GENE TARGETING IN PLANTS USING DNA VIRUSES

Systems and methods for gene targeting in plants, including systems and methods that include the use of geminiviruses and customizable endonucleases.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims benefit of priority from U.S. Provisional Application Ser. No. 61/790,581, filed on Mar. 15, 2013, U.S. Provisional Application Ser. No. 61/772,704, filed on Mar. 5, 2013, and U.S. Provisional Application No. 61/661,542, filed on Jun. 19, 2012, all of which are incorporated herein by reference in their entirety.

STATEMENT AS TO FEDERALLY SPONSORED RESEARCH

This invention was made with government support under DBI-0923827 awarded by the National Science Foundation. The government has certain rights in the invention.

TECHNICAL FIELD

This document relates to materials and methods for gene targeting in plants, and particularly to methods for gene targeting that include using geminiviruses and customizable endonucleases.

BACKGROUND

The precise modification of higher eukaryotic genomes, including plant genomes, is a highly sought after technology for basic research and biotechnology applications. Precise genome modification—referred to herein as gene targeting (GT)—relies on the DNA-repair machinery of the target cell, and on an exogenously supplied repair template (also referred to as a “donor sequence”). Through the activity of the homologous recombination (HR) pathway, homologous sequences carried by the repair template can recombine with a chromosomal target. Consequently, any modified sequence carried by the repair template will be stably integrated into the genome. Attempts to implement GT in plants often are plagued by extremely low HR frequencies. The majority of the time, donor DNA molecules integrate illegitimately via non-homologous end joining (NHEJ). This process occurs regardless of the size of the homologous “arms,” as increasing the length of homology to approximately 22 kb results in no significant enhancement in GT (Thykjaer et al., Plant Mol. Biol., 35:523-530, 1997).

Other studies have aimed at increasing the efficiency of GT in plants. Some methods are based on the use of customizable endonucleases, such as zinc finger nucleases (ZFN5), meganucleases (MN5), and transcription activator-like (TAL) effector nucleases (TALE nucleases). A targeted DNA double-strand break (DSB) can stimulate recombination by a factor of 100 between transforming T-DNA and a native chromosomal locus (Puchta et al., Proc. Natl. Acad. Sci. USA, 93:5055-5060, 1996). Through the coordinated delivery of a repair template and a customizable endonuclease, high-frequency GT may be achieved in plants (Townsend et al., Nature, 459:442-445, 2009). Such methods are designed for use in protoplasts, which enables direct delivery of repair templates and nuclease-expressing plasmids to individual cells though PEG transformation or electroporation. However, the ability to practice GT is limited to labs with the expertise and equipment for tissue culturing and plant regeneration.

SUMMARY

Gene targeting in plant cells has been performed primarily by two techniques: (1) direct transfer of DNA into plant cells by either electroporation/PEG transformation of protoplasts, or by biolistic bombardment of DNA into various plant tissues; and (2) by Agrobacterium-mediated transformation. In these methods, the exogenously supplied DNA is either T-DNA, PCR-derived, or plasmid-derived.

This document is based in part on the development of a novel and effective in planta method for gene targeting that combines the use of geminiviral-based gene targeting vectors and a targeted DNA double strand break engineered by a co-delivered endonuclease. This is the first account demonstrating concurrent use of these techniques as a gene targeting methodology, which is likely to have vast implications in all areas of plant biology. For example, this technology can be used to accelerate the rate of functional genetic studies in plants. The technology also can be used to engineer plants with improved characteristics, including enhanced nutritional quality, increased resistance to disease and stress, and heightened production of commercially valuable compounds.

There are several benefits to using geminiviruses and endonucleases for gene targeting in plants, including (i) the ability of the virus to stably propagate the gene targeting vector from cell-to-cell within the plant, (ii) the ability of the virus to replicate the gene targeting vector to high copy numbers within plant cell nuclei (on average 1000 copies per cell, but numbers can reach up to 30,000), and (iii) the circular nature of the geminivirus genome, as circular DNA is thought to participate less frequently in illegitimate recombination. These properties contribute to an effective, reliable and reproducible procedure for gene targeting in plant cells.

The methods provided herein enable practitioners to achieve high frequency gene targeting by creating a chromosome break in a target locus while simultaneously using the viral replication machinery to make repair templates to achieve gene targeting. The viral repair templates can be generated either by infecting plants with engineered viruses or by using deconstructed viral vectors. The latter vectors replicate viral DNA and thereby produce the repair template, but they do not generate a productive infection.

In a first aspect, this disclosure features a method for modifying the genetic material of a plant cell. The method can include (a) introducing into the cell a virus nucleic acid comprising a repair template that is heterologous to the virus and is targeted to a first sequence that is endogenous to the plant cell; and (b) inducing a double strand break at or near the sequence to which the repair template is targeted, wherein said double strand break is generated by an endonuclease targeted to a second endogenous plant sequence at or near the first sequence that is targeted by the repair template, wherein homologous recombination occurs between the first endogenous plant sequence and the repair template.

The virus nucleic acid can be a plant DNA virus nucleic acid. The virus nucleic acid can be a geminivirus nucleic acid. The endonuclease can be a zinc finger nuclease, a transcription activator-like effector nuclease, a meganuclease, or a CRISPR/Cas system endonuclease. The endonuclease can be encoded by a transgene sequence stably integrated into the genetic material of the plant, or can be expressed transiently. When the endonuclease is encoded by a transgene, the transgene can be operably linked to a promoter that is constitutive, cell specific, inducible, or activated by alternative splicing of a suicide exon. The virus nucleic acid can include a sequence encoding the endonuclease. The method can further include introducing into the plant cell an RNA virus nucleic acid comprising a nucleotide sequence encoding the endonuclease. The RNA virus nucleic acid can be introduced into the plant cell after or simultaneous with step (a). The RNA virus nucleic acid can be from a tobacco rattle virus, a potato virus X, a pea early browning virus, or a barley stripe mosaic virus. The plant can be a monocotyledonous plant (e.g., wheat, maize, a grass such as purple false brome (Brachypodium distachyon), Haynaldia villosa, or Setaria), or a dicotyledonous plant (e.g., tomato, soybean, tobacco, potato, or Arabidopsis).

Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention pertains. Although methods and materials similar or equivalent to those described herein can be used to practice the invention, suitable methods and materials are described below. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting.

The details of one or more embodiments of the invention are set forth in the accompanying drawings and the description below. Other features, objects, and advantages of the invention will be apparent from the description and drawings, and from the claims.

DESCRIPTION OF DRAWINGS

FIG. 1 is an illustration of the cabbage leaf curl virus (CaLCuV) genome. CaLCuV contains a bipartite genome, with the DNA A component encoding proteins necessary for viral replication and encapsidation, and the DNA B component encoding proteins necessary for cell-to-cell movement. The coat protein nucleotide sequence (CP) can be replaced by up to 800 nucleotides of repair template DNA sequence. See, Gutierrez, Physiol. Mol. Plant Pathol. 6060:219-230, 2002.

FIG. 2 is a schematic of an experimental approach for gene targeting using engineered geminiviruses and transgenic Arabidopsis plants encoding a stably integrated zinc finger nuclease (ZFN) transgene. Repair of the ZFN-induced DSB using a repair template on the CaLCuV A genome results in the stable incorporation of a unique 18 bp sequence into the ADH1 gene.

FIG. 3 is an illustration of a nested PCR method that can be used to detect gene-targeted ADH1 alleles. Genomic DNA from somatic Arabidopsis cells—exposed to estradiol and infected with CaLCuV—is used as a template for PCR amplification of the ADH1 locus. Amplicons are gel purified and used as templates for a second PCR, with one primer specific for the GT modification. Dashed lines represent the outer limit of homology carried by the repair template.

FIG. 4A is a diagram of pCPCbLCVA.007, which contains the entire genome of the CaLCuV A component flanked by direct repeats of the common region for viral excision from the plasmid. To modify pCPCbLVCA.007 for carrying gene fragments, the coding region of the coat protein gene, AR1, was replaced with a polylinker. The AR1 promoter, the translational start (ATG) and the putative polyadenylation sites are retained. To initiate infection, this plasmid is co-transformed with pCPCbLCVB.002. Virus derived from these vectors moves from cell-to-cell within Arabidopsis plants but, without the coat protein gene, it is not transmissible.

FIG. 4B is a diagram of pCPCbLCVB.002, which contains the entire genome of the CaLCuV B component flanked by direct repeats of the common region for viral excision from the plasmid. Bombardment of the B component alone can be used as a negative control for DNA contamination (no virus should be replicated). See, Muangsan and Robertson, Meth. Mol. Biol. 265:101-15, 2004.

FIG. 5 is a picture of gels with amplicons generated from an enrichment PCR designed to detect ZFN-induced mutations at the ADH1 gene after induction by β-estradiol. DNA was assessed for NHEJ mutations from (i) non-induced and non-infected plants (−Estradiol, −Virus), (ii) induced and non-infected plants (+Estradiol, −Virus), (iii) non-induced and infected plants (−Estradiol, +Virus), and (iv) induced and infected plants (+Estradiol, +Virus). D, digested; UD, undigested.

FIG. 6 is a diagram of the CaLCuV A plasmid (left panel) and a series of pictures of gels showing the stability of repair template sequences in infected plants (right panels). Genomic DNA from infected plants was used as a template for PCR amplification of the repair template sequence. Primers NB153 and NB158 (left panel) recognize sequences in the viral genome and amplify across the repair template. Five differently sized repair templates were analyzed. Repair templates with sizes 400 nt, 600 nt, 800 nt, and 1000 nt contained ADH1 homology sequences, while 715 nt contained gus::nptII homology sequence. PCR amplicons (right panel) were run out on a 1% agarose gel. Controls for 1000 nt and 800 nt used plasmid DNA as a template for PCR (CaLCuVA.ADH1-1000 and CaLCuVA.ADH1-800, respectively).

FIG. 7 is a series of pictures of agarose gels showing PCR detection of amplicons from modified ADH1 loci. Genomic DNA from infected plants exposed to β-estradiol (left panels; +Virus, +Estradiol) or not exposed to β-estradiol (right panel; +Virus, −Estradiol) was subjected to nested PCR using primers designed to detect the 5′ modification junction (5′ check), the 3′ modification junction (3′ check), and amplification of the starting template (input).

FIG. 8 is a series of pictures showing evidence of GT at the gus::nptII gene. Co-infected plants (CaLCuVA.GUS-FIX and CaLCuVB with TRV-Zif268) were stained in X-Gluc and chlorophyll was removed. Images of selected plants are shown. Arrows point to blue-staining cells.

FIG. 9 is an illustration of a strategy for creating a geminivirus replicon (GVR) system for transient protein expression, and subsequently transient genome editing, in plants. LSL T-DNA functions as a template for Rep-assisted replicative release of replicons (top). LIR, SIR, and Rep/RepA nucleotide sequences were derived from Bean yellow dwarf virus (BeYDV, GenBank accession number DQ458791.1). Following delivery of LSL T-DNA to plant cell nuclei by Agrobacterium, Rep protein mediates replicational release of single-stranded DNA (ssDNA) replicons (middle). Complementary strand synthesis is carried out by host polymerases, resulting in transcriptionally-competent double-stranded DNA (dsDNA) replicons (bottom). Transcription of protein coding sequence is driven by the nearby LIR and further promoted with an upstream 2×35S promoter. SD, DEM2 splice donor; SA, DEM2 splice acceptor, LB, left border; RB, right border.

FIG. 10A is an illustration of an approach for cloning customizable endonucleases into pLSL. The pZHY013 entry vector, encodes unique restriction enzyme sites (XbaI, BamHI, NheI and BglII) for sequential cloning of nucleotide sequences for TALE or ZF binding domains.

FIG. 10B is an illustration of vectors for Gateway cloning of customizable endonucleases and repair templates into pLSL. FokI nucleotide sequences encode obligate heterodimeric proteins (EL-KK). Noteworthy, an AatII enzyme site permits cloning of Cas9 or MN nucleotide sequences upstream of Nos terminator sequence (Nos-T).

FIG. 10C is the full sequence of the LSL region (SEQ ID NO:78) located between the left and right T-DNA borders in pLSL. The hygromycin resistance gene, located between the left border and the upstream LIR, is not shown. The highly-conserved nonanucleotide sequence (TAATATTAC), required for Rep-initiated rolling circle replication, is underlined in both LIR elements.

FIG. 11 is an illustration showing the general structure of the replicase expressing T-DNA plasmids used in the experiments described herein. Rep/RepA nucleotide sequences (both wild type and LxCxQ) were cloned into pMDC32 (2×35S promoter) or pFZ19 (XVE promoter).

FIG. 12 is an image of plant tissue expressing GUS enzyme. LSL T-DNA, encoding NLS-tagged beta-glucuronidase (pLSLGUS), was delivered to Nicotiana tabacum var. xanthi leaf tissue with p35SREP (right side of leaf) or without p35SREP (left side of leaf) by syringe infiltration of Agrobacterium. Transformed leaf tissue was stained seven days post infiltration (dpi) with X-Gluc, and chlorophyll was removed to better visualize staining.

FIG. 13 is a series of images of plant tissue expressing GFP. Leaf tissue transformed with pLSLGFP, with and without delivery of p35SREP, or transformed with pLSLGUS with delivery of p35SREP, was visualized 3, 7, and 12 dpi.

FIG. 14 is an image of a representative leaf seven dpi, demonstrating tissue health. Leaf tissue from WT Nicotiana tabacum plants was syringe infiltrated with Agrobacterium containing pLSLGUS (right), or coinfiltrated with Agrobacterium containing pLSLGUS and p35SREP. Leaf tissue was removed from the plant seven dpi and imaged. Slight browning in tissue transformed with p35SREP was observed.

FIG. 15 is an illustration (top) and example (bottom) of detecting GVRs encoding GUS and GFP nucleotide sequences in plant cells. To assay for the presence of GVRs, genomic DNA was extracted three dpi and used as template for PCR. Primers were designed to amplify LIR sequence contained on the replicon. Amplicons were present only when p35SREP was co-transformed with pLSL, suggesting the presence of GVRs.

FIG. 16 is an illustration of target loci for Zif268::FokI, the T30 TALE nuclease pair, and the CRISPR/Cas system. ZFN target sequence is present within a stably integrated, and defective gus::nptII reporter gene (top). The T30 TALE nuclease and CRISPR/Cas target sequences are present within the endogenous acetolactate synthase genes (ALS), SuRA (middle) and SuRB (bottom). AI, artificial intron IV of ST-LS1 gene from Solanum tuberosum.

FIG. 17 is an image of a gel from a PCR designed to detect GVRs containing ZFN (pLSLZ.D), TALE nuclease (pLSLT), and CRISPR/Cas (pLSLC) sequences.

FIG. 18 is an image of a gel (middle) from a PCR-digest (top) designed to detect ZFN-induced mutations at the gus::nptII gene. Plant DNA was isolated from leaf tissue seven dpi. Amplicons encompassing the ZFN target site were digested overnight with MseI and separated on an agarose gel. Cleavage-resistant bands were cloned into pJet1.2 and sequenced (bottom).

FIG. 19 is an image of a gel (middle) from an enrichment PCR (top) designed to detect TALE nuclease-induced mutations at the ALS loci. Plant DNA was pre-digested overnight with AluI before PCR amplification of SuRA and SurB loci. Amplicons were digested overnight with AluI, separated on an agarose gel, and cleavage-resistant bands were cloned into pJet1.2 and sequenced (bottom).

FIG. 20 is an image of a gel (middle) from a PCR-digest (top) designed to detect Cas9-included mutations at the ALS loci. Plant DNA was isolated from leaf tissue five dpi and the CRISPR/Cas target site was amplified by PCR. The resulting amplicons were digested with AlwI, separated on an agarose gel, and cleavage resistant bands were cloned and sequenced (bottom).

FIG. 21 is a schematic outlining the approach to correct a non-functional gus::nptII reporter. Repair template sequence, present within pLSLZ.D, encodes 1 kb homology arms isogenic to gus::nptII sequence, as well as 600 bp of sequence designed to restore gus::nptII protein function.

FIG. 22 shows selected images leaf tissue with GUS-expressing cells. To visualize cells expressing functional GUS protein, leaf tissue was stained in X-Gluc solution for 24 to 48 hours at 37° C., and chlorophyll was removed. Images shown are selected examples from tissue transformed with p35SZ.D (left), pLSLZ.D (center), and both pLSLZ.D and p35SREP (right).

FIG. 23 is an image of a gel (bottom) from a PCR (top) designed to detect GUS::NPTII genes. PCR was performed on genomic DNA extracted from leaf tissue seven dpi. Primers were designed to be complementary to sequence downstream of the NPTII coding sequence and homologous to the sequence within the repair template (top). A high number of amplicons of the expected size (1.078 kb) were observed only from genomic DNA isolated from tissue transformed with pLSLZ.D and p35SREP.

FIG. 24 is a graph plotting the density of GUS-expressing cells across multiple transgenic lines (identified as 1.7, 4.3, 9.1, and 11.3). Error bars represent SEM of at least three biological replicates.

FIG. 25 is a series of graphs plotting the density of GUS-expressing cells with different transformed vectors. Error bars represent SEM of at least three biological replicates.

FIG. 26 is a series of images of leaf tissue with GUS-expressing cells following Agrobacterium-mediated delivery of pLSLZ.D and p35SREP to transgenic lines 1.7, 4.3, and 11.3, as indicated.

FIG. 27 is a series of images of leaf tissue with GUS-expressing cells following Agrobacterium-mediated delivery of pLSLZ.D to transgenic lines 1.7 and 11.3, as indicated.

FIG. 28 is a series of images of leaf tissue with GUS-expressing cells following Agrobacterium-mediated delivery of p35SREP to transgenic lines 1.7, 4.3, and 11.3, as indicated.

FIG. 29 is a series of images of leaf tissue with GUS-expressing cells following Agrobacterium-mediated delivery of pLSLD and p35SREP to transgenic lines 1.7, 4.3, and 11.3, as indicated.

FIG. 30 is a series of images of leaf tissue with GUS-expressing cells following Agrobacterium-mediated delivery of p35SZ.D and p35SREP to transgenic lines 1.7, 4.3, and 11.3, as indicated.

FIG. 31 is an illustration of the approach used to create a SuRB::NPTII fusion protein (top) and an image of two gels from PCRs designed to genotype candidate recombinant plants (bottom). Primers were designed to detect the 5′ modification junction (5′ check) and the 3′ modification junction (3′ check).

FIG. 32 is an image of a gel from a PCR designed to detect BeYDV-based GVRs in potato cells. Genomic DNA from plants co-transformed with p35SREP and pLSLGFP was evaluated for replicational release (top), and for the presence of Rep/RepA nucleotide sequence (bottom).

FIG. 33 is an image of a gel from a PCR designed to detect Rep/RepA RNA transcripts in potato plants transformed with p35SREP.

FIG. 34 is a pair of images of potato leaves expressing GUS enzyme. Potato leaves were transformed with Agrobacterium containing pLSLGUS (left) or a mixture of Agrobacterium containing pLSLGUS and p35SREP (right). Leaf tissue was stained in X-Gluc solution and chlorophyll was removed.

FIG. 35 is a series of images of tomato leaf tissue with GUS-expressing cells. Tomato leaf tissue was infiltrated with Agrobacterium containing pLSLGUS (right) or a mixture of Agrobacterium containing pLSLGUS and p35SREP (left and middle). To visualize cells expressing functional GUS protein, infected leaf tissue was stained in X-Gluc solution for 24 hours at 37° C., and chlorophyll was removed. Black arrows indicate areas of GUS activity.

FIG. 36 is an illustration showing the general structure of the Wheat dwarf virus LSL T-DNA. Rep/RepA nucleotide sequence is present within the LIR elements. Rep/RepA gene expression is initiated from the complementary sense LIR promoter.

FIG. 37 is a pair of images of wheat calli tissue expressing GFP. GFP sequence was delivered to calli by particle bombardment of plasmid DNA containing BeYDV LSL sequences (left) or WDV LSL sequences (right). Images were taken three dpi.

FIG. 38 is a set of images of Setaria calli expressing GFP. GFP sequence was delivered to calli by particle bombardment of plasmid DNA containing BeYDV LSL sequences (left) or WDV LSL sequences (right). Images were taken three dpi.

FIG. 39 is a set of images of corn embryos expressing GFP. GFP sequence was delivered to calli by particle bombardment of plasmid DNA containing BeYDV LSL sequences (left), WDV LSL sequences (middle), or control (right). Images were taken three dpi.

FIG. 40 is an illustration describing an approach to correct a non-functional gus::nptII reporter gene in rice (top) and pictures of GUS activity in rice leaves (bottom).

DETAILED DESCRIPTION

This document provides a highly efficient, virus-based system and methods for targeted modification of plant genomes. The in planta system and methods for GT include the use of customizable endonucleases in combination with plant DNA viruses. Plant DNA viruses, including geminiviruses, have many attributes that may be advantageous for in planta GT, including their ability to replicate to high copy numbers in plant cell nuclei. Importantly, these viruses can be modified to encode a desired nucleotide sequence, such as a repair template sequence targeted to a particular sequence in a plant genome. First generation geminiviruses, or “full viruses” (viruses that retain only the useful “blocks” of sequence), can carry up to about 800 nucleotides (nt), while deconstructed geminiviruses (viruses that encode only the proteins needed for viral replication) have a much larger cargo capacity. This document describes how customizable nucleases and plant DNA viruses enable in planta GT, and provides materials and methods for achieving such GT. The methods can be used with both monocotyledonous plants (e.g., banana, grasses (e.g., Brachypodium distachyon), wheat, oats, barley, maize, Haynaldia villosa, palms, orchids, onions, pineapple, rice, and sorghum) and dicotyledonous plants (e.g., Arabidopsis, beans, Brassica, carnations, chrysanthemums, citrus plants, coffee, cotton, eucalyptus, impatiens, melons, peas, peppers, Petunia, poplars, potatoes, roses, soybeans, squash, strawberry, sugar beets, tobacco, tomatoes, and woody tree species).

In general, the system and methods described herein include two components: a plant DNA virus (e.g., geminivirus) vector containing a repair template targeted to an endogenous plant sequence, and an endonuclease that also is targeted to a site near or within the target sequence. The endonuclease can be activated to create targeted DNA double-strand breaks at the desired locus, and the plant cell can repair the double-strand break using the repair template present in the geminivirus, thereby incorporating the modification stably into the plant genome.

Geminiviruses are a large family of plant viruses that contain circular, single-stranded DNA genomes. Examples of geminiviruses include the cabbage leaf curl virus, tomato golden mosaic virus, bean yellow dwarf virus, African cassava mosaic virus, wheat dwarf virus, miscanthus streak mastrevirus, tobacco yellow dwarf virus, tomato yellow leaf curl virus, bean golden mosaic virus, beet curly top virus, maize streak virus, and tomato pseudo-curly top virus. As described herein, geminivirus sequences can be used as gene targeting vectors. For example, the geminivirus genome can be engineered to contain a desired modification flanked by sequences of homology to a target locus. In some cases, this can be accomplished by replacing non-essential geminivirus nucleotide sequence (e.g., CP sequence) with a desired repair template. Other methods for adding sequence to viral vectors include, without limitation, those discussed in Peretz et al. (Plant Physiol., 145:1251-1263, 2007).

The repair template contains homology to a particular sequence within the genome of a plant. Typically, a repair template includes a nucleic acid that will replace an endogenous target sequence within the plant, flanked by sequences homologous to endogenous sequences on either side of the target. When a non-essential (e.g., CP) sequence within a geminivirus vector is replaced with a repair template, the repair template can have a length up to about 800 nt (e.g., 100 nt, 200 nt, 300 nt, 400 nt, 500 nt, 600 nt, 700 nt, 800 nt, or any length between about 100 nt and about 800 nt). Within the repair template, the flanking homologous sequences can have any suitable length (e.g., about 25 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, 250 nt, 300 nt, 350 nt, 400 nt, or any length between about 25 nt and about 400 nt). Repair templates and DNA virus plasmids can be prepared using techniques that are standard in the art, including those described below.

The second component of the system and methods described herein is an endonuclease that can be customized to target a particular nucleotide sequence and generate a double strand break at or near that sequence. Examples of such customizable endonucleases include ZFNs, MNs, and TALE nucleases, as well as Clustered Regularly Interspersed Short Palindromic Repeats/CRISPR-associated (CRISPR/Cas) systems. See, for example, Sander et al., Nature Methods, 8:67-69, 2011; Jacoby et al., Nucl. Acids Res., 10.1093/nar/gkr1303, 2012); Christian et al., Genetics, 186:757-761, 2010; U.S. Publication No. 2011/0145940; Cong et al., Science 339:819-823, 2013; and Mali et al., Science 339:823-826, 2013, for a discussion of each. In particular, CRISPR/Cas molecules are components of a prokaryotic adaptive immune system that is functionally analogous to eukaryotic RNA interference, using RNA base pairing to direct DNA or RNA cleavage. Directing DNA DSBs requires two components: the Cas9 protein, which functions as an endonuclease, and CRISPR RNA (crRNA) and tracer RNA (tracrRNA) sequences that aid in directing the Cas9/RNA complex to target DNA sequence (Makarova et al., Nat Rev Microbiol, 9(6):467-477, 2011). The modification of a single targeting RNA can be sufficient to alter the nucleotide target of a Cas protein. In some cases, crRNA and tracrRNA can be engineered as a single cr/tracrRNA hybrid to direct Cas9 cleavage activity (Jinek et al., Science, 337(6096):816-821, 2012). Like TALE nucleases, for example, the components of a CRISPR/Cas system (the Cas9 endonuclease and the crRNA and tracrRNA, or the cr/tracrRNA hybrid) can be delivered to a cell in a geminivirus construct.

In some embodiments of the systems and methods provided herein, the sequence encoding the endonuclease can be stably integrated into the plant genome that will be infected with a geminivirus containing a repair template. See, for example, FIG. 2, which depicts a plant genome into which a sequence encoding an ADH1 targeted ZFN has been stably integrated. The coding sequence can be operably linked to a promoter that is inducible, constitutive, cell specific, or activated by alternative splicing of a suicide exon. For example, as shown in FIG. 2, the ADH1 ZFN coding sequence is operably linked to an XVE promoter, which can be activated by estradiol. The plant can be infected with a geminivirus containing a repair template (indicated by the black bar flanked by white bars in the “CaLCuV”), and expression of the ZFN can be activated by treating the plant with estradiol. The ZFN protein then can cleave the DNA at the target sequence, facilitating HR on either side of the repair template to be integrated.

Alternatively, the endonuclease coding sequence can be contained in the same geminivirus construct as the repair template, or can be present in a second plasmid that is separately delivered to the plant, either sequentially or simultaneously with the geminivirus construct. For example, in some embodiments, plants can be transfected or infected with a second viral vector, such as an RNA virus vector (e.g., a tobacco rattle virus (TRV) vector, a potato virus X vector, a pea early browning virus vector, or a barley stripe mosaic virus vector) that encodes the endonuclease. As an example, TRV is a bipartite RNA plant virus that can be used to transiently deliver protein coding sequences to plant cells. For example, the TRV genome can be modified to encode a ZFN or TALE nuclease by replacing TRV nucleotide sequence with a subgenomic promoter and the ORF for the endonuclease. The inclusion of a TRV vector can be useful because TRV infects dividing cells and therefore can modify germ line cells specifically. In such cases, expression of the endonuclease encoded by the TRV can occur in germ line cells, such that HR at the target site is heritable.

In embodiments in which a geminivirus vector contains both a repair template and an endonuclease encoding sequence, it is noted that that the geminivirus can be deconstructed such that it encodes only the proteins needed for viral replication. Since a deconstructed geminivirus vector has a much larger capacity for carrying sequences that are heterologous to the virus, it is noted that the repair template may be longer than 800 nt. An exemplary system using a deconstructed vector is described in the Example below.

The construct(s) containing the repair template and, in some cases, the endonuclease encoding sequence, can be delivered to a plant cell using, for example, biolistic bombardment. Alternatively, the repair template and endonuclease sequences can be delivered using Agrobacterium-mediated transformation, insect vectors, grafting, or DNA abrasion, according to methods that are standard in the art, including those described herein.

After a plant is infected or transfected with a repair template (and, in some cases, an endonuclease encoding sequence), any suitable method can be used to determine whether GT has occurred at the target site. In some embodiments, a phenotypic change can indicate that a repair template sequence has been integrated into the target site. Such is the case for the gus::nptII plants that were repaired with a geminivirus containing a GUS sequence, as described below. PCR-based methods also can be used to ascertain whether a genomic target site contains a repair template sequence, and/or whether precise recombination has occurred at the 5′ and 3′ ends of the repair template. A schematic depicting an example of such a technique is provided in FIG. 3, and the work described below also demonstrates GT in Arabidopsis using PCR-based techniques. In some of these experiments, plants expressing a ZFN were infected with geminiviruses producing repair templates (also referred to herein as donor molecules), and recombination between the repair template and the target gene on the plant chromosome was observed in somatic cell genomic DNA from infected plants expressing an active endonuclease. In particular, following systemic infection of an engineered geminivirus containing a unique 18 bp modification flanked by 400 bases of homology to the ADH1 target locus, ZFN expression was induced. Following ZFN expression, genomic DNA from somatic cells was extracted and assessed for GT events. Results from the enrichment PCR suggested successful GT of the ADH1 loci using geminiviruses and ZFNs. Additional experiments are described that involve quantifying the frequency of gene targeting in somatic cells, and demonstrating gene targeting by phenotypic analysis.

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

Examples Cloning of Genetic Elements into pCPCbLCVA.007

The cabbage leaf curl virus (CaLCuV) is a bipartite, circular single-stranded DNA virus that can infect Arabidopsis plants when delivered by microprojectile bombardment. Initiating viral infection requires the delivery of two plasmids containing sequence for both genomes (A and B components; FIG. 1). The viral sequences are partially duplicated, containing two direct repeats of the origin of replication flanking the viral genome. Consequently, delivery of these plasmids to plant cell nuclei results in replicational release of full-length, circular geminivirus genomes.

To construct CaLCuV A components encoding repair template sequence, the coat protein (AR-1) coding sequence was replaced with desired sequence. AR-1 is required for insect-transmission of the virus, but it is not required for viral amplification and systemic spreading. Because of this, approximately 800 nucleotides can be added to the A component genome without preventing its ability to infect. Viral vectors encoding repair templates targeting the ADH1 and gus::nptII loci use the pCPCbLCVA.007 backbone. pCPCbLCVA.007 is a plasmid initially designed for viral induced gene silencing (VIGS). It encodes a partially duplicated A component with the AR-1 nucleotide sequence replaced with a multicloning site (MCS). Co-bombardment of Arabidopsis plants with pCPCbLCVA.007 (FIG. 4A) and pCPCbLCVB.002 (FIG. 4B) (encoding a partially duplicated B genome) results in a high-frequency of successful infection (75-100%).

Constructing First Generation Viral Vectors with Repair Templates Targeting ADH1

An ADH1-targeting repair template was constructed for ligation into pCPCbLCVA.007. The template for amplifying the ADH1 repair template was genomic DNA from Arabidopsis thaliana (ecotype Columbia). To isolate genomic DNA, about 100 mg of leaf tissue was frozen in liquid nitrogen and ground to a fine powder. 500 μl of CTAB buffer (2.0 g hexadecyl trimethyl-ammonium bromide (CTAB)), 10 mL 1M tris pH 8.0, 4 mL 0.5M ethylenediaminetetraacetic acid di-sodium salt (EDTA), 28 mL 5M NaCl, 40 mL dH2O, pH adjusted to 5.0 per 100 mL of solution) was added and the samples were incubated at 65° C. for 20 min. Samples were centrifuged for 5 minutes at 12,000 RPM and the supernatant was transferred to a clean microfuge tube. 500 μA of chloroform was added and the samples were inverted for 5 minutes at room temperature. Samples were centrifuged for 1 minute at 12,000 RPM and the supernatant was transferred to a clean microfuge tube. 800 μl of ice-cold 100% ethanol was added and the samples were centrifuged for 1 minute at 15,000 RPM. The supernatant was decanted and the genomic DNA pellet was washed once in 75% ethanol. Samples were centrifuged for 30 seconds at 13,000 RPM and the supernatant was completely removed. Last, the genomic DNA was resuspended in 50 μl of dH2O. Repair templates targeting ADH1 were designed to encode a unique 18 bp modification sequence (5′-GAGCTCAGTACTGC ATGC-3′; SEQ ID NO:1) flanked by arms of homology to the ADH1-ZFN target site. Several repair templates were constructed with varying lengths of homology for each arm. In total, four repair templates were made with 491, 391, 291, or 191 nucleotides of homology in each arm. Notably, the modification was designed to remove the native ZFN binding site, which prevents cleavage of the repair template before and after GT. To generate ADH1 repair templates for cloning into pCPCbLCVA.007, left and right homology sequences were amplified from Arabidopsis genomic DNA using primers NB177+NB128 and NB178+NB129 for 491 bp homology arms, NB104+NB128 and NB112+NB129 for 391 bp homology arms, NB105+NB207 and NB113+NB208 for 291 bp homology arms, and NB106+NB207 and NB114+NB208 for 191 bp homology arms, respectively. Primer sequences are provided in Table 1. Importantly, the reverse primers for the left homology arm and the forward primers for the right homology arm contained complementary 18 bp linkers encoding the modification sequence. Also, the forward primers for the left homology arm and the reverse primer for the right homology arm contained linkers encoding XbaI and BglII restriction enzyme sites, respectively. PCR reactions were performed in a 25 μl PCR mix composed of 2.5 μl of 10×NEB Standard Taq buffer, 0.5 μl of 10 mM dNTPs, 0.5 μl of 10 μM primer 1, 0.5 μl of 10 μM primer 2, 18.8 μl of dH2O, 0.2 μl of Taq polymerase, and 2 μl of genomic DNA (˜200 ng). The PCR conditions were 5 minutes at 94° C. followed by 30 cycles of 30 seconds at 94° C., 30 seconds at 55° C., and 1 minute at 72° C. The resulting amplicons were resolved by agarose electrophoresis using a 1% gel. DNA bands of expected sizes were excised from the agarose gel and purified using the QIAquick Gel Extraction Kit (Qiagen) following manufacturer's protocols. Purified DNA fragments containing the left and right homology arms were then fused together in an overlap-extension PCR (OE-PCR). Fusion reactions were performed in a 24 μl PCR mix composed of 2.5 μl of 10× cloned Pfu buffer, 0.5 μl of 10 mM dNTPs, 14.5 μl of dH2O, 0.5 μl of Pfu enzyme, and 3 μl each of the purified amplicons. Fusion conditions were 5 minutes at 94° C. followed by 10 cycles of 30 seconds at 94° C., 30 seconds at 50° C., and 1 minute at 72° C. Following the fusion PCR, 0.5 μl of 10 μM primer 1 and 0.5 μl of 10 μM primer 2 were added and the samples were run in another PCR. The PCR conditions were 5 minutes at 94° C. followed by 30 cycles of 30 seconds at 94° C., 30 seconds at 56° C., and 1 minute at 72° C. Following OE-PCR, 10 μl of the PCR solution and 1 μg of pCPCbLCVA.007 were digested with XbaI and BglII following standard procedures. The resulting digested amplicons and vector were resolved by agarose electrophoresis using a 1% gel. DNA bands of expected sizes were excised from the agarose gel and purified and ligated together in a 10 μl reaction using T4 DNA ligase (New England Biolabs) following the manufacturer's procedures. DH5α E. coli were transformed with 2 μl of the ligation mix following standard procedures and plated onto LB media containing 50 μg/ml of carbenicillin. DNA sequences of resulting clones were confirmed by sequencing to encode the expected repair template. These vectors are henceforth referred to as CaLCuVA.ADH1-1000, CaLCuVA.ADH1-800, CaLCuVA.ADH1-600, and CaLCuVA.ADH1-400.

Constructing First Generation Viral Vectors with Repair Templates Targeting Gus::nptII

The following describes methods for constructing GUS-FIX repair templates for ligation into pCPCbLCVA.007. The chromosomal target for the repair template is a GUS transgene with ˜300 bp of nucleotide sequence removed from the 3′ end and replaced with a Zif268 target site. GUS-FIX repair templates were designed to contain flanking arms of homology to the target locus (200 bp each) and a 300 bp modification sequence. As a consequence of GT, the coding sequence of GUS is restored. Cells actively expressing GUS can be phenotypically detected by an enzymatic assay. To generate GUS-FIX repair templates for cloning into pCPCbLCVA.007, the left homology arm (also containing the 300 bp of GUS-FIX sequence) and the right homology arm were amplified from pDW1269 plasmid DNA using primers NB274+NB271 and NB272+NB275, respectively. Importantly, the left and right homology arms contained complementary sequences to enable their fusion in OE-PCR. PCR reactions to generate the fragments were performed in a 25 μl mix composed of 2.5 μl of 10×NEB Standard Taq buffer, 0.5 μl of 10 mM dNTPs, 0.5 μl of 10 μM primer 1, 0.5 μl of 10 μM primer 2, 18.8 μl of dH2O, 0.2 μl of Taq polymerase, and 2 μl of genomic DNA (˜200 ng). The PCR conditions were 5 minutes at 94° C. followed by 30 cycles of 30 seconds at 94° C., 30 seconds at 55° C., and 1 minute at 72° C. The resulting amplicons were resolved by agarose electrophoresis using a 1% gel. DNA bands of expected sizes were purified and ligated together in an OE-PCR. Fusion reactions were performed in a 24 μl mix composed of 2.5 μl of 10× cloned Pfu buffer, 0.5 μl of 10 mM dNTPs, 14.5 μl of dH2O, 0.5 μl of Pfu enzyme, and 3 μl each of the purified amplicons. Fusion conditions were 5 minutes at 94° C. followed by 10 cycles of 30 seconds at 94° C., 30 seconds at 50° C., and 1 minute at 72° C. Next, 0.5 μl of 10 μM primer NB274 and 0.5 μl of 10 μM primer NB275 were directly added to the fusion reactions and immediately run in another PCR. The PCR conditions were 5 minutes at 94° C. followed by 30 cycles of 30 seconds at 94° C., 30 seconds at 56° C., and 1 minute at 72° C. Following OE-PCR, 10 μl of solution and 1 μg of pCPCbLCVA.007 were digested with XbaI and BglII following standard procedures. The resulting digested amplicons and vector were resolved by agarose electrophoresis using a 1% gel. DNA bands of expected sizes were purified and ligated in a 10 μl reaction using T4 DNA ligase. DH5α E. coli were transformed with 2 μl of the ligation mix following standard procedures, and plated onto LB media containing 50 μg/ml of carbenicillin. The DNA sequence of a resulting clone was confirmed to encode the GUS-FIX repair template sequence. This vector is referred to as CaLCuVA.GUS-FIX.

Growing Arabidopsis Plants

To prepare Arabidopsis plants for biolistic bombardment, 500-1,000 Arabidopsis seeds (10-20 mg) were stratified in 0.1% agarose for 3 days at 4° C. Seeds were dispensed onto the surface of BM2 soil (J.R. Johnson Supply; Minneapolis, Minn.) in each of the four corners of 2.5×2.5 inch pots. Pots were placed in a plastic flat and 1 L of 10-20-10 Peters Professional (Scotts) fertilizer solution was added. Flats were covered with a clear plastic dome and moved to a growth chamber under 12 h light/12 h dark conditions. Plants were grown at 22-24° C. for 2 weeks before removing the dome, and then grown for an additional 1-2 weeks with watering when needed. Watering was stopped approximately 7 days before bombardment. Plants were bombarded when they reached the five- to six-leaf-stage (approximately four weeks).

Infecting Arabidopsis Plants by Biolistic Bombardment

Biolistic bombardment was carried out closely following the protocol described by Muangsan et al., Meth. Mol. Biol., 265:101-115, 2004. Briefly, to prepare microprojectile particles for five bombardments, 5 μg of each plasmid (CaLCuVA and CaLCuVB) was added to a tube containing 50 μl of 60 mg/mL gold beads and briefly vortexed. 50 μl of 2.5 M CaCl2 was directly added to the samples and immediately pipetted in and out of a tip to break up conglomerates. 20 μl of 0.1 M spermidine was added and the samples were immediately vortexed for 5 min. The samples were centrifuged at 10,000 RPM for 10 seconds and the supernatant was removed. The gold-bead pellet was resuspended in 250 μl of 100% ethanol and then centrifuged at 10,000 rpm for 10 sec. Supernatants were removed and the samples were resuspended in 65 μl of 100% ethanol. The particles were then stored on ice until bombardment. To prepare the assembly for the microprojectile particles, macrocarrier holders and macrocarriers were soaked in 95% ethanol, air-dried, and assembled. 10 μl of resuspended particles were then spotted onto the center of the macrocarrier and allowed to air-dry.

Biolistic bombardment was carried out in a horizontal laminar flow hood using a PDS-1000 He system (Bio-Rad). To prepare the PDS-1000 He system, a non-sterile rupture disk (1100 psi) was dipped in 100% isopropanol and placed into the upper assembly. The macrocarrier launch assembly (MCLA) was then prepared by dipping a metal stopping screen in 95% ethanol, and then placing the dried screen onto the opening of the lower assembly. The macrocarrier and macrocarrier holder were inverted and placed above the stopping screen. The retaining ring was screwed in, and the MCLA was placed into the top rack of the chamber. A single pot containing four plants was then placed in the chamber directly beneath the MCLA. A vacuum of 28 in was created, and helium was added to the upper chamber until the rupture disk burst. Bombarded plants were then removed from the chamber and returned to a covered flat. Between bombardments of different constructs, the chamber was cleaned with 70% ethanol. This procedure was repeated for additional infections. By following these methods, infection was successfully initiated in majority of the bombarded plants (75-100%).

Growing Infected Arabidopsis Plants

Immediately after bombardment, infected Arabidopsis plants were placed in a flat with approximately 1 L of fertilizer solution and moved back to the growth chamber. A clear plastic dome was used to cover the plants for seven days post infection. Infection was noticeable 8-10 dpi by curling of rosette leaves. At 14 dpi, plants containing an XVE ADH1-ZFN transgene were induced by exposure to β-estradiol (Sigma E2758) by spraying and watering. The spray contained 0.01% Silwet L-77 (Vac-In-Stuff) and 20 μM β-estradiol, while the water contained only 20 μM β-estradiol. Induction was carried out by continuously spraying (approximately once a day) and watering (approximately twice a week) for 10-14 days.

Isolating Genomic DNA from Infected Arabidopsis Plants

About two weeks after induction, genomic DNA was extracted from somatic plant tissue. A single rosette leaf and cauline leaf were collected from each infected plant. Care was taken when choosing leaves in order to minimize the likelihood of detecting recombination between plasmid molecules and genomic DNA. Criteria for choosing rosette leaves were 1) healthy leaf tissue with no obvious necrotic lesions, and 2) leaves growing on the periphery of the pot—away from damage caused by biolistic bombardment. Plant genomic DNA was extracted following the CTAB procedure as described above.

Assessing β-Estradiol Induction of the ADH1-ZFN Transgene

To determine if induction of nuclease expression by β-estradiol was successful, enrichment PCR was performed on purified genomic DNA. Enrichment PCR is designed to detect ZFN-induced NHEJ mutations at the ADH1 target locus—an indirect assay for verifying nuclease activity. This procedure relies on a restriction enzyme site positioned in or near the target site spacer sequence. In essence, if the nuclease is not active, then target site amplicons will be completely digested by the restriction enzyme. On the other hand, if the nuclease is active there will be a population of target site amplicons with destroyed restriction enzymes sites that will not be digested by the restriction enzyme. Thus, detection of a digestion-resistant band suggests that the nuclease was actively creating DSBs.

For these assays, 1 μg of genomic DNA from induced and non-induced plants was digested with BstXI (NEB) in a 10 μl solution following standard procedures. Immediately following digestion, 2 μl of the solution was used as a template for PCR in a reaction containing of 2.5 μl of 10×NEB Standard Taq buffer, 0.5 μl of 10 mM dNTPs, 0.5 μl of 10 μM primer NB161, 0.5 μl of 10 μM primer NB154, 18.8 μl of dH2O, 0.2 μl of Taq polymerase, and 2 μl of the digested solution (˜200 ng genomic DNA). The PCR conditions were 5 minutes at 94° C. followed by 35 cycles of 30 seconds at 94° C., 30 seconds at 55° C., and 1 minute at 72° C. 10 μl of the PCR reaction was then digested with BstXI. The entire digested sample and the corresponding PCR sample were loaded side-by-side onto a 1.2% agarose gel. In general, plants that were not exposed to estradiol had very faint, or undetectable, digestion-resistant amplicons (FIG. 5, bottom row, digested (“D”) lanes). Conversely, plants exposed to β-estradiol had much stronger resistant bands (FIG. 5, top row, digested (“D”) lanes). From these data, it was concluded that the timing of ADH1-targeted DSBs was controlled by β-estradiol.

Assessing Repair Template Stability in Infected Plants

To ensure that the repair template was stably replicated in infected plants, PCR was performed on purified genomic DNA. Notably, DNA isolated from infected plants is a mixture of plant genomic DNA and virus genomic DNA. Primers were designed to recognize viral sequence (non-repair template sequence) in the CaLCuV A plasmid (FIG. 6, left panel), and to amplify across the entire repair template sequence. PCR reactions contained 2.5 μl of 10×NEB Standard Taq buffer, 0.5 μl of 10 mM dNTPs, 0.5 μl of 10 μM primer NB153, 0.5 μl of 10 μM primer NB158, 18.8 μl of dH2O, 0.2 μl of Taq polymerase, and 2 μl of purified genomic DNA (˜200 ng). The PCR conditions were 5 minutes at 94° C. followed by 35 cycles of 30 seconds at 94° C., 30 seconds at 55° C., and 1 minute at 72° C. 10 μl of the PCR sample was loaded onto a 1.0% agarose gel. FIG. 6 (right panel) shows the resulting amplicons from infected plants carrying repair templates ranging from 400 nt to 1000 nt. These results suggested that repair templates equal to or less than 715 bp were stably replicated in plant cells. For this reason, only viruses carrying repair templates equal or less than 715 bp were assessed in the subsequent experiments. Based on these experiments, it was concluded that first generation geminiviral vectors effectively amplified and disseminated repair templates in Arabidopsis plants.

Detecting GT at the ADH1 Locus

Nested PCR was performed to detect modified ADH1 loci. Primers were designed to amplify the ADH1 locus approximately 700 bp upstream and downstream of the ZFN target sequence. The resulting amplicons were then used as a template for a nested PCR, with primers that specifically recognize the unique 18 bp modification sequence and ADH1 sequence outside the homology arms carried by the virus. In detail, the ADH1 locus was amplified in a PCR reaction containing 2.5 μl of 10×NEB Standard Taq buffer, 0.5 μl of 10 mM dNTPs, 0.5 μl of 10 μM primer NB257, 0.5 μl of 10 μM primer NB258, 18.8 μl of dH2O, 0.2 μl of Taq polymerase, and 2 μl of purified genomic DNA (˜200 ng). The PCR conditions were 5 minutes at 94° C. followed by 15 cycles of 30 seconds at 94° C., 30 seconds at 55° C., and 1 minute at 72° C. Amplicons were column purified using the QIAquick Gel Extraction Kit. Purified amplicons were then used as templates for three nested PCRs. The first PCR checked for the 5′ modification junction using primers NB154 and NB264. The second PCR checked for the 3′ modification junction using primers NB263 and NB155. The third PCR was a control for template amplification and used primers NB155 and NB154. To minimize template switching, PCR was performed using Expand Long Template PCR system (Roche) in a reaction containing 2.5 μl buffer 1, 0.5 μl 10 mM dNTPs, 0.5 μl of 10 μM primer 1, 0.5 μl of 10 μM primer 2, 0.2 μl of the Taq/Tgo polymerase mix, 17.8 μl dH2O, and 3 μl of purified amplicons. The PCR conditions were 5 minutes at 94° C. followed by 30 cycles of 30 seconds at 94° C., 30 seconds at 55° C., and 1 minute at 72° C. Amplicons were run on a 1% agarose gel. In select plants (KU70 −/−, ADH1-ZFN +1+ background) that were infected with virus and exposed to β-estradiol, a noticeable amplicon band was present in both the 5′ and 3′ junction PCRs (FIG. 7). Importantly, plants (Columbia background) that were only infected with the virus did not have detectable amplicons for the 5′ and 3′ junction PCR. From these results it was concluded that geminiviruses and ZFNs can stimulate GT at an endogenous locus in somatic leaf tissue.

Delivery of Zif268-ZFN for GT at the Gus::nptII Locus

GT was stimulated at the gus::nptII transgene. To detect GT by phenotype, plants containing a stably integrated gus::nptII transgene were infected with CaLCuVA.GUS-FIX and CaLCuVB following the procedures described above. Notably, immediately following the truncated GUS nucleotide sequence was a target site for Zif268. For these experiments, Zif268::FokI was transiently delivered to plants 8 dpi by TRV. TRV is a bipartite RNA plant virus that can be used to transiently deliver protein coding sequences to plant cells. In the present experiments, TRV was modified to express Zif268::FokI by replacing the 2b and 2c nucleotide sequences with a subgenomic promoter and the ORF for the Zif268::FokI. Infection was carried out by syringe infiltration of Agrobacterium carrying T-DNA coding for both TRV genomes. Briefly, GV3101 Agrobacterium carrying T-DNA encoding for TRV1 and TRV2-Zif268 were grown overnight at 28° C. in 3 mL of LB medium containing 50 μg/mL kanamycin and 50 μg/mL gentamycin. One mL of the culture was transferred to 100 mL LB medium containing 50 μg/mL kanamycin and 50 μg/mL gentamycin and grown overnight at 28° C. until they reached an OD of approximately 1.0. Solutions were then centrifuged at 7000 RPM for 10 minutes and resuspended in 50 mL of MMAi solution (0.5 g MS salts, 0.195 g MES, 2 g sucrose, 100 μl of 200 mM acetosyringone per 100 mL at pH 5.6) followed by shaking at 50 rpm for 2 hours. Solutions of Agrobacterium containing TRV1 and TRV2-Zif268 were mixed in a 1:1 ratio and syringe infiltrated into three rosette leaves per plant. TRV and geminivirus infected plants were moved to a growth chamber under 12 h light/12 h dark conditions at 22-24° C. for 15 days.

Detecting GT at the Gus::nptII Locus

To detect evidence for GT at the gus::nptII locus, plants were analyzed for cells expressing functional GUS protein. Fifteen days after TRV infection and 23 days after geminivirus infection, plants were stained overnight at 37° C. in an X-Gluc solution (0.052 g X-Gluc (GoldBio), 5 mL 1M sodium phosphate, 0.1 mL Triton X per 100 mL). Plants were removed from the stain and incubated in 75% ethanol for 2-3 days to remove chlorophyll (which helped with visualizing the blue staining) Plants were visualized using a stereoscope. If GT occurred, spots of blue were observed where one or multiple cells had reconstituted GUS expression. Such blue spots also were observed in tissue that developed after biolistic bombardment. FIG. 8 shows images of plants co-infected with CaLCuVA.GUS-FIX and CaLCuVB (or with either plasmid alone) that were stained in X-gluc. The spotty patches of blue staining in the rosette leaves and in the newly developed tissue suggested that GT had occurred. These results indicated that geminiviruses and ZFNs can stimulate GT at a gus::nptII transgene in plant somatic tissue.

Approach for Generating Bean Yellow Dwarf Virus Replicon Vectors

An exemplary method for generating bean yellow dwarf virus (BeYDV) replicons in plant cells involves delivery of one or two plasmids or T-DNA molecules that encode the trans-acting replication-associated proteins, Rep/RepA, and direct duplications of the large intergenic region (LIR) flanking sequence encoding the small intergenic region (SIR; FIGS. 9-10). Normally, virus replication is initiated by Rep protein binding to LIR sequence on a circular dsDNA genome. However, if the geminivirus genome is linearized and contains flanking LIR sequences (also referred to as an LSL vector), Rep proteins bind to the LIR sequences and release circularized, single-stranded geminiviral replicons (GVRs). Replicons can then be used as a template for replicase-mediated genome amplification. Consequently, any sequence present inside the flanking LIRs will be present in the replicon. Eliminating coat protein and movement protein sequence abolishes cell-cell movement, but significantly lessens genome-size restraints imposed by plasmodesmata. To compensate for loss of cell-cell movement, Agrobacterium was used to direct GVR production in specific cells. To facilitate cloning of endonuclease and repair template sequence into an LSL destination vector, MultiSite Gateway cloning technology (Invitrogen) was implemented.

Constructing an LSL Destination T-DNA Plasmid

The following describes methods for constructing a BeYDV-derived LSL destination T-DNA plasmid (pLSL; FIGS. 9-10). Assembly of the complete LSL nucleotide sequence was accomplished by cloning smaller “blocks” of LSL sequence into pBluescript KS+ plasmids before cloning into a pCAMBIA1300 T-DNA backbone. The first block was designed to contain LIR::DEM2 splice acceptor (last 62 nt of the DEM2 intron)::tobacco etch virus (TEV) 5′ UTR (last 93 nt of the TEV 5′ UTR)::attR1::chloramphenicol resistance gene (CmR). The second block contained ccdB::attR2::SIR. The third block contained 2×35S::TEV 5′ UTR (first 38 nt of the TEV 5′ UTR)::DEM2 splice donor (first 32 nt of the DEM2 intron)::LIR. LIR and SIR sequences were obtained from the mild BeYDV isolate (GenBank accession number DQ458791.1). To generate attR1::CmR sequence for block 1, pFZ19 was used as a template for PCR amplification using primers NB326 and NB327. PCR solutions contained 2.5 μl of 10× cloned Pfu buffer, 0.5 μl of 10 mM dNTPs, 0.5 μl of 10 μM primer NB326, 0.5 μl of 10 μM primer NB327, 18.5 μl of dH2O, 0.5 μl of Pfu enzyme, and 2 μl of plasmid DNA (˜20 ng). PCR cycling included 5 minutes at 94° C., followed by 30 cycles of 30 seconds at 94° C., 30 seconds at 55° C., and 2 minutes at 72° C. PCR amplicons were column purified using the QIAquick gel extraction kit. Purified amplicons were then used in an OE-PCR with NB330 and NB331 to generate the complete nucleotide sequence for block 1. OE-PCR solutions contained 2.5 μl of 10× cloned Pfu buffer, 0.5 μl of 10 mM dNTPs, 0.5 μl of 10 μM primer NB327, 0.5 μl of 10 μM primer NB325, 14.5 μl of dH2O, 0.5 μl of Pfu enzyme, and 2 μl of purified amplicons, NB330 (2 ng) and NB331 (2 ng). PCR cycling consisted of 5 minutes at 94° C., followed by 30 cycles of 30 seconds at 94° C., 30 seconds at 55° C., and 4 minutes at 72° C. Amplicons and 1 μg of pBluescript KS+ vector were digested with Kpnl and XbaI. Digested fragments were purified and ligated following standard procedures. The resulting ligation was transformed into DH5α E. coli cells following standard procedures. Herein, the sequence verified plasmid containing block 1 is termed pBlock1. Blocks 2 and 3 were constructed using similar methods. Construction of Block 2 first required amplification and purification of ccdB::attR2 from pFZ19 using primers NB328 and NB332. Purified amplicons were added to an OE-PCR with NB344 and primers NB328+NB329 to generate the complete nucleotide sequence for block 2. Purified amplicons were ligated into pBluescript KS+ with XbaI and SacI and transformed into ccdB-resistant XL-1 Blue cells to generate pBlock2. Construction of block 3 first required PCR amplification of 2×35S sequence from pMDC32 using primers NB333+NB334. To generate the complete nucleotide sequence for block 3, purified amplicons were used in an OE-PCR with NB335 and NB336 using primers NB333 and NB337. Purified amplicons were ligated into pBluescript KS+ with XhoI and SacI, and transformed into DH5α cells to generate pBlock3. Nucleotide sequences for the two LIR elements in pBlock1 and pBlock3 were designed to contain inverted homodimeric BsaI to facilitate cloning of the conserved hairpin structure. To complete the hairpin structure, pBlock1 and pBlock3 were digested with BsaI and gel purified. Primers NB338 and NB339 were dephosphorylated, annealed, ligated into pBlock1 and pBlock3 vector backbones, and transformed into DH5α to generate pBlock1 HP and pBlock3HP. To construct the final LSL vector, pBlock1HP, pBlock2, pBlock3HP, pCAMBIA1300 were digested with SbfI+XbaI, XbaI+XhoI, XhoI+SbfI, and SbfI, respectively. Fragments of the expected sizes were gel purified, ligated, and transformed into ccdB-resistant XL-1 Blue cells following standard protocols for 4-way ligations. The resulting plasmid (pLSL, FIG. 10C) was sequence verified and used as a destination vector for MultiSite Gateway cloning.

Constructing a Nuclease-Entry Plasmid

A nuclease-entry vector was constructed for MultiSite Gateway cloning into pLSL (pNJB091; FIG. 10B). Four unique restriction enzyme sites immediately upstream of two FokI coding sequences allows for sequential cloning of custom-designed DNA binding domains. To construct pNJB091, pZHY013 (a modified pCR8 entry vector encoding FokI heterodimer sequences; FIG. 10A) and NB318 were digested with BsmI and EcoRV. Digested fragments were gel purified, ligated and transformed into DH5α cells following standard protocols.

Constructing a Donor-Entry Plasmid

A donor-entry vector was constructed for MultiSite Gateway cloning into pLSL (pNJB080; FIG. 10B). Two unique pairs of restriction enzyme sites flanking ccdB and CmR selection markers permit efficient cloning of repair templates. To construct pNJB80, sequence encoding the CmR and ccdB genes was amplified by PCR from pFZ19 using NB316+NB317 primers. Amplicons were purified and used in an OE-PCR with NB314 and primers NB315 and NB317. PCR solutions contained 2.5 μl of 10× cloned Pfu buffer, 0.5 μl of 10 mM dNTPs, 0.5 μl of 10 μM NB315, 0.5 μl of 10 μM NB317, 16.5 μl of dH2O, 0.5 μl of Pfu enzyme, 2 μl of purified amplicons, and 2 μl of 10 μM NB314. PCR cycling included 5 minutes at 94° C., followed by 30 cycles of 30 seconds at 94° C., 30 seconds at 55° C., and 3 minutes at 72° C. Resulting amplicons were gel purified following standard procedures. Amplicons and pZHY558 were digested with ApaI and BsrGI, ligated, and transformed into ccdB-resistant cells following standard procedures. MultiSite Gateway recombination with pNEL1R5 into pLSL positions repair template sequence between two transcriptional-termination sequences (upstream Nos-T sequence and downstream SIR sequence). The studies herein may benefit from flanking termination sequences. For example, transcriptional gene silencing is facilitated through production of RNA molecules with homology to an endogenous gene. Reducing read-through transcription of repair template sequence may decrease unintentional silencing of targeted genes.

Constructing Replicase-Expressing T-DNA Plasmids

To initiate replicational release of GVRs from LSL T-DNA, trans-acting Rep/RepA proteins must be expressed. Here, two Rep/RepA T-DNA expression plasmids were constructed. The first plasmid encodes the Rep/RepA coding sequence downstream of an estradiol-inducible XVE promoter (pXVEREP), such that when integrated into the plant genome, Rep/RepA expression can be induced by exposing plant tissue to β-estradiol. The second plasmid encodes Rep/RepA downstream of a 2×35S promoter (p35SREP). For each plasmid, WT RepA and mutant RepA (RepA LxCxQ; Liu et al., Virology 256:270-279, 1999) versions are created (pXVEREPLxCxQ and p35SREPLxCxQ). Normally, RepA interacts with the host cell's retinoblastoma (RB) protein, sequestering its repressive activity on E2F. This promotes entry into S phase, and, in turn, provides the invading geminivirus with replication machinery needed to amplify its genome. The studies described herein may benefit from a RepA protein that does not interact with RB. For example, in actively dividing meristem cells or germline cells, factors required for replicon amplification should already be present. Thus, there may be little need to inactivate RB in these cell types. Furthermore, expression of RepA LxCxQ may result in decreased toxicity in these cell types—which may facilitate recovery of modified seeds.

To generate pXVEREP, p35SREP, pXVEREPLxCxQ, and p35SREPLxCxQ (FIG. 11), WT and mutant Rep/RepA coding sequences were amplified by OE-PCR using NB319, NB320, and NB322, and primers NB323 and NB324 (WT Rep/RepA), or using NB319, NB321, and NB322, and primers NB323 and NB324 (mutant Rep/RepA). PCR solutions consisted of 2.5 μl of 10× cloned Pfu buffer, 0.5 μl of 10 mM dNTPs, 0.5 μl of 10 μM NB323, 0.5 μl of 10 μM NB324, 14.5 μl of dH2O, 0.5 μl of Pfu enzyme, and 2 μl of each DNA component. PCR cycling included 5 minutes at 94° C., followed by 30 cycles of 30 seconds at 94° C., 30 seconds at 55° C., and 3 minutes at 72° C. Resulting amplicons were purified using the QIAquick gel extraction kit. One μl of purified amplicons was combined with 150 ng of pFZ19 or pMDC32 (2×35S Ti-DNA vector) and recombination was stimulated using LR clonase (Invitrogen) as described by the manufacturer's protocol. Plasmid from the resulting solution was transformed into DH5α, and cells were plated on LB plates containing 50 μg/mL kanamycin.

Demonstrating Transient Delivery of Reporter Proteins in Nicotiana tabacum Leaf Tissue Using GVRs

Functionality of the system was tested by attempting to transiently express reporter proteins in somatic leaf tissue. To this end, pLSL was modified to encode NLS-tagged green fluorescent protein (pLSLGFP) or beta-glucuronidase (pLSLGUS). GFP and GUS nucleotide sequence were amplified from, respectively, pTC23 and pNB67 using primers NB362 and NB363, and primers NB448 and NB449. Forward and reverse primers contained XbaI and AatII restriction enzyme sites, respectively for cloning into pNB091. The resulting vectors were used in a MultiSite Gateway recombination reaction with pLSL and pNB098 (a modified version of pNB080 with a repair template to correct a non-functional gus::nptII transgene) to generate pLSLGFP and pLSLGUS. These vectors were sequence verified and transformed into Agrobacterium tumefaciens GV3101 by the freeze-thaw method. Single colonies of transformed Agrobacterium were grown overnight in a shaker at 28° C. in 5 mL of LB starter culture with 50 μg/ml kanamycin and 50 μg/ml gentamicin. The next day, 1 ml was used to inoculate 50 mL of LB culture with 50 μg/ml kanamycin and 50 μg/ml gentamicin. After reaching an OD600 of 1 (approximately 16 hours), cells were pelleted, and resuspended to an OD600 of 0.2 in infiltration buffer (10 mM 2-(N-morpholino) ethanesulfonic acid (MES), and 10 mM MgSO4, pH 5.6). Resuspended cultures were incubated at room temperature for 2 hours before infiltration. To demonstrate transient expression of GUS, half leaves were fully infiltrated with Agrobacterium containing pLSLGUS or a 1:1 mixture of Agrobacterium containing pLSLGUS and p35SREP. Seven dpi infected leaf tissue was excised from the plant and stained in X-Gluc for 24 hours at 37° C. Chlorophyll was removed using 80% ethanol, and leaf images were taken (FIG. 12). To demonstrate transient expression of GFP, three leaves were syringe infiltrated with Agrobacterium containing pLSLGFP, or infiltrated with a 1:1 mixture of Agrobacterium containing pLSLGFP and p35SREP or pLSLGUS and p35SREP. Images capturing GFP fluorescence were taken 3, 7, and 12 dpi (FIG. 13). Both GUS and GFP expression were markedly enhanced when p35SREP was co-delivered. Notably, a slight browning of leaf tissue was observed 7 dpi due to replicase expression (FIG. 14). To correlate enhanced protein expression with replicon production, Rep-assisted replicational release was evaluated by PCR (FIG. 15, top). To this end, DNA was extracted from leaf tissue infiltrated with Agrobacterium containing pLSLGFP or pLSLGUS, or infiltrated with a 1:1 mixture of Agrobacterium containing pLSLGFP or pLSLGUS and p35SREP. Circular replicons were detected by PCR using primers NB415 and NB416. Template switching was minimized by using the Expand Long Template PCR mix (Roche) following manufacturer's protocols. Strong amplification of LIR sequence only from samples co-transformed with p35SREP suggests that GVRs were present in the transformed cells (FIG. 15, bottom). Taken together, these data illustrate that GVRs can facilitate transient delivery of reporter proteins.

Demonstrating Targeted Mutagenesis by Delivery of ZFNs in Nicotiana tabacum Leaf Tissue Using GVRs

To demonstrate targeted mutagenesis, pLSL was modified to encode a Zif268::FokI ZFN. Zif268::FokI sequence was amplified from pDW1345 using primers NB379 and NB380. Forward and reverse primers contained XbaI and AatII restriction enzyme sites for cloning into pNJB091. The resulting vector was used in a MultiSite Gateway recombination reaction with pLSL and pNB098 to generate pLSLZ.D. The resulting vectors were sequence verified and transformed into Agrobacterium tumefaciens GV3101 by the freeze-thaw method. Target sequence for Zif268 is present within a gus::nptII reporter gene that is stably integrated in the genome of N. tabacum plants (FIG. 16). Leaf tissue was syringe infiltrated with Agrobacterium containing pLSLZ.D, or coinfiltrated with Agrobacterium containing pLSLZ.D and p35SREP. Plant DNA was extracted seven dpi, replicational release was verified (FIG. 17), and Zif268 target sequence was analyzed for ZFN-induced non-homologous end joining (NHEJ) mutations. To this end, a 484 bp DNA sequence, encoding the Zif268 target sequence, was amplified by PCR using primers NB422 and NB424. The resulting amplicons were purified and used as a template in a second PCR with primers NB396 and NB307 (FIG. 18). The PCR product was digested overnight with MseI and separated on an agarose gel. Cleavage-resistant products, present only in the pLSLZD and p35SREP lane, were cloned and sequenced (FIG. 18). Six out of eight sequenced clones contained mutations at the Zif268 target sequence. Five out of the six sequences encoded distinct NHEJ mutations suggesting GVR-mediated delivery of Zif268:FokI occurred in multiple somatic cells. Furthermore, densitometry analysis of cleavage-resistant amplicons indicates approximately 10% of reporter genes encode NHEJ mutations. Together, these results suggest GVRs enable targeted mutagenesis by the transient delivery of ZFN protein.

Demonstrating Targeted Mutagenesis by Delivery of TALE Nucleases in Nicotiana tabacum Leaf Tissue Using GVRs

Replicon-mediated expression of a ZFN monomer is predicted to be efficient due to its relatively small coding sequence (the Zif268::FokI gene is 897 nt) and minimal sequence repeats. To assess whether GVRs can facilitate delivery of large and repetitive TALE nuclease sequence, pLSL was modified to encode two TALE nuclease sequences separated by a T2A translational-skipping sequence (pLSLT). Target sequence for the TALE nuclease pair is present within two endogenous ALS genes, SuRA and SuRB (Zhang et al., Plant Physiol. 161:20-27, 2012, FIG. 16). WT N. tabacum leaves were syringe infiltrated with Agrobacterium containing pLSLT, or coinfiltrated with Agrobacterium containing pLSLT and p35SREP. Plant DNA was extracted seven dpi, replicational release was verified (FIG. 17), and SuRA and SuRB loci were amplified following an initial digestion of genomic DNA with AluI. Resulting amplicons were digested with AluI overnight and separated on an agarose gel (FIG. 19). Sequencing of cleavage-resistant amplicons confirmed TALE nuclease-induced NHEJ mutations in seven out of eleven clones. These results suggest GVR-mediated TALE nuclease expression can be achieved.

Demonstrating Targeted Mutagenesis by Delivery of CRISPR/Cas Elements in Nicotiana tabacum Leaf Tissue Using GVRs

The CRISPR/Cas system functions to protect bacteria and archaea against invading foreign nucleic acid. It was previously demonstrated that targeted DNA double-strand breaks (DSBs) could be created in mammalian cells by expression of the Cas9 endonuclease and a programmable guide RNA (gRNA). We tested whether the CRISPR/Cas system is functional in plant cells using GVRs to deliver the components necessary for targeted DNA cleavage. The LSL T-DNA was modified to encode a plant codon-optimized Cas9 followed by gRNA driven by an AtU6 RNA polymerase III promoter. The gRNA was designed to recognize a site in SuRA and SuRB approximately 100 bp downstream of the T30 TALEN target (FIG. 16). Genomic DNA was extracted five dpi, replicational release was verified (FIG. 17; pLSLC), and PCR products encompassing the gRNA target were subjected to AlwI digestion (FIG. 20). DNA sequencing of AlwI resistant products derived from the sample transformed with pLSLC and p35SREP confirmed the presence of mutations at the predicted target site in five out of seven clones. Notably, one of the mutant amplicons contained an intact AlwI site but also had a four by deletion; recovery of this mutant was likely due to incomplete digestion of the PCR amplicon. The data demonstrate that the CRISPR/Cas system can be used to make targeted modifications to plant genomes and that GVRs can simultaneously deliver gRNA and the Cas9 endonuclease.

Demonstrating GT in Nicotiana tabacum Using GVRs

GVRs were assessed for their ability to achieve GT through the coordinated delivery of nucleases and repair templates. The target for modification was the defective gus::nptII gene, which can be repaired by correcting a 600 bp deletion that removes part of the coding sequences of both GUS and NPTII. Following Zif268::FokI in pLSLZ.D is a us::NPTII repair template (FIG. 21). Cells having undergone GT will stain blue when incubated in a solution with the GUS substrate X-Gluc. Random integration of the repair template or read-through transcription from viral promoters should not produce functional GUS protein due to 703 nt missing from the 5′ coding sequence. This was confirmed by delivering pLSLZ.D and p35SREP to non-transgenic leaf tissue; no GUS activity was observed (data not shown). To compare the performance of GVRs with the delivery of conventional T-DNA technology, a T-DNA vector was engineered to encode Zif268::FokI and a us:NPTII repair template (p35SZ.D). To this end, Multisite Gateway recombination was performed using plasmids pMDC32, pNB098 and pNB091. Due to the two-component design GVRs—requiring co-delivery of pLSLZD and p35SREP, a direct comparison of GT frequencies with p35SZD results in a performance bias, favoring the system that requires transfer of the least number of T-DNAs. While this may be an influencing factor, co-transformation of T-DNA in Nicotiana species is efficient (McCormac et al., Transgenic Res. 25:549-561, 2001), likely leading to minimal loss of performance with GVRs. Five to seven dpi, infiltrated leaf tissue was stained in X-Gluc and chlorophyll was removed. Relative to p35SZ.D, a substantial enhancement in the number of GUS-expressing cells in leaf tissue transformed with pLSLZD and p35SREP (FIG. 22) was observed. To molecularly verify repair of reporter gene coding sequences, PCR was performed using primers NB394 and NB423, which bind to sequence within the 600 bp modification and are complementary to sequence downstream of the homology encoded on the repair template. A ˜1,000 bp product, present only in the lane with p35SREP and pLSLZ.D suggested the presence of repaired reporter genes (FIG. 23). To quantify the relative enhancement of GT, the density of blue sectors was quantified from four transgenic plant lines (1.7, 4.3, 9.1, and 11.3). A significant enhancement in blue sectors with pLSLZ.D and p35SREP was observed across all four plant lines (FIG. 24) was observed. Table 2 indicates the total number of blue sectors in leaf tissue transgenic lines.

Exploring Elements of GVRs Necessary for High frequency GT

There are several features of GVRs that may promote GT, including high levels of nuclease expression, high levels of repair template production and pleotropic Rep and RepA activity. To individually test these features, we paired two experimental samples on a single leaf to minimize variation caused by differences in leaf age and health, and quantified the density of blue sectors that result from GT. To determine the contribution of ZFN expression on GT, the coding sequence Zif268::FokI was replaced with GFP. Consistent with the stimulatory effect DSBs have on recombination, we observed a significant decrease in blue sectors when Zif268::FokI was removed (FIG. 25, top left). To determine if Rep-mediated replication of the GVRs contributes to GT, we compared the co-delivery of pLSLZ.D and p35SREP with the co-delivery of p35SZ.D and p35SREP. The decrease in blue sectors observed after removing the cis-acting LIR and SIR elements suggests that GVR replication contributes to enhanced rates of GT (FIG. 25, top right). Finally, to determine if there are pleotropic consequences of Rep and RepA expression on GT, we compared frequencies of GT using our standard T-DNA vector (p35SZ.D) with and without p35SREP. Here, we observed a significant increase in blue sectors when p35SREP was delivered, suggesting that pleotropic Rep and/or RepA activity promotes GT (FIG. 25, bottom left). See also FIGS. 26-30 for additional images of leaf tissue with GUS activity.

Mastrevirus RepA is known to interact with plant cell proteins, including the retinoblastoma-related protein pRBR. By sequestering pRBR's repressive activity against E2F, S-phase progression is promoted, providing the necessary factors for genome replication. One explanation for our results showing a pleotropic activity of replicase proteins on GT is that, in somatic leaf tissue, RepA promotes cell-cycle progression from G0/G1 to S phase and thereby provides improved cellular conditions for homologous recombination. To test this hypothesis, we introduced a single amino acid substitution within the conserved pRBR-interacting domain of RepA (designated LxCxQ) which reduces binding affinity to pRBR. A significant decrease in GT was observed when LxCxQ RepA T-DNA was delivered (FIG. 25, bottom right), suggesting that progression into S-phase stimulates GT.

Demonstrating Methods for Regeneration of Recombinant Nicotiana tabacum Plants

To regenerate modified Nicotiana tabacum plants, the leaf disc transformation protocol was implemented (Horsch et al., Science 227:1229-31, 1985). The target gene was the endogenous SuRB gene. A repair template, present downstream of the T30 TALEN pair on pLSLT, contained 1 kb of sequence homologous to the SuRB locus flanking NPTII coding sequence. As a consequence of GT, the NPTII coding sequence is placed in-frame with the SuRB coding sequence, resulting in the production of a SuRB::NPTII fusion protein. Agrobacterium containing pLSLT and p35SREP were grown overnight at 28° C. in LB with 50 μg/ml kanamycin and 50 μg/ml gentamycin. Cells were pelleted and resuspended to an OD600 of 1 in LB. Leaf discs from WT tobacco plants were transferred into the Agrobacterium cultures for 10 minutes and then plated onto co-cultivation media as described elsewhere (Gallois and Marinho, Methods Mol. Biol. 49:39-48, 1995). Three days after transformation, discs were transferred to regeneration plates containing 50 μg/ml kanamycin and 1 mg/L 6-Benzylaminopurine. Shoots that appear about four weeks after transformation were assessed for the presence of the SuRB:NPTII fusion gene by PCR (FIG. 31). Amplification of a ˜1.2 kb product (plant #6) suggests this plant was produced from a cell that has undergone GT. Amplification of the 5′ junction may suggest that the GT event was ‘one-sided’ (e.g. following invasion of the repair template by a free 3′ end of the chromosomal DNA, the NPTII sequence is copied and then the break is sealed by illegitimate recombination).

Demonstrating Replicational Release in Potato

Functionality of BeYDV replicons in economically-valuable crops was investigated. To this end, experiments were first undertaken to demonstrate replicational release in potato cells (Solanum tuberosum cultivar Deseree). Potato leaf tissue was excised from aseptically-growing plants, and co-transformed with Agrobacterium containing p35SREP and pLSLGFP. Following co-transformation, leaf tissue was plated on cocultivation media for 2 days to allow for T-DNA transfer and integration. Leaf tissue was then washed in MS media containing 250 μg/mL cefotaxime, and plated on regeneration media containing 50 μg/mL hygromycin. Genomic DNA from several lines of hygromycin-resistant potato plants (Line 1, 3, 4, 5, 9, 10, 11, 12) was isolated and assessed for the presence of p35SREP T-DNA and circular replicons. Amplification of a 440 bp sequence from Rep/RepA and a 714 bp sequence from replicon nucleotide sequence from plant line 10 suggests GVRs are present in potato cells (FIG. 32). Interestingly, expression of Rep/RepA does not elicit an observable hypersensitive response. This was demonstrated by verifying expression of Rep/RepA in phenotypically-normal hygromycin-resistant plants by RT-PCR using primers that detect Rep/RepA RNA sequence (FIG. 33).

Demonstrating Transient Delivery of Reporter Proteins in Tomato Leaf Tissue Using GVRs

To demonstrate functionality of BeYDV-based GVRs in tomato (Solarium lycopersicum cv. M82), pLSLGUS and p35SREP were transformed into Agrobacterium tumefaciens (AGL1) by the freeze-thaw method. Agrobacterium was grown overnight at 28° C. to an OD600 of 1 and diluted in LB media to an OD600 of 0.2. Half leaves were fully infiltrated with Agrobacterium encoding pLSLGUS or coinfiltrated with pLSLGUS and p35SREP. To detect cells expressing GUS enzyme, leaf tissue was stained eleven dpi in X-Gluc solution. Chlorophyll was removed using 80% ethanol, and leaf images were taken (FIG. 34). The presence of GUS-expressing cells only in tissue transformed with pLSLGUS and p35SREP (FIG. 35) suggested GVRs can drive transient protein expression in tomato leaf tissue.

Demonstrating Functionality of Wheat Dwarf Virus Replicons in Wheat, Setaria, and Maize

To expand the use of GVRs for genome editing in monocotyledonous plants, an LSL T-DNA was constructed with cis-acting replication sequences from the Wheat dwarf virus (WDV) (FIG. 36). Rep/RepA coding sequence was positioned inside the flanking LIR sequences, just downstream of the complementary sense LIR promoter. To demonstrate transient protein expression, WDV LSL plasmids containing the GFP gene (WDV-GFP) were delivered to wheat (Triticum aestivum cultivar Bobwhite), Setaria (Setaria viridis) and maize (Zea mays cultivar A188), by particle bombardment. Three days post bombardment, tissue was assessed for GFP expression. Enhanced expression of GFP was observed in wheat calli (FIG. 37), Setaria calli (FIG. 38), and corn embryos (FIG. 39) when delivered WDV-GFP. One explanation for these results may be that WDV replicons are replicating and promoting GFP expression.

Demonstrating GT in Rice Using WDV Replicons

To determine if WDV can facilitate the delivery of TALENs and repair templates for GT in rice, a WDV replicon was engineered to contain the T30 TALEN pair followed by a repair template designed to correct the non-functional gus::nptII gene (FIG. 40, top). Leaf tissue from transgenic rice plants, containing a stably integrated gus::nptII gene, was exposed to Agrobacterium containing WDV T-DNA plasmids with or without repair template sequence. Transformation conditions were performed as previously described by Andrieu et al. (Rice, 5:23, 2012). Leaf tissue also was transformed with conventional T-DNA containing the T30 TALEN pair followed by the us::NPTII repair template. Blue sectors observed in leaf tissue delivered GVR T-DNA and conventional T-DNA suggests that gus::nptII gene function was restored through GT in a subset of leaf cells (FIG. 40, bottom).

OTHER EMBODIMENTS

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

TABLE 1 Primer Sequence SEQ ID NO NB104 gcggtaccctcgagtctagatctgtctttttccaaatatttattg 2 NB105 gcggtaccctcgagtctagatttttgtggtggttgcagc 3 NB106 gcggtaccctcgagtctagacaagattctcttcacttctc 4 NB112 aggctagcgagctcagatctccttgtcaagaggagcatc 5 NB113 aggctagcgagctcagatcttttgccattaatggagaatcttg 6 NB114 aggctagcgagctcagatctcaatgacgacactccccac 7 NB128 gcatgcagtactgagctcgccgaagatacgtggaaac 8 NB129 gagctcagtactgcatgcgctggagggtaatagaaac 9 NB153 gattaggctagcgagctcagatct 10 NB154 cggacagattattcgatgcaaagg 11 NB155 gacaaaccacaactgacaatacaga 12 NB158 gcggtaccctcgagtctaga 13 NB161 tcaccatcgtgaatcatccctcct 14 NB177 tcgagtctagacacaatcacacaaaactaacaaaag 15 NB178 gctcagatctgcaccaagaccaaaaatggcaac 16 NB207 gcatgcagtactgagctcacgtggaaacaacggtgtttg 17 NB208 gagctcagtactgcatgctaatagaaacactaatcttc 18 NB257 tgccacgtggacgaatactagcaa 19 NB258 gcttgaatcatggcctgaacgctt 20 NB263 gagctcagtactgcat 21 NB264 gcatgcagtactgagc 22 NB271 ttacggtttttcaccgaagttcat 23 NB272 ttcggtgaaaaaccgtaaaccgacctgtccggtgccctg 24 NB274 actgatctagacactggcggaagcaacgcgta 25 NB275 tcagtagatctgccatgatggatactttctcg 26 NB307 gccatgatggatactttctcg 27 NB314 caacttttgtatacaaagttggcattataaaaaagcattgctcatcaatttgttgcaacgaacaggtcactatcagtcaaaataaa 28 atcattatt NB315 aagctcgggcccaataatgattttattttg 29 NB316 ctttgtatacaaaagttgccgagctcgcggccgcattaggcaccccag 30 NB317 aactttgtacaagaaagctgggtcgtcgacctgcagactggctgtg 31 NB318 aggaagtgagacggaaatttaataacggcgagataaacttttaataggacgtccgatcgttcaaacatttggcaataaagtttct 32 taagattgaatcctgttgccggtcttgcgatgattatcatataatttctgttgaattacgttaagcatgtaataattaacatg taatgcatgacgttatttatgagatgggtttttatgattagagtcccgcaattatacatttaatacgcgatagaaaacaaaat atagcgcgcaaactaggataaattatcgcgcgcggtgtcatctatgttactagatcgggaattgatcccccctcgacagctt ccggaaagggcgaattcgcaactttgtatacaaaagttgaacgagaaacgtaaaatgatataaatatcaatatattaaattag attttgcataaaaaacagactacataatactgtaaaacacaacatatccagtcactatgccatccagctgatatcccctat NB319 cgacggccagtcttaagctcgggccccaaataatgattttattttgactgatagtgacctgttcgttgcaacaaattgatgagca 33 atgcttttttataatgccaactttgtacaaaaaagcaggctccgaattcatgccttctgctagtaagaacttcagactccaat ctaaatatgttttccttacctatcccaagtgctcatctcaaagagatgatttattccagtttctctgggagaaactcacacct tttcttattttcttccttggtgttgcttctgagcttcatcaagatggcactacccactatcatgctcttctccagcttgataa aaaaccttgtattagggatccttcttttttcgattttgaaggaaatcaccctaatatccagccagctagaaactctaaacaag tccttgattacatatcaaaggacggagatattaaaaccagaggagatttccgagatcataaggtctctcctcgcaaatctgac NB320 attaaaaccagaggagatttccgagatcataaggtctctcctcgcaaatctgacgcacgatggagaactattatccagactgca 34 acgtctaaggaggaatatcttgacatgatcaaggaagaattccctcatgaatgggcaacaaagcttcaatggctggaatattc agccaacaaattattccctccacaacctgaaccgtatgtgtcgcccttcacagaatcagatcttcgctgccacgaagatctac actcctggagggaaacccatctataccatgtaagcatagacgcttatacttacatacatcctgtctcataccaacaagctcaa tctgaccttgaatggatggccgatttaaccaggacaatggaaggaatggaatccgacaccccagcctctacatctgcggacca actcgtaccggaaagaccacctgggctagaagtctcggacgacacaactattggaacggtaccatcgatttcaccaactacgat NB321 attaaaaccagaggagatttccgagatcataaggtctctcctcgcaaatctgacgcacgatggagaactattatccagactgcaa 35 cgtctaaggaggaatatcttgacatgatcaaggaagaattccctcatgaatgggcaacaaagcttcaatggctggaatattca gccaacaaattattccctccacaacctgaaccgtatgtgtcgcccttcacagaatcagatcttcgctgccaccaagatctaca ctcctggagggaaacccatctataccatgtaagcatagacgcttatacttacatacatcctgtctcataccaacaagctcaat ctgaccttgaatggatggccgatttaaccaggacaatggaaggaatggaatccgacaccccagcctctacatctgcggaccaac tcgtaccggaaagaccacctgggctagaagtctcggacgacacaactattggaacggtaccatcgatttcaccaactacgat NB322 gtaccggaaagaccacctgggctagaagtctcggacgacacaactattggaacggtaccatcgatttcaccaactacgatgaaca 36 cgccacctataatatcatcgacgacatccccttcaagttcgtcccattgtggaagcaattaataggttgccagtctgatttca ctgtcaaccctaaatatggaaaaaagaagaaaataaaaggtgggatcccttctataattctttgcaatcctgacgaagactgg atgttatcaatgacaagtcaacagaaggattactttaaagataattgcgtcacccactacatgtgtgacggggagactttttt tgctcgggaatcgtcgagtcactgaacgtgcctgaattcgacccagctttcttgtacaaagttggcattataaaaaataatt gctcatcaatttgttgcaacgaacaggtcactatcagtcaaaataaaatcattatttgccatccagctgatatcccctatagtg NB323 cgacggccagtcttaagctc 37 NB324 cactataggggatatcagct 38 NB325 agcttggtacccctgcaggtagcagaaggcatg 39 NB326 ataagcacaagttttatccggc 40 NB327 ggatcctctagattacgccccgcctgc 41 NB328 cgtaatctagaggatccggcttactaaaagc 42 NB329 tgttgaccgagctcctgcagaagcttctcgag 43 NB330 cctgcaggtagcagaaggcatgttgttgtgactccgaggggttgcctcaaactctatcttataaccggcgtggaggcatggagg 44 caggggtattttggtcattttaatagatagtggaaaatgacgtggaatttacttaaagacgaagtcgagacctttgcgactct agaggtctcaaatttaatattaccggcgtggcccccccttatcgcgagtgctttagcacgagcggtccagatttaaagtagaa aatttcccgcccactagggttaaaggtgttcacactataaaagcatatacgatgtgatggtatttgatggagcgtatattgta tcaggtatttccgttggatacgaattattcgtacgaccctccctaagattcttgattgtttataaaaccaaatctcattgtct ttgttgtgtattgtttgcaggacgtcgagagttctcaacacaacatatacaaaacaaacgaatctcaagca NB331 acaaaacaaacgaatctcaagcaatcaagcattctacttctattgcagcaatttaaatcatttctacaagtttgtacaaaaaag 45 ctgaacgagaaacgtaaaatgatataaatatcaatatattaaattagattttgcataaaaaacagactacataatactgtaaa acacaacatatccagtcactatggcggccgcattaggcaccccaggctttacactttatgcttccggctcgtataatgtgtgg attttgagttaggatccgtcgagattttcaggagctaaggaagctaaaatggagaaaaaaatcactggatataccaccgttga tatatcccaatggcatcgtaaagaacattttgaggcattttcagtcagttgctcaatgtacctataaccagaccgttcagctgg atattacggcctttttaaagaccgtaaagaaaaataagcacaagttttatccggcctttattcaca NB332 ctatggtcgacctgcagactggctgtg 46 NB333 gggatcccactcgagggtcaacatggtggagcacg 47 NB334 ctagagtcgaggtcctctcca 48 NB335 aggtggctcctacaaatgccatcattgcgataaaggaaaggccatcgttgaagatgcctctgccgacagtggtcccaaagatgg 49 acccccacccacgaggagcatcgtggaaaaagaagacgttccaaccacgtcttcaaagcaagtggattgatgtgatatctcca ctgacgtaagggatgacgcacaatcccactatccttcgcaagacccttcctctatataaggaagttcatttcatttggagagg acctcgactctagccttcctctatataaggaagttcatttcatttggagaggtaagtttcacttcacacattattactgtctt ctaatacaaggttttttatcaagctggagaagagcatgatagtgggtagtgccatcttgatgaagctcagaagcaacaccaag gaagaaaataagaaaaggtgtgagtttctcccagagaaactggaataaatcatctctttgagatgagcacttgggataggtaag NB336 tgagatgagcacttgggataggtaaggaaaacatatttagattggagtctgaagttcttactagcagaaggcatgttgttgtga 50 ctccgaggggttgcctcaaactctatcttataaccggcgtggaggcatggaggcaggggtattttggtcattttaatagatag tggaaaatgacgtggaatttacttaaagacgaagtcgagacctttgcgactctagaggtctcaatttaatattaccggcgtgg cccccccttatcgcgagtgctttagcacgagcggtccagatttaaagtagaaaatttcccgcccactagggttaaaggtgttca cactataaaagcatatacgatgtgatggtatttgatggagcgtatattgtatcaggtatttccgttggatacgaattattcg tacgaccctcatagtttaaactgaaggcgggaaacgacaatctgatccaagctcaagctaagcttgcatgcctgcaggatatcg NB337 cgatatcctgcaggcatgcaagcttagc 51 NB338 aagtctttgcgacaagggggggcccacgccg 52 NB339 aattcggcgtgggcccccccttgtcgcaaag 53 NB344 cacagccagtctgcaggtcgaccatagtgactggatatgttgtgttttacagtattatgtagtctgttttttatgcaaaatcta 54 atttaatatattgatatttatatcattttacgtttctcgttcagctttcttgtacaaagtggtgagtgtacttcaagtcagtg ggaaatcaataaaatgattattttatgaatatatttcattgtgcaagtagatagaaattacatatgttacataacacacgaaa taaacaaaaaaagacaatccaaaaacaaacaccccaaaaaaaataatcactttagataaactcgtatgaggagaggcacgttc agtgactcgacgattcccgagcaaaaaaagtctccccgtcacacatgtagtgggtgacgcaattatctttaaagtaatccttc tgttgacttgtcattgataacatccagtcttcgtcaggattgcaaagaattatagaagggatcccactcgagaagcttctgcag NB362 acctcgactctagaatgaagactaatctttttctctttc 55 NB363 gaacgatcggacgtcttaaagctcatcatgtttgtatag 56 NB379 gcccttcaccatggcttcctcccctccaaagaaaaag 57 NB380 gaacgatcggacgtcctattaaaagtttatctcaccgtta 58 NB394 tgccgccgtgttccggctgtcagc 59 NB396 aaggtgcacgggaatatttcgcgc 60 NB415 gtttcacttcacacattattactg 61 NB416 tgttgagaactctcgacgtcctgc 62 NB422 gtgtgaacaacgaactgaactggc 63 NB423 agagcgcccaatacgcaaaccgc 64 NB424 cagcgagtcagtgagcgaggaagc 65 NB448 agctagtctagaatgttacgtcctgtagaaacc 66 NB449 gtacgtgacgtctcattgtttgcctccctgc 67 NB478 gacggtgcagaaagtgaagta 68 NB479 tatggcccaggagtgtctaa 69 NB488 caagctaagcttgcatgcctgcagggtgtttgacaggatatattggcg 70 NB489 tccatgccgcctcctttagc 71 NB490 gctaaaggaggcggcatgga 72 NB491 accacttcaagaactctgtagc 73 NB492 gctacagagttcttgaagtggtg 74 NB493 aggcacgttcagtgactcgacgaagtagatgccgaccggatctgtcg 75 NB494 gaagttcttactagcagaaggcatcggatctgcgaaagctcgagag 76 NB495 gtcacaacaacatgccttctgctacctgcaggcgtaatcatggtcatagc 77

TABLE 2 Delivered T-DNA: p35SZ.D pLSLZ.D + p35SREP pLSLD + p35SREP Transgenic plant line ID: 1.7 4.3 9.1 11.3 1.7 4.3 9.1 11.3 1.7 4.3 11.3 Leaf 1 (blue sectors/cm2) 0.00 1.85 0.93 0.56 479.26 85.37 218.89 372.96 0.19 7.41 1.11 Leaf 2 (blue sectors/cm2) 1.48 7.78 13.89 2.96 160.93 96.67 77.22 147.96 0.00 0.00 Leaf 3 (blue sectors/cm2) 0.93 22.96 0.00 1.48 170.19 68.15 120.37 61.67 2.04 Leaf 4 (blue sectors/cm2) 0.74 1.11 287.22 25.00 38.15 2.22 Leaf 5 (blue sectors/cm2) 1.11 6.11 101.48 70.37 109.07 Leaf 6 (blue sectors/cm2) 10.00 13.33 90.74 96.48 Leaf 7 (blue sectors/cm2) 36.67 74.63 Leaf 8 (blue sectors/cm2) 27.96 p35SZ.D + Delivered T-DNA: pLSLZ.D p35SZ.D + p35SREP p35SREPLxCxQ Transgenic plant line ID: 1.7 9.1 11.3 1.7 4.3 9.1 11.3 1.7 9.1 11.3 Leaf 1 (blue sectors/cm2) 7.04 47.96 5.00 16.11 19.26 4.26 67.41 3.89 0.00 0.00 Leaf 2 (blue sectors/cm2) 7.04 16.48 19.81 0.93 Leaf 3 (blue sectors/cm2) 0.93 6.67 7.04 0.00 Leaf 4 (blue sectors/cm2) 23.15 0.00 Leaf 5 (blue sectors/cm2) 3.70 0.00 Leaf 6 (blue sectors/cm2) 2.22 Leaf 7 (blue sectors/cm2) 11.11 Leaf 8 (blue sectors/cm2) 0.19 Leaf 9 (blue sectors/cm2) 0.93 Leaf 10 (blue sectors/cm2) 8.15

Claims

1. A method for modifying the genetic material of a plant cell, comprising:

(a) introducing into the cell a virus nucleic acid comprising a donor sequence that is heterologous to the virus and is targeted to a first sequence that is endogenous to the plant cell; and
(b) inducing a double strand break at or near the sequence to which the donor sequence is targeted, wherein said double strand break is generated by an endonuclease targeted to a second endogenous plant sequence at or near the first sequence that is targeted by the donor sequence,
wherein homologous recombination occurs between the first endogenous plant sequence and the donor sequence.

2. The method of claim 1, wherein the virus nucleic acid is a plant DNA virus nucleic acid.

3. The method of claim 1, wherein the virus nucleic acid is a geminivirus nucleic acid.

4. The method of claim 1, wherein the endonuclease is a zinc finger nuclease, a transcription activator-like effector nuclease, a meganuclease, or a CRISPR/Cas system endonuclease.

5. The method of claim 1, wherein the endonuclease is encoded by a transgene sequence stably integrated into the genetic material of the plant, or is expressed transiently.

6. The method of claim 5, wherein the transgene encoding the endonuclease is operably linked to a promoter that is constitutive, cell specific, inducible, or activated by alternative splicing of a suicide exon.

7. The method of claim 1, wherein the virus nucleic acid comprises a sequence encoding the endonuclease.

8. The method of claim 1, further comprising introducing into the plant cell an RNA virus nucleic acid comprising a nucleotide sequence encoding the endonuclease.

9. The method of claim 8, wherein the RNA virus nucleic acid is introduced into the plant cell after or simultaneous with step (a).

10. The method of claim 8, wherein the RNA virus nucleic acid is from a tobacco rattle virus, a potato virus X, a pea early browning virus, or a barley stripe mosaic virus.

11. The method of claim 1, wherein the plant is monocotyledonous.

12. The method of claim 11, wherein the plant is wheat, maize, or Setaria.

13. The method of claim 1, wherein the plant is dicotyledonous.

14. The method of claim 13, wherein the plant is tomato, soybean, tobacco, potato, or Arabidopsis.

Patent History
Publication number: 20150225734
Type: Application
Filed: Jun 19, 2013
Publication Date: Aug 13, 2015
Applicant: Regents of the University of Minnesota (Minneapolis, MN)
Inventors: Daniel F. Voytas (Falcon Heights, MN), Nicholas Baltes (New Brighton, MN)
Application Number: 14/409,148
Classifications
International Classification: C12N 15/82 (20060101); C12N 15/90 (20060101);