CA2396898A1 - Methods and compositions to modulate expression in plants - Google Patents

Methods and compositions to modulate expression in plants Download PDF

Info

Publication number
CA2396898A1
CA2396898A1 CA002396898A CA2396898A CA2396898A1 CA 2396898 A1 CA2396898 A1 CA 2396898A1 CA 002396898 A CA002396898 A CA 002396898A CA 2396898 A CA2396898 A CA 2396898A CA 2396898 A1 CA2396898 A1 CA 2396898A1
Authority
CA
Canada
Prior art keywords
zinc finger
protein
target
expression
finger protein
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA002396898A
Other languages
French (fr)
Inventor
Carlos F. Barbas, Iii.
Justin T. Stege
Xueni Guan
Bipin Dalmia
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Syngenta Participations AG
Scripps Research Institute
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Publication of CA2396898A1 publication Critical patent/CA2396898A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/415Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from plants
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/82Vectors or expression systems specially adapted for eukaryotic hosts for plant cells, e.g. plant artificial chromosomes (PACs)
    • C12N15/8216Methods for controlling, regulating or enhancing expression of transgenes in plant cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/82Vectors or expression systems specially adapted for eukaryotic hosts for plant cells, e.g. plant artificial chromosomes (PACs)
    • C12N15/8241Phenotypically and genetically modified plants via recombinant DNA technology
    • C12N15/8261Phenotypically and genetically modified plants via recombinant DNA technology with agronomic (input) traits, e.g. crop yield
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/82Vectors or expression systems specially adapted for eukaryotic hosts for plant cells, e.g. plant artificial chromosomes (PACs)
    • C12N15/8241Phenotypically and genetically modified plants via recombinant DNA technology
    • C12N15/8261Phenotypically and genetically modified plants via recombinant DNA technology with agronomic (input) traits, e.g. crop yield
    • C12N15/8287Phenotypically and genetically modified plants via recombinant DNA technology with agronomic (input) traits, e.g. crop yield for fertility modification, e.g. apomixis
    • C12N15/8289Male sterility
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A40/00Adaptation technologies in agriculture, forestry, livestock or agroalimentary production
    • Y02A40/10Adaptation technologies in agriculture, forestry, livestock or agroalimentary production in agriculture
    • Y02A40/146Genetically Modified [GMO] plants, e.g. transgenic plants

Abstract

The invention relates to the field of plant and agricultural technology. Mor e specifically, the invention relates to the use of zinc finger proteins and fusions of said proteins to regulate gene expression and metabolic pathways in plants.

Description

METHODS AND COMPOSITIONS TO MODULATE EXPRESSION IN
PLANTS
This invention was made with U.S. government support under Contract No.
GM53910 by the National Institutes of Health. The U.S. government has certain rights in the invention.
Related Applications This is a continuation-in-part of U.S. Application Serial No. 09/620,897, now pending, which claims priority, under 35 U.S.C. ~ 119(e), ofU.S. Provisional Patent Application Serial No. 60/177,468, filed January 21, 2000. The disclosure of the above referenced applications is incorporated by reference in its entirety.
Technical Field The invention relates to the field of plant and agricultural technology. More specifically, the invention relates to the use of zinc finger proteins and fusions of said proteins to regulate gene expression and metabolic pathways in plants.
Back~,round Art Genetic expression in general, and in plants in particular, is controlled at both transcription and translation levels. Regulation of transcription often involves regulatory factors which contain zinc finger domains of a particular family -e.g., domains which comprise approximately 30 amino acids containing two cysteine and two histidine residues folded around a zinc ion and providing an alpha helical recognition sequence specific at the "fingertip" for a particular 3-nucleotide sequence.
The nature of such zinc fingers is set forth, for example, in WO 98/54311, the contents of which are incorporated herein by reference. As there are 64 possible three nucleotide targets for binding by zinc fingers, it is theoretically possible to design 64 individual zinc fingers, each of which would bind specifically to only one of the 64 possible triplets. By combining multiple zinc finger motifs, a larger target sequence could be bound specifically.
It has been calculated that a nucleotide sequence containing only 18 nucleotides would serve as an unique address within even a 68 billion pair genome;
thus, an 18 nucleotide sequence would clearly serve as an unique target within the human genome (3.5 billion bp) or maize (2 billion bp). Thus, a hexadactyl zinc finger protein, properly designed, could target any arbitrary unique sequence within the human or maize genome. Considering the complexity of human and maize genome, this specially designed zinc finger protein could target any unique sequence within these and other organisms.
In the context of regulatory transcription factors, zinc finger domains which are responsible for specifically targeting a particular nucleotide sequence within a gene are generally coupled to additional amino acid sequences which serve to modulate expression either by activating (amplifying) or repressing it. Thus, typical transcription regulatory factors comprise both a zinc finger domain responsible for targeting the appropriate position of the genome and a functional portion which controls transcription of the gene once the fusion protein is bound.
Synthetic zinc finger proteins have been synthesized and found to have binding affinity similar to those found in native transcription factors.
Further, zinc finger proteins have been designed which are specific for TGA or for one of the triplets of the formula GNN. Thus, zinc finger proteins can be designed to target unique sequences of the formula (GNN)6 or sequences containing 18 nucleotides wherein some of the GNN triplets have been substituted by TGA. As the design of zinc finger proteins progresses, appropriate zinc finger domains can be designed for any desired target sequence.
There is evidence to show that specific synthetic zinc finger proteins can transiently regulate reporter gene, e.g., luciferase, expression in cultured mammalian cells when fused with a transcriptional activation or repression domain (Liu et al., P~oc. Natl. Acad. Sci. USA, 94:5525-5530 (1997); Wu et al., PYOC. Natl. Acad.
Sci.
USA, 92:344-348 (1995); and Beerli et al., Pr~oc. Natl. Acad. Sci. USA, 95:14628-14633 (1998)). However, there has been no data showing: (1) if these synthetic zinc finger proteins can be used to manipulate endogenous gene expression; and (2) if such transient regulation can be stabilized. There has also been no scientific evidence predicting if this technology will perform well in a whole living organism as it may in tissue culture system.
In addition to all these unknown factors, a plant cell is considered different than a mammalian cell in numerous aspects even though they share most of the basic features of living organisms. First, plant cells have different subcellular biological structures, such as cell walls, which make the mechanism and procedure of transformation of foreign gene into plant cells significantly different from mammalian cells. Second, the genetic recombination mechanism and frequency in plant cells differ from that in mammalian cells as well. For any given transgene to be expressed and functional in any living cell, the very critical step is integration into host genome, the mechanism of which differs between plant and mammalian cells. Generally, plant cells have much lower integration frequency. Third, most plant cells have specialized metabolic pathway and enzymes catalyzing these pathways so that a gene functioning in a mammalian cell is not necessarily functional in a plant cell. Fourth, the preference of genetic codon usage is different amongst plant, mammalian, and other biological systems.
It is highly desirable to control expression of target genes in plants whether these genes are native to the plant or constitute modifications of the native plant genetic complement. The present invention provides such means, and is exemplified by the control of expression of three genes in plants: (1) the reporter gene luciferase in tobacco and maize cells; (2) the APETALA3 (AP3) gene in Arabidopsis plant, and (3) the gene encoding myo inositol 1-phosphate synthase (MIPS) in maize, all of which are representatives of economically valuable genes.
Thus, there remains a need for methods and compositions to control, at will, gene expression and other functions and activities in plants, plant tissues, and plant cells. The present invention address these and other related needs in the art.
Surnmary of the Invention The invention relates to the field of plant and agricultural technology. In one aspect, the present invention is directed to a method to modulate the expression of a target gene in plant cells, which method comprises providing plant cells with a zinc finger protein, said zinc finger protein being capable of binding, and preferably, specifically binding, to a target nucleotide sequence, or a complementary strand thereof, within a target gene, and allowing said zinc finger protein binding to said target nucleotide sequence, whereby the expression of said target gene in said plant cells is modulated.
The zinc finger protein can be provided to the plant cells via any suitable methods known in the art. For example, the zinc finger protein can be exogenously added to the plant cells and the plant cells are maintained under conditions such that the zinc forger protein binds to the target nucleotide sequence and regulates the expression of the target gene in the plant cells. Alternatively, a nucleotide sequence, e.g., DNA or RNA, encoding the zinc forger protein can be expressed in the plant cells and the plant cells are maintained under conditions such that the expressed zinc finger protein binds to the target nucleotide sequence and regulates the expression of the target gene in the plant cells.
A preferred method to modulate the expression of a target gene in plant cells comprises the following steps: a) providing plant cells with an expression system for a zinc finger protein, said zinc finger protein being capable of binding, and preferably specifically binding, to a target nucleotide sequence, or a complementary strand thereof, within a target gene; and b) culturing said plant cells under conditions wherein said zinc finger protein is produced and binds to said target nucleotide sequence, whereby expression of said target gene in said plant cells is modulated.
Any target nucleotide sequence can be modulated by the present method. For example, the target nucleotide sequence can be endogenous or exogenous to the target gene. Tn a specific embodiment, the target nucleotide sequence is endogenous to the plant but is in a non-naturally-occurring location. The target nucleotide sequence can be located in any suitable place in relation to the target gene. For example, the target nucleotide sequence can be upstream or downstream of the coding region ofthe target gene. Alternatively, the target nucleotide sequence is within the coding region of the target gene. Preferably, the target nucleotide sequence is a promoter of a regulatory protein. Also preferably, the target nucleotide sequence comprises 3, 6, 9, 12, 15 or 18 nucleotides. More preferably, the target nucleotide sequence comprises 18 nucleotides and wherein the zinc finger protein is a hexadactyl zinc finger protein.
Further preferably, the targeted nucleotide sequence is of the formula (GNN)", and wherein N is any one of the A, T, C or G and n is an integer from 1 to 6. More preferably, the targeted nucleotide sequence is of the formula (GNN)6, and wherein N
is any one of the A, T, C or G.
Plant cells containing any copy number of the target nucleotide sequence can be used in the present methods. For example, the plant cells comprising at least two copies of the same or different target nucleotide sequence can be used.
Alternatively, each target 'nucleotide sequence can be located within a different target gene so that more than one different target genes can be modulated.
Any target gene can be modulated by the present method. For example, the target gene can encode a product that affects biosynthesis, modification, cellular trafficking, metabolism and degradation of a peptide, a protein, an oligonucleotide, a nucleic acid, a vitamin, an oligosaccharide, a carbohydrate, a lipid, or a small molecule. Exemplary proteins or peptides include enzymes, transport proteins such as ion channels and pumps, nutrient or storage proteins, contractile or motile proteins such as actins and myosins, structural proteins, defense proteins or regulatory proteins such as antibodies, hormones and growth factors. Exemplary nucleic acids include DNA, such as A-, B- or Z-form DNA, and RNA such as mRNA, tRNA and rRNA.
The nucleic acids can be single-, double- and triple-stranded nucleic acids.
Exemplary vitamins include water-soluble vitamins such as thiamine, riboflavin, nicotinic acid, pantothenic acid, pyridoxine, biotin, folate, vitamin B12 and ascorbic acid, and fat-soluble vitamins such as vitamin A, vitamin D, vitamin E, and vitamin K. Exemplary lipids include triacylglycerols such as tristearin, tripalinitin and triolein, waxes, phosphoglycerides such as phosphatidylethanolamine, phosphatidylcholine, phosphatidylserine, phosphatidylinositol and cardiolipin, sphingolipids such as sphingomyelin, cerebrosides and gangliosides, sterols such as cholesterol and stigmasterol and sterol fatty acid esters. The fatty acids can be saturated fatty acids such as lauric acid, myristic acid, palmitic acid, stearic acid, arachidic acid and lignoceric acid, or can be unsaturated fatty acids such as palmitoleic acid, oleic acid, linoleic acid, linolenic acid and arachidonic acid. In a specific embodiment, the target gene encodes a protein or an RNA. In another specific embodiment, the target gene is a reporter gene, e.g., luciferase, the AP3 gene or the nayo inositol 1 phosphate synthase gene. In still another specific embodiment, the present method is used for treating a disorder in the plant cells, wherein the disorder is associated with abnormal expression of the target gene.
When the target gene encodes a target protein, the present method can be used to modulate the expression of said encoded target protein. Expression of any target protein can be modulated by the present method in plant cells. The protein whose expression being modulated can be endogenous or exogenous to the plant cell.
The modulation can be activation or inhibition.
In a specific embodiment, the protein whose expression being modulated is an antibody. In another specific embodiment, the protein whose expression being modulated participates in a metabolic pathway or controls a metabolic pathway, e.g., an anabolic or a catabolic pathway. The present method can be used for modulating metabolic pathways of any desirable molecules such as vitamins, taste molecules, e.g., S

bad taste molecules, anti-oxidants, sugars and flavanoids. The metabolic pathway being modulated can be endogenous or exogenous to the plant cell. In still another specific embodiment, target gene encodes a structural protein, e.g., an enzyme or a co-factor in a metabolic pathway, or a regulatory protein. In yet another specific embodiment, the metabolic pathway being modulated enhances an input or output trait in a plant or seed.
Although in some cases, a zinc finger protein itself is sufficient for modulating gene expression, the zinc finger protein is preferably fused to a protein which activates or represses gene expression, e.g., an activator domain of a regulatory protein or an active domain of a nucleic acid modifying protein.
For modulating the expression of the target gene, the zinc finger protein can bind to the target nucleotide sequence within the target gene. Alternatively, the zinc finger protein can bind to the complementary strand of the target nucleotide sequence.
Additionally, the zinc finger protein can specifically bind to an effector domain of the target sequence and whereby the expression of the target gene is modulated by competitive inhibition of said effector domain. In such case, the zinc finger protein preferably does not comprise an effector domain.
The zinc forger protein used in the present methods can comprise a plurality of finger regions. The zinc finger protein can comprise linker regions among the plurality of forger regions. For example, the zinc finger protein used in the present method can contain any number of the 3-finger region. Preferably, the zinc finger protein can comprise at least two 3-finger regions that are separated and linked together with a linker region. The linker region can be any suitable length, e.g., from about 2 to about 10 amino acid residues in length. Preferably, the linker region between any said two 3-finger region is about 5 amino acid residues in length.
In addition to the typical zinc finger domains, the zinc finger protein can further comprise other desirable domains such as effector domains active in the host plant cells. Any types of zinc finger protein can be used in the present method. But preferably, the zinc finger protein comprising a framework from a plant zinc forger protein can be used. Alternatively, synthetic zinc finger proteins or non-naturally-occurring zinc finger proteins can be used.
In a specific embodiment, the zinc finger protein used in the present methods is ZFPml, ZFPm2, ZFPm3, ZFPm4 or ZFPAp3, alone or fused to an effector domain.

In another specific embodiment, the zinc finger protein used in the present methods is not any zinc finger protein that is disclosed in U.S. Patent No.
6,140,466 or WO 98/54311, e.g., a zinc finger-nucleotide binding polypeptide variant comprising at least three zinc finger modules that bind to a target cellular nucleotide sequence and modulate the transcriptional function of the cellular nucleotide sequence, wherein the amino acid sequence of each zinc finger module that binds a target cellular nucleotide comprises two cysteines and two histidines whereby both cysteines are amino proximal to both histidines and wherein each of three modules of said variant has at least one amino acid sequence modification.
The present methods can be used to modulate gene expression in any plant cells, e.g., monocot or dicot plant cells. The plant cells can be in any suitable forms.
Fox example, the plant cells can be included within an intact plant, and preferably the plant cells constitute all the cells of an intact plant. Alternatively, the plant cells can be contained in an in vitf-o culture, and preferably be cultured in planta.
The plant cells can also be in the form of protoplasts or spheroplasts.
The present method can be used to achieve any desirable degree of the modulation of a target gene expression. Preferably, the modulation of the gene expression is at least two fold, e.g., at least five fold repression or at least two fold activation. Also preferably, the modulation changes the phenotype of the plant cells, the tissues) of the plant or the whole plant.
When the zinc forger protein is provided to plant cells with an expression system for a zinc finger protein, any suitable promoters can be used in directing expression of the zinc forger protein. The expression system can comprise a constitutive promoter or an inducible promoter. In addition, the zinc finger protein can be expressed transiently or stably.
Preferably, the expression system can comprise a tissue-specific promoter.
Also preferably, the expression of the zinc finger protein is controlled by a tissue-specific promoter and whereby tissue-specific modulation of the target gene expression is obtained. In this way, the zinc finger protein can be expressed in any desirable plant tissue, such as calli, meristem, leave, root or organ explant in tissue culture.
Also preferably, the zinc finger protein can be expressed in a specific organelle, such as a mitochondria, a nucleus, a plastid or a vacuole.
Exemplary plastids include chloroplast, leucoplast, aravloplast and chromoplast.

When the'zinc finger protein is provided to plant cells with an expression system for a zinc finger protein, the nucleotide sequence encoding the zinc finger protein can be targeted to or stably integrated in a specific organelle. Such nucleotide sequence can be targeted to a specific organelle by any methods known in the art. For example, the zinc finger protein can be targeted to plastid via a plastid transit peptide, to chloroplast via a chloroplast transit peptide, to mitochondria) via a mitochondria) target peptide or to nucleus via a nuclear targeting peptide.
When the zinc finger protein is provided to plant cells with an expression system for a zinc finger protein, the nucleotide sequence encoding the zinc finger protein can comprise preferred codons of the host plant, e.g., preferred translational start codon of the host plant.
In another aspect, the present invention is directed to a method of modulating a level of a compound in a plant cell, which method comprises expressing in a plant cell a zinc forger protein that binds or specifically binds to a target nucleotide sequence within a target gene to modulate expression of said target gene which is involved in a compound's metabolism in said plant cell, whereby the level of said compound in said plant cell is modulated. The level of any compound, e.g., phytic acid, or target gene, e.g., AP3, can be modulated by the present method in plant cells.
Preferably, the zinc finger protein used in the present methods is not any zinc finger protein that is disclosed in U.S. Patent No. 6,140,466 or WO 98/54311, e.g., a zinc finger-nucleotide binding polypeptide variant comprising at least three zinc finger modules that bind to a target cellular nucleotide sequence and modulate the transcriptional function of the cellular nucleotide sequence, wherein the amino acid sequence of each zinc finger module that binds a target cellular nucleotide comprises two cysteines and two histidines whereby both cysteines are amino proximal to both histidines and Wherein each of three modules of said variant has at least one amino acid sequence modification.
In still another aspect, the present invention is directed to an expression vector comprising a nucleotide sequence encoding a zinc finger protein, said zinc finger protein is capable of binding or specifically binding to a target nucleotide sequence, or a complementary strand thereof, within a target gene whose expression is to be modulated by said zinc finger protein. A plant that is regenerated from a plant transformed with the above expression vector is also provided. Preferably, the zinc finger protein used in the present expression vectors is not any zinc finger protein that is disclosed in U.S. Patent No. 6,140,466 or WO 98/54311, e.g., a zinc finger-nucleotide binding polypeptide variant comprising at least three zinc finger modules that bind to a target cellular nucleotide sequence and modulate the transcriptional function of the cellular nucleotide sequence, wherein the amino acid sequence of each zinc finger module that binds a target cellular nucleotide comprises two cysteines and two histidines whereby both cysteines are amino proximal to both histidines and wherein each of three modules of said variant has at least one amino acid sequence modification.
In yet another aspect, the present invention is directed to a genetically modified plant cell, which cell comprises an expression system for a zinc finger protein, said zinc forger protein is capable of binding, preferably specifically binding, to a target nucleotide sequence, or a complementary strand thereof, within a target gene whose expression is to be modulated by said zinc finger protein.
In yet another aspect, the present invention is directed to a zinc finger protein that is ZFPml, ZFPm2, ZFPm3, ZFPm4, or ZFPAp3, preferably in combination with positive and negative regulating domains, or a fusion protein comprises a zinc forger of 2C7 and an effector domain of SID (2C7-S~ fusion protein).
In yet another aspect, the present invention is directed to an isolated nucleic acid fragment comprising a sequence of nucleotides encoding ZFPml, ZFPm2, ZFPm3, ZFPm4, ZFPAp3 or a 2C7-SID fusion protein. Plasmids and cells containing the nucleic acid fragments and methods for producing ZFPml, ZFPm2, ZFPm3, ZFPm4, ZFPAp3 proteins or 2C7-SID fusion proteins using the plasmids and cells are also provided.
In yet another aspect, the present invention is directed to an antibody that specifically binds to the above-described zinc finger protein.
In yet another aspect, the present invention is directed to a zinc finger protein comprising a zinc finger nucleic acid binding domain and an effector domain, wherein said effector domain comprises an active domain of a restriction enzyme, an active domain of a nucleic acid modifying protein, e.g., a nucleic acid methylase, a label or a modification.
In yet~another aspect, the present invention is directed to an assay method for determining a suitable position in a gene for regulation of expression in plant cells, which method comprises: a) providing a target gene which contains a nucleotide sequence encoding a reporter protein within the coding region of said target gene and a target nucleotide sequence at a predetermined location within said target gene; b) contacting said target gene with a regulatory factor comprising a zinc finger protein specific for said target nucleotide sequence; and c) assessing the level of expression of said reporter gene in the presence and absence of said contacting; wherein a change in the level of expression of said reporter gene in the presence as opposed to the absence of said contacting identifies said position of said target nucleotide sequence as a position suitable for controlling expression of said target gene in plant cells.
Preferably, the zinc finger protein used in the present assay methods is not any zinc finger protein that is disclosed in U.S. Patent No. 6,140,466 or WO
98/54311, e.g., a zinc finger-nucleotide binding polypeptide variant comprising at least three zinc finger modules that bind to a target cellular nucleotide sequence and modulate the transcriptional function of the cellular nucleotide sequence, wherein the amino acid sequence of each zinc finger module that binds a target cellular nucleotide comprises two cysteines and two histidines whereby both cysteines are amino proximal to both histidines and wherein each of three modules of said variant has at least one amino acid sequence modification.
Brief Description of the Drawings Fig. 1 A Constructs for analyzing activation of reporter gene in maize HE89 (F19556) cells.
Fig. 1 B Activation of reporter gene in maize HE89 (F19556) cells. Maize Ubiquitin promoter was used to drive the expression of 2C7-VP64 fusion protein (activator). Reporter I (p5'C7F) is generally activated 150 to 250 fold. Reporter II activation is between 40 to 70 fold.
Fig. 2 Reporter plasmids for evaluating different binding site positions.
~pAluc: Full length CsVMV; p5'C7A: Full length CsVMV with 6x2c7 binding sites at the 5' end; p3'C7A: Full length CsVMV with 6x2c7 binding sites at the 3' end of the 5'UTR; pC7bE: CsVMV
with 6x2c7 binding sites replacing -112 to -63; p5'C7C: CsVMV (-222 to +72) with 6x2c7 binding sites at the 5' end; p5'C7D: CsVMV
(-178 to +72) with 6x2c7 binding sites at the 5' end; p5'C7F:
CsVMV (-112 to +72) with 6x2c7 binding sites at the 5' end;

pc7rbTATA: Minimal promoter with a TATA box with 6x2c7 binding sites at the 5' end; prbTATA: Minimal promoter with a TATA box with no ZFP binding sites.
Fig. 3 Zinc finger protein-effector fusion constructs for evaluation of the position effect of ZFP binding site in tobacco cells. ZC7-SID: 6 finger ZFP fused to the Sin3 interaction domain (SDJ); 2C7-SKD: 6 finger ZFP fused to the super krab domain (SKD); 2C7-VP64: 6 finger ZFP fused to 4 repeats of the minimal VP 16 activation domain (VP64); C7-VP64: 3 finger ZFP fused to 4 repeats of the minimal VP16 activation domain (VP64); C7-GFP: 3 finger ZFP
fused to the Green Fluorescent Protein (GFP); GFP: Free GFP.
Fig. 4 A Effects of zinc finger protein binding site on reporter gene expression levels and activation and activation of reporter gene with 3-( blue) or 6-zinc (cross hatch) finger proteins. A) Effects of different binding site positions on activation by a 3-finger effector (c7-vp64, solid bars) or a 6-finger protein (2c7-vp64, striped bars).
Fig. 4 B Activation of a minimal TATA containing reporter.
Fig. 5 Repression of reporters with 3- or 6-finger zinc finger fusion proteins.
Fig. 6 DNA recognition helix sequences of ZFPml, ZFPm2, ZFPm3, ZFPm4, and ZFPAp3.
Fig. 7 ELISA analysis of ZFPm1 DNA-binding specificity. Six-finger protein ZFPml was purified through affinity column from as MBP
fusion protein from E. coli. Specificity of binding was analyzed by measuring the binding activity in total lysates to immobilized biotinylated hairpin oligonucleotides containing the indicated 18-by targets. Assay were performed in duplicate. Binding site tested:
Ap3, m12, m34, 8 other non-target oligos (a-h) and no oligo control Fig. 8 ELISA analysis of ZFPm2 DNA-binding specificity. Six-finger protein ZFPm2 was purified through affinity column from as MBP
fusion protein from E. coli. Specificity of binding was analyzed by measuring the binding activity in total lysates to immobilized biotinylated hairpin oligonucleotides containing the indicated 18-by targets. Assay were performed in duplicate. Binding site tested:
Ap3, m12, m34, 8 other non-target oligos (a-h) and no oligo control Fig. 9 ELISA analysis of ZFPm3 DNA-binding specificity. Six-finger protein ZFPm3 was purified through affinity column from as MBP
fusion protein from E. coli. Specificity of binding was analyzed by measuring the binding activity in total lysates to immobilized biotinylated hairpin oligonucleotides containing the indicated 18-by targets. Assay were performed in duplicate. Binding site tested:
Ap3, m12, m34, 8 other non-target oligos (a-h) and no oligo control Fig. 10 ELISA analysis of ZFPm4 DNA-binding specificity. Six-forger protein ZFPm4 was purified through affinity column from as MBP
fusion protein from E. coli. Specificity of binding was analyzed by measuring the binding activity in total lysates to immobilized biotinylated hairpin oligonucleotides containing the indicated 18-by targets. Assay were performed in duplicate. Binding site tested:
Ap3, m12, m34, 8 other non-target oligos (a-h) and no oligo control Fig. 11 ELISA analysis of ZFPmAp3 DNA-binding specificity. Six-finger protein ZFPAp3 was purified through affinity column from as MBP
fusion protein from E. coli. Specificity of binding was analyzed by measuring the binding activity in total lysates to immobilized biotinylated hairpin oligonucleotides containing the indicated 18-by targets. Assay were performed in duplicate. Binding site tested:
Ap3, m12, m34, 8 other non-target oligos (a-h) and no oligo control Fig. 12 Gel Shift analysis of ZFPml DNA-binding affinity. Six-finger protein ZFPml was purified through affinity column from as MBP
fusion protein from E. coli. A. Affinity of binding was determined from a gel shift analysis of the binding of labeled m12 oligo to decreasing concentrations of the purified ZFPml protein. B. The affinity of ZFPml was calculated to be approximately 2nM from the concentration where one half of the labeled oligo is bound to the ZFPml protein.
Fig. 13 Gel Shift analysis of ZFPm2 DNA-binding affinity. Six-finger protein ZFPm2 was purified through affinity column from as MBP
fusion protein from E. coli. Affinity of binding was determined from a gel shift analysis of the binding of labeled m12 oligoTto decreasing concentrations of the purified ZFPm2 protein. The affinity of ZFPm2 was calculated to be approximately 7.SnM from the concentration where one half of the labeled oligo is bound to the ZFPm2 protein.
Fig. 14 Gel Shift analysis of ZFPm3 DNA-binding affinity. Six-finger protein ZFPm3 was purified through affinity column from as MBP
fusion protein from E. coli. Affinity of binding was determined from a gel shift analysis of the binding of labeled m34 oligo to decreasing concentrations of the purified ZFPm3 protein. The affinity of ZFPm3 was calculated to be approximately 0.18nM from the concentration where one half of the labeled oligo is bound to the ZFPm3 protein.
Fig. 15 Gel Shift analysis of ZFPm4 DNA-binding affinity. Six-finger protein ZFPm4 was purified through affinity column from as MBP
fusion protein from E. coli. Affinity of binding was determined from a gel shift analysis of the binding of labeled m34 oligo to decreasing concentrations of the purified ZFPm4 protein. The affinity of ZFPm4 was calculated to be approximately 0.25nM from the concentration where one half of the labeled oligo is bound to the ZFPm4 protein.
Fig. 16 Gel Shift analysis of ZFPAp3 DNA-binding affinity. Six-finger protein ZFPAp3 was purified through affinity column from as MBP
fusion protein from E. coli. Affinity of binding was determined from a gel shift analysis of the binding of labeled Ap3 oligo to decreasing concentrations of the purified ZFPAp3 protein. The affinity of ZFPAp3 was calculated to be approximately 2.3nM from the concentration where one half of the labeled oligo is bound to the ZFPAp3 protein.

Fig. 17 In vivo characterization of newly synthesized zinc finger protein ZFPml, ZFPm2, ZFPm3, and ZFPm4 in plant reporter system. The four activation fusion constructs are: ZmUbi::ZFPml-VP64//nos, ZmUbi::ZFPm2-VP64//nos, ZmUbi::ZFPm3-VP64//nos, and ZmUbi::ZFPm4-VP64//nos. The reporter constructs are similar to reporter I in Fig. 1 except the 2C7 binding site was replaced.by MIPS gene specific ZFP binding site m12 for ZFPml and ZFPm2 and by the second MIPS gene specific ZFP binding site m34 for ZFPm3 and ZFPm4.
Fig. 18 ZFP-Effector fusion constructs for Arabidopsis transformation.
ZFP-Effector constructs for Arabidopsis transformation. A.
Transient transformation vectors: pND3011 and pND3012 are transcriptional repressors. pND3014 and pND3013 are transcriptional activators. pND0001 is control vector. B. Stable transformation vectors for Agrobacteria mediated plant transformation method.
Fig. 19 Transient activation of endogenous AP3 gene in Arabidopsis leaf cells. RT-PCR was used to detect AP3 expression in Arabidopsis leaf protoplasts transformed with a GFP (pND0001, control), m4-VP64 (pND3013, non-specific activation control), and Ap3-VP64 (pND3014, AP3 specific activation). When no RNA was used in the RT-PCR reaction, no bands were seen (-). Transient transformation of a GFP or m4-VP64 construct have no effect on AP3 expression. The AP3-specific activator Ap3-VP64 causes a clear activation of AP3 expression.
Fig. 20 Endogenous gene AP3 specifically repressed by the expression of ZFPAp3-repressor fusion protein in transgenic plant ND0052-2e.
A. PCR identification of transgene ZFPAp3 in transgenic event ND0052-2e and wt plant. B. RT-PCR evaluation of endogenous gene AP3 expression level in transgenic event ND0052-2e and wild-type (wt) plant. In plant ND0052-2e, the expression of AP3 gene is significantly repressed by the expression of ZFPAp3-repressor (ZFPAp3-SID) fusion protein (Quantitative PCR shows a 46 fold of CA 02396898 2002-07-lE
US010181' 04-04-2002 2082 14';33 FR MOF 3AN DIEGO _8S8 720 5125 TO 7564tJ27801a20014 P.0 repression). The cDNA used in RT.PGR analysis was isolated from the flowers of each plant.
Fig. 21 Quantitative analysis of endogenous gens AP3 gene expression level in transgenie ZFPAp3 plants. The expression level of AP3 gene in transgenic plant is normalized to the wild type plant (defined as scale of 1).
Repression event NA0052?e shows a 46-fold of repression and ND0052-257 shows a 5=fold of repression. Activation event ND0051 shows a 2-fold of activation. Each sample was performed on triplicate assays.
Fig: 22 ZFP-Effector fusion constructs for maize transformation. A. MMapS gene specific activation constructs. B. MIPS gene speci5e repression constructs.
Fig. 23 Transient activation of endogenous MIPS gene in maize cells.
Quantitative PCR was used to detect NITPS expression in maize cells transformed with a GFP (control), ml-VP64 (pND3015), m2 VP64 (p~1173023), m3 VP64 (pND302A), and m4-VP64 (pND301~. The activator ZFPmA-VP64 causes activation of MIfS expression.
Fig_ 24 Nucleotide sequences of various nucleic acids and oligonucleotides disclosed in the present application.
),Modes for Carryinp Out the TiZvegltion A. Aefinit3ons Unless defined otherwise, all technical and scientific teams used herein have the~same meaning as is commonly understood by one of ordinary skill in the art to which this invontioa belongs. All patents, applications, published applications and. other publications and sequences from GenBank and other databases referrod to herein are incorporated by refece~ce in their entirety. If a definition set forth us this section is contrary to or otherwise incons;stcnt with a definition set forth in applications, published applications and other publications and sequences from GenBank and other data bases that are herein incorporated by reference, the definition set forth in this section prevails over the definition that is incorporated herein by reference.
As used herein, "a" or "an" means "at least one" or "one or mon "
IS
Eepfanasteii 4.Aa~. n~
AMENDED SHEET

As used herein, "zinc finger protein, (zinc forger polypeptide, or ZFP)"
refers to a polypeptide having nucleic acid, e.g., DNA, binding domains that are stabilized by zinc. The individual DNA binding domains are typically referred to as "fingers,"
such that a zinc forger protein or polypeptide has at least one forger, more typically two fingers, more preferably three fingers, or even more preferably four or five fingers, to at least six or more forgers. Each finger binds from two to four base pairs of DNA, typically three or four base pairs of DNA. A ZFP binds to a nucleic acid sequence called a target nucleic acid sequence. Each forger usually comprises an approximately 30 amino acids, zinc-chelating, DNA-binding subdomain. An exemplary motif of one class, the Cys2-His2 class (C2H2 motif), is -CYS-(X)2-4-CYS-(X)12-HiS-(X)3-5-His, where X is any amino acid, and a single zinc finger of this class consists of an alpha helix containing the two invariant histidine residues and the two cysteine residues of a single beta turn that binds a zinc cation (see, e.g., Berg et al., Science, 271:1081-1085 (1996)). A zinc finger protein can have at least two DNA-binding domains, one of which is a zinc finger polypeptide, linked to the other domain via a flexible linker. The two domains can be identical or different.
Both domains can be zinc finger proteins, either identical or different zinc finger proteins.
Alternatively, one domain can be a non-zinc finger DNA-binding protein, such as one from a transcription factor.
As used herein, "framework (or backbone) derived from a naturally occurring zinc finger protein" means that the protein or peptide sequence within the naturally occurring zinc finger protein that is involved in non-sequence specific binding with a target nucleotide sequence is not substantially changed from its natural sequence. For example, such framework (or backbone) derived from the naturally occurnng zinc finger protein maintains at least 50%, and preferably, 60%, 70%, 80%, 90%, 95%, 99% or 100% identity compared to its natural sequence in the non-sequence specific binding region. Alternatively, the nucleic acid encoding such framework (or backbone) derived from the naturally occurring zinc finger protein can be hybridizable with the nucleic acid encoding the naturally occurnng zinc finger protein, either entirely or within the non-sequence specific binding region, under low, medium or high stringency condition. Preferably, the nucleic acid encoding such framework (or backbone) derived from the naturally occurnng zinc finger protein is hybridizable with the nucleic acid encoding the naturally occurnng zinc finger protein, either entirely or within the non-sequence specific binding region, under high stringency condition.
As used herein, "gene" refers to a nucleic acid molecule or portion thereof which comprises a coding sequence, optionally containing introns, and control regions which regulate the expression of the coding sequence and the transcription of untranslated portions of the transcript. Thus, the term "gene" includes, besides coding sequence, regulatory sequence such as the promoter, enhancer, 5' untranslated regions, 3' untranslated region, termination signals, poly adenylation region and the like. Regulatory sequence of a gene may be located proximal to, within, or distal to the coding region.
As used herein, "taxget gene" refers to a gene whose expression is to be modulated by a zinc finger protein in plant cells.
As used herein, "plant" refers to any of various photosynthetic, eucaryotic mufti-cellular organisms of the kingdom Plantae, characteristically producing embryos, containing chloroplasts, having cellulose cell walls and lacking locomotion.
As used herein, "plant" includes any plant or part of a plant at any stage of development, including seeds, suspension cultures, embryos, meristematic regions, callus tissue, leaves, roots, shoots, gametophytes, sporophytes, pollen, microspores, and progeny thereof. Also included are cuttings, and cell or tissue cultures.
As used in conjunction with the present invention, the term "plant tissue" includes, but is not limited to, whole plants, plant cells, plant organs, e.g., leafs, stems, roots, menstems, plant seeds, protoplasts, callus, cell cultures, and any groups of plant cells organized into structural and/or functional units.
As used herein, "modulate the expression of a target gene in plant cells"
refers to increasing (activation) or decreasing (repression) the expression of the target gene in plant cells with a zinc forger protein, alone or in combination with other transcription andlor translational regulatory factors, or nucleic acids encoding such zinc finger protein, in plant cells.
As used herein, "providing plant cells with a zinc finger protein" refers to the provisional to the plant cells, whether in culture or in whole plant, functional zinc finger protein that is capable of modulating a target gene in the plant cells.
The functional zinc finger protein can be provided, i.e., delivered, to the plant cells by any means. For example, the zinc finger protein can be delivered directly into the plant cells. Alternatively and preferably, nucleic acids, e.g., DNA or mRNA, encoding such zinc finger protein can be delivered into the plant cells and the plant cells are maintained under the conditions that functional zinc finger protein can be produced within the plant cells.
As used herein, a "target nucleotide sequence" refers to a portion of double-stranded polynucleotide acid, e.g., RNA, DNA, PNA (peptide nucleic acid) or combinations thereof, to which it is advantageous to bind a protein. In one ~~
embodiment, a "target nucleotide sequence" is all or part of a transcriptional control element for a gene for which a desired phenotypic result can be attained by altering the degree of its expression. A transcriptional control element includes positive and negative control elements such as a promoter, an enhancer, other response elements, e.g., steroid response element, heat shock response element, metal response element, a repressor binding site, operator, and/or a silencer. The transcriptional control element can be viral, eukaryotic, or prokaryotic. A "target nucleotide sequence" also includes a downstream or an upstream sequence which can bind a protein and thereby modulate, typically prevent transcription.
As used herein, "specifically binds to a target nucleotide sequence" means that the binding affinity of a zinc finger protein to a specified target nucleic acid sequence is statistically higher than the binding affinity of the same zinc forger protein to a generally comparable, but non-target nucleic acid sequence, e.g., a GNN
sequence without matching code sequence for the particular zinc finger protein.
Normally, the binding affinity of a zinc finger protein to a specified target nucleic acid sequence is at least 1.5 fold, and preferably 2 fold or 5 fold, of the binding affinity of the same zinc finger protein to a non-target nucleic acid sequence. It also refers to binding of a zinc-finger-protein-nucleic-acid-binding domain to a specified nucleic acid target sequence to a detectably greater degree, e.g., at least 1.5-fold over background, than its binding to non-target nucleic acid sequences and to the substantial exclusion of non-target nucleic acids. The zinc finger protein's Kd to each nucleotide sequence can be compared to assess the binding specificity of the zinc finger protein to a particular target nucleotide sequence.
As used herein, a "target nucleotide sequence within a target gene" refers to a functional relationship between the target nucleotide sequence and the target gene in that binding of a zinc-finger-protein to the target nucleotide sequence will modulate the expression of the target gene. The target nucleotide sequence can be physically located anywhere inside the boundaries of the target gene, e.g., 5' ends, coding region, 3' ends, upstream,and downstream regions outside of cDNA encoded region, or inside enhancer or other regulatory region, and can be proximal or distal to the target gene.
As used herein, "culturing plant cells" refers to the cultivation or growth of the plant cells. Such cultivation or growth can be in vitro, e.g., in culture medium, or in vivo, e.g., in planta.
As used herein, "endogenous" refers to nucleic acid or protein sequence naturally associated with a target gene or a host cell into which it is introduced.
As used herein, "exogenous" refers to nucleic acid or protein sequence not naturally associated with a target gene or a host cell into which it is introduced, including non-naturally occurring multiple copies of a naturally occurnng nucleic acid, e.g., DNA sequence, or naturally occurring nucleic acid sequence located in a non-naturally occurring genome location.
As used herein, "effector (or effector protein)" refer to constructs or their encoded products which are able to regulate gene expression either by activation or repression or which exert other effects on a target nucleic acid. The effector protein may include a zinc finger binding region only, but more commonly also includes a "functional domain" such as a "regulatory domain."
As used herein, "regulatory domain" refer to the portion of the effector protein or effector which enhances or represses gene expression.
As used herein, "compound" refer to any substance that is metabolized in plant cells and its metabolism can be modulated by a zinc-finger-protein in plant cells.
As used herein, "genetically modified plant (or transgenic plant)" refers to a plant which comprises within its genome an exogenous polynucleotide.
Generally, and preferably, the exogenous polynucleotide is stably integrated within the genome such that the polynucleotide is passed on to successive generations. The exogenous polynucleotide may be integrated into the genome alone or as part of a recombinant expression cassette. "Transgenic" is used herein to include any cell, cell line, callus, tissue, plant part or plant, the genotype of which has been altered by the presence of exogenous nucleic acid including those transgenics initially so altered as well as those created by sexual crosses or asexual propagation from the initial transgenic.
The term "transgenic" as used herein does not encompass the alteration of the genome (chromosomal or extra-chromosomal) by conventional plant breeding methods or by naturally occurring events such as random cross-fertilization, non-recombinant viral infection, non-recombinant bacterial transformation, non-recombinant transposition, or spontaneous mutation. -As used herein, "essential gene" refers to a gene encoding a protein that is essential to the growth or survival of the plant, e.g., a biosynthetic enzyme, receptor, signal transduction protein, structural gene product, or transport protein.
As used herein, "expression cassette" refers to a DNA sequence capable of directing expression of a particular nucleotide sequence in an appropriate host cell, comprising a promoter operably linked to the nucleotide sequence of interest which is operably linked to termination signals. It also typically comprises sequences required for proper translation of the nucleotide sequence. The coding region usually codes for a protein of interest but may also code for a functional RNA of interest, for example antisense RNA or a nontranslated RNA, in the sense or antisense direction. The expression cassette comprising the nucleotide sequence of interest may be chimeric, meaning that at least one of its components is heterologous with respect to at least one of its other components. The zinc finger-effector fusions of the present invention are chimeric. The expression cassette may also be one which is naturally occurring but has been obtained in.a recombinant form useful for heterologous expression.
Typically, however, the expression cassette is heterologous with respect to the host, i. e., the particular DNA sequence of the expression cassette does not occur naturally in the host cell and must have been introduced into the host cell or an ancestor of the host cell by a transformation event. The expression of the nucleotide sequence in the expression cassette may be under the control of a constitutive promoter or of an inducible promoter which initiates transcription only when the host cell is exposed to some particular external stimulus. In the case of a multicellular organism, such as a plant, the promoter can also be specific to a particular tissue or organ or stage of development. In the case of a plastid expression cassette, for expression of the nucleotide sequence from a plastid genome, additional elements, i.e., ribosome binding sites, may be required.
As used herein, "minimal promoter" refers to a promoter element, particularly a TATA element, that is inactive or that has greatly reduced promoter activity in the absence of upstream activation. In the presence of a suitable transcription factor, the minimal promoter functions to permit transcription.
As used herein, "significant increase" refer to an increase in gene expression, enzymatic or other biological activity, specificity or affinity, or effector or phenotypic activity, that is larger than the margin of error inherent in the measurement technique, preferably an increase by about 2-fold or greater of the activity without the ZFP or ligand inducer, more preferably an increase by about 5-fold or greater, and most preferably an increase by about 10-fold or greater.
As used herein, "repressor protein" refer to a protein that binds to operator of DNA or to RNA to prevent transcription or translation, respectively.
As used herein, "repression" refer to inhibition of transcription or translation by binding of repressor protein to specific site on DNA or mRNA. Preferably, repression includes a significant change in transcription or translation level of at least 1.5 fold, more preferably at least two fold, and even more preferably at least five fold.
As used herein, "activator protein" refer to a protein that binds to operator of DNA or to RNA to enhance transcription or translation, respectively.
As used herein, "activation" refer to enhancement of transcription or translation by binding of activator protein to specific site on DNA or mRNA.
Preferably, activation includes a significant change in transcription or translation level of at least 1.5 fold, more preferably at least two fold, and even more preferably at least five fold.
As used herein, "conservatively modified variant" refer to amino acid and nucleic acid sequences containing individual substitutions, deletions or additions that alter, add or delete a single amino acid or nucleotide or a small percentage of amino acids or nucleotides in the sequence, where the alteration results in the substitution of an amino acid with a chemically similar amino acid. Conservative substitution tables providing functionally similar amino acids are well known in the art. Such conservatively modified variants are in addition to and do not exclude polymorphic variants and alleles of the invention. The following groups each contain amino acids that are conservative substitutions for one another: 1) Alanine (A), Glycine (G); 2) Serine (S), Threonine (T); 3) Aspartic acid (D), Glutamic acid (E); 4) Asparagine (I~, Glutamine (Q); 5) Cysteine (C), Methionine (M); 6) Arginine (R), Lysine (K), Histidine (H); 7) Isoleucine (1), Leucine (L), Valine (V); and 8) Phenylalanine (F), Tyrosine (Y), Tryptophan (W). ,(see, e.g., Creighton, Proteins (1984) for a discussion of amino acid properties).
As used herein, a combination refers to any association between two or among more items.

As used herein, a composition refers to any mixture. It may be a solution, a suspension, liquid, powder, paste, aqueous, non-aqueous or any combination thereof.
As used herein, derivative or analog of a molecule refers to a portion derived from or a modified version of the molecule.
As used herein, operably linked, operatively linked or operationally associated refers to the functional relationship of DNA with regulatory and effector sequences of nucleotides, such as promoters, enhancers, transcriptional and translational stop sites, and other signal sequences. For example, operative linkage of DNA to a promoter refers to the physical and functional relationship between the DNA and the promoter such that the transcription of such DNA is initiated from the promoter by an RNA
polymerase that specifically recognizes, binds to and transcribes the DNA. To optimize expression and/or in vitro transcription, it may be helpful to remove, add or alter 5' untranslated portions of the clones to eliminate extra, potentially inappropriate alternative translation initiation (i.e., start) codons or other sequences that may interfere with or reduce expression, either at the level of transcription or translation..
Alternatively, consensus ribosome binding sites (see, e.g., I~ozak, J. Biol.
Chern., 266:19867-19870 (1991)) can be inserted immediately 5' of the start codon and may enhance expression. The desirability of (or need for) such modification may be empirically determined.
As used herein, a promoter region or promoter element refers to a segment of DNA or RNA that controls transcription of the DNA or RNA to which it is operatively linked. The promoter region includes specific sequences that are sufficient for RNA polymerase recognition, binding and transcription initiation. This portion of the promoter region is referred to as the promoter. In addition, the promoter region includes sequences that modulate this recognition, binding and transcription initiation activity of RNA polymerase. These sequences may be cis acting or may be responsive to traps acting factors. Promoters, depending upon the nature of the regulation, may be constitutive or regulated.
As used herein: stringency of hybridization in determining percentage mismatch is as follows: (1) high stringency: 0.1 x SSPE, 0.1% SDS, 65°C; (2) medium stringency: 0.2 x SSPE, 0.1% SDS, 50°C; and (3) low stringency:
1.0 x SSPE, 0.1% SDS, SO°C. Equivalent stringencies may be achieved using alternative buffers, salts and temperatures. The term substantially identical or homologous or similar varies with the context as understood by those skilled in the relevant art and generally means at least 70%, preferably means at least 80%, more preferably at least 90%, and most preferably at least 95% identity.
As used herein, substantially identical to a product means sufficiently similar so that the property of interest is sufficiently unchanged so that the substantially identical product can be used in place of the product.
As used herein, isolated means that a substance is either present in a preparation at a concentration higher than that substance is found in nature or in its naturally occurring state or that the substance is present in a preparation that contains other materials with which the substance is not associated with in nature.
As used herein, substantially pure means sufficiently homogeneous to appear free of readily detectable impurities as determined by standard methods of analysis, such as thin layer chromatography (TLC), gel electrophoresis and high performance liquid chromatography (HPLC), used by those of skill in the art to assess such purity, or sufficiently pure such that further purification would not detestably alter the physical and chemical properties, such as enzymatic and biological activities, of the substance. Methods for purification of the compounds to produce substantially chemically pure compounds are known to those of skill in the art. A
substantially chemically pure compound may, however, be a mixture of stereoisomers or isomers.
In such instances, further purification might increase the specific activity of the compound.
As used herein, vector or plasmid refers to discrete elements that are used to introduce heterologous DNA into cells for either expression or replication thereof.
Selection and use of such vehicles are well known within the skill of the artisan. An expression vector includes vectors capable of expressing DNAs that are operatively linked with regulatory sequences, such as promoter regions, that are capable of effecting expression of such DNA fragments. Thus, an expression vector refers to a recombinant DNA or RNA construct, such as a plasmid, a phage, recombinant virus or other vector that, upon introduction into an appropriate host cell, results in expression of the cloned DNA. Appropriate expression vectors are well known to those of skill in the art and include those that are replicable in eukaryotic cells and/or prokaryotic cells and those that remain episomal or those which integrate into the host cell genome.

As used herein, "macromolecule" refers to a molecule that, without attaching to another molecule, is capable of generating an antibody that specifically binds to the macromolecule.
As used herein, "small molecule" refers to a molecule that, without forming homo-aggregates or without attaching to a macromolecule or adjuvant, is incapable of generating an antibody that specifically binds to the small molecule.
Preferably, the small molecule has a molecular weight that is about or less than 10,000 daltons. More preferably, the small molecule has a molecular weight that is about or less than 5,000 dalton.
As used herein, "vitamin" refers to a trace organic substance required in certain biological species. Most vitamins function as components of certain coenzymes.
As used herein, "lipid" refers to water-insoluble, oily or greasy organic substances that are extractable from cells and tissues by nonpolar solvents, such as chloroform or ether.
As used herein, a "receptor" refers to a molecule that has an affinity for a given ligand. Receptors may be naturally-occurring or synthetic molecules.
Receptors may also be referred to in the art as anti-ligands. As used herein, the receptor and anti-ligand are interchangeable. Receptors can be used in their unaltered state or as aggregates with other species. Receptors may be attached, covalently or noncovalently, or in physical contact with, to a binding member, either directly or indirectly via a specific binding substance or linker. Examples of receptors, include, but are not limited to: antibodies, cell membrane receptors surface receptors and internalizing receptors, monoclonal antibodies and antisera reactive with specific antigenic determinants such as on viruses, cells, or other materials, drugs, polynucleotides, nucleic acids, peptides, cofactors, lectins, sugars, polysaccharides, cells, cellular membranes, and organelles.
As used herein, "nutrient or storage protein" refers to a protein that is used by the cell as the nutrient source or storage form for such nutrient. Non-limiting examples of nutrient or storage proteins include gliadin, ovalbumin, casein, and ferritin.
As used herein, "contractile or motile protein" refers to a protein that endows cells and organisms with the ability to contract, to change shape, or to move about.

' Non-limiting examples of contractile or motile proteins include actin, myosin, tubulin and dynein.
As used herein, "structural protein" refers to a protein that serves as supporting filaments, cables, or sheets to give biological structures strength or protection. Non-limiting examples of structural proteins include keratin, fibroin, collagen, elastin and proteoglycans.
As used herein, "defense protein" refers to a protein that defends organisms against invasion by other species or protect them from injury. Non-limiting examples of defense proteins include antibodies, fibrinogen, thrombin, botulinus toxin, diphtheria toxin, snake venoms and ricin.
As used herein, "regulatory protein" refers to a protein that helps regulate cellular or physiological activity. Non-limiting examples of regulatory proteins include insulin, growth hormones, corticotropin and repressors.
For clarity of disclosure, and not by way of limitation, the detailed description of the invention is divided into the subsections that follow.
B. Methods for modulating gene expression in plant In one aspect, the present invention is directed to a method to modulate the expression of a target gene in plant cells, which method comprises providing plant cells with a zinc finger protein, said zinc finger protein being capable of binding or specifically binding to a target nucleotide sequence, or a complementary strand thereof, within a target gene, and allowing said zinc finger protein binding to said target nucleotide sequence, whereby the expression of said target gene in said plant cells is modulated.
Plant The present invention can be used over a broad range of plant types, 1 preferably the class of higher plants amenable to transformation techniques, particularly monocots and dicots. Particularly preferred are monocots such as the species of the Family Grarriineae including Sorghum bicolor and Zea mays. The present method can also be used in species from the following genera:
Cucurbita, Rosa, Vitis, Juglans, Fragaria, Lotus, Medicago, Onobrychis, Trifolium, Trigonella, Vigna, Citrus, Linum, Geranium, Manihot, Daucus, Arabidopsis, Brassica, Raphanus, Sinapis, Atropa, Capsicum, Datura, Hyoscyamus, Lycopersicon, Nicotiana, Solanum, Petunia, Digitalis, Majorana, Ciahorium, Helianthus, Lactuca, Bromus, Asparagus, Antirrhinum, Heterocallis, Nemesis, Pelargonium, Panieum, Pennisetum, Ranunculus, Senecio, Salpiglossis, Cucumis, Browaalia, Glycine, Pisum, Phaseolus, Lolium, Oryza, Avena, Hordeum, Secale, and Triticum.
Preferred plant cells include those from corn (Zea mays), canola (Brassica napus, Brassica raga ssp.), particularly those Brassica species useful as sources of seed oil, alfalfa (Medicago sativa), rice (Oryza sativa), rye (Secale cereale), sorghum (Sorghum bicolor, Sorghum vulgare), millet (e.g., pearl millet (Pennisetum glaucum), proso millet (Panicum nailiaceum), foxtail millet (Setaria italics), finger millet (Eleusine coracarta), sunflower (Helianthus annuus), safflower (Carthamus tinctorius), wheat (Triticum aestivum), duckweed (Lemma) soybean (Glycine max), tobacco (Nicotiana tabacum), potato (Solanum tuberosum), peanut (Arachis hypogaea), cotton (Gossypium barbadense, Gossypium hirsutum), sweet potato (Qpomoea batatus), cassava (Manihot esculenta), coffee (Cqfea spp.), coconut (Cocos nucijra), pineapple (Ananas comosus), citrus tree (Citrus spp.), cocoa (Theobroma cacao), tea (Camellia sinensis), banana (Muss spp.), avocado (Persea americana), fig (Ficus casica), guava (Psidium guajava), mango (Mangifera indica), olive (Olea europaea), papaya (Carica papaya), cashew (Anacardium occidentale), macadamia (Macadamia integr~fblia), almond (Prunus amygdalus), sugar beet (Beta vulgaris), sugarcane (Saccharum spp.), oats, barley, vegetables, ornamentals, and conifers.
Preferred vegetables include tomatoes (Lycopersicon esculentum), lettuce (e.g., Lactuca sativa), green beans (Phaseolus vulgaris), lima beans (Phaseolus limensis), peas (Lathyrus spp.), and members of the genus Cucumis such as cucumber (C. sativus), cantaloupe (C cantalupensis), and musk melon (C. melo).
Preferred ornamentals include azalea (Rhododendrom spp.), hydrangea (Macrophylla hydrangea), hibiscus (Hibiscus rosasanensis), roses (Ross spp.), tulips (Tulips .spp.), daffodils (Narcissus spp.), petunias (Petunia hybrids), carnation (Dianthus caryophyllus), poinsettia (Euphorbiapulcherrima), and chrysanthemum.
Conifers that may be employed in practicing the present method include, for example, pines such as loblolly pine (Pinus taeda), slash pine (Pinus elliotii), ponderosa pine (Pinus ponderosa), lodgepole pine (Pinus contorts), and Monterey pine (Pinus radiata); Douglas-fir (Pseudotsuga menziesii); Western hemlock (Isuga canadensis); Sitka spruce (Picea glauoa); redwood (Sequoia sempervirens); true firs such as silver fix (Abies amabilis) and balsam fir (Abies balsamea); and cedars such as Western red cedar (Thuja plicata) and Alaska yellow-cedar (Chamaecyparis nootkatensis).
Leguminous plants include beans and peas. Beans include guar, locust bean, fenugreek, soybean, garden bean, cowpea, mungbean, lima bean, fava bean, lentils, chickpea, etc. Legumes include, but are not limited to, Arachis, e.g., peanuts, Vicia, e.g., crown vetch, hairy vetch, adzuki bean, mung bean, and chickpea, Lupinus, e.g., lupine, trifolium, Phaseolus, e.g., common bean and lima bean, Pisum, e.g., field bean, Melilotus, e.g., clover, Medicago, e.g., alfalfa, Lotus, e.g., trefoil, lens, e.g., lentil, and false indigo. Preferred forage and turf grass for use in the methods of the invention include alfalfa, orchard grass, tall fescue, perennial ryegrass, creeping bent grass, and redtop.
More preferably, plants that can be modulated by the present method are crop plants and model plant, e.g., corn, rice, alfalfa, sunflower, canola, soybean, cotton, peanut, sorghtun, wheat, tobacco, lemma, etc. Most preferably, gene expression in corn, rice, lemma, and soybean plants are modulated by the present method.
Zinc finer proteins used in the present method Any zinc finger proteins can be used in the present method. In addition, zinc finger proteins disclosed or designed and predicted according to the procedures in " WO 98/54311 can be used. WO 98154311 discloses technology which allows the design of zinc finger protein domains which bind specific nucleotide sequences which are unique to a target gene. It has been calculated that a sequence comprising nucleotides is sufficient to specify an unique location in the genome of higher organisms. Typically, therefore, the zinc forger protein domains are hexadactyl, i.e., contain 6 zinc forgers, each with its specifically designed alpha helix for interaction with a particular triplet. However, in some instances, a shorter or longer nucleotide target sequence may be desirable. Thus, the zinc finger domains in the proteins may contain from 2-12 fingers, preferably, 3-8 fingers, more preferably 5-7 fingers, and most preferably 6 fingers.
When a mufti-finger protein binds to a polynucleotide duplex, e.g., DNA, RNA, PNA or any hybrids thereof, its forgers typically line up along the polynucleotide duplex with a periodicity of about one finger per 3 bases of nucleotide sequence. The binding sites of individual zinc fingers (or subsites) typically span three to four bases, and subsites of adjacent fingers usually overlap by one base.

Accordingly, a three-finger zinc finger protein XYZ binds~to the 10 base pair site abcdefghij (where these letters indicate one of the duplex DNA) with the subsits of finger X being ghij, finger Y being defg and finger Z being abcd. For example, as known in the art, to design a three-finger zinc finger protein to bind to the targeted 10 base site abcdefXX~~X (wherein each "X" represents a base that would be specified in a particular application), zinc forgers Y and Z would have the same polypeptide sequence as found in the original zinc finger discussed above (perhaps a wild type zinc fingers which bind defg and abcd, respectively). Finger X would have a mutated polypeptide sequence. Preferably, finger X would have mutations at one or more of the base-contacting positions, z.e., finger X would have the same polypeptide sequence as a wild type zinc finger except that at least one of the four amino residues at the primary positions would differ. Similarly, to design a three-finger zinc protein that would bind to a 10 base sequence abcXXXXhij (wherein each "X" is base that would be specified in a particular application), fingers X and Z have the same sequence as the wild type zinc fingers which bind ghij and abcd, respectively, while finger Y would have residues at one or more base-coating positions which differ from those in a wild type finger. The present method can employ mufti-fingered proteins in which more than one finger differs from a wild type zinc finger. The present method can also employ mufti-fingered protein in which the amino acid sequence in all the fingers have been changed, including those designed by combinatorial chemistry or other protein design and binding assays.
It is also possible to design or select a zinc finger protein to bind to a targeted polynucleotide in which more than four bases have been altered. ~In this case, more than one finger of the binding protein must be altered. For example, in the 10 base sequence XXXdefgXXX, a three-finger binding protein could be designed in which fingers X and Z differ from the corresponding fingers in a wild type zinc finger, while finger Y will have the same polypeptide sequence as the corresponding finger in the wild type fingers which binds to the subsite defg. Binding proteins having more than three fingers can also be designed for base sequences of longer length. For example, a four finger-protein will optimally bind to a 13 base sequence, while a five-finger protein will optimally bind to a 16 base sequence. A mufti-finger protein can also be designed in which some of the fingers are not involved in binding to the selected DNA. Slight variations are also possible in the spacing of the fingers and framework.

While the present method can employ any valid recognition code, the zing finger protein nucleic acid binding domain/nucleoside binding partner pairs disclosed in United States Patent No. 5,789,538, WO 96/06166, and WO 00/23464 can also be used.
S Methods for designing and identifying a zinc finger protein with desired nucleic acid binding characteristics also include those described in W098/53060, which reports a method for preparing a nucleic acid binding protein of the Cys2-His2 zinc finger class capable of binding to a nucleic acid quadruplet in a target nucleic acid sequence.
Zinc finger proteins useful in the present method can comprise at least one zinc finger polypeptide linked via a linker, preferably a flexible linker, to at least a second DNA binding domain, which optionally is a second zinc finger polypeptide.
The zinc finger protein may contain more than two DNA-binding domains, as well as one or more regulator domains. The zinc forger polypeptides used in the present method can be engineered to recognize a selected target site in the gene of choice.
Typically, a backbone from any suitable C2H2-ZFP, such as SPA, SPIC, or ZIF268, is used as the scaffold for the engineered zinc finger polypeptides (see, e.g., Jacobs, EMBO J. (1992) 11:4507 ; and Desjarlais & Berg, Proc. Natl. Acad. Sci. USA
(1993) 90:2256-2260). A number of methods can then be used to design and select a zinc finger polypeptide with high affinity for its target. A zinc finger polypeptide can be designed or selected to bind to any suitable target site in the target gene, with high affinity.
Any suitable method known in the art can be used to design and construct nucleic acids encoding zinc finger polypeptides, e.g., phage display, random mutagenesis, combinatorial libraries, computer/rational design, affinity selection, PCR, cloning from cDNA or genomic libraries, synthetic construction and the like.
(see, e.g., U.S. Pat. No. 5,786,538; Wu et al., Proc. Natl. Acad. Sci. USA
(1995) 1 92:344-348; Jamieson et al., Biochemistl. (1994) 33:5689-5695; Rebar & Pabo, Science (1994) 263:671-673; Choo & Klug, Proc. Natl. Acad. Sci. USA (1994) 91:
11168-11172; Pomerantz et al., Science, 267:93-96 (1995); Pomerantz et al., Proc.
Natl. Acad. Sci. USA (1995) 92:9752-9756; Liu et al., Proc. Natl. Acad. Sci.
USA
(1997) 94:5525-5530; and Desjarlais & Berg,(1994) Proc. Natl. Acad. Sci. USA
91:11-99-11103).

Zinc finger proteins useful in the method can be made by any recombinant DNA technology method for gene construction. For example, PCR based construction can be used. Ligation of desired fragments can also be performed, using linkers or appropriately complementary restriction sites. One can also synthesize entire finger domain or parts thereof by any protein synthesis method.
Preferred for cost and flexibility is the use of PCR primers that encode a finger sequence or part thereof with known base pair specificity, and that can be reused or recombined to create new combinations of fingers and ZFP sequences The amino acid linker should be flexible, a beta turn structure is preferred, to allow each three finger domain to independently bind to its target sequence and avoid steric hindrance of each other's binding. Linkers can be designed and empirically tested.
If recognition code is incomplete, or if desired, in one embodiment, the ZFP
can be designed to bind to non-contiguous target sequences. For example, a target 1 S sequence for a six-finger ZFP can be a nine base pair sequence (recognized by three fingers) with intervening bases (that do not contact the zinc finger nucleic acid binding domain) between a second nine base pair sequence (recognized by a second set of three fingers). The number of intervening bases can vary, such that one can compensate for this intervening distance with an appropriately designed amino acid linker between the two three-finger parts of ZFP. A range of intervening nucleic acid bases in a target binding site is preferably 20 or less bases, more preferably 10 or less, and even more preferably 6 or less bases. It is of course recognized that the linker must maintain the reading frame between the linked parts of ZFP protein.
A minimum length of a linker is the length that would allow the two zinc finger domains to be connected without providing steric hindrance to the domains or the linker. A linker that provides more than the minimum length is a "flexible linker."
Determining the length of minimum linkers and flexible linkers can be performed using physical or computer models of DNA-binding proteins bound to their respective target sites as are known in the art.
The six-finger zinc finger peptides can use a conventional "TGEKP" linker to connect two three-finger zinc finger peptides or to add additional forgers to a three-finger protein. Other zinc finger peptide linkers, both natural and synthetic, are also suitable.

A useful zinc finger framework is that of ZIF268 (see WO00/23464 and references cited therein.), however, others are suitable. Examples of known zinc finger nucleotide binding polypeptides that can be truncated, expanded, and/or mutagenized in order to change the function of a nucleotide sequence containing a zinc forger nucleotide binding motif includes TFIIIA and zifz68. Other zinc finger nucleotide binding proteins are known to those of skill in the art. The marine HiS2 zinc finger protein Zif268 is structurally well characterized of the zinc finger proteins (Pavletich and Pabo, Science (1991) 252:809-817; Elrod-Erickson et al., Structure (London) (1996) 4:1171-1180; and Swirnoff et al., Mol. Cell. Biol.
(1995) 15 :2275-2287). DNA recognition in each of the three zinc finger domains of this protein is mediated by residues in the N-terminus of the alpha-helix contacting primarily three nucleotides on a single strand.of the DNA. The operator binding site for this three finger protein is 5'-GCGTGGGCG-'3. Structural studies of Zif268 and other related zinc forger-DNA complexes (Elrod-Erickson et al., Structure (London) (1998) 6:451-464; Kim and Berg, Nature Structural Biology (1996) 3:940-945;
Pavletich and Pabo, Science (1993) 261:1701-1707; Houbaviy et al., Proc. Natl.
Acad. Sci. USA (1996) 93:13577-13582; Fairall et al., Nature (London) (1993) 366:483-487; Wuttke et al., J. Mol. Biol. (1997) 273:183-206; Nolte et al., Proc. Natl.
Acad. Sci. USA (1998) 95:2938-2943; and Narayan et al., J. Biol. Chem. (1997) 272:7801-7809) have shown that residues from primarily three positions on the a-helix, -1, 3, and 6, are involved in specific base contacts. Typically, the residue at position -1 of the a-helix contacts the 3' base of that finger's subsite while positions 3 and 6 contact the middle base and the 5' base, respectively.
However, it should be noted that at least in some cases, zinc finger domains appear to specify overlapping 4 by sites rather than individual 3 by sites. In Zif268, residues in addition to those found at helix positions -1, 3, and 6 are involved in contacting DNA (Elrod-Erickson et al., Structure (1996) 4:1171-1180).
Specifically, an aspartate in helix position 2 of the middle finger plays several roles in recognition and makes a variety of contacts. The carboxylate of the aspartate side chain hydrogen bonds with arginine at position -1, stabilizing its interaction with the 3'-guanine of its target site. This aspartate may also participate in water-mediated contacts with the guanine's complementary cytosine. In addition, this carboxylate is observed to make a direct contact to the N4 of the cytosine base on the opposite strand of the 5'-guanine base of the finger 1 binding site. It is this interaction which is the chemical basis for target site overlap. -Any suitable method of protein purification known to those of skill in the art can be used to purify the zinc finger proteins of the invention (see Sambrook et al., Molecular Clonin~~ A Laboratory Manual (2d ed., Cold Spring Harbor Laboratory Press, Plainview, New York) (1989)). In addition, any suitable host can be used, e.g., bacterial cells, insect cells, yeast cells, mammalian cells, and the like.
In one embodiment, expression of the zinc forger protein fused to a maltose binding protein (MBP-zinc finger protein) in bacterial strain JM 109 allows for purification through an amylose column (NEB). High expression levels of the zinc forger protein can be obtained by induction with IPTG since the MBP-zinc forger protein fusion in the pMal-c2 expression plasmid is under the control of the IPTG
inducible tac promoter (NEB). Bacteria containing the MBP-zinc finger protein fusion plasmids are inoculated in to 2xYT medium containing lO~LM ZnCl2, 0.02%
glucose, plus 50 ptg/ml ampicillin and shaken at 37°C. At mid-exponential growth IPTG is added to 0.3 mM and the cultures are allowed to shake. After 3 hours the bacteria are harvested by centrifugation, disrupted by sonication, and then insoluble material is removed by centrifugation. The MBP-zinc finger proteins are captured on an arnylose-bound resin, washed extensively with buffer containing 20 mM Tris-HCl (pH 7-5), 200 mM NaCI, 5 mM DTT and 50 ~tM ZnCl2, then eluted with maltose in essentially the same buffer (purification is based on a standard protocol from NEB).
Purified proteins are quantitated and stored for biochemical analysis.
The biochemical properties of the purified proteins, e.g., Kd, can be characterized by any suitable assay. In one embodiment, Kd is characterized via electrophoretic mobility shift assays ("EMSA") (Buratowski & Chodosh, in Current Protocols in Molecular Biology pp. 12.2.1-12.2.7 (Ausubel ed., 1996)). By "Kd"
is meant the dissociation constant for the compound, i.e., the concentration of a compound (e.g., a zinc finger protein) that gives half maximal binding of the compound to its target (i.e., half of the compound molecules are bound to the target) under given conditions (i.e., when [target] « Kd), as measured using a given assay system. Any assay system can be used, as long as it gives an accurate measurement of the actual Kd of the zinc finger protein. In one embodiment, the Kd for the zinc finger proteins of the invention is measured using an electrophoretic mobility shift assay as described herein.

As described in the Example Section below, the observed affinities of 6-finger proteins tested were determined to be: ZFPml=2.OnM, ZFPm2=7.5 nM, ZFPm3=
0.18 nM, ZFPm4=0.25 nM and ZFPAp3=2.3 nM (Fig. 12-16). Most of these affinities compare favorably with existing 6 finger proteins. The 2C7 derivative of the Spl zinc finger was reported to have a specificity of 0.46 nM and the e2c zinc finger was reported to have an affinity of 0.5 nM. Consequently, zinc forgers used in the invention typically have affinity 0.1 to 1.0 nanomolar range, more typically 0.18 to 0.75 nanomolar, and to picomoloar range and even to femtomolar range.
Typically, naturally occurring zinc fingers have Kd in the nanomolar range.
Zinc .10 forger proteins useful in the invention may even have an affinity for the target site that is in the femtamolar range, e.g., 100 femtamoles, 10 femtamoles, or less, in some cases as low as one femtamole.
In a specific embodiment, the zinc finger protein used in the present methods comprises a framework (or backbone) derived from a naturally occurring zinc forger protein. Framework (or backbone) derived from any naturally occurnng zinc finger protein can be used. For example, the zinc finger protein comprises a framework (or backbone) derived from a zinc finger protein comprising a C2H2 motif can be used.
Preferably, the protein or peptide sequence within the ~i sheet of the C2H2 motif is not substantially changed, or not changed, from its natural sequence.
' In another specific embodiment, the zinc finger protein used in the present methods comprises a framework (or backbone) derived from a zinc finger protein that is naturally functional in plant cells. For example, the zinc finger protein used in the present methods can comprise a C3H zinc finger (Terol et al., Gene, 260 1-2 :45-53 (2000)), a QALGGH motif (Takatsuji, Plant. Mol. Biol., 39 6 :1073-8 (1999)), a RING-H2 zinc finger motif (Jensen et al., FEBS Lett., 436 2 :283-7 (1998)), a amino acid C2H2 motif (Chou et al., Proc. Natl. Acad. Sci. USA, 95 9 :5293-8 (1998)), a zinc finger motif of Arabidopsis LSD1 (Dietrich et al., Cell, 88 5 :685-94 (1997)) and a zinc forger motif of BBF/Dof domain proteins (De Paolis et al., Plant J., 10 2 :215-23 (1996)).
In another specific embodiment, the zinc finger protein used in the present methods comprises a framework (or backbone) derived from a zinc finger protein that is known in the art as of January 19, 2001.
For example, the zinc finger protein used in the present methods can comprise a framework (or backbone) derived from the zinc finger protein disclosed in the following U.S. patents and PCT patent publications: U.S. Patent No. 6,160,091, 6,140,466, 6,140,081, 5,831,008, 5,811,304 and 5,789,538 and WO 00/42219, WO
00/41566, WO 00/27878, WO 00/23464, WO 00/20622, WO 00/20556, WO
99/62952, WO 99148909, WO 99/46293, WO 99/45132, WO 99/42474, WO
99/21991, WO 98/54311, WO 98/53061, WO 98/45326, WO 96/11267, WO
96106168 and WO 95/19431.
The zinc finger protein used in the present methods can also comprise a framework (or backbone) derived from the zinc finger protein disclosed in the following references: 1) Cousins RJ, Lanningham-Foster L., "Regulation of cysteine-rich intestinal protein, a zinc finger protein, by mediators of the immune response." J
Infect Dis. 2000 Sep;l82 Suppl 1:581-4; 2) Bouhouche N, Syvanen M, Kado CL, "The origin of prokaryotic C2H2 zinc finger regulators." Trends Microbiol.

Feb;B(2):77-81; 3: Klug A., "Zinc finger peptides for the regulation of gene expression." J Mol Biol. 1999 Oct 22;293(2):215-8; 4) Cook T, Gebelein B, Urrutia R., "Sp l and its likes: biochemical and functional predictions for a growing family of zinc finger transcription factors." Ann N Y Acad Sci. 1999 Jun 30;880:94-102;
5) Takatsuji H., "Zinc-finger proteins: the classical zinc forger emerges in contemporary plant science." Plant Mol Biol. 1999 Apr;39(6):1073-8; 6) Huang S., "The retinoblastoma protein-interacting zinc finger gene RIZ in 1p36-linked cancers."
Front Biosci. 1999 Jun 15;4:D528-32; 7: Krempler A, Brenig B., "Zinc finger proteins: watchdogs in muscle development." Mol Gen Genet. 1999 Mar;261 (2):209-15; 8) Hoffmann A, Ciani E, Houssami S, Brabet P, Journot L, Spengler D., "Induction of type I PACAP receptor expression by the new zinc finger protein Zac1 and p53." Ann N Y Acad Sci. 1998 Dec 11;865:49-58; 9) Kroricke KD, Kolb-Bachofen V., "Measurement of nitric oxide-mediated effects on zinc homeostasis and zinc finger transcription factors." Methods Enzymol. 1999;301:126-35; 10) Tseng H., "Basonuclin, a zinc finger protein associated with epithelial expansion and pxoliferation." Front Biosci. 1998 Sep 1;3:D985-8; 11) Takatsuji H., "Zinc-forger transcription factors in plants." Cell Mol Life Sci. 1998 Jun;54(6):582-96;
12) Chilton BS, Hewetson A., "Zinc finger proteins RUSH in where others fear to tread."
Biol Reprod. 1998 Feb;58(2):285-94; 13) Urrutia R., "Exploring the role of homeobox and zinc finger proteins in pancreatic cell proliferation, differentiation, and apoptosis." Int J Pancreatol. 1997 Aug;22(1):1-14; 14) Mizuno K, Higuchi O., "LIM domains:
double zinc finger motifs involved in protein-protein interactions"
Tanpakushitsu Kakusan Koso. 1997 Oct;42(13):2061-71; 15: Losson R., "KR.AB zinc finger proteins and nuclear receptors: a possible cross-talk." Biol Chem. 1997 Ju1;378(7):579-81; 16) Eis PS., "Fluorescence studies of zinc finger peptides and proteins." Methods Enzymol. 1997;278:330-43; 17) Tamaoki T, Hashimoto T., "ZFH/ATBF1 gene family: transcription factors containing both homeo- and zinc finger-domains"
Tanpakushitsu Kakusan Koso. 1996 Sep;41 (11):1550-9; 18: Hollemann T, Bellefroid E, Stick R, Pieler T., "Zinc forger proteins in early Xenopus development."
Int J Dev Biol. 1996 Feb;40(1):291-5; 19) Kroncke KD, Kolb-Bachofen V., "Detection of nitric oxide interaction with zinc finger proteins." Methods Enzymol. 1996;269:279-84; 20) Lopingco MC, Perkins AS., "Molecular analysis of Evil, a zinc finger oncogene involved in myeloid leukemia." Curr Top Microbiol Irnmunol. 1996;211:211-22:
21) Hromas R, Davis B, Rauscher FJ 3rd, Klemsz M, Tenen D, Hoffinan S, Xu D, Morris JF., "Hematopoietic transcriptional regulation by the myeloid zinc finger gene, MZF-l ." Curr Top Microbiol Immunol. 1996;211:159-64; 22) Nagaoka M, Sugiura Y., Conversion of zinc finger protein to artificial site-specific nuclease:
application to chromosome mapping and sequencing" Nippon Rinsho. 1995 Oct;53(10):2589-96;
23) Sarkar B. "Metal replacement in DNA-binding zinc forger proteins and its relevance to mutagenicity and carcinogenicity through free radical generation."
Nutrition. 1995 Sep-Oct;l l(5 Suppl):646-9; 24) Gashler A, Sukhatme, VP., "Early growth response protein 1 (Egr-1): prototype of a zinc-finger family of transcription factors." Prog Nucleic Acid Res Mol Biol. 1995;50:191-224; 25) Knegtel RM, van Tilborg MA, Boelens R, Kaptein R., "NMR structural studies on the zinc finger domains of nuclear hormone receptors." EXS. 1995;73:279-95; 26) Sanchez-Garcia I, Rabbitts TH., "The LIM domain: a new structural motif found in zinc-finger-like proteins." Trends Genet. 1994 Sep;10(9):315-20; 27) Pieler T, Bellefroid E., "Perspectives on zinc finger protein function and evolution--an update." Mol Biol Rep. 1994 Ju1;20(1):1-8; 28) Madden SL, Rauscher FJ 3d., "Positive and negative regulation of transcription and cell growth mediated by the EGR family of zinc-finger gene products." Ann N Y Acad Sci. 1993 Jun 11;684:75-84; 29) Thiesen HJ, Bach C., "DNA recognition of C2H2 zinc-forger proteins. Evidence for a zinc-finger-specific DNA recognition code." Ann N Y Acad Sci. 1993 Jun 11;684:246-9; 30) Freemont PS., "The RING finger. A novel protein sequence motif related to the zinc finger."
Ann N Y Acad Sci. 1993 Jun 11;684:174-92; 31) Werner H, Roberts CT Jr, LeRoith D., "The regulation of IGF-I receptor gene expression by positive and negative zinc-finger transcription factors." Adv Exp Med Biol. 1993;343:91-103; 32) Berg JM., "Spl and the subfamily of zinc finger proteins with guanine-rich binding sites." Proc Natl Acad Sci U S A. 1992 Dec 1;89(23):11109-10; 33) Reddy BA, Etkin LD, Freemont PS., "A novel zinc finger coiled-coil domain in a family of nuclear proteins." Trends Biochem Sci. 1992 Sep;l7(9):344-5; 34) Freedman LP., "Anatomy of the steroid receptor zinc finger region." Endocr Rev. 1992 May;13(2):129-45; 35) el-Baradi T, Pieler T., "Zinc finger proteins: what we know and what we would like to know." Mech Dev. 1991 Nov;35(3):155-69; 36) Schule R, Evans RM., "Cross-coupling of signal transduction pathways: zinc finger meets leucine zipper."
Trends Genet. 1991 Nov-Dec;7(11-12):377-81; 37) Summers MF., Zinc forger motif for single-stranded nucleic acids? Investigations by nuclear magnetic resonance."
J Cell Biochem. 1991 Jan;45(1):41-8; 38) Ashworth A, Denny P., "Zinc finger protein genes in the mouse genome." Mamm Genome. 1991;1(3):196-200; 39) South TL, Kim B, Hare DR, Summers MF., "Zinc fingers and molecular recognition. Structure and nucleic acid binding studies of an HIV zinc finger-like domain." Biochem Pharmacol.
1990 Jul 1;40(1):123-9; 40) Berg JM., "Zinc forger domains: hypotheses and current knowledge." Annu Rev Biophys Biophys Chem. 1990;19:405-21; 41) Copeland NG, Jenkins NA., "Retroviral integration in murine myeloid tumors to identify Evi-l, a novel locus encoding a zinc-finger protein." Adv Cancer Res. 1990;54:141-57;
42) Erickson RP, Verga V., "Is zinc-finger Y the sex-determining gene?" Am J Hum Genet. 1989 Nov;45(5):671-4; and 43) Struhl K., "Helix-turn-helix, zinc-finger, and leucine-zipper motifs for eukaryotic transcriptional regulatory proteins."
Trends Biochem Sci. 1989 Apr;l4(4):137-40.
Effector domains Although in some cases, a zinc forger protein itself is sufficient for modulating gene expression, the zinc finger protein is preferably fused to an effector domain (or regulatory domain or functional domain), i.e., a protein domain which activates or represses gene expression, e.g., an activator domain of a regulatory protein or an active domain of a nucleic acid modifying protein. The foregoing terms - i.e., "effector domain (or effector)," "regulatory domain," and "functional domain"
may refer to the materials on either the nucleic acid or protein level as will be clear from the context.

The effector domain can have an activity such as transcriptional modulation activity, DNA modifying activity, protein modifying activity and the like when tethered to a DNA binding domain, i.e., a zinc finger protein. Examples of regulatory domains include proteins or effector domains of proteins such as transcription factors and co-factors, e.g., KRAB, MAD, ERD, SID, nuclear factor kappa B subunit p65, early growth response factor 1, and nuclear hormone receptors, VP16 and VP64, endonucleases, integrases, recombinases, methyltransferases, histone acetyltransferases, histone deacetylases, mutases, restriction enzymes, etc.
Activators and repressors include co-activators and co-repressors (see, e.g., Utley et al., Nature (1998) 394:498-502; and WO 00/03026). Effector domains can include DNA-binding domains from a protein that is not a zinc finger protein, such as a restriction enzyme, a nuclear hormone receptor, a homeodomain protein such as engrailed or antenopedia, a bacterial helix-turn-helix motif protein such as lambda repressor and tet repressor, Gal4, TATA binding protein, helix-loop-helix motif proteins such as myc and myo D, leucine zipper type proteins such as fos and jun, and beta sheet motif proteins such as met, arc, and rnnt repressors. Particularly preferred activator is the C1 activator domain of maize (Goff et al., Genes Dev. (1991) 5(2):298-309).
In a specific embodiment, the zinc finger protein having an effector domain is one that is responsive to a ligand. The effector domain can effect such a response.
Use of ligand inducible binding domain-effector fusions is generally known as a gene switch. Therefore, the ZFP domains discussed here can be used as part of the ligand-binding domain in gene switches. Example of such ligand-responsive domains include hormone receptor ligand binding domains, e.g., estrogen receptor domain, ecydysone receptor system, glucocorticosteroid (Parker, CuYr. Opin. Cell Biol.
(1993) 5:499-504; Beato et al., Cell (1995) 83:851-857; Mangelsdorf et al., Cell (1995) 83:835-839; and Yamamoto, Annu. Rev. Genet. (1985) 19:209-252), inducible promoters of trehalose biosynthetic genes (WO 99/46370), chemically inducible arabidopsis PR-1 promoter (WO 98/03536; and U.S. Patent No. 5,689,044) and a chemically inducible promoter of a cucumber chitinase/lysozyme gene (LT.S.
Patent No. 5,654,414). Preferred inducers are small, inorganic, biodegradable, molecules.
The zinc forger protein can be covalently or non-covalently associated with one or more regulatory domains. Alternatively, two or more regulatory domains, whether identical or different ones, can be linked together. The regulatory domains can be covalently linked to'the zinc finger protein nucleic acid binding domain, e.g., via an amino acid linker, as part of a fusion protein. The zinc finger proteins can also be associated with a regulatory domain via a non-covalent dimerization domain, e.g., a leucine zipper, a STAT protein N terminal domain, or an FK506 binding protein (see, e.g., O'Shea, Science (1991) 254:539; Barahmand-Pour et al., Curr. Top.
Microbiol. Immunol. (1996) 211:121-128; Klemm et al., Annu. Rev. Immunol.
(1998) 16:569-592; Klemm et al., Annu. Rev. Immunol. (1998) 16:569-592; Ho et al., Nature (1996) 382:822-826; and Pomeranz et al., Biochem. (1998) 37:965). The regulatory domain can be associated with the zinc finger protein domain at any suitable position, including the C- or N-terminus of the zinc finger protein.
Common regulatory domains for addition to the zinc finger protein made using the methods of the invention include, e.g., DNA-binding domains from transcription factors, effector domains from transcription factors (activators, repressors, co-activators, co-repressors), silencers, nuclear hormone receptors, and chromatin associated proteins and their modifiers, e.g., methylases, kinases, acetylases and deacetylases.
Transcription factor polypeptides from which one can obtain a regulatory domain include those that are involved in regulated and basal transcription.
Such polypeptides include transcription factors, their effector domains, coactivators, silencers, nuclear hormone receptors (see, e.g., Goodrich et al., Cell (1996) 84:825-830) for a review of proteins and nucleic acid elements involved in transcription.
Transcription factors in general are reviewed in Bames and Adcock, Clin. Exp.
Allerg v 25 Suppl. 2:46-49 (1995) and Roeder, Methods Enz. (1996) 273:165-171.
Databases dedicated to transcription factors are also known (see, e.g., Williams, Science (1995) 269:630). Nuclear hon- none receptor transcription factors are described in, for example, Rosen et al., J. Med. Chem. (1995) 38:4855-4874.
The CBBP family of transcription factors are reviewed in Wedel et al., Imrnunobiology (1995) 193:171-185. Coactivators and co-repressors that mediate transcription regulation by nuclear hormone receptors are reviewed in, for example, Meier, Eur. J.
Endocrinol. (1996) 134(2):158-9; Kaiser et al., Trends Biochem. Sci. (1996) 21:342-345; and Utley et al., Nature (1998) 394:498-502. GATA transcription factors, which are involved in regulation of hematopoiesis, are described in, for example, Simon, Nat. Genet. (1995) 11:9-11; and Weiss et al., Exp. Hematol. 23:99-107. TATA
box binding protein (T13P) and its associated TAF polypeptides (which include TAF30, TAF55, TAF80, TAF1 10, TAFI 50, and TAF250) are described in Goodrich & Tjian, Curr. Opin. Cell Biol. (1994) 6:403-409 and Hurley, Curr. Opin. Struct. Biol.
(i996) 6:69-75. The STAT family of transcription factors are reviewed in, for example, Barahmand-Pour et al., Curr. Top. Microbiol. Immunol. (1996) 211:121-128.
Transcription factors involved in disease are reviewed in Aso et al., J Clin.
Invest.
(1996) 97:1561-1569.
In one embodiment, the KRAB repression domain from the human KOX- I
protein is used as a transcriptional repressor (Thiesen et al., New Biologist (1990) 2:363-374; Margolin et al., Proc. Natl. Acad. Sci. USA (1994) 91:4509-4513;
Pengue et al., Nucl. Acids Res. (1994) 22:2908-2914; and Witzgall et al., Proc. Natl.
Acad.
Sci. USA (1994) 91:4514-4518). In another embodiment, KAP-1, a KRAB co-represcor, is used with KR.AB (Friedman et al., Genes Dev. (1996) 10:2067-2078).
Alternatively, KAP- I can be used alone with a zinc finger protein. Other preferred transcription factors and transcription factor domains that act as transcriptional repressors include MAD (see, e.g., Sommer et al., JBiol. Chem. (1998) 273:6632-6642; Gupta et al., Oncogene (1998) 16:1149-1159; Queva et al., Oncogene (1998) 16:967-977; Larsson et al., Oncogene (1997) :737-748; Laherty et al., Cell (1997) 89:349-356; and Cultraro et al., Mol Cell. Biol. (1997) 17:2353-2359); FKHR
(forkhead in rhapdosarcoma gene ) (Ginsberg et al., Cancer Res. (1998) 15:3542-3546; and Epstein et al., Mol. Cell. Biol. (1998) 18:4118-4130); EGR- I (early growth response gene product- 1) (Yan et al., Proc. Natl. Acad. Sci. USA (1998) 95:8298-8303; and Liu et al., Cancer Gene Tlaer. (1998) 5:3-28); the ets2 repressor factor repressor domain (ERD) (Sgouras et al., EMBO J (1995) 14:4781-4793); and the MAD smSIN3 interaction domain (SID) (Ayer et al., Mol. Cell. Biol. (1996) 16:5772-5781).
In another embodiment, the HSV VP 16 activation domain is used as a transcriptional activator (see, e.g., Hagmann et al., J Virol. (1997) 71:5952-5962).
Other preferred transcription factors that could supply activation domains include the VP64 activation domain (Selpel et al., EMBO J (1996) 11:4961-4968); nuclear hormone receptors (see, e.g., Torchia et al., Curr. Opin. Cell. Biol. (1998) 10:373-383); the p65 subunit of nuclear factor kappa B (Bitko & Barik, J Virol.
(1998) 72:5610-5618 and Doyle & Hunt, Neuroreport (1997) 8:2937-2942); and EGR-I
(early growth response gene product-1) (Yan et al., Proc. Nad. Acad. Sci. USA
(1998) 95:8298-8303; and Liu et al., Cancer Gene Ther. (1998) 5:3-28).

Kinases, phosphatases, and other proteins that modify polypeptides involved in gene regulation are also useful as regulatory domains for zinc finger proteins. - Such modifiers are often involved in switching on or off transcription mediated by, for example, hormones. Kinases involved in transcription regulation are reviewed in Davis, Mol. Reprod. Dev. (1995) 42:459-467, Jackson et al., Adv. Second Messenger Phosphoprotein Res. (1993) 28:279-286, and Boulikas, Crit. Rev. Eukaryot. Gene Expr. (1995) 5:1-77, while phosphatases axe reviewed in, for example, Schonthal &
Semin, Cancer Biol. (1995) 6:239-248. Nuclear tyrosine kinases are described in Wang, Trends Biochem. Sci. (1994) 19:373-376.
Useful domains can also be obtained from the gene products of oncogenes (e.g., myc, jun, fos, myb, max, mad, rel, ets, bcl, myb, mos family members) and their associated factors and modifiers. Oncogenes are described in, for example, Cooper, Oncogenes, 2nd ed., The Jones and Bartlett Series in Biology, Boston, MA, Jones and Bartlett Publishers, 1995. The ets transcription factors are reviewed in Waslylk et al., Eur. JBiochem. (1993) 211:7-18. Myc oncogenes are reviewed in, for example, Ryan et al., Biochern. J. (1996) 314:713-721. The Jun and fos transcription factors are described in, for example, The Fos and Jun Families of Transcription Factors, Angel & Herrlich, eds. (1994). The max oncogene is reviewed in Hurlin et al., Cold Spring Harb. Symp. Quant. Biol. 59:109-116. The myb gene family is reviewed in Kanei-Ishii et al., Curr. Top. Microbiol. Immunol. (1996) 211:89-98. The mos family is reviewed in Yew et al., Curr. Opin. Genet. Dev. (1993) 3:19-25.
In still another embodiment, histone acetyltransferase is used as a transcriptional activator (see, e.g., Jin & Scotto, Mol. Cell. Biol. (1998) 18:4377-4384;
Wolffle, Science (1996) 272:371-372; Taunton et al., Science (1996) 272:408-411;
and Hassig et al., Proc. Natl. Acad. Sci. USA (1998) 95:3519-3524). In another embodiment, histone deacetylase is used as a transcriptional repressor (see, e.g., Jin &
Scotto, Mol. Cell. Biol. (1998) 18:4377-4384; Syntichaki & Thireos, JBiol.
Chern.
(1998) 273:24414-24419; Sakaguchi et al., Genes Dev. (1998) 12:2831-2841; and Martinez et al., JBiol. Chem. (1998) 273:23781-23785).
In a preferred embodiment, the plant effectors listed in the following Table 1 can be used:
Table 1. Exemplary plant effectors.

Gene Or anismActivatorRe ressorRegion GenBankplasmidReference g p Cloned Accession (aa APETALAArabidopsiyes 194-255527109 ppl Plant Molecular s Effector (AP1) Biology 40:419-429 DNA Maize yes 135-238X66076 Dofl The Plant EffectorCell binding 10:75-89 with one (1998) finger (Doff ) Dof2 Maize yes 112-221S~9852~mDof2 The plant EffectorCell 10:75-89 (1998) Dof2 Arabidopsi 115-204BAA3319AtDofl Blast Search s 7 Effector Viral Herpes yes VP16 ProteinVirus Effector (VP16) CI maize yes C1 Effector G-box Arabidopsiyes I-110 P42774 GBF1 EMBO

bindings Effector11 (4):1275-factor 1289 (1992) (GBF-1 ) Mybstl Potato 173-358 MybstlEffeEMBO

ctor 13(22):

(1994) PCP1 Potato 206-496X82328 PCP1 Mol. Gen.

EffectorGenet.247:

(1995) PWRKY1 Parsley yes 1-187 U48831 Pwrkyl EMBO

Effector18(17):

(1999) PWRKY3 Parsley 1-237 Pwrky3 EMBO

Effector18(17):

(1999) AFTI Arabidopsiyes I-248 U02565 AFTI FEBS
(14-3-3 s EffectorLetters protein) 443:

(1999) ZAP1 Arabidopsiyes 168-282X92976 Zapl Nucleic EffectorAcids' Research 24(23):

(1996) Target genes and target nucleotide seguences Expression of any target genes in plant can be modulated by the present method. For example, the expression of the APETALA3 (AP3) gene of Arabidopsis can be modulated. The APETALA3 (AP3) gene of Arabidopsis is a member of the ADS domain proteins that are required to specify the flower organ types and are involved in regulation of floral development. The success illustrated below in targeting this gene for endogenous gene regulation is important for several reasons.
First, Arabidopsis is a well-studied model organism in which genetic studies are easily and rapidly performed and is considered a typical case. Second, AP3 has been well studied in its role during flower development to specify floral organ identity (see, Yanofsky, Annu. Rev. Plant Physiol. Plant Mol. Biol. (1995) 46:167-188; and Irish, Developmental Biology (1999) 209:211-220) and detailed analysis of the AP3 promoter and its transcription have already been published (Irish et al., The Plc~nt Cell (1995) 7:1635-1644) so that the approach is illustrative for other plants as well.
Finally, both ap3 mutants and AP3 overexpressors have previously been shown to have visible flower phenotypic changes (Jack et al., Cell (1992) 68:683-687;
and Jack et al., Cell (1994) 76:703-716), which makes success verifiable. Thus, the AP3 gene is an excellent target for demonstrating this technology as a tool for controlling gene expression in plants in general.
In another example, the expression of the maize MIPS gene can be modulated.
The maize MIPS protein catalyzes the conversion of glucose-6-phosphate to rnyoinositol 1-phosphate which is an early step in the synthesis of phytic acid. Low phytic acid content in feed for many species of animals is considered to have a great advantage from environmental and nutritional standpoints since phytic acid metabolism in plant plays a role in the regulation of phosphate and mineral concentration. Controlling the expression of the MIPS gene would therefore provide a means to assure plants with low phytic acid content and act as a dominant trait. The present invention permits such control, as well as control of genetic expression generally in plants.
The target nucleotide sequence is any location within the target gene whose expression is to be regulated which provides a suitable location for controlling expression. The target nucleotide sequence may be within the coding region or upstream or downstream thereof. For activation, upstream from ATG translation start codon is preferred, most preferably upstream of TATTA box but not exceeding 1000 by from the start of transcription. For repression, upstream from the ATG
translation start codon is also preferred, but preferably downstream from TATTA box.
The targeted nucleotide sequence can also be a short portion of duplex nucleic acid, e.g., RNA, DNA, PNA or any hybrids thereof, having from about 8 to about base pairs and having a defined sequence for which there is some desirable purpose in determining its presence or absence within a larger polynucleotide. For example, it may be desirable to determine whether a particular promoter or control region is found within the genome of a particular organism. A labeled protein, e.g., bound with a radioactive or fluorescent label, containing zinc fingers which binds to a polynucleotide having this particular sequence can be used to determine whether the genetic material of the organism contains this particular sequence.
The target sequence may reside endogenously in the target gene, e.g., M1PS in maize and AP3 in Arabidopsis as shown in the Example Section below, or may be inserted into the gene, e.g., heterologous, as is illustrated below for luciferase in tobacco, for example, using techniques such as homologous recombination.
Where a gene contains a suitable target nucleotide sequence in a region which is appropriate for controlling expression, the regulatory factors employed in the methods of the invention can target the endogenous nucleotide sequence.
However, if the target gene lacks an appropriate unique nucleotide sequence or contains such a sequence only in a position where binding to a regulatory factor would be ineffective in controlling expression, it may be necessary to provide a "heterologous"
targeted nucleotide sequence. By "heterologous" targeted nucleotide sequence is meant either a sequence completely foreign to the gene to be targeted or a sequence which resides in the gene itself, but in a different position from that wherein it is inserted as a target.
Thus, it is possible to completely control the nature and position of the targeted nucleotide sequence.
The target sequence may be any given sequence of interest for which a complementary zinc finger protein is designed. Target genes include both structural and regulatory genes. When the target gene is a regulatory gene, the expression of the gene that is regulated by the target regulatory gene can also be regulated by the zinc finger protein, albeit indirectly. Therefore, expression of single genes or gene families can be controlled by the present methods.
The target gene may, as is the case for the MIPS gene and AP3 gene, be endogenous to the plant cells or plant wherein expression is regulated or may be a transgene which has been inserted into the cells or plants in order to provide a production system for a desired protein or which has been added to the genetic compliment in order to modulate the metabolism of the plant or plant cells, for example as illustrated below for the C7 binding site inserted into tobacco genome.
Selection of ZFP effector and target nucleotide seguence The assay system exemplified in Example 1 (see Example Section below) can be used in selecting desirable ZFP, effector or target nucleotide sequence.
The assay system used in the Example 1 generally contains two constructs. One construct contains a reporter gene, e.g., P-glucuronidase (GUS), chloramphenicol acetyl transferase (CAT), or green fluorescent protein (GFP), operably linked to a promoter having a ZFP target nucleotide sequence so that the expression of the reporter gene is amenable to regulation by the ZFP. The other construct contains ZFP gene, and optionally fused to an effector domain. Upon introduction into and expression within plant or plant cells, whether transiently or stably, the ZFP, and optionally the ZFP-effector fusion protein, will regulate the expression of the reporter gene, provided that the ZFP and the target nucleotide sequence have matching "code" sequence, e.g., the pairs listed in Table 2. In such a system, if only one component of the system is changed and the rest of the components are kept constant with the known operative ones, the assay system can be used to screen or select fox new ZFP, effector or target nucleotide sequence with desirable specificity or binding affinity.
For example, if a system is started with a ZFP, optionally with an effector and a target nucleotide sequence that are compatible and substitute the ZFP with a new amino acid sequence, it will be possible to determine whether the new amino acid sequence can function as a ZFP with the desired specificity and affinity to the particular target nucleotide sequence by comparing the assay readout with the known ZFP and the new amino acid sequence. New effector and target nucleotide sequence can be identified using similar procedure. If desired, the assay can be conducted quantitatively to identify ZFP, effector or target nucleotide sequence with particular specificity and binding affinity.
Since ZFPs can tolerate changes in the finger region, one can start with a preferred pairs of ZFP and target nucleotide sequence, e.g., the pairs listed in Table 2, and mutagenize the finger region of the ZFP to identify those variants that can still function as a ZFP, but with different specificity to binding affinity. It is often necessary to select ZFP finger sequence by testing against actual DNA
sequence, since the juxtaposition of triplets may affect optimal ZFP finger sequence. In addition, particular positions in the finger region, e.g., -1, 3, 6, should be the focused in the mutagenesis analysis because these positions are thought to be critical for the binding between a ZFP and its target nucleotide sequence. Accordingly, ZFPs with different level of specificities and binding affinities can be obtained and these ZFPs can be used in fine-tuned control of a target gene expression in plant.
C. Expression of zinc finger protein in plant and uses thereof The zinc finger protein can be provided to the plant cells via any suitable methods known in the art. For example, the zinc finger protein can be exogenously added to the plant cells and the plant cells are maintained under conditions such that the zinc finger protein binds to the target nucleotide sequence and regulates the expression of the target gene in the plant cells. Alternatively, a nucleotide sequence encoding the zinc finger protein can be expressed in the plant cells and the plant cells are maintained under conditions such that the expressed zinc forger protein binds to the target nucleotide sequence and regulates the expression of the target gene in the plant cells.
The zinc finger gene can be expressed in plant with any suitable plant expression vectors. Typical vectors useful for expression of genes in higher plants are well known in the art and include vectors derived from the tumor-inducing (Ti) plasmid of Agrobacterium tumefaciens described by Rogers et al., Meth. in Enzymol.
(1987) 153:253-277. These vectors are plant integrating vectors in that upon transformation, the vectors integrate a portion of vector DNA into the genome of the host plant. Exemplary A. tumefaciens vectors useful herein are plasmids pKYLX6 ' and pKYLX7 of Schardl et al., Gehe (1987) 61:1-11 and Berger et al., Proc.
Natl.
Acad. Sci. USA (1989) 86:8402-8406. Another useful vector herein is plasmid pBI
101.2 that is available from Clontech Laboratories, Inc. (Palo Alto, CA).
In addition to regulatory domains, often the zinc finger protein can be expressed as a fusion protein such as maltose binding protein ("MBP"), glutathione S
transferase (GST), hexahistidine, c-myc, and the FLAG epitope, for ease of purification, monitoring expression, or monitoring cellular and subcellular localization.

The nucleic acid sequence encoding a ZFP can be modified to improve ' expression of the ZFP in plants by using codon preference. When the nucleic acid is prepared or altered synthetically, advantage can be taken of known codon preferences of the intended plant host where the nucleic acid is to be expressed. For example, although nucleic acid sequences of the present invention may be expressed in both monocotyledonous and dicotyledonous plant species, sequences can be modified to account for the specific codon preferences and GC content preferences of monocotyledons or dicotyledons as these preferences have been shown to differ (hurray et al., Nucl. Acids Res. (1989) 17: 477-498). Thus, the maize preferred codon for a particular amino acid may be derived from known gene sequences from maize. Maize codon usage for 28 genes from maize plants are listed in Table 4 of hurray et al., supra.
In one embodiment, the transgene may include a reporter protein such as luciferase, which can be helpful in providing an assay system for determining the position in a target gene in which the targeted nucleotide sequence should reside.
Thus, the invention also includes an assay system for determination of a suitable region for targeting by regulatory factors, wherein said assay method comprises providing a chimeric gene comprising a nucleotide sequence encoding a reporter protein optionally fused to coding regions of said gene; said nucleotide sequence operably linked to control sequences endogenous to said gene. The chimeric gene is then provided a targeted nucleotide sequence at various locations within the endogenous portions and the effect on production of the reporter protein of contact with the appropriate zinc finger containing regulatory protein is determined.
It may be desirable in some instances to modify plant cells or plants with families of transgenes representing, for example, a metabolic pathway. In those instances, it may be desirable to design the constructs so that the family can be regulated as a whole, e.g., by designing the control regions of the members of the family with similar or identical targets for the zinc finger protein portion of the effector protein. Such sharing of target sequences in gene families may occur naturally in endogenously produced metabolic sequences.
In most instances, it is desirable to provide the expression system for the effector protein with control sequences that are tissue specific so that the desired gene regulation can occur selectively in the desired portion of the plant. For example, to repress MIPS expression, it is desirable to provide the effector protein with control sequences that are selectively effective in seeds. With respect to the AP3 gene, effector proteins for regulation of expression would be designed for selective -expression in flowering portions of the plant. However, in some instances, it may be desirable to have the genetic control expressible in all tissues in instances where an insect resistance gene is the target. In such cases, it may be desirable to place the expression system for the effector protein under control of an inducible promoter so that inducer can be supplied to the plant only when the need arises, for example, activation of an insect resistance gene.
In another embodiment, ZFPs can be used to create functional "gene knockouts" and "gain of function" mutations in a host cell or plant by repression or activation of the target gene expression. Repression or activation may be of a structural gene, e.g., one encoding a protein having for example enzymatic activity, or of a regulatory gene, e.g., one encoding a protein that in turn regulates expression of a structural gene. Expression of a negative regulatory protein can cause a functional gene knockout of one or more genes, under its control. Conversely, a zinc finger having a negative regulatory domain can repress a positive regulatory protein to knockout or prevent expression of one or more genes under control of the positive regulatory protein.
The present invention further provides recombinant expression cassettes comprising a ZFP-encoding nucleic acid. A nucleic acid sequence coding for the desired polynucleotide of the present invention can be used to construct a recombinant expression cassette which can be introduced into the desired host cell. A
recombinant expression cassette will typically comprise a polynucleotide encoding creation zinc finger proteins, e.g., ZFPml, ZFPm2, ZFPm3, ZFPm4 and ZFPAp3, operably linked to transcriptional initiation regulatory sequences which will direct the transcription of the polynucleotide in the intended host cell, such as tissues of a transformed plant.
For example, plant expression vectors may include (1) a cloned plant gene under the transcriptional control of 5' and 3' regulatory sequences and (2) a dominant selectable marker. Such plant expression vectors may also contain, if desired, a promoter regulatory region (e.g., one confernng inducible or constitutive, environmentally- or developmentally-regulated, or cell- or tissue-specific/selective expression), a transcription initiation start site, a ribosome binding site, an RNA
processing signal, a transcription termination site, and/or a polyadenylation signal.

A plant promoter fragment can be employed which will direct expression of a polynucleotide of the present invention in all tissues of a regenerated plant.
Such promoters are referred to herein as "constitutive" promoters and are active under most environmental conditions and states of development or cell differentiation.
Examples of constitutive promoters include the rice actin 1 promoter (L1.5. Patent No.
5,641,876), the cauliflower mosaic virus (CaMV) 355 transcription initiation region (Odell et al., Nature (1985) 313:810-812), the P- or 2'- promoter derived from T-DNA
of Agrobacterium tumefaciens, the ubiquitin I promoter, the Smas promoter, the cinnamyl alcohol dehydrogenase promoter (IJ.S. Patent No. 5,683,439), the Nos promoter, the pEmu promoter, the rubisco promoter, the GRP 1 - 8 promoter, and other transcription initiation regions from various plant genes known to those of skilled artisans.
Alternatively, the plant promoter can direct expression of a polynucleotide of the present invention in a specific tissue or may be otherwise order more precise environmental or developmental control. Such promoters are referred to here as "inducible" promoters. Environmental conditions that may effect transcription by inducible promoters include pathogen attack, anaerobic conditions, or the presence of light. Examples of inducible promoters are the AdhI promoter which is inducible by hypoxia or cold stress, the Hsp70 promoter which is inducible by heat stress, and the PPDK promoter which is inducible by light. Examples of promoters under developmental control include promoters that initiate transcription only, or preferentially, in certain tissues, such as leaves, roots, fruit, seeds, or flowers. An exemplary promoter is the anther specific promoter 5126 (U.5. Patent Nos.
5,689,049 and 5,689,051). The operation of a promoter may also vary depending on its location in the genome. Thus, an inducible promoter may become fully or partially constitutive in certain locations.
Both heterologous and non-heterologous (i.e., endogenous) promoters can be employed to direct expression of the nucleic acids of the present invention.
These promoters can also be used, for example, in recombinant expression cassettes to drive expression of antisense nucleic acids to reduce, increase, or alter concentration and/or composition of the proteins of the present invention in a desired tissue.
Thus, in some embodiments, the nucleic acid construct will comprise a promoter functional in a plant cell, such as in Zea mays, operably linked to a polynucleotide of the present invention.

In some embodiments, isolated nucleic acids which serve as promoter or enhancer elements can be introduced in the appropriate position (generally upstream) of a non- heterologous form of a polynucleotide of the present invention so as to up or down regulate expression of a polynucleotide of the present invention. For example, endogenous promoters can be altered in vivo by mutation, deletion, and/or substitution (see, Kmiec, U.S. Patent No. 5,565,350; WO 93/22443), or isolated promoters can be introduced into a plant cell in the proper orientation and distance from a gene of the present invention so as to control the expression of the gene. Gene expression can be modulated under conditions suitable for plant growth so as to alter the total concentration and/or alter the composition of the polypeptides of the present invention in plant cell.
The selection of the promoter used in expression cassettes will determine the spatial and temporal expression pattern of the transgene in the transgenic plant, i.e., the ZFP gene is only expressed in the desired tissue or at a certain time in plant development or growth. Selected promoters will express transgenes in specific cell types (such as leaf epidermal cells, mesophyll cells, root cortex cells) or in specific tissues or organs (roots, leaves or flowers, for example) and the selection will reflect the desired location of accumulation of the gene product. Alternatively, the selected promoter may drive expression of the gene under various inducing conditions.
Additionally, the zinc finger encoding gene can be under control of and activated by a promoter responsive to the presence of a pathogen or to plant stress, e.g., cold stress, salt stress, etc., so that the induced ZFP can modulate a gene that counteracts the pathogen, stress, etc. Promoters vary in their strength, i. e., ability to promote transcription. Depending upon the host cell system utilized, any one of a number of suitable promoters can be used, including the gene's native promoter.
A variety of promoters will be useful in the invention, particularly to control the expression of the ZFP and ZFP-effector fusions, the choice of which will depend in part upon the desired level of protein expression and desired tissue-specific, temporal specific, or environmental cue-specific control, if any in a plant cell.
Constitutive and tissue specific promoters are of particular interest. Such constitutive promoters include, for example, the core promoter of the Rsyn7, the core CaMV

promoter (Odell et al., Nature (1985) 313:810-812), CaMV 19S (Grounder and Kohli, Mol Gen Genet 1989 Dec; 220(1):95-101), rice actin (Wang et al., Mol. Cell.
Biol., 12, 3399-3406 (1992); U.S. Patent No. 5,641,876; and McElroy et al., Plant Cell (1985) 2:163-171); ubiquitin (Christensen et al., Plant Mol. Biol.
(1989)12:619-632 and Christensen et al., Plant Mol. Biol. (1992) 18:675-689), pEMCT (Last et al., -Theor. Appl. Genet. (1991) 81:581-588), MAS (Veltenet al., EMBO J. (1984) 3:2723-2730), nos (Ebert et al., Proc. Natl. Acad. Sci. U.S.A. 84, 5745-5749 (1987)), Adh (Dennis and Berg, Nucleic Acids Res., 13:7945-7957 (1985)), and constitutive promoters described in, for example, U.S. Patent Nos. 5,608,149; 5,608,144;
5,604,121; 5,569,597; 5,466,785; 5,399,680; 5,268,463; and 5, 608,142.
Ubiquitin is a gene product known to accumulate in many cell types and its promoter has been cloned from several species for use in transgenic plants (e.g.
sunflower - Binet et al., Plant Science (1991) 79:87-94; maize - Christensen et al., Plant Molec. Biol. (1989) 12: 619-632; and Arabidopsis - Norns et al., Plant Mol.
Biol. (1993) 21:895-906). The maize ubiquitin promoter has been developed in transgenic monocot systems and its sequence and vectors constructed for monocot transformation are disclosed in the patent publication EP 0 342 926 (to Lubrizol) which is herein incorporated by reference. Taylor et al. (Plant Cell Rep.
(1993) 12:491-495) describe a vector (pAHC25) that comprises the maize ubiquitin promoter and first intron and its high activity in cell suspensions of numerous monocotyledons when introduced via microprojectile bombardment. The Arabidopsis ubiquitin promoter is also ideal for use with the nucleotide sequences of the present invention.
The ubiquitin promoter is suitable for gene expression in transgenic plants, both monocotyledons and dicotyledons. Suitable vectors are derivatives of pAHC25 or any of the transformation vectors described in this application, modified by the introduction of the appropriate ubiquitin promoter andlor intron sequences.
Construction of the plasmid pCGN1761 is described in the published patent application EP 0 392 225 (Example 23), which is hereby incorporated by reference.
pCGN1761 contains the "double" CaMV 35S promoter and the tml transcriptional terminator with a unique EcoRI site between the promoter and the terminator and has a pUC-type backbone. A derivative of pCGN1761 is constructed which has a modified polylinker which includes Notl and Xhol sites in addition to the existing EcoRI site. This derivative is designated pCGN1761ENX. pCGN1761ENX is useful for the cloning of cDNA sequences or coding sequences (including microbial ORF
sequences) within its polylinker for the purpose of their expression under the control of the 35S promoter in transgenic plants. The entire 35S promoter-coding sequence-tml terminator cassette of such a construction can be excised by HindlIl, Sphl, SaII, and Xbal sites 5' to the promoter and Xbal, BamHI and BgII sites 3' to the terminator for transfer to transformation vectors such as those described below.
Furthermore, the double 35S promoter fragment can be removed by 5' excision with HindlIl, Sphl, SaII, ~'bal, or Pstl, and 3' excision with any of the polylinker restriction sites (EcoRI, Notl or ~Yho~ for replacement with another promoter. If desired, modifications around the cloning sites can be made by the introduction of sequences that may enhance translation. This is particularly useful when overexpression is desired. For example, pCGN1761ENX may be modified by optimization of the translational initiation site as described in Example 37 of U.S. Patent No. 5,639,949, incorporated herein by reference.
Several isoforms of actin are known to be expressed in most cell types and consequently the actin promoter is a good choice for a constitutive promoter.
In particular, the promoter from the rice Actl gene has been cloned and characterized (McElroy et al., Plant Cell (1990) 2:163-171). A 1.3 kb fragment of the promoter was found to contain all the regulatory elements required for expression in rice protoplasts. Furthermore, numerous expression vectors based on the Actl promoter have been constructed specifically for use in monocotyledons (McElroy et al., Mol.
Gen. Genet. (1991) 231:150-160). These incorporate the Actl intron 1, Adhl 5' flanking sequence and Adhl intron 1 (from the maize alcohol dehydrogenase gene) and sequence from the CaMV 35S promoter. Vectors showing highest expression were fusions of 35S and Actl intron or the Actl 5' flanking sequence and the Actl intron. Optimization of sequences around the initiating ATG (of the GUS
reporter gene) also enhanced expression. The promoter expression cassettes described by McF~lroy et al. (Mol. Gen. Genet. (1991) 231:150-160) can be easily modified for gene expression and are particularly suitable for use in monocotyledonous hosts. For example, promoter-containing fragments is removed from the McElroy constructions and used to replace the double 35S promoter in pCGN1761ENX, which is then available for the insertion of specific gene sequences. The fusion genes thus constructed can then be transferred to appropriate transformation vectors. In a separate report, the rice Actl promoter with its first intron has also been found to direct high expression in cultured barley cells (Chibbar et al., Plant Cell Rep. (1993) 12:506-509).
Tissue-specific promoters can be utilized to target enhanced expression within a particular plant tissue (See generally, copending U.S. Provisional Patent Application entitled "Promoters for Regulation of Plant Gene Expression, Attorney Docket No.
1360.002PRV (Schwegman, Lundberg, Woessner & Kluth), filed June 23, 2000, the content of which is herein incorporated by reference in its entirety. Examples of tissue specific promoters which have been described include the lectin (Vodkin, Prog.
Clinc: Biol. Res., 138:87-98 (1983); and Lindstrom et al., Dev. Genet., 11:160-(1990)), corn alcohol dehydrogenase -1 (Dennis et al., Nucleic Acids Res., 12:3983-4000 (1984)), corn light harvesting complex (Becker, PlantMol Biol., 2~: 49-60 (1992); and Bansal et al., Proc. Natl. Acad. Sei. U.S.A., 89:3654-3658 {1992)), corn heat shock protein (Odell et al., Nature (1985) 313:810-812; and Marrs et al., Dev.
Genet., 14 1 :27-41 (1993)), small subunit RuBP carboxylase (Waksman et al., Nucleic Acids Res., 15 17 :7181 (1987); and Berry-Lowe et al., J. Mol. Appl.
Genet., 1 6 :483-498 (1982)), Ti plasmid mannopine synthase (Ni et al., Plant Mol.
Biol., 30 1 :77-96 (1996)), Ti plasmid nopaline synthase (Bevan, Nz~cleic Acids Res., 11 2 :369-385 (1983)), petunia chalcone isomerase (Van Tunen et al., EMBO J., 7:1257-1263 (1988)), bean glycine _ .rich protein 1 (Keller et al., Genes Dev., 3:1639-1646 {1989)), truncated CaMV 35s (Odell et al., Nature (1985) 313:810-812), potato patatin (Wenzler et al., Plant Mol: Biol., 13:347-354 (1989)), root cell (Yamamoto et al., Nucleic Acids Res., 18:7449 (1990)), maize zero (Reina et al., Nucleic Acids Res., 18:6425 (1990); Kriz et al., Mol. Gen. Genet., 207:90-98 1987; Wandelt and Feix, Nucleic Acids Res., 17:2354 (1989); Langridge and Feix, Cell, 34:1015-1022 (1983);
and Reina et al., Nucleic Acids Res., 18:7449 (1990)), globulin-1 (Belanger et al., Genetics, 129:863-872 (1991)), a-tubulin, cab (Sullivan et al., Mol. Gen.
Genet., 215:431-440 (1989)), PEPCase (Cushman et al., Plant Cell, 1 7 :715-25 (1989)), R
gene complex-associated promoters (Chandler et al., Plant Cell, 1:1175-1183 (1989)), and chalcone -synthase promoters (Franken et al., EMBO J., 10:2605-2612, 1991)).
Tissue-specific promoters disclosed in the following references may also be used: Yamamoto et al., Plant J (1997) 12(2):255-265; Kawamata et al., Plant Cell Physiol. (1997) 38(7):792-803; Hansen et al., Mol. Gen Genet. (1997) 254(3):337);
Russell et al., Trarasgenic Res. (1997) 6(2):15 7-168; Rinehart et al., Plant Physiol.
(1996) 112(3):1331; Van Camp et al., Plant Physiol. (1996) 112(2):525-535;
Canevascini et al., Plant Physiol. (1996) 112(2):513-524; Yamamoto et al., Plant Cell Pkysiol. (1994) 35(5):773 -778; Lam, Results Probl. Cell Differ. (1994) 20:181-196;
Orozco et al., Plant Mol. Biol. (1993) 23(6):1129-1138; Matsuoka et al., Proc Natl.

Acad. Sci. USA (1993) 90(20):9586-9590; and Guevara-Garcia et al., Plant J.
(1993) 4(3):495-505. Such promoters can be modified, if necessary, for weak expression.
A maize gene encoding phosphoenol carboxylase (PEPC) has been described by Hudspeth & Grula (Plant Molec Biol 12: 579-589 (1989)). Using standard molecular biological techniques the promoter for this gene can be used to drive the expression of any gene in a leaf specific manner in transgenic plants. Other leaf specific promoters are known in the art, and include those described in, for example, Yamamoto et al., Plant J. (1997) 12(2):255-265; Kwon et al., Plant Physiol.
(1994) 105:357-367; Yamamoto et al., Plant Cell Physiol. (1994) 35(5):773-778; Gotor et al., Plant J. (1993) 3:509-518; Orozco et al., Plant Mol. Biol. (1993) 23(6):1129-1138; and Matsuoka et al., Proc. Natl. Acad. Sci. USA (1993) 90(20):9586-9590.
Another pattern of gene expression is root expression. A suitable root promoter is the promoter of the maize metallothionein-like (MTL) gene described by de Framond (FEBS (1991) 290:103-106) and also in U.S. Patent No. 5,466,785, incorporated herein by reference. This "MTL" promoter is transferred to a suitable vector such as pCGN1761ENX for the insertion of a selected gene and subsequent transfer of the entire promoter-gene-terminator cassette to a transformation vector of interest. Other examples of root-specific promoters which have been described include the RB7 promoter from Nicotiana tabacum (U.S. Patent Nos. 5,459,252 and 5,750,386).
8 describes the isolation of the maize calcium-dependent protein kinase (CDPK) gene which is expressed in pollen cells. The gene sequence and promoter extend up to 1400 by from the start of transcription. Using standard molecular biological techniques, this promoter or parts thereof, can be transferred to a vector such as pCGN1761 where it can replace the 35S promoter and be used to drive the expression of a nucleic acid sequence of the invention in a pollen-specific manner.
Inducible promoters and other types of regulated promoters can also be used.
For example, PR-1 promoters, ethanol-inducible promoters, glucocorticoid-inducible promoters, wound-inducible promoters, promoters for pith-preferred expression and promoters for receptor mediated transactivation in the presence of a chemical ligand can be used.
The double 35S promoter in pCGN1761ENX may be replaced with any other promoter of choice that will result in suitably high expression levels. By way of example, one of the chemically regulatable promoters described in U.S. Patent No.

5,614,395, such as the tobacco PR-la promoter, may replace the double 35S
promoter. Alternately, the Arabidopsis PR-1 promoter described in Lebel et al:, Plant J. (1998) 16:223-233 may be used. The promoter of choice is preferably excised from its source by restriction enzymes, but can alternatively be PCR-amplified using primers that carry appropriate terminal restriction sites. Should PCR-amplification be undertaken, then the promoter should be re-sequenced to check for amplification errors after the cloning of the amplified promoter in the target vector. The chemically/pathogen regulatable tobacco PR-1 a promoter is cleaved from plasmid pCIB1004 (for construction, see example 21 of EP 0 332 104, which is hereby incorporated by reference) and transferred to plasmid pCGN1761ENX (U.S. Patent No. 5,639,949). pCIB 1004 is cleaved with Ncol and the resultant 3' overhang of the linearized fragment is rendered blunt by treatment with T4 DNA polymerase. The fragment is then cleaved with HindlIl and the resultant PR-1 a promoter-containing fragment is gel purified and cloned into pCGN1761ENX from which the double 35S
promoter has been removed. This is done by cleavage with Xhol and blunting with T4 polymerase, followed by cleavage with HindlIl and isolation of the larger vector-terminator containing fragment into which the pCIB1004 promoter fragment is cloned. This generates a pCGN1761ENX derivative with the PR-1 a promoter and the tml terminator and an intervening polylinker with unique EcoRI and Notl sites.
The selected coding sequence can be inserted into this vector, and the fusion products (i. e.
promoter-gene-terminator) can subsequently be transferred to any selected transformation vector, including those described infra. Various chemical regulators may be employed to induce expression of the selected coding sequence in the plants transformed according to the present invention, including the benzothiadiazole, isonicotinic acid, and salicylic acid compounds disclosed in U.S. Patent Nos.
5,523,31 l and 5,614,395.
A promoter inducible by certain alcohols or ketones, such as ethanol, may also be used to confer inducible expression of a coding sequence of the present invention.
Such a promoter is for example the alcA gene promoter from Aspergillus nidulans (Caddick et al., (1998) Nat. Biotechnol 16:177-180). In A. nidulans, the alcA
gene encodes alcohol dehydrogenase I, the expression of which is regulated by the AlcR
transcription factors in presence of the chemical inducer. For the purposes of the present invention, the CAT coding sequences in plasmid palcA:CAT comprising a alcA gene promoter sequence fused to a minimal 35S promoter (Caddick et al., Nat.

Biotechnol (1998) 16:177-180) are replaced by a coding sequence of the present invention to form an expression cassette having the coding sequence under the control of the alcA gene promoter. This is earned out using methods well known in the art.
Induction of expression of a nucleic acid sequence of the present invention using systems based on steroid hormones is also contemplated. For example, a glucocorticoid-mediated induction system is used (Aoyama and Chua, The Plant ,Tournal (1997) 11:605-612) and gene expression is induced by application of a glucocorticoid, for example a synthetic glucocorticoid, preferably dexamethasone, preferably at a concentration ranging from O.lmM to lmM, more preferably from l OmM to 100mM. For the purposes of the present invention, the luciferase gene sequences are replaced by a nucleic acid sequence of the invention to form an expression cassette having a nucleic acid sequence of the invention under the control of six copies of the GAL4 upstream activating sequences fused to the 35S
minimal promoter. This is carried out using methods well known in the art. The traps-acting factor comprises the GAL4 DNA-binding domain (Keegan et al., Science (1986) 231:699-704) fused to the transactivating domain of the herpes viral protein (Triezenberg et al., Genes Devel. (1988) 2:718-729) fused to the hormone-binding domain of the rat glucocorticoid receptor (Picard et al., (1988) Cell 54:1073-1080).
The expression of the fusion protein is controlled by any promoter suitable for expression in plants known in the art or described here. This expression cassette is also comprised in the plant comprising a nucleic acid sequence of the invention fused to the 6xGALA~/minimal promoter. Thus, tissue- or organ-specificity of the fusion protein is achieved leading to inducible tissue- or organ-specificity of the insecticidal toxin.
Wound-inducible promoters may also be suitable for gene expression.
Numerous such promoters have been described (e.g. Xu et al., Plant Molec.
Biol.
(1993) 22:573-588; Logemann et al., Plant Cell (1989) 1:151-158; Rohrmeier &
Lehle, Plant Molec. Biol. (1993) 22:783-792; Firek et al., Plant Molec. Biol.
(1993) 22:129-142; and Warner et al., Plant J. (1993) 3:19'1-201) and all are suitable for use with the instant invention. Logemann et al. describe the 5' upstream sequences of the dicotyledonous potato wunl gene. Xu et al. show that a wound-inducible promoter from the dicotyledon potato (pint) is active in the monocotyledon rice. Further, Rohrmeier &
Lehle describe the cloning of the maize Wipl cDNA which is wound induced and which can be used to isolate the cognate promoter using standard techniques.
Similar, Firek et al. and Warner et al. have described a wound-induced gene from the monocotyledon Asparagus o~cinalis, which is expressed at local wound and pathogen invasion sites. Using cloning techniques well known in the art, these promoters can be transferred to suitable vectors, fused to the genes pertaining to this invention, and used to express these genes at the sites of plant wounding.
WO 93/07278, which is herein incorporated by reference, describes the isolation of the maize trpA gene, which is preferentially expressed in pith cells. The gene sequence and promoter extending up to -1726 by from the start of transcription are presented. Using standard molecular biological techniques, this promoter, or parts thereof, can be transferred to a vector such as pCGN1761 where it can replace the 35S
promoter and be used to drive the expression of a foreign gene in a pith-preferred manner. In fact, fragments containing the pith-preferred promoter or parts thereof can be transferred to anywector and modified for utility in transgenic plants.
U.S. Patent No. 5,880,333 describes a system whereby class II hormone receptors such as Ecdysone Receptor (EcR) and Ultraspiracle (LTSP), which function together as a heterodimer, regulate the expression of a target polypeptide in a plant cell in the presence of an appropriate chemical ligand, e.g., tebufenozide.
Particularly preferred are the inducible PRl promoter, maize ubiquitin promoter, and rice actin promoter.
Any combination of constitutive or inducible and non-tissue specific or tissue specific may be used to control ZFP expression.
The desired control may be temporal, developmental or environmentally controlled using the appropriate promoter. Environmentally controlled promoters are those that respond to assault by pathogen, pathogen toxin, or other external compound (e.g., intentionally applied small molecule inducer). An example of a temporal or developmental promoter is a fruit ripening-dependent promoter.
Thus, the present invention provides compositions, and methods for making, heterologous promoters and/or enhancers operably linked to a ZFP and ZFP-effector fusion encoding polynucleotide of the present invention.
By selection of different strength of promoter to express ZFP-effector fusion protein, one can control the effect of ZFP in gene expression regulation in many levels. Generally, The stronger the promoter is, the stronger the effect of ZFP-effector protein is. However, moderate level of gene expression activation or repression is desirable since over-expression of certain foreign protein or over-activation of certain endogenous gene in certain cell type may cause toxicity effect. As in the example of AP3 gene activation, the Ubiquitin promoter (strong promoter) was originally used to express ZFPAp3-VP64 activation fusion protein. We have switched to a weaker promoter AP3 endogenous promoter to express the same constructs to achieve even clear phenotype changes.
Methods for identifying promoters with a particular expression pattern, in terms of, e.g., tissue type, cell type, stage of development, and/or environmental conditions, are well known in the art. See, e.g., The Maize Handbook, Chapters 115, Freeling and Walbot, Eds., Springer, New York (1994); Corn and Corn Improvement, Pedition, Chapter 6, Sprague and Dudley, Eds., American Society of Agronomy, Madison, Wisconsin (1988).
A typical step in promoter isolation is identification of gene products that are expressed with some degree of specificity in the target tissue. Amongst the range of methodologies are: differential hybridization to cDNA libraries; subtractive hybridization; differential display; differential 2-D protein gel electrophoresis; DNA
probe arrays; and isolation of proteins known to be expressed with some specificity in the target tissue. Such methods are well known to those of skill in the art.
Commercially available products for identifying promoters are known in the art such as Clontech's (Palo Alto, CA) Universal GenomeWalker Kit.
For the protein-based methods, it is helpful to obtain the amino acid sequence for at least a portion of the identified protein, and then to use the protein sequence as the basis for preparing a nucleic acid that can be used as a probe to identify either genomic DNA directly, or preferably, to identify a cDNA clone from a library prepared from the target tissue. Once such a cDNA clone has been identified, that sequence can be used to identify the sequence at the 5' end of the transcript of the indicated gene. For differential hybridization, subtractive hybridization and differential display, the nucleic acid sequence identified as enriched in the target tissue is used to identify the sequence at the 5' end of the transcript of the indicated gene. Once such sequences are identified, starting either from protein sequences or nucleic acid sequences, any of these sequences identified as being from the gene transcript can be used to screen a genomic library prepared from the target organism.
Methods for identifying and confirming the transcriptional start site are well known in the art.

In the process of isolating promoters expressed under particular environmental conditions or stresses, or in specific tissues, or at particular developmental stages, a number of genes are identified that are expressed under the desired circumstances, in the desired tissue, or at the desired stage. Further analysis will reveal expression of each particular gene in one or more other tissues of the plant. One can identify a promoter with activity in the desired tissue or condition but that do not have activity in any other common tissue.
To identify the promoter sequence, the 5' portions of the clones can be analyzed for sequences characteristic of promoter sequences. For instance, promoter sequence elements include the TATA box consensus sequence (TATAAT), which is usually an AT-rich stretch of 5-10 by located approximately 20 to 40 base pairs upstreaW of the transcription start site. Identification of the TATA box is well known in the art. For example, one way to predict the location of this element is to identify the transcription start site using standard RNA-mapping techniques such as primer extension, S I analysis, and/or RNase protection. To confirm the presence of the AT-rich sequence, a structure-function analysis can be performed involving mutagenesis of the putative region and quantification of the mutation's effect on expression of a linked downstreamweporter gene. See, e.g., The Maize Handbook, Chapter 114, Freeling and Walbot, Eds., Springer, New York (1994).
In plants, further upstream from the TATA box, at positions -80 to -100, there is typically a promoter element (i.e., the CART box) with a series of adenines surrounding the trinucleotide G (or T) N G (Messing et al., in Genetic Engineering in Plants, Kosage, Meredith and Hollaender, Eds., pp. 221-227 1983). In maize, there is no well conserved CAAT box but there are several short, conserved protein-binding motifs upstream of the TATA box. These include motifs for the traps-acting transcription factors involved in light regulation, anaerobic induction, hormonal regulation, or anthocyanin biosynthesis, as appropriate for each gene.
Once promoter and/or gene sequences are known, a region of suitable size is selected from the genomic DNA that is 5' to the transcriptional start, or the translational start site, and such sequences are then linked to a coding sequence. If the transcriptional start site is used as the point of fusion, any of a number of possible 5' untranslated regions can be used in between the transcriptional start site and the partial coding sequence. If the translational start site at the 3' end of the specific promoter is used, then it is linked directly to the methionine start codon of a coding sequence.
If polypeptide expression is desired, it is generally desirable to include a polyadenylation region at the 3'-end of a polynucleotide coding region. The polyadenylation region can be derived from the natural gene, from a variety of other plant genes, or from T-DNA. The 3' end sequence to be added can be derived from, for example, the nopaline synthase or octopine synthase genes, or alternatively from another plant gene, or less preferably from any other eukaryotic gene.
An intron sequence can be added to the S' untranslated region or the coding sequence of the partial coding sequence to increase the amount of the mature message that accumulates in the cytosol. Inclusion of a spliceable intron in the transcription unit in both plant and animal expression constructs has been shown to increase gene expression at both the mRNA and protein levels up to 1,000-fold (Buchman and Berg, Mol. Cell Biol. (1988) 8:4395-4405; and Callis et al., Genes Dev. (1987) 1:1183-1200). Such intron enhancement of gene expression is typically greatest when placed near the 5' end of the transcription unit. Use of maize introns Adhl-S intron 1, 2, and 6, the Bronze-I intron are known in the art (See generally, The Maize Handbook, Chapter 116, Freeling and Walbot, Eds, Springer, New York (1994)) Plant transformation protocols as well as protocols for introducing nucleotide sequences into plants may vary depending on the type of plant or plant cell, i.e., monocot or dicot, targeted for transformation. Suitable methods of introducing nucleotide sequences into plant cells and subsequent insertion into the plant genome include rnicroinjection (Crossway et al., Biotechniques (1986) 4:320-334), electroporation (Riggs et al., Proc. Nc~tl. Acad Sci. USA (1986) 83:5602-5606), Agrobacterium-mediated transformation (Townsend et al., U.S. Patent No.
5,563,055), direct gene transfer (Paszkowski et al., EMBO J. (1984) 3:2717-2722), and ballistic particle acceleration (see, for example, Sanford et al., U.S.
Patent No.
4,945,050; Tomes et al. (1995). "Direct DNA Transfer into Intact Plant Cells via Microprojectile Bombardment,"~in Plant Cell, Tissue, and Organ Culture:
Fundamental Methods, ed. Gamborg and Phillips (Springer-Verlag, Berlin); and McCabe et al., Biotechnology (1988) 6:923-926). Also see Weissingeretal., Ann.
Rev.
Genet. (1988) 22:421-477; Sanford et al., Particulate Science and Technology (1987) 5:27-37 (onion); Christou et al., PlantPhysiol. (1988) 87:671-674 (soybean);
McCabe et al., BiolTechnology (1988) 6:923-926 (soybean); Finer and McMullen, In Vitro Cell Dev. Biol. (1991) 27P: 175-182 (soybean); Singh et al., Theor. Appl.
Genet.
(1998) 96:319-324 (soybean); Datta et al., Biotechnology (1990) 8:736-740 (rice);
Klein et al., Proc. Natl. Acad Sci. USA (1988) 85:4305-4309 (maize); Klein et al., Biotechnology (1988) 6:559-563 (maize); Tomes, U.S. Patent No. 5,240,855;
Buising et al., U.S. Patent Nos. 5,322, 783 and 5,324,646; Tomes et al. (1995).
"Direct DNA
Transfer into Intact Plant Cells via Microprojectile Bombardment," in Plant Cell, Tissue, and Organ Culture: Fundamental Methods, ed. Gamborg (Springer-Verlag, Berlin) (maize); Klein et al., Plant Physiol. (1988) 91:440-444 (maize); Fromm et al., Biotechnology (1990) 8:833-839 (maize); Hooykaas-Van Slogteren et al., Nature (London) (1984) 311:763-764; Bowen et al., U.S. Patent No. 5,736,369 (cereals);
Bytebier et al., Proc. Natl. Acad Sci. USA (1987) 84:5345-5349 (Liliaceae). In The Experimental Manipulation of Ovule Tissues, ed. Chapman et al. (Longman, New York), pp. 197-209 (pollen); Kaeppler et al., Plant Cell Reports (1990) 9:415-and Kaeppler et al., Theor. Appl. Genet. (1992) 84:560-566 (whisker- mediated transformation); D'Halluin et al., Plant Cell (1992) 4:1495-1505 (electroporation); Li et al., Plant Cell Reports (1993) 12:250-255 and Christou and Ford, Annals gfBotany (1995) 75:407-413 (rice); Osjoda et al:, Nature Biotechnology (1996) 14:745-(maize via Agrobacterium tumefaciens); all of which are herein incorporated by reference.
The zinc finger protein with optional effector domain can be targeted to a specific organelle within the plant cell. Targeting can be achieved with providing the ZFP an appropriate targeting peptide sequence, such as a secretory signal peptide (for secretion or cell wall or membrane targeting, a plastid transit peptide, a chloroplast transit peptide, a mitochondrial target peptide, a vacuole targeting peptide, or a nuclear targeting peptide, and the like (Reiss et al., Mol. Gen. Genet. (1987) 209(1):116-121; Settles and Martienssen, Trends Cell Biol (1998) 12:494-501;
Scott et al., JBiol Chern (2000) 10:1074; and Luque and Correas, J Cell Sci (2000) 113:2485-2495).
For examples, certain plastid organelle targeting sequences are disclosed in WO 00/12732 (see also de Castro Silva Filho et al., Plant Mol. Biol. (1996) 30:769-1780; Schnell, D.J. et al., JBiol. Chem. (1991) 266(5):3335-3342; and Lamppa et al., JBiol. Chena. (1988) 263:14996-14999). Plastids are a class of plant organelles derived from proplastids .and include chloroplasts, leucoplasts, aravloplasts, and chromoplasts. The plastids are major sites of biosynthesis in plants. In addition to photosynthesis in the chlordplast, plastids are also sites of lipid biosynthesis, nitrate reduction to ammonium, and starch storage. And while plastids contain their own circular genome, most of the proteins localized to the plastids are encoded by the nuclear genome and are imported into the organelle from the cytoplasm.
As described in the Example Section below, a nuclear localization peptide (see Figure 3) was used to target the ZFP-effector fusions to the nucleus.
The modified plant may be grown into plants in accordance with conventional ways (See, e.g., McCormick et al., Plant Cell. Reports (1986) 81-84). These plants may then be grown, and either pollinated with the same-transformed strain or different strains, and the resulting hybrid having~the desired phenotypic characteristic identified. Two or more generations may be grown to ensure that the subj ect phenotypic characteristic is stably maintained and inherited and then seeds harvested to ensure the desired phenotype or other property has been achieved.
The method of the invention is particularly appealing to the plant breeder because it has the effect of providing a dominant trait, which minimizes the level of crossbreeding necessary to develop a phenotypically desirable species which is also commercially valuable. Typically, modification of the plant genome by conventional methods creates heterozygotes where the modified gene is phenotypically recessive.
Crossbreeding is required to obtain homozygous forms where the recessive characteristic is found in the phenotype. This crossbreeding is laborious and time consuming. The need for such crossbreeding is eliminated in the case of the present invention which provides an immediate phenotypic effect.
When the target gene encodes a target protein, the present method can be used to modulate the expression of said encoded target protein. Expression of any target protein can be modulated by the present method in plant cells. The protein whose expression being modulated can be endogenous or exogenous to the plant cell.
The modulation can be activation or inhibition.
In a specific embodiment, the protein whose expression being modulated is an antibody. In another specific embodiment, the protein whose expression being modulated participates in a metabolic pathway or controls a metabolic pathway, e.g., an anabolic or a catabolic pathway. The present method can be used for modulating metabolic pathways of any desirable molecules such as vitamins, taste molecules, e.g., bad taste molecules, anti-oxidants, sugars and ~flavanoids. The metabolic pathway being modulated can be endogenous or exogenous to the plant cell. In still another specific embodiment, target gene encodes a structural protein, e.g., an enzyme or a co-factor in a metabolic pathway, or a regulatory protein. In yet another specific -embodiment, The metabolic pathway being modulated enhances an input or output trait in a plant or seed.
Enzyme activity means herein the ability of an enzyme to catalyae the conversion of a substrate into a product. A substrate for the enzyme comprises the natural substrate of the enzyme but also comprises analogues of the natural substrate, which can also be converted, by the enzyme into a product or into an analogue of a product. The activity of the enzyme is measured for example by determining the amount of product in the reaction after a certain period of time, or by determining the amount of substrate remaining in the reaction mixture after a certain period of time.
The activity of the enzyme is also measured by determining the amount of an unused co-factor of the reaction remaining in the reaction mixture after a certain period of time or by determining the amount of used co-factor in the reaction mixture after a certain period of time. The activity of the enzyme is also measured by determining the amount of a donor of free energy or energy-rich molecule (e.g., ATP, phosphoenolpyruvate, acetyl phosphate or phosphocreatine) remaining in the reaction mixture after a certain period of time or by determining the amount of a used donor of free energy or energy-rich molecule (e.g., ADP, pyruvate, acetate or creatine) in the reaction mixture after a certain period of time.
Co-factor is a natural reactant, such as an organic molecule or a metal ion, required in an enzyme-catalyzed reaction. A co-factor can be regenerated and reused.
Exemplary co-factors include NAD(P), riboflavin (including FAD and FMN), folate, molybdopterin, thiamin, biotin, lipoic acid, pantothenic acid and coenzyme A, S-adenosylmethionine, pyridoxal phosphate, ubiquitinone, menaquinone.
D. Genetically modified plant cells, tissues and seeds In yet another aspect, the present invention is directed to a genetically modified plant cell, which cell comprises an expression system for a zinc finger protein, said zinc forger protein is capable of binding, and preferably specifically binding, to a target nucleotide sequence, or a complementary strand thereof, within a target gene whose expression is to be modulated by said zinc finger protein.
The genetically modified plant cell can comprise any desirable target nucleotide sequence and target gene. Within the genetically modified plant cell, the target nucleotide sequence can be endogenous or exogenous to the targeted gene. The target gene can be endogenous or exogenous to the plant cells. The genetically -modified plant cell can exist in culture or can be contained in an intact plant.
Within the genetically modified plant cell, the zinc forger protein can be used to control the expression of any target genes. In a specific embodiment, the zinc finger protein controls its own expression by binding to a target sequence within..the zinc forger protein gene. In another specific embodiment, the zinc finger protein controls its own expression by binding to a first target sequence within the zinc forger protein gene and controls the expression of the target gene by binding to a second target sequence within the target gene. Preferably, the first target sequence within the zinc finger protein gene is different from the second target sequence within the target gene. The expression of the zinc forger protein gene can be further controlled by second promoter, e.g., an inducible promoter. The zinc forger protein contained in the genetically modified plant cell can contain any number of zinc finger sequence.
Preferably, the zinc finger protein comprises at least two zinc forger sequences, e.g., from about 2 to about 6 zinc finger sequences or from about 3 to about 6 zinc finger sequences., Any plant cell can be genetically modified to comprise an expression system for a zinc finger protein, so that the expression of a target gene is modulated by said zinc finger protein. Exemplary plants include tobacco, tomato, potato, banana, soybean, pepper, wheat, rye, rice, spinach, carrot, maize and corn.
A method to modulate expression in a plant cell is provided, which method comprises culturing the genetically modified plant cell(s). Preferably, such genetically modified plant cells) is cultured in plants.
In a specific embodiment, a transgenic plant cell is provided, which plant cell is transformed with a nucleic acid comprising a functional geminiviral replicase gene operably linked to a fruit ripening-dependent promoter. In another specific embodiment, a genetically modified plant cell is provided, which cell comprises a heterologous zinc finger protein that specifically binds to a target nucleotide sequence in said plant cell wherein said heterologous zinc finger protein is constitutively or inducibly expressed.
A genetically modified plant tissue is also provided herein, which tissue comprises the above-described genetically modified plant cell(s).

A genetically modified plant seed, e.g., a tobacco, tomato, potato, banana, soybean, pepper, wheat, rye, rice, spinach, carrot, maize and corn seed, is further-provide herein, which seed comprises the above-described genetically modified plant cell(s).
S A transgenic plant seed is further provide herein, which seed is transformed with a nucleic acid comprising a functional geminiviral replicase gene operably linked to a fruit ripening-dependent promoter.
In a specific embodiment, the genetically modified plant cell, tissue, seed and whole plant does not contain a zinc finger protein that is disclosed in U.S.
Patent No.
~ 6,140,466 or WO 98/54311, e.g., a zinc finger-nucleotide binding polypeptide variant comprising at least three zinc forger modules that bind to a target cellular nucleotide sequence and modulate the transcriptional function of the cellular nucleotide sequence, wherein the amino acid sequence of each zinc finger module that binds a target cellular nucleotide comprises two cysteines and two histidines whereby both cysteines are amino proximal to both histidines and wherein each of three modules of said variant has at least one amino acid sequence modification.
E. Nucleic acids encoding ZFPml, ZFPm2, ZFPm3, ZFPm4 and ZFPAp3 In yet another aspect, the present invention is directed to an isolated nucleic acid fragment, comprising a sequence of nucleotides encoding ZFPml (SEQ ID
N0:14), ZFPm2 (SEQ ID NO:15), ZFPm3 (SEQ ID N0:16), ZFPm4 (SEQ m N0:17) or ZFPAp3 (SEQ ID N0:18). The isolated nucleic acid fragment can be DNA or RNA. An isolated nucleic acid fragment, which is hybridizable to the nucleic acid fragment encoding ZFPml, ZFPm2, ZFPm3, ZFPm4 or ZFPAp3 under low, medium and high stringency condition is also provided. Preferably, the isolated nucleic acid fragment hybridizes to the nucleic acid fragment. encoding ZFPmI, ZFPm2, ZFPm3, ZFPm4 or ZFPAp3 under high stringency condition. Plasmids comprising the nucleic acid fragment encoding ZFPml, ZFPm2, ZFPm3, ZFPm4 or ZFPAp3 and cells comprising such plasmids are further provided. Any suitable cells can be used and preferably, bacterial cells, yeast cells, fungal cells, plant cells, insect cells and animal cells are used.
The nucleic acid fragment encoding ZFPml, ZFPm2, ZFPm3, ZFPm4 or ZFPAp3 can be prepared by any methods known in the art, e.g., recombinant production, chemical synthesis or a combination thereof (See generally, Cu~ent Protocols in Molecular Biology (1998) ~ 20, John Wiley & Sons, Inc; Knorre, Design and Targeted Reactions of Oligonucleotide Derivative, CRC Press, 1994; and Staut (Ed.), Nucleic Acid Chemstry: Improved and New Synthetic Procedures, Methods and Techniques, John Wiley & Sons, Inc., 1978). In a specific embodiment, a method for producing a ZFPml, ZFPm2, ZFPm3, ZFPm4 or ZFPAp3 protein is provided, which method comprises growing the cells harboring plasmids containing the nucleic acid fragment encoding ZFPml, ZFPm2, ZFPm3, ZFPm4 or ZFPAp3 under conditions whereby these zinc forger proteins are expressed by the cell; and recovering the expressed zinc finger protein.
The isolated nucleic acids encoding ZFPml, ZFPm2, ZFPm3, ZFPm4 or ZFPAp3 typically consists of at least 25 (continuous) nucleotides, 50 nucleotides, 100 nucleotides, 150 nucleotides, or 200 nucleotides, or a full-length coding sequence. In another embodiment, the nucleic acids are smaller than 35, 200, or 500 nucleotides in length. Nucleic acids can be single or double stranded. Nucleic acids that hybridize to or are complementary to the foregoing sequences, in particular the inverse complement to nucleic acids that hybridize to the foregoing sequences (i.e., the inverse complement of a nucleic acid strand has the complementary sequence running in reverse orientation to the strand so that the inverse complement would hybridize without mismatches to the nucleic acid strand) are also provided. In specific aspects, nucleic acids are provided which comprise a sequence complementary to (specifically are the inverse complement of) at least 10, 25, 50, 100, or 200 nucleotides or the entire coding region of a ZFPml, ZFPm2, ZFPm3, ZFPm4 or ZFPAp3 coding sequence.
The nucleic acids encoding ZFPml, ZFPm2, ZFPm3, ZFPm4 or ZFPAp3 provided herein include those with nucleotide sequences encoding substantially the same amino acid sequences as found in Figures 6, and those encoding amino acid sequences with functionally equivalent amino acids.
F. ZFPml, ZFPm2, ZFPm3, ZFPm4 and ZFPAp3 proteins Tn yet another aspect, the present invention is directed to a zinc finger protein that is ZFPm1 (SEQ )D N0:38), ZFPm2 (SEQ m N0:39), ZFPm3 (SEQ ID N0:40), ZFPm4 (SEQ m N0:41) or ZFPAp3 (SEQ m N0:42), preferably in combination with positive and negative regulating domains. The ZFPml, ZFPm2, ZFPm3, and ZFPm4 zinc fingers are specific for the MIPS gene, meaning they can specifically and strongly bind to nucleotide sequence within the MIPS gene. ZFPAp3 is designed to bind to AP3 in Arabidopsis. The positive regulatory domain VP64 (Beerli et al.;
Proc. Natl. Acad. Sci. USA (1998) 95:14628-14633) is fused to the C-terminal of each zinc finger domain. The negative regulatory domains SID (mSin3 interaction domain) (Ayer et al., Mol. Cell. Biol. (1996) 16:5772-5781) or SKD (a modified Kruppel-associated box) (Margolin et al., Proc. Natl. Acad. Sci. USA (1994) 91.:4509-4513) are fused to the N-terminal of each zinc finger domain.
In yet another aspect, the present invention is directed to a zinc finger protein comprising a zinc finger nucleic acid binding domain and an effector domain, wherein said effector domain comprises an active domain of a restriction enzyme, an active domain of a nucleic acid modifying protein, e.g., a nucleic acid methylase, a label or a modification.
The zinc finger proteins can be made by any methods known in the art. The zinc finger proteins can be produced by chemical synthesis (see e.g., Fmoc Solid Phase Peptide Synthesis: A Practical Approach, Chan and White (Ed.), Oxford University Press, 2000; Peptide Synthesis Protocols, Vol. 35, Pennington and Dunn (Ed.), Humana Press, 1995; and Chemical Approaches to the Syntlaesis of Peptides and Proteins, Lloyd-Williams et al. (Ed.), CRC Press, Inc., 1997), recombinant production (See generally, Current Protocols in Molecular Biology (1998) ~ 20, John Wiley & Sons, Inc.), or a combination thereof. Preferably, the zinc finger proteins are produced by recombinant production.
Functional fragments, analogs or derivatives of the ZFPml, ZFPm2, ZFPm3, ZFPm4 or ZFPAp3 polypeptides are also provided. Preferably, such fragments, analogs or derivatives can be recognized an antibody raised against a ZFPml, ZFPm2, ZFPm3, ZFPm4 or ZFPAp3 polypeptide. Also preferably, such fragments, analogs or derivatives comprise an amino acid sequence that has at least 60% identity, more preferably at least 90% identity to the ZFPml (SEQ >D N0:38), ZFPm2 (SEQ )D
N0:39), ZFPm3 (SEQ >D N0:40), ZFPm4 (SEQ ID N0:41) or ZFPAp3 (SEQ )D
N0:42) polypeptide (See Figure 6).
G. Antibodies recognizing ZFPml, ZFPm2, ZFPm3, ZFPm4 and ZFPAp3 proteins An antibody that specifically binds to the ZFPml, ZFPm2, ZFPm3, ZFPm4 or ZFPAp3 proteins is also provided. Antibodies, whether polyclonal or monoclonal antibodies, can be raised against the zinc finger proteins by any methods known in the art (see e.g., Antibody Production: Essential Techniques, Delves, Wiley, John 8~ Sons, Inc., 1997; Basic Methods in Antibody Production and Characterization, Howard and Bethell, CRC Press, Inc., 1999; and Monoclonal Antibody Production Techniques and Applications: Hybridoma Techniques, Schook, Marcel Dekker, 1987). These antibodies can be used to assess the expression level and localization of ZFP
protein in cells, e.g., plant cells.
For production of the antibody, various host animals can be immunized by injection with the ZFPml, ZFPm2, ZFPm3, ZFPm4 or ZFPAp3 proteins, or a derivative of the foregoing, such as a cross-linked zinc finger protein. Such host animals include but are not limited to rabbits, mice, rats, and the like.
Various adjuvants can be used to increase the immunological response, depending on the host species, and include but are not limited to Freund's (complete and incomplete), mineral gels such as aluminum hydroxide, surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, dinitrophenol, and potentially useful human adjuvants such as bacille Calmette-Guerin (BCG) and corynebacterium parvum.
For preparation of monoclonal antibodies directed towards ZFPml, ZFPm2, ZFPm3, ZFPm4 or ZFPAp3 proteins or domains, derivatives, fragments or analogs thereof, any technique that provides for the production of antibody molecules by continuous cell lines in culture may be used. Such techniques include but are not restricted to the hybridoma technique originally developed by Kohler and Milstein (Nature 256:495-497 (1975)), the trioma technique, the human B-cell hybridoma technique (I~ozbor et al., Irnntunology Today 4:72 (1983)), and the EBV
hybridoma technique to produce human monoclonal antibodies (Cole et al., in Monoclonal Arttibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96 (1985)). In an additional embodiment, monoclonal antibodies can be produced in germ-free animals (W089/12690). Human antibodies may be used and can be obtained by using human hybridomas (Cote et al., Proc. Natl. Acad. Sci. USA 80:2026-2030 (1983)) or by transforming human B cells with EBV virus in vitro (Cole et al., in Monoclonal Antibodies arid Cancer Therapy, Alan R. Liss, Inc., pp. 77-96 (1985)).
Techniques developed for the production of "chimeric antibodies" (Morrison et al., Proc.
Natl.
Acad. Sci. USA 81:6851-6855 (1984); Neuberger et al., Nature 312:604-608 (1984);
and Takeda et al., Nature 314:452-454 (1985)) by splicing the genes from a mouse antibody molecule specific for the ZFPml, ZFPm2, ZFPm3, ZFPm4 or ZFPAp3 protein together with genes from a human antibody molecule of appropriate biological activity can be used; such antibodies are within the scope of this invention.
Techniques described for the production of single chain antibodies (IJ.S.
patent 4,946,778) can be adapted to produce ZFPml, ZFPm2, ZFPm3, ZFPm4 or ZFPAp3 protein-specific single chain antibodies. An additional embodiment utilizes the techniques described for the construction of Fab expression libraries (Huse et al., Science 246:1275-1281 (1989)) to allow rapid and easy identification of monoclonal Fab fragments with the desired specificity for ZFPml, ZFPm2, ZFPm3, ZFPm4 or ZFPAp3 proteins, or domains, derivatives, or analogs thereof. Non-human antibodies can be "humanized" by known methods (see, e.g., U.S. Patent No. 5,225,539).
Antibody fragments that contain the idiotypes of ZFPml, ZFPm2, ZFPm3, ZFPm4 or ZFPAp3 proteins can be generated by techniques known in the art in accordance with the methods of the present invention. For example, such fragments include but are not limited to: the F(ab')2 fragment which can be produced by pepsin digestion of the antibody molecule; the Fab' fragments that can be generated by reducing the disulfide bridges of the F(ab')2 fragment, the Fab fragments that can be generated by treating the antibody molecular with papain and a reducing agent, and Fv fragments.
In the production of antibodies, screening for the desired antibody can be accomplished by techniques known in the art in accordance with the methods of the present invention, e.g., ELISA (enzyme-linked immunosorbent assay). To select antibodies specific to a particular domain of the ZFPml, ZFPm2, ZFPm3, ZFPm4 or ZFPAp3 proteins, one may assay generated hybridomas for a product that binds to the fragment of a ZFPml, ZFPm2, ZFPm3, ZFPm4 or ZFPAp3 proteins that contain such a domain.
H. Nucleic acids encoding ZFP-SID fusion proteins In yet another aspect, the present invention is directed to an isolated nucleic acid fragment, comprising a sequence of nucleotides encoding a fusion protein comprises a zinc finger domain, e.g., 2C7 and an effector domain of SID
(mSin3A
Interaction Domain). A fusion protein comprising the 2C7 and the SID domains is termed as a 2C7-SID fusion protein. The isolated nucleic acid fragment can be DNA
or RNA. An isolated nucleic acid fragment, which is hybridizable to the nucleic acid fragment encoding the 2C7-SID fiision protein under low, medium and high stringency condition is also provided. Preferably, the isolated nucleic acid fragment hybridizes to the nucleic acid fragment encoding the 2C7-SID fusion protein under high stringency condition. Also preferbaly, the isolated nucleic acid fragment has the nucleotide sequence set forth in SEQ ID NO:S or SEQ ID N0:66. Plasmids comprising the nucleic acid fragment encoding the 2C7-SID fusion protein and cells comprising such plasmids are further provided. Any suitable cells can be used and preferably, bacterial cells, yeast cells, fungal cells, plant cells, insect cells and animal cells are used.
The nucleic acid fragment encoding the 2C7-SID fusion protein can be prepared by any methods known in the art, e.g., recombinant production, chemical synthesis or a combination thereof (See generally, Current Protocols in Molecular Biology (1998) ~ 20, John Wiley & Sons, Inc; Knorre, Design and Targeted Reactions of Oligonucleotide Derivative, CRC Press, 1994; and Staut (Ed.), Nucleic Acid Chemstry: Improved and New Synthetic Procedures, Methods and Techniques, John Wiley & Sons, Inc., 1978). In a specific embodiment, a method for producing a SID fusion protein is provided, which method comprises growing the cells harboring plasmids containing the nucleic acid fragment encoding the 2C7-SID fusion protein under conditions whereby these zinc finger proteins are expressed by the cell;
and recovering the expressed 2C7-SID fusion protein.
The isolated nucleic acids encoding the 2C7-SID fusion protein typically consists of at least 25 (continuous) nucleotides, 50 nucleotides, 100 nucleotides, 150 nucleotides, or 200 nucleotides, or a full-length coding sequence. In another embodiment, the nucleic acids are smaller than 35, 200, or 500 nucleotides in length.
Nucleic acids can be single or double stranded. Nucleic acids that hybridize to or are complementary to the foregoing sequences, in particular the inverse complement to nucleic acids that hybridize to the foregoing sequences (i.e., the inverse complement of a nucleic acid strand has the complementary sequence running in reverse orientation to the strand so that the inverse complement would hybridize without mismatches to the nucleic acid strand) are also provided. In specific aspects, nucleic acids are provided which comprise a sequence complementary to (specifically axe the inverse complement of) at least 10, 25, 50, 100, or 200 nucleotides or the entire coding region of the 2C7-SID fusion protein coding sequence.

The nucleic acids encoding the 2C7-SID fusion protein provided herein include those with nucleotide sequences encoding substantially the same amino ~.cid sequences as found in Figures 6 and 24, and those encoding amino acid sequences with functionally equivalent amino acids.
Any suitable SID domains can be used. Preferably, the SID domain is derived from a SID domain of the MAD 1 protein.
I. ZFP-SID fusion proteins In yet another aspect, the present invention is directed to a ZFP-SID, e.g., 2C7-SID, fusion protein comprising a zinc finger of 2C7 and an effector domain of SID. The negative regulatory domains SID (mSin3 interaction domain) (Ayer et al., Mol. Cell. Biol. (1996) 16:5772-5781) or SKD (a modified Kruppel-associated box) (Margolin et al., Proc. Natl. Acad. Sci. USA (1994) 91:4509-4513) can be fused to the C- or N-terminal of the 2C7 zinc finger domain. Preferably, the SID domain is fused to the N-terminal of the 2C7 zinc finger domain. Also preferably, the 2C7-SID
fusion protein comprises a nuclear localization signal.
The 2C7-SID fusion protein can be made by any methods known in the art. It can be produced by chemical synthesis (see e.g., Frnoc Solid Phase Peptide Synthesis:
A Practical Approach, Chan and White (Ed.), Oxford University Press, 2000;
Peptide Synthesis Protocols, Vol. 35, Pennington and Dunn (Ed.), Humana Press, 1995;
and Chemical Approaches to the Syntlaesis of Peptides arad Proteins, Lloyd-Williams et al.
(Ed.), CRC Press, Inc., 1997), recombinant production (See generally, Current Protocols in Molecular Biology (1998) ~ 20, John Wiley & Sons, Inc.), or a combination thereof. Preferably, the 2C7-SI17 fusion proteins are produced by recombinant production.
Functional fragments, analogs or derivatives of the 2C7-SID fusion proteins or polypeptides are also provided. Preferably, such fragments, analogs or derivatives can be recognized an antibody raised against a 2C7-SID fusion protein or polypeptide.
Also preferably, such fragments, analogs or derivatives comprise an amino acid sequence that has at least 60% identity, more preferably at least 90% identity to the 2C7-SID fusion protein encoded by the nucleotide sequence set forth in SEQ ID
NO:S
or SEQ ID N0:66.
J. Antibodies recognizing ZFP-SID fusion protein An antibody that specifically binds to a ZFP-Sm, e.g., 2C7-Sm, fusion proteins is also provided. Antibodies, whether polyclonal or monoclonal antibodies, can be raised against the 2C7-Sm fusion proteins by any methods known in the art (see e.g., Antibody Production: Essential Techniques, Delves, Wiley, John &
Sons, Inc., 1997; Basic Methods in Antibody Production and Characterization, Howard and Bethell, CRC Press, Inc., 1999; and Monoclonal Antibody Production Techniques and Applications: Hybridoma Techniques, Schook, Marcel Dekker, 1987). These antibodies can be used to assess the expression level and localization of ZFP
protein in cells, e.g., plant cells.
For production of the antibody, various host animals can be immunized by injection with the 2C7-Sm fusion proteins, or a derivative of the foregoing, such as a cross-linked fusion protein. Such host animals include but are not limited to rabbits, mice, rats, and the like. Various adjuvants can be used to increase the immunological response, depending on the host species, and include but are not limited to Freund's (complete and incomplete), mineral gels such as aluminum hydroxide, surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, dinitrophenol, and potentially useful human adjuvants such as bacille Calmette-Guerin (BCG) and corynebacterium parvum.
For preparation of monoclonal antibodies directed towards 2C7-Sm fusion proteins or domains, derivatives, fragments or analogs thereof, any technique that provides for the production of antibody molecules by continuous cell lines in culture may be used. Such techniques include but are not restricted to the hybridoma technique originally developed by I~ohler and Milstein (Nature 256:495-497 (1975)), the trioma technique, the human B-cell hybridoma technique (Kozbor et al., Immunology Today 4:72 (1983)), and the EBV hybridoma technique to produce human monoclonal antibodies (Cole et al., ira Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96 (1985)). In an additional embodiment, monoclonal antibodies can be produced in germ-free animals (W089/12690). Human antibodies may be used and can be obtained by using human hybridomas (Cote et al., Proc. Natl. Acad. Sci. USA X0:2026-2030 (1983)) or by transforming human B
cells with EBV virus in vitro (Cole et al., in Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96 (1985)). Techniques developed for the production of "chimeric antibodies" (Mornson et al., Proc. Natl. Acad. Sci. USA 81:6851-6855 (1984); Neuberger et al., Nature 312:604-608 (1984); and Takeda et al., Nature 314:452-454 (1985)) by splicing the genes from a mouse antibody molecule specific for the 2C7-SID fusion proteins together with genes from a human antibody molecule of appropriate biological activity can be used; such antibodies are within the scope of this invention.
Techniques described for the production of single chain antibodies (U.S.
patent 4,946,778) can be adapted to produce 2C7-SID fusion protein-specific single chain antibodies. An additional embodiment utilizes the techniques described for the construction of Fab -expression libraries (Huse et al., Science 246:1275-1281 (1989)) to allow rapid and easy identification of monoclonal Fab fragments with the desired specificity for 2C7-SID fusion proteins, or domains, derivatives, or analogs thereof.
Non-human antibodies can be "humanized" by known methods (see, e.g., U.S.
Patent No. 5,225,539).
Antibody fragments that contain the idiotypes of 2C7-SID fusion proteins can be generated by techniques known in the art in accordance with the methods of the present invention. For example, such fragments include but are not limited to:
the F(ab')2 fragment.which can be produced by pepsin digestion of the antibody molecule; the Fab' fragments that can be generated by reducing the disulfide bridges of the F(ab')2 fragment, the Fab fragments that can be generated by treating the antibody molecular with papain and a reducing agent, and Fv fragments.
In the production of antibodies, screening for the desired antibody can be accomplished by techniques known in the art in accordance with the methods of the present invention, e.g., ELISA (enzyme-linked immunosorbent assay). To select antibodies specific to a particular domain of the 2C7-SID fusion proteins, one may assay generated hybridomas for a product that binds to the fragment of a 2C7-SID
fusion protein that contains such a domain.
Literature Cited Ayer, D. E., Laherty, C. D., Lawrence, Q. A., Armstrong, A. P. and Eisenman, ' R. N. (1996) Mol. Cell. Biol. 16, 5772-5781 Barbas, C..F., Gottesfeld, J. M., and Wright, P. E. WO 95/19431 Zinc finger protein derivatives and methods therefor Beato, M., Herrlich, P., and G. Schutz Steroid hormone receptors: Many actors In search of a plot Cell 1995 83: 851-857 Beerli, R., Segal, D., Dreier B., and Barbas, C. F. (1998) Proc. Natl. Acad.
Sci.
USA 95, 14628-14633 Byzova, M., Franken, J., Aarts, M., Alineida-Engler, J., Engler, G., Mariani, C., Campagne, M., and Angenent, G. (1999) Arabidopsis STERILE APETALA, a multifunctional gene regulating inflorescence, flower, and ovule development Gene and Development 13~ 1002-1014 Callis, J., Raasch, J. A., and Vierstra, R. D. (1990) Ubiquitin extension proteins of Arabidopsis thaliana J. Biol. Chem. 265, 12486-12493 Calvert, L. A. Ospina, M. D., and Shepherd, R. J. (1995) Characterization of cassava vein mosaic virus: a distinct plant papraretrovirus J. Gen. Virol. 76, Chourey, P. S. and Zurawski, D. B. (1981) Callus formation from protoplasts of a maize cell culture Theor. Appl. Genet. 59, 341-344 1981 Cornejo, M. J., Luth, D., Blankenship, K. M., Anderson, O. D., and Blechl, A.
E. (1993) Activity of a maize ubiquitin promoter in transgenic rice Plarzt Mol Biol 23 (3), 567-81 Gibbs, A. J., (1977) CMIlAAB Descriptiozis of plant viruses 184 Irish, V., and Yamamoto, Y. (1995) Conservation of floral homeotic gene function between Arabidopsis and Arztirrhinum The Plant Cell 7, 1635-1644 Irish, V. F. (1999) Patterning the flower Developnzental Biology 209, 211-220 Jack, T., Brockman, L., and Meyerowitz, E. (1992) The homeotic gene APETALA3 of Arabidopsis thaliana encodes a ADS box and is expressed in petals and stamens Gell 68, 683-687 Jack, T., Fox, G., and Meyerowitz, E. (1994) Arabidopsis homeotic gene APETALA3 ectopic expression: transcriptional and post-transcriptional regulation determine floral organ identity Cell 76, 703-716 Kim, J. and Pabo, C. O. (1998) Getting a handhold on DNA: Design of poly-zinc finger proteins with femtomolar dissociation constants Proc. Natl. Acad.
Sci.
USA 95, 2812-2817 Liu, Q., Segal, D., Ghiara, J., and Barbas, C. F. (1997) Design of polydactyl zinc-finger proteins for unique addressing within complex genomes Proc. Natl.
Acad.
Sci. USA 94, 5525-5530 Luque, C. M. and Correas, I: (2000) A constitutive region is responsible for nuclear targeting of 4.1R: modulation by alternative sequences results in differential intracellular localization J Cell Sci 113, 2485-2495 Mangelsdorf, D. J., Thummel, C., Beato, M., Herrlich, P., Schutz, G., S Umesono, K., Blumberg, B., Kastner, P., Mark, M., and Chambon, P (1995) The Nuclear Receptor Superfamily: the Second Decade Cell 1995 83: 835-839 Margolin, J. F., Friedman, J. R., Meyer, W. K. Vassing, H., Thiesen, H. and Rauscher, F. J. (1994) Proc. Natl. Acad. Sci. USA 91; 4509-4513 '~ Norns, S. R., Meyer, S. E. and Callis, J. (1993) The intron ofArabidopsis thaliana polyubiquitin genes is conserved in location and is a quantitative determinant of chimeric gene expression Plant Mole. Biol. 21, 895-906 Parker, M. G. (1993) Curr. Opin. Cell Biol. S, 499-504 Pineiro, M., and Coupland, G. (1998) The control of flowering time and floral identity in Arabidopsis Plant Physiol. 117, 1-8 1S Reiss, B., Wasmann, C. C., and Bohnert, Ii. J. (1987) Regions in the transit peptide of SSU essential for transport into chloroplasts Mol. Gen. Genet.
209(1), 116-Scott, S.V., Nice, D. C. 3rd, Nau, J. J., Weisman, L. S., Kamada, Y., Keizer-Gunnink, I,. Funakoshi, T., Veenhuis, M., Ohsumi, Y., Klionsky, D. J. (2000) Apgl3p and VacBp are part of a complex of phosphoproteins that are required for cytoplasm to vacuole targeting JBiol Chern 10, 1074 Segal, D., Dreier, B., Beerli, R. R., and Barbas, C. F. (1999) Toward controlling gene expression at will: Selection and design of zinc finger domains recognizing each of the S'-GNN-3' DNA target sequences Proc. Natl. Acad. Sci.
USA
2S 96, 2758-2763 Settles, A. M. and Martienssen, R. (1998) Old and new pathways of protein export in chloroplasts and bacteria Trends Cell Biol 12, 494-SOI
Talamond, P., Gallon, G., and Treche, S. (1998) Rapid and sensitive liquid chromatographic method using a conductivity detector for the determination of phytic acid in food J. of ChromatogYaphy 805, 143-147 Verdaguer, B. (1996) Isolation and expression in transgenic tobacco and rice plants, of the Casseva Vein Mosaic Virus (CsVMV) promoter Plant Mol. Biol. 31, Verdaguer, B. (1998) Functional organization of the Casseva Vein Mosaic Virus (CsVMV) promoter Plant Mol. Biol. 37, 1055-1067 -Weigel, D. (19950 The genetics of flower development: From floral induction to ovule morphogenesis Anrau Rev. Genetics 29, 19-39 Wu, H., Yang, W., and Barbas, C. F. (1995) Building zinc fingers by selection: Toward a therapeutic application Proc. Natl. Aead. Sci. USA 92, 344-Yamamoto, K. R. (1985) Steroid Receptor Regulated Transcription of Specific Genes and Gene Networks Annu. Rev. Genet. 19: 209-252.
Yanofsky, M. (1995) Annu. Rev. Plant Physiol. Plafat Mol. Biol. 46, 167-188 All references cited herein are incorporated~by reference as if each were specifically incorporated by reference.
The following examples are intended to illustrate but not to limit the invention.
K. EXAMPLES
The invention is illustrated by materials and methods for controlling genetic expression in plants, e.g., the luciferase transgene, Arabidopsis AP3 gene;
and the P
maize MIPS gene. "Providing" herein means maintaining a plant cell, either in planta or ex planta, either in a plant or in cell or tissue culture, under conditions in which expression and production of a desired zinc finger protein is produced in the plant cell. The regulatory factor, or "effector protein," will at least comprise a zinc finger component which binds specifically to a nucleotide sequence contained within the target gene such that the binding of the zinc finger protein modulates expression of the target gene (See generally Kim and Pabo; Pr~oc. Natl. Acad. Sci. U.S.A., 95:2812-2817 (1998)) and may also contain a regulatory domain. Thus, the zinc forger protein can be fused to an additional amino acid sequence which provides regulation,.
although as stated above, in some instances, binding of the zinc finger portion alone has an effect on expression. This is especially the case when the binding target is in the TATTA box region where transcription is initiated.
Example 1 ZFP effector fusion protein function on luciferase reporter gene in maize cells To establish this Zinc Finger Protein ("ZFP") technology in plants, we have shown that transient trans-activation and trans-repression of a reporter gene can be achieved in representative plant cells. These transient regulation is mediated by a previously synthesized synthetic zinc finger proteins, 2C7. The binding site of zinc finger protein 2C7 sequence was originally from the type 1 human immune deficiency virus (HIV-1) genome. The synthetic zinc finger protein 2C7 has been show to be able to specifically bind to this site and activate reporter gene's expression in human Hela cell line (Liu et al., Proc. Natl. Acad. Sci. USA (1997) 94:5525-5530;
and Wu et al., Proc. Natl. Acad. Sci. USA (1995) 92:344-348). The claimed method comprises providing plant cells, either in culture or in intact plants, with a functional ZFP, or an expression system for production of a suitable ZFP. However, by "culturing" is meant maintaining a plant cell, either in planta or ex planta, either in a plant or in cell or tissue culture, under conditions in which expression and production of a desired zinc finger protein is produced in the plant cell.
A. Reporter constructs Luciferase reporter plasmids were constructed from the Cassava Vein Mosaic Virus promoter (CsVMV) (SEQ ID NO:1) (Calvert et al., J. Gen. Virol. (1995) 76:1271-1278; Verdaguer et al., Plant Mol. Biol. (1996) 31:1129-1139, Verdaguer et al., Plant Mol. Biol. (1998) 37:1055-1067). Six tandem repeats of 2C7 binding sites (6X2C7) (SEQ ID N0:2) was inserted at the upstream of reporter. Reporter I
(p5'C7F) is a deleted and thus inactive version of promoter CsVMV which contains nucleotide sequence -112 by to +72 by of the full length CsVMV promoter with 2C7 binding site is inserted at the 5' end. Reporter II (pc7rbTATA) contains a minimal promoter (SEQ ID N0:3) with 6X2C7 binding site inserted at the 5' end.
Reporter I contains a longer promoter sequence and is stronger than the promoter in reporter I (Fig. 1).
B. Effector constructs Maize ubiquitin promoter ZmUbi (Cornejo et al., Plant Mol Biol (1993) 23(3):567-81) was used to express the ZFP-effector fusion proteins. The activation construct (pND3008) (SEQ ID N0:4) consists of several functional domains: zinc finger protein domain 2C7, nuclear localization signal, the transcriptional activation domain VP64 which is fused to the relative C-terminal of ZFP2C7 and HA epitope tag sequence (Fig. 1). The function of HA tag is for the detection of zinc finger protein expression through Western procedures. The repression construct (pND3018) (SEQ ID NO:S) consists of repression domain SID, nuclear localization signal, ZFP2C7 domain, and HA epitope tag sequence. The repression domain SID is fused to the relative N-terminal of ZFP2C7 domain.
C. _Transient assay Maize protoplasts were prepared from maize cell line HE89 (F19556) and transformed using standard procedure (Chourey and Zurawski, TheoY. Appl.
Genet.
59:341-344 (1981)). For each transformation reaction, lug of luciferase reporter constructs and l0ug activator were co-transformed into one million purified protoplasts. The transformed protoplasts were incubated at 28°C in dark for 24 - 48 hrs. Collect cells by centrifugation at SOOg for Smin. Aspirate off media.
Resuspend in 80u1 1.2x Passive Lysis Buffer (Promega). Freeze at -80°C for >1 Omin. Thaw completely at room temp. Vortex and spin at 3500RPM for Smin at 4°C.
Collect supernatant. Assay 20u1 extract for luciferase with the Luciferase Assay Kit (Promega).
The luciferase activity of the target reporter is normalized to the protein contents of each sample to generate the specific activity. This will correct for any variations in transformation efficiency and cell extractions.
D. Function of ZFP effector fusion protein in maize protoplasts The effects of the 2C7-VP64 ZFP-activator fusion was tested on Reporter I and II reporters (Fig. 1). With Reporter I (p5'c7F), 146 to 250 fold of activation is achieved with the 2C7-Vp64 activator. With Reporter II (pc7rbTATA), 40 to 70 fold of activation is achieved with the 2C7-Vp64 activator.
The effect of the 2C7-SID ZFP-repressor fusions on tested on Reporter I. The 2C7-SID effector produces almost 2 fold of repression.
Example 2 Position effect of zinc finger protein binding site on the activation of luciferase reporter constructs in tobacco cells A. R~orter constructs A series of luciferase reporter plasmids were constructed from the full length and partial deletion version of Cassava Vein Mosaic Virus promoter (CsVMV
promoter with six tandem repeats ofl8bp 2C7 (GCG TGG GCG GCG TGG GCG) (SEQ ID N0:43) binding sites (6X2C7) inserted at different locations within fragments of this promoter (Fig. 2) (Verdaguer et al., Plant Mol. Biol. (1998) -37:1055-1067). Plasmid pAluc contains a full-length CsVMV promoter (same as in pND3008, see SEQ ID NO:1) driving the expression of luciferase gene. Plasmid p5'C7A contains the same full-length CsVMV promoter with 6X2C7 inserted at the 5' end using Xba I site. Plasmid p3'C7A is almost identical to p5'C7A except the 6X2C7 was inserted at the 3' end using Xba I site. Plasmid pC78E contains the deleted CsVMV promoter with 6X2C7 binding site replacing nucleotide sequence -112 to -63 of CsVMV. Plasmid p5'C7C contains a partial deleted CsVMV (-222 to +72) with 6X2C7 inserted at the 5' end. Plasmid p5'C7D contains a partial deleted CsVMV (-178 to +72) with 6X2C7 inserted at the 5' end. Plasmid p5'C7F and pc7rbTATA are the same in example 1A. Plasmid prbTATA is identical to pc7rbTATA except no 6X2C7 binding site. The promoter activity in each construct is further decreased than the previous version as the promoter sequence gets shorter. For example, the activity of p5'C7A is higher than p5'C7C, p5'C7D is higher than p5'C7F, and their activities are all higher than the minimal promoter construct pc7rbTATA.
B. Effector constructs Tobacco mosaic virus (TMV) (Gibbs, CMIlAAB Descripti~ns of plant viruses (1977) 184) was used as an expression vector for the ZFP-effector fusion proteins.
The coding region for the coat protein was replaced by the ZFP fusion sequence (Fig.
3). Plasmid p2C7-SID consists of TMV promoter and SDI-2C7 repression domain (same as in pND3018). Plasmid p2C7-VP64 consists of TMV promoter and 2C7-VP64 activation domain (same as in pND3008). Plasmid pC7-VP64 is similar to p2C7-VP64 except the 6 finger zinc finger protein 2C7 (in pND3008 and pND3018) is replaced by 3 finger protein C7 (SEQ ID N0:6). Plasmid pC7-GFP consists TMV
promoter and GFP fragment (Ref). It was used as background control.
Arabidopsis ubiquitin promoter UBQ3 was also used to express the ZFP fusion proteins(constructs did not show here and the results are similar).
C. Transient assay TMV viral RNA was made from the effector constructs with the T7 Megascript Kit (Ambion). Collect log phase tobacco BY-2 cells in conical tubes.
Wash with 0.4M mannitol. Digest cell walls by adding 20 ml Enzyme Solution (1%

Cellulase Onezuka RS (Karlan Research USA #2019), 0.1 % Pectolyase Y-23 (Karlan Research USA #8006), 0.4M mannitol, pH 5.8). Incubate in deep petri dish (NUNC
4031) at 22-28°C in dark with continuous or occasional swirling for 2hrs. Wash 2x with 0.4M mannitol. Collect by centrifugation at 300g for 2min. Remove as much supernatant as possible and resuspend in 15-25m1 Electroporation Buffer (0.3M
mannitol, SmM MES, 70mM KCl, pH 5.8). Count cells on hemacytometer and adjust concentration to 1.6x1061m1. Add 0.3m1 cells to a 0.4cm electroporation cuvette. Add 100u1 Electroporation Buffer containing lug reporter plasmid DNA, O.lug/ul Renilla Luciferase (RL) control plasmid with no ZFP binding sites, and 25ug viral RNA, mix gently with pipette, and immediately electroporate (R=~ C=125uF, V=300V).
Incubate on ice for 30min. Incubate at RT for Srnin. Transfer 100u1 eletroporated cells to 3m1 Protoplast Culture Media (2.2g/1 Murashige and Skoog Plant Salt Base-Gibco BRL #11117-074, O.lg/1 myo-inositol, lmg/1 thiamin-HCl, 0.2mg/12,4-D, l Og/1 sucrose, 0.4M mannitol, and pH 5.8). Incubate cells in 3m1 media in 15m1 conical tube placed on side or in 1-2ml media in a small petri dish (Coming #25050-35). Incubate at 28°C in dark for 48 hrs. Collect cells by centrifugation at SOOg for Smin. Aspirate off media. Resuspend in 80u1 1.2x Passive Lysis Buffer (Promega).
Freeze at -80°C for >1 Omin. Thaw completely at room temp. Vortex and spin at 3500RPM for Smin at 4°C. Collect supernatant. Assay 20u1 extract for luciferase and RL activity with the Dual Luciferase Assay Kit (Promega).
In this transient assay, the luciferase activity of the target reporter is normalized to the RL activity of the control reporter by division. This will correct for any variations in transformation efficiency and cell extract concentration.
The normalized activity of an effector is presented as the normalized activity of reporter plus effector relative to the activity of the target reporter without effector expression.
Thus, a relative activity of 1 means there is no effector function.
D. Effect of binding site position and ZFP-activator fusion protein function in tobacco protoplasts The effects of the C7-VP64 and 2C7-VP64 ZFP-activator fusions were tested on these reporters (Fig. 4). The results show zinc finger domain 2C7-VP64 fusion protein activated the expression of luciferase reporter gene in both tobacco cells and in maize cells. The activation level was dependent on the position of zinc finger protein binding sites. Generally, the closer of the binding site is to the TATA box, the higher of the activation level. Three-finger zinc finger protein (C7) generally gives higher activation level than the six-finger zinc finger protein (2C7). It is probably due to the fact of doubling the total numbers of activator when a 3-finger protein was used.
With a minimal promoter, Reporter II (pc7rbTATA), almost 100-fold of activation is achieved with the C7-Vp64 activator. The 2C7-VP64 activator gives 23-fold of activation. A stronger promoter, p5'c7F (Reporter I), is activated 10 fold by and 5 fold by 2C7-VP64. From this experiment, it is clear that the zinc finger-binding site is preferred upstream of TATA box.
E. Effect of ZFP-r~ressors in tobacco protoplasts To determine whether repression can be achieved, the effects of the 2C7-SID, 2C7-SKD, and e2c-SID ZFP-repressor fusions on three different reporter plasmids:
Reporter I (p5'C7F), Reporter II (pc7rbTATA), and pC78E. Where studied. The 2C7-SID effector produces almost 5-fold repression of all the reporters (Fig.
S). The 2C7-SKD shows no specific repression of any of the reporters. Similarly, the e2c-SID repressor does not cause much repression of the reporters (Fig. 5).
Thus, ZFP fusions to the SID repressor domain provide effective, specific repression of their target genes. However, the repression domain SKD and the e2c zinc finger protein are not functional in plant cells.
Example 3 Selection of zinc finer protein binding sites for endogenous gene MIPS and AP3 The genes for AP3 (U30729) and MIfS (AF056326) were examined for suitable zinc finger binding sites. At the time of construction, the zinc finger library covered all the GNN triplets and the TGA triplet as shown in the recognition code provided in Table 2) (see also Segal et al., Proc. Natl. Acad. Sci. Z7SA
(1999) 96:2758-2763. Potential zinc finger-binding sites were selected by searching for 6 triplets (18 consecutive base pairs) with either GNN or TGA.
Table 2. GNN triplets and the TGA triplet recognition code Target Sequence ZFP domain GAA SQSSNLV (SEQ ID N0:44) GAC SDPGNLV (SEQ ID N0:45) GAG SRSDNLVR (SEQ ID N0:46) GA STSGNLV (SEQ ID N0:47) GCA SQSGDLRR (SEQ m N0:48) GCC SDCRDLAR (SEQ m N0:49) GCG SRSDDLVR (SEQ m N0:50) GCT STSGELV (SEQ m N0:51) GGA SQSSHLVR (SEQ m N0:52) or SQRAHLER (SEQ m N0:53) GGC SDPGHLVR (SEQ m N0:54) GGG , SRSDKLVR (SEQ m N0:55) GGT STSGHLVR (SEQ DJ N0:56) GTA SQSSSLVR (SEQ m N0:57) GTC SDPGALVR (SEQ m N0:58) GTG SRSDVLVR (SEQ m N0:59) or SRI~DSLVR (SEQ B7 N0:60) GTT STSGSLVR (SEQ ID N0:61) TGA SQAGHLAS (SEQ m N0:62) For the AP3 gene, only one target site was identified as a possible ZFP
binding site using the current code. This site (5'-tac ttc ttc aac tcc atc-3') was located from -112 to -95 relative to the start of translation, about 70 base pairs upstream of the start of transcription. ZFPAp3 was selected to bind the compliment of this sequence (5'-GAT GGA GTT GAA GAA GTA-3') (SEQ m N0:7). Of course, with a complete code falls in place in combo with present leadings, any sequence can be targeted.
For the MIPS gene, there were two possible regions in which ZFP could be targeted using current code. One was at the very 5' end of the cDNA from position -85 to -65 relative to the translational start site (5'-GCC TCC TTC CTC CTC TCA
CTC-3') (SEQ m N0:8). Two possible zinc fingers could be made to this site.
ZFPml binds the compliment of this sequence from -68 to -85 (5'- TGA GAG GAG
GAA GGA GGC-3') (SEQ m N0:9) and ZFPm2 binds from -65 to =82 (5'- GAG
TGA GAG GAG GAA GGA-3') (SEQ ID NO:10) of the compliment. Another location for ZFP binding sites was within the translated region of the gene from 294 to 317 (5'-GCC AAC TAC TAC GGC TCC CTC ACC-3') (SEQ m NO:11) after the translational start site. Two ZFPs were selected to bind the compliment of this site.
ZFPm3 binds from 311 to 294 (5'- GGA GCC GTA GTA GTT GGC-3') (SEQ m N0:12) and ZFPm4 binds from 317 to 300 (5'- GGT GAG GGA GCC GTA GTA-3') (SEQ m N0:13). All four of these zinc finger proteins of the six-finger (hectadactyl) type were constructed. The activity of these different zinc fingers when fused to transcriptional regulatory domains should provide us with more information about the optimal binding position. This information will aid in selecting binding sites for new target genes.
Exam,~le 4 Construction of new zinc finger protein ZFPAp3 ZFPml ZFPm2, ZFP
and ZFPm4 Since the specificity of the ZFP is determined by the DNA binding alpha helix, polydactyl zinc finger proteins with novel DNA specificity can be constructed by modifying the recognition helices of existing zinc finger proteins. A human zinc finger protein SplC has been selected to serve as a framework in the present example. It has been demonstrated that the SplC protein can provide a good framework for zinc finger domain modification (Beerli et,al., Proc. Natl.
Acad. Sci.
USA (1998) 95:14628-14633).
New zinc finger proteins were constructed in two steps. First, three-finger ZFPs are constructed by PCR from overlapping oligonucleotides. These three-finger ZFPs are then fused together to create a six-finger (polydactyl) zinc finger protein to bind a specific l8bp sequence. PCR construction of three-finger proteins was carried out in two sequential PCR reactions. In PCRl, the F2-b (finger 2-backward) and F2-f (finger 2- forward) primers were used as a template for PCR extension reaction with the F1-fl (forger 1- first forward) and F3-bl (finger 3- first backward) primers.
For example, in a 100u1 reaction, lug F1-fl, lug F3Tbl, O.lug F2-f, O.lug F2-b, 8u12.5mM dNTP mix, 10u1 l Ox Taq Buffer with lSmM MgCl2 (Perkin Elmer), O.SuI Su/ul AmpliTaq Gold (Perkin Elmer). PCR conditions 1) 94°C for 30sec; 2) 9~°C for 30sec; 3) 60°C for 30sec; 4) 72°C for 30sec; 5) repeat steps 2-4, 25 times; 6) 72°C for lmin. In the second reaction, PCR2, lul of the PCR extension product from PCRl is used as a template for PCR with F1-f2 (finger 1 second forward) and f3-b2 (finger 3 second backward) to extend the construct. In a 100u1 reaction, lug F1-fZ, lug F3-b2, lul PCRl reaction, 8u12.5mM dNTP mix, 10u1 lOx Taq Buffer with lSmM MgCl2 (Perkin Eliner), O.Sul Su/ul AmpliTaq Gold (Perkin Elmer). Amplify with same conditions used in PCRl. The 320bp PCR product was digested with SfiI
and the 300bp fragment was isolated and ligated into a derivative of the pMal-vector (New England Biolabs). When transformed into XLl-Blue E. coli, this vector provided IPTG-inducible expression of the zinc finger fused to the Maltose Binding Protein (MBP). The MBP fusion allows easy purification and detection of the zinc finger protein. Similarly, the second half of zinc finger protein domain (also ' containing three fingers) was synthesized using PCR extension reaction.
The following provides details of a typical construction of a zinc finger, in this case, construction of ZFPm2: The following primers were used to construct the ZFPm2a protein: F1-fl (ggt aag tcc ttc AGC CGC AGC GAT AAC CTG GTG CGC
cac cag cgt acc cac acg ggt gaa aaa ccg tat aaa tgc cca gag), Fl-f2 (gag gag gag gag gtg gcc cag gcg gcc ctc gag ccc ggg gag aag ccc tat get tgt ccg gaa tgt ggt aag tcc ttc AGC CGC AGC), F2-f (GC CAG GCC GGC CAC CTG GCC AGC cat caa cgc act cat act ggc gag aag cca tac aaa.tgt cca gaa tgt ggc), F2-b (GCT GGC CAG GTG
GCC
GGC CTG GCT aaa aga ttt gcc gca ctc tgg gca ttt ata cgg ttt ttc acc), F3-bl (CCG
GAC GAG ATT GTC AGA CCG AGA gaa aga ctt gcc aca ttc tgg aca ttt gta tgg c), F3-b2 (gag gag gag gag ctg gcc ggc ctg gcc act agt ttt ttt acc ggt gtg agt acg ttg gtg CCG GAC GAG ATT GTC AGA CCG). These primers were used as described above to generate the 3-finger protein, ZFPm2a (SEQ ID N0:63):
1 gaggaggagg aggtggccca ggcggccctc gagcccgggg agaagcccta 50 51 tgcttgtccg gaatgtggta agtccttcAG CCGCAGCGAT AACCTGGTGC 100 101 GCcaccagcg tacccacacg ggtgaaaaac cgtataaatg cccagagtgc 150 1 S 1 ggcaaatctt ttAGCCAGGC CGGCCACCTG GCCAGCcatc aacgcactca 200 201 tactggcgag aagccataca aatgtccaga atgtggcaag tctttcTCTC 250 251 GGTCTGACAA TCTCGTCCGG caccaacgta ctcacaccgg taaaaaaact 300 301 agtggccagg ccggccagct cctcctcctc 330 This protein has the framework of the SplC zinc finger protein (lower case sequences) with the DNA recognition helices replaced with the appropriate sequences (upper case sequences) to generate a zinc finger with a new binding specificity (5'-GAG TGA GAG). A similar set of primers was used to generate the ZFPm2b (SEQ
ID N0:64) protein:
1 gaggaggagg aggtggccca ggcggccctc gagcccgggg agaagcccta S 1 tgcttgtccg gaatgtggta agtccttcTC TCAGAGCTCT CACCTGGTGC
~3 101 GCcaccagcg tacccacacg ggtgaaaaac cgtataaatg cccagagtgc 1:
151 ggcaaatctt ttAGCCAGTC CAGCAACCTG GTGCGCcatc aacgcactca 2~
201 tactggcgag aagccataca aatgtccaga atgtggcaag tctttcTCTC 2:
251 GGTCTGACAA TCTCGTCCGG caccaacgta ctcacaccgg taaaaaaact 3~
301 agtggccagg ccggccagct cctcctcctc 3 This protein also has a SplC framework with altered recognition helices to bind (5'-GAG GAA GGA-3'). The first set of three finger protein (ZFPa) was fused to C-terminal to the second set of three finger protein (ZFPb) by ligating the ZFPa SpeIiXmaI 0.3kb fragment and ZFPb XhoI-BsrFI 0.3kb fragment into the pMal-C2 vector digested with XhoI and SpeI. The resulting six-finger zinc finger protein should specifically bind the target 18 by sequence 5'-GAG TGA GAG GAG GAA
GGA-3' (SEQ ID N0:65).
Using the same method, three-finger proteins with a SplC framework were made to bind the ml, m3, m4, and Ap3 target sites. These three-finger proteins were fused together to create the six-finger zinc finger. Restriction sites were designed in to allow ligation of the fragments. The five constructs that carried the ZFP
genes were named as pMAL-ml, pMAL-m2, pMAL-m3, pMAL-m4, and pMAL-Ap3. The coding regions are the 500 by fragment from Sfi digestion of each pMal plasmid and are named ZFPml (SEQ ID NO:14), ZFPm2 (SEQ ID NO:15), ZFPm3 (SEQ ff~
N0:16), ZFPm4 (SEQ ID NO:17), and ZFPAp3 (SEQ ID N0:18).
Example 5 Expression and Purification of ZFPAp3 ZFPm1 ZFPm2, ZFPm3, and ZFPm4 proteins in E. coli A. Pr~aration of zinc finer protein crude extracts Zinc Finger Protein fusions were prepared as follows, primarily to obtain material for use in raising anti-ZFP antibodies and to assess binding affinity and specificity in vitro.
E, coli (XLl-Blue and (K12TB1) containing the zinc finger expression plasmid was first collected for each construct. Grow 3m1 culture of bacteria overnight in SB (1 Og/1 MOPS, 30g/1 Bacto Peptone, 20g/1 Yeast extract) + SOug/ml Carbenicillin + 1% Glucose at 37°C. Start Sml culture w/ 0.25m1 of overnight culture in SB + SOug/ml Carbenicillin + 0.2% Glucose + 90um ZnCl2 at 37°C for 2hrs. Add IPTG to a concentration of 0.3mM. Shake at 37°C for 2hrs. Pellet bacteria at 2~OOg for Smin. Resuspend pellet in 0.3m1 Zn Buffer A (lOmM Tris base, 90mM KCI, 1mM MgCL2, pH 7.5) +SmM DTT. Freeze suspension at -80°C. Freeze thaw 6 times between dry ice/ethanol bath and 37°C water bath. Spin 14K for Smin at 4°C.
The supernatant is then ready for ELISA assay.
B. Purification of ZFPA~3 ZFPml ZFPm2 ZFPm3 and ZFPm4 All five zinc-finger proteins were affinity purified using New England Biolab's pMAL protein fusion purification Kit. One hundred ml of E. coli (K12TB1) that carried construct pMAL-ml, pMAL-m2, pMAL-m3, pMAL-m4, and pMAL-Ap3 separately was grown at 30°C for overnight under the selection of Carbenicillin (same condition as above). Cells were collected through centrifugation and lysed through brief sonication on ice. The ZFPml-MBP fusion protein was purified from this total lysate using New England Biolab's Kit (#800). The purified samples were quantitated and loaded on SDS-PAGE to estimate the quality. All five proteins were shown as single band on SDS-PAGE gel.
C. ZFP antibod~production Two antibodies were generated from the purified zinc forger proteins.
Antibody I was made against the ZFPml antigen. Antibody II was made against an equal mixture of the ZFPm3 and ZFPm4 antigens. Both antibodies were tittered using ELISA procedure and Western Blots of zinc finger protein expressing cells.
Example 6 Characterization of DNA binding specificit~of ZFPAp3 ZFPml ZFPm2 ZFPm3, and ZFPm4 Zinc finger protein can be characterized by any methods known in the art. For example, the zinc finger protein can be characterized by in vitro assay such as ELISA
and in vivo assay such as gel shifting assay. ELISA assay can be used to characterized biding specificity of a particular zinc finger protein for a variety of target nucleotide sequences. Gel shifting assay can be sued to characterize binding affinity, i.e., obtaining binding constant, of a particular zinc finger protein for a particular target nucleotide sequence..

In this study, crude extracts of the three-finger and six-finger proteins were used for ELISA assay to evaluate the newly synthesized zinc fingex protein's DNA
binding specificity. The assay was also repeated with the purified six-finger proteins.
A. ELISA procedures Coat ELISA plates (Costar 3690) with streptavidin. Use 25u1/well of 8ug/ml streptavidin in PBS. Incubate at 37°C for lhr. Wash 2x with dH20. Bind wells with 25u1/well of 4ng/ul biotinylated target oligonucleotide in PBS. Incubate at 37°C for lhr. Wash 2x with dH20. Block with I75ullwell of 3% BSA in Zn Buffer A.
Incubate at 37°C for lhr. Tap plates dry. Perform 1:2 serial dilutions of extract in Binding Buffer (1%BSA, SmM DTT, .l2ug/ul Salmon Sperm DNA in ZnBA).
Usually from 1:4 to 1:512 for mufti-target ELISA or 1:4 to 1:32 for screening clones for specific DNA binding activity. Incubate lhr at room temperature. Wash.8x with dH20. Add primary antibody: 25u1/well of 1:1000 dilution of mouse anti-MBP
(Sigma M-6295) in 1%BSA ZnBA (no DTT). Incubate 30min at room temperature.
Wash 8x with dH20. Add secondary antibody: 25u1/well of 1:1000 dilution of goat anti-mouse/Alkaline Phosphatase (Pierce 31324) in 1%BSA ZnBA (no DTT).
Incubate 30min at room temperature. Wash 8x with dH20. Develop with 25u1 developer (1 tablet of Sigma 104 Phosphatase Substrate (Sigma 104-105)/ Sml AP
Developing Buffer [100m1 diethanolamine+O.OSg MgCl2 + 97.5 Sodium Azide, adjust to SOOmI with ddH20, pH 9.88]). Incubate at room temperature for 5-60min until color reaction is yellow. Read A405 with a plate reader.
B. ZFP specificity evaluation Hairpin oligonucleotides with a 5' biotin conjugate were synthesized. The oligo m12 (5'-Biotin-GGa gcc tcc ttc ctc ctc tca ctc GGG TTTT CCC gag tga gag gag gaa gga ggc tCC-3') (SEQ ID N0:19) has the target sites for ZFPml and ZFPm2.
Oligo m34 (5'-Biotin-GGa gcc aac tac tac ggc tcc ctc acc GGG TTTT CCC ggt gag gga gcc gta gta gtt ggc tCC-3') (SEQ ID N0:20) has the target sites for ZFPm3 and ZFPm4. The oligo Ap3 has the target site for the ZFPAp3 protein (5'-Biotin-GGt tac ttc ttc aac tcc atc GGG TTTT CCC gat gga gtt gaa gaa gta aCC-3') (SEQ ID
N0:21) .
The individual three-finger ZFPs were evaluated on ELISA with the m12, m34, and Ap3 target oligos. In every case, the three-finger proteins bound their target oligo with significantly higher affinity than the non-target oligos. The eight non-target oligos are: NRI-1 (SEQ m N0:22), NRI-2 (SEQ 117 N0:23), hHD-I (SEQ DJ
N0:24), hHD-II (SEQ ID N0:2S), cScl-g (SEQ ID N0:26), c5p3-g (SEQ ID N0:27), B3c2 (SEQ ID N0:28), and e2c-g (SEQ ID N0:29). All five six-finger proteins, ZFPml, ZFPm2, ZFPm3, ZFPm4, and ZFPAp3 were tested more extensively by ELISA with the m12, m34, Ap3, and the above eight non-target oligos. The six-finger proteins bound their targets better than any of the non-target oligos.
ZFPml, ZFPm4 and ZFPAp3 showed even higher specificity than ZFPm2 and ZFPm3 (Fig. 7-11). M12 site is a sequence that contains overlapping ml and m2 binding sites:
Similarly, m34 contains overlapping m3 and m4 sequences.
To evaluate the specificity of new zinc finger protein, the affinity of each protein was determined for binding to the target sequence and several non-target sequences at appropriate protein concentration. For example, to protein ZFPml and ZFPm2, the non-target oligoes are: Oligo m34 (SEQ ID N0:20), Oligo Ap3 (SEQ ID
N0:21), NRI-1 (SEQ ID N0:22), NRI-2 (SEQ ID N0:23), hHI~-I (SEQ ID N0:24), hHD-II (SEQ ID N0:25), c5c1-g (SEQ ID N0:26), c5p3-g (SEQ ID N0:27), B3c2 (SEQ ID N0:28), and e2c-g (SEQ ID N0:29). Similarly, to protein ZFPm3 and ZFPm4, the non-target oligoes are Oligo m12 (SEQ ID N0:19), oligo Ap3 (SEQ ID
N0:21), and the eight other oligoes. To protein ZFPAp3, the non-target oligoes are Oligo m12, oligo m34 (SEQ ID N0:20), and the eight other oligoes. The affinity reading for target oligo was further normalized to the no oligo control which has the least affinity. Table 3 showed the relative affinity of each zinc finger protein to the listed 11 oligo sequences. This relative affinity represents the fold of specificity of each protein to its target (specific or non-specific). The higher number of the fold of specificity means the more specific of this protein to that particular oligo.
For ZFPml, ZFPm2, ZFPm3, ZFPm4, and ZFPAp3, the fold of specificity to its own target oligo are 18, 23, 13, 16, and 27, respectively, the highest number among all 11 oligoes tested. The result indicates that the six-finger protein, as well as the three-finger proteins, bound their target oligo with significantly higher affinity than the non-target oligoes in every case.

...

.--..-.

cn N N N 01 ~n M

U

N

M
d' M N M

b0 i M M M M N N

U

i U
I~ N M M N

U

~ H

_ U

Wit'N M N N

'.

U ,.., .

U _ O

bO

N ~ v~ M .~ N N

H ~

~rj N

U d' N ~ M ~t ~

H

v7 I~ d ~ 00 M M ~C O

r~ r-1 ~ 0 N M ,~

~

M
M l~ N

O ~ N O

~

M W .r .--, 'r ~ 'r M

y Q N N N N

Refinements in binding affinity and specificity can be further obtained by random mutagenesis, as by PCR mutagenesis to introduce mutations in the ZFP-to improve ZFP specificity or affinity.
C. DNA binding affinit~of ZFPml ZFPm2, ZFPm3, ZFPm4, and ZFPAp3 The affinity of the purified six-finger proteins was measured using gel shift assay. The target oligos used in the ELISA assays were radioactively labeled using [~y32P]-ddATP and terminal transferase. Ten serial dilutions of 1:3 with the purified protein (starting with ~lmg/ml) in binding buffer (1pM labeled oligo, 10%
Glycerol, 0.8% BSA, 0.lug/ul salmon sperm DNA in Zn Buffer A) were performed. The ZFP
was allowed to bind at room temperature for lhr. The samples were then run on 6% non-denaturing PAGE gel in TBE. The gel was dried and exposed with Phosphoimager. The affinity (Kd) of the ZFP was calculated from this gel. The Kd value is the concentration of the protein at which half of the labeled oligo is shifted to a higher molecular weight on the gel by binding to the ZFP. The affinities of these 6-finger proteins were determined to be: ZFPml=2.OnM, ZFPm2=7.5 nM, ZFPm3=
0.18 nM, ZFPm4=0.25 nM and ZFPAp3=2.3 nM (Fig. 12-16). Most of these affinities compare favorably with the existed 6 finger proteins. The 2C7 derivative of the Spl zinc forger has a specificity of 0.46 nM and the e2c zinc forger constructed earlier (Beerli et al., Proc. Natl. Acczcl. Sci. USA (1998) 95:14628-14633) has an affinity of 0.5 nM.
D Evaluation of zinc finer protein ZFPml ZFPm2 ZFPm3 and ZFPm4 in reporter I system (In vivo characterization) To evaluate the in vivo binding ability of the zinc forger proteins, ZFPml, ZFPm2, ZFPm3, and ZFPm4 were cloned into plasmid reporter I system. Two steps are involved: First step is to modify the reporter construct by replacing 2C7 binding site with MIPS gene specific binding site m12 and m34. The second step is to modify the activators (activation constructs) by replacing the 2C7 fragment with each of these four new zinc finger proteins. To evaluate ZFPml, the ZFPm1-VP64 activation construct is co-transformed with ml2-luciferase reporter construct. The luciferase reading from this activation transformation is compared to the BG luciferase activity (no activator). The ZFPm2, ZFPm3, and ZFPm4 are evaluated using the similar steps.
The results show that all four new zinc finger proteins can mediate the activation of reporter gene. In this case, an endogenous target gene sequences were used as a heterologous target gene to activate the protein Luc fusion by piggy backing the VP64 regulator protein activator domain to a ZFP specific for the endogenous target gene sequence. The activation level is comparable to the 2C7 zinc finger protein (Fig. 17).
Example 7 Evaluation of AP3-specific ZFP-activators with transient assays in .
Arabidopsis leaf prOtOplasls A. Transformation of AP3-specific ZFP-activators into Arabidomsis leaf nrotoplasts The ZFPAp3 domain was fused to the N terminal of the VP64 activation domain and then cloned into plant expression vector under the control of the promoter (Fig. 18).
For transient expression of AP3 activator in Arabidopsis protoplasts, cut 10 fully expanded Arabidopsis leaves into lmm strips and add to l Oml enzyme solution (2.5 mg/ml Yakult Macerozyme R10, 15 mg/ml Yakult Cellulase R10, 0.4M
Mannitol, 10 mM MES-KOH (pH 5.6), l9mM CaCl2, 0.35 ul/ml beta-Mercaptoethanol, 0.1% BSA). Incubate 30 min in the dark at room temp under vacuum (run vacuum pump for 3-4 min then hold vacuum and turn off pump).
Incubate 1.5 hours on orbital shaker at room temp 40 rpm, in the dark.
Incubate 5 min on orbital shaker at room temp 80 rpm, in the dark. Filter protoplasts through 70 um nylon mesh. Dilute with 4 mL 200 mM CaCl2. Spin at 60 g 4°C, 5 min.
Remove supernatant. Re-suspend pellet in 3mL wash solution (333 mM Mannitol, 133mM
CaCl2). Spin at 40g for 5 minute 4°C. Remove supernatant. Suspend pellet in 2 mL
WS solution (154 mM NaCI, 125 mM CaCl2, 5 mM KCI, 5 mM Glucose, 1.5 mM
MES pH 5.6 with KOH). Incubate on ice 30 min. Count cells. Spin in 40g, 4°C, S
min. Remove supernatant. Re-suspend to 2 x 106 cells per mL ~ solution (0.6 M Mannitol, 22.5 mM MgCl2, 7.5 mM MES-KOH pH 5.6). Add hug of effector plasmid DNA to a 2m1 centrifuge tube. Add 60 uL protoplast solution. Add 69uL
of a 40% PEG, 400 mM Mannitol, 100 mM Ca(N03)Z solution. Mix thoroughly but gently by rolling tube. Incubate 30 min. at room temp. Dilute with 200 uL WS
solution, mix gently, and incubate 10 min at room temp. Add 400 uL WS mix gently, incubate 10 min. room temp. Add 800 uL W5, mix gently. Spin in microcentrifuge at 60 x g, room temp. for S min. Remove supernatant from all tubes. Re-suspend with S00 uL of 400mM mannitol/WS 4:1 solution. Spin in microcentrifuge~at 60 x g, room temp. for 5 min. Remove supernatant. Re-suspend with 600 uL of culture medium (500 mM Mannitol, 10 mM KCI, 4 mM MES-KOH pH 5.6). Plate in 12 well Falcon culture plates. Incubate at room temp under fluorescent light for 24hrs.
Harvest the protoplasts using a large bore pipette Spin in microcentrifuge at 400g, room temp, 5 mm.
B. RT-PCR analy-s of endogenous gene AP3 expression level Extract RNA with Qiagen Plant RNeasy kit. RTPCR 200ng of RNA with Qiagen 1-step RTPCR kit in a 25u1 reaction [5u1 Sx Buffer, lul dNTP's, 0.125u1 100uM Ap3-F (SEQ ID N0:30) (5'-GGCGAGAGGGAAGATCCAG-3'), 0.125u11 100uM Ap3-4R (5'-CTCCTCTAATACGACTCACTATAGGGACACTCACCTAGCCTCTG-3')(SEQ
ID N0:37), lul Enzyme mix). Thermocycler setting: 50° C 30', 95°
C 15', (94° C
30", 60° C 30", 72° C 1') x39 cycles, 72° C 10'.
C. PCR analysis of effector fusion protein expression level Using the RNA prepared above, the amount of activator expression can also be determined. AP3-specific effector expression can be detected by RTPCR as described above using a generic zinc finger forward primer, NZlibS' (GGCCCAGGCGGCCCTCGAGC) (SEQ ID N0:31) and an Ap3-specific reverse primer Ap3f4-R, (CTAACCAAGGAGCCACTGGTG) (SEQ ID N0:32). Similarly, the m4-specific effector expression can be detected using NZlibS' and a m4-specific reverse primer, m4f3-R (CCTCGCAAGATCACGACAATC) (SEQ ID N0:33).
D. Effects of expressinJ-g ZFPA~3-activator fusion protein on the transcriRtional level of endogenous gene AP3 The AP3 gene is expressed exclusively in the developing flower, so no expression was expected in the leaf derived protoplasts. Three constructs, AP3 activation (pND3014), m4 activation (pND3013) and GFP control (pND0001) were transformed into Arabidopsis leaf cells (protoplasts) respectively. The ZFPm4-specific constructs was used as a control in this experiment to show that the gene specific transcription factors generated with this technology affect only their target gene since the ZFPm4-specific activator (ZFPm4-VP64) should have no effect on the endogenous AP3 gene. The result show that only in the cells that are transformed with AP3 activation constructs there are AP3 transcripts detected (Fig. 19). Thus the ZFP

' ",.,x .".. ~»~G 14 : s~ t-R MOFO SqN D I EGO 858 720 S 125 Tt' X564!#27881;#200 US010181:
04-04-2002 _ ~ , domain is able to direct the activation donain (VP64) to the specific endogenous target gene.
F~camulc $
t r essi d a a a 'vati f 3 c reset n ' 'c ab A. T sfo afro of ific -r si r r tdo is An Agrobacteria transformation vector that contains expression cassette of UBQ3::S1D-ZFPAp3llnos with hygromycin as selection marker was created and named as pND0052. Similarly, expression cassette ofLBQ3::ZFPAp3-VP64/Inos (ZFPAp3 activation construct) was cloned into this Agrobacteria transformation vector as well and __ _ need as pNT)0051 (1~ig:1$). These pTasxriids were then tz~ansforcned into AriFbfdops~r plants using Agrobacteria mediated transformation methods. About 20 transgenic plants were generated.
B. SCR ~alwsis of effeetct~ r n pretein ee~ci sion ~pyel The expression of ZFPAp3 gene in each transgenic event was identified by RT-PCR (Example 7, section G7.
C. R - ~ 's rd us c a re The transcriptional level of endogcnons AP3 gene in each transgenic event was detected by RT PCR (Example ?, section B).
D. antitative al sis ere a ressio eve 't 've P
The TaqMan assay was used for quantifying AP3 gene expression in transformed Arabidopsfs plant: The guantitative PCR probe for AP3 (SIrQ 1D N0:34) and two primers (SEQ ID NO:35 and SEQ >D N4:36) were designed using Primer Express (P'erlan Elmer).
RNA samples were prepared from average of two flower heads using RNAwiz method (Example 7, Section B). TaqMan PCR reaction was done using 250ng total RNA and TaqMan One-step Its'-PCR Master mix reagent. Thcmaal cycling conditions are:
48°C, 30min.; 95°C, l Omin.; then followed by 40 cycles of 95°C,15";
60°C, lmin.. Data were analyzed using relative quantification of the comparative Ct method. For comparative Ct method, thioredoxin3 gene was used as a relative internal standard at this time.

Emvf angst AMENDED SHEET

E. Effects of expressing ZFPAp3-repressor fusion protein on the transcriptional level of endogenous gene AP3 Both RT-PCR and quantitative PCR with AP3 specific primer was performed on these transgenic events (above example 7A). The endogenous gene AP3 expression level has been changed in several events, especially for three events:
ND0052-2e, ND0052-257, and ND0051-la.
Plant ND0052-2e contains very high expression level of ZFPAp3-SID
transgene (Fig. 20A). The transcriptional level of endogenous AP3 gene has been significantly down regulated (Fig. 20B). Quantitative analysis shows a nearly 50 fold of repression was achieved in this plant (Fig. 21).
The transcriptional level of endogenous AP3 gene in Plant ND0052-257 is less repressed compared to plant ND0052-2e, about 5 fold (Fig. 21).
Only one event containing activation construct was confirmed and designated as ND0051-la. Curiously, there was a two-fold activation of the endogenous AP3 gene in this plant (Fig. 21). The phenotype of this plant looks normal. We have not been able to detect the protein expression level in this plant. Considering the significantly less number of activation events that were finally identified (ten to twenty times less than the number of repression events, Table I), it is reasonable to assume that over-activation and over-expression of AP3 gene is toxic to the plant.
This toxic effect could cause lose of transgenic events during selection and regeneration. Consistent with this notion is that our transient regulation data indicates that activation is much more efficient than repression. Expression of ZFPAp3-construct with a weaker promoter or with an inducible promoter would or might increase transgenic events.
F. Phenotype changes caused by expressing ZFPAp3-repressor fusion protein in tran~enic Arabielopsis Among the repression events, there are three independent events that showed significant phenotype changes: flower structures and fertility. They are named as ND0052-2e, ND0052-2d, and ND0052-257. Molecular analysis has been performed on the ND0052-2e and ND0052-257 (Example 8, section E).
The first event, ND0051-2e, did not have a typical flower formation compared to the wildtype flower: the petals are under developed (short and narrow) and partially converted to sepal-like structure; the stamens are much smaller in size compared to the wildtype plant. However, there may be some pullens on the anther. The carpet appears normal except that the size is significantly bigger and developed much faster than the stamens. Generally, the phenotype of this flower is similar to the flower-s from previously characterized mutant ap3 (Jack et al., Cell (1992) 68:683-687, Jack et al., Cell (1994) 76:703-716) and sap (Byzova et al., Gene and Development (1999) 13:1002-1014) but is definitely not identical. This plant is sterile.
Comparing with the morphology of this flower with the wild type flower, it appears that the un proportional development of stamen and stigma is the cause of sterility. More detailed analysis can be carried out in this area.
The second event is named as ND0052-257. This is a sterile plant too. The flower is very similar to flower from ND0052-2e. However, there seems missing one petal in the two flowers that we have dissected.
The third event is named as ND0051-2d. Same as the previous two, it is a sterile plant. There is no silique formation in this plant. The plant died during flowering stage.
Wild type flowers have four organ types (sepal, petal, stamen, carpel) arranged in concentric whorls. The AP3 gene is involved in the determination of organ identity. Misexpression of the AP3 gene results in homeotic mutations where whorl-specific organ identity will be altered. While we expected the phenotypes of the activated and repressed AP3 plants that generated through zinc finger protein technology should mimic the phenotype of previously described AP3 overexpressors, ap3 mutants (Jack et al., Cell (1992) 68:683-687, Jack et al., Cell (1994) 76:703-716), and sap (Byzova et al., Gene and Development (1999) 13:1002-1014), we also think the phenotype can not be identical since the mechanism of repression AP3 gene is different.
. The promoter driving the expression of zinc finger protein is ubiquitin3 (UBQ3) promoter. UBQ3 is a constitutive promoter and the expression pattern and strength is very different from the native AP3 promoter.
We are analyzing more transgenic plants (both repression events and activation events) now. We will choose several plants (4 to 5) with different expression level of zinc finger protein through Western analysis. The level of expression can then be correlated with the floral phenotype and effector expression levels.
AP3 gene is one of the many genes that are involved in floral organ determination (Weigel, Annu. Rev. Genetics (1995) 29:19-39; and Pineiro and r , (~_Qe~-~~2 ' - CA 02396898 2002-07-18 US010181 Coupland, Plant Physiol (1998) 117:108).11 is very likely that there are other genes, especially the down stream genes, that play a role in this very complex process as well. We plan to conduct gene chip assay (Affymetrix) on the final selected AP3 events.
This assay should provide us with more insight into the Moral development and regulation process.
More sterile plants have been identified other than the three events mentioned above. It is estimated that flit chance of getting,stcrile phenotype with our repression construct is about 5 to 10°l0.
a a ' 'on 3 a a ex ress'on ' snit a id sts flo promoter AP3 ~~b f~o~.~ ~~~~A~ ~~~~~) was iso~atsd by PCR according to Irish and Yamamoto (Plant Cell 7 (1'0),1635-1644 (1995)). 'this fragment was used to replace the UBQ3 fragment in the pNA00S 1 construct. The final eonstxvet was na~oned as pND304S (AraAP3 promote~::ZFPAp3-YP64//nos). pND3045 was transformed into Arabidopsis as described in previous section. About 400 plants were generated through selection media. There are several plants that have shown sterile phenotype already.
Molecular analysis on these events can be performed as previously descn'bed.
High numbers of transgenic plants have been easily selected with this tissue specific activation construct. With our previous activation construct pND0051 (with constitute promoter UaQ3), we only obtained three transgenic plants and none of them shows high Level of ZFP expression and atone of them shows phenotypic changes, These results support our previous hypothesis that over-activation and over-expression of Ap3 gene is toxic to the plant.
Examoie 1010 valuation of la 'o o endo en us a a ressio a ' the zinc ~ r ro e' having less than six fineers The Ap3-VP64 6-forger construct appears to work effectively and specifically to activate and repress AP3 expression in Arabidopsis protoplasts or even the whole plant 'this is a good system to test the activity of zinc~finger protein with less than 6 EmvfangsiAMENDED SHEET

fingers, such as 3-finger effector constructs. ' The 6-finger ZFPAp3 protein was constructed from two 3-finger proteins (See Example 3). ZPFAp3a is a 3-finger protein with fingers 4-6 of ZFPAp3. Similarly, ZFPAp3b has fingers 1 to 3 of ZFPAp3.
To test these 3-finger constructs, the Ap3-VP64 expression plasmid is digested with SfiI to remove the 6-finger Ap3 coding region. The 300bp SfiI fragment of the AP3 3-finger proteins is ligated in this digested vector to generate Ap3a-VP64 and Ap3b-VP64 constructs. The activation of AP3 expression in protoplasts is tested with these constructs using the method described in Example 7 and 8. Generally, 3-finger protein is less specific than the 6-finger protein. But, this may not be the case for all the targets (genes). If some of these 3-finger effectors are able to specifically and effectively activate AP3 expression, this will indicate that 6 finger proteins are not required for all targets. This will also demonstrate the versatility of this technology with respect to the number of zinc fingers required.
Example 11 Evaluation of ZFP-activator/re~ressor fusion protein function in maize MIPS is an endogenous maize enzyme. Its gene expression level can be monitored at three levels after transforming the maize cells with ZFPmips-activator or ZFPmips-repressor. The first level is on the transcriptional level.
Quantitative PCR
can be used to analyze the abundant of MIPS transcripts. The second level is on the protein expression level. We have generated MIPS specific antibody and can analyze the amount of MIPS protein expressed on Western blot. The third level is on the function level of MIPS enzyme. The activity of MIPS enzyme can be monitored by the concentration of its product, phytic acid through HPLC (Talamond et al., J. of Chromatography (1998) 805:143-147) and colorimetric method (Sigma, #670-A).
A Construction of maize MIfS-specific ZFP-effectors Four activation constructs were generated for ZFPml, ZFPm2, ZFPm3, and ZFPm4 (ZFPmips) by fusing the ZFPmips domain to the N terminal of the VP64 activation domain. Two repression constructs were generated for ZFPml and ZFPm4 by fusing the ZFPmips domain to the C terminal of the SID repression domain.
Then the ZFPmips-effector cassette was cloned into monocot expression vector under the control~of the maize ubiquitin (ZmUbi) promoter (same as in pND3008). A "nos"
fragment was placed at the 3'end of this cassette as transcription terminator (same as in pND3008). These six constructs are named as pND3015, pND3023, pND3024, pND3016, pND3019, and pND3017 (Fig.22).
Our transient regulation result in maize cells indicates that activation is more efficient than repression. So we have transformed all eight constructs into maize cells.
A successful activation of MIPS gene in maize is an indication of success of this zinc finger protein technology in regulating another endogenous gene in another plant species. Maize is monocot and considered as crop plant with MIPS as an agronomic trait.
B Function of ZFPmips-effectors in maize A maize type II cell line HE89 was used to evaluate the ZFPmips-effector fusion constructs function (Fig.22). Protoplasts from HE89 suspension cells were isolated and transformed using standard procedures (Chourey and Zurawski, 1981).
The activation constructs pND3015, pND3023, pND3024, and pND3016 were transformed into freshly prepared protoplasts. The transcription level of MIPS
gene was detected through quantitative PCR. With pND3016 (ZFPm4-VP64 activation constructs), at least 2-fold of activation is detected (Fig. 23). We also measured phytic acid concentration in maize tissues that were transformed by the activation construct of pND3016. We found that the phytic acid concentration is increased from 304 pg/ml to 569 pg/ml (detection was carried out with HPLC).
We have demonstrated herein that zinc finger protein approach can be used in both up-regulation (as example of AP3 in Arabidopsis, MIPS in maize) and down-regulation (as example of AP3 in Arabidopsis) of endogenous gene expression in plant.
The above examples are included for illustrative purposes only and is not intended to limit the scope of the invention. Since modifications will be apparent to those of skill in this art, it is intended that this invention be limited only by the scope of the appended claims.

Claims (138)

We Claim:
1. A method to modulate the expression of a target gene in plant cells, which method comprises providing plant cells with a zinc finger protein, said zinc finger protein being capable of specifically binding to a target nucleotide sequence, or a complementary strand thereof, within a target gene, and allowing said zinc finger protein binding to said target nucleotide sequence, whereby the expression of said target gene in said plant cells is modulated.
2. The method of claim 1, wherein the zinc finger protein is exogenously added to the plant cells and the plant cells are maintained under conditions such that the zinc finger protein binds to the target nucleotide sequence and regulates the expression of the target gene in the plant cells.
3. The method of claim 1, wherein a nucleotide sequence encoding the zinc finger protein is expressed in the plant cells and the plant cells are maintained under conditions such that the expressed zinc finger protein binds to the target nucleotide sequence and regulates the expression of the target gene in the plant cells.
4. A method to modulate the expression of a target gene in plant cells, which method comprises:

a) providing plant cells with an expression system for a zinc finger protein, said zinc finger protein being capable of specifically binding to a target nucleotide sequence, or a complementary strand thereof, within a target gene;
and . b) culturing said plant cells under conditions wherein said zinc finger protein is produced and binds to said target nucleotide sequence, whereby expression of said target gene in said plant cells is modulated.
5. The method of claim 1, wherein the target nucleotide sequence is endogenous or exogenous to the target gene.
6. The method of claim 1, wherein the target nucleotide sequence is upstream of, downstream of, or within the coding region of the target gene.
7. The method of claim 1, wherein the target nucleotide sequence is DNA, RNA, PNA or a combination thereof.
8. The method of claim 1, wherein the target nucleotide sequence is a promoter of a regulatory protein.
9. The method of claim 1, wherein the target nucleotide sequence comprises 3, 6, 9, 12, 15 or 18 nucleotides.
10. The method of claim 1, wherein the target nucleotide sequence comprises 18 nucleotides and wherein the zinc finger protein is a hexadactyl zinc finger protein.
11. The method of claim 1, wherein the targeted nucleotide sequence is of the formula (GNN)n, and wherein N is any one of the A, T, C or G and n is an integer from 1 to 6.
12. The method of claim 11, wherein the n is 6.
13. The method of claim 1, wherein the target nucleotide sequence is endogenous to the plant but is in a non-naturally-occurring location.
14. The method of claim 1, wherein the plant cells comprise at least two copies of the same or different target nucleotide sequence(s).
15. The method of claim 14, wherein each target nucleotide sequence is located within a different target gene, whereby more than one different target genes are modulated.
16. The method of claim 1, wherein the target gene encodes a product that affects biosynthesis, modification, cellular trafficking, metabolism and degradation of a peptide, a protein, an oligonucleotide, a nucleic acid, a vitamin, an oligosaccharide, a carbohydrate, a lipid, or a small molecule.
17. The method of claim 16, wherein the target gene encodes a protein or an RNA.
18. The method of claim 1, wherein the target gene encodes myoinositol 1-phosphate synthase.
19. The method of claim 1, wherein the target gene encodes a protein and, the expression of said encoded protein is modulated.
20. The method of claim 19, wherein the protein whose expression being modulated is heterologous to the plant cell.
21. The method of claim 20, wherein the protein whose expression being modulated is an antibody.
22. The method of claim 19, wherein the expression of the protein is activated.
23. The method of claim 19, wherein the protein whose expression being modulated participates in a metabolic pathway or controls a metabolic pathway.
24. The method of claim 23, wherein the metabolic pathway is an anabolic or a catabolic pathway.
25. The method of claim 23, wherein the metabolic pathway is for a molecule selected from the group consisting of a vitamin, a taste molecule, an anti-oxidant, a sugar and a flavanoid molecule.
26. The method of claim 25, wherein the taste molecule is a bad taste molecule.
27. The method of claim 23, wherein the metabolic pathway is heterologous to the plant cell.
28. The method of claim 23, wherein the metabolic pathway enhances an input or output trait in a plant or seed.
29. The method of claim 19, wherein the target gene encodes an enzyme, a transport protein, a nutrient or storage protein, a contractile or motile protein, a structural protein, a defense protein or a regulatory protein.
30. The method of claim 19, wherein the target gene encodes an enzyme or a co-factor in a metabolic pathway.
31. The method of claim 1, which method is used for treating a disorder in the plant cells, wherein the disorder is associated with abnormal expression of the target gene.
32. The method of claim 1, wherein the zinc finger protein is linked to a protein which activates or represses gene expression.
33. The method of claim 32, wherein the zinc finger protein is linked to the protein which activates or represses gene expression as a fusion protein.
34. The method of claim 32, wherein the protein which activates gene expression comprises an activator domain of a regulatory protein.
35. The method of claim 32, wherein the protein which activates or represses gene expression comprises an active domain of a nucleic acid modifying protein.
36. The method of claim 1, wherein the zinc finger protein binds to the complementary strand of the target nucleotide sequence.
37. The method of claim 1, wherein the zinc finger protein specifically binds to an effector domain of the target sequence and whereby the expression of the target gene is modulated by competitive inhibition of said effector domain.
38. The method of claim 37, wherein the zinc finger protein does not comprise an effector domain.
39. The method of claim 1, wherein the zinc finger protein comprises an effector domain active in the host plant cells.
40. The method of claim 1, wherein the zinc finger protein comprises a plurality of finger regions.
41. The method of claim 40, which comprises linker regions among the plurality of finger regions.
42. The method of claim 1, wherein the zinc finger protein comprises at least two 3-finger region and the linker region between any said two 3-finger region is from about 2 to about 10 amino acid residues in length.
43. The method of claim 42, wherein the linker region between any said two 3-finger region is about 5 amino acid residues in length.
44. The method of claim 1, wherein the zinc finger protein comprises a framework from a plant zinc finger protein.
45. The method of claim 1, wherein the zinc finger protein is a synthetic zinc finger protein, or a non-naturally-occurring zinc finger protein in the host plant.
46. The method of claim 1, wherein the zinc finger protein is selected from the group consisting of ZFPm1, ZFPm2, ZFPm3, ZFPm4 and ZFPAp3.
47. The method of claim 1, wherein the zinc finger protein is not a zinc finger-nucleotide binding polypeptide variant comprising at least three zinc finger modules that bind to a target cellular nucleotide sequence and modulate the transcriptional function of the cellular nucleotide sequence, wherein the amino acid sequence of each zinc finger module that binds a target cellular nucleotide comprises two cysteines and two histidines whereby both cysteines are amino proximal to both histidines and wherein each of three modules of said variant has at least one amino acid sequence modification.
48. The method of claim 1, wherein the plant cells are monocot or dicot plant cells.
49. The method of claim 1, wherein the plant cells are included within an intact plant or constitute all the cells of an intact plant.
50. The method of claim 1, wherein the plant cells are protoplasts or spheroplasts.
51. The method of claim 1, wherein the modulation of the gene expression is activation or repression.
52. The method of claim 1, wherein the modulation of the gene expression is at least two fold.
53. The method of claim 52, wherein the modulation is at least five fold repression.
54. The method of claim 52, wherein the modulation is at least two fold activation.
55. The method of claim 1, wherein the modulation changes the phenotype of the plant cells, the tissue of the plant or the whole plant.
56. The method of claim 4, wherein the plant cells are contained in an in vitro culture.
57. The method of claim 4, wherein the culturing is in planta.
58. The method of claim 4 wherein the expression system comprises an inducible promoter.
59. The method of claim 4, wherein the expression of the zinc finger-protein is controlled by a tissue-specific promoter and whereby tissue-specific modulation of the target gene expression is obtained.
60. The method of claim 59, wherein the tissue is selected from the group consisting of calli, meristem, leave, root and organ explant in tissue culture.
61. The method of claim 4, wherein the zinc finger protein is expressed in a specific organelle.
62. The method of claim 61, wherein the organelle is selected from the group consisting of a mitochondria, a nucleus, a plastid and a vacuole.
63. The method of claim 62, wherein the plastid is selected from the group consisting of a chloroplast, a leucoplast, an aravloplast and a chromoplast.
64. The method of claim 4, wherein the zinc finger protein is stably integrated in a specific organelle.
65. The method of claim 4, wherein the zinc finger protein is targeted to a specific organelle.
66. The method of claim 65, wherein the zinc finger protein is targeted to plastid via a plastid transit peptide, to chloroplast via a chloroplast transit peptide, to mitochondrial via a mitochondrial target peptide or to nucleus via a nuclear targeting peptide.
67. The method of claim 4, wherein the expression is transient or stable.
68. The method of claim 4, wherein the zinc finger protein comprises preferred codons of the host plant.
69. The method of claim 68, wherein the zinc finger protein comprises preferred translational start codon of the host plant.
70. A method of modulating a level of a compound in a plant cell, which method comprises expressing in a plant cell a zinc finger protein that specifically binds to a target nucleotide sequence within a target gene to modulate expression of said target gene which is involved in a compound's metabolism in said plant cell, whereby the level of said compound in said plant cell is modulated.
71. The method of claim 70, wherein the compound is phytic acid.
72. The method of claim 70, wherein the target gene encodes AP3.
73. The method of claim 70, wherein the zinc finger protein is not a zinc finger-nucleotide binding polypeptide variant comprising at least three zinc finger modules that bind to a target cellular nucleotide sequence and modulate the transcriptional function of the cellular nucleotide sequence, wherein the amino acid sequence of each zinc finger module that binds a target cellular nucleotide comprises two cysteines and two histidines whereby both cysteines are amino proximal to both histidines and wherein each of three modules of said variant has at least one amino acid sequence modification.
74. An expression vector for modulating gene expression in plant cells, which expression vector comprises a nucleotide sequence encoding a zinc finger protein, said zinc finger protein is capable of specifically binding to a target nucleotide sequence, or a complementary strand thereof, within a target gene whose expression is to be modulated by said zinc finger protein in plant cells.
75. The expression vector of claim 74, wherein the zinc finger protein is not a zinc finger-nucleotide binding polypeptide variant comprising at least three zinc finger modules that bind to a target cellular nucleotide sequence and modulate the transcriptional function of the cellular nucleotide sequence, wherein the amino acid sequence of each zinc finger module that binds a target cellular nucleotide comprises two cysteines and two histidines whereby both cysteines are amino proximal to both histidines and wherein each of three modules of said variant has at least one amino acid sequence modification.
76. A genetically modified plant cell, which cell comprises an expression system for a zinc finger protein, said zinc finger protein is capable of specifically binding to a target nucleotide sequence, or a complementary strand thereof, within a target gene whose expression is to be modulated by said zinc finger protein.
77. The genetically modified plant cell of claim 76, wherein the target nucleotide sequence is endogenous or exogenous to the targeted gene.
78. The genetically modified plant cell of claim 76, wherein the target gene is endogenous or exogenous to the plant cells.
79. The genetically modified plant cell of claim 76, which is contained in an intact plant.
80. The genetically modified plant cell of claim 76, wherein the zinc finger protein controls its own expression by binding to a target sequence within the zinc finger protein gene.
81. The genetically modified plant cell of claim 76, wherein the zinc finger protein controls its own expression by binding to a first target sequence within the zinc finger protein gene and controls the expression of the target gene by binding to a second target sequence within the target gene.
82. The genetically modified plant cell of claim 81, wherein the first target sequence within the zinc finger protein gene is different from the second target sequence within the target gene.
83. The genetically modified plant cell of claim 76, wherein the zinc finger protein gene is further controlled by a second promoter.
84. The genetically modified plant cell of claim 83, wherein the second promoter is inducible.
85. The genetically modified plant cell of claim 76, wherein the zinc finger protein comprises at least two zinc finger sequences.
86. The genetically modified plant cell of claim 85, wherein the zinc finger protein comprises from about 2 to about 6 zinc finger sequences.
87. The genetically modified plant cell of claim 85, wherein the zinc finger protein comprises from about 3 to about 6 zinc finger sequences.
88. The genetically modified plant cell of claim 76, which is selected from the group consisting of a tobacco, tomato, potato, banana, soybean, pepper, wheat, rye, rice, spinach, carrot, maize and corn cell.
89. The genetically modified plant cell of claim 76, wherein the zinc finger protein expressed in the plant cell is not a zinc finger-nucleotide binding polypeptide variant comprising at least three zinc finger modules that bind to a target cellular nucleotide sequence and modulate the transcriptional function of the cellular nucleotide sequence, wherein the amino acid sequence of each zinc finger module that binds a target cellular nucleotide comprises two cysteines and two histidines whereby both cysteines are amino proximal to both histidines and wherein each of three modules of said variant has at least one amino acid sequence modification.
90. A genetically modified plant cell transformed with a nucleic acid comprising a functional geminiviral replicase gene operably linked to a fruit ripening-dependent promoter.
91. A genetically modified plant cell, which cell comprises an exogenous zinc finger protein that specifically binds to a target nucleotide sequence in said plant cell wherein said exogenous zinc finger protein is constitutively expressed.
92. A genetically modified plant cell, which cell comprises an exogenous zinc finger protein that specifically binds to a target nucleotide sequence in said plant cell wherein said exogenous zinc finger protein is inducibly expressed.
93. A genetically modified plant tissue, which tissue comprises the genetically modified plant cell of claim 76.
94. A genetically modified plant seed, which seed comprises the genetically modified plant cell of claim 76.
95. The genetically modified plant seed of claim 94, which is selected from the group consisting of a tobacco, tomato, potato, banana, soybean, pepper, wheat, rye, rice, spinach, carrot, maize and corn seed.
96. A genetically modified plant seed transformed with a nucleic acid comprising a functional geminiviral replicase gene operably linked to a fruit ripening-dependent promoter.
97. A genetically modified plant seed, which seed comprises the genetically modified plant cell of claim 89.
98. A plant that is regenerated from a plant transformed with the expression vector of claim 74.
99. A method to modulate expression in a plant cell, which method comprises culturing the plant cell of claim 76.
100. The method of claim 99, wherein the plant cell is cultured in planta.
101. A zinc finger protein comprising a zinc finger nucleic acid binding domain and an effector domain, wherein said effector domain comprises an active domain of a restriction enzyme, an active domain of a nucleic acid modifying protein, a label or a modification.
102. The zinc finger protein of claim 101, wherein the nucleic acid modifying protein is a nucleic acid methylase.
103. A zinc finger protein that is selected from the group consisting of ZFPm1, ZFPm2, ZFPm3, ZFPm4 and ZFPAp3.
104. A zinc finger protein that is recognizable by an antibody that specifically binds to a zinc finger protein selected from the group consisting of ZFPm1, ZFPm2, ZFPm3, ZFPm4 and ZFPAp3.
105. The zinc finger protein of claim 104, which retains gene expression modulation activity of ZFPm1, ZFPm2, ZFPm3, ZFPm4 or ZFPAp3.
106. An antibody that specifically binds to a zinc finger protein selected from the group consisting of ZFPm1, ZFPm2, ZFPm3, ZFPm4 and ZFPAp3.
107. The antibody of claim 106, which is a monoclonal antibody.
108. An isolated nucleic acid fragment, comprising a sequence of nucleotides encoding a zinc finger protein that is selected from the group consisting of ZFPm1, ZFPm2, ZFPm3, ZFPm4 and ZFPAp3.
109. The isolated nucleic acid fragment of claim 108, which is DNA or RNA.
110. An isolated nucleic acid fragment, which is hybridizable to the nucleic acid fragment of claim 108.
111. The isolated nucleic acid fragment of claim 110, which is hybridizable to the nucleic acid fragment of claim 108 under high stringency condition.
112. A plasmid, comprising the nucleic acid fragment of claim 108.
113. A cell, comprising the plasmid of claim 112.
114. The cell of claim 113, wherein the cell is selected from the group consisting of a bacterial cell, a yeast cell, a fungal cell, a plant cell, an insect cell and an animal cell.
115. A method for producing a zinc finger protein, comprising: growing the cell of claim 113 under conditions whereby the zinc finger protein is expressed by the cell; and recovering the expressed zinc finger protein.
116. An assay method for determining a suitable position in a gene for regulating gene expression in plant cells, which method comprises:
a) providing a target gene which contains a nucleotide sequence encoding a reporter protein within the coding region of said target gene and a target nucleotide sequence at a predetermined location within said target gene;
b) contacting said target gene with a regulatory factor comprising a zinc finger protein specific for said target nucleotide sequence; and c) assessing the level of expression of said reporter gene in the presence and absence of said contacting;
wherein a change in the level of expression of said reporter gene in the presence as opposed to the absence of said contacting identifies said position of said target nucleotide sequence as a position suitable for controlling expression of said target gene in plant cells.
117. The assay method of claim 116, wherein the zinc finger protein is not a zinc finger-nucleotide binding polypeptide variant comprising at least three zinc finger modules that bind to a target cellular nucleotide sequence and modulate the transcriptional function of the cellular nucleotide sequence, wherein the amino acid sequence of each zinc finger module that binds a target cellular nucleotide comprises two cysteines and two histidines whereby both cysteines are amino proximal to both histidines and wherein each of three modules of said variant has at least one amino acid. sequence modification.
118. A fusion protein, which fusion protein comprises a zinc finger of 2C7 and an effector domain of SID.
119. A zinc finger protein that is recognizable by an antibody that specifically binds to a fusion protein comprising a zinc finger of 2C7 and an effector domain of SID3.
120. The zinc finger protein of claim 119, which retains gene expression modulation activity of the fusion protein comprising a zinc finger of 2C7 and an effector domain of SID3.
121. The fusion protein of claim 118, which is encoded by the nucleotide sequence set forth in SEQ ID NO:5 or SEQ ID NO:66.
122. An antibody that specifically binds to a fusion protein comprising a zinc finger of 2C7 and an effector domain of SID3.
123. The antibody of claim 122, which is a monoclonal antibody.
124. An isolated nucleic acid fragment, comprising a sequence of nucleotides encoding a fusion protein comprising a zinc finger of 2C7 and an effector domain of SID3.~
125. The isolated nucleic acid fragment of claim 124, which is DNA or RNA.
126. An isolated nucleic acid fragment, which is hybridizable to the nucleic acid fragment of claim 124.
127. The isolated nucleic acid fragment of claim 126, which is hybridizable to the nucleic acid fragment of claim 124 under high stringency condition.
128. The isolated nucleic acid fragment of claim 124, which has the nucleotide sequence set forth in SEQ ID NO:5 or SEQ ID NO:66.
129. A plasmid, comprising the nucleic acid fragment of claim 124.
130. A cell, comprising the plasmid of claim 129.
131. The cell of claim 130, wherein the cell is selected from the group consisting of a bacterial cell, a yeast cell, a fungal.cell, a plant cell, an insect cell and an animal cell.
132. A method for producing a fusion protein, comprising: growing the cell of claim 130 under conditions whereby the fusion protein is expressed by the cell; and recovering the expressed fusion protein.
133. The method of claim 1, wherein the zinc finger protein comprises a framework (or backbone) derived from a naturally occurring zinc finger protein.
134. The method of claim 1, wherein the zinc finger protein comprises a framework (or backbone) derived from a zinc finger protein comprising a C2H2 motif.
135. The method of claim 134, wherein the protein or peptide sequence within the .beta. sheet of the C2H2 motif is not changed from its natural sequence.
136. The method of claim 1, wherein the zinc finger protein comprises a framework (or backbone) derived from a zinc finger protein that is naturally functional in plant cells.
137. The method of claim 136, wherein the framework (or backbone) comprises a motif selected from the group consisting of a C3H zinc finger, a QALGGH motif, a RING-H2 zinc finger motif, a 9 amino acid C2H2 motif, a zinc finger motif of Arabidopsis LSD1 and a zinc finger motif of BBF/Dof domain proteins.
138. The method of claim 1, wherein the zinc finger protein comprises a framework (or backbone) derived from a zinc finger protein that is known in the art as of January 19, 2001.
CA002396898A 2000-01-21 2001-01-19 Methods and compositions to modulate expression in plants Abandoned CA2396898A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US17746800P 2000-01-21 2000-01-21
US60/177,468 2000-01-21
US62089700A 2000-07-21 2000-07-21
US09/620,897 2000-07-21
PCT/US2001/001817 WO2001052620A2 (en) 2000-01-21 2001-01-19 Methods and compositions to modulate expression in plants

Publications (1)

Publication Number Publication Date
CA2396898A1 true CA2396898A1 (en) 2001-07-26

Family

ID=26873328

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002396898A Abandoned CA2396898A1 (en) 2000-01-21 2001-01-19 Methods and compositions to modulate expression in plants

Country Status (5)

Country Link
EP (1) EP1276869A2 (en)
AR (1) AR048989A1 (en)
AU (1) AU2964101A (en)
CA (1) CA2396898A1 (en)
WO (1) WO2001052620A2 (en)

Families Citing this family (75)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7285416B2 (en) 2000-01-24 2007-10-23 Gendaq Limited Regulated gene expression in plants
US7262055B2 (en) 1998-08-25 2007-08-28 Gendaq Limited Regulated gene expression in plants
US7329728B1 (en) 1999-10-25 2008-02-12 The Scripps Research Institute Ligand activated transcriptional regulator proteins
AU2001253914B2 (en) 2000-04-28 2006-06-08 Sangamo Therapeutics, Inc. Targeted modification of chromatin structure
BRPI0407514A (en) 2003-02-17 2006-02-14 Metanomics Gmbh method for preparing a non-human organism, polynucleotide, method for preparing a vector, vector, host cell, polypeptide, antibody, antisense polynucleic acid, method for preparing a plant, plant cell, transgenic plant tissue, useful animal cell , useful animal or a transgenic microorganism non-human organism, transgenic microorganism, yield or a propagating material of a useful plant or animal, biomass of a microorganism, method for preparing fine chemicals, and, use of polypeptide
ATE514781T1 (en) 2003-04-15 2011-07-15 Basf Plant Science Gmbh NUCLEIC ACID SEQUENCES CODING FOR PROTEINS RELATED TO ABIOTIC STRESS RESPONSE, AS WELL AS PLANT CELLS AND PLANTS WITH INCREASED ENVIRONMENTAL STRESS TOLERANCE
WO2005014828A2 (en) 2003-08-01 2005-02-17 Basf Plant Science Gmbh Process for the production of fine chemicals in plants
EP1765857A2 (en) 2004-07-02 2007-03-28 Metanomics GmbH Process for the production of fine chemicals
CN101001956B (en) 2004-07-31 2014-09-24 梅坦诺米克斯有限公司 Preparation of organisms with faster growth and/or higher yield
AU2005286427B2 (en) 2004-09-24 2011-09-15 Basf Plant Science Gmbh Plant cells and plants with increased tolerance to environmental stress
EP1794184A2 (en) 2004-09-24 2007-06-13 BASF Plant Science GmbH Nucleic acid sequences encoding proteins associated with abiotic stress response and plant cells and plants with increased tolerance to environmental stress
MX2007007040A (en) 2004-12-17 2008-10-24 Metanomics Gmbh Process for the control of production of fine chemicals.
EP2573188A2 (en) 2005-03-02 2013-03-27 Metanomics GmbH Process for the production of fine chemicals
CA2598792A1 (en) 2005-03-02 2006-09-08 Metanomics Gmbh Process for the production of fine chemicals
WO2007020198A2 (en) 2005-08-12 2007-02-22 Basf Plant Science Gmbh Nucleic acid sequences encoding proteins associated with abiotic stress response and plant cells and plants with increased tolerance to environmental stress
CN101356279A (en) 2005-11-10 2009-01-28 先锋高级育种国际公司 DOF (DNA binding with one finger) sequences and methods of use
GB0526449D0 (en) * 2005-12-23 2006-02-08 Medical Res Council Polypeptide targeting
EP2343375A3 (en) 2006-03-24 2012-02-08 BASF Plant Science GmbH Proteins associated with abiotic stress response and homologs
EP2090662A3 (en) 2006-04-05 2012-10-31 Metanomics GmbH Process for the production of a fine chemical
US7557266B2 (en) 2006-04-19 2009-07-07 Pioneer Hi-Bred International, Inc. Isolated polynucleotide molecules corresponding to mutant and wild-type alleles of the maize D9 gene and methods of use
CA2652598A1 (en) 2006-05-17 2007-11-29 Pioneer Hi-Bred International, Inc. Artificial plant minichromosomes
EP2029619B1 (en) 2006-05-31 2013-01-09 Metanomics GmbH Manipulation of the nitrogen metabolism using ammonium transporter or glucose 6-phosphate deshydrogenases or farnesyl phosphate synthetase (fpp)
CN103397043A (en) 2006-08-04 2013-11-20 维莱尼姆公司 Glucanases, nucleic acids encoding same and method for making and using same
EP2620495B1 (en) 2006-09-21 2016-03-09 BASF Enzymes LLC Phytases, nucleic acids encoding them and methods for making and using them
CA2663959A1 (en) 2006-10-13 2008-04-17 Basf Plant Science Gmbh Plants with increased yield
US20100124583A1 (en) 2008-04-30 2010-05-20 Xyleco, Inc. Processing biomass
DK2412812T3 (en) 2006-12-14 2015-05-26 Dow Agrosciences Llc Optimized non-canonical zinc finger proteins
US8343747B2 (en) 2006-12-21 2013-01-01 Verenium Corporation Amylases and glucoamylases, nucleic acids encoding them and methods for making and using them
AU2008214568B2 (en) 2007-02-16 2013-04-18 Basf Plant Science Gmbh Nucleic acid sequences for regulation of embryo-specific expression in monocotyledonous plants
US7642436B2 (en) 2007-04-13 2010-01-05 Ball Horticultural Company Petunia mutant allele
AR067318A1 (en) 2007-05-22 2009-10-07 Basf Plant Science Gmbh PLANTS WITH HIGHER TOLERANCE AND / OR INCREASED RESISTANCE TO ENVIRONMENTAL STRESS AND HIGHER BIOMASS PRODUCTION
CA2687635A1 (en) 2007-05-22 2008-11-27 Basf Plant Science Gmbh Plant cells and plants with increased tolerance and/or resistance to environmental stress and increased biomass production-ko
CA2687211A1 (en) 2007-05-25 2008-12-04 Cropdesign N.V. Yield enhancement in plants by modulation of maize alfins
WO2008157263A2 (en) 2007-06-15 2008-12-24 Arkansas State University Methods of delivery of molecules to cells using a ricin subunit and compositions relating to same
US8664475B2 (en) 2007-09-18 2014-03-04 Basf Plant Science Gmbh Plants with increased yield
EP2594647A3 (en) 2007-09-21 2013-07-24 BASF Plant Science GmbH Plants with increased yield
BRPI0820114B1 (en) 2007-12-03 2022-10-04 Syngenta Participations Ag METHOD OF INCREASE THE SENSITIVITY OF AN ESCHERICHIA COLI-DERIVED PHYTASE TO A PEPSIN
EP2604622A3 (en) 2007-12-19 2013-10-09 BASF Plant Science GmbH Plants with increased yield and/or increased tolerance to environmental stress (IY-BM)
EP2240587A2 (en) 2007-12-21 2010-10-20 BASF Plant Science GmbH Plants with increased yield (ko nue)
AU2009218478A1 (en) 2008-02-27 2009-09-03 Basf Plant Science Gmbh Plants with increased yield
CA3051188A1 (en) 2008-04-30 2009-11-05 Xyleco, Inc. Processing biomass
US7964774B2 (en) 2008-05-14 2011-06-21 Monsanto Technology Llc Plants and seeds of spring canola variety SCV384196
EP2352822A2 (en) 2008-08-19 2011-08-10 BASF Plant Science GmbH Plants with increased yield by increasing or generating one or more activities in a plant or a part thereof
BRPI0919153A2 (en) 2008-09-23 2019-05-07 Basf Plant Science Gmbh method for producing a high yielding plant, nucleic acid molecule, nucleic acid construct, vector, process for producing a polypeptide, polypeptide, antibody, plant cell nucleus, plant cell, plant tissue, propagating material, pollen , progeny, material collected, or a plant, or part of a plant, a transgenic plant, process for identifying a compound, method for producing an agricultural composition, composition, use of nucleic acid, and method for identifying a plant with an increased yield, and to increase the yield of a plant population
EP2350289A1 (en) 2008-10-23 2011-08-03 BASF Plant Science GmbH Plants with increased yield (nue)
AU2009306369A1 (en) 2008-10-23 2010-04-29 Basf Plant Science Gmbh A method for producing a transgenic cell with increased gamma-aminobutyric acid (GABA) content
AU2010204723A1 (en) 2009-01-14 2011-08-18 The Salk Institute For Biological Studies Methods for screening and compounds that protect against amyloid diseases
CA2758824A1 (en) 2009-04-17 2010-10-21 Basf Plant Science Company Gmbh Plant promoter operable in endosperm and uses thereof
CA2760700A1 (en) 2009-05-04 2010-11-11 Pioneer Hi-Bred International, Inc. Yield enhancement in plants by modulation of ap2 transcription factor
WO2010135588A2 (en) 2009-05-21 2010-11-25 Verenium Corporation Phytases, nucleic acids encoding them and methods for making and using them
US8440891B2 (en) 2009-09-22 2013-05-14 Board of Trustees of the University of Akransas, N.A. Rice cultivar CL 142-AR
US8440892B2 (en) 2009-10-15 2013-05-14 Board Of Trustees Of The University Of Arkansas, N.A. Rice cultivar CL 181-AR
WO2011082310A2 (en) 2009-12-30 2011-07-07 Pioneer Hi-Bred International, Inc. Methods and compositions for targeted polynucleotide modification
US20110167516A1 (en) 2009-12-30 2011-07-07 Pioneer Hi-Bred International, Inc. Methods and compositions for the introduction and regulated expression of genes in plants
EA201291178A1 (en) 2010-05-06 2013-04-30 Пайонир Хай-Бред Интернэшнл, Инк. GENE AND PROTEIN ACC SYNTHASE 3 MAISA AND THEIR APPLICATION
US9096871B2 (en) 2010-10-06 2015-08-04 Bp Corporation North America Inc. Variant CBH I polypeptides with reduced product inhibition
US8785729B2 (en) 2011-08-09 2014-07-22 Nunhems, B.V. Lettuce variety redglace
US8754293B2 (en) 2011-09-09 2014-06-17 Nunhems B.V. Lettuce variety intred
JP2014530607A (en) * 2011-10-11 2014-11-20 アリオフタ・アーゲーAliophtha Ag Regulation of receptor expression by delivery of artificial transcription factors
US20130167262A1 (en) 2011-12-21 2013-06-27 The Curators Of The University Of Missouri Soybean variety s05-11268
US9204603B2 (en) 2011-12-21 2015-12-08 The Curators Of The University Of Missouri Soybean variety S05-11482
WO2013134651A1 (en) 2012-03-09 2013-09-12 Board Of Trustees Of Michigan State University Method of enhancing plant drought tolerance by expression of ndr1
US8878009B2 (en) 2012-04-26 2014-11-04 Monsanto Technology, LLP Plants and seeds of spring canola variety SCV318181
US8859857B2 (en) 2012-04-26 2014-10-14 Monsanto Technology Llc Plants and seeds of spring canola variety SCV259778
US8835720B2 (en) 2012-04-26 2014-09-16 Monsanto Technology Llc Plants and seeds of spring canola variety SCV967592
EP3166965B1 (en) 2014-07-10 2020-09-09 Benson Hill, Inc. Compositions and methods for increasing plant growth and yield
EP3365450A2 (en) 2015-10-23 2018-08-29 Donald Danforth Plant Science Center Genes encoding c4 transporter proteins for use in increasing crop plant yield
BR112018008705B1 (en) 2015-10-30 2023-12-26 Pioneer Hi-Bred International, Inc METHOD FOR PRODUCING A TRANSGENIC PLANT
JP6871260B2 (en) 2015-11-06 2021-05-12 パイオニア ハイ−ブレッド インターナショナル, インコーポレイテッド Improved plant transformation methods and compositions
EP3833747A1 (en) 2018-06-28 2021-06-16 Pioneer Hi-Bred International, Inc. Methods for selecting transformed plants
CN108949786B (en) * 2018-06-29 2021-07-23 山东农业大学 Application of arabidopsis E3 ubiquitin ligase encoding gene ATL27 in regulation and control of plant salt tolerance
CA3111090A1 (en) 2018-10-31 2020-05-07 Pioneer Hi-Bred International, Inc. Compositions and methods for ochrobactrum-mediated plant transformation
CN109912704B (en) * 2019-03-22 2022-03-18 济南大学 Corn zinc finger binding protein interaction factor gene Actin and recombinant expression vector and application thereof
CN111205356B (en) * 2020-01-15 2023-03-21 湖北大学 Gene for regulating and controlling plant florescence and encoding protein and application thereof
CN114561387B (en) * 2022-02-28 2023-08-15 北京大学现代农业研究院 Peanut promoter and application thereof

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6140466A (en) * 1994-01-18 2000-10-31 The Scripps Research Institute Zinc finger protein derivatives and methods therefor
US6534261B1 (en) * 1999-01-12 2003-03-18 Sangamo Biosciences, Inc. Regulation of endogenous gene expression in cells using zinc finger proteins
AU778150B2 (en) * 1999-05-28 2004-11-18 Gendaq Limited Molecular switches

Also Published As

Publication number Publication date
EP1276869A2 (en) 2003-01-22
WO2001052620A2 (en) 2001-07-26
AR048989A1 (en) 2006-06-21
WO2001052620A3 (en) 2002-02-07
AU2964101A (en) 2001-07-31

Similar Documents

Publication Publication Date Title
US7378510B2 (en) Synthetic zinc finger protein encoding sequences and methods of producing the same
CA2396898A1 (en) Methods and compositions to modulate expression in plants
US10017775B2 (en) Engineered zinc finger proteins targeting plant genes involved in fatty acid biosynthesis
EP1303608A2 (en) Zinc finger domain recognition code and uses thereof
US20030082561A1 (en) Zinc finger domain recognition code and uses thereof
AU2008203310A1 (en) Cell proliferation-related polypeptides and uses therefor
Wang et al. Maize GSK3-like kinase ZmSK2 is involved in embryonic development
CA2256121A1 (en) Cyclin-dependent kinase inhibitors as plant growth regulators
AU2006203634B2 (en) Methods and compositions to modulate expression in plants
US7553667B2 (en) Regulation of gene expression using chromatin remodelling factors
US7582807B2 (en) Regulation of gene expression using chromatin remodelling factors
AU2015203725B2 (en) Engineered zinc finger proteins targeting plant genes involved in fatty acid biosynthesis
US20040091990A1 (en) Compositions and methods for regulation of plant gamma-tocopherol methyltransferase
AU2006200381A1 (en) Regulation of Gene Expression Using Chromatin Remodelling Factors

Legal Events

Date Code Title Description
EEER Examination request
FZDE Dead