target site means, wherein by target site integration a target site is created.
[11]
A method according to any one of claims 7 to 10, wherein one or more of the exogenous sequences comprises a coding sequence and the cell expresses the product of the coding sequence.
[12]
12. Method for deleting one or more sequences inserted into a cell's genome, the method comprising:
(a) integrating a plurality of exogenous sequences as defined in claims 7 to 11; and (b) expressing the appropriate nucleases in the cell, so that one or more of the exogenous sequences are deleted from the genome.
[13]
13. Method for providing a genomically altered cell, the method comprising:
(a) integrating or deleting one or more exogenous sequences in at least one first cell according to the method as defined in any one of claims 7 to 12;
(b) allowing the first cell to develop into a sexually mature organism; and (c) crossing the organism with a second organism comprising genomic changes to generate a second cell with at least one of the genomic changes of the first and second organisms.
[14]
14. The method of claim 13, wherein the genomic changes in the second cell comprise a plurality of heterologous genes at a single genomic site in the second cell.
[15]
A method according to any one of claims 7 to 14, wherein the cell further comprises modifications of its genome outside the region comprising the integrated nucleic acid molecule.
3/3
[16]
16. Method according to any one of claims 7 to
14, wherein the cell is a plant cell.
类似技术:
公开号 | 公开日 | 专利标题
JP5902631B2|2016-04-13|Targeted genome modification
JP6886433B2|2021-06-16|Methods and compositions for incorporating exogenous sequences into the plant genome
JP6442442B2|2018-12-19|Zinc finger nuclease-mediated homologous recombination
JP6127297B2|2017-05-17|Targeted integration into the ZP15 locus
AU2013256240B2|2018-09-20|Targeted modification of malate dehydrogenase
US20180163218A1|2018-06-14|Reconstruction of site specific nuclease binding sites
AU2017216446A1|2017-08-31|Zinc finger nuclease-mediated homologous recombination
BRPI0922641B1|2021-10-26|METHOD OF INTEGRATION OF ONE OR MORE EXOGENOUS NUCLEIC ACID SEQUENCES INTO THE GENOME OF A PLANT CELL
BRPI0716427B1|2017-08-29|METHOD FOR INTRODUCING AN EXOGENOUS SEQUENCE IN THE GENOME OF A PLANT CELL
同族专利:
公开号 | 公开日
CN106086047B|2021-07-06|
UA118328C2|2019-01-10|
US10260062B2|2019-04-16|
CO6561839A2|2012-11-15|
CN102812034A|2012-12-05|
CN102812034B|2016-08-03|
CA2787494C|2019-09-17|
EA031322B1|2018-12-28|
IL221029D0|2012-09-24|
AU2011207769A1|2012-08-02|
AR080009A1|2012-03-07|
CN106086047A|2016-11-09|
KR20120117890A|2012-10-24|
CA2787494A1|2011-07-28|
MX336846B|2016-02-03|
MX2012008490A|2013-04-05|
WO2011090804A1|2011-07-28|
CL2012002033A1|2012-12-14|
IL221029A|2019-10-31|
JP5902631B2|2016-04-13|
NZ601247A|2014-10-31|
EA201201036A1|2013-01-30|
EP2526112A1|2012-11-28|
JP2013517770A|2013-05-20|
EP2526112B1|2018-10-17|
AU2011207769B2|2015-05-28|
KR101866578B1|2018-06-11|
US20110189775A1|2011-08-04|
EP2526112A4|2013-12-04|
引用文献:
公开号 | 申请日 | 公开日 | 申请人 | 专利标题

US4217344A|1976-06-23|1980-08-12|L'oreal|Compositions containing aqueous dispersions of lipid spheres|
US4235871A|1978-02-24|1980-11-25|Papahadjopoulos Demetrios P|Method of encapsulating biologically active materials in lipid vesicles|
US4186183A|1978-03-29|1980-01-29|The United States Of America As Represented By The Secretary Of The Army|Liposome carriers in chemotherapy of leishmaniasis|
US4261975A|1979-09-19|1981-04-14|Merck & Co., Inc.|Viral liposome particle|
US4485054A|1982-10-04|1984-11-27|Lipoderm Pharmaceuticals Limited|Method of encapsulating biologically active materials in multilamellar lipid vesicles |
US4501728A|1983-01-06|1985-02-26|Technology Unlimited, Inc.|Masking of liposomes from RES recognition|
US4946787A|1985-01-07|1990-08-07|Syntex Inc.|N--dialkyloxy)- and N--dialkenyloxy)-alk-1-yl-N,N,N-tetrasubstituted ammonium lipids and uses therefor|
US4897355A|1985-01-07|1990-01-30|Syntex Inc.|N[ω,-dialkyloxy]- and N-[ω,-dialkenyloxy]-alk-1-yl-N,N,N-tetrasubstituted ammonium lipids and uses therefor|
US5049386A|1985-01-07|1991-09-17|Syntex Inc.|N-ω,-dialkyloxy)- and N--dialkenyloxy)Alk-1-YL-N,N,N-tetrasubstituted ammonium lipids and uses therefor|
US4774085A|1985-07-09|1988-09-27|501 Board of Regents, Univ. of Texas|Pharmaceutical administration systems containing a mixture of immunomodulators|
US4837028A|1986-12-24|1989-06-06|Liposome Technology, Inc.|Liposomes with enhanced circulation time|
US5140081A|1989-03-21|1992-08-18|Ciba-Geigy Corporation|Peroxide free radical initiators containing hindered amine moieties with low basicity|
US5264618A|1990-04-19|1993-11-23|Vical, Inc.|Cationic lipids for intracellular delivery of biologically active molecules|
AU7979491A|1990-05-03|1991-11-27|Vical, Inc.|Intracellular delivery of biologically active substances by means of self-assembling lipid complexes|
SE466987B|1990-10-18|1992-05-11|Stiftelsen Ct Foer Dentaltekni|DEVICE FOR DEEP-SELECTIVE NON-INVASIVE, LOCAL SEATING OF ELECTRICAL IMPEDANCE IN ORGANIC AND BIOLOGICAL MATERIALS AND PROBE FOR SEATING ELECTRICAL IMPEDANCE|
US5487994A|1992-04-03|1996-01-30|The Johns Hopkins University|Insertion and deletion mutants of FokI restriction endonuclease|
US5436150A|1992-04-03|1995-07-25|The Johns Hopkins University|Functional domains in flavobacterium okeanokoities restriction endonuclease|
US5356802A|1992-04-03|1994-10-18|The Johns Hopkins University|Functional domains in flavobacterium okeanokoites restriction endonuclease|
JP4012243B2|1994-01-18|2007-11-21|ザスクリップスリサーチインスティチュート|Zinc finger protein derivatives and methods therefor|
US6242568B1|1994-01-18|2001-06-05|The Scripps Research Institute|Zinc finger protein derivatives and methods therefor|
US6140466A|1994-01-18|2000-10-31|The Scripps Research Institute|Zinc finger protein derivatives and methods therefor|
US5877302A|1994-03-23|1999-03-02|Case Western Reserve University|Compacted nucleic acids and their delivery to cells|
AU698152B2|1994-08-20|1998-10-22|Gendaq Limited|Improvements in or relating to binding proteins for recognition of DNA|
US5789538A|1995-02-03|1998-08-04|Massachusetts Institute Of Technology|Zinc finger proteins with high affinity new DNA binding specificities|
US5925523A|1996-08-23|1999-07-20|President & Fellows Of Harvard College|Intraction trap assay, reagents and uses thereof|
GB9703369D0|1997-02-18|1997-04-09|Lindqvist Bjorn H|Process|
GB2338237B|1997-02-18|2001-02-28|Actinova Ltd|In vitro peptide or protein expression library|
GB9710809D0|1997-05-23|1997-07-23|Medical Res Council|Nucleic acid binding proteins|
GB9710807D0|1997-05-23|1997-07-23|Medical Res Council|Nucleic acid binding proteins|
US7102055B1|1997-11-18|2006-09-05|Pioneer Hi-Bred International, Inc.|Compositions and methods for the targeted insertion of a nucleotide sequence of interest into the genome of a plant|
US6410248B1|1998-01-30|2002-06-25|Massachusetts Institute Of Technology|General strategy for selecting high-affinity zinc finger proteins for diverse DNA target sites|
WO1999045132A1|1998-03-02|1999-09-10|Massachusetts Institute Of Technology|Poly zinc finger proteins with improved linkers|
US6140081A|1998-10-16|2000-10-31|The Scripps Research Institute|Zinc finger binding domains for GNN|
GB9824544D0|1998-11-09|1999-01-06|Medical Res Council|Screening system|
US7013219B2|1999-01-12|2006-03-14|Sangamo Biosciences, Inc.|Regulation of endogenous gene expression in cells using zinc finger proteins|
US6453242B1|1999-01-12|2002-09-17|Sangamo Biosciences, Inc.|Selection of sites for targeting by zinc finger proteins and methods of designing zinc finger proteins to bind to preselected sites|
US6534261B1|1999-01-12|2003-03-18|Sangamo Biosciences, Inc.|Regulation of endogenous gene expression in cells using zinc finger proteins|
US6599692B1|1999-09-14|2003-07-29|Sangamo Bioscience, Inc.|Functional genomics using zinc finger proteins|
AT309536T|1999-12-06|2005-11-15|Sangamo Biosciences Inc|METHODS OF USING RANDOMIZED ZINCFINGER PROTEIN LIBRARIES FOR IDENTIFYING GENERAL FUNCTIONS|
US6435261B1|1999-12-30|2002-08-20|Wayne D. Ailiff|Mechanism for inverting the cope of a molding flask|
AT355368T|2000-01-24|2006-03-15|Gendaq Ltd|NUCLEIC ACID BINDING POLYPEPTIDE IS IDENTIFIED BY FLEXIBLE LINKER-LINKED NUCLEIC ACID ACID|
WO2001059450A2|2000-02-08|2001-08-16|Sangamo Biosciences, Inc.|Cells expressing zinc finger protein for drug discovery|
US20020061512A1|2000-02-18|2002-05-23|Kim Jin-Soo|Zinc finger domains and methods of identifying same|
US20030044787A1|2000-05-16|2003-03-06|Joung J. Keith|Methods and compositions for interaction trap assays|
JP2002060786A|2000-08-23|2002-02-26|Kao Corp|Germicidal stainproofing agent for hard surface|
US6794136B1|2000-11-20|2004-09-21|Sangamo Biosciences, Inc.|Iterative optimization in the design of binding proteins|
GB0108491D0|2001-04-04|2001-05-23|Gendaq Ltd|Engineering zinc fingers|
DE10131786A1|2001-07-04|2003-01-16|Sungene Gmbh & Co Kgaa|Recombination systems and methods for removing nucleic acid sequences from the genome of eukaryotic organisms|
EP1421177A4|2001-08-20|2006-06-07|Scripps Research Inst|Zinc finger binding domains for cnn|
CA2474486C|2002-01-23|2013-05-14|The University Of Utah Research Foundation|Targeted chromosomal mutagenesis using zinc finger nucleases|
US20030232410A1|2002-03-21|2003-12-18|Monika Liljedahl|Methods and compositions for using zinc finger endonucleases to enhance homologous recombination|
CA2497913C|2002-09-05|2014-06-03|California Institute Of Technology|Use of chimeric nucleases to stimulate gene targeting|
US8409861B2|2003-08-08|2013-04-02|Sangamo Biosciences, Inc.|Targeted deletion of cellular DNA sequences|
JP4555292B2|2003-08-08|2010-09-29|サンガモバイオサイエンシズインコーポレイテッド|Methods and compositions for targeted cleavage and recombination|
US7888121B2|2003-08-08|2011-02-15|Sangamo Biosciences, Inc.|Methods and compositions for targeted cleavage and recombination|
BRPI0509460B8|2004-04-30|2017-06-20|Dow Agrosciences Llc|herbicide resistance genes|
WO2007014181A2|2005-07-25|2007-02-01|Johns Hopkins University|Site-specific modification of the human genome using custom-designed zinc finger nucleases|
JP2009502170A|2005-07-26|2009-01-29|サンガモバイオサイエンシズインコーポレイテッド|Targeted integration and expression of foreign nucleic acid sequences|
CA2651494C|2006-05-25|2015-09-29|Sangamo Biosciences, Inc.|Engineered cleavage half-domains|
AT536371T|2006-05-25|2011-12-15|Sangamo Biosciences Inc|PROCESS AND COMPOSITIONS FOR GENERIC ACTIVATION|
CN105296527B|2006-08-11|2020-11-27|陶氏益农公司|Zinc finger nuclease-mediated homologous recombination|
WO2008060510A2|2006-11-13|2008-05-22|Sangamo Biosciences, Inc.|Zinc finger nuclease for targeting the human glucocorticoid receptor locus|
KR101574529B1|2006-12-14|2015-12-08|다우 아그로사이언시즈 엘엘씨|Optimized non-canonical zinc finger proteins|
US8110379B2|2007-04-26|2012-02-07|Sangamo Biosciences, Inc.|Targeted integration into the PPP1R12C locus|
BRPI0817560A2|2007-09-27|2015-07-14|Dow Agrosciences Llc|Engineered zinc claw proteins that target 5-enolpiruvil shiquimato-3-phosphate synthase genes|
EP2188384B1|2007-09-27|2015-07-15|Sangamo BioSciences, Inc.|Rapid in vivo identification of biologically active nucleases|
EP2195438B1|2007-10-05|2013-01-23|Dow AgroSciences LLC|Methods for transferring molecular substances into plant cells|
EP2597155B1|2007-10-25|2016-11-23|Sangamo BioSciences, Inc.|Methods and compositions for targeted integration|
CA2720903C|2008-04-14|2019-01-15|Sangamo Biosciences, Inc.|Linear donor constructs for targeted integration|
ES2654923T3|2008-12-17|2018-02-15|Dow Agrosciences Llc|Directed integration in Zp15 location|
MX2012008485A|2010-01-22|2012-08-17|Dow Agrosciences Llc|Excision of transgenes in genetically modified organisms.|
WO2011090804A1|2010-01-22|2011-07-28|Dow Agrosciences Llc|Targeted genomic alteration|WO2011090804A1|2010-01-22|2011-07-28|Dow Agrosciences Llc|Targeted genomic alteration|
US8771985B2|2010-04-26|2014-07-08|Sangamo Biosciences, Inc.|Genome editing of a Rosa locus using zinc-finger nucleases|
EP2597943B1|2010-07-29|2018-02-07|Dow AgroSciences LLC|Strains of agrobacterium modified to increase plant transformation frequency|
BR112013024337A2|2011-03-23|2017-09-26|Du Pont|complex transgenic trace locus in a plant, plant or seed, method for producing in a plant a complex transgenic trace locus and expression construct|
RU2658437C2|2012-05-02|2018-06-21|ДАУ АГРОСАЙЕНСИЗ ЭлЭлСи|Targeted modification of malate dehydrogenase|
US10174331B2|2012-05-07|2019-01-08|Sangamo Therapeutics, Inc.|Methods and compositions for nuclease-mediated targeted integration of transgenes|
WO2014036219A2|2012-08-29|2014-03-06|Sangamo Biosciences, Inc.|Methods and compositions for treatment of a genetic condition|
BR112015004948A2|2012-09-07|2017-11-21|Dow Agrosciences Llc|fad2 performance loci and target corresponding site-specific binding proteins capable of inducing targeted breakdowns|
JP6775953B2|2012-09-07|2020-10-28|ダウ アグロサイエンシィズ エルエルシー|FAD3 performance locus and corresponding target site-specific binding proteins capable of inducing targeted cleavage|
EP2893025B1|2012-09-07|2019-03-13|Dow AgroSciences LLC|Engineered transgene integration platformfor gene targeting and trait stacking|
KR102192599B1|2013-04-05|2020-12-18|다우 아그로사이언시즈 엘엘씨|Methods and compositions for integration of an exogenous sequence within the genome of plants|
US9873894B2|2013-05-15|2018-01-23|Sangamo Therapeutics, Inc.|Methods and compositions for treatment of a genetic condition|
CA2926822A1|2013-11-04|2015-05-07|Dow Agrosciences Llc|Optimal soybean loci|
NZ719494A|2013-11-04|2017-09-29|Dow Agrosciences Llc|Optimal maize loci|
KR102269769B1|2013-11-04|2021-06-28|코르테바 애그리사이언스 엘엘씨|Optimal maize loci|
AU2014341934B2|2013-11-04|2017-12-07|Corteva Agriscience Llc|Optimal soybean loci|
US10774338B2|2014-01-16|2020-09-15|The Regents Of The University Of California|Generation of heritable chimeric plant traits|
SI3194570T1|2014-09-16|2022-01-31|Sangamo Therapeutics, Inc.|Methods and compositions for nuclease-mediated genome engineering and correction in hematopoietic stem cells|
CN107249605A|2014-11-17|2017-10-13|阿迪塞特生物股份有限公司|The gamma delta T cells of engineering|
US20160244784A1|2015-02-15|2016-08-25|Massachusetts Institute Of Technology|Population-Hastened Assembly Genetic Engineering|
KR102194612B1|2015-03-16|2020-12-23|인스티튜트 오브 제네틱스 앤드 디벨롭멘털 바이오롤지,차이니즈 아카데미 오브 사이언시스|Site-specific modification method of plant genome using non-genetic material|
US20180142249A1|2016-11-21|2018-05-24|Dow Agrosciences Llc|Site specific integration of a transgne using intra-genomic recombination via a non-homologous end joining repair pathway|
CN108220339A|2016-12-14|2018-06-29|深圳华大生命科学研究院|The method of cell transformation|
EP3555285A4|2016-12-14|2020-07-08|Dow AgroSciences LLC|Reconstruction of site specific nuclease binding sites|
CN107784199A|2017-10-18|2018-03-09|中国科学院昆明植物研究所|A kind of organelle gene group screening technique based on STb gene sequencing result|
AU2018370120A1|2017-11-15|2020-06-25|Adicet Bio, Inc.|Methods for selective expansion of d3 yd T-cell populations and compositions thereof|
EP3890757A1|2018-12-03|2021-10-13|Adicet Bio Inc.|Methods for selective in vivo expansion of gamma delta t-cell populations and compositions thereof|
WO2021028359A1|2019-08-09|2021-02-18|Sangamo Therapeutics France|Controlled expression of chimeric antigen receptors in t cells|
法律状态:
2020-02-11| B15I| Others concerning applications: loss of priority|Free format text: PERDA DA PRIORIDADE US 61/336,457 DE 22/01/2010 REIVINDICADA NO PCT/US2011/000125 POR NAO CUMPRIMENTO DA EXIGENCIA PUBLICADA NA RPI 2545 DE 15/10/2019 PARA APRESENTACAO DE DOCUMENTO DE CESSAO CORRETO. CESSAO APRESENTADA NAO POSSUI ASSINATURA DE TODOS OS INVENTORES DA PRIORIDADE REIVINDICADA. |
2020-02-27| B06F| Objections, documents and/or translations needed after an examination request according [chapter 6.6 patent gazette]|
2020-06-23| B12F| Other appeals [chapter 12.6 patent gazette]|
2021-06-08| B15K| Others concerning applications: alteration of classification|Free format text: AS CLASSIFICACOES ANTERIORES ERAM: C07H 21/02 , C12P 21/06 Ipc: A01H 1/06 (2006.01), C12P 21/06 (2006.01), C12N 15 |
2021-07-06| B06U| Preliminary requirement: requests with searches performed by other patent offices: procedure suspended [chapter 6.21 patent gazette]|
2021-10-13| B350| Update of information on the portal [chapter 15.35 patent gazette]|
2021-11-23| B06A| Patent application procedure suspended [chapter 6.1 patent gazette]|
2022-03-08| B11B| Dismissal acc. art. 36, par 1 of ipl - no reply within 90 days to fullfil the necessary requirements|
优先权:
申请号 | 申请日 | 专利标题
US33645710P| true| 2010-01-22|2010-01-22|
PCT/US2011/000125|WO2011090804A1|2010-01-22|2011-01-24|Targeted genomic alteration|
[返回顶部]