WO2003096797A2 - Method of increasing plant organ and seed size in a plant - Google Patents

Method of increasing plant organ and seed size in a plant Download PDF

Info

Publication number
WO2003096797A2
WO2003096797A2 PCT/US2003/014989 US0314989W WO03096797A2 WO 2003096797 A2 WO2003096797 A2 WO 2003096797A2 US 0314989 W US0314989 W US 0314989W WO 03096797 A2 WO03096797 A2 WO 03096797A2
Authority
WO
WIPO (PCT)
Prior art keywords
seq
plant
dna
promoter
sequence
Prior art date
Application number
PCT/US2003/014989
Other languages
French (fr)
Other versions
WO2003096797A3 (en
Inventor
Mary Fernandes
Zhidong Xie
Stanton B. Dotson
Original Assignee
Monsanto Technology Llc
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 Monsanto Technology Llc filed Critical Monsanto Technology Llc
Priority to AU2003237834A priority Critical patent/AU2003237834A1/en
Priority to EP03736593A priority patent/EP1504106B1/en
Priority to DK03736593.9T priority patent/DK1504106T3/en
Priority to US10/513,167 priority patent/US7335812B2/en
Priority to AT03736593T priority patent/ATE473281T1/en
Priority to DE60333280T priority patent/DE60333280D1/en
Publication of WO2003096797A2 publication Critical patent/WO2003096797A2/en
Publication of WO2003096797A3 publication Critical patent/WO2003096797A3/en
Priority to US12/001,901 priority patent/US7994402B2/en

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)
    • C12N15/8241Phenotypically 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
    • 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

Definitions

  • This invention relates to plant molecular biology.
  • this invention relates to transgenic plants having increased or enhanced seed and organ sizes.
  • a plant goes through a development cycle, which includes seed germination, maturation of plant, reproduction, and finally senescence that leads to death of a plant.
  • Several biological processes are common to different stages of plant development. Desired effects such as growth of tissue organ are achieved in nature by fine-tuning of the metabolism of the organism.
  • the final phase of growth is senescence which is a highly regulated, genetically controlled and active process (Thomas H., and Stoddart J. L., Ann. Rev. Plant Physiol (31) 83- 111, 1980).
  • Senescence is mostly studied in plant leaves and is regarded as a series of events concerned with cellular disassembly and the mobilization of released material to other plant parts such as seeds, storage organs or developing leaves and flowers (Nooden L.D. In Senescence and Aging in Plants, Academic press, 391-439, 1988).
  • Leaf senescence can be initiated by seed development in certain species of plants. This was demonstrated in soybean by surgically removing flowers or physically restricting pod growth to observe the delay in leaf senescence (Nooden L.D. In Senescence and Aging in Plants, Academic press, 330-368,1988; Miceli F, Crafts-Brandner S.J., Egli D.B. Crop Sci. (35), 1080-1085, 1995).
  • partitioning of resources between vegetative and reproductive development involves a complex interplay of generative and degenerative processes, requiring differential expression of genes.
  • SAGs Synchronization Associated Genes
  • the SAG 13 gene was first described by Lohman et al. in 1994 (Lohman K.M., Gan S.,
  • the present invention relates to transgenic plants with increased organ size when compared to a non transformed plant of the same species.
  • the present invention provides a transgenic plant with increased seed size when compared to a non transformed plant of the same species.
  • One embodiment of the invention provides a method of increasing seed and organ size of a plant by transforming the plant with a DNA construct comprising a promoter that functions in plants, operably linked to a DNA molecule that encodes a protein, where in said DNA molecule is selected from the from the group consisting of SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9, SEQ ID NO: 11, SEQ ID NO: 13, SEQ ID NO: 15, SEQ ID NO: 17, SEQ ID NO: 19, SEQ ID NO: 21, SEQ ID NO: 23, SEQ ID NO: 25, SEQ ID NO: 27, SEQ ID NO: 29, SEQ ID NO: 31, SEQ ID NO: 33, SEQ ID NO: 35, SEQ ID NO: 37, SEQ ID NO: 39, SEQ ID NO: 41, SEQ ID NO: 43, SEQ ID NO: 45, SEQ ID NO: 47, SEQ ID NO: 49, SEQ ID NO: 51, SEQ ID NO: 53, SEQ ID NO: 66, S
  • the method also includes the DNA construct operably linked to a 3' termination region and selecting a desired plant from a population of transformed plants containing said DNA construct; wherein said desired plant exhibits increased seed and organ size compared to a plant of a same plant species not transformed to contain said DNA construct.
  • the method may also encompass various promoters including a caulimovirus promoter, or a heterologous plant constitutive promoter, or a tissue specific or an organ enhanced promoter.
  • the present invention provides a method for increasing seed and other plant organ sizes by transforming a plant with a DNA construct comprising a promoter that functions in plants, operably linked to a DNA molecule that encodes a protein, wherein said protein comprises at least an N-terminal 50% portion of a polypeptide selected from the group consisting of : SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10, SEQ ID NO: 12, SEQ ID NO: 14, SEQ ID NO: 16, SEQ ID NO: 18, SEQ ID NO: 20, SEQ ID NO: 22, SEQ ID NO: 24, SEQ ID NO: 26, SEQ ID NO: 28, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 34, SEQ ID NO: 36, SEQ ID NO: 38, SEQ ID NO: 40, SEQ ID NO: 42, SEQ ID NO: 44, SEQ ID NO: 46, SEQ ID NO: 48, SEQ ID NO: 50, SEQ ID NO: 52, SEQ
  • the present invention provides an isolated DNA construct comprising a promoter capable of functioning in a plant cell, operably linked to a structural nucleic acid sequence encoding SEQ ID NO: 2 or SEQ ID NO: 4, and a 3' non translated nucleic acid sequence capable of causing transcriptional termination and the addition of polyadenylated nucleotides to the 3 'end of the transcribed mRNA sequence.
  • This nucleic acid sequence may optionally include an intron, a 5' untranslated leader sequence or another nucleic acid sequence designed to enhance transcription and/or translation.
  • Another aspect of the present invention provides a method for improving the seed and organ size in a plant comprising the steps of:
  • SEQ ID NO: 22 SEQ ID NO: 24, SEQ ID NO: 26, SEQ ID NO: 28, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 34, SEQ ID NO: 36, SEQ ID NO: 38, SEQ ID NO: 40, SEQ ID NO: 42, SEQ ID NO: 44, SEQ ID NO: 46, SEQ ID NO: 48, SEQ ID NO: 50, SEQ ID NO: 52, SEQ ID NO: 54, SEQ ID NO: 67, SEQ ID NO: 69, and SEQ ID NO: 71 operably linked to
  • step (b) obtaining transformed plant cells containing the nucleic acid of step (a);
  • transgenic plant with increased seed and organ size when compared to a non transformed plant of a same plant species, said transgenic plant comprising a DNA construct, wherein said DNA construct comprises a promoter that functions in plants, operably linked to a DNA molecule that encodes a protein, wherein said DNA molecule is selected from the group consisting of: SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9, SEQ ID NO: 11, SEQ ID NO: 13, SEQ ID NO: 15, SEQ ID NO: 17, SEQ ID NO: 19, SEQ ID NO: 21, SEQ ID NO: 23, SEQ ID NO: 25, SEQ ID NO: 27, SEQ ID NO: 29, SEQ ID NO: 31, SEQ ID NO: 33, SEQ ID NO: 35, SEQ ID NO: 37, SEQ ID NO: 39, SEQ ID NO: 41, SEQ ID NO: 43, SEQ ID NO: 45, SEQ ID NO: 47, SEQ ID NO: 49, SEQ ID NO: 51,
  • transgenic plant with increased seed and organ size when compared to a non transformed plant of a same plant species, the transgenic plant comprising a DNA construct, wherein said DNA construct encodes a protein that comprises at least an N-terminal 50% portion selected from the group consisting of: SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10, SEQ ID NO: 12, SEQ ID NO: 14, SEQ ID NO: 16, SEQ ID NO: 18, SEQ ID NO: 20, SEQ ID NO: 22, SEQ ID NO: 24, SEQ ID NO: 26, SEQ ID NO: 28, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 34, SEQ ID NO: 36, SEQ ID NO: 38, SEQ ID NO: 40, SEQ ID NO: 42, SEQ ID NO: 44, SEQ ID NO: 46, SEQ ID NO: 48, SEQ ID NO: 50, SEQ ID NO: 52, SEQ ID NO: 54, SEQ ID NO: 67, S
  • nucleotide sequence encodes a polypeptide having an amino acid sequence that has at least 50% sequence identity, preferably at least 60%, more preferably at least 70% sequence identity, even more preferably at least 80% or 90% sequence identity, and most preferably at least 95% to 98% sequence identity to a polypeptide selected from group consisting of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10, SEQ ID NO: 12, SEQ ID NO: 14, SEQ ID NO: 16, SEQ ID NO: 18, SEQ ID NO: 20, SEQ ID NO: 22, SEQ ID NO: 24, SEQ ID NO: 26, SEQ ID NO: 28, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 34, SEQ ID NO: 36, SEQ ID NO: 38, SEQ ID NO: 40, SEQ ID NO:
  • Figure 1 shows a plasmid map for plant transformation vector pMON 23435 with its elements.
  • Figure 2 shows a plasmid map for plant transformation vector pMON 57521 with its elements.
  • Figure 3 shows a plasmid map for plant transformation vector pMON 54955 with its elements.
  • Figure 4 shows a plasmid map for plant transformation vector pMON 73955 with its elements.
  • Figure 5 shows a plasmid map for plant transformation vector pMON 73963 with its elements.
  • Figure 6 shows a plasmid map for bacterial transformation vector pMON 63132 with its elements.
  • Figure 7 shows a plasmid map for bacterial transformation vector pMON 63133 with its elements.
  • Figure 8 shows a plasmid map for bacterial transformation vector pMON 6134 with its elements.
  • Figure 9 shows a plasmid map for bacterial transformation vector pMON 6135 with its elements.
  • Figure 10 shows primers used for amplification of gene and for sequencing of amplicons.
  • Figure 11 shows putative products from steroid reductase activity.
  • Figure 12 shows a potential steroid reductase reaction.
  • the present invention provides a method of increasing seed and organ size of a plant by transforming the plant with a DNA construct comprising a promoter that functions in plants, operably linked to a DNA molecule that encodes a protein, where in said DNA molecule is selected from the from the group consisting of SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9, SEQ ID NO: 11, SEQ ID NO: 13, SEQ ID NO: 15, SEQ ID NO: 17, SEQ ID NO: 19, SEQ ID NO: 21, SEQ ID NO: 23, SEQ ID NO: 25, SEQ ID NO: 27, SEQ ID NO: 29, SEQ ID NO: 31, SEQ ID NO: 33, SEQ ID NO: 35, SEQ ID NO: 37, SEQ ID NO: 39, SEQ ID NO: 41, SEQ ID NO: 43, SEQ ID NO: 45, SEQ ID NO: 47, SEQ ID NO: 49, SEQ ID NO: 51, SEQ ID NO: 53, SEQ ID NO: 66, SEQ ID
  • the present invention provides a method for increasing seed and other plant organ sizes by transforming a plant with a DNA construct comprising a promoter that functions in plants, operably linked to a DNA molecule that encodes a protein, wherein said protein comprises at least an N-terminal 50% portion of a polypeptide selected from the group consisting of : SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10, SEQ ID NO: 12, SEQ ID NO: 14, SEQ ID NO: 16, SEQ ID NO: 18, SEQ ID NO: 20, SEQ ID NO: 22, SEQ ID NO: 24, SEQ ID NO: 26, SEQ ID NO: 28, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 34, SEQ ID NO: 36, SEQ ID NO: 38, SEQ ID NO: 40, SEQ ID NO: 42, SEQ ID NO: 44, SEQ ID NO: 46, SEQ ID NO: 48, SEQ ID NO: 50, SEQ ID NO: 52, SEQ
  • nucleic acid molecule as used herein means a deoxyribonucleic acid (DNA) molecule or ribonucleic acid (RNA) molecule. Both DNA and RNA molecules are constructed from nucleotides linked end to end, wherein each of the nucleotides contains a phosphate group, a sugar moiety, and either a purine or a pyrimidine base. Nucleic acid molecules can be a single or double-stranded polymer of nucleotides read from the 5' to the 3' end. Nucleic acid molecules may also optionally contain synthetic, non-natural or altered nucleotide bases that permit correct read through by a polymerase and do not alter expression of a polypeptide encoded by that nucleic acid molecule.
  • an isolated nucleic acid molecule means a nucleic acid molecule that is no longer accompanied by some of materials with which it is associated in its natural state or to a nucleic acid molecule the structure of which is not identical to that of any of naturally occurring nucleic acid molecule.
  • an isolated nucleic acid molecule examples include: (1) DNAs which have the sequence of part of a naturally occurring genomic DNA molecule but are not flanked by two coding sequences that flank that part of the molecule in the genome of the organism in which it naturally occurs; (2) a nucleic acid molecule incorporated into a vector or into the genomic DNA of a prokaryote or eukaryote in a manner such that the resulting molecule is not identical to any naturally occurring vector or genomic DNA; (3) a separate molecule such as a cDNA, a genomic fragment, a fragment produced by polymerase chain reaction (PCR), or a restriction fragment; (4) recombinant DNAs; and (5) synthetic DNAs.
  • An isolated nucleic acid molecule may also be comprised of one or more segments of cDNA, genomic DNA or synthetic DNA.
  • the isolated nucleic acid molecules of the present invention also include known types of modifications, for example, labels which are known in the art, methylation, "caps", substitution of one or more of the naturally occurring nucleotides with an analog.
  • internucleotide modifications for example, those with uncharged linkages (methyl phosphonates, phosphotriesters, phosphoamidates, carbamates, etc.) and with charged linkages (phosphorothioates, phosphorodithioates, etc.), those containing pendant moieties, such as, proteins (including nucleases, toxins, antibodies, signal peptides, poly-L-lysine, etc.), those with intercalators (acridine, psoralen, etc.), those containing chelators (metals, radioactive metals, boron, oxidative metals, etc.), those containing alkylators, and those with modified linkages.
  • uncharged linkages methyl phosphonates, phosphotriesters, phosphoamidates, carbamates, etc.
  • charged linkages phosphorothioates, phosphorodithioates, etc.
  • pendant moieties such as, proteins (including nucleases, toxins, antibodies, signal peptides, poly-
  • nucleotide sequence means the order contiguous of nucleic acid molecule of both the sense and antisense strands or as a duplex strand. It includes, but is not limited to, self-replicating plasmids, synthetic polynucleotides, chromosomal sequences, and infectious polymers of DNA or RNA.
  • a nucleotide sequence is said to be the "complement” of another nucleotide sequence if they exhibit complete complementarity.
  • molecules are said to exhibit "complete complementarity" when every nucleotide of one of the sequences is complementary to a nucleotide of the other.
  • a coding sequence and "a structural nucleotide sequence” mean a nucleotide sequence, which is translated into a polypeptide, usually via mRNA, when placed under the control of appropriate regulatory sequences. The boundaries of the coding sequence are determined by a translation start codon at the 5 '-terminus and a translation stop codon at the 3 '-terminus.
  • a coding sequence can include, but is not limited to, genomic DNA, cDNA, and recombinant nucleotide sequences.
  • polypeptides of the invention like other polypeptides, have different domains which perfo ⁇ n different functions. Thus, the coding sequences need not be full length, so long as the desired functional domain of the polypeptide is expressed.
  • the distinguishing features of polypeptides of the present invention are discussed in detail in Examples.
  • the term "recombinant DNAs" as used herein means DNAs that contains a genetically engineered modification through manipulation via mutagenesis, restriction enzymes, and the like.
  • synthetic DNAs as used herein means DNAs assembled from oligonucleotide building blocks that are chemically synthesized using procedures known to those skilled in the art. These building blocks are ligated and annealed to form DNA segments, which are then enzymatically assembled to construct the entire DNA. "Chemically synthesized”, as related to a sequence of DNA, means that the component nucleotides were assembled in vitro. Manual chemical synthesis of DNA may be accomplished using well-established procedures, or automated chemical synthesis can be performed using one of a number of commercially available machines.
  • polypeptide and protein mean a polymer composed of amino acids connected by peptide bonds.
  • An amino acid unit in a polypeptide (or protein) is called a residue.
  • polypeptide and protein also applies to any amino acid polymers in which one or more amino acid residue is an artificial chemical analogue- of a corresponding naturally occurring amino acid, as well as to any naturally occurring amino acid polymers.
  • the essential nature of such analogues of naturally occurring amino acids is that, when incorporated into a polypeptide, that polypeptide is specifically reactive to antibodies elicited to the same polypeptide but consisting entirely of naturally occurring amino acids.
  • proteins or polypeptides may undergo modification, including but not limited to, disulfide bond formation, gamma-carboxylation of glutamic acid residues, glycosylation, lipid attachment, phosphorylation, oligomerization, hydroxylation and ADP-ribosylation.
  • modification including but not limited to, disulfide bond formation, gamma-carboxylation of glutamic acid residues, glycosylation, lipid attachment, phosphorylation, oligomerization, hydroxylation and ADP-ribosylation.
  • Exemplary modifications are described in most basic texts, such as, for example, Proteins - Structure and Molecular Properties, 2nd ed., T. E. Creighton, W. H. Freeman and Company, New York (1993), herein inco ⁇ orated by reference in its entirety. Many detailed reviews are available on this subject, such as, for example, those provided by Wold, F., Post-translational Protein Modifications. Perspectives and Prospects, pp.
  • polypeptides blockages of the amino or carboxyl group in a polypeptide, or both, by a covalent modification, is common in naturally occurring and synthetic polypeptides and such modifications may be present in polypeptides of the present invention, as well.
  • a methionine residue at the NH 2 terminus may be deleted.
  • this invention contemplates the use of both the methionine containing and the methionine-less amino terminal variants of the polypeptide of the invention.
  • amino acid and “amino acids” refer to all naturally occurring amino acids and, unless otherwise limited, known analogs of natural amino acids that can function in a similar manner as naturally occurring amino acids. This definition is meant to include ⁇ norleucine, ornithine, homocysteine, and homoserine.
  • amino acid sequence means the sequence of amino acids in a polypeptide (or protein) that is written starting with the amino-terminal (N-terminal) residue and ending with the carboxyl-terminal (C-terminal) residue.
  • an amino acid subsequence means a portion of the amino acid sequence of a polypeptide.
  • An amino acid subsequence generally has a length of 3 to 50 amino acid residues.
  • substantially purified polypeptide and “substantially purified protein”, as used herein, means a polypeptide or protein that is separated substantially from all other molecules normally associated with it in its native state and is the predominant species present in a preparation.
  • a substantially purified molecule may be greater than 60% free, preferably 75% free, more preferably 90% free, and most preferably 95% free from the other molecules (exclusive of solvent) present in the natural mixture.
  • Percentage of sequence identity is determined by comparing two optimally aligned sequences over a comparison window, wherein the portion of the polynucleotide or amino acid sequence in the comparison window may comprise additions or deletions (i.e., gaps) as compared to the reference sequence (which does not comprise additions or deletions) for optimal alignment of the two sequences.
  • the percentage is calculated by determining the number of positions at which the identical nucleic acid base or amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the window of comparison and multiplying the result by 100 to yield the percentage of sequence identity.
  • identity refers to amino acid or nucleic acid sequences that when compared using the local homology algorithm of Smith and Waterman (T. F. Smith and M. S. Waterman, J. Mol. Biol. (147) 195-197 (1981). in the SSEARCH3 3.0t75 (W.R. Pearson, Genomics (11)635- 650 (1991) or BLAST 2.2.1 (Altschul, Stephen F., Thomas L. Madden, Alejandro A. Schaffer, Jinghui Zhang, Zheng Zhang, Webb Miller, and David J. Lipman , Nucleic Acids Res. (25)3389- 3402 (1997) programs which are exactly alike.
  • similarity refers to two amino acids, which are similar as defined by similarity matrix BLOSSUM62 (S. Henikoff and J. G. Henikoff, , Proc. Natl. Acad. Sci. U.S.A. (89) 10915-10919 (1992) which is used in BLAST 2.2.1 (Altschul, Stephen F., Thomas L. Madden, Alejandro A. Schaffer, Jinghui Zhang, Zheng Zhang, Webb Miller, and David J. Lipman , Nucleic Acids Res. (25)3389-3402 (1997).
  • the BLAST uses alias "positive” for similarity.
  • percent identity for a pair of protein sequences refers to the number of identical amino acid residues in a two-sequence alignment reported by BLAST, divided by the total number of amino acid residues in the same alignment, expressed in percentage.
  • percent similarity for a pair of protein sequences refers to the number of similar (“Positive” in BLAST output) amino acid residues in a two-sequence alignment reported by BLAST, divided by the total number of amino acid residues in the same alignment, expressed in percentage.
  • Polypeptides which are "substantially similar” share sequences as noted above except that residue positions which are not identical may differ by conservative amino acid changes.
  • Conservative amino acid substitutions refer to the interchangeability of residues having similar side chains.
  • Constant amino acid substitutions mean substitutions of one or more amino acids in a native amino acid sequence with another amino acid(s) having similar side chains, resulting in a silent change. conserveed substitutes for an amino acid within a native amino acid sequence can be selected from other members of the group to which the naturally occurring amino acid belongs.
  • a group of amino acids having aliphatic side chains is glycine, alanine, valine, leucine, and isoleucine; a group of amino acids having aliphatic-hydroxyl side chains is serine and threonine; a group of amino acids having amide-containing side chains is asparagine and glutamine; a group of amino acids having aromatic side chains is phenylalanine, tyrosine, and tryptophan; a group of amino acids having basic side chains is lysine, arginine, and histidine; and a group of amino acids having sulfur-containing side chains is cysteine and methionine.
  • Preferred conservative amino acids substitution groups are: valine-leucine, valine- isoleucine, phenylalanine-tyrosine, lysine-arginine, alanine-valine, aspartic acid-glutamic acid, and asparagine-glutamine.
  • nucleotide sequences can be appropriately adjusted to determine corresponding sequence identity of two nucleotide sequences encoding the polypeptides of the present invention by taking into account codon degeneracy, conservative amino acid substitutions, reading frame positioning and the like. Substantial identity of nucleotide sequences for these purposes normally means sequence identity of at least 35%).
  • codon degeneracy means divergence in the genetic code permitting variation of the nucleotide sequence without affecting the amino acid sequence of an encoded polypeptide.
  • codon-bias exhibited by a specific host cell in usage of nucleotide codons to specify a given amino acid.
  • the present invention provides an isolated nucleic acid molecule in a DNA construct comprising a promoter operably linked to nucleotide sequence or complement thereof, wherein the nucleotide sequence hybridizes under stringent conditions to the complement of a second nucleotide sequence encoding a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10, SEQ ID NO: 12, SEQ ID NO: 14, SEQ ID NO: 16, SEQ ID NO: 18, SEQ ID NO: 20, SEQ ID NO: 22, SEQ ID NO: 24, SEQ ID NO: 26, SEQ ID NO: 28, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 34, SEQ ID NO: 36, SEQ ID NO:
  • the present invention also provides a method for obtaining an isolated nucleic acid molecule of the present invention, the method comprising the steps of: (a) probing a cDNA or genomic library with a hybridization probe comprising a nucleotide sequence encoding all or a portion of the amino acid sequence of a polypeptide, wherein the amino acid sequence of the polypeptide is selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 34, SEQ ID NO: 36, SEQ ID NO: 38, SEQ ID NO: 40, SEQ ID NO: 42, SEQ ID NO: 44, SEQ ID NO: 46, SEQ ID NO: 48, SEQ ID NO: 52, SEQ ID NO: 54, SEQ ID NO: 67, SEQ ID NO: 69, and SEQ ID NO: 71 (b) identifying a DNA clone that hybridizes under stringent conditions to hybridization probe; (c) isolating the DNA clone identified in step (b); and (d)
  • Hybridization conditions are sequence dependent and will be different in different circumstances.
  • stringent conditions are selected to be about 5°C lower than the thermal melting point (Tm) for the specific sequence at a defined ionic strength and pH.
  • the "thermal melting point” is the temperature (under defined ionic strength and pH) at which 50% of a target molecule hybridizes to a completely complementary molecule.
  • Appropriate stringent conditions which promote DNA hybridization for example, 6.0 X sodium chloride/sodium citrate (SSC) at about 45°C, followed by a wash of 2.0 X SSC at 50°C, are known to those skilled in the art or can be found in Current Protocols in Molecular Biology, John Wiley & Sons, N.Y.
  • the salt concentration in the wash step can be selected from a low stringent condition of about 2.0 X SSC at 50°C to a high stringency of about 0.2 X SSC at 50°C.
  • the temperature in the wash step can be increased from low stringent conditions at room temperature, about 22°C, to high stringent conditions at about 65°C. Both temperature and salt concentration may be varied, or either the temperature or the salt concentration may be held constant while the other variable is changed.
  • stringent conditions include at least one wash in 2.0 X SSC at a temperature of at least about 50°C for 20 minutes, or equivalent conditions.
  • nucleic acid molecules of the present invention may be combined with other non- native, or “heterologous” sequences in a variety of ways.
  • heterologous sequences it is meant any sequence that is not naturally found joined to the nucleotide sequence encoding polypeptide of the present invention, including, for example, combinations of nucleotide sequences from the same plant which are not naturally found joined together, or the two sequences originate from two different species.
  • homologous refers to two or more genes that are derived from a single gene in a common ancestor.
  • homoology is attributed to decent from a common ancestor.
  • orthologous refers to different homologous sequences in different species that arose from a common ancestral gene during speciation; may or may not be responsible for a similar function.
  • paralogous refers to homologous sequences within a single species that arose by gene duplication.
  • domain refers to a discrete portion of a protein assumed to fold independently of the rest of the protein and possessing its own function. Domain is also used to refer a discrete portion of nucleotide when translated provides a portion of protein which is assumed to fold independently of the rest of the translated nucleotide sequence and possessing its own function.
  • Motif refers to a short conserved region in a protein or nucleotide sequence. Motifs are frequently highly conserved parts of domains.
  • the term "genome” as it applies to plant cells encompasses not only chromosomal DNA found within the nucleus, but organelle DNA found within subcellular components of the cell. DNAs of the present invention introduced into plant cells can therefore be either chromosomally integrated or organelle-localized.
  • operably linked as used in reference to a regulatory sequence and a structural nucleotide sequence, means that the regulatory sequence causes regulated expression of the operably linked structural nucleotide sequence.
  • “Expression” means the transcription and stable accumulation of sense or antisense RNA derived from the nucleic acid molecule of the present invention. Expression may also refer to translation of mRNA into a polypeptide.
  • RNA transcript means RNA transcript that includes the mRNA and so can be translated into polypeptide or protein by the cell.
  • Antisense RNA means a RNA transcript that is complementary to all or part of a target primary transcript or mRNA and that blocks the expression of a target gene (U.S. Pat. No. 5,107,065, incorporated herein by reference). The complementarity of an antisense RNA may be with any part of the specific gene transcript, i.e., at the 5' non-coding sequence, 3' non- translated sequence, introns, or the coding sequence.
  • RNA transcript means the product resulting from RNA polymerase-catalyzed transcription of a DNA sequence. When the RNA transcript is a perfect complementary copy of the DNA sequence, it is referred to as the primary transcript or it may be a RNA sequence derived from post-transcriptional processing of the primary transcript and is referred to as the mature RNA.
  • overexpression means the expression of a polypeptide encoded by an exogenous nucleic acid molecule introduced into a host cell, wherein said polypeptide is either not normally present in the host cell, or wherein said polypeptide is present in said host cell at a higher level than that normally expressed from the endogenous gene encoding said polypeptide.
  • ectopic expression it is meant that expression of a nucleic acid molecule encoding a polypeptide in a cell type other than a cell type in which the nucleic acid molecule is normally expressed, at a time other than a time at which the nucleic acid molecule is normally expressed or at a expression level other than the level at which the nucleic acid molecule normally is expressed.
  • Antisense inhibition means the production of antisense RNA transcripts capable of suppressing the expression of the target polypeptide.
  • Co-suppression means the production of sense RNA transcripts capable of suppressing the expression of identical or substantially similar foreign or endogenous genes (U.S. Patent No. 5,231,020, incorporated herein by reference).
  • a gene means the segment of DNA that is involved in producing a polypeptide. Such segment of DNA includes regulatory sequences preceding (5' non-coding sequences) and following (3' non-coding sequences) the coding region as well as intervening sequences (introns) between individual coding segments (exons).
  • a "Native gene” means a gene as found in nature with its own regulatory sequences.
  • Chimeric gene means any gene that is not a native gene, comprising regulatory and coding sequences that are not found together in nature. Accordingly, a chimeric gene may comprise regulatory sequences and coding sequences that are derived from different sources, or regulatory sequences and coding sequences derived from the same source, but arranged in a manner different than that found in nature.
  • Endogenous gene or "endogenous DNA molecule” means a native gene in its natural location in the genome of an organism.
  • a “foreign gene” means a gene not normally found in the host organism, but that is introduced into the host organism by gene transfer.
  • Foreign genes can comprise native genes inserted into a non-native organism, or chimeric genes.
  • a “transgene” is a gene that has been introduced into the genome by a transformation procedure.
  • Regulatory sequences mean nucleotide sequences located upstream (5' non-coding sequences), within, or downstream (3' non-translated sequences) of a structural nucleotide sequence, and which influence the transcription, RNA processing or stability, or translation of the associated structural nucleotide sequence. Regulatory sequences may include promoters, translation leader sequences, introns, and polyadenylation recognition sequences.
  • promoter sequence means a nucleotide sequence that is capable of, when located in cis to a structural nucleotide sequence encoding a polypeptide, functioning in a way that directs expression of one or more mRNA molecules that encodes the polypeptide. Such promoter regions are typically found upstream of the trinucleotide ATG sequence at the start site of a polypeptide-coding region. Promoter sequences can also include sequences from which transcription of transfer RNA (tRNA) or ribosomal RNA (rRNA) sequences are initiated. Transcription involves the synthesis of a RNA chain representing one strand of a DNA duplex.
  • tRNA transfer RNA
  • rRNA ribosomal RNA
  • RNA polymerase By “representing” it is meant that the RNA is identical in sequence with one strand of the DNA; it is complementary to the other DNA strand, which provides the template for its synthesis. Transcription takes place by the usual process of complementary base pairing, catalyzed and scrutinized by the enzyme RNA polymerase. The reaction can be divided into three stages described as initiation, elongation and termination. Initiation begins with the binding of RNA polymerase to the double stranded (DS or ds) DNA. The sequence of DNA required for the initiation reaction defines the promoter. The site at which the first nucleotide is incorporated is called the start-site or start-point of transcription. Elongation describes the phase during which the enzyme moves along the DNA and extends the growing RNA chain.
  • Elongation involves the disruption of the DNA double stranded structure in which a transiently unwound region exists as a hybrid RNA-DNA duplex and a displaced single strand of DNA. Termination involves recognition of the point at which no further bases should be added to the chain. To terminate transcription, the formation of phosphodiester bonds must cease and the transcription complex must come apart. When the last base is added to the RNA chain, the RNA-DNA hybrid is disrupted, the DNA reforms into a duplex state, and the RNA polymerase enzyme and RNA molecule are both released from the DNA. The sequence of DNA required for the termination reaction is called the terminator.
  • the promoter sequence consists of proximal and more distal upstream elements, the latter elements often referred to as enhancers.
  • an “enhancer” is a DNA sequence that can stimulate promoter activity and may be an innate element of the promoter or a heterologous element inserted to enhance the level or tissue-specificity of a promoter. Promoters may be derived in their entirety from a native gene, or be composed of different elements derived from different promoters found in nature, or even comprise synthetic DNA segments. It is understood by those skilled in the art that different promoters may direct the expression of a gene in different tissues or cell types, or at different stages of development, or in response to different environmental conditions.
  • Promoters which are known or are found to cause transcription of DNA in plant cells, can be used in the present invention. Such promoters may be obtained from a variety of sources such as plants and plant viruses. A number of promoters, including constitutive promoters, inducible promoters and tissue-specific promoters, that are active in plant cells have been described in the literature. In addition to promoters that are known to cause transcription of DNA in plant cells, other promoters may be identified for use in the current invention by screening a plant cDNA library for genes that are selectively or preferably expressed in the target tissues and then determine the promoter regions.
  • constitutive promoter means a regulatory sequence which causes expression of a structural nucleotide sequence in most cells or tissues at most times. Constitutive promoters are active under most environmental conditions and states of development or cell differentiation. A variety of constitutive promoters are well known in the art.
  • constitutive promoters that are active in plant cells include but are not limited to the nopaline synthase (NOS) promoters; DNA plant virus promoters including, but not limited to the caulimovirus promoters for example, cauliflower mosaic virus (CaMV) 19S and 35S, and figwort mosaic virus promoters; the bacilliform virus promoter for example sugar cane bacilliform virus, rice tungro bacilliform virus, among others; plant actin promoters, such as the Arabidopsis and rice actin gene promoter (see, e.g., Huang et al, Plant Mol. Biol. 33:125-139 (1997), US Patent No. 5,641,876), herein incorporated by reference in its entirety). These promoters when used in a DNA construct are heterologous to the linked gene sequence when they are derived from a different organism, plant species, or a different gene.
  • NOS nopaline synthase
  • DNA plant virus promoters including, but not limited to the caul
  • inducible promoter means a regulatory sequence which causes conditional expression of a structural nucleotide sequence under the influence of changing environmental conditions or developmental conditions.
  • inducible promoters include but are not limited to the senescence-induced promoter for the senescence-associated gene, SAG 12, (Gan and Amasino, Science 270: 1986-1988 (1995), herein incorporated by reference in its entirety); the light-inducible promoter from the small subunit of ribulose-l,5-bis-phosphate carboxylase (ssRUBISCO); the drought-inducible promoter of maize (Busk et al, Plant J.
  • auxin-response elements El promoter fragment (AuxREs) in the soybean (Glycine max L.) (Liu et al, Plant Physiol. 775:397-407 (1997), herein incorporated by reference in its entirety); the auxin-responsive Arabidopsis GST6 promoter (also responsive to salicylic acid and hydrogen peroxide) (Chen et al, Plant J. 10: 955-966 (1996), herein inco ⁇ orated by reference in its entirety); the auxin- inducible parC promoter from tobacco (Sakai et al, Plant Cell Physiol.
  • tissue-specific promoter or "organ enhanced promoter” means a regulatory sequence that causes transcriptions or enhanced transcriptions of DNA in specific cells or tissues at specific times during plant development, such as in vegetative tissues or reproductive tissues.
  • tissue-specific promoters under developmental control include promoters that initiate transcription only (or primarily only) in certain tissues, such as vegetative tissues, e.g., roots, leaves or stems, or reproductive tissues, such as fruit, ovules, seeds, pollen, pistols, flowers, or any embryonic tissue.
  • Reproductive tissue specific promoters may be, e.g., ovule- specific, embryo-specific, endosperm-specific, integument-specific, seed coat-specific, pollen- specific, petal-specific, sepal-specific, or some combination thereof.
  • tissue-specific promoter may drive expression of operably linked sequences in tissues other than the target tissue.
  • a tissue-specific promoter is one that drives expression preferentially in the target tissue, but may also lead to some expression in other tissues as well.
  • tuber-specific promoters include but are not limited to the class I and II patatin promoters (Bevan et al, EMBO J. 8: 1899-1906 (1986); Koster-Topfer et al, Mol Gen Genet. 219: 390-396 (1989); Mignery et al, Gene. 62: 27-44 (1988); Jefferson et al, Plant Mol.
  • leaf-specific promoters include but are not limited to the ribulose biphosphate carboxylase (RBCS or RuBISCO) promoters (see, e.g., Matsuoka et al, Plant J. 6:311-319 (1994), herein inco ⁇ orated by reference in its entirely); the light harvesting chlorophyll a/b binding protein gene promoter (see, e.g., Shhna et ⁇ /., R/ ⁇ ..tR/nw ⁇ /. 775:477-483 (1997); Cass ⁇ et al, Plant Physiol.
  • RBCS or RuBISCO ribulose biphosphate carboxylase
  • Root-specific promoter examples include but are not limited to the promoter for the acid chitinase gene (Samac et al, Plant Mol. Biol 25: 587-596 (1994), herein inco ⁇ orated by reference in its entirety); the root specific subdomains of the CaMV35S promoter that have been identified (Lam et al, Proc. Natl. Acad. Sci.
  • Another class of useful vegetative tissue-specific promoters is meristematic (root tip and shoot apex) promoters.
  • meristematic (root tip and shoot apex) promoters For example, the "SHOOTMERISTEMLESS” and “SCARECROW” promoters, which are active in the developing shoot or root apical meristems (Di Laurenzio et al, Cell 86:423- 433 (1996); Long, Nature 379:66-69 (1996); herein inco ⁇ orated by reference in their entireties), can be used.
  • Another example of a useful promoter is that which controls the expression of 3-hydroxy-3- methylglutaryl coenzyme A reductase HMG2 gene, whose expression is restricted to meristematic and floral (secretory zone of the stigma, mature pollen grains, gynoecium vascular tissue, and fertilized ovules) tissues (see, e.g., Enjuto et al, Plant Cel 7:517-527 (1995), herein incorporated by reference in its entirety).
  • Another example of a useful promoter is that which controls the expression of knl-related genes from maize and other species which show meristem-specific expression (see, e.g., Granger et al, Plant Mol Biol.
  • KNAT1 Arabidopsis thaliana KNAT1 promoter.
  • KNAT1 transcript is localized primarily to the shoot apical meristem; the expression of KNATI in the shoot meristem decreases during the floral transition and is restricted to the cortex of the inflorescence stem (see, e.g., Lincoln et al, Plant Cell
  • Suitable seed-specific promoters can be derived from the following genes: MAC1 from maize (Sheridan et al, Genetics 742:1009-1020 (1996), herein inco ⁇ orated by reference in its entirety); Cat3 from maize (GenBank No. L05934, Abler et al, Plant Mol. Biol. 22:10131-1038
  • GenBank No. U39944; Ray et al, Proc. Natl. Acad. Sci. USA 91:5161-5165 (1994), all of which are herein incorporated by reference in their entireties) can also be used.
  • the egg and central cell specific MEA (FIS1) and FIS2 promoters are also useful reproductive tissue-specific promoters (Luo et al., Proc. Natl. Acad. Sci. USA, 97:10637-10642 (2000); Dahlle-Calzada, et al., Genes
  • a maize pollen-specific promoter has been identified in maize (Guerrero et al., Mol. Gen.
  • Promoters derived from genes encoding embryonic storage proteins which includes the gene encoding the 2S storage protein from Brassica napus (Dasgupta et al, Gene 133:301-302 (1993), herein incorporated by reference in its entirety); the 2S seed storage protein gene family from Arabidopsis; the gene encoding oleosin 20kD from Brassica napus (GenBank No.
  • Promoters derived from zein encoding genes can be also used.
  • the zeins are a group of storage proteins found in maize endosperm.
  • promoters known to function include the promoters for the following genes: waxy, Brittle, Shrunken 2, Branching enzymes I and II, starch synthases, debranching enzymes, oleosins, glutelins, and sucrose synthases.
  • a particularly preferred promoter for maize endosperm expression is the promoter for the glutelin gene from rice, more particularly the Osgt-1 promoter (Zheng et al, Mol. Cell Biol. 13: 5829-5842 (1993), herein inco ⁇ orated by reference in its entirety).
  • promoters suitable for expression in wheat include those promoters for the ADPglucose pyrophosphorylase (ADPGPP) subunits, the granule bound and other starch synthases, the branching and debranching enzymes, the embryogenesis-abundant proteins, the gliadins, and the glutenins.
  • promoters in rice include those promoters for the ADPGPP subunits, the granule bound and other starch synthases, the branching enzymes, the debranching enzymes, sucrose synthases, and the glutelins.
  • a particularly preferred promoter is the promoter for rice glutelin, Osgt-1.
  • promoters for barley include those for the ADPGPP subunits, the granule bound and other starch synthases, the branching enzymes, the debranching enzymes, sucrose synthases, the hordeins, the embryo globulins, and the aleurone specific proteins.
  • a tomato promoter active during fruit ripening, senescence and abscission of leaves and, to a lesser extent, of flowers can be used (Blume et al., Plant J. 72:731-746 (1997), herein incorporated by reference in its entirety).
  • Other exemplary promoters include the pistol specific promoter in the potato (Solarium tuberosum L.) SK2 gene, encoding a pistil-specific basic endochitinase (Ficker et al, Plant Mol Biol. 35:425-431 (1997), herein inco ⁇ orated by reference in its entirety); the Blec4 gene from pea (Pisum sativum cv.
  • tissue specific E8 promoter from tomato is also useful for directing gene expression in fruits. It is recognized that additional promoters that may be utilized are described, for example, in U.S. Patent Nos. 5,378,619, 5,391,725, 5,428,147, 5,447,858, 5,608,144, 5,608,144, 5,614,399, 5,633,441, 5,633,435, and 4,633,436, all of which are herein inco ⁇ orated in their entirety.
  • tissue specific enhancer may be used (Fromm et al., The Plant Cell 7:977-984 (1989), herein inco ⁇ orated by reference in its entirety). It is further recognized that since in most cases the exact boundaries of regulatory sequences have not been completely defined, DNA fragments of different lengths may have identical promoter activity.
  • the "translation leader sequence” means a DNA sequence located between the promoter sequence of a gene and the coding sequence.
  • the translation leader sequence is present in the fully processed mRNA upstream of the translation start sequence.
  • the translation leader sequence may affect processing of the primary transcript to mRNA, mRNA stability or translation efficiency. Examples of translation leader sequences have been described (US Patent No. 5,659,122 and Turner and Foster, Molecular Biotechnology 3:225 (1995) herein inco ⁇ orated by reference in its entirety).
  • the "3' non-translated sequences” means DNA sequences located downstream of a structural nucleotide sequence and include sequences encoding polyadenylation and other regulatory signals capable of affecting mRNA processing or gene expression.
  • the polyadenylation signal functions in plants to cause the addition of polyadenylate nucleotides to the 3' end of the mRNA precursor.
  • the polyadenylation sequence can be derived from the natural gene, from a variety of plant genes, or from T-DNA.
  • An example of the polyadenylation sequence is the nopaline synthase 3' sequence (NOS 3'; Fraley et al, Proc. Natl. Acad. Sci. USA 80: 4803-4807 (1983), herein inco ⁇ orated by reference in its entirety).
  • NOS 3' Fraley et al, Proc. Natl. Acad. Sci. USA 80: 4803-4807 (1983
  • the use of different 3' non-translated sequences is exemplified by Ingelbrecht et al, Plant Cell 7:671-680 (1989), herein inco ⁇ orated by reference in its entirety.
  • the isolated nucleic acid molecules of the present invention may also include introns.
  • an intron sequence is inserted between the promoter sequence and the structural gene sequence or, optionally, may be inserted in the structural coding sequence to provide an interrupted coding sequence.
  • An example of such an intron sequence is the HSP 70 intron described in WO 93/19189, herein inco ⁇ orated by reference in its entirety.
  • the laboratory procedures in recombinant DNA technology used herein are those well known and commonly employed in the art. Standard techniques are used for cloning, DNA and RNA isolation, amplification and purification. Generally enzymatic reactions involving DNA ligase, DNA polymerase, restriction endonucleases and the like are performed according to the manufacturer's specifications. These techniques and various other techniques are generally performed according to Sambrook et al., Molecular Cloning - A Laboratory Manual, 2nd. ed., Cold Spring Harbor Laboratory, Cold Spring Harbor, New York (1989).
  • nucleic acid molecules encoding polypeptides of the present invention from soybean, corn, rice and other species are described in detail in Examples. All or a substantial portion of the nucleic acid molecules of the present invention may be used to isolate cDNAs and nucleic acid molecules encoding homologous polypeptides from the same or other plant species.
  • a "substantial portion" of a nucleotide sequence comprises enough of the sequence to afford specific identification and/or isolation of a nucleic acid molecule comprising the sequence.
  • Nucleotide sequences can be evaluated either manually by one skilled in the art, or by using computer-based sequence comparison and identification tools that employ algorithms such as BLAST (Basic Local Alignment Search Tool; Altschul et al. JMol Biol 275:403-410 (1993). In general, a sequence of thirty or more contiguous nucleotides is necessary in order to putatively identify a nucleotide sequence as homologous to a gene.
  • gene-specific oligonucleotide probes comprising 30 or more contiguous nucleotides may be used in sequence-dependent methods of gene identification (e.g., Southern hybridization) and isolation (e.g., in situ hybridization of bacterial colonies or bacteriophage plaques).
  • short oligonucleotides of 12 or more nucleotides may be used as amplification primers in PCR in order to obtain a particular nucleic acid molecule comprising the primers.
  • sequence-dependent protocols include, but are not limited to, methods of nucleic acid molecule hybridization, and methods of DNA and RNA amplification as exemplified by various uses of nucleic acid molecule amplification technologies (e.g., polymerase chain reaction, ligase chain reaction).
  • nucleic acid molecules encoding other polypeptide of the present invention could be isolated directly by using all or a substantial portion of the nucleic acid molecules of the present invention as DNA hybridization probes to screen cDNA or genomic libraries from any desired plant employing methodology well known to those skilled in the art. Methods for forming such libraries are well known in the art. Specific oligonucleotide probes based upon the nucleic acid molecules of the present invention can be designed and synthesized by methods known in the art.
  • nucleic acid molecules can be used directly to synthesize DNA probes by methods known to the skilled artisan such as random primer DNA labeling, nick translation, or end-labeling techniques, or RNA probes using available in vitro transcription systems.
  • specific primers can be designed and used to amplify a part or all of the sequences.
  • the resulting amplification products can be labeled directly during amplification reactions or labeled after amplification reactions, and used as probes to isolate full-length cDNA or genomic DNAs under conditions of appropriate stringency.
  • the nucleic acid molecules of interest can be amplified from nucleic acid samples using amplification techniques.
  • the disclosed nucleic acid molecules may be used to define a pair of primers that can be used with the polymerase chain reaction (Mullis, et al, Cold Spring Harbor Symp. Quant. Biol. 57:263-273 (1986); Erlich et al, EP 50,424; EP 84,796, EP 258,017, EP 237,362; Mullis, EP 201,184; Mullis et al, US 4,683,202; Erlich, US 4,582,788; and Saiki, R.
  • PCR and other in vitro amplification methods may also be useful, for example, to clone nucleotide sequences that encode for polypeptides to be expressed, to make nucleic acid molecules to use as probes for detecting the presence of the desired mRNA in samples, for nucleic acid sequencing, or for other purposes.
  • two short segments of the nucleic acid molecules of the present invention may be used in polymerase chain reaction protocols to amplify longer nucleic acid molecules encoding homologues of a polypeptide of the present invention from DNA or RNA.
  • the skilled artisan can follow the RACE protocol (Frohman et al., Proc. Natl. Acad. Sci. USA 85:8998 (1988), herein inco ⁇ orated by reference in its entirety) to generate cDNAs by using PCR to amplify copies of the region between a single point in the transcript and the 3 ' or 5' end.
  • Primers oriented in the 3' and 5' directions can be designed from the nucleic acid molecules of the present invention.
  • 3 'RACE or 5 'RACE systems Using commercially available 3 'RACE or 5 'RACE systems (Gibco BRL, Life Technologies, Gaithersburg, Maryland U.S.A.), specific 3' or 5' cDNA fragments can be isolated (Ohara et al., Proc. Natl. Acad. Sci. USA 86:5673 (1989); Loh et al., Science 243:217 (1989), both of which are herein inco ⁇ orated by reference in their entireties). Products generated by the 3' and 5' RACE procedures can be combined to generate full-length cDNAs (Frohman and Martin, Techniques ! : 165 (1989), herein incorporated by reference in its entirety).
  • Nucleic acid molecules of interest may also be synthesized, either completely or in part, especially where it is desirable to provide plant-preferred sequences, by well-known techniques as described in the technical literature. See, e.g., Carruthers et al., Cold Spring Harbor Symp. Quant. Biol. 47:411-418 (1982), and Adams et al., J. Am. Chem. Soc. 105:661 (1983), both of which are herein inco ⁇ orated by reference in their entireties. Thus, all or a portion of the nucleic acid molecules of the present invention may be synthesized using codons preferred by a selected plant host. Plant-preferred codons may be determined, for example, from the codons used most frequently in the proteins expressed in a particular plant host species. Other modifications of the gene sequences may result in mutants having slightly altered activity
  • nucleotide sequences encoding polypeptide of the present invention facilitates immunological screening of cDNA expression libraries.
  • Synthetic polypeptides representing portions of the amino acid sequences of polypeptides of the present invention may be synthesized. These polypeptides can be used to immunize animals to produce polyclonal or monoclonal antibodies with specificity for polypeptides comprising the amino acid sequences. These antibodies can be then be used to screen cDNA expression libraries to isolate full-length cDNA clones of interest (Lemer, Adv. Immunol. 36: 1 (1984); Sambrook et al., Molecular Cloning: A Laboratory Manual; Cold Spring Harbor Laboratory Press: Cold Spring Harbor, (1989)).
  • Another aspect of the present invention relates to methods of making a DNA construct by obtaining a nucleic acid molecule comprising a nucleotide sequence encoding a polypeptide of the present invention, the amino acid sequence of which has at least 50% sequence identity to a polypeptide selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10, SEQ ID NO: 12, SEQ ID NO: 14, SEQ ID NO: 16, SEQ ID NO: 18, SEQ ID NO: 20, SEQ ID NO: 22, SEQ ID NO: 24, SEQ ID NO: 26, SEQ ID NO: 28, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 34, SEQ ID NO: 36, SEQ ID NO: 38, SEQ ID NO: 40, SEQ ID NO: 42, SEQ ID NO: 44, SEQ ID NO: 46, SEQ ID NO: 48, SEQ ID NO: 50, SEQ ID NO: 52, SEQ ID NO: 54, SEQ ID NO:
  • nucleic acid molecule comprising a nucleotide sequence or complement thereof, wherein the nucleotide sequence encodes a polypeptide having an amino acid sequence that has at least 50% sequence identity, preferably at least 60%, more preferably at least 70% sequence identity, even more preferably at least 80% or 90% sequence identity, and most preferably at least 95% to 98% sequence identity to a polypeptide selected from group consisting of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10, SEQ ID NO: 12, SEQ ID NO: 14, SEQ ID NO: 16, SEQ ID NO: 18, SEQ ID NO: 20, SEQ ID NO: 22, SEQ ID NO: 24, SEQ ID NO: 26, SEQ ID NO: 28, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 34, SEQ ID NO: 36, SEQ ID NO: 38, SEQ ID NO: 40, SEQ ID NO: 42, SEQ ID NO: 44, SEQ ID NO:
  • One method of the present invention is for obtaining a nucleic acid molecule encoding all or a substantial portion of the amino acid sequence of a polypeptide of the present invention comprising: (a) probing a cDNA or genomic library with a hybridization probe comprising a nucleotide sequence encoding all or a substantial portion of a polypeptide having an amino acid sequence set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10, SEQ ID NO: 12, SEQ ID NO: 14, SEQ ID NO: 16, SEQ ID NO: 18, SEQ ID NO: 20, SEQ ID NO: 22, SEQ ID NO: 24, SEQ ID NO: 26, SEQ ID NO: 28, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 34, SEQ ID NO: 36, SEQ ID NO: 38, SEQ ID NO: 40, SEQ ID NO: 42, SEQ ID NO: 44, SEQ ID NO: 46, SEQ ID NO: 48, SEQ
  • Another method of the present invention for obtaining a nucleic acid molecule encoding all or a substantial portion of the amino acid sequence of a polypeptide of the present invention comprising: (a) synthesizing a first and a second oligonucleotide primers, wherein the sequences of the first and second oligonucleotide primers encode two different portions of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10, SEQ ID NO: 12, SEQ ID NO: 14, SEQ ID NO: 16, SEQ ID NO: 18, SEQ ID NO: 20, SEQ ID NO: 22, SEQ ID NO: 24, SEQ ID NO: 26, SEQ ID NO: 28, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 34, SEQ ID NO: 36, SEQ ID NO: 38, SEQ ID NO: 40, SEQ ID NO: 42, SEQ ID NO: 44, SEQ ID NO
  • nucleic acid molecules of the present invention may also be used as probes for genetically and physically mapping the genes that they are a part of, and as markers for traits linked to those genes. Such information may be useful in plant breeding in order to develop lines with desired phenotypes.
  • the nucleic acid molecules of the present invention may be used as restriction fragment length polymo ⁇ hism (RFLP) markers. Southern blots (Maniatis et al., Molecular Cloning: A Laboratory Manual, Second Edition (1989) Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y., herein inco ⁇ orated by reference in its entirety) of restriction-digested plant genomic DNA may be probed with the nucleic acid fragments of the present invention.
  • the resulting banding patterns may then be subjected to genetic analyses using computer programs such as MapMaker (Lander et al., Genomics 7:174-181 (1987), herein inco ⁇ orated by reference in its entirety) in order to construct a genetic map.
  • the nucleic acid fragments of the present invention may be used to probe Southern blots containing restriction endonuclease-treated genomic DNAs of a set of individuals representing parent and progeny of a defined genetic cross. Segregation of the DNA polymo ⁇ hisms is noted and used to calculate the position of the nucleotide sequence of the present invention in the genetic map previously obtained using this population (Botstein et al, Am. J. Hum. Genet. 32:314-331 (1980), herein inco ⁇ orated by reference in its entirety).
  • Nucleic acid probes derived from the nucleic acid molecules of the present invention may also be used for physical mapping (i.e., placement of sequences on physical maps; see Hoheisel et al, In: Nonmammalian Genomic Analysis: A Practical Guide, Academic press 1996, pp. 319- 346, and references cited therein).
  • nucleic acid probes derived from the nucleic acid molecules of the present invention may be used in direct fluorescence in situ hybridization (FISH) mapping (Trask, Trends Genet. 7:149-154 (1991), herein incorporated by reference in its entirety).
  • FISH direct fluorescence in situ hybridization
  • current methods of FISH mapping favor use of large clones (several to several hundred KB; see Laan et al, Genome Res. 5:13-20 (1995), herein inco ⁇ orated by reference in its entirety)
  • improvements in sensitivity may allow performance of FISH mapping using shorter probes.
  • nucleic acid amplification-based methods of genetic and physical mapping may be carried out using the nucleotide molecules of the present invention.
  • examples include allele-specific amplification (Kazazian et al., J. Lab. Clin. Med. 77:95-96 (1989), herein inco ⁇ orated by reference in its entirety), polymo ⁇ hism of PCR-amplified fragments (CAPS; Sheffield et al, Genomics 76:325-332 (1993), herein inco ⁇ orated by reference in its entirety), allele-specific ligation (Landegren et al, Science 241:1011-1080 (1988) herein inco ⁇ orated by reference in its entirety), nucleotide extension reactions (Sokolov et al., Nucleic Acid Res.
  • Isolated nucleic acid molecules of the present invention may find use in the identification of loss of function mutant phenotypes of a plant, due to a mutation in one or more endogenous genes encoding polypeptides of the present invention. This can be accomplished either by using targeted gene disruption protocols or by identifying specific mutants for these genes contained in a population of plants carrying mutations in all possible genes (Ballinger and Benzer, Proc. Natl. Acad Sci USA 5(5:9402-9406 (1989); Koes et al, Proc. Natl. Acad. Sci. USA 92: 149-8153 (1995); Bensen et al, Plant Cell 7/75-84 (1995) all of which are incorporated herein by reference in their entirety).
  • short segments of the nucleic acid molecules of the present invention may be used in polymerase chain reaction protocols in conjunction with a mutation tag sequence primer on DNAs prepared from a population of plants in which mutator transposons or some other mutation-causing DNA element has been introduced. The amplification of a specific DNA fragment with these primers indicates the insertion of the mutation tag element in or near the plant gene encoding polypeptide(s) of the present invention.
  • the nucleic acid molecules of the present invention may be used as a hybridization probe against PCR amplification products generated from the mutation population using the mutation tag sequence primer in conjunction with an arbitrary genomic site primer, such as that for a restriction enzyme site-anchored synthetic adapter.
  • Isolated nucleic acid molecules of the present invention can be mutated by a variety of methods well known in the art (Shortle D. et al., Annu. Rev. Genet. 15: 265, 1981; Itakura K. et al., Ann Rev. Biochem 53: 323, 1984; Botstein D. & Shortle D., Science 229: 1193, 1985; Smith M., Ann. Rev. Genet. 19: 423 1985; and Sambrook, et al, In: Molecular Cloning, A Laboratory Manual, 2nd Edition, Cold Spring Harbor Press, Cold Spring Harbor, New York 1989, herein incorporated by reference in its entirety).
  • mutations can also be introduced by causing point mutation or site directed mutagenesis by using commercially available kits such as QuickChangeTM from Stratagene (11011 North Torrey Pines Road, La Jolla, CA 92037).
  • site directed mutations can include but are not limited to, truncations, substitutions, additions, terminations, fusions of polypeptides or nucleic acid.
  • a plant containing a mutation in the endogenous gene encoding polypeptides of the present invention can be identified and obtained.
  • Such plant can also be obtained by in situ site directed mutagenesis. This mutant plant can then be used to determine or confirm the natural function of the polypeptides of the present invention disclosed herein.
  • C-terminal region refers to the region of a peptide, polypeptide, or protein chain from the middle thereof to the end that carries the amino acid having a free carboxyl group.
  • N-terminal region refers to the region of a peptide, polypeptide, or protein chain from the amino acid having a free amino group to the middle of the chain.
  • Antisense Oligonucleotides or Polynucleotides of the Present Invention provides an antisense oligonucleotide or polynucleotide encoding an RNA molecule which hybridizes to at least a portion of an RNA transcript of an endogenous gene encoding a polypeptide of the present invention, wherein the RNA molecule hybridizes with the RNA transcript such that expression of the endogenous gene is altered.
  • the present invention in another aspect, provides DNA construct wherein a promoter that functions in plant is operably linked to an antisense oligonucleotide or polynucleotide encoding an RNA molecule which hybridizes under stringent hybridization conditions to at least a portion of an RNA transcript of an endogenous gene encoding a polypeptide the amino acid sequence of which has at least 50% sequence identity to a member selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10, SEQ ID NO: 12, SEQ ID NO: 14, SEQ ID NO: 16, SEQ ID NO: 18, SEQ ID NO: 20, SEQ ID NO: 22, SEQ ID NO: 24, SEQ ID NO: 26, SEQ ID NO: 28, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 34, SEQ ID NO: 36, SEQ ID NO: 38, SEQ ID NO: 40, SEQ ID NO: 42, SEQ ID NO: 44, SEQ ID NO
  • the antisense oligonucleotide or polynucleotide can be full length or preferably has about six to about 100 nucleotides.
  • the antisense oligonucleotide or polynucleotide hybridizes under stringent conditions to either at least a corresponding portion of one strand of a nucleotide sequence selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9, SEQ ID NO: 11, SEQ ID NO: 13, SEQ ID NO: 15, SEQ ID NO: 17, SEQ ID NO: 19, SEQ ID NO: 21, SEQ ID NO: 23, SEQ ID NO: 25, SEQ ID NO: 27, SEQ ID NO: 29, SEQ ID NO: 31, SEQ ID NO: 33, SEQ ID NO: 35, SEQ ID NO: 37, SEQ ID NO: 39, SEQ ID NO: 41, SEQ ID NO: 43, SEQ ID NO: 45, SEQ ID NO: 47, SEQ ID NO:
  • the antisense oligonucleotide or polynucleotide hybridizes under stringent conditions to a corresponding portion of the 5' non- coding region or 3' non-translated region of the RNA transcript.
  • the antisense oligonucleotide or polynucleotide further comprises a sequence encoding a catalytic RNA or riboenzyme.
  • the antisense oligonucleotides or polynucleotides of the present invention may find particular use in antisense technology to suppress endogenous gene expression to control sizes of organs in transgenic plants.
  • a nucleic acid molecule derived from a nucleotide sequence selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9, SEQ ID NO: 11, SEQ ID NO: 13, SEQ ID NO: 15, SEQ ID NO: 17, SEQ ID NO: 19, SEQ ID NO: 21, SEQ ID NO: 23, SEQ ID NO: 25, SEQ ID NO: 27, SEQ ID NO: 29, SEQ ID NO: 31, SEQ ID NO: 33, SEQ ID NO: 35, SEQ ID NO: 37, SEQ ID NO: 39, SEQ ID NO: 41, SEQ ID NO: 43, SEQ ID NO: 45, SEQ ID NO: 47, SEQ ID NO: 49, SEQ ID NO: 51, SEQ ID NO: 53, SEQ ID NO: 66, SEQ ID NO: 68, and SEQ ID NO: 70 is cloned and operably linked to a promoter such that the antisense strand of RNA will be
  • antisense RNA inhibits gene expression by preventing the accumulation of mRNA that encodes the enzyme of interest (see, e.g., Sheehy et al, Proc. Nat. Acad. Sci. USA 55:8805-8809 (1988), and U.S. Patent No. 4,801,340; both of which are herein incorporated by reference in their entireties).
  • the antisense oligonucleotide or polynucleotide to be introduced generally will be substantially identical to at least a portion of the endogenous gene or genes of the present invention to be repressed.
  • the sequence however, needs not to be perfectly identical to inhibit or suppress expression of the endogenous gene or genes of the present invention.
  • the introduced sequence also needs not be full length relative to either the primary transcription product or fully processed mRNA. Generally, higher homology can be used to compensate for the use of a shorter sequence. Furthermore, the introduced sequence need not have the same intron or exon pattern, and homology of non-coding segments may he equally effective. Normally, a sequence of between about 6 nucleotides and about full length nucleotides should be used, though a sequence of at least about 500 to about 1700 nucleotides is preferred, a sequence of at least about 200 nucleotides is more preferred, and a sequence of about 6 to about 100 nucleotides is especially preferred.
  • the antisense oligonucleotide or polynucleotide is substantially complementary to at least a corresponding portion of one strand of a nucleotide sequence selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9, SEQ ID NO: 11, SEQ ID NO: 13, SEQ ID NO: 15, SEQ ID NO: 17, SEQ ID NO: 19, SEQ ID NO: 21, SEQ ID NO: 23, SEQ ID NO: 25, SEQ ID NO: 27, SEQ ID NO: 29, SEQ ID NO: 31, SEQ ID NO: 33, SEQ ID NO: 35, SEQ ID NO: 37, SEQ ID NO: 39, SEQ ID NO: 41, SEQ ID NO: 43, SEQ ID NO: 45, SEQ ID NO: 47, SEQ ID NO: 49, SEQ ID NO: 51, SEQ ID NO: 53, SEQ ID NO: 66, SEQ ID NO: 68, SEQ ID NO: 70, or
  • the antisense oligonucleotide or polynucleotide is substantially complementary to a corresponding portion of the 5' non-coding portion or 3' non-coding portion of one strand of a DNA molecule which has substantial sequence identity to a sequence selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9, SEQ ID NO: 11, SEQ ID NO: 13, SEQ ID NO: 15, SEQ ID NO: 17, SEQ ID NO: 19, SEQ ID NO: 21, SEQ ID NO: 23, SEQ ID NO: 25, SEQ ID NO: 27, SEQ ID NO: 29, SEQ ID NO: 31, SEQ ID NO: 33, SEQ ID NO: 35, SEQ ID NO: 37, SEQ ID NO: 39, SEQ ID NO: 41, SEQ ID NO: 43, SEQ ID NO: 45, SEQ ID NO: 47, SEQ ID NO: 49, SEQ ID NO: 51, SEQ ID NO: 53, SEQ ID NO: 66,
  • the antisense oligonucleotides or polynucleotides of the present invention may further comprise a nucleotide sequence encoding a catalytic RNA molecule or a ribozyme. It is known to a skilled person in the art that catalytic RNA molecules or ribozymes can also be used to inhibit expression of genes of the present invention. It is possible to design ribozymes that specifically pair with virtually any target RNA and cleave the phosphodiester backbone at a specific location, thereby functionally inactivating the target RNA. In carrying out this cleavage, the ribozyme is not itself altered, and is thus capable of recycling and cleaving other molecules, making it a true enzyme. The inclusion of ribozyme sequences within antisense RNAs confers RNA-cleaving activity upon them, thereby increasing the activity of the recombinant DNA constructs.
  • RNAs A number of classes of ribozymes have been identified.
  • One class of ribozymes is derived from a number of small circular RNAs, which are capable of self-cleavage and replication in plants.
  • the RNAs replicate either alone (viroid RNAs) or with a helper virus (satellite RNAs). Examples include RNAs from avocado sunblotch viroid and the satellite RNAs from tobacco ringspot virus, lucerne transient streak virus, velvet tobacco mottle virus, Solanum nodiflorum mottle virus and subterranean clover mottle virus.
  • the design and use of target RNA- specific ribozymes is described in Haseloff et al. Nature 334:585-591 (1988), herein incorporated by reference in its entirety.
  • the present invention also provides antibodies that specifically bind to the polypeptides of the present invention and recombinant DNA constructs that comprise nucleic acid molecules of the present invention.
  • transgenic plant means a plant that contains an exogenous nucleic acid, which can be derived from the same plant species or from a different species.
  • exogenous it is meant that a nucleic acid molecule originates from outside the plant to which the nucleic acid molecule is introduced.
  • An exogenous nucleic acid molecule can have a naturally occurring or non-naturally occurring nucleotide sequence.
  • an exogenous nucleic acid molecule can be a heterologous nucleic acid molecule derived from a different plant species than the plant into which the nucleic acid molecule is introduced or can be a nucleic acid molecule derived from the same plant species as the plant into which it is introduced.
  • the term "genome” as it applies to plant cells encompasses not only chromosomal DNA found within the nucleus, but organelle DNA found within subcellular components of the cell. DNAs of the present invention introduced into plant cells can therefore be either chromosomally integrated or organelle-localized.
  • the term "genome” as it applies to bacteria encompasses both the chromosome and plasmids within a bacterial host cell. Encoding DNAs of the present invention introduced into bacterial host cells can therefore be either chromosomally integrated or plasmid-localized.
  • Exogenous nucleic acid molecules may be transferred into a plant cell by the use of a recombinant DNA construct (or vector) designed for such a pu ⁇ ose.
  • the present invention also provides a plant recombinant DNA construct (or vector) for producing transgenic plants, wherein the plant recombinant DNA construct (or vector) comprises a sense oligonucleotide or polynucleotide or an antisense oligonucleotide or polynucleotide.
  • Methods that are well known to those skilled in the art may be used to prepare the plant recombinant DNA construct (or vector) of the present invention. These methods include in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination.
  • a plant recombinant DNA construct (or vector) of the present invention contains a sense oligonucleotide or polynucleotide or an antisense oligonucleotide or polynucleotide and operably linked regulatory sequences or control elements.
  • exemplary regulatory sequences include but are not limited to promoters, translation leader sequences, introns, 3' non-translated sequences.
  • the promoters can be constitutive, inducible, or preferably tissue-specific promoters.
  • a plant recombinant DNA construct (vector) of the present invention will typically comprise a selectable marker that confers a selectable phenotype on plant cells.
  • Selectable markers may also be used to select for plants or plant cells that contain the exogenous nucleic acid molecules of the present invention.
  • the marker may encode biocide resistance, antibiotic resistance (e.g., kanamycin, G418 bleomycin, hygromycin, etc.), or herbicide resistance (e.g., glyphosate, etc.).
  • selectable markers include, but are not limited to, a neo gene (Potrykus et al, Mol. Gen. Genet.
  • a plant recombinant DNA construct (vector) of the present invention may also include a screenable marker. Screenable markers may be used to monitor expression. Exemplary screenable markers include a ⁇ -glucuronidase or uidA gene (GUS) which encodes an enzyme for which various chromogenic substrates are known (Jefferson, Plant Mol. Biol, Rep. 5:387-405 (1987); Jefferson et al, EMBO J.
  • GUS ⁇ -glucuronidase or uidA gene
  • selectable or screenable marker genes are also genes which encode a secretable marker whose secretion can be detected as a means of identifying or selecting for transformed cells. Examples include markers that encode a secretable antigen that can be identified by antibody interaction, or even secretable enzymes which can be detected catalytically.
  • Secretable proteins fall into a number of classes, including small, diffusible proteins detectable, e.g., by ELISA, small active enzymes detectable in extracellular solution (e.g., ⁇ -amylase, ⁇ -lactamase, phosphinothricin transferase), or proteins which are inserted or trapped in the cell wall (such as proteins which include a leader sequence such as that found in the expression unit of extension or tobacco PR-S).
  • small active enzymes detectable in extracellular solution e.g., ⁇ -amylase, ⁇ -lactamase, phosphinothricin transferase
  • proteins which are inserted or trapped in the cell wall such as proteins which include a leader sequence such as that found in the expression unit of extension or tobacco PR-S.
  • Other possible selectable and/or screenable marker genes will be apparent to those of skill in the art.
  • reporter gene In addition to a selectable marker, it may be desirous to use a reporter gene. In some instances a reporter gene may be used with or without a selectable marker. Reporter genes are genes which are typically not present in the recipient organism or tissue and typically encode for proteins resulting in some phenotypic change or enzymatic property. Examples of such genes are provided in K. Wising et al. Ann. Rev. Genetics, 22, 421 (1988), which is inco ⁇ orated herein by reference. Preferred reporter genes include the beta-glucuronidase (GUS) of the uidA locus of E. coli, the chloramphenicol acetyl transferase gene from Tn9 of E.
  • GUS beta-glucuronidase
  • an assay for detecting reporter gene expression may then be performed at a suitable time after said gene has been introduced into recipient cells.
  • a preferred such assay entails the use of the gene encoding beta-glucuronidase (GUS) of the uidA locus of E. coli as described by Jefferson et al., (Biochem. Soc. Trans. 15, 17-19 (1987)) to identify transformed cells.
  • GUS beta-glucuronidase
  • the various components of the construct or fragments thereof will normally be inserted into a convenient cloning vector, e.g., a plasmid that is capable of replication in a bacterial host, e.g., E. coli.
  • a convenient cloning vector e.g., a plasmid that is capable of replication in a bacterial host, e.g., E. coli.
  • the cloning vector with the desired insert may be isolated and subjected to further manipulation, such as restriction digestion, insertion of new fragments or nucleotides, ligation, deletion, mutation, resection, etc. so as to tailor the components of the desired sequence.
  • a plant recombinant DNA construct (vector) of the present invention may also include a chloroplast transit peptide, in order to target the polypeptide of the present invention to the plastid.
  • plastid means the class of plant cell organelles that includes amyloplasts, chloroplasts, chromoplasts, elaioplasts, eoplasts, etioplasts, leucoplasts, and proplastids. These organelles are self-replicating, and contain what is commonly referred to as the "chloroplast genome," a circular DNA molecule that ranges in size from about 120 to about 217 kb, depending upon the plant species, and which usually contains an inverted repeat region.
  • chloroplast transit peptide examples include the small subunit of ribulose- 1 ,5-biphosphate carboxylase (ssRUBISCO, SSU), 5-enolpyruvateshikimate-3-phosphate synthase (EPSPS), ferredoxin, ferredoxin oxidoreductase, the light-harvesting-complex protein I and protein II, and thioredoxin F.
  • chloroplast polypeptides include the small subunit of ribulose- 1 ,5-biphosphate carboxylase (ssRUBISCO, SSU), 5-enolpyruvateshikimate-3-phosphate synthase (EPSPS), ferredoxin, ferredoxin oxidoreductase, the light-harvesting-complex protein I and protein II, and thioredoxin F.
  • non-plastid polypeptides may be targeted to the chloroplast by use of polypeptide fusions with a CTP and that a CTP sequence is sufficient to target a polypeptide to the plastid.
  • a CTP sequence is sufficient to target a polypeptide to the plastid.
  • Those skilled in the art will also recognize that various other recombinant DNA constructs can be made that utilize the functionality of a particular plastid transit peptide to import the enzyme into the plant cell plastid depending on the promoter tissue specificity.
  • the present invention also provides a transgenic plant comprising in its genome an exogenous nucleic acid molecule which comprises: (A) a 5' non-coding sequence which functions in the cell to cause the production of an RNA molecule; which is operably linked to (B) an antisense oligonucleotide or polynucleotide or a sense oligonucleotide or polynucleotide of this invention; which is operably linked to (C) a 3' non-translated sequence that functions in said cell to cause termination of transcription.
  • Transgenic plants of the present invention preferably have inco ⁇ orated into their genome or transformed into their chloroplast or plastid genomes an exogenous nucleic acid molecule (or "transgene") that comprises a sense oligonucleotide or polynucleotide or an antisense oligonucleotide or polynucleotide.
  • Transgenic plants are also meant to comprise progeny (descendant, offspring, etc.) of any generation of such a transgenic plant.
  • a seed of any generation of all such transgenic plants wherein said seed comprises a sense oligonucleotide or polynucleotide or an antisense oligonucleotide or polynucleotide of the present invention is also an important aspect of the invention.
  • the DNA constructs of the present invention may be introduced into the genome of a desired plant host by a variety of conventional transformation techniques, which are well known to those skilled in the art.
  • Preferred methods of transformation of plant cells or tissues are the Agrobacterium mediated transformation method and the biolistics or particle-gun mediated transformation method.
  • Suitable plant transformation vectors for the purpose of Agrobacterium mediated transformation include those derived from a Ti plasmid of Agrobacterium tumefaciens, as well as those disclosed, e.g., by Herrera-Estrella et al., Nature 303:209 (1983); Bevan, Nucleic Acids Res.
  • a plasmid expression vector suitable for the introduction of an antisense oligonucleotide or polynucleotide or a sense oligonucleotide or polynucleotide in monocots using electroporation or particle-gun mediated transformation is composed of the following: a promoter that is inducible or constitutive or tissue-specific; an intron that provides a splice site to facilitate expression of the gene, such as the Hsp70 intron (U.S. Patent Number 5,859,347); and a 3' polyadenylation sequence such as the nopaline synthase 3' sequence (NOS 3'; Fraley et al., Proc. Natl. Acad. Sci. USA 80: 4803-4807(1983)).
  • This expression cassette may be assembled on high copy replicons suitable for the production of large quantities of DNA.
  • US Patent No. 6,147,278 herein inco ⁇ orated by reference in its entirety, and contains a gene encoding an EPSPS enzyme conferring glyphosate resistance (denominated aroA; CP4), which is an excellent selection marker gene for many plants.
  • the gene is fused to the Arabidopsis EPSPS chloroplast transit peptide (CTP2) and expressed from the FMV promoter as described therein.
  • the cells (or protoplasts) can be cultured to regenerate into whole plants.
  • Such regeneration techniques rely on manipulation of certain phytohormones in a tissue culture growth medium, typically relying on a biocide and/or herbicide marker which has been introduced together with the desired nucleotide sequences.
  • Choice of methodology for the regeneration step is not critical, with suitable protocols being available for hosts from Leguminosae (alfalfa, soybean, clover, etc.), Umbelliferae (carrot, celery, parsnip), Cruciferae (cabbage, radish, canola/rapeseed, etc.), Cucurbitaceae (melons and cucumber), Gramineae (wheat, barley, rice, maize, etc.), Solanaceae (potato, tobacco, tomato, peppers), various floral crops, such as sunflower, and nut-bearing trees, such as almonds, cashews, walnuts, and pecans. See, for example, Ammirato et al., Handbook of Plant Cell Culture - Crop Species.
  • a transgenic plant formed using Agrobacterium transformation methods typically contains a single exogenous gene on one chromosome. Such transgenic plants can be referred to as being heterozygous for the added exogenous gene.
  • transgenic plant that is homozygous for the added exogenous gene; i.e., a transgenic plant that contains two added exogenous genes, one gene at the same locus on each chromosome of a chromosome pair.
  • a homozygous transgenic plant can be obtained by sexually mating (selfing) an independent segregant transgenic plant that contains a single exogenous gene, germinating some of the seed produced and analyzing the resulting plants produced for the exogenous gene of interest.
  • transgenic plants containing the exogenous nucleic acid molecule that encodes a polypeptide of interest are well known in the art.
  • the regenerated plants are self-pollinated to provide homozygous transgenic plants, as discussed above. Otherwise, pollen obtained from the regenerated plants is crossed to seed-grown plants of agronomically important lines. Conversely, pollen from plants of these important lines is used to pollinate regenerated plants.
  • a transgenic plant of the present invention containing desired polypeptides of the present invention are cultivated using methods well known to one skilled in the art.
  • Plants that can be made to have larger organ size by practice of the present invention include, but are not limited to, Ac ⁇ ci ⁇ , alfalfa, aneth, apple, apricot, artichoke, arugula, asparagus, avocado, banana, barley, beans, beet, blackberry, blueberry, broccoli, brussels sprouts, cabbage, canola, cantaloupe, carrot, cassava, cauliflower, celery, cherry, cilantro, citrus, Clementines, coffee, corn, cotton, cucumber, Douglas fir, eggplant, endive, escarole, eucalyptus, fennel, figs, gourd, grape, grapefruit, honey dew, jicama, kiwifruit, lettuce, leeks, lemon, lime, loblolly pine, mango, melon, mushroom, nut, oat, okra, onion, orange, an ornamental plant, papaya, parsley, pea, peach, peanut, pear, pepper, persimmon, pine
  • the present invention also provides cells of the transgenic plants of the present invention which could be used for regenerating a plant or any organ of plant with present invention.
  • the present invention also further provides a method for altering a specific organ or organs in a plant to generate smaller plant or plant organ, the method comprising the steps of: a) introducing into the genome of the plant an exogenous nucleic acid molecule comprising in the 5' to 3' direction i) a promoter that functions in the cells of said plant, said promoter operably linked to; ii) an antisense oligonucleotide or polynucleotide or a sense oligonucleotide or polynucleotide of the present invention, said antisense oligonucleotide or polynucleotide operably linked to; iii) a 3' non-translated nucleotide sequence that functions in said cells of said plant to cause transcriptional termination; b) obtaining transformed plant cells containing the exogenous nucleic acid molecule of step (a); and c) regenerating from said transformed plant cells a transformed plant in which the expression of an endogenous gene of the
  • Arabidopsis thaliana var Columbia seeds were obtained from Lehle seeds (LEHLE SEEDS 1102 South Industrial Blvd., Suite D, Round Rock TX 78681 USA). For growing seeds into plants, 2.5 inch pots are prepared with soil covered with bridal veil or a mesh screen, making sure that the soil is not packed too tightly and the mesh is in contact with the soil surface (this ensures that the germinating seedlings will be able to get through the mesh. Seeds are sown and covered with a germination dome. Seeds are vernalized for 3-4 days. Plants are grown under conditions of 16 hours light / 8 hours dark at 20-22° C, 70% humidity.
  • MF16 5'SAG13 and MF17 3'SAG13 were designed based on the SAG 13 sequence information (AF192276) from Genbank and custom synthesized by Gibco BRL, Life Technologies, Gaithersburg, Maryland U.S. A.
  • MF16 5'SAG13 is ATA TTT AAC AAG CCA TGG CAA AGG A
  • MF17 3'SAG13 is ATA TGT GTT TGA ATT CAT AGT CTT GAA identified as SEQ ID NO: 55 and SEQ ID NO: 56 respectively in the sequence list, which annealed on SAG 13 gene to introduce Nco 1 site at 5 'end and Eco RI site at 3 'end of the gene.
  • PCR was then performed to amplify the S.4G-73 cDNA using the above prepared cDNA as the template, and MF16 5'SAG13 and MF17 3'SAG13 as the primers.
  • the thermal cycling conditions were as follows: 94°C, 40 second, followed 30 cycles of 94°C, 25 seconds; 55°C, 30 seconds and 68°C, 2 minutes 30 seconds ( All reagents and equipment for PCR can be procured from Applied Biosystems, 850 Lincoln Center Drive, Foster City, CA 94404, USA).
  • the amplified cDNA was purified by gel-electrophoresis to obtain the gene identified as SEQ ID NO: 1.
  • RNA is isolated from appropriate crop and other desired plant species by pooling tissues of different developmental stages of all vegetative and reproductive organs. Sequences can be cloned out from total RNA by methodology shown in above paragraph.
  • DNA is isolated from the desired microorganism. Isolation of DNA from microorganism is well known in the art (Sambrook, et al, In: Molecular Cloning, A Laboratory Manual, 2nd Edition, Cold Spring Harbor Press, Cold Spring Harbor, New York 1989, herein inco ⁇ orated by reference in its entirety). This DNA along with oligonucleotide PCR primers can be used in a polymerase chain reaction by any one skilled in the art to isolate genes of the invention.
  • mutations can also be introduced by causing a point mutation or site directed mutagenesis in nucleotide SEQ ID NO: 3 by using commercially available kits such as QuickChangeTM Site Directed Mutagenesis Kit from Stratagene (11011 North Torrey Pines Road, La Jolla, CA 92037) to result in a peptide with about 90% of the N-terminal of SEQ ID NO: 4 by altering a nucleotide sequence to encode a stop codon at amino acid position 241 + 3, or to result in a peptide with about 80% of the N- terminal of SEQ ID NO: 4 by altering a nucleotide sequence to encode a stop codon at amino acid position 214 + 3, or to result in about 70% of the N-terminal of SEQ ID NO: 4 by altering a nucleotide sequence to encode a stop codon at amino acid position 188 ⁇ 3, or to result in about 60%) of the N-terminal of SEQ ID NO: 4 by altering a nucleotide
  • mutations can also be introduced in situ by causing point mutation or site directed mutagenesis in nucleotide SEQ ID NO: 3 by using in situ mutagenesis technology provided by Valigen Inc. (Newtown PA. USA) (US Patent Number 6,211,351; US Patent Number 6,271,360, WO 01/24615 Al, and WO 01/25460 A2, herein incorporated by reference in its entirety) to result in a peptide with about 90% of the N-terminal of SEQ ID NO: 4 by altering a nucleotide sequence to encode a stop codon at amino acid position 241 ⁇ 3, or to result in a peptide with about 80% of the N-terminal of SEQ ID NO: 4 by altering a nucleotide sequence to encode a stop codon at amino acid position 214 + 3, or to result in about 70% of the N-terminal of SEQ ID NO: 4 by altering a nucleotide sequence to encode a stop codon at amino acid position 188 + 3, or to result in about 60% of
  • DNA segments that provide replication function and antibiotic selection in bacterial cells for example, an E. coli origin of replication such as or/322, a broad host range origin of replication such as orN or oriRi, and a coding region for a selectable marker such as Spc/Str that encodes for Tn7 aminoglycoside adenyltransferase (aadA) conferring resistance to spectinomycin or streptomycin, or a gentamicin (Gm, Gent) selectable marker.
  • the host bacterial strain is Agrobacterium tumefaciens ABI or LBA4404.
  • the genetic elements of the DNA constructs are assembled to have in operable linkage a promoter that functions in plants. Additionally an antibiotic or herbicide marker cassette, an epitope tag (for example Flag® peptide catalog number F-3290, SIGMA, P.O. Box 14508 St. Louis, MO 63178 USA) at the 3' termination region of gene of interest is included in the DNA construct.
  • the multiple cloning site in this DNA construct encodes Bglll, Ncol, EcoRI, Sail, and Xhol.
  • the epitope tag region was encoded with Sail and Xhol restriction sites for optional removal of the epitope tag.
  • the Ncol site encodes a Kozak sequence for efficient translation of the protein products.
  • DNA constructs used in the method of the current invention comprise any promoter known to function to cause the transcription in plant cells and any antibiotic tolerance encoding polynucleotide sequence known to confer antibiotic tolerance to plant cells.
  • the antibiotic tolerance polynucleotide sequences include, but are not limited to polynucleotide sequences encoding for proteins involved in antibiotic tolerance to kanamycin, neomycin, hygromycin, and other antibiotics known in the art.
  • Antibiotic tolerance gene in such a vector can be replaced by herbicide tolerance encoding for 5-enolpyruvylshikimate-3 -phosphate synthase (EPSPS, described in U.S.
  • EPSPS 5-enolpyruvylshikimate-3 -phosphate synthase
  • Patent Number 4,810,648 phytoene desaturase (crtl (Misawa et al, (1993) Plant Journal 4:833-840, and (1994) Plant Jour 6:481-489) for tolerance to norflurazon, acetohydroxy acid synthase (AHAS, Sathasiivan et al. (1990) Nucl. Acids Res. 18:2188-2193) and the bar gene for tolerance to glufosinate (DeBlock, et al. (1987) E R ⁇ J. 6:2513-2519.
  • Herbicides for which transgenic plant tolerance has been demonstrated and the method of the present invention can be applied include, but are not limited to: glyphosate, glufosinate, sulfonylureas, imidazolinones, bromoxynil, delapon, cyclohezanedione, protopo ⁇ hyrionogen oxidase inhibitors, and isoxaslutole herbicides.
  • P- ⁇ 35S promoter U.S. Patent No.5,539,142, 5,196,525, 5,322,938 and 5,164,316 herein incorporated by reference in its entirety.
  • P-E35S promoter can be replaced by P-CaMV.35S promoter (U.S. Patent No. 5,858,742, herein inco ⁇ orated by reference in its entirety), or by enhanced P-CaMV.35S from Cauliflower mosaic virus containing a duplication of the -90-300 region as described in U. S. Patent No.
  • Figwort mosaic virus promoter P-FMV, as described in U. S. Patent No. 5,378,619, herein inco ⁇ orated by reference in its entirety; or the P-AtEFla (P-AtEFl or EFla) a promoter region from the Arabidopsis thaliana elongation factor gene la; the GboxlO and Gboxl lmotif (Fumiharu et al., (1999) Plant J. 18:443-448); or the DC3 promoter region from carrot (Seffens et al., Develop. Genet. 11:65-76); or the TP12 promoter (GenBank accession no.
  • DNA molecules encoding plastid transit peptides for example, the Arabidopsis EPSPS chloroplast transit peptide, At.CTP2 as described in U. S. Patent No. 5,633,435, herein inco ⁇ orated by reference in its entirety.
  • the method of the present invention enables one of skill in the art of plant molecular biology to design and assemble plant expression cassettes that contain promoters of known and unknown function.
  • the genetic elements of the DNA construct further comprise 5' leader polynucleotides for example, the Hsp70 non-translated leader sequence from Petunia hybrida as described in U. S. Patent No. 5,362,865, herein inco ⁇ orated by reference in its entirety.
  • the genetic elements further comprise herbicide tolerance genes that include, but are not limited to, for example, the aroA:CP4 coding region for EPSPS glyphosate resistant enzyme isolated from Agrobacterium tumefaciens (AGRTU) strain CP4 as described in U. S. Patent No. 5,633,435, herein incorporated by reference in its entirety.
  • the genetic elements of the DNA construct further comprise 3' termination regions that include, but are not limited to, the E9 3' termination region of the pea RbcS gene that functions as a polyadenylation signal; the nos is the 3' end of the nopaline synthase gene that functions as a polyadenylation signal.
  • the genetic elements of the DNA construct further comprise the right border (RB) and left borders (LB) of the Ti plasmid of Agrobacterium tumefaciens octopine and nopaline strains.
  • construct pMON575221 was transformed in Arabidopsis plants by Agrobacterium mediated transformation procedure.
  • Amplified and purified product SEQ ID NO: 1 can also be cloned in antisense orientation in appropriate cloning sites of vector pMON23435 to express the opposite strand of SEQ ID NO:l by one skilled in the art.
  • EXAMPLE 4 Cloning of DNA molecules of the present invention: This example illustrates how all other DNA molecules as shown in table from example 9 are isolated from different plant species by designing appropriate PCR primers based on the DNA sequence information provided in the table of Example 8. For isolating these DNA molecules one skilled in the art will isolate total RNA from a crop or other desired plant species by pooling tissues of different developmental stages of all vegetative and reproductive organs. DNA molecules are cloned out from total RNA by methodology shown in Example 2.
  • Isolated DNA molecule sequences can be cloned at appropriate cloning sites in sense or antisense orientation of a plant expression vector shown in Example 3 or a similar vector capable of ectopically expressing the gene of interest of the present invention in sense or antisense orientation as known in the art.
  • This example illustrates how Agrobacterium cells are transformed and how transformed cells are cultured. Transformation ; 1. Electroporate 2 ⁇ l of DNA construct into 20 ⁇ l of ABI competent cells;
  • ABI Cell Culture 1. Pick 3 colonies per ABI plate and grow each in 4ml LB media containing Spectinomycin (75 ⁇ g/ml), Kanamycin (50 ⁇ g/ml), Chloramphenicol (25 ⁇ g/ml);
  • Glycerol Stocks. & DNA Preps 1. Make three 1 ml ABI glycerol stocks per 4 ml culture, using 500 ⁇ l of culture and 500 ⁇ l of 40% glycerol. Freeze and store at -80°C. 2. Miniprep remaining culture (about 2.5ml), using a Qiagen miniprep kit and protocol (Qiagen Genomics, Inc., Seattle, WA), ensuring add PB buffer wash step and EB buffer (lOmM Tris- Cl, pH 8.5) to 70°C before eluting DNA from column. The resulting volume per miniprep sample should be 50 ⁇ l.
  • Arabidopsis plants may be transformed by any one of many available methods.
  • Arabidopsis plants may be transformed using in planta transformation method by vacuum infiltration (see, Bechtold et al, In planta Agrobacterium mediated gene transfer by infiltration of adult Arabidopsis thaliana plants. CR Acad. Sci. Paris Sciences de la vie/life sciences 316: 1194-1199 (1993), herein inco ⁇ orated by reference in its entirety). Plants can be grown as described in Example 1.
  • Asrobacterium Preparation (Small scale and Large scale cultures ' ): Agrobacterium strain ABI is streaked onto an LB plate containing Spectinomycin 100 mg/L, Streptomycin 100 mg/L, Chloramphenicol 25mg/L, and Kanamycin 50mg/L (denoted SSCK). Two days prior to infiltration, a loop of Agrobacterium is placed into a tube containing 10 mis LB/SSCK and put on a shaker in the dark at 28°C to grow overnight. The following day, the Agrobacterium is diluted 1:50 in 400 mis of bacterial grown medium such as SSCK and put on a shaker at 28°C to grow for 16-20 hours. Infiltration
  • a very small amount of seed can be first plated out to check for contamination. If there is contamination, re-sterilized seeds for ⁇ 4 more hours and check for contamination again.
  • the second sterilization is usually not necessary, but sometimes the seed harbors a fungal contaminant and repeat sterilizations are needed. (The sterilization duration generally is shorter than 16 hours because of significantly decreased germination rates starting at 24 hr. sterilization duration).
  • Example 7 Following subsections of Example 7 are used for describing phenotypic changes after transformation and growth of Arabidopsis plants as described in Example 6.
  • Three different events obtained from transformation of pMON57521 (Figure 2) into Arabidopsis thaliana ecotype Columbia were selected and grown side by side with wild type plants. Growth conditions were 16 hr light, 8 hr night, 21 degrees Centigrade and 70% relative humidity. Plants were observed at all growth stages and photographed using an Olympus camera C-2500 L (Olympus America Inc., 2 Corporate Center Drive, Melville, NY 11747) as described by the manufacturer.
  • Olympus America Inc. 2 Corporate Center Drive, Melville, NY 11747
  • Plants transformed with Construct pMON57521 were shown to exhibit 2 to 3 times larger flower and floral organs such as stamen and pistils when compared to the wild type, non transformed plants of the same species. Transgenic plants were also observed to have at least 2 times more lateral roots and 2 to 3 times thicker stem size when compared to the lateral roots and stems of wild type, non transformed plants of the same species. Approximately 2 fold increased trichome numbers and distribution were also observed. Over expression of the polypeptide encoded by the sequence in the pMON57521 construct of the present invention in the transformed plants, yielded plants that exhibited a 100 % increase in individual seed size and weight.
  • Seeds from transformed plants exhibit increased seed size that was approximately twice as large as the seeds from wild type plants as shown in following table. Image sizes were determined by the Pro or Lucis software ( Optronics, 175 Cremona Drive, Goleta, CA 93117) based on the image pixel value of wild type and transformed plants' seed under the same resolution. Plant lines 8752-1, 8752-2, 8752-6, and 8752-7 in Table 1 correspond to different transgenic plant lines produced by transforming wild Arabidopsis plants with pMON57521.
  • Seed Weight The seed from plants containing pMON57521 lines was also found to be heavier than WT seed. Seed weight was measured from 2 lines produced from event 8752 (8752-1 and 8752-6) and 2 lines from event 8748 (8748-2 and 8748-7) and compared with wild type. Shown below is a representative seed weight analysis. For each line, 3 replications were measured (i.e. 50 seeds were counted three times and measured each time). This analysis was repeated with different seed counts (eg. 100 seed or 150 seed) and was found to be highly reproducible. For example, weight per 100 seed is similarly high ⁇ 0.0025 gm. for wild type and 0.0046 gm. for seeds from plants transformed with sequence of the present invention.
  • Plant lines 8752-1, 8752-6, 8748-2, and 8748- 7 in Table 2 correspond to different transgenic plant lines produced by transforming wild Arabidopsis plants with pMON57521. Table 2: Seed Weight
  • (C) Seed Number Lines over expressing sequence of the present invention were found to have fewer seed per silique as compared to wild type plants. The average number of seed /silique of equivalent maturity was 34 for transgenic plants of the present invention as compared to 52 for wild type plants. The analysis was repeated three times with highly reproducible results within and between events.
  • Seed Yield There was an increase of > 20% seed yield in transgenic plants of the present invention. Seed yield when measured in terms of weight of total seeds per plant.
  • silique number at growth stage 6.5 was estimated for both WT and transgenics and total silique number is shown in the following table. This indicates again that silique number was at least 28% greater in lines over-expressing the polypeptide molecules corresponding SEQ ID NO: 2 (transformed lines) as compared to wild type lines at the same growth stage. As the plant proceeds to senescence, the relative increase appears to stay consistent and together with the increase in seed weight in the transgenic plant, contributes to the overall increase in seed yield per plant. Growth stage estimation was based on "Growth stage- based phenotypic analysis of Arabidopsis: A model for high throughput functional genomics in plants. Plant Cell. 13(7):1499, 2001"
  • WT wild type plant and Transformed line is transgenic plant line expressing polypeptide molecules corresponding to SEQ ID. NO: 2.
  • This example describes increase of seeds in transgenic soybean plants expressing polypeptide molecule corresponding to SEQ ID NO: 2. Soybean plants were transformed with pMON73955 ( Figure 7) to constitutively express
  • Soybean transformation is performed essentially as described in WO 00/42207, herein inco ⁇ orated by reference in its entirety. RI seed from 10 events out of 44 events were advanced in Puerto Rico (PR) based on the gene copy number in plants. Soybean transformation was performed essentially as described in WO 00/42207, herein inco ⁇ orated by reference in its entirety. Preliminary data indicates that 1 of the 10 events in PR showed a phenotype similar to that seen for the Arabidopsis over-expression of SEQ ID NO: 2 (pMON 57521 Figure 2) i.e. more pods and more branches as well as short, fat pods with fewer seed.
  • Table 7 R2 seed weights of individual lines from the large-seeded transgenic event "A expressing SEQ ID NO: 2.
  • This example describes over expression of the protein in bacterial cells for purification of the protein so as to screen for the activity of polypeptide molecules.
  • A Cloning of the nucleotide molecules of the present invention for expression of corresponding peptides.
  • pMON57521 Figure 2
  • pMON73963 Figure 3
  • Nostoc punctiforme Nostoc genomic DNA is used as a DNA template source for polymerase chain reaction amplification (PCR) of DNA for cloning into bacterial cells so as to express the Arabidopsis thaliana protein corresponding to SEQ ID NO: 2, SEQ ID NO: 4, or Nostoc protein corresponding to SEQ ID NO: 50.
  • PCR polymerase chain reaction amplification
  • thermocycler needed for performing PCR reaction was procured from Applied Biosystems (Perkin-Elmer Corp., Applied Biosystems Div., Foster City, CA).
  • the polynucleotide sequence of the amplicon produced from Arabidopsis is shown in SEQ ID NO:l and SEQ ID NO: 3.
  • PCR primer pairs SEQ ID NO: 57 and SEQ ID NO: 58 were used for performing the reaction to obtain the Arabidopsis amplicon.
  • Arabidopsis amplicons were subcloned into a pET-28b vector (E coli expression vector, Novagen, Madison, WI, USA) and were sequenced by using sequencing primers SEQ ID NO: 59 and SEQ ID NO: 60 to confirm the sequence of the cloned polynucleotide molecule.
  • Nostoc amplicons were generated by using Nostoc genomic DNA as template with primers pairs SEQ ID NO: 61 and SEQ ID NO: 63 or SEQ ID NO: 62 and SEQ ID NO: 63.
  • Nostoc amplicons were also subcloned in to the pET-28b vector and were sequenced by using sequencing primers SEQ ID NO: 64 and SEQ ID NO: 65 to confirm the polynucleotide sequence of the cloned polynucleotide molecule.
  • Primer Sequence for amplification and sequencing of molecules of invention from Arabidopsis and Nostoc are shown in Figure 10. These constructs contained a candidate from Arabidopsis SEQ ID NO: 3 and the closest Nostoc homologue SEQ ID NO: 49, both with and without an N-terminal His-tag. An N-terminal His-tag was chosen based on the crystal structures (Nakajima et al.
  • Protein extracts were prepared by freeze thawing and sonication as described Cull and McHenry (Cull M and McHenry CS. Methods in Enzymology 182, 147-153, 1990). Pellets were resuspended in an appropriate volume of either 50 mM Tris-HCI pH 7.4 250 mM NaCI and 500 ⁇ M EDTA with 0.05% CHAPS and ImM ABESF or 50 mM KH 2 PO 4 pH-7.2 250 mM NaCI and 250 ⁇ M EDTA with 0.05% CHAPS and ImM ABESF, buffer.
  • Protein concentrations were 1.5 to 1.7 mg /mL,based on the method of Bradford (Bradford M, Anal Biochem, 72, 248, 1976 and Bio-Rad Laboratories Procedure bulletin 1123) with BSA standard (Bradford reagent and BSA was from Bio-Rad Laboratories Headquarters 1000 Alfred Nobel Drive Hercules, CA 94547).
  • the assays described below used 10 or 15 uL of this solution
  • Spectrophotometric assays were used for determination of enzyme activities of purified proteins. Assays were based on earlier described methods (Portsteffen et al. Phyochemistry, 37, 391, 1994). Consumption or production of NADPH was observed at 340 nm. Assays were performed in a Varian Gary 50 Bio spectrophotometer (Varian Instruments inc. AA, ICP, UV, Fluorescence Products 679 Springvale RoadMulgrave, Victoria 3170Australia).
  • NADPH solutions were freshly made and checked by UV-vis for degradation.
  • Enzyme solutions were supplemented with ImM MgCl 2 and ImM Zn Cl 2 which were at 1.5 to 1.7 mg /mL concentrations.
  • the assays used 10 or 15 ⁇ L of this solution (ca. 15 to 22 ⁇ g per reaction).
  • Stock solutions of the substrates (20 to 500mM) were made up in a reaction buffer or as noted.
  • Steroids solutions were made in 100% EtOH. From these stock solutions additions to the assays were made directly. Precipitation of steroids was observed.
  • the soluble concentration of steroids in reaction mixture is not accurately known in each case.
  • good estimate of substrate (sterol) concentration would be 100 to 200 ⁇ M for a saturated solution under these conditions.
  • Table 8 Zero Order Result Table using acetone as substrate (60mM) shows evidence of reductase activity. Turnover (consumption of NADPH) is observed in the reductive direction. Slopes (for acetone, lines 1 and 2) indicate the difference from control is 10 fold. Controls (lines 3 and 4) were identical except for the omission of acetone.
  • Start start time
  • Stop stop time
  • Slope change in absorbance over time
  • a start is starting absorbance
  • a stop is ending absorbance.
  • Table 9 Zero Order Result Table. This table shows elected data for 5 ⁇ -androstane-3,17- dione in a reductase assay. This example shows the results for three reactions with acetone, three reactions with 5 ⁇ -androstane-3,17-dione and two controls. Table 8 shows the initial rates as estimated by linear fit between 0 and 5 minutes. The results for 5 ⁇ -androstane-3,17-dione shown here are the best of any substrate at the same concentration (approx 200 ⁇ M). 4- androstene-3,17-dione is the next best substrate with a rate on the order of 2.5 to 4 times slower than this under the same conditions (not shown). Acetone results at similar concentrations (Table 7 shows results at high acetone concentration) are on the order of 10 to 20 times slower and nearly at background levels. Similar results have been obtained with multiple batches of enzyme and substrate. Sample Start Stop Slope A A S.D.
  • Start start time
  • Stop stop time
  • Slope change in absorbance ovei • time
  • a start is starting absorbance
  • a stop is ending absorbance
  • RNA is purified from tissue using Trizol reagent from Life Technologies (Gibco BRL, Life Technologies, Gaithersburg, Maryland U.S.A.), as recommended by the manufacturer.
  • Poly A+ RNA (mRNA) is purified using magnetic oligo dT beads essentially as recommended by the manufacturer (Dynabeads, Dynal Corporation, Lake Success, New York U.S.A.).
  • the cDNA libraries are plated on LB agar containing the appropriate antibiotics for selection and incubated at 37° for sufficient time to allow the growth of individual colonies.
  • the plasmid DNA is isolated from each clone using Qiaprep plasmid isolation kits, using the conditions recommended by the manufacturer (Qiagen Inc., Santa Clara, California U.S.A.).
  • the template plasmid DNA clones are used for subsequent sequencing.
  • a commercially available sequencing kit such as the ABI PRISM dRhodamine Terminator Cycle Sequencing Ready Reaction Kit with AmpliTaq® DNA Polymerase, FS, is used under the conditions recommended by the manufacturer (PE Applied Biosystems, Foster City, CA).
  • the cDNAs of the present invention are generated by sequencing initiated from the 5' end or 3' end of each cDNA clone. Entire inserts or only part of the inserts (ESTs or expressed sequenced tags) are sequenced.
  • a number of DNA sequencing techniques are known in the art, including fluorescence- based sequencing methodologies. These methods have the detection, automation and instrumentation capability necessary for the analysis of large volumes of sequence data.
  • the 377 and 3700 DNA Sequencer Perkin-Elmer Co ⁇ ., Applied Biosystems Div., Foster City, CA
  • fluorescent dye-labeled sequence reaction products are detected and data entered directly into the computer, producing a chromatogram that is subsequently viewed, stored, and analyzed using the corresponding software programs.
  • These methods are known to those of skill in the art and have been described and reviewed (Birren et al, Genome Analysis: Analyzing DNA,1, Cold Spring Harbor, New York, the entirety of which is herein inco ⁇ orated by reference).
  • the generated ESTs (including any full-length cDNA inserts or complete coding sequences) are combined with ESTs and full-length cDNA sequences in public databases such as GenBank. Duplicate sequences are removed, and duplicate sequence identification numbers are replaced. The combined dataset is then clustered and assembled using Pangea Systems tool identified as CAT v.3.2. First, the EST sequences are screened and filtered, e.g. high frequency words are masked to prevent spurious clustering; sequence common to known contaminants such as cloning bacteria are masked; high frequency repeated sequences and simple sequences are masked; unmasked sequences of less than 100 bp are eliminated.
  • the screened and filtered ESTs are combined and subjected to a word-based clustering algorithm which calculates sequence pair distances based on word frequencies and uses a single linkage method to group like sequences into clusters of more than one sequence, as appropriate.
  • Clustered sequences are assembled individually using an iterative method based on PHRAP/CRAW/MAP providing one or more self-consistent consensus sequences and inconsistent singleton sequences.
  • the assembled clustered sequence files are checked for completeness and parsed to create data representing each consensus contiguous sequence (contig), the initial EST sequences, and the relative position of each EST in a respective contig.
  • the sequence of the 5' most clone is identified from each contig.
  • the initial sequences that are not included in a contig are separated out.
  • SEQ ID NO: 1 exhibits 52.857% percent identity with its closest known functional gene, identified using the BLAST 2.2.1 software with BLOSUM62 matrix and "no Filter” options (Genbank accession number gi

Abstract

The present invention provides methods and DNA molecules useful for producing transgenic plants with agronomically desired traits based on altered sizes of plant organs and tissues.

Description

MOBT:234P 11899.0234.00PC00
1 -
METHOD OF INCREASING PLANT ORGAN AND SEED SIZE IN A PLANT
CROSS REFERENCE TO RELATED APPLICATIONS
This Application claims the benefit of U.S. Provisional Application No.60/381,100, filed 5/15/2002.
FIELD OF THE INVENTION This invention relates to plant molecular biology. In particularly, this invention relates to transgenic plants having increased or enhanced seed and organ sizes.
BACKGROUND OF THE INVENTION
Recent advances in genetic engineering have provided the prerequisite tools to transform plants to contain foreign (often referred to as "heterogeneous or heterologous ") or improved endogenous genes. The introduction of such a gene in a plant can desirably lead to an improvement of an already existing pathway in plant tissues or introduction of a novel pathway to modify desired product levels, increase metabolic efficiency, and/or save on energy cost to the cell. Plants with unique physiological and biochemical traits and characteristics, such as herbicide resistance and insect resistance, have already been produced. The ability to create traits that play an essential role in plant growth and development, crop yield potential and stability, and crop quality and composition are particularly desirable targets for the crop plant improvement.
Normally a plant goes through a development cycle, which includes seed germination, maturation of plant, reproduction, and finally senescence that leads to death of a plant. Several biological processes are common to different stages of plant development. Desired effects such as growth of tissue organ are achieved in nature by fine-tuning of the metabolism of the organism. The final phase of growth is senescence which is a highly regulated, genetically controlled and active process (Thomas H., and Stoddart J. L., Ann. Rev. Plant Physiol (31) 83- 111, 1980). Senescence is mostly studied in plant leaves and is regarded as a series of events concerned with cellular disassembly and the mobilization of released material to other plant parts such as seeds, storage organs or developing leaves and flowers (Nooden L.D. In Senescence and Aging in Plants, Academic press, 391-439, 1988). Leaf senescence can be initiated by seed development in certain species of plants. This was demonstrated in soybean by surgically removing flowers or physically restricting pod growth to observe the delay in leaf senescence (Nooden L.D. In Senescence and Aging in Plants, Academic press, 330-368,1988; Miceli F, Crafts-Brandner S.J., Egli D.B. Crop Sci. (35), 1080-1085, 1995). During senescence, partitioning of resources between vegetative and reproductive development involves a complex interplay of generative and degenerative processes, requiring differential expression of genes.
Differentially expressed genes during senescence are usually referred as "Senescence Associated Genes" or SAGs (Hensel L.L, Grbic V., Baumgarten D.A., Bleecker A.B., The Plant Cell (5) 553-564 1993). All SAG genes may not be functionally related, but they all are involved in similar physiological processes. In the past, senescence studies were directed towards understanding processes to generally enhance knowledge and applying this information relating to senescence in agriculture to enhancing yield and reducing post harvest losses (Hensel L.L, Grbic V., Baumgarten D.A., Bleecker A.B., The Plant Cell (5) 553-564 1993; Gan S., Amasino R.M., Science, (270) 1986-1988, 1995; Gan S., Amasino R.M., Plant Physiol., (113) 313-319, 1997; Guarente L., Ruvkun G., Amasino R.M., Proc. Natl Acad. Sci USA (95) 1034-1036, 1998; US Patent Number 5 68 9042; PCT/US00/03494; and PCT/USOO/18364, July 2000).
The SAG 13 gene was first described by Lohman et al. in 1994 (Lohman K.M., Gan S.,
John M.C., Amasino R.M., Physiologia Plantarum (92) 322-328, 1994) and then by Weaver et al. in 1998 (Weaver L.M. Gan S., Quirino B, Amasino R.M., Plant Mol. Biol. 455-469, 1998) as one of the genes associated with senescence. SAG genes by definition are up regulated during age-mediated senescence. SAG 13 was observed to be induced strongly shortly before visible senescence marked by yellowing of green leaves.
SUMMARY OF THE INVENTION The present invention relates to transgenic plants with increased organ size when compared to a non transformed plant of the same species. In one preferred embodiment, the present invention provides a transgenic plant with increased seed size when compared to a non transformed plant of the same species. One embodiment of the invention provides a method of increasing seed and organ size of a plant by transforming the plant with a DNA construct comprising a promoter that functions in plants, operably linked to a DNA molecule that encodes a protein, where in said DNA molecule is selected from the from the group consisting of SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9, SEQ ID NO: 11, SEQ ID NO: 13, SEQ ID NO: 15, SEQ ID NO: 17, SEQ ID NO: 19, SEQ ID NO: 21, SEQ ID NO: 23, SEQ ID NO: 25, SEQ ID NO: 27, SEQ ID NO: 29, SEQ ID NO: 31, SEQ ID NO: 33, SEQ ID NO: 35, SEQ ID NO: 37, SEQ ID NO: 39, SEQ ID NO: 41, SEQ ID NO: 43, SEQ ID NO: 45, SEQ ID NO: 47, SEQ ID NO: 49, SEQ ID NO: 51, SEQ ID NO: 53, SEQ ID NO: 66, SEQ ID NO: 68, and SEQ ID NO: 70 and wherein said promoter is heterologous to said DNA molecule. The method also includes the DNA construct operably linked to a 3' termination region and selecting a desired plant from a population of transformed plants containing said DNA construct; wherein said desired plant exhibits increased seed and organ size compared to a plant of a same plant species not transformed to contain said DNA construct. The method may also encompass various promoters including a caulimovirus promoter, or a heterologous plant constitutive promoter, or a tissue specific or an organ enhanced promoter.
In another embodiment, the present invention provides a method for increasing seed and other plant organ sizes by transforming a plant with a DNA construct comprising a promoter that functions in plants, operably linked to a DNA molecule that encodes a protein, wherein said protein comprises at least an N-terminal 50% portion of a polypeptide selected from the group consisting of : SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10, SEQ ID NO: 12, SEQ ID NO: 14, SEQ ID NO: 16, SEQ ID NO: 18, SEQ ID NO: 20, SEQ ID NO: 22, SEQ ID NO: 24, SEQ ID NO: 26, SEQ ID NO: 28, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 34, SEQ ID NO: 36, SEQ ID NO: 38, SEQ ID NO: 40, SEQ ID NO: 42, SEQ ID NO: 44, SEQ ID NO: 46, SEQ ID NO: 48, SEQ ID NO: 50, SEQ ID NO: 52, SEQ ID NO: 54, SEQ ID NO: 67, SEQ ID NO: 69, and SEQ ID NO: 71 wherein said DNA construct is operably linked to a 3 'termination region; and selecting a plant from a population of transformed plants containing said DNA construct; wherein said plant exhibits increased seed and organ size compared to a plant of a same species not transformed to contain said DNA construct. In a further embodiment, the present invention provides an isolated DNA construct comprising a promoter capable of functioning in a plant cell, operably linked to a structural nucleic acid sequence encoding SEQ ID NO: 2 or SEQ ID NO: 4, and a 3' non translated nucleic acid sequence capable of causing transcriptional termination and the addition of polyadenylated nucleotides to the 3 'end of the transcribed mRNA sequence. This nucleic acid sequence may optionally include an intron, a 5' untranslated leader sequence or another nucleic acid sequence designed to enhance transcription and/or translation.
Another aspect of the present invention provides a method for improving the seed and organ size in a plant comprising the steps of:
(a) inserting into the genome of a plant a DNA construct comprising in the 5' to 3' direction: (i) a promoter that functions in the cell of a selected plant tissue operably linked to;
(ii) a structural nucleic acid sequence that encodes a protein product selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID
NO: 10, SEQ ID NO: 12, SEQ ID NO: 14, SEQ ID NO: 16, SEQ ID NO: 18, SEQ ID NO: 20,
SEQ ID NO: 22, SEQ ID NO: 24, SEQ ID NO: 26, SEQ ID NO: 28, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 34, SEQ ID NO: 36, SEQ ID NO: 38, SEQ ID NO: 40, SEQ ID NO: 42, SEQ ID NO: 44, SEQ ID NO: 46, SEQ ID NO: 48, SEQ ID NO: 50, SEQ ID NO: 52, SEQ ID NO: 54, SEQ ID NO: 67, SEQ ID NO: 69, and SEQ ID NO: 71 operably linked to
(iii) a 3' non translated nucleic acid sequences that functions in a plant cell to cause transcriptional termination and addition of polyadenylated nucleotides to the 3' end of an RNA sequence;
(b) obtaining transformed plant cells containing the nucleic acid of step (a); and
(c) regenerating from transformed plant cells a transformed plant that over-expresses the gene product in the plant cell.
One aspect of invention provides a transgenic plant with increased seed and organ size when compared to a non transformed plant of a same plant species, said transgenic plant comprising a DNA construct, wherein said DNA construct comprises a promoter that functions in plants, operably linked to a DNA molecule that encodes a protein, wherein said DNA molecule is selected from the group consisting of: SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9, SEQ ID NO: 11, SEQ ID NO: 13, SEQ ID NO: 15, SEQ ID NO: 17, SEQ ID NO: 19, SEQ ID NO: 21, SEQ ID NO: 23, SEQ ID NO: 25, SEQ ID NO: 27, SEQ ID NO: 29, SEQ ID NO: 31, SEQ ID NO: 33, SEQ ID NO: 35, SEQ ID NO: 37, SEQ ID NO: 39, SEQ ID NO: 41, SEQ ID NO: 43, SEQ ID NO: 45, SEQ ID NO: 47, SEQ ID NO: 49, SEQ ID NO: 51, SEQ ID NO: 53, SEQ ID NO: 66, SEQ ID NO: 68, and SEQ ID NO: 70 operably linked to a 3 'termination region, wherein said promoter is heterologus to said DNA molecules. Another aspect of invention provides a transgenic plant with increased seed and organ size when compared to a non transformed plant of a same plant species, the transgenic plant comprising a DNA construct, wherein said DNA construct encodes a protein that comprises at least an N-terminal 50% portion selected from the group consisting of: SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10, SEQ ID NO: 12, SEQ ID NO: 14, SEQ ID NO: 16, SEQ ID NO: 18, SEQ ID NO: 20, SEQ ID NO: 22, SEQ ID NO: 24, SEQ ID NO: 26, SEQ ID NO: 28, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 34, SEQ ID NO: 36, SEQ ID NO: 38, SEQ ID NO: 40, SEQ ID NO: 42, SEQ ID NO: 44, SEQ ID NO: 46, SEQ ID NO: 48, SEQ ID NO: 50, SEQ ID NO: 52, SEQ ID NO: 54, SEQ ID NO: 67, SEQ ID NO: 69, and SEQ ID NO: 71. Yet another aspect of the present invention provides a DNA construct comprising a promoter operably linked to an isolated nucleic acid molecule comprising a nucleotide sequence or complement thereof, wherein the nucleotide sequence encodes a polypeptide having an amino acid sequence that has at least 50% sequence identity, preferably at least 60%, more preferably at least 70% sequence identity, even more preferably at least 80% or 90% sequence identity, and most preferably at least 95% to 98% sequence identity to a polypeptide selected from group consisting of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10, SEQ ID NO: 12, SEQ ID NO: 14, SEQ ID NO: 16, SEQ ID NO: 18, SEQ ID NO: 20, SEQ ID NO: 22, SEQ ID NO: 24, SEQ ID NO: 26, SEQ ID NO: 28, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 34, SEQ ID NO: 36, SEQ ID NO: 38, SEQ ID NO: 40, SEQ ID NO: 42, SEQ ID NO: 44, SEQ ID NO: 46, SEQ ID NO: 48, SEQ ID NO: 50, SEQ ID NO: 52, SEQ ID NO: 54, SEQ ID NO: 67, SEQ ID NO: 69, SEQ ID NO: 71 where in said promoter is heterologus to the said nucleic acid molecules. BRIEF DESCRIPTION OF THE FIGURES
Figure 1 shows a plasmid map for plant transformation vector pMON 23435 with its elements. Figure 2 shows a plasmid map for plant transformation vector pMON 57521 with its elements.
Figure 3 shows a plasmid map for plant transformation vector pMON 54955 with its elements. Figure 4 shows a plasmid map for plant transformation vector pMON 73955 with its elements.
Figure 5 shows a plasmid map for plant transformation vector pMON 73963 with its elements.
Figure 6 shows a plasmid map for bacterial transformation vector pMON 63132 with its elements.
Figure 7 shows a plasmid map for bacterial transformation vector pMON 63133 with its elements.
Figure 8 shows a plasmid map for bacterial transformation vector pMON 6134 with its elements. Figure 9 shows a plasmid map for bacterial transformation vector pMON 6135 with its elements.
Figure 10 shows primers used for amplification of gene and for sequencing of amplicons.
Figure 11 shows putative products from steroid reductase activity.
Figure 12 shows a potential steroid reductase reaction.
DETAILED DESCRIPTION OF THE INVENTION
Isolated Nucleic Acid Molecules of the Present Invention
The present invention provides a method of increasing seed and organ size of a plant by transforming the plant with a DNA construct comprising a promoter that functions in plants, operably linked to a DNA molecule that encodes a protein, where in said DNA molecule is selected from the from the group consisting of SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9, SEQ ID NO: 11, SEQ ID NO: 13, SEQ ID NO: 15, SEQ ID NO: 17, SEQ ID NO: 19, SEQ ID NO: 21, SEQ ID NO: 23, SEQ ID NO: 25, SEQ ID NO: 27, SEQ ID NO: 29, SEQ ID NO: 31, SEQ ID NO: 33, SEQ ID NO: 35, SEQ ID NO: 37, SEQ ID NO: 39, SEQ ID NO: 41, SEQ ID NO: 43, SEQ ID NO: 45, SEQ ID NO: 47, SEQ ID NO: 49, SEQ ID NO: 51, SEQ ID NO: 53, SEQ ID NO: 66, SEQ ID NO: 68, SEQ ID NO: 70 and wherein said promoter is heterologous to said DNA molecule.
In another embodiment, the present invention provides a method for increasing seed and other plant organ sizes by transforming a plant with a DNA construct comprising a promoter that functions in plants, operably linked to a DNA molecule that encodes a protein, wherein said protein comprises at least an N-terminal 50% portion of a polypeptide selected from the group consisting of : SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10, SEQ ID NO: 12, SEQ ID NO: 14, SEQ ID NO: 16, SEQ ID NO: 18, SEQ ID NO: 20, SEQ ID NO: 22, SEQ ID NO: 24, SEQ ID NO: 26, SEQ ID NO: 28, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 34, SEQ ID NO: 36, SEQ ID NO: 38, SEQ ID NO: 40, SEQ ID NO: 42, SEQ ID NO: 44, SEQ ID NO: 46, SEQ ID NO: 48, SEQ ID NO: 50, SEQ ID NO: 52, SEQ ID NO: 54, SEQ ID NO: 67, SEQ ID NO: 69, or SEQ ID NO: 71 operably linked to a 3'termination region.
The term "nucleic acid molecule" as used herein means a deoxyribonucleic acid (DNA) molecule or ribonucleic acid (RNA) molecule. Both DNA and RNA molecules are constructed from nucleotides linked end to end, wherein each of the nucleotides contains a phosphate group, a sugar moiety, and either a purine or a pyrimidine base. Nucleic acid molecules can be a single or double-stranded polymer of nucleotides read from the 5' to the 3' end. Nucleic acid molecules may also optionally contain synthetic, non-natural or altered nucleotide bases that permit correct read through by a polymerase and do not alter expression of a polypeptide encoded by that nucleic acid molecule.
The term "an isolated nucleic acid molecule" as used herein means a nucleic acid molecule that is no longer accompanied by some of materials with which it is associated in its natural state or to a nucleic acid molecule the structure of which is not identical to that of any of naturally occurring nucleic acid molecule. Examples of an isolated nucleic acid molecule include: (1) DNAs which have the sequence of part of a naturally occurring genomic DNA molecule but are not flanked by two coding sequences that flank that part of the molecule in the genome of the organism in which it naturally occurs; (2) a nucleic acid molecule incorporated into a vector or into the genomic DNA of a prokaryote or eukaryote in a manner such that the resulting molecule is not identical to any naturally occurring vector or genomic DNA; (3) a separate molecule such as a cDNA, a genomic fragment, a fragment produced by polymerase chain reaction (PCR), or a restriction fragment; (4) recombinant DNAs; and (5) synthetic DNAs. An isolated nucleic acid molecule may also be comprised of one or more segments of cDNA, genomic DNA or synthetic DNA.
It is also contemplated by the inventors that the isolated nucleic acid molecules of the present invention also include known types of modifications, for example, labels which are known in the art, methylation, "caps", substitution of one or more of the naturally occurring nucleotides with an analog. Other known modifications include internucleotide modifications, for example, those with uncharged linkages (methyl phosphonates, phosphotriesters, phosphoamidates, carbamates, etc.) and with charged linkages (phosphorothioates, phosphorodithioates, etc.), those containing pendant moieties, such as, proteins (including nucleases, toxins, antibodies, signal peptides, poly-L-lysine, etc.), those with intercalators (acridine, psoralen, etc.), those containing chelators (metals, radioactive metals, boron, oxidative metals, etc.), those containing alkylators, and those with modified linkages.
The term "nucleotide sequence" as used herein means the order contiguous of nucleic acid molecule of both the sense and antisense strands or as a duplex strand. It includes, but is not limited to, self-replicating plasmids, synthetic polynucleotides, chromosomal sequences, and infectious polymers of DNA or RNA.
A nucleotide sequence is said to be the "complement" of another nucleotide sequence if they exhibit complete complementarity. As used herein, molecules are said to exhibit "complete complementarity" when every nucleotide of one of the sequences is complementary to a nucleotide of the other.
As used herein both terms "a coding sequence" and "a structural nucleotide sequence" mean a nucleotide sequence, which is translated into a polypeptide, usually via mRNA, when placed under the control of appropriate regulatory sequences. The boundaries of the coding sequence are determined by a translation start codon at the 5 '-terminus and a translation stop codon at the 3 '-terminus. A coding sequence can include, but is not limited to, genomic DNA, cDNA, and recombinant nucleotide sequences.
The polypeptides of the invention, like other polypeptides, have different domains which perfoπn different functions. Thus, the coding sequences need not be full length, so long as the desired functional domain of the polypeptide is expressed. The distinguishing features of polypeptides of the present invention are discussed in detail in Examples. The term " recombinant DNAs" as used herein means DNAs that contains a genetically engineered modification through manipulation via mutagenesis, restriction enzymes, and the like.
The term "synthetic DNAs" as used herein means DNAs assembled from oligonucleotide building blocks that are chemically synthesized using procedures known to those skilled in the art. These building blocks are ligated and annealed to form DNA segments, which are then enzymatically assembled to construct the entire DNA. "Chemically synthesized", as related to a sequence of DNA, means that the component nucleotides were assembled in vitro. Manual chemical synthesis of DNA may be accomplished using well-established procedures, or automated chemical synthesis can be performed using one of a number of commercially available machines.
Both terms "polypeptide" and "protein", as used herein, mean a polymer composed of amino acids connected by peptide bonds. An amino acid unit in a polypeptide (or protein) is called a residue. The terms "polypeptide" and "protein" also applies to any amino acid polymers in which one or more amino acid residue is an artificial chemical analogue- of a corresponding naturally occurring amino acid, as well as to any naturally occurring amino acid polymers. The essential nature of such analogues of naturally occurring amino acids is that, when incorporated into a polypeptide, that polypeptide is specifically reactive to antibodies elicited to the same polypeptide but consisting entirely of naturally occurring amino acids. It is well known in the art that proteins or polypeptides may undergo modification, including but not limited to, disulfide bond formation, gamma-carboxylation of glutamic acid residues, glycosylation, lipid attachment, phosphorylation, oligomerization, hydroxylation and ADP-ribosylation. Exemplary modifications are described in most basic texts, such as, for example, Proteins - Structure and Molecular Properties, 2nd ed., T. E. Creighton, W. H. Freeman and Company, New York (1993), herein incoφorated by reference in its entirety. Many detailed reviews are available on this subject, such as, for example, those provided by Wold, F., Post-translational Protein Modifications. Perspectives and Prospects, pp. 1-12 in Post-translational Covalent Modification of Proteins, B. C. Johnson, Ed., Academic Press, New York (1983); Seifter et al., Meth. Enzymol. 182:626-M (1990) and Rattan et al., Protein Synthesis: Post-translational Modifications and Aging, Ann. N.Y. Acad. Sci. 663:48-62 (1992), herein incorporated by reference in their entirety. Modifications can occur anywhere in a polypeptide, including the peptide backbone, the amino acid side-chains and the amino or carboxyl termini. In fact, blockage of the amino or carboxyl group in a polypeptide, or both, by a covalent modification, is common in naturally occurring and synthetic polypeptides and such modifications may be present in polypeptides of the present invention, as well. For instance, the amino terminal residue of polypeptides made in E. coli or other cells, prior to proteolytic processing, almost invariably will be N-formylmethionine. During post-translational modification of the polypeptide, a methionine residue at the NH2 terminus may be deleted. Accordingly, this invention contemplates the use of both the methionine containing and the methionine-less amino terminal variants of the polypeptide of the invention. Thus, as used herein, the terms "protein" and "polypeptide" include any protein or polypeptide that is modified by any biological or non- biological process.
The terms "amino acid" and "amino acids" refer to all naturally occurring amino acids and, unless otherwise limited, known analogs of natural amino acids that can function in a similar manner as naturally occurring amino acids. This definition is meant to include norleucine, ornithine, homocysteine, and homoserine.
The term "amino acid sequence" means the sequence of amino acids in a polypeptide (or protein) that is written starting with the amino-terminal (N-terminal) residue and ending with the carboxyl-terminal (C-terminal) residue.
The term "an amino acid subsequence" means a portion of the amino acid sequence of a polypeptide. An amino acid subsequence generally has a length of 3 to 50 amino acid residues.
Both terms "substantially purified polypeptide" and "substantially purified protein", as used herein, means a polypeptide or protein that is separated substantially from all other molecules normally associated with it in its native state and is the predominant species present in a preparation. A substantially purified molecule may be greater than 60% free, preferably 75% free, more preferably 90% free, and most preferably 95% free from the other molecules (exclusive of solvent) present in the natural mixture.
Percentage of sequence identity is determined by comparing two optimally aligned sequences over a comparison window, wherein the portion of the polynucleotide or amino acid sequence in the comparison window may comprise additions or deletions (i.e., gaps) as compared to the reference sequence (which does not comprise additions or deletions) for optimal alignment of the two sequences. The percentage is calculated by determining the number of positions at which the identical nucleic acid base or amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the window of comparison and multiplying the result by 100 to yield the percentage of sequence identity. The term "identity" refers to amino acid or nucleic acid sequences that when compared using the local homology algorithm of Smith and Waterman (T. F. Smith and M. S. Waterman, J. Mol. Biol. (147) 195-197 (1981). in the SSEARCH3 3.0t75 (W.R. Pearson, Genomics (11)635- 650 (1991) or BLAST 2.2.1 (Altschul, Stephen F., Thomas L. Madden, Alejandro A. Schaffer, Jinghui Zhang, Zheng Zhang, Webb Miller, and David J. Lipman , Nucleic Acids Res. (25)3389- 3402 (1997) programs which are exactly alike.
The term "similarity" refers to two amino acids, which are similar as defined by similarity matrix BLOSSUM62 (S. Henikoff and J. G. Henikoff, , Proc. Natl. Acad. Sci. U.S.A. (89) 10915-10919 (1992) which is used in BLAST 2.2.1 (Altschul, Stephen F., Thomas L. Madden, Alejandro A. Schaffer, Jinghui Zhang, Zheng Zhang, Webb Miller, and David J. Lipman , Nucleic Acids Res. (25)3389-3402 (1997). The BLAST uses alias "positive" for similarity. These two terms, similarity and positive-ness, are interchangeable.
The term "percent identity" for a pair of protein sequences refers to the number of identical amino acid residues in a two-sequence alignment reported by BLAST, divided by the total number of amino acid residues in the same alignment, expressed in percentage. The term "percent similarity" for a pair of protein sequences refers to the number of similar ("Positive" in BLAST output) amino acid residues in a two-sequence alignment reported by BLAST, divided by the total number of amino acid residues in the same alignment, expressed in percentage.
Both terms "substantially identical" and "substantial identity", as reference to two amino acid sequences or two nucleotide sequences, means that one amino acid sequence or one nucleotide sequence has at least 50% sequence identity compared to the other amino acid sequence or nucleotide sequence as a reference sequence using the Gap program in the WISCONSIN PACKAGE version 10.0-UNIX from Genetics Computer Group, Inc. based on the method of Needleman and Wunsch (J. Mol. Biol. 48:443-453 (1970), herein incorporated by reference in its entirety) using the set of default parameters for pair-wise comparison (for amino acid sequence comparison: Gap Creation Penalty = 8, Gap Extension Penalty = 2; for nucleotide sequence comparison: Gap Creation Penalty = 50; Gap Extension Penalty = 3).
Polypeptides, which are "substantially similar" share sequences as noted above except that residue positions which are not identical may differ by conservative amino acid changes. Conservative amino acid substitutions refer to the interchangeability of residues having similar side chains. "Conservative amino acid substitutions" mean substitutions of one or more amino acids in a native amino acid sequence with another amino acid(s) having similar side chains, resulting in a silent change. Conserved substitutes for an amino acid within a native amino acid sequence can be selected from other members of the group to which the naturally occurring amino acid belongs. For example, a group of amino acids having aliphatic side chains is glycine, alanine, valine, leucine, and isoleucine; a group of amino acids having aliphatic-hydroxyl side chains is serine and threonine; a group of amino acids having amide-containing side chains is asparagine and glutamine; a group of amino acids having aromatic side chains is phenylalanine, tyrosine, and tryptophan; a group of amino acids having basic side chains is lysine, arginine, and histidine; and a group of amino acids having sulfur-containing side chains is cysteine and methionine. Preferred conservative amino acids substitution groups are: valine-leucine, valine- isoleucine, phenylalanine-tyrosine, lysine-arginine, alanine-valine, aspartic acid-glutamic acid, and asparagine-glutamine.
One skilled in the art will recognize that the values of the above substantial identity of nucleotide sequences can be appropriately adjusted to determine corresponding sequence identity of two nucleotide sequences encoding the polypeptides of the present invention by taking into account codon degeneracy, conservative amino acid substitutions, reading frame positioning and the like. Substantial identity of nucleotide sequences for these purposes normally means sequence identity of at least 35%). The term "codon degeneracy" means divergence in the genetic code permitting variation of the nucleotide sequence without affecting the amino acid sequence of an encoded polypeptide. The skilled artisan is well aware of the "codon-bias" exhibited by a specific host cell in usage of nucleotide codons to specify a given amino acid. Therefore, when synthesizing a gene for ectopic expression in a host cell, it is desirable to design the gene such that its frequency of codon usage approaches the frequency of preferred codon usage of the host cell. In another aspect, the present invention provides an isolated nucleic acid molecule in a DNA construct comprising a promoter operably linked to nucleotide sequence or complement thereof, wherein the nucleotide sequence hybridizes under stringent conditions to the complement of a second nucleotide sequence encoding a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10, SEQ ID NO: 12, SEQ ID NO: 14, SEQ ID NO: 16, SEQ ID NO: 18, SEQ ID NO: 20, SEQ ID NO: 22, SEQ ID NO: 24, SEQ ID NO: 26, SEQ ID NO: 28, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 34, SEQ ID NO: 36, SEQ ID NO: 38, SEQ ID NO: 40, SEQ ID NO: 42, SEQ ID NO: 44, SEQ ID NO: 46, SEQ ID NO: 48, SEQ ID NO: 50, SEQ ID NO: 52, SEQ ID NO: 54, SEQ ID NO: 67, SEQ ID NO: 69, and SEQ ID NO: 71.
The present invention also provides a method for obtaining an isolated nucleic acid molecule of the present invention, the method comprising the steps of: (a) probing a cDNA or genomic library with a hybridization probe comprising a nucleotide sequence encoding all or a portion of the amino acid sequence of a polypeptide, wherein the amino acid sequence of the polypeptide is selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 34, SEQ ID NO: 36, SEQ ID NO: 38, SEQ ID NO: 40, SEQ ID NO: 42, SEQ ID NO: 44, SEQ ID NO: 46, SEQ ID NO: 48, SEQ ID NO: 52, SEQ ID NO: 54, SEQ ID NO: 67, SEQ ID NO: 69, and SEQ ID NO: 71 (b) identifying a DNA clone that hybridizes under stringent conditions to hybridization probe; (c) isolating the DNA clone identified in step (b); and (d) sequencing the cDNA insert or genomic fragment contained in the DNA clone isolated in step (c) wherein the sequenced nucleic acid molecule encodes all or a substantial portion of the amino acid sequence of the present invention.
Hybridization conditions are sequence dependent and will be different in different circumstances. As used herein "stringent conditions" are selected to be about 5°C lower than the thermal melting point (Tm) for the specific sequence at a defined ionic strength and pH. The "thermal melting point" is the temperature (under defined ionic strength and pH) at which 50% of a target molecule hybridizes to a completely complementary molecule. Appropriate stringent conditions which promote DNA hybridization, for example, 6.0 X sodium chloride/sodium citrate (SSC) at about 45°C, followed by a wash of 2.0 X SSC at 50°C, are known to those skilled in the art or can be found in Current Protocols in Molecular Biology, John Wiley & Sons, N.Y. (1989), 6.3.1-6.3.6, incorporated herein by reference in its entirety. For example, the salt concentration in the wash step can be selected from a low stringent condition of about 2.0 X SSC at 50°C to a high stringency of about 0.2 X SSC at 50°C. In addition, the temperature in the wash step can be increased from low stringent conditions at room temperature, about 22°C, to high stringent conditions at about 65°C. Both temperature and salt concentration may be varied, or either the temperature or the salt concentration may be held constant while the other variable is changed. For the purposes of this disclosure, stringent conditions include at least one wash in 2.0 X SSC at a temperature of at least about 50°C for 20 minutes, or equivalent conditions.
The nucleic acid molecules of the present invention may be combined with other non- native, or "heterologous" sequences in a variety of ways. By "heterologous" sequences it is meant any sequence that is not naturally found joined to the nucleotide sequence encoding polypeptide of the present invention, including, for example, combinations of nucleotide sequences from the same plant which are not naturally found joined together, or the two sequences originate from two different species.
The term "homologous" refers to two or more genes that are derived from a single gene in a common ancestor. The term "homology is attributed to decent from a common ancestor. (J. J. Doyle and B. S. Gaut, Plant Molecular Biology (42)1-23, 2000.
The term "orthologous" refers to different homologous sequences in different species that arose from a common ancestral gene during speciation; may or may not be responsible for a similar function. The term paralogous refers to homologous sequences within a single species that arose by gene duplication.
The term "domain" refers to a discrete portion of a protein assumed to fold independently of the rest of the protein and possessing its own function. Domain is also used to refer a discrete portion of nucleotide when translated provides a portion of protein which is assumed to fold independently of the rest of the translated nucleotide sequence and possessing its own function.
The term "Motif refers to a short conserved region in a protein or nucleotide sequence. Motifs are frequently highly conserved parts of domains.
The term "genome" as it applies to plant cells encompasses not only chromosomal DNA found within the nucleus, but organelle DNA found within subcellular components of the cell. DNAs of the present invention introduced into plant cells can therefore be either chromosomally integrated or organelle-localized. The term "operably linked", as used in reference to a regulatory sequence and a structural nucleotide sequence, means that the regulatory sequence causes regulated expression of the operably linked structural nucleotide sequence. "Expression" means the transcription and stable accumulation of sense or antisense RNA derived from the nucleic acid molecule of the present invention. Expression may also refer to translation of mRNA into a polypeptide. "Sense" RNA means RNA transcript that includes the mRNA and so can be translated into polypeptide or protein by the cell. "Antisense RNA" means a RNA transcript that is complementary to all or part of a target primary transcript or mRNA and that blocks the expression of a target gene (U.S. Pat. No. 5,107,065, incorporated herein by reference). The complementarity of an antisense RNA may be with any part of the specific gene transcript, i.e., at the 5' non-coding sequence, 3' non- translated sequence, introns, or the coding sequence. "RNA transcript" means the product resulting from RNA polymerase-catalyzed transcription of a DNA sequence. When the RNA transcript is a perfect complementary copy of the DNA sequence, it is referred to as the primary transcript or it may be a RNA sequence derived from post-transcriptional processing of the primary transcript and is referred to as the mature RNA.
The term "overexpression" means the expression of a polypeptide encoded by an exogenous nucleic acid molecule introduced into a host cell, wherein said polypeptide is either not normally present in the host cell, or wherein said polypeptide is present in said host cell at a higher level than that normally expressed from the endogenous gene encoding said polypeptide. By "ectopic expression" it is meant that expression of a nucleic acid molecule encoding a polypeptide in a cell type other than a cell type in which the nucleic acid molecule is normally expressed, at a time other than a time at which the nucleic acid molecule is normally expressed or at a expression level other than the level at which the nucleic acid molecule normally is expressed. "Antisense inhibition" means the production of antisense RNA transcripts capable of suppressing the expression of the target polypeptide. "Co-suppression" means the production of sense RNA transcripts capable of suppressing the expression of identical or substantially similar foreign or endogenous genes (U.S. Patent No. 5,231,020, incorporated herein by reference).
The term "a gene" means the segment of DNA that is involved in producing a polypeptide. Such segment of DNA includes regulatory sequences preceding (5' non-coding sequences) and following (3' non-coding sequences) the coding region as well as intervening sequences (introns) between individual coding segments (exons). A "Native gene" means a gene as found in nature with its own regulatory sequences. "Chimeric gene" means any gene that is not a native gene, comprising regulatory and coding sequences that are not found together in nature. Accordingly, a chimeric gene may comprise regulatory sequences and coding sequences that are derived from different sources, or regulatory sequences and coding sequences derived from the same source, but arranged in a manner different than that found in nature. "Endogenous gene" or "endogenous DNA molecule" means a native gene in its natural location in the genome of an organism. A "foreign gene" means a gene not normally found in the host organism, but that is introduced into the host organism by gene transfer. Foreign genes can comprise native genes inserted into a non-native organism, or chimeric genes. A "transgene" is a gene that has been introduced into the genome by a transformation procedure.
"Regulatory sequences" mean nucleotide sequences located upstream (5' non-coding sequences), within, or downstream (3' non-translated sequences) of a structural nucleotide sequence, and which influence the transcription, RNA processing or stability, or translation of the associated structural nucleotide sequence. Regulatory sequences may include promoters, translation leader sequences, introns, and polyadenylation recognition sequences.
The term "promoter sequence" means a nucleotide sequence that is capable of, when located in cis to a structural nucleotide sequence encoding a polypeptide, functioning in a way that directs expression of one or more mRNA molecules that encodes the polypeptide. Such promoter regions are typically found upstream of the trinucleotide ATG sequence at the start site of a polypeptide-coding region. Promoter sequences can also include sequences from which transcription of transfer RNA (tRNA) or ribosomal RNA (rRNA) sequences are initiated. Transcription involves the synthesis of a RNA chain representing one strand of a DNA duplex. By "representing" it is meant that the RNA is identical in sequence with one strand of the DNA; it is complementary to the other DNA strand, which provides the template for its synthesis. Transcription takes place by the usual process of complementary base pairing, catalyzed and scrutinized by the enzyme RNA polymerase. The reaction can be divided into three stages described as initiation, elongation and termination. Initiation begins with the binding of RNA polymerase to the double stranded (DS or ds) DNA. The sequence of DNA required for the initiation reaction defines the promoter. The site at which the first nucleotide is incorporated is called the start-site or start-point of transcription. Elongation describes the phase during which the enzyme moves along the DNA and extends the growing RNA chain. Elongation involves the disruption of the DNA double stranded structure in which a transiently unwound region exists as a hybrid RNA-DNA duplex and a displaced single strand of DNA. Termination involves recognition of the point at which no further bases should be added to the chain. To terminate transcription, the formation of phosphodiester bonds must cease and the transcription complex must come apart. When the last base is added to the RNA chain, the RNA-DNA hybrid is disrupted, the DNA reforms into a duplex state, and the RNA polymerase enzyme and RNA molecule are both released from the DNA. The sequence of DNA required for the termination reaction is called the terminator. The promoter sequence consists of proximal and more distal upstream elements, the latter elements often referred to as enhancers. Accordingly, an "enhancer" is a DNA sequence that can stimulate promoter activity and may be an innate element of the promoter or a heterologous element inserted to enhance the level or tissue-specificity of a promoter. Promoters may be derived in their entirety from a native gene, or be composed of different elements derived from different promoters found in nature, or even comprise synthetic DNA segments. It is understood by those skilled in the art that different promoters may direct the expression of a gene in different tissues or cell types, or at different stages of development, or in response to different environmental conditions.
Promoters, which are known or are found to cause transcription of DNA in plant cells, can be used in the present invention. Such promoters may be obtained from a variety of sources such as plants and plant viruses. A number of promoters, including constitutive promoters, inducible promoters and tissue-specific promoters, that are active in plant cells have been described in the literature. In addition to promoters that are known to cause transcription of DNA in plant cells, other promoters may be identified for use in the current invention by screening a plant cDNA library for genes that are selectively or preferably expressed in the target tissues and then determine the promoter regions.
The term "constitutive promoter" means a regulatory sequence which causes expression of a structural nucleotide sequence in most cells or tissues at most times. Constitutive promoters are active under most environmental conditions and states of development or cell differentiation. A variety of constitutive promoters are well known in the art. Examples of constitutive promoters that are active in plant cells include but are not limited to the nopaline synthase (NOS) promoters; DNA plant virus promoters including, but not limited to the caulimovirus promoters for example, cauliflower mosaic virus (CaMV) 19S and 35S, and figwort mosaic virus promoters; the bacilliform virus promoter for example sugar cane bacilliform virus, rice tungro bacilliform virus, among others; plant actin promoters, such as the Arabidopsis and rice actin gene promoter (see, e.g., Huang et al, Plant Mol. Biol. 33:125-139 (1997), US Patent No. 5,641,876), herein incorporated by reference in its entirety). These promoters when used in a DNA construct are heterologous to the linked gene sequence when they are derived from a different organism, plant species, or a different gene.
The term "inducible promoter" means a regulatory sequence which causes conditional expression of a structural nucleotide sequence under the influence of changing environmental conditions or developmental conditions. Examples of inducible promoters include but are not limited to the senescence-induced promoter for the senescence-associated gene, SAG 12, (Gan and Amasino, Science 270: 1986-1988 (1995), herein incorporated by reference in its entirety); the light-inducible promoter from the small subunit of ribulose-l,5-bis-phosphate carboxylase (ssRUBISCO); the drought-inducible promoter of maize (Busk et al, Plant J. 77:1285-1295 (1997), herein incoφorated by reference in its entirety); the cold, drought, and high salt inducible promoter from potato (Kirch, Plant Mol. Biol. 33:897-909 (1997), herein incorporated by reference in its entirety); a nitrate-inducible promoter derived from the spinach nitrite reductase gene (Back et al, Plant Mol. Biol. 17:9 (1991), herein incoφorated by its entirety); salicylic acid inducible promoter (Uknes et al, Plant Cell 5:159-169 (1993); Bi et al, Plant J. 5:235-245 (1995) herein incoφorated by reference in their entireties); the auxin-response elements El promoter fragment (AuxREs) in the soybean (Glycine max L.) (Liu et al, Plant Physiol. 775:397-407 (1997), herein incorporated by reference in its entirety); the auxin-responsive Arabidopsis GST6 promoter (also responsive to salicylic acid and hydrogen peroxide) (Chen et al, Plant J. 10: 955-966 (1996), herein incoφorated by reference in its entirety); the auxin- inducible parC promoter from tobacco (Sakai et al, Plant Cell Physiol. 37:906-913 (1996), herein incoφorated by reference in its entirety); a plant biotin response element (Streit et al, Mol. Plant Microbe Interact. 70:933-937 (1997), herein incoφorated by reference in its entirety); the promoter responsive to the stress hormone abscisic acid (Sheen et al, Science 274:1900-1902 (1996), herein incorporated by reference in its entirety); the maize In2-2 promoter activated by benzenesulfonamide herbicide safeners (De Veylder et al, Plant Cell Physiol 35:568-577 (1997), herein incoφorated by reference in its entirety); a tetracycline- inducible promoter, such as the promoter for the Avena sativa L. (oat) arginine decarboxylase gene (Masgrau et al, Plant J. 77:465-473 (1997), herein incoφorated by reference in its entirety); and a salicylic acid-responsive element (Stange et al, Plant J. 77:1315-1324 (1997), herein incorporated by reference in its entirety).
The term "tissue-specific promoter" or "organ enhanced promoter" means a regulatory sequence that causes transcriptions or enhanced transcriptions of DNA in specific cells or tissues at specific times during plant development, such as in vegetative tissues or reproductive tissues. Examples of tissue-specific promoters under developmental control include promoters that initiate transcription only (or primarily only) in certain tissues, such as vegetative tissues, e.g., roots, leaves or stems, or reproductive tissues, such as fruit, ovules, seeds, pollen, pistols, flowers, or any embryonic tissue. Reproductive tissue specific promoters may be, e.g., ovule- specific, embryo-specific, endosperm-specific, integument-specific, seed coat-specific, pollen- specific, petal-specific, sepal-specific, or some combination thereof. One skilled in the art will recognize that a tissue-specific promoter may drive expression of operably linked sequences in tissues other than the target tissue. Thus, as used herein a tissue-specific promoter is one that drives expression preferentially in the target tissue, but may also lead to some expression in other tissues as well.
A variety of promoters specifically active in vegetative tissues, such as leaves, stems, roots and tubers, can be used to express the nucleic acid molecules of the present invention. Examples of tuber-specific promoters include but are not limited to the class I and II patatin promoters (Bevan et al, EMBO J. 8: 1899-1906 (1986); Koster-Topfer et al, Mol Gen Genet. 219: 390-396 (1989); Mignery et al, Gene. 62: 27-44 (1988); Jefferson et al, Plant Mol. Biol 14: 995-1006 (1990), herein incoφorated by reference in their entireties), the promoter for the potato tuber ADPGPP genes, both the large and small subunits; the sucrose synthase promoter (Salanoubat and Belliard, Gene. 60: 47-56 (1987), Salanoubat and Belliard, Gene. 84: 181-185 (1989), herein incoφorated by reference in their entirety); and the promoter for the major tuber proteins including the 22 kd protein complexes and proteinase inhibitors (Hannapel, Plant Physiol. 101: 703-704 (1993), herein incoφorated by reference in its entirety). Examples of leaf- specific promoters include but are not limited to the ribulose biphosphate carboxylase (RBCS or RuBISCO) promoters (see, e.g., Matsuoka et al, Plant J. 6:311-319 (1994), herein incoφorated by reference in its entirely); the light harvesting chlorophyll a/b binding protein gene promoter (see, e.g., Shhna et β/., R/α..tR/nwø/. 775:477-483 (1997); Cassλ et al, Plant Physiol. 116:1533- 1538 (1998), herein incoφorated by reference in their entireties); and the Arabidopsis thaliana myb-related gene promoter (Atmyb5) (Li et al, FEBS Lett. 379:111-121 (1996), herein incoφorated by reference in its entirety). Examples of root-specific promoter include but are not limited to the promoter for the acid chitinase gene (Samac et al, Plant Mol. Biol 25: 587-596 (1994), herein incoφorated by reference in its entirety); the root specific subdomains of the CaMV35S promoter that have been identified (Lam et al, Proc. Natl. Acad. Sci. (U.S.A.) 5(5:7890-7894 (1989), herein incorporated by reference in its entirety); the ORF13 promoter from Agrobacterium rhizogenes which exhibits high activity in roots (Hansen et al, Mol. Gen. Genet. 254:331-343 (1997), herein incoφorated by reference in its entirety); the promoter for the tobacco root-specific gene TobRB7 (Yamamoto et al, Plant Cell 3:371-382 (1991), herein incoφorated by reference in its entirety); and the root cell specific promoters reported by Conkling et al. (Conkling et al, Plant Physiol. 93: 1203-1211 (1990), herein incorporated by reference in its entirety). Another class of useful vegetative tissue-specific promoters is meristematic (root tip and shoot apex) promoters. For example, the "SHOOTMERISTEMLESS" and "SCARECROW" promoters, which are active in the developing shoot or root apical meristems (Di Laurenzio et al, Cell 86:423- 433 (1996); Long, Nature 379:66-69 (1996); herein incoφorated by reference in their entireties), can be used. Another example of a useful promoter is that which controls the expression of 3-hydroxy-3- methylglutaryl coenzyme A reductase HMG2 gene, whose expression is restricted to meristematic and floral (secretory zone of the stigma, mature pollen grains, gynoecium vascular tissue, and fertilized ovules) tissues (see, e.g., Enjuto et al, Plant Cel 7:517-527 (1995), herein incorporated by reference in its entirety). Also another example of a useful promoter is that which controls the expression of knl-related genes from maize and other species which show meristem-specific expression (see, e.g., Granger et al, Plant Mol Biol. 37:373-378 (1996); Kerstetter et al, Plant Cell (5:1877-1887 (1994); Hake et al, Philos. Trans. R. Soc. Lond. B. Biol. Sci. 350:45-51 (1995), herein incoφorated by reference in their entireties). Another example of a meristematic promoter is the Arabidopsis thaliana KNAT1 promoter. In the shoot apex, KNAT1 transcript is localized primarily to the shoot apical meristem; the expression of KNATI in the shoot meristem decreases during the floral transition and is restricted to the cortex of the inflorescence stem (see, e.g., Lincoln et al, Plant Cell
(5:1859-1876 (1994), herein incoφorated by reference in its entirety).
Suitable seed-specific promoters can be derived from the following genes: MAC1 from maize (Sheridan et al, Genetics 742:1009-1020 (1996), herein incoφorated by reference in its entirety); Cat3 from maize (GenBank No. L05934, Abler et al, Plant Mol. Biol. 22:10131-1038
(1993), herein incoφorated by reference in its entirety); vivparous-1 from Arabidopsis (Genbank
No. U93215); Atimycl from Arabidopsis (Urao et al, Plant Mol Biol 32:571-57 (1996);
Conceicao et al, Plant 5:493-505 (1994), herein incorporated by reference in their entireties); napA from Brassica napus (GenBank No. J02798); the napin gene family from Brassica napus (Sjodahl et al, Planta 197:264-211 (1995), herein incoφorated by reference in its entirety).
The ovule-specific promoter for BEL1 gene (Reiser et al Cell 53:735-742 (1995),
GenBank No. U39944; Ray et al, Proc. Natl. Acad. Sci. USA 91:5161-5165 (1994), all of which are herein incorporated by reference in their entireties) can also be used. The egg and central cell specific MEA (FIS1) and FIS2 promoters are also useful reproductive tissue-specific promoters (Luo et al., Proc. Natl. Acad. Sci. USA, 97:10637-10642 (2000); Vielle-Calzada, et al., Genes
Dev. 13:2971-2982 (1999); herein incorporated by reference in their entireties).
A maize pollen-specific promoter has been identified in maize (Guerrero et al., Mol. Gen.
Genet. 224:161-168 (1990), herein incoφorated by reference in its entirety). Other genes specifically expressed in pollen have been described (see, e.g., Wakeley et al., Plant Mol. Biol. 37:187-192 (1998); Ficker et al., Mol. Gen. Genet. 257: 132-142 (1998); Kulikauskas et al., Plant
Mol. Biol. 34:809-814 (1997); Treacy et al., Plant Mol. Biol. 34:603-611 (1997); all of which are herein incorporated by reference in their entireties).
Promoters derived from genes encoding embryonic storage proteins, which includes the gene encoding the 2S storage protein from Brassica napus (Dasgupta et al, Gene 133:301-302 (1993), herein incorporated by reference in its entirety); the 2S seed storage protein gene family from Arabidopsis; the gene encoding oleosin 20kD from Brassica napus (GenBank No.
M63985); the genes encoding oleosin A (GenBank No. U09118) and oleosin B (GenBank No.
U09119) from soybean; the gene encoding oleosin from Arabidopsis (GenBank No. Z17657); the gene encoding oleosin 18kD from maize (GenBank No. J05212, Lee, Plant Mol. Biol. 26:1981-1987 (1994), herein incoφorated by reference in its entirety); and the gene encoding low molecular weight sulphur rich protein from soybean (Choi et al., Mol. Gen. Genet. 246:266- 268 (1995), herein incoφorated by reference in its entirety), can also be used.
Promoters derived from zein encoding genes (including the 15 kD, 16 kD, 19 kD, 22 kD, 27 kD, and gamma genes) (Pedersen et al, Cell 29: 1015-1026 (1982), herein incorporated by reference in its entirety) can be also used. The zeins are a group of storage proteins found in maize endosperm.
Other promoters known to function, for example, in maize, include the promoters for the following genes: waxy, Brittle, Shrunken 2, Branching enzymes I and II, starch synthases, debranching enzymes, oleosins, glutelins, and sucrose synthases. A particularly preferred promoter for maize endosperm expression is the promoter for the glutelin gene from rice, more particularly the Osgt-1 promoter (Zheng et al, Mol. Cell Biol. 13: 5829-5842 (1993), herein incoφorated by reference in its entirety). Examples of promoters suitable for expression in wheat include those promoters for the ADPglucose pyrophosphorylase (ADPGPP) subunits, the granule bound and other starch synthases, the branching and debranching enzymes, the embryogenesis-abundant proteins, the gliadins, and the glutenins. Examples of such promoters in rice include those promoters for the ADPGPP subunits, the granule bound and other starch synthases, the branching enzymes, the debranching enzymes, sucrose synthases, and the glutelins. A particularly preferred promoter is the promoter for rice glutelin, Osgt-1. Examples of such promoters for barley include those for the ADPGPP subunits, the granule bound and other starch synthases, the branching enzymes, the debranching enzymes, sucrose synthases, the hordeins, the embryo globulins, and the aleurone specific proteins.
A tomato promoter active during fruit ripening, senescence and abscission of leaves and, to a lesser extent, of flowers can be used (Blume et al., Plant J. 72:731-746 (1997), herein incorporated by reference in its entirety). Other exemplary promoters include the pistol specific promoter in the potato (Solarium tuberosum L.) SK2 gene, encoding a pistil-specific basic endochitinase (Ficker et al, Plant Mol Biol. 35:425-431 (1997), herein incoφorated by reference in its entirety); the Blec4 gene from pea (Pisum sativum cv. Alaska), active in epidermal tissue of vegetative and floral shoot apices of transgenic alfalfa. This makes it a useful tool to target the expression of foreign genes to the epidermal layer of actively growing shoots. The tissue specific E8 promoter from tomato is also useful for directing gene expression in fruits. It is recognized that additional promoters that may be utilized are described, for example, in U.S. Patent Nos. 5,378,619, 5,391,725, 5,428,147, 5,447,858, 5,608,144, 5,608,144, 5,614,399, 5,633,441, 5,633,435, and 4,633,436, all of which are herein incoφorated in their entirety. In addition, a tissue specific enhancer may be used (Fromm et al., The Plant Cell 7:977-984 (1989), herein incoφorated by reference in its entirety). It is further recognized that since in most cases the exact boundaries of regulatory sequences have not been completely defined, DNA fragments of different lengths may have identical promoter activity.
The "translation leader sequence" means a DNA sequence located between the promoter sequence of a gene and the coding sequence. The translation leader sequence is present in the fully processed mRNA upstream of the translation start sequence. The translation leader sequence may affect processing of the primary transcript to mRNA, mRNA stability or translation efficiency. Examples of translation leader sequences have been described (US Patent No. 5,659,122 and Turner and Foster, Molecular Biotechnology 3:225 (1995) herein incoφorated by reference in its entirety). The "3' non-translated sequences" means DNA sequences located downstream of a structural nucleotide sequence and include sequences encoding polyadenylation and other regulatory signals capable of affecting mRNA processing or gene expression. The polyadenylation signal functions in plants to cause the addition of polyadenylate nucleotides to the 3' end of the mRNA precursor. The polyadenylation sequence can be derived from the natural gene, from a variety of plant genes, or from T-DNA. An example of the polyadenylation sequence is the nopaline synthase 3' sequence (NOS 3'; Fraley et al, Proc. Natl. Acad. Sci. USA 80: 4803-4807 (1983), herein incoφorated by reference in its entirety). The use of different 3' non-translated sequences is exemplified by Ingelbrecht et al, Plant Cell 7:671-680 (1989), herein incoφorated by reference in its entirety. The isolated nucleic acid molecules of the present invention may also include introns.
Generally, optimal expression in monocotyledonous and some dicotyledonous plants is obtained when an intron sequence is inserted between the promoter sequence and the structural gene sequence or, optionally, may be inserted in the structural coding sequence to provide an interrupted coding sequence. An example of such an intron sequence is the HSP 70 intron described in WO 93/19189, herein incoφorated by reference in its entirety. The laboratory procedures in recombinant DNA technology used herein are those well known and commonly employed in the art. Standard techniques are used for cloning, DNA and RNA isolation, amplification and purification. Generally enzymatic reactions involving DNA ligase, DNA polymerase, restriction endonucleases and the like are performed according to the manufacturer's specifications. These techniques and various other techniques are generally performed according to Sambrook et al., Molecular Cloning - A Laboratory Manual, 2nd. ed., Cold Spring Harbor Laboratory, Cold Spring Harbor, New York (1989).
Isolation and identification of nucleic acid molecules encoding polypeptides of the present invention from soybean, corn, rice and other species are described in detail in Examples. All or a substantial portion of the nucleic acid molecules of the present invention may be used to isolate cDNAs and nucleic acid molecules encoding homologous polypeptides from the same or other plant species.
A "substantial portion" of a nucleotide sequence comprises enough of the sequence to afford specific identification and/or isolation of a nucleic acid molecule comprising the sequence. Nucleotide sequences can be evaluated either manually by one skilled in the art, or by using computer-based sequence comparison and identification tools that employ algorithms such as BLAST (Basic Local Alignment Search Tool; Altschul et al. JMol Biol 275:403-410 (1993). In general, a sequence of thirty or more contiguous nucleotides is necessary in order to putatively identify a nucleotide sequence as homologous to a gene. Moreover, with respect to nucleotide sequences, gene-specific oligonucleotide probes comprising 30 or more contiguous nucleotides may be used in sequence-dependent methods of gene identification (e.g., Southern hybridization) and isolation (e.g., in situ hybridization of bacterial colonies or bacteriophage plaques). In addition, short oligonucleotides of 12 or more nucleotides may be used as amplification primers in PCR in order to obtain a particular nucleic acid molecule comprising the primers. The skilled artisan, having the benefit of the sequences as reported herein, may now use all or a substantial portion of the disclosed sequences for puφoses known to those skilled in this art. Accordingly, the instant invention comprises the complete sequences as reported in the accompanying Sequence Listing, as well as substantial portions of those sequences as defined above.
Isolation of nucleic acid molecules encoding homologous polypeptides using sequence- dependent protocols is well known in the art. Examples of sequence-dependent protocols include, but are not limited to, methods of nucleic acid molecule hybridization, and methods of DNA and RNA amplification as exemplified by various uses of nucleic acid molecule amplification technologies (e.g., polymerase chain reaction, ligase chain reaction).
For example, structural nucleic acid molecules encoding other polypeptide of the present invention, either as cDNAs or genomic DNAs, could be isolated directly by using all or a substantial portion of the nucleic acid molecules of the present invention as DNA hybridization probes to screen cDNA or genomic libraries from any desired plant employing methodology well known to those skilled in the art. Methods for forming such libraries are well known in the art. Specific oligonucleotide probes based upon the nucleic acid molecules of the present invention can be designed and synthesized by methods known in the art. Moreover, the entire sequences of the nucleic acid molecules can be used directly to synthesize DNA probes by methods known to the skilled artisan such as random primer DNA labeling, nick translation, or end-labeling techniques, or RNA probes using available in vitro transcription systems. In addition, specific primers can be designed and used to amplify a part or all of the sequences. The resulting amplification products can be labeled directly during amplification reactions or labeled after amplification reactions, and used as probes to isolate full-length cDNA or genomic DNAs under conditions of appropriate stringency.
Alternatively, the nucleic acid molecules of interest can be amplified from nucleic acid samples using amplification techniques. For instance, the disclosed nucleic acid molecules may be used to define a pair of primers that can be used with the polymerase chain reaction (Mullis, et al, Cold Spring Harbor Symp. Quant. Biol. 57:263-273 (1986); Erlich et al, EP 50,424; EP 84,796, EP 258,017, EP 237,362; Mullis, EP 201,184; Mullis et al, US 4,683,202; Erlich, US 4,582,788; and Saiki, R. et al, US 4,683,194, all of which are herein incoφorated by reference in their entireties) to amplify and obtain any desired nucleic acid molecule directly from mRNA, from cDNA, from genomic libraries or cDNA libraries. PCR and other in vitro amplification methods may also be useful, for example, to clone nucleotide sequences that encode for polypeptides to be expressed, to make nucleic acid molecules to use as probes for detecting the presence of the desired mRNA in samples, for nucleic acid sequencing, or for other purposes. In addition, two short segments of the nucleic acid molecules of the present invention may be used in polymerase chain reaction protocols to amplify longer nucleic acid molecules encoding homologues of a polypeptide of the present invention from DNA or RNA. For example, the skilled artisan can follow the RACE protocol (Frohman et al., Proc. Natl. Acad. Sci. USA 85:8998 (1988), herein incoφorated by reference in its entirety) to generate cDNAs by using PCR to amplify copies of the region between a single point in the transcript and the 3 ' or 5' end. Primers oriented in the 3' and 5' directions can be designed from the nucleic acid molecules of the present invention. Using commercially available 3 'RACE or 5 'RACE systems (Gibco BRL, Life Technologies, Gaithersburg, Maryland U.S.A.), specific 3' or 5' cDNA fragments can be isolated (Ohara et al., Proc. Natl. Acad. Sci. USA 86:5673 (1989); Loh et al., Science 243:217 (1989), both of which are herein incoφorated by reference in their entireties). Products generated by the 3' and 5' RACE procedures can be combined to generate full-length cDNAs (Frohman and Martin, Techniques ! : 165 (1989), herein incorporated by reference in its entirety). Nucleic acid molecules of interest may also be synthesized, either completely or in part, especially where it is desirable to provide plant-preferred sequences, by well-known techniques as described in the technical literature. See, e.g., Carruthers et al., Cold Spring Harbor Symp. Quant. Biol. 47:411-418 (1982), and Adams et al., J. Am. Chem. Soc. 105:661 (1983), both of which are herein incoφorated by reference in their entireties. Thus, all or a portion of the nucleic acid molecules of the present invention may be synthesized using codons preferred by a selected plant host. Plant-preferred codons may be determined, for example, from the codons used most frequently in the proteins expressed in a particular plant host species. Other modifications of the gene sequences may result in mutants having slightly altered activity
Availability of the nucleotide sequences encoding polypeptide of the present invention facilitates immunological screening of cDNA expression libraries. Synthetic polypeptides representing portions of the amino acid sequences of polypeptides of the present invention may be synthesized. These polypeptides can be used to immunize animals to produce polyclonal or monoclonal antibodies with specificity for polypeptides comprising the amino acid sequences. These antibodies can be then be used to screen cDNA expression libraries to isolate full-length cDNA clones of interest (Lemer, Adv. Immunol. 36: 1 (1984); Sambrook et al., Molecular Cloning: A Laboratory Manual; Cold Spring Harbor Laboratory Press: Cold Spring Harbor, (1989)). It is understood that people skilled in the art are familiar with the standard resource materials which describe specific conditions and procedures for the construction, manipulation and isolation of antibodies (see, for example, Harlow and Lane, In Antibodies: A Laboratory Manual, Cold Spring Harbor Press, Cold Spring Harbor, New York (1988)). Another aspect of the present invention relates to methods of making a DNA construct by obtaining a nucleic acid molecule comprising a nucleotide sequence encoding a polypeptide of the present invention, the amino acid sequence of which has at least 50% sequence identity to a polypeptide selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10, SEQ ID NO: 12, SEQ ID NO: 14, SEQ ID NO: 16, SEQ ID NO: 18, SEQ ID NO: 20, SEQ ID NO: 22, SEQ ID NO: 24, SEQ ID NO: 26, SEQ ID NO: 28, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 34, SEQ ID NO: 36, SEQ ID NO: 38, SEQ ID NO: 40, SEQ ID NO: 42, SEQ ID NO: 44, SEQ ID NO: 46, SEQ ID NO: 48, SEQ ID NO: 50, SEQ ID NO: 52, SEQ ID NO: 54, SEQ ID NO: 67, SEQ ID NO: 69, and SEQ ID NO: 71. One aspect of the present invention provides an isolated nucleic acid molecule comprising a nucleotide sequence or complement thereof, wherein the nucleotide sequence encodes a polypeptide having an amino acid sequence that has at least 50% sequence identity, preferably at least 60%, more preferably at least 70% sequence identity, even more preferably at least 80% or 90% sequence identity, and most preferably at least 95% to 98% sequence identity to a polypeptide selected from group consisting of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10, SEQ ID NO: 12, SEQ ID NO: 14, SEQ ID NO: 16, SEQ ID NO: 18, SEQ ID NO: 20, SEQ ID NO: 22, SEQ ID NO: 24, SEQ ID NO: 26, SEQ ID NO: 28, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 34, SEQ ID NO: 36, SEQ ID NO: 38, SEQ ID NO: 40, SEQ ID NO: 42, SEQ ID NO: 44, SEQ ID NO: 46, SEQ ID NO: 48, SEQ ID NO: 50, SEQ ID NO: 52, SEQ ID NO: 54, SEQ ID NO: 67, SEQ ID NO: 69, and SEQ ID NO: 71.
One method of the present invention is for obtaining a nucleic acid molecule encoding all or a substantial portion of the amino acid sequence of a polypeptide of the present invention comprising: (a) probing a cDNA or genomic library with a hybridization probe comprising a nucleotide sequence encoding all or a substantial portion of a polypeptide having an amino acid sequence set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10, SEQ ID NO: 12, SEQ ID NO: 14, SEQ ID NO: 16, SEQ ID NO: 18, SEQ ID NO: 20, SEQ ID NO: 22, SEQ ID NO: 24, SEQ ID NO: 26, SEQ ID NO: 28, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 34, SEQ ID NO: 36, SEQ ID NO: 38, SEQ ID NO: 40, SEQ ID NO: 42, SEQ ID NO: 44, SEQ ID NO: 46, SEQ ID NO: 48, SEQ ID NO: 50, SEQ ID NO: 52, SEQ ID NO: 54, SEQ ID NO: 67, SEQ ID NO: 69, SEQ ID NO: 71 or an amino acid sequence set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10, SEQ ID NO: 12, SEQ ID NO: 14, SEQ ID NO: 16, SEQ ID NO: 18, SEQ ID NO: 20, SEQ ID NO: 22, SEQ ID NO: 24, SEQ ID NO: 26, SEQ ID NO: 28, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 34, SEQ ID NO: 36, SEQ ID NO: 38, SEQ ID NO: 40, SEQ ID NO: 42, SEQ ID NO: 44, SEQ ID NO: 46, SEQ ID NO: 48, SEQ ID NO: 50, SEQ ID NO: 52, SEQ ID NO: 54, SEQ ID NO: 67, SEQ ID NO: 69, SEQ ID NO: 71 with conservative amino acid substitutions; (b) identifying a DNA clone that hybridizes under stringent conditions to the hybridization probe; (c) isolating the DNA clone identified in step (b); and (d) sequencing the cDNA or genomic fragment that comprises the clone isolated in step (c) wherein the sequenced nucleic acid molecule encodes all or a substantial portion of the amino acid sequence of polypeptide of the present invention.
Another method of the present invention for obtaining a nucleic acid molecule encoding all or a substantial portion of the amino acid sequence of a polypeptide of the present invention comprising: (a) synthesizing a first and a second oligonucleotide primers, wherein the sequences of the first and second oligonucleotide primers encode two different portions of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10, SEQ ID NO: 12, SEQ ID NO: 14, SEQ ID NO: 16, SEQ ID NO: 18, SEQ ID NO: 20, SEQ ID NO: 22, SEQ ID NO: 24, SEQ ID NO: 26, SEQ ID NO: 28, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 34, SEQ ID NO: 36, SEQ ID NO: 38, SEQ ID NO: 40, SEQ ID NO: 42, SEQ ID NO: 44, SEQ ID NO: 46, SEQ ID NO: 48, SEQ ID NO: 50, SEQ ID NO: 52, SEQ ID NO: 54, SEQ ID NO: 67, SEQ ID NO: 69, SEQ ID NO: 71; and (b) amplifying and obtaining the nucleic acid molecule directly from mRNA samples, from genomic libraries or from cDNA libraries using the first and second oligonucleotide primers of step (a) wherein the nucleic acid molecule encodes all or a substantial portion of the amino acid sequence polypeptide of the present invention. All or a substantial portion of the nucleic acid molecules of the present invention may also be used as probes for genetically and physically mapping the genes that they are a part of, and as markers for traits linked to those genes. Such information may be useful in plant breeding in order to develop lines with desired phenotypes. For example, the nucleic acid molecules of the present invention may be used as restriction fragment length polymoφhism (RFLP) markers. Southern blots (Maniatis et al., Molecular Cloning: A Laboratory Manual, Second Edition (1989) Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y., herein incoφorated by reference in its entirety) of restriction-digested plant genomic DNA may be probed with the nucleic acid fragments of the present invention. The resulting banding patterns may then be subjected to genetic analyses using computer programs such as MapMaker (Lander et al., Genomics 7:174-181 (1987), herein incoφorated by reference in its entirety) in order to construct a genetic map. In addition, the nucleic acid fragments of the present invention may be used to probe Southern blots containing restriction endonuclease-treated genomic DNAs of a set of individuals representing parent and progeny of a defined genetic cross. Segregation of the DNA polymoφhisms is noted and used to calculate the position of the nucleotide sequence of the present invention in the genetic map previously obtained using this population (Botstein et al, Am. J. Hum. Genet. 32:314-331 (1980), herein incoφorated by reference in its entirety).
The production and use of plant gene-derived probes for use in genetic mapping is described in Bernatzky and Tanksley, Plant Mol. Biol. Reporter 4:31-41 (1986), herein incorporated by reference in its entirety. Numerous publications describe genetic mapping of specific cDNA clones using the methodology outlined above or variations thereof. For example, F2 intercross populations, backcross populations, randomly mated populations, near isogenic lines, exotic germplasms, and other sets of individuals may be used for mapping. Such methodologies are well known to those skilled in the art.
Nucleic acid probes derived from the nucleic acid molecules of the present invention may also be used for physical mapping (i.e., placement of sequences on physical maps; see Hoheisel et al, In: Nonmammalian Genomic Analysis: A Practical Guide, Academic press 1996, pp. 319- 346, and references cited therein).
In another embodiment, nucleic acid probes derived from the nucleic acid molecules of the present invention may be used in direct fluorescence in situ hybridization (FISH) mapping (Trask, Trends Genet. 7:149-154 (1991), herein incorporated by reference in its entirety). Although current methods of FISH mapping favor use of large clones (several to several hundred KB; see Laan et al, Genome Res. 5:13-20 (1995), herein incoφorated by reference in its entirety), improvements in sensitivity may allow performance of FISH mapping using shorter probes.
A variety of nucleic acid amplification-based methods of genetic and physical mapping may be carried out using the nucleotide molecules of the present invention. Examples include allele-specific amplification (Kazazian et al., J. Lab. Clin. Med. 77:95-96 (1989), herein incoφorated by reference in its entirety), polymoφhism of PCR-amplified fragments (CAPS; Sheffield et al, Genomics 76:325-332 (1993), herein incoφorated by reference in its entirety), allele-specific ligation (Landegren et al, Science 241:1011-1080 (1988) herein incoφorated by reference in its entirety), nucleotide extension reactions (Sokolov et al., Nucleic Acid Res. 75:3671 (1990) herein incoφorated by reference in its entirety), Radiation Hybrid Mapping (Walter et al, Nat. Genet. 7:22-28 (1997) herein incorporated by reference in its entirety) and Happy Mapping (Dear and Cook, Nucleic Acid Res. 77:6795-6807 (1989) herein incoφorated by reference in its entirety). For these methods, the sequence of a nucleic acid fragment is used to design and produce primer pairs for use in the amplification reaction or in primer extension reactions. The design of such primers is well known to those skilled in the art. In methods employing PCR-based genetic mapping, it may be necessary to identify DNA sequence differences between the parents of the mapping cross in the region corresponding to the nucleotide sequence. This, however, is generally not necessary for mapping methods.
Isolated nucleic acid molecules of the present invention may find use in the identification of loss of function mutant phenotypes of a plant, due to a mutation in one or more endogenous genes encoding polypeptides of the present invention. This can be accomplished either by using targeted gene disruption protocols or by identifying specific mutants for these genes contained in a population of plants carrying mutations in all possible genes (Ballinger and Benzer, Proc. Natl. Acad Sci USA 5(5:9402-9406 (1989); Koes et al, Proc. Natl. Acad. Sci. USA 92: 149-8153 (1995); Bensen et al, Plant Cell 7/75-84 (1995) all of which are incorporated herein by reference in their entirety). The latter approach may be accomplished in two ways. First, short segments of the nucleic acid molecules of the present invention may be used in polymerase chain reaction protocols in conjunction with a mutation tag sequence primer on DNAs prepared from a population of plants in which mutator transposons or some other mutation-causing DNA element has been introduced. The amplification of a specific DNA fragment with these primers indicates the insertion of the mutation tag element in or near the plant gene encoding polypeptide(s) of the present invention. Alternatively, the nucleic acid molecules of the present invention may be used as a hybridization probe against PCR amplification products generated from the mutation population using the mutation tag sequence primer in conjunction with an arbitrary genomic site primer, such as that for a restriction enzyme site-anchored synthetic adapter. Isolated nucleic acid molecules of the present invention can be mutated by a variety of methods well known in the art (Shortle D. et al., Annu. Rev. Genet. 15: 265, 1981; Itakura K. et al., Ann Rev. Biochem 53: 323, 1984; Botstein D. & Shortle D., Science 229: 1193, 1985; Smith M., Ann. Rev. Genet. 19: 423 1985; and Sambrook, et al, In: Molecular Cloning, A Laboratory Manual, 2nd Edition, Cold Spring Harbor Press, Cold Spring Harbor, New York 1989, herein incorporated by reference in its entirety). These mutations can also be introduced by causing point mutation or site directed mutagenesis by using commercially available kits such as QuickChange™ from Stratagene (11011 North Torrey Pines Road, La Jolla, CA 92037).For example site directed mutations can include but are not limited to, truncations, substitutions, additions, terminations, fusions of polypeptides or nucleic acid. With either method, a plant containing a mutation in the endogenous gene encoding polypeptides of the present invention can be identified and obtained. Such plant can also be obtained by in situ site directed mutagenesis. This mutant plant can then be used to determine or confirm the natural function of the polypeptides of the present invention disclosed herein.
Methods for introducing genetic mutations into plant genes are well known. For instance, seeds or other plant material can be treated with a mutagenic chemical substance, according to standard techniques. Such chemical substances include, but are not limited to, the following: diethyl sulfate, ethylene imine, ethyl methanesulfonate and N-nitroso-N-ethylurea. Alternatively, ionizing radiation from sources such as, for example, X-rays or gamma rays can be used. Desired mutants are selected by assaying for increased seed mass, oil content and other properties. "C-terminal region" refers to the region of a peptide, polypeptide, or protein chain from the middle thereof to the end that carries the amino acid having a free carboxyl group.
"N-terminal region" refers to the region of a peptide, polypeptide, or protein chain from the amino acid having a free amino group to the middle of the chain. Antisense Oligonucleotides or Polynucleotides of the Present Invention The present invention, in another aspect, provides an antisense oligonucleotide or polynucleotide encoding an RNA molecule which hybridizes to at least a portion of an RNA transcript of an endogenous gene encoding a polypeptide of the present invention, wherein the RNA molecule hybridizes with the RNA transcript such that expression of the endogenous gene is altered. The present invention, in another aspect, provides DNA construct wherein a promoter that functions in plant is operably linked to an antisense oligonucleotide or polynucleotide encoding an RNA molecule which hybridizes under stringent hybridization conditions to at least a portion of an RNA transcript of an endogenous gene encoding a polypeptide the amino acid sequence of which has at least 50% sequence identity to a member selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10, SEQ ID NO: 12, SEQ ID NO: 14, SEQ ID NO: 16, SEQ ID NO: 18, SEQ ID NO: 20, SEQ ID NO: 22, SEQ ID NO: 24, SEQ ID NO: 26, SEQ ID NO: 28, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 34, SEQ ID NO: 36, SEQ ID NO: 38, SEQ ID NO: 40, SEQ ID NO: 42, SEQ ID NO: 44, SEQ ID NO: 46, SEQ ID NO: 48, SEQ ID NO: 50, SEQ ID NO: 52, SEQ ID NO: 54, SEQ ID NO: 67, SEQ ID NO: 69, and SEQ ID NO: 71. The antisense oligonucleotide or polynucleotide can be full length or preferably has about six to about 100 nucleotides. In one embodiment of this invention, the antisense oligonucleotide or polynucleotide hybridizes under stringent conditions to either at least a corresponding portion of one strand of a nucleotide sequence selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9, SEQ ID NO: 11, SEQ ID NO: 13, SEQ ID NO: 15, SEQ ID NO: 17, SEQ ID NO: 19, SEQ ID NO: 21, SEQ ID NO: 23, SEQ ID NO: 25, SEQ ID NO: 27, SEQ ID NO: 29, SEQ ID NO: 31, SEQ ID NO: 33, SEQ ID NO: 35, SEQ ID NO: 37, SEQ ID NO: 39, SEQ ID NO: 41, SEQ ID NO: 43, SEQ ID NO: 45, SEQ ID NO: 47, SEQ ID NO: 49, SEQ ID NO: 51, SEQ ID NO: 53, SEQ ID NO: 66, SEQ ID NO: 68, SEQ ID NO: 70:, or the RNA transcript transcribed from the a nucleotide sequence selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9, SEQ ID NO: 11, SEQ ID NO: 13, SEQ ID NO: 15, SEQ ID NO: 17, SEQ ID NO: 19, SEQ ID NO: 21, SEQ ID NO: 23, SEQ ID NO: 25, SEQ ID NO: 27, SEQ ID NO: 29, SEQ ID NO: 31, SEQ ID NO: 33, SEQ ID NO: 35, SEQ ID NO: 37, SEQ ID NO: 39, SEQ ID NO: 41, SEQ ID NO: 43, SEQ ID NO: 45, SEQ ID NO: 47, SEQ ID NO: 49, SEQ ID NO: 51, SEQ ID NO: 53 SEQ ID NO: 66, SEQ ID NO: 68, and SEQ ID NO: 70. In another embodiment, the antisense oligonucleotide or polynucleotide hybridizes under stringent conditions to a corresponding portion of the 5' non- coding region or 3' non-translated region of the RNA transcript. In another embodiment, the antisense oligonucleotide or polynucleotide further comprises a sequence encoding a catalytic RNA or riboenzyme. The antisense oligonucleotides or polynucleotides of the present invention may find particular use in antisense technology to suppress endogenous gene expression to control sizes of organs in transgenic plants. To accomplish this, a nucleic acid molecule derived from a nucleotide sequence selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9, SEQ ID NO: 11, SEQ ID NO: 13, SEQ ID NO: 15, SEQ ID NO: 17, SEQ ID NO: 19, SEQ ID NO: 21, SEQ ID NO: 23, SEQ ID NO: 25, SEQ ID NO: 27, SEQ ID NO: 29, SEQ ID NO: 31, SEQ ID NO: 33, SEQ ID NO: 35, SEQ ID NO: 37, SEQ ID NO: 39, SEQ ID NO: 41, SEQ ID NO: 43, SEQ ID NO: 45, SEQ ID NO: 47, SEQ ID NO: 49, SEQ ID NO: 51, SEQ ID NO: 53, SEQ ID NO: 66, SEQ ID NO: 68, and SEQ ID NO: 70 is cloned and operably linked to a promoter such that the antisense strand of RNA will be transcribed. The construct is then transformed into plants and the antisense strand of RNA is produced. In plant cells, it has been suggested that antisense RNA inhibits gene expression by preventing the accumulation of mRNA that encodes the enzyme of interest (see, e.g., Sheehy et al, Proc. Nat. Acad. Sci. USA 55:8805-8809 (1988), and U.S. Patent No. 4,801,340; both of which are herein incorporated by reference in their entireties).
The antisense oligonucleotide or polynucleotide to be introduced generally will be substantially identical to at least a portion of the endogenous gene or genes of the present invention to be repressed. The sequence, however, needs not to be perfectly identical to inhibit or suppress expression of the endogenous gene or genes of the present invention.
For antisense suppression, the introduced sequence also needs not be full length relative to either the primary transcription product or fully processed mRNA. Generally, higher homology can be used to compensate for the use of a shorter sequence. Furthermore, the introduced sequence need not have the same intron or exon pattern, and homology of non-coding segments may he equally effective. Normally, a sequence of between about 6 nucleotides and about full length nucleotides should be used, though a sequence of at least about 500 to about 1700 nucleotides is preferred, a sequence of at least about 200 nucleotides is more preferred, and a sequence of about 6 to about 100 nucleotides is especially preferred.
In another embodiment of this invention, the antisense oligonucleotide or polynucleotide is substantially complementary to at least a corresponding portion of one strand of a nucleotide sequence selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9, SEQ ID NO: 11, SEQ ID NO: 13, SEQ ID NO: 15, SEQ ID NO: 17, SEQ ID NO: 19, SEQ ID NO: 21, SEQ ID NO: 23, SEQ ID NO: 25, SEQ ID NO: 27, SEQ ID NO: 29, SEQ ID NO: 31, SEQ ID NO: 33, SEQ ID NO: 35, SEQ ID NO: 37, SEQ ID NO: 39, SEQ ID NO: 41, SEQ ID NO: 43, SEQ ID NO: 45, SEQ ID NO: 47, SEQ ID NO: 49, SEQ ID NO: 51, SEQ ID NO: 53, SEQ ID NO: 66, SEQ ID NO: 68, SEQ ID NO: 70, or the RNA transcript transcribed from the a nucleotide sequence selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9, SEQ ID NO: 11, SEQ ID NO: 13, SEQ ID NO: 15, SEQ ID NO: 17, SEQ ID NO: 19, SEQ ID NO: 21, SEQ ID NO: 23, SEQ ID NO: 25, SEQ ID NO: 27, SEQ ID NO: 29, SEQ ID NO: 31, SEQ ID NO: 33, SEQ ID NO: 35, SEQ ID NO: 37, SEQ ID NO: 39, SEQ ID NO: 41, SEQ ID NO: 43, SEQ ID NO: 45, SEQ ID NO: 47, SEQ ID NO: 49, SEQ ID NO: 51, SEQ ID NO: 53, SEQ ID NO: 66, SEQ ID NO: 68, SEQ ID NO: 70. In another embodiment, the antisense oligonucleotide or polynucleotide is substantially complementary to a corresponding portion of the 5' non-coding portion or 3' non-coding portion of one strand of a DNA molecule which has substantial sequence identity to a sequence selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9, SEQ ID NO: 11, SEQ ID NO: 13, SEQ ID NO: 15, SEQ ID NO: 17, SEQ ID NO: 19, SEQ ID NO: 21, SEQ ID NO: 23, SEQ ID NO: 25, SEQ ID NO: 27, SEQ ID NO: 29, SEQ ID NO: 31, SEQ ID NO: 33, SEQ ID NO: 35, SEQ ID NO: 37, SEQ ID NO: 39, SEQ ID NO: 41, SEQ ID NO: 43, SEQ ID NO: 45, SEQ ID NO: 47, SEQ ID NO: 49, SEQ ID NO: 51, SEQ ID NO: 53, SEQ ID NO: 66, SEQ ID NO: 68, or SEQ ID NO: 70.
The antisense oligonucleotides or polynucleotides of the present invention may further comprise a nucleotide sequence encoding a catalytic RNA molecule or a ribozyme. It is known to a skilled person in the art that catalytic RNA molecules or ribozymes can also be used to inhibit expression of genes of the present invention. It is possible to design ribozymes that specifically pair with virtually any target RNA and cleave the phosphodiester backbone at a specific location, thereby functionally inactivating the target RNA. In carrying out this cleavage, the ribozyme is not itself altered, and is thus capable of recycling and cleaving other molecules, making it a true enzyme. The inclusion of ribozyme sequences within antisense RNAs confers RNA-cleaving activity upon them, thereby increasing the activity of the recombinant DNA constructs.
A number of classes of ribozymes have been identified. One class of ribozymes is derived from a number of small circular RNAs, which are capable of self-cleavage and replication in plants. The RNAs replicate either alone (viroid RNAs) or with a helper virus (satellite RNAs). Examples include RNAs from avocado sunblotch viroid and the satellite RNAs from tobacco ringspot virus, lucerne transient streak virus, velvet tobacco mottle virus, Solanum nodiflorum mottle virus and subterranean clover mottle virus. The design and use of target RNA- specific ribozymes is described in Haseloff et al. Nature 334:585-591 (1988), herein incorporated by reference in its entirety.
The present invention also provides antibodies that specifically bind to the polypeptides of the present invention and recombinant DNA constructs that comprise nucleic acid molecules of the present invention.
Plant Recombinant DNA Constructs and Transformed Plants The term "transgenic plant" means a plant that contains an exogenous nucleic acid, which can be derived from the same plant species or from a different species. By "exogenous" it is meant that a nucleic acid molecule originates from outside the plant to which the nucleic acid molecule is introduced. An exogenous nucleic acid molecule can have a naturally occurring or non-naturally occurring nucleotide sequence. One skilled in the art understands that an exogenous nucleic acid molecule can be a heterologous nucleic acid molecule derived from a different plant species than the plant into which the nucleic acid molecule is introduced or can be a nucleic acid molecule derived from the same plant species as the plant into which it is introduced.
The term "genome" as it applies to plant cells encompasses not only chromosomal DNA found within the nucleus, but organelle DNA found within subcellular components of the cell. DNAs of the present invention introduced into plant cells can therefore be either chromosomally integrated or organelle-localized. The term "genome" as it applies to bacteria encompasses both the chromosome and plasmids within a bacterial host cell. Encoding DNAs of the present invention introduced into bacterial host cells can therefore be either chromosomally integrated or plasmid-localized.
Exogenous nucleic acid molecules may be transferred into a plant cell by the use of a recombinant DNA construct (or vector) designed for such a puφose. The present invention also provides a plant recombinant DNA construct (or vector) for producing transgenic plants, wherein the plant recombinant DNA construct (or vector) comprises a sense oligonucleotide or polynucleotide or an antisense oligonucleotide or polynucleotide. Methods that are well known to those skilled in the art may be used to prepare the plant recombinant DNA construct (or vector) of the present invention. These methods include in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination. Such techniques are described in Sambrook et al., Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Press, Plainview, N.Y. (1989); and Ausubel et al., Current Protocols in Molecular Biology, John Wiley & Sons, New York, N.Y. (1989).
A plant recombinant DNA construct (or vector) of the present invention contains a sense oligonucleotide or polynucleotide or an antisense oligonucleotide or polynucleotide and operably linked regulatory sequences or control elements. Exemplary regulatory sequences include but are not limited to promoters, translation leader sequences, introns, 3' non-translated sequences. The promoters can be constitutive, inducible, or preferably tissue-specific promoters.
A plant recombinant DNA construct (vector) of the present invention will typically comprise a selectable marker that confers a selectable phenotype on plant cells. Selectable markers may also be used to select for plants or plant cells that contain the exogenous nucleic acid molecules of the present invention. The marker may encode biocide resistance, antibiotic resistance (e.g., kanamycin, G418 bleomycin, hygromycin, etc.), or herbicide resistance (e.g., glyphosate, etc.). Examples of selectable markers include, but are not limited to, a neo gene (Potrykus et al, Mol. Gen. Genet. 7PP:183-188 (1985)) which codes for kanamycin resistance and can be selected for using kanamycin, G418, etc.; a bar gene which codes for bialaphos resistance; a mutant EPSP synthase gene (Hinchee et al, Bio/Technology (5:915-922 (1988)) which encodes glyphosate resistance; a nitrilase gene which confers resistance to bromoxynil (Stalker et al., J. Biol. Chem. 2(53:6310-6314 (1988)); a mutant acetolactate synthase gene (ALS) which confers imidazolinone or sulphonylurea resistance (European Patent Application 154,204 (Sept. 11, 1985)); and a methotrexate resistant DHFR gene (Thillet et al, J. Biol. Chem. 263:12500-12508 (1988)). A plant recombinant DNA construct (vector) of the present invention may also include a screenable marker. Screenable markers may be used to monitor expression. Exemplary screenable markers include a β-glucuronidase or uidA gene (GUS) which encodes an enzyme for which various chromogenic substrates are known (Jefferson, Plant Mol. Biol, Rep. 5:387-405 (1987); Jefferson et al, EMBO J. (5.3901-3907 (1987)); an R-locus gene, which encodes a product that regulates the production of anthocyanin pigments (red color) in plant tissues (Dellaporta et al, Stadler Symposium 77:263-282 (1988)); a β-lactamase gene (Sutcliffe et al, Proc. Natl. Acad. Sci. (U.S.A.) 75:3737-3741 (1978)), a gene which encodes an enzyme for which various chromogenic substrates are known (e.g., PADAC, a chromogenic cephalosporin); a luciferase gene (Ow et ah, Science 234:856-859 (1986)) a xylE gene (Zukowsky et al, Proc. Natl. Acad. Sci. (U.S.A.) 50:1101-1105 (1983)) which encodes a catechol dioxygenase that can convert chromogenic catechols; an α-amylase gene (Ikatu et al, Bio/Technol 5:241-242 (1990)); a tyrosinase gene (Katz et al, J. Gen. Microbiol. 129:2103-2114 (1983)) which encodes an enzyme capable of oxidizing tyrosine to DOPA and dopaquinone which in turn condenses to melanin; an α-galactosidase, which will turn a chromogenic α-galactose substrate.
Included within the terms "selectable or screenable marker genes" are also genes which encode a secretable marker whose secretion can be detected as a means of identifying or selecting for transformed cells. Examples include markers that encode a secretable antigen that can be identified by antibody interaction, or even secretable enzymes which can be detected catalytically. Secretable proteins fall into a number of classes, including small, diffusible proteins detectable, e.g., by ELISA, small active enzymes detectable in extracellular solution (e.g., α-amylase, β-lactamase, phosphinothricin transferase), or proteins which are inserted or trapped in the cell wall (such as proteins which include a leader sequence such as that found in the expression unit of extension or tobacco PR-S). Other possible selectable and/or screenable marker genes will be apparent to those of skill in the art.
In addition to a selectable marker, it may be desirous to use a reporter gene. In some instances a reporter gene may be used with or without a selectable marker. Reporter genes are genes which are typically not present in the recipient organism or tissue and typically encode for proteins resulting in some phenotypic change or enzymatic property. Examples of such genes are provided in K. Wising et al. Ann. Rev. Genetics, 22, 421 (1988), which is incoφorated herein by reference. Preferred reporter genes include the beta-glucuronidase (GUS) of the uidA locus of E. coli, the chloramphenicol acetyl transferase gene from Tn9 of E. coli, the green fluorescent protein from the bioluminescent jellyfish Aequorea victoria, and the luciferase genes from firefly Photinus pyralis. An assay for detecting reporter gene expression may then be performed at a suitable time after said gene has been introduced into recipient cells. A preferred such assay entails the use of the gene encoding beta-glucuronidase (GUS) of the uidA locus of E. coli as described by Jefferson et al., (Biochem. Soc. Trans. 15, 17-19 (1987)) to identify transformed cells. In preparing the recombinant DNA constructs (vectors) of the present invention, the various components of the construct or fragments thereof will normally be inserted into a convenient cloning vector, e.g., a plasmid that is capable of replication in a bacterial host, e.g., E. coli. Numerous cloning vectors exist that have been described in the literature, many of which are commercially available. After each cloning, the cloning vector with the desired insert may be isolated and subjected to further manipulation, such as restriction digestion, insertion of new fragments or nucleotides, ligation, deletion, mutation, resection, etc. so as to tailor the components of the desired sequence. Once the construct has been completed, it may then be transferred to an appropriate vector for further manipulation in accordance with the manner of transformation of the host cell.
A plant recombinant DNA construct (vector) of the present invention may also include a chloroplast transit peptide, in order to target the polypeptide of the present invention to the plastid. The term "plastid" means the class of plant cell organelles that includes amyloplasts, chloroplasts, chromoplasts, elaioplasts, eoplasts, etioplasts, leucoplasts, and proplastids. These organelles are self-replicating, and contain what is commonly referred to as the "chloroplast genome," a circular DNA molecule that ranges in size from about 120 to about 217 kb, depending upon the plant species, and which usually contains an inverted repeat region. Many plastid-localized polypeptides are expressed from nuclear genes as precursors and are targeted to the plastid by a chloroplast transit peptide (CTP), which is removed during the import steps. Examples of such chloroplast polypeptides include the small subunit of ribulose- 1 ,5-biphosphate carboxylase (ssRUBISCO, SSU), 5-enolpyruvateshikimate-3-phosphate synthase (EPSPS), ferredoxin, ferredoxin oxidoreductase, the light-harvesting-complex protein I and protein II, and thioredoxin F. It has been demonstrated that non-plastid polypeptides may be targeted to the chloroplast by use of polypeptide fusions with a CTP and that a CTP sequence is sufficient to target a polypeptide to the plastid. Those skilled in the art will also recognize that various other recombinant DNA constructs can be made that utilize the functionality of a particular plastid transit peptide to import the enzyme into the plant cell plastid depending on the promoter tissue specificity.
The present invention also provides a transgenic plant comprising in its genome an exogenous nucleic acid molecule which comprises: (A) a 5' non-coding sequence which functions in the cell to cause the production of an RNA molecule; which is operably linked to (B) an antisense oligonucleotide or polynucleotide or a sense oligonucleotide or polynucleotide of this invention; which is operably linked to (C) a 3' non-translated sequence that functions in said cell to cause termination of transcription.
Transgenic plants of the present invention preferably have incoφorated into their genome or transformed into their chloroplast or plastid genomes an exogenous nucleic acid molecule (or "transgene") that comprises a sense oligonucleotide or polynucleotide or an antisense oligonucleotide or polynucleotide. Transgenic plants are also meant to comprise progeny (descendant, offspring, etc.) of any generation of such a transgenic plant. A seed of any generation of all such transgenic plants wherein said seed comprises a sense oligonucleotide or polynucleotide or an antisense oligonucleotide or polynucleotide of the present invention is also an important aspect of the invention.
The DNA constructs of the present invention may be introduced into the genome of a desired plant host by a variety of conventional transformation techniques, which are well known to those skilled in the art. Preferred methods of transformation of plant cells or tissues are the Agrobacterium mediated transformation method and the biolistics or particle-gun mediated transformation method. Suitable plant transformation vectors for the purpose of Agrobacterium mediated transformation include those derived from a Ti plasmid of Agrobacterium tumefaciens, as well as those disclosed, e.g., by Herrera-Estrella et al., Nature 303:209 (1983); Bevan, Nucleic Acids Res. 12: 8711-8721 (1984); Klee et al., Bio-Technology 3(7): 637-642 (1985); and EPO publication 120,516. In addition to plant transformation vectors derived from the Ti or root- inducing (Ri) plasmids of Agrobacterium, alternative methods can be used to insert the DNA constructs of this invention into plant cells. Such methods may involve, but are not limited to, for example, the use of liposomes, electroporation, chemicals that increase free DNA uptake, free DNA delivery via microprojectile bombardment, and transformation using viruses or pollen. A plasmid expression vector suitable for the introduction of an antisense oligonucleotide or polynucleotide or a sense oligonucleotide or polynucleotide in monocots using electroporation or particle-gun mediated transformation is composed of the following: a promoter that is inducible or constitutive or tissue-specific; an intron that provides a splice site to facilitate expression of the gene, such as the Hsp70 intron (U.S. Patent Number 5,859,347); and a 3' polyadenylation sequence such as the nopaline synthase 3' sequence (NOS 3'; Fraley et al., Proc. Natl. Acad. Sci. USA 80: 4803-4807(1983)). This expression cassette may be assembled on high copy replicons suitable for the production of large quantities of DNA.
An example of a useful Ti plasmid cassette vector for plant transformation is described in
US Patent No. 6,147,278 herein incoφorated by reference in its entirety, and contains a gene encoding an EPSPS enzyme conferring glyphosate resistance (denominated aroA; CP4), which is an excellent selection marker gene for many plants. The gene is fused to the Arabidopsis EPSPS chloroplast transit peptide (CTP2) and expressed from the FMV promoter as described therein.
When adequate numbers of cells (or protoplasts) containing an antisense oligonucleotide or polynucleotide or a sense oligonucleotide or polynucleotide of the present invention are obtained, the cells (or protoplasts) can be cultured to regenerate into whole plants. Such regeneration techniques rely on manipulation of certain phytohormones in a tissue culture growth medium, typically relying on a biocide and/or herbicide marker which has been introduced together with the desired nucleotide sequences. Choice of methodology for the regeneration step is not critical, with suitable protocols being available for hosts from Leguminosae (alfalfa, soybean, clover, etc.), Umbelliferae (carrot, celery, parsnip), Cruciferae (cabbage, radish, canola/rapeseed, etc.), Cucurbitaceae (melons and cucumber), Gramineae (wheat, barley, rice, maize, etc.), Solanaceae (potato, tobacco, tomato, peppers), various floral crops, such as sunflower, and nut-bearing trees, such as almonds, cashews, walnuts, and pecans. See, for example, Ammirato et al., Handbook of Plant Cell Culture - Crop Species. Macmillan Publ. Co. (1984); Shimamoto et al., Nature 338:274-276 (1989); Fromm, UCLA Symposium on Molecular Strategies for Crop Improvement, April 16-22, 1990. Keystone, CO (1990); Vasil et al., Bio/Technology 8:429-434 (1990); Vasil et al., Bio/Technology 10:667-674 (1992); Hayashimoto, Plant Physiol. 93:857-863 (1990); and Datta et al., Bio-technology 8:736-740 (1990). Plant regeneration from cultured protoplasts is described in Evans et al., Protoplasts Isolation and Culture, Handbook of Plant Cell Culture, pp. 124-176, MacMillilan Publishing Company, New York, 1983; and Binding, Regeneration of Plants, Plant Protoplasts, pp. 21-73, CRC Press, Boca Raton, 1985. Regeneration can also be obtained from plant callus, explants, organs, or parts thereof. Such regeneration techniques are described generally in Klee et al., Ann. Rev. Plant Phys. 38:467-486 (1987). A transgenic plant formed using Agrobacterium transformation methods typically contains a single exogenous gene on one chromosome. Such transgenic plants can be referred to as being heterozygous for the added exogenous gene. More preferred is a transgenic plant that is homozygous for the added exogenous gene; i.e., a transgenic plant that contains two added exogenous genes, one gene at the same locus on each chromosome of a chromosome pair. A homozygous transgenic plant can be obtained by sexually mating (selfing) an independent segregant transgenic plant that contains a single exogenous gene, germinating some of the seed produced and analyzing the resulting plants produced for the exogenous gene of interest.
The development or regeneration of transgenic plants containing the exogenous nucleic acid molecule that encodes a polypeptide of interest is well known in the art. Preferably, the regenerated plants are self-pollinated to provide homozygous transgenic plants, as discussed above. Otherwise, pollen obtained from the regenerated plants is crossed to seed-grown plants of agronomically important lines. Conversely, pollen from plants of these important lines is used to pollinate regenerated plants. A transgenic plant of the present invention containing desired polypeptides of the present invention are cultivated using methods well known to one skilled in the art. Plants that can be made to have larger organ size by practice of the present invention include, but are not limited to, Acαciα, alfalfa, aneth, apple, apricot, artichoke, arugula, asparagus, avocado, banana, barley, beans, beet, blackberry, blueberry, broccoli, brussels sprouts, cabbage, canola, cantaloupe, carrot, cassava, cauliflower, celery, cherry, cilantro, citrus, Clementines, coffee, corn, cotton, cucumber, Douglas fir, eggplant, endive, escarole, eucalyptus, fennel, figs, gourd, grape, grapefruit, honey dew, jicama, kiwifruit, lettuce, leeks, lemon, lime, loblolly pine, mango, melon, mushroom, nut, oat, okra, onion, orange, an ornamental plant, papaya, parsley, pea, peach, peanut, pear, pepper, persimmon, pine, pineapple, plantain, plum, pomegranate, poplar, potato, pumpkin, quince, radiata pine, radicchio, radish, raspberry, rice, rye, sorghum, southern pine, soybean, spinach, squash, strawberry, sugarbeet, sugarcane, sunflower, sweet potato, sweetgum, tangerine, tea, tobacco, tomato, turf, a vine, watermelon, wheat, yams, and zucchini.
The present invention also provides cells of the transgenic plants of the present invention which could be used for regenerating a plant or any organ of plant with present invention.
The present invention also further provides a method for altering a specific organ or organs in a plant to generate smaller plant or plant organ, the method comprising the steps of: a) introducing into the genome of the plant an exogenous nucleic acid molecule comprising in the 5' to 3' direction i) a promoter that functions in the cells of said plant, said promoter operably linked to; ii) an antisense oligonucleotide or polynucleotide or a sense oligonucleotide or polynucleotide of the present invention, said antisense oligonucleotide or polynucleotide operably linked to; iii) a 3' non-translated nucleotide sequence that functions in said cells of said plant to cause transcriptional termination; b) obtaining transformed plant cells containing the exogenous nucleic acid molecule of step (a); and c) regenerating from said transformed plant cells a transformed plant in which the expression of an endogenous gene of the present invention is suppressed or inhibited.
The following examples are provided to better elucidate the practice of the present invention and should not be inteφreted in any was to limit the scope of the present invention. Those skilled in the art will recognize that various modifications, truncations, etc., can be made to the methods and genes described herein while not departing from the spirit and scope of the present invention. In the following examples references are made to proprietary databases and proprietary libraries, e.g., of DNA clones, available to the inventors from Monsanto Company.
EXAMPLES EXAMPLE 1
Stock Plant Material and Growth Conditions
Arabidopsis thaliana var Columbia seeds were obtained from Lehle seeds (LEHLE SEEDS 1102 South Industrial Blvd., Suite D, Round Rock TX 78681 USA). For growing seeds into plants, 2.5 inch pots are prepared with soil covered with bridal veil or a mesh screen, making sure that the soil is not packed too tightly and the mesh is in contact with the soil surface (this ensures that the germinating seedlings will be able to get through the mesh. Seeds are sown and covered with a germination dome. Seeds are vernalized for 3-4 days. Plants are grown under conditions of 16 hours light / 8 hours dark at 20-22° C, 70% humidity. They are watered twice weekly, and fertilize from below with 1/2 X (half of the strength recommended by the manufacturer) Peters 20-20-20 fertilizer (from Hummert International, Earth City, MO). Micronutrients are added (Hummert's Dyna-grain Soluble Trace Elements) (in full strength recommended by the manufacturer) every other week. After about 1-2 weeks, dome is removed and pots are thinned to one or two plants per pot. Clip the primary bolt, when it develops, to encourage more secondary bolt formation. In 5-7 days the plants will be ready for infiltration.
Isolation of Gene and its modification: Senescing leaves were removed from plants grown as indicated in above Example 1. Senescing leaves were flash frozen in liquid nitrogen until ready for isolation of RNA. RNA was prepared from senescent Arabidopsis leaves by the Trizol method (Gibco BRL, Life Technologies, Gaithersburg, Maryland U.S. A.) essentially as recommended by the manufacturer. SAG13 cDNA was isolated by reverse transcription from the above senescent leaf RNA using Superscript II kit (Gibco BRL, Life Technologies, Gaithersburg, Maryland U.S. A.) according to the manufacturer's directions. ..To isolate DNA molecules of the present invention, two gene specific primers, MF16 5'SAG13 and MF17 3'SAG13, were designed based on the SAG 13 sequence information (AF192276) from Genbank and custom synthesized by Gibco BRL, Life Technologies, Gaithersburg, Maryland U.S. A. The sequence of MF16 5'SAG13 is ATA TTT AAC AAG CCA TGG CAA AGG A, and MF17 3'SAG13 is ATA TGT GTT TGA ATT CAT AGT CTT GAA identified as SEQ ID NO: 55 and SEQ ID NO: 56 respectively in the sequence list, which annealed on SAG 13 gene to introduce Nco 1 site at 5 'end and Eco RI site at 3 'end of the gene. PCR was then performed to amplify the S.4G-73 cDNA using the above prepared cDNA as the template, and MF16 5'SAG13 and MF17 3'SAG13 as the primers. The thermal cycling conditions were as follows: 94°C, 40 second, followed 30 cycles of 94°C, 25 seconds; 55°C, 30 seconds and 68°C, 2 minutes 30 seconds ( All reagents and equipment for PCR can be procured from Applied Biosystems, 850 Lincoln Center Drive, Foster City, CA 94404, USA). The amplified cDNA was purified by gel-electrophoresis to obtain the gene identified as SEQ ID NO: 1.
All other sequences shown in table from Example 8 are isolated from different plant species by designing appropriate PCR primers based on the sequence information provided in table of Example 8. For isolating these sequences, total RNA is isolated from appropriate crop and other desired plant species by pooling tissues of different developmental stages of all vegetative and reproductive organs. Sequences can be cloned out from total RNA by methodology shown in above paragraph. In order to isolate genes of the invention from microorganisms DNA is isolated from the desired microorganism. Isolation of DNA from microorganism is well known in the art (Sambrook, et al, In: Molecular Cloning, A Laboratory Manual, 2nd Edition, Cold Spring Harbor Press, Cold Spring Harbor, New York 1989, herein incoφorated by reference in its entirety). This DNA along with oligonucleotide PCR primers can be used in a polymerase chain reaction by any one skilled in the art to isolate genes of the invention.
When the amplified product SEQ ID NO: 1 was sequenced, it was discovered to have an extra "T" between position 622-628, as compared to SEQ ID NO: 3. Addition of an extra "T" in the amplified product caused the generation of termination codon at position 211 of SEQ ID NO: 4 and a change of amino acid at position 210 from aspartic acid (D) to arginine (R). It is known in the art that polymerase chain reaction can cause point mutations (Cline J, Braman JC, Hogrefe HH; Nucleic Acids Res .; 24(18):3546-51, 1996) as shown between SEQ ID No: 1 and SEQ ID NO: 3. However such a mutation can be created within an isolated nucleic acid molecule at a desired position by a number of methods know in the art (Shortle D. et al., Annu. Rev. Genet. 15: 265, 1981; Itakura K. et al., Ann Rev. Biochem 53: 323, 1984; Botstein D. & Shortle D., Science 229: 1193, 1985; Smith M., Ann. Rev. Genet. 19: 423 1985; and Sambrook, et al, In: Molecular Cloning, A Laboratory Manual, 2nd Edition, Cold Spring Harbor Press, Cold Spring Harbor, New York 1989, herein incorporated by reference in its entirety). These mutations can also be introduced by causing a point mutation or site directed mutagenesis in nucleotide SEQ ID NO: 3 by using commercially available kits such as QuickChange™ Site Directed Mutagenesis Kit from Stratagene (11011 North Torrey Pines Road, La Jolla, CA 92037) to result in a peptide with about 90% of the N-terminal of SEQ ID NO: 4 by altering a nucleotide sequence to encode a stop codon at amino acid position 241 + 3, or to result in a peptide with about 80% of the N- terminal of SEQ ID NO: 4 by altering a nucleotide sequence to encode a stop codon at amino acid position 214 + 3, or to result in about 70% of the N-terminal of SEQ ID NO: 4 by altering a nucleotide sequence to encode a stop codon at amino acid position 188 ± 3, or to result in about 60%) of the N-terminal of SEQ ID NO: 4 by altering a nucleotide sequence to encode a stop codon at amino acid position 161 ± 3, or to result in about 50% of the N-terminal of SEQ ID NO: 4 by altering a nucleotide sequence to encode a stop codon at amino acid position 134 ± 3 of the reading frame. These mutations can also be introduced in situ by causing point mutation or site directed mutagenesis in nucleotide SEQ ID NO: 3 by using in situ mutagenesis technology provided by Valigen Inc. (Newtown PA. USA) (US Patent Number 6,211,351; US Patent Number 6,271,360, WO 01/24615 Al, and WO 01/25460 A2, herein incorporated by reference in its entirety) to result in a peptide with about 90% of the N-terminal of SEQ ID NO: 4 by altering a nucleotide sequence to encode a stop codon at amino acid position 241 ± 3, or to result in a peptide with about 80% of the N-terminal of SEQ ID NO: 4 by altering a nucleotide sequence to encode a stop codon at amino acid position 214 + 3, or to result in about 70% of the N-terminal of SEQ ID NO: 4 by altering a nucleotide sequence to encode a stop codon at amino acid position 188 + 3, or to result in about 60% of the N-terminal of SEQ ID NO: 4 by altering a nucleotide sequence to encode a stop codon at amino acid position 161 + 3, or to result in about 50% of the N-terminal of SEQ ID NO: 4 by altering a nucleotide sequence to encode a stop codon at amino acid position 134 + 3 of the reading frame. Additional mutations can be introduced to provide a translational product that is larger or smaller than SEQ ID NO: 1. It can be determined that these modified polynucleotide sequences will provide a desired phenotype without undue experimentation EXAMPLE 2
Genetic elements of cloning vector for expressing protein in plants (Figure 1 and 4) The DNA constructs used are double border plant transformation constructs that contain
DNA segments that provide replication function and antibiotic selection in bacterial cells, for example, an E. coli origin of replication such as or/322, a broad host range origin of replication such as orN or oriRi, and a coding region for a selectable marker such as Spc/Str that encodes for Tn7 aminoglycoside adenyltransferase (aadA) conferring resistance to spectinomycin or streptomycin, or a gentamicin (Gm, Gent) selectable marker. For plant transformation, the host bacterial strain is Agrobacterium tumefaciens ABI or LBA4404.
The genetic elements of the DNA constructs are assembled to have in operable linkage a promoter that functions in plants. Additionally an antibiotic or herbicide marker cassette, an epitope tag ( For example Flag® peptide catalog number F-3290, SIGMA, P.O. Box 14508 St. Louis, MO 63178 USA) at the 3' termination region of gene of interest is included in the DNA construct. The multiple cloning site in this DNA construct encodes Bglll, Ncol, EcoRI, Sail, and Xhol. The epitope tag region was encoded with Sail and Xhol restriction sites for optional removal of the epitope tag. The Ncol site encodes a Kozak sequence for efficient translation of the protein products. DNA constructs used in the method of the current invention comprise any promoter known to function to cause the transcription in plant cells and any antibiotic tolerance encoding polynucleotide sequence known to confer antibiotic tolerance to plant cells. The antibiotic tolerance polynucleotide sequences include, but are not limited to polynucleotide sequences encoding for proteins involved in antibiotic tolerance to kanamycin, neomycin, hygromycin, and other antibiotics known in the art. Antibiotic tolerance gene in such a vector can be replaced by herbicide tolerance encoding for 5-enolpyruvylshikimate-3 -phosphate synthase (EPSPS, described in U.S. Patent Numbers 5,627,061, and 5,633,435, herein incoφorated by reference in its entirety; Padgette et al. (1996) Herbicide Resistant Crops, Lewis Publishers, 53-85, and in Penaloza-Vazquez, et al. (1995) Plant Cell Reports 14:482-487) and aroA (U.S. Patent Number 5,094,945) for glyphosate tolerance, bromoxynil nitrilase (Bxn) for Bromoxynil tolerance (U.S. Patent Number 4,810,648), phytoene desaturase (crtl (Misawa et al, (1993) Plant Journal 4:833-840, and (1994) Plant Jour 6:481-489) for tolerance to norflurazon, acetohydroxy acid synthase (AHAS, Sathasiivan et al. (1990) Nucl. Acids Res. 18:2188-2193) and the bar gene for tolerance to glufosinate (DeBlock, et al. (1987) E R< J. 6:2513-2519. Herbicides for which transgenic plant tolerance has been demonstrated and the method of the present invention can be applied include, but are not limited to: glyphosate, glufosinate, sulfonylureas, imidazolinones, bromoxynil, delapon, cyclohezanedione, protopoφhyrionogen oxidase inhibitors, and isoxaslutole herbicides.
Genetic elements of transgene DNA constructs used for plant transformation and expression of transgenes in plants include, but are not limited to: the P-Ε35S promoter (U.S. Patent No.5,539,142, 5,196,525, 5,322,938 and 5,164,316 herein incorporated by reference in its entirety). P-E35S promoter can be replaced by P-CaMV.35S promoter (U.S. Patent No. 5,858,742, herein incoφorated by reference in its entirety), or by enhanced P-CaMV.35S from Cauliflower mosaic virus containing a duplication of the -90-300 region as described in U. S. Patent No. 5,424,200, herein incorporated by reference in its entirety; or the Figwort mosaic virus promoter, P-FMV, as described in U. S. Patent No. 5,378,619, herein incoφorated by reference in its entirety; or the P-AtEFla (P-AtEFl or EFla) a promoter region from the Arabidopsis thaliana elongation factor gene la; the GboxlO and Gboxl lmotif (Fumiharu et al., (1999) Plant J. 18:443-448); or the DC3 promoter region from carrot (Seffens et al., Develop. Genet. 11:65-76); or the TP12 promoter (GenBank accession no. U68483); DNA molecules encoding plastid transit peptides, for example, the Arabidopsis EPSPS chloroplast transit peptide, At.CTP2 as described in U. S. Patent No. 5,633,435, herein incoφorated by reference in its entirety. The method of the present invention enables one of skill in the art of plant molecular biology to design and assemble plant expression cassettes that contain promoters of known and unknown function. The genetic elements of the DNA construct further comprise 5' leader polynucleotides for example, the Hsp70 non-translated leader sequence from Petunia hybrida as described in U. S. Patent No. 5,362,865, herein incoφorated by reference in its entirety. The genetic elements further comprise herbicide tolerance genes that include, but are not limited to, for example, the aroA:CP4 coding region for EPSPS glyphosate resistant enzyme isolated from Agrobacterium tumefaciens (AGRTU) strain CP4 as described in U. S. Patent No. 5,633,435, herein incorporated by reference in its entirety. The genetic elements of the DNA construct further comprise 3' termination regions that include, but are not limited to, the E9 3' termination region of the pea RbcS gene that functions as a polyadenylation signal; the nos is the 3' end of the nopaline synthase gene that functions as a polyadenylation signal. The genetic elements of the DNA construct further comprise the right border (RB) and left borders (LB) of the Ti plasmid of Agrobacterium tumefaciens octopine and nopaline strains. EXAMPLE 3
Cloning of isolated DNA Molecule: Amplified and purified product SEQ ID NO: 1 from Example 1 was digested by Ncol and EcoRl restriction enzymes (BRL/Life Technologies, Inc., Gainthersburg, MD). The digested product was purified again by gel electrophoresis before ligating to binary vector pMON23435 that had been linearized by Nco 1 and Eco RI and T4 DNA ligase (BRL/Life Technologies, Inc., Gaithersburg, MD). The ligation reaction was performed according to the manufacturer's instructions. The resulting plasmid was confirmed by restriction mapping and sequencing. After ligation of the Nco-EcoRl fragment of SEQ ID NO: 1 into vector pMON23435, the new construct was referred to as construct pMON57521. Construct pMON57521 was transformed in Arabidopsis plants by Agrobacterium mediated transformation procedure.
Amplified and purified product SEQ ID NO: 1 can also be cloned in antisense orientation in appropriate cloning sites of vector pMON23435 to express the opposite strand of SEQ ID NO:l by one skilled in the art. EXAMPLE 4 Cloning of DNA molecules of the present invention: This example illustrates how all other DNA molecules as shown in table from example 9 are isolated from different plant species by designing appropriate PCR primers based on the DNA sequence information provided in the table of Example 8. For isolating these DNA molecules one skilled in the art will isolate total RNA from a crop or other desired plant species by pooling tissues of different developmental stages of all vegetative and reproductive organs. DNA molecules are cloned out from total RNA by methodology shown in Example 2. In order to isolate genes of the invention from microorganisms one will have to isolate DNA from desired microorganism. Isolation of DNA from microorganism is well known in the art (Sambrook, et al, In: Molecular Cloning, A Laboratory Manual, 2nd Edition, Cold Spring Harbor Press, Cold Spring Harbor, New York 1989, herein incoφorated by reference in its entirety). This DNA along with oligonucleotide PCR primers can be used in a polymerase chain reaction by any one skilled in the art to isolate genes of the invention.
Design of primers and reaction conditions are determined as described in the art. (PCR Strategies, Edited by Michael A. Innis; David H. Gelfand; John J. Sninsky; Academic Press 1995 and PCR Protocols, A Guide to Method and Applications, Edited by Michael A. Innis; David H. Gelfand; John Sninsky; and Thomas J. White, Academic Press 1990, herein incoφorated by reference in its entirety). All reagents for isolating sequences of the invention can be procured from Gibco BRL, Life Technologies, Gaithersburg, Maryland U.S.A. Isolated DNA molecule sequences can be cloned at appropriate cloning sites in sense or antisense orientation of a plant expression vector shown in Example 3 or a similar vector capable of ectopically expressing the gene of interest of the present invention in sense or antisense orientation as known in the art. EXAMPLE 5
This example illustrates how Agrobacterium cells are transformed and how transformed cells are cultured. Transformation ; 1. Electroporate 2 μl of DNA construct into 20 μl of ABI competent cells;
2. Pipette transformed cells directly onto LB plates containing Spectinomycin (75 μg/ml), Kanamycin (50μg/ml), Chloramphenicol (25μg/ml). Add 50 μl of SOC media to plate and spread;
3. Incubate plated transformation at 28°C for 2 days (or can grow over weekend), ABI Cell Culture: 1. Pick 3 colonies per ABI plate and grow each in 4ml LB media containing Spectinomycin (75μg/ml), Kanamycin (50μg/ml), Chloramphenicol (25μg/ml);
2. Incubate 4 ml cultures of at 28°C, shaking, for 2 days. (Culture tubes should be at an angle). Glycerol Stocks. & DNA Preps; 1. Make three 1 ml ABI glycerol stocks per 4 ml culture, using 500μl of culture and 500μl of 40% glycerol. Freeze and store at -80°C. 2. Miniprep remaining culture (about 2.5ml), using a Qiagen miniprep kit and protocol (Qiagen Genomics, Inc., Seattle, WA), ensuring add PB buffer wash step and EB buffer (lOmM Tris- Cl, pH 8.5) to 70°C before eluting DNA from column. The resulting volume per miniprep sample should be 50μl.
Digest Confirmation:
1. Using the Pollux and construct maps, choose two digests to perform: one to verify insert integrity, one to verify vector integrity (will need to refer to plasmid maps);
2. Digest 17μl miniprep DNA per digest, resulting in a final digest volume of 20μl; 3. Run 20μl of each digest on 1% agarose gel vs. 1Kb DNA ladder; and
4. For 2 of 3 confirmed clones, streak LB plates containing Spectinomycin (75 μg/ml), Kanamycin (50μg/ml), Chloramphenicol (25μg/ml) from ABI glycerol stocks and allow to grow at 28°C for 2 days (or can grow over weekend).
All other reagents used in example 4 can be procured from Sigma Chemical Company. Saint Louis, MO, USA EXAMPLE 6
This example demonstrates how to transform Arabidopsis plants with gene constructs of present invention Arabidopsis plants may be transformed by any one of many available methods. For example, Arabidopsis plants may be transformed using in planta transformation method by vacuum infiltration (see, Bechtold et al, In planta Agrobacterium mediated gene transfer by infiltration of adult Arabidopsis thaliana plants. CR Acad. Sci. Paris Sciences de la vie/life sciences 316: 1194-1199 (1993), herein incoφorated by reference in its entirety). Plants can be grown as described in Example 1. Asrobacterium Preparation (Small scale and Large scale cultures'): Agrobacterium strain ABI is streaked onto an LB plate containing Spectinomycin 100 mg/L, Streptomycin 100 mg/L, Chloramphenicol 25mg/L, and Kanamycin 50mg/L (denoted SSCK). Two days prior to infiltration, a loop of Agrobacterium is placed into a tube containing 10 mis LB/SSCK and put on a shaker in the dark at 28°C to grow overnight. The following day, the Agrobacterium is diluted 1:50 in 400 mis of bacterial grown medium such as SSCK and put on a shaker at 28°C to grow for 16-20 hours. Infiltration
Harvest the Agrobacterium cells by pouring into a 500 ml centrifuge bottle and spinning at 3500 rpm for 20-25 minutes. Pour off the supernatant. Dry the pellet and then resuspend in 25 ml Infiltration Medium (MS Basal Salts 0.5%, Gambόrg's B-5 Vitamins 1%, Sucrose 5%, MES 0.5 g/L, pH 5.7) with 0.44 nM benzylaminopurine (BAP) (10 μl of a 1.0 mg/L stock in DMSO per liter) and 0.02% Vac-In-Stuff (Silwet L-77) from Lehle Seeds (Round Rock, TX). The BAP and Silwet L-77 are added fresh the day of infiltration. Add 200 μl of Silwet L-77, and 20 μl of BAP (0.5 mg/L stock). Using Infiltration Medium as your blank, take the OD6oo of a 1 :10 dilution of the Agrobacterium suspensions. Calculate the volume needed for 400 ml of Agrobacterium suspension/infiltration medium, OD600 = 0.6, for the vacuum infiltration. Equation: (final volume') * (final OD600) = Volume needed for final OD600 of 0.6 OD600 Place resuspended culture in a Rubbermaid container inside a vacuum dessicator. Invert pots containing plants to be infiltrated into the solution so that the entire plant is covered, including the rosette, but not too much of the soil is submerged. Soak the plants with water for at least 30 min. prior to infiltration. (This keeps the soil from soaking up the Agrobacterium suspension).
Draw a vacuum of ~ 23-27 in. Hg for 10 min. Quickly release the vacuum. Briefly drain the pots, place them on their sides in a diaper-lined tray, cover the tray with a dome to maintain humidity, and return to growth chamber. The following day, uncover the pots, set them upright, and remove the diaper. Do not water plants for ~ 5 days. After the 5 days are up, allow the plants to be watered and to continue to grow under the same conditions as before. (The leaves that were infiltrated may degenerate but the plant should survive until it is finished flowering). Harvesting and Sterilizing Seed Cone the plants, individually, by using the Lehle Aracons (Lehle Seeds, Round Rock, TX) approximately 2 weeks after infiltration. After all of the seed is matured and has set (~ 4 weeks post-infitration), remove the plants from water to dry down the seeds. Approximately 2 weeks later harvest the seeds by cutting the branches below the cone. Clean the seed by using a sieve to catch the silique and branch material and allow the seed to go through. Place the seed in an envelope or in 15ml conical tubes.
Transfer desired amount of seeds to 15ml conical tubes prior to sterilization. Loosen the lid to the conicals and place them on their side in a vacuum dessicator with a beaker containing 400 ml of bleach Clorox (Clorox Company, Oakland, CA) and 4 ml of Hydrochloric Acid. (Add the HCl to the Clorox in a fume hood). Pull a vacuum just to seal the dessicator, and close the suction (i.e. so that the dessicator is still under a vacuum but the vacuum is not still being directly pulled) for ~ 16 hrs. After sterilization, release the vacuum and place tubes containing seed in a sterile hood (keep caps loose so gas can still be released).
Plate ("sprinkle") the seed on selection plates containing MS Basal Salts 4.3 g/L, Gamborg'a B-5 (500 X) 2.0 g/L, Sucrose 10 g/L, MES 0.5 g/L, and 8 g/L Phytagar (Life Technologies, Inc., Rockville, MD) with Carbenicillin 250mg/L, Cefotaxime 100 mg/L. Selection levels will either be kanamycin 60 mg/L, Glyphosate 60μM, or Bialaphos lOmg/L.
A very small amount of seed can be first plated out to check for contamination. If there is contamination, re-sterilized seeds for ~ 4 more hours and check for contamination again. The second sterilization is usually not necessary, but sometimes the seed harbors a fungal contaminant and repeat sterilizations are needed. (The sterilization duration generally is shorter than 16 hours because of significantly decreased germination rates starting at 24 hr. sterilization duration). Seal plates with parafilm and place in a cold room to vernalize for ~ 2-4 days. After seeds are vernalized, place in percival with cool white bulbs. Transfer to Soil
After 5-10 days at -26 °C and a 16/8 light-dark cycle, the transformants will be visible as green plants. After another 1-2 weeks, plants will have at least one set of true leaves. Transfer plants to soil, cover with a germination dome, and move to a growth chamber with normal Arabidopsis growth conditions. Keep covered until new growth is apparent (usually 5-7 days). EXAMPLE 7
Following subsections of Example 7 are used for describing phenotypic changes after transformation and growth of Arabidopsis plants as described in Example 6. Three different events obtained from transformation of pMON57521 (Figure 2) into Arabidopsis thaliana ecotype Columbia were selected and grown side by side with wild type plants. Growth conditions were 16 hr light, 8 hr night, 21 degrees Centigrade and 70% relative humidity. Plants were observed at all growth stages and photographed using an Olympus camera C-2500 L (Olympus America Inc., 2 Corporate Center Drive, Melville, NY 11747) as described by the manufacturer. Microscopic images of plant organ were acquired after proper dissection on a stereoscopic microscope Nikon SMZ 1500 (NIKON, 1300 Walt Whitman Road, Melville, NY 11747) equipped with MagnaFire Digital imaging system. Image quantitation was done as per manufacturer's instructions by using MagnaFire Digital imaging system's software s Pro™ or Lucis™ sof wares (Optronics, 175 Cremona Drive, Goleta, CA 93117). The three events showed high phenotypic consistency in preliminary experiments and hence only two events were chosen for further downstream work.
Plants transformed with Construct pMON57521 were shown to exhibit 2 to 3 times larger flower and floral organs such as stamen and pistils when compared to the wild type, non transformed plants of the same species. Transgenic plants were also observed to have at least 2 times more lateral roots and 2 to 3 times thicker stem size when compared to the lateral roots and stems of wild type, non transformed plants of the same species. Approximately 2 fold increased trichome numbers and distribution were also observed. Over expression of the polypeptide encoded by the sequence in the pMON57521 construct of the present invention in the transformed plants, yielded plants that exhibited a 100 % increase in individual seed size and weight. Approximately 20 % increase in seed yield/plant was also observed for plants transformed with pMON57521 when compared to seed yield/plant of wild type, non transformed plants of the same species. The lines were followed up to 6 generations after transformation and appeared to be highly consistent in maintaining the observed phenotypic changes.
(A) Seed Size: Seeds from transformed plants exhibit increased seed size that was approximately twice as large as the seeds from wild type plants as shown in following table. Image sizes were determined by the Pro or Lucis software ( Optronics, 175 Cremona Drive, Goleta, CA 93117) based on the image pixel value of wild type and transformed plants' seed under the same resolution. Plant lines 8752-1, 8752-2, 8752-6, and 8752-7 in Table 1 correspond to different transgenic plant lines produced by transforming wild Arabidopsis plants with pMON57521.
Table 1: Seed Size
Figure imgf000055_0001
(B) Seed Weight: The seed from plants containing pMON57521 lines was also found to be heavier than WT seed. Seed weight was measured from 2 lines produced from event 8752 (8752-1 and 8752-6) and 2 lines from event 8748 (8748-2 and 8748-7) and compared with wild type. Shown below is a representative seed weight analysis. For each line, 3 replications were measured (i.e. 50 seeds were counted three times and measured each time). This analysis was repeated with different seed counts (eg. 100 seed or 150 seed) and was found to be highly reproducible. For example, weight per 100 seed is similarly high ~ 0.0025 gm. for wild type and 0.0046 gm. for seeds from plants transformed with sequence of the present invention. Data was also highly reproducible within a given line and across lines. The average weight of a single seed from a line transformed with sequence of the present invention was extrapolated to be -0.048 mg as opposed to 0.026 mg for a WT seed. Plant lines 8752-1, 8752-6, 8748-2, and 8748- 7 in Table 2 correspond to different transgenic plant lines produced by transforming wild Arabidopsis plants with pMON57521. Table 2: Seed Weight
Figure imgf000056_0001
(C) Seed Number: Lines over expressing sequence of the present invention were found to have fewer seed per silique as compared to wild type plants. The average number of seed /silique of equivalent maturity was 34 for transgenic plants of the present invention as compared to 52 for wild type plants. The analysis was repeated three times with highly reproducible results within and between events.
Table 3: Seed Number
Figure imgf000057_0001
(D) Seed Yield: There was an increase of > 20% seed yield in transgenic plants of the present invention. Seed yield when measured in terms of weight of total seeds per plant.
Table 4: Seed Yield
Figure imgf000057_0002
Figure imgf000058_0001
EXAMPLE 8
This example describes changes in the branching pattern after transformation and growth of Arabidopsis plants as described in Example 6. Arabidopsis plants over-expressing polypeptide molecules corresponding to SEQ ID NO:
2 appear to have more branches than wild type plants. This results in a net increase in silique number. Although each transgenic silique appears to have fewer seed than an equivalent wild type silique, the increase in branching appears to compensate resulting in a net increase in overall seed yield. The branching pattern of Arabidopsis over-expressing lines was compared to wild type plants and the transgenic plants were shown to have more branches than WT plants. All measurements were taken at growth stage 6.5 which corresponds to an Arabidopsis growth stage where the plant is still actively growing (mid-flowering). By growth stage 6.9 the plants have started senescing and flower production has stopped with < 5 open flowers. Thus the growth stage 6.5 gives a fairly accurate estimate of the growth rate of the plant prior to senescence. An average of the silique number at growth stage 6.5 was estimated for both WT and transgenics and total silique number is shown in the following table. This indicates again that silique number was at least 28% greater in lines over-expressing the polypeptide molecules corresponding SEQ ID NO: 2 (transformed lines) as compared to wild type lines at the same growth stage. As the plant proceeds to senescence, the relative increase appears to stay consistent and together with the increase in seed weight in the transgenic plant, contributes to the overall increase in seed yield per plant. Growth stage estimation was based on "Growth stage- based phenotypic analysis of Arabidopsis: A model for high throughput functional genomics in plants. Plant Cell. 13(7):1499, 2001"
Table 5 Number of Silique. WT is wild type plant and Transformed line is transgenic plant line expressing polypeptide molecules corresponding to SEQ ID. NO: 2.
Figure imgf000059_0001
EXAMPLE 9
This example describes increase of seeds in transgenic soybean plants expressing polypeptide molecule corresponding to SEQ ID NO: 2. Soybean plants were transformed with pMON73955 (Figure 7) to constitutively express
SEQ ID NO: 2. Soybean transformation is performed essentially as described in WO 00/42207, herein incoφorated by reference in its entirety. RI seed from 10 events out of 44 events were advanced in Puerto Rico (PR) based on the gene copy number in plants. Soybean transformation was performed essentially as described in WO 00/42207, herein incoφorated by reference in its entirety. Preliminary data indicates that 1 of the 10 events in PR showed a phenotype similar to that seen for the Arabidopsis over-expression of SEQ ID NO: 2 (pMON 57521 Figure 2) i.e. more pods and more branches as well as short, fat pods with fewer seed. More two-seeded pods were observed and the seed was larger than seed from wild type and negative plants (but seed size was not doubled as seen in Arabidopsis). In addition the positives (transgenic line expressing genes of interest) from this event "A" produced more seed than the negatives (transgenic line NOT expressing genes of interest) as shown in Table 7. Table 6 shows the seed weight of individual plants (events) from the RI generation and seed weight in the R2 generation. R2 seed weight in this table is an average of seed weight of all lines from the original RI event. All numbers are expressed as a percentage of wild-type (WT). Detailed R2 seed weight and seed number data from single lines (positives versus negatives) of the best performing event "A" is shown in Table 7. In general a good correlation is observed between positives and negatives for seed size and seed yield.
Table 6: RI and R2 seed weights of 10 independent soybean transgenic events expressing SEQ ID NO: 2. Values are expressed as a percentage of wild-type where WT = 100%)
Figure imgf000060_0001
Table 7: R2 seed weights of individual lines from the large-seeded transgenic event "A expressing SEQ ID NO: 2.
Figure imgf000060_0002
lA-1 NEG }75 |14 |18.67 |
EXAMPLE 10
This example describes over expression of the protein in bacterial cells for purification of the protein so as to screen for the activity of polypeptide molecules. (A) Cloning of the nucleotide molecules of the present invention for expression of corresponding peptides. pMON57521 (Figure 2), pMON73963 (Figure 3), or Nostoc punctiforme (Nostoc) genomic DNA is used as a DNA template source for polymerase chain reaction amplification (PCR) of DNA for cloning into bacterial cells so as to express the Arabidopsis thaliana protein corresponding to SEQ ID NO: 2, SEQ ID NO: 4, or Nostoc protein corresponding to SEQ ID NO: 50. Design of primers and PCR reaction conditions are determined as described in the article PCR Strategies, Edited by Michael A. Innis; David H. Gelfand; & Johm J. Sninsky; Academic Press 1995 and PCR Protocols, A Guide to Method and Applications, Edited by Michael A. Innis; David H. Gelfand; Johm J. Sninsky; & Thomas J. White Academic Press 1990, herein incoφorated by reference in its entirety. All reagents for performing the PCR reaction can be procured from Gibco BRL, Life Technologies, Gaithersburg, Maryland U.S. A. The thermocycler needed for performing PCR reaction was procured from Applied Biosystems (Perkin-Elmer Corp., Applied Biosystems Div., Foster City, CA). The polynucleotide sequence of the amplicon produced from Arabidopsis is shown in SEQ ID NO:l and SEQ ID NO: 3. PCR primer pairs SEQ ID NO: 57 and SEQ ID NO: 58 were used for performing the reaction to obtain the Arabidopsis amplicon. Arabidopsis amplicons were subcloned into a pET-28b vector (E coli expression vector, Novagen, Madison, WI, USA) and were sequenced by using sequencing primers SEQ ID NO: 59 and SEQ ID NO: 60 to confirm the sequence of the cloned polynucleotide molecule. Similarly Nostoc amplicons were generated by using Nostoc genomic DNA as template with primers pairs SEQ ID NO: 61 and SEQ ID NO: 63 or SEQ ID NO: 62 and SEQ ID NO: 63. Nostoc amplicons were also subcloned in to the pET-28b vector and were sequenced by using sequencing primers SEQ ID NO: 64 and SEQ ID NO: 65 to confirm the polynucleotide sequence of the cloned polynucleotide molecule. Primer Sequence for amplification and sequencing of molecules of invention from Arabidopsis and Nostoc are shown in Figure 10. These constructs contained a candidate from Arabidopsis SEQ ID NO: 3 and the closest Nostoc homologue SEQ ID NO: 49, both with and without an N-terminal His-tag. An N-terminal His-tag was chosen based on the crystal structures (Nakajima et al. PNAS 95, 4876, 1998) of the related proteins (tropinone reductases) which suggested that the N terminus would not interfere with the dimerization domain. The resultant vectors pMON 63132 (Figure 4, Nostoc Histag), pMON 63133 (Figure 5, A. thaliana), pMON 63134 (Figure 6, A. thaliana Histag) , and pMON 63135 Figure 7 Nostoc) were used in the following examples for expression of the protein molecules. As described herein any molecules of present invention can be cloned for expression of its corresponding peptide or protein molecule.
(B) Over expression/Purification of Protein molecules of the present invention: Appropriate clones (as shown in Figures 4-7) expressing all four versions of above- mentioned proteins in E. coli were identified by appropriate antibiotic selection as described by the manufacturer (Novagen 441 Charmany Drive Madison, WI, 53719, USA). Expressed protein bands of the predicted sizes were identified by analytical SDS poly acrylamide gel electrophoresis (Laemmli, U.K., Nature, 227, 680, 1970) to confirm the expression of desired protein. A gel with the His tagged and Non tagged proteins for all four proteins clearly shows the size differences. Protein obtained from the plasmid pMON63134 (Figure 6) was chosen for assay work described below. Protein extracts were prepared by freeze thawing and sonication as described Cull and McHenry (Cull M and McHenry CS. Methods in Enzymology 182, 147-153, 1990). Pellets were resuspended in an appropriate volume of either 50 mM Tris-HCI pH 7.4 250 mM NaCI and 500 μM EDTA with 0.05% CHAPS and ImM ABESF or 50 mM KH2PO4 pH-7.2 250 mM NaCI and 250 μM EDTA with 0.05% CHAPS and ImM ABESF, buffer. Crude extracts containing His-tagged protein were applied to equilibrated His-Trap (Pharmacia Piscataway, NJ) columns and purified by standard procedures as described by the manufacturer of the columns. Steps included a 10 mM imidazole elution /wash and elution in 250 mM imidazole. As a result the Arabidopsis tagged protein was partially purified (approximatey 85-90 % by SDS-PAGE stained with Coomasie Blue). Purified samples were gel filtered into 50 mM KH2PO4 pH-7.2 250 mM NaCI and 250 μM EDTA, 10% glycerol and stored at -80°C until use. Protein concentrations were 1.5 to 1.7 mg /mL,based on the method of Bradford (Bradford M, Anal Biochem, 72, 248, 1976 and Bio-Rad Laboratories Procedure bulletin 1123) with BSA standard (Bradford reagent and BSA was from Bio-Rad Laboratories Headquarters 1000 Alfred Nobel Drive Hercules, CA 94547). The assays described below used 10 or 15 uL of this solution
(15 to 22 μg per reaction).
(C) Catalytic or Enymatic activity determination of peptide/protein molecules of present invention.
Spectrophotometric assays were used for determination of enzyme activities of purified proteins. Assays were based on earlier described methods (Portsteffen et al. Phyochemistry, 37, 391, 1994). Consumption or production of NADPH was observed at 340 nm. Assays were performed in a Varian Gary 50 Bio spectrophotometer (Varian Instruments inc. AA, ICP, UV, Fluorescence Products 679 Springvale RoadMulgrave, Victoria 3170Australia). All assays were run at 30°C for 10 to 30 minutes in lmL volumes using 100 mM KH2PO4 pH 6.5, 250 μM EDTA or 50 mM KH2PO4 pH-7.2, 250 mM NaCI, and 250 μM EDTA as reaction buffer. If metals were added they were supplemented to reactions in cuvettes from 100 mM stocks of either ZnCl2 and or MgCl2. Substrates included those found in table 8. All assays contained 3 to 30μg of protein 200 to 400 μM NADPH or NADP and 100 μM to 10 mM substrate (acetone 60mM). Controls contained no substrate to determine the background rate of consumption and degradation of NADPH. NADPH solutions were freshly made and checked by UV-vis for degradation. Enzyme solutions were supplemented with ImM MgCl2 and ImM Zn Cl2 which were at 1.5 to 1.7 mg /mL concentrations. The assays used 10 or 15 μL of this solution (ca. 15 to 22 μg per reaction). Stock solutions of the substrates (20 to 500mM) were made up in a reaction buffer or as noted. Steroids solutions were made in 100% EtOH. From these stock solutions additions to the assays were made directly. Precipitation of steroids was observed. Thus, the soluble concentration of steroids in reaction mixture is not accurately known in each case. However, good estimate of substrate (sterol) concentration would be 100 to 200 μM for a saturated solution under these conditions. There was a clear difference in rate observed by changing concentration from 26 μM to 260 μM indicating that saturation may be between the two points both for enzyme activity and substrate solubility. At these concentrations precipitation did not have an effect on the absorbance changes at 340 nm as evidenced by controls containing substrate and NADPH. In addition to cell extract control, a substrate control was also employed because solutions of some substrates tested can have a lower pH at described assay conditions which may affect rates of NADPH degradation and enzyme activity under assay conditions. . Results of the substrates tested can be found in Table 10 below.
These results (Table 8 to 10) show that the A. thaliana protein molecule SEQ ID NO: 4 has significant steroid reductase activity. Combined with other biological data these results suggest that SEQ ID NO: 4 is likely a steroid reductase with true substrates that are most likely brassinosteroids. These results do not preclude the possibility that another compound class might act as substrate but results suggest that the substrate is a ketone moiety potentially in a steroid ring system.
Table 8: Zero Order Result Table using acetone as substrate (60mM) shows evidence of reductase activity. Turnover (consumption of NADPH) is observed in the reductive direction. Slopes (for acetone, lines 1 and 2) indicate the difference from control is 10 fold. Controls (lines 3 and 4) were identical except for the omission of acetone.
Sample Start Stop Slope A A S.D.
(min) (min) (Abs/min) Start Stop
Acetone 0.000 10.00 -0.0118 0.8869 0.76-0 0.0016
Acetone 0.000 10.00 -0.0111 0.8655 0.7502 0.0025
Control 0.000 10.00 -0.0006 0.8609 0.8547 0.0006
Control 0.000 10.00 -0.0010 0.8773 0.8683 0.0011
Start=start time, Stop=stop time, Slope is change in absorbance over time, A start is starting absorbance, A stop is ending absorbance.
Table 9: Zero Order Result Table. This table shows elected data for 5α-androstane-3,17- dione in a reductase assay. This example shows the results for three reactions with acetone, three reactions with 5α-androstane-3,17-dione and two controls. Table 8 shows the initial rates as estimated by linear fit between 0 and 5 minutes. The results for 5α-androstane-3,17-dione shown here are the best of any substrate at the same concentration (approx 200 μM). 4- androstene-3,17-dione is the next best substrate with a rate on the order of 2.5 to 4 times slower than this under the same conditions (not shown). Acetone results at similar concentrations (Table 7 shows results at high acetone concentration) are on the order of 10 to 20 times slower and nearly at background levels. Similar results have been obtained with multiple batches of enzyme and substrate. Sample Start Stop Slope A A S.D.
(min) (min) Abs/min) Start Stop
5-alpha 0.000 5.000 -0.0632 1.4399 1.1322 0.0018
5 -alpha 0.000 5.000 -0.0611 1.4105 1.1092 0.0022
5-alpha 0.000 5.000 -0.0652 1.4710 1.1652 0.0030 acetone 0.000 5.000 -0.0049 1.5050 1.4793 0.0018 acetone 0.000 5.000 -0.0047 1.3902 1.3961 0.0012 acetone 0.000 5.000 -0.0041 1.4584 1.4352 0.0014
Control 0.000 5.000 -0.0030 1.5256 1.5118 0.0013
Control 0.000 5.000 -0.0035 1.4898 1.4743 0.0018
Start=start time, Stop=stop time, Slope is change in absorbance ovei time, A start is starting absorbance, A stop is ending absorbance
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0001
EXAMPLE 11
In the following example a large number of DNA sequences were searched using BlastA
(Altschul, Stephen F., Thomas L. Madden, Alejandro A. Schaffer, Jinghui Zhang, Zheng Zhang, Webb Miller, and David J. Lipman , Nucleic Acids Res. (25)3389-3402 (1997)) to find various homologs, paralogs or orthologs of the gene of the present invention. These sequences are determined from cDNA libraries prepared from a variety plant species and tissues.
For construction of cDNA libraries, tissue is harvested and immediately frozen in liquid nitrogen and stored at -80°C until total RNA extraction. Total RNA is purified from tissue using Trizol reagent from Life Technologies (Gibco BRL, Life Technologies, Gaithersburg, Maryland U.S.A.), as recommended by the manufacturer. Poly A+ RNA (mRNA) is purified using magnetic oligo dT beads essentially as recommended by the manufacturer (Dynabeads, Dynal Corporation, Lake Success, New York U.S.A.).
Construction of plant cDNA libraries is well known in the art and a number of cloning strategies exist. A number of cDNA library construction kits are commercially available. The
Superscript™ Plasmid System for cDNA synthesis and Plasmid Cloning (Gibco BRL, Life
Technologies, Gaithersburg, Maryland U.S. A.) is used, following the conditions suggested by the manufacturer.
The cDNA libraries are plated on LB agar containing the appropriate antibiotics for selection and incubated at 37° for sufficient time to allow the growth of individual colonies.
Single selective media colonies are individually placed in each well of a 96-well microtiter plates containing LB liquid including the selective antibiotics. The plates are incubated overnight at approximately 37°C with gentle shaking to promote growth of the cultures.
The plasmid DNA is isolated from each clone using Qiaprep plasmid isolation kits, using the conditions recommended by the manufacturer (Qiagen Inc., Santa Clara, California U.S.A.). The template plasmid DNA clones are used for subsequent sequencing. For sequencing the cDNA libraries, a commercially available sequencing kit, such as the ABI PRISM dRhodamine Terminator Cycle Sequencing Ready Reaction Kit with AmpliTaq® DNA Polymerase, FS, is used under the conditions recommended by the manufacturer (PE Applied Biosystems, Foster City, CA). The cDNAs of the present invention are generated by sequencing initiated from the 5' end or 3' end of each cDNA clone. Entire inserts or only part of the inserts (ESTs or expressed sequenced tags) are sequenced.
A number of DNA sequencing techniques are known in the art, including fluorescence- based sequencing methodologies. These methods have the detection, automation and instrumentation capability necessary for the analysis of large volumes of sequence data. Currently, the 377 and 3700 DNA Sequencer (Perkin-Elmer Coφ., Applied Biosystems Div., Foster City, CA) allows the most rapid electrophoresis and data collection. With these types of automated systems, fluorescent dye-labeled sequence reaction products are detected and data entered directly into the computer, producing a chromatogram that is subsequently viewed, stored, and analyzed using the corresponding software programs. These methods are known to those of skill in the art and have been described and reviewed (Birren et al, Genome Analysis: Analyzing DNA,1, Cold Spring Harbor, New York, the entirety of which is herein incoφorated by reference).
The generated ESTs (including any full-length cDNA inserts or complete coding sequences) are combined with ESTs and full-length cDNA sequences in public databases such as GenBank. Duplicate sequences are removed, and duplicate sequence identification numbers are replaced. The combined dataset is then clustered and assembled using Pangea Systems tool identified as CAT v.3.2. First, the EST sequences are screened and filtered, e.g. high frequency words are masked to prevent spurious clustering; sequence common to known contaminants such as cloning bacteria are masked; high frequency repeated sequences and simple sequences are masked; unmasked sequences of less than 100 bp are eliminated. The screened and filtered ESTs are combined and subjected to a word-based clustering algorithm which calculates sequence pair distances based on word frequencies and uses a single linkage method to group like sequences into clusters of more than one sequence, as appropriate. Clustered sequences are assembled individually using an iterative method based on PHRAP/CRAW/MAP providing one or more self-consistent consensus sequences and inconsistent singleton sequences. The assembled clustered sequence files are checked for completeness and parsed to create data representing each consensus contiguous sequence (contig), the initial EST sequences, and the relative position of each EST in a respective contig. The sequence of the 5' most clone is identified from each contig. The initial sequences that are not included in a contig are separated out.
Above described databases . with nucleotide and peptide sequences were queried with sequences of the present invention to get following homologues, orthologs or paralogs as shown in following table. The BLAST 2.2.1 software (Altschul, Stephen F., Thomas L. Madden, Alejandro A. Schaffer, Jinghui Zhang, Zheng Zhang, Webb Miller, and David J. Lipman , Nucleic Acids Res. (25)3389-3402 (1997), with BLOSUM62 matrix and "no Filter" options, was used in the queries. As when necessary, frame-shifts in the DNA sequences of the homologues were detected by aligning the DNA sequence of the homologue in question to the protein sequence of present invention, using the "frame+_n2p" program with default parameters in the GenCore software package (Compugen Inc., 1998). Such frame-shifts were conceptually corrected to yield open reading frames. The "translate" program with default parameters in the same package was used to translate open reading frames to corresponding peptide sequences based on standard genetic codes.
SEQ ID NO: 1 exhibits 52.857% percent identity with its closest known functional gene, identified using the BLAST 2.2.1 software with BLOSUM62 matrix and "no Filter" options (Genbank accession number gi|1717752|sp|P50162|TRNl_DATST). SEQ ID NO: 1 exhibits identity of 111 amino acid residues over its entire length of 210 amino acids with tropionone reductase of Datura stramonium (Genbank accession number gi|1717752|sp|P50162|TRNl_DATST). Due to this relationship, it is possible that enzymes with similar activities can also function in present invention, also DNA molecules encoding proteins that are closely related to tropionone reductase are an aspect of the present invention. Table 11.
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000071_0001

Claims

CLAIMS:
1. A method of increasing seed and organ size of a plant comprising the steps of: a) transforming said plant with a DNA construct comprising a promoter that functions in plants, operably linked to a DNA molecule that encodes a protein, wherein said DNA molecule is selected from the group consisting of: SEQ ID
NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9, SEQ ID NO: 11, SEQ ID NO: 13, SEQ ID NO: 15, SEQ ID NO: 17, SEQ ID NO: 19, SEQ ID NO: 21, SEQ ID NO: 23, SEQ ID NO: 25, SEQ ID NO: 27, SEQ ID NO: 29, SEQ ID NO: 31, SEQ ID NO: 33, SEQ ID NO: 35, SEQ ID NO: 37, SEQ ID NO: 39, SEQ ID NO: 41, SEQ ID NO: 43, SEQ ID NO: 45, SEQ ID NO: 47,
SEQ ID NO: 49, SEQ ID NO: 51, SEQ ID NO: 53, SEQ ID NO: 66, SEQ ID NO: 68, and SEQ ID NO: 70 which is operably linked to a 3 'termination region; and x b) selecting a desired plant from a population of transformed plants containing said DNA construct; wherein said desired plant exhibits increased seed and organ size compared to a plant of a same plant species not transformed to contain said DNA construct.
2. The method of claim 1, wherein said promoter is a caulimovirus promoter.
3. The method of claim 1, wherein said promoter comprises a heterologous plant constitutive promoter.
4. The method of claim 1, wherein said promoter is a tissue specific promoter or an organ enhanced promoter.
5. A method of increasing seed and organ size of a plant comprising the steps of: a) transforming said plant with a DNA construct comprising a promoter that functions in plants, operably linked to a DNA molecule that encodes a protein, wherein said protein comprises at least an N-terminal 50% portion of a polypeptide selected from the group consisting of : SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10, SEQ ID NO: 12, SEQ ID NO: 14, SEQ ID NO: 16, SEQ ID NO: 18 SEQ ID NO: 20, SEQ ID NO: 22, SEQ ID NO: 24, SEQ ID NO: 26, SEQ ID NO: 28 SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 34, SEQ ID NO: 36, SEQ ID NO: 38 SEQ ID NO: 40, SEQ ID NO: 42, SEQ ID NO: 44, SEQ ID NO: 46, SEQ ID NO: 48 SEQ ID NO: 50, SEQ ID NO: 52, SEQ ID NO: 54, SEQ ID NO: 67, SEQ ID NO: 69; and SEQ ID NO: 71, wherein said DNA construct is operably linked to a 3'termination region; and b) selecting a plant from a population of transformed plants containing said DNA construct; wherein said plant exhibits increased seed and organ size compared to a plant of a same plant species not transformed to contain said DNA construct.
6. The method of claim 5, wherein said plant species is selected from the group consisting of: Acacia, alfalfa, aneth, apple, apricot, artichoke, arugula, asparagus, avocado, banana, barley, beans, beet, blackberry, blueberry,, broccoli, brussels sprouts, cabbage, canola, cantaloupe, carrot, cassava, cauliflower, celery, cherry, cilantro, citrus, Clementines, coffee, corn, cotton, cucumber, Douglas fir, eggplant, endive, escarole, eucalyptus, fennel, figs, gourd, grape, grapefruit, honey dew, jicama, kiwifruit, lettuce, leeks, lemon, lime, loblolly pine, mango, melon, millet, mushroom, nut, oat, okra, onion, orange, an ornamental plant, papaya, parsley, pea, peach, peanut, pear, pepper, persimmon, pine, pineapple, plantain, plum, pomegranate, poplar, potato, pumpkin, quince, radiata pine, radicchio, radish, raspberry, rice, rye, sorghum, southern pine, soybean, spinach, squash, strawberry, sugarbeet, sugarcane, sunflower, sweet potato, sweetgum, tangerine, tea, tobacco, tomato, turf, a vine, watermelon, wheat, yams, and zucchini.
7. A transgenic plant with increased seed and organ size compared to a non transformed plant of a same plant species, said transgenic plant comprising a DNA construct, wherein said
DNA construct encodes a protein comprising at least an N-terminal 50% portion selected from the group consisting of: SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10, SEQ ID NO: 12, SEQ ID NO: 14, SEQ ID NO: 16, SEQ ID NO: 18, SEQ ID NO: 20, SEQ ID NO: 22, SEQ ID NO: 24, SEQ ID NO: 26, SEQ ID NO: 28, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 34, SEQ ID NO: 36, SEQ ID NO: 38, SEQ ID NO: 40, SEQ ID NO: 42, SEQ ID NO: 44, SEQ ID NO: 46, SEQ ID NO: 48, SEQ ID NO: 50, SEQ ID NO: 52, SEQ ID NO: 54, SEQ ID NO: 67, SEQ ID NO: 69, and SEQ ID NO: 71.
8. The transgenic plant of claim 1, wherein said DNA construct encodes a protein comprising at least an N-terminal 60% portion selected from the group consisting of: SEQ ID
NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10, SEQ ID NO: 12, SEQ ID NO: 14, SEQ ID NO: 16, SEQ ID NO: 18, SEQ ID NO: 20, SEQ ID NO: 22, SEQ ID NO: 24, SEQ ID NO: 26, SEQ ID NO: 28, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 34, SEQ ID NO: 36, SEQ ID NO: 38, SEQ ID NO: 40, SEQ ID NO: 42, SEQ ID NO: 44, SEQ ID NO: 46, SEQ ID NO: 48, SEQ ID NO: 50, SEQ ID NO: 52, SEQ ID NO: 54, SEQ ID NO: 67, SEQ ID NO: 69, and SEQ ID NO: 71.
9. The transgenic plant of claim 7, wherein said plant species is selected from the group consisting of: Acacia, alfalfa, aneth, apple, apricot, artichoke, arugula, asparagus, avocado, banana, barley, beans, beet, blackberry, blueberry, broccoli, brussels sprouts, cabbage, canola, cantaloupe, carrot, cassava, cauliflower, celery, cherry, cilantro, citrus, Clementines, coffee, corn, cotton, cucumber, Douglas fir, eggplant, endive, escarole, eucalyptus, fennel, figs, gourd, grape, grapefruit, honey dew, jicama, kiwifruit, lettuce, leeks, lemon, lime, loblolly pine, mango, melon, millet, mushroom, nut, oat, okra, onion, orange, an ornamental plant, papaya, parsley, pea, peach, peanut, pear, pepper, persimmon, pine, pineapple, plantain, plum, pomegranate, poplar, potato, pumpkin, quince, radiata pine, radicchio, radish, raspberry, rice, rye, sorghum, southern pine, soybean, spinach, squash, strawberry, sugarbeet, sugarcane, sunflower, sweet potato, sweetgum, tangerine, tea, tobacco, tomato, turf, a vine, watermelon, wheat, yams, and zucchini.
10. A progeny plant produced from said transgenic plant of claim 7.
PCT/US2003/014989 2002-05-15 2003-05-14 Method of increasing plant organ and seed size in a plant WO2003096797A2 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
AU2003237834A AU2003237834A1 (en) 2002-05-15 2003-05-14 Method of increasing plant organ and seed size in a plant
EP03736593A EP1504106B1 (en) 2002-05-15 2003-05-14 Method of increasing plant seed weight
DK03736593.9T DK1504106T3 (en) 2002-05-15 2003-05-14 Method of increasing the weight of a plant seed
US10/513,167 US7335812B2 (en) 2002-05-15 2003-05-14 Method of increasing plant organ and seed size in a plant
AT03736593T ATE473281T1 (en) 2002-05-15 2003-05-14 METHOD FOR INCREASE SEED WEIGHT IN A PLANT
DE60333280T DE60333280D1 (en) 2002-05-15 2003-05-14 PROCESS FOR INCREASING THE SEED WEIGHT IN A PLANT
US12/001,901 US7994402B2 (en) 2002-05-15 2007-12-13 Method of increasing plant organ and seed size in a plant

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US38110002P 2002-05-15 2002-05-15
US60/381,100 2002-05-15

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US10513167 A-371-Of-International 2003-05-14
US12/001,901 Division US7994402B2 (en) 2002-05-15 2007-12-13 Method of increasing plant organ and seed size in a plant

Publications (2)

Publication Number Publication Date
WO2003096797A2 true WO2003096797A2 (en) 2003-11-27
WO2003096797A3 WO2003096797A3 (en) 2004-11-18

Family

ID=29550070

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2003/014989 WO2003096797A2 (en) 2002-05-15 2003-05-14 Method of increasing plant organ and seed size in a plant

Country Status (9)

Country Link
US (2) US7335812B2 (en)
EP (1) EP1504106B1 (en)
AR (2) AR040020A1 (en)
AT (1) ATE473281T1 (en)
AU (1) AU2003237834A1 (en)
DE (1) DE60333280D1 (en)
DK (1) DK1504106T3 (en)
ES (1) ES2346863T3 (en)
WO (1) WO2003096797A2 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2353387A1 (en) 2010-02-05 2011-08-10 Bayer CropScience AG Use of succinate dehydrogenase (SDH) inhibitors in the treatment of plant types in the sweet grass family
WO2014161908A1 (en) 2013-04-05 2014-10-09 Bayer Cropscience Nv Brassica plants comprising mutant da1 alleles

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012112529A1 (en) 2011-02-14 2012-08-23 Xyleco, Inc. Processing biomass
US9297021B2 (en) 2011-11-02 2016-03-29 University Of North Texas MtNIP regulated plants with significantly increased size and biomass
JP2021501231A (en) 2017-10-27 2021-01-14 キシレコ インコーポレイテッド Biomass processing method

Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0050424A1 (en) 1980-09-24 1982-04-28 Cetus Corporation Method for producing an analytical antibody probe, an analytical antibody probe, and a method for analysing a sample for certain antibodies
EP0084796A2 (en) 1982-01-22 1983-08-03 Cetus Corporation HLA typing method and cDNA probes used therein
EP0120516A2 (en) 1983-02-24 1984-10-03 Rijksuniversiteit Leiden A process for the incorporation of foreign DNA into the genome of dicotyledonous plants; Agrobacterium tumefaciens bacteria and a process for the production thereof
EP0154204A2 (en) 1984-03-06 1985-09-11 Mgi Pharma, Inc. Herbicide resistance in plants
US4582788A (en) 1982-01-22 1986-04-15 Cetus Corporation HLA typing method and cDNA probes used therein
EP0201184A2 (en) 1985-03-28 1986-11-12 F. Hoffmann-La Roche Ag Process for amplifying nucleic acid sequences
US4683194A (en) 1984-05-29 1987-07-28 Cetus Corporation Method for detection of polymorphic restriction sites and nucleic acid sequences
EP0237362A1 (en) 1986-03-13 1987-09-16 F. Hoffmann-La Roche Ag Process for detecting specific nucleotide variations and genetic polymorphisms present in nucleic acids and kits therefor
EP0258017A2 (en) 1986-08-22 1988-03-02 F. Hoffmann-La Roche Ag Purified thermostable enzyme and process for amplifying, detecting, and/or cloning nucleic acid sequences using said enzyme
US5107065A (en) 1986-03-28 1992-04-21 Calgene, Inc. Anti-sense regulation of gene expression in plant cells
US5231020A (en) 1989-03-30 1993-07-27 Dna Plant Technology Corporation Genetic engineering of novel plant phenotypes
US5641876A (en) 1990-01-05 1997-06-24 Cornell Research Foundation, Inc. Rice actin gene and promoter
US5689042A (en) 1995-03-29 1997-11-18 Wisconsin Alumni Research Foundation Transgenic plants with altered senescence characteristics
US5859347A (en) 1992-03-19 1999-01-12 Monsanto Company Enhanced expression in plants
US6147278A (en) 1985-10-25 2000-11-14 Monsanto Company Plant vectors

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE69816059T2 (en) * 1997-02-10 2004-04-22 National Research Council Of Canada, Ottawa GENE THAT CODES FOR THE PYRUVATE DEHYDROGENASE KINASE OF PLANTS
CA2263067A1 (en) 1999-02-26 2000-08-26 The Australian National University Method of modifying plant morphology, biochemistry and physiology
JP2003525051A (en) * 2000-03-01 2003-08-26 リサーチ・アンド・ディベロップメント・インスティチユート・インコーポレイテッド Transgenic plants with improved seed yield, biomass, and harvest index
AU8681101A (en) * 2000-08-24 2002-03-04 Scripps Research Inst Stress-regulated genes of plants, transgenic plants containing same, and methodsof use
AU2001289843A1 (en) * 2001-08-28 2002-02-13 Bayer Cropscience Ag Polypeptides for identifying herbicidally active compounds
US20090183270A1 (en) * 2002-10-02 2009-07-16 Adams Thomas R Transgenic plants with enhanced agronomic traits

Patent Citations (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0050424A1 (en) 1980-09-24 1982-04-28 Cetus Corporation Method for producing an analytical antibody probe, an analytical antibody probe, and a method for analysing a sample for certain antibodies
EP0084796A2 (en) 1982-01-22 1983-08-03 Cetus Corporation HLA typing method and cDNA probes used therein
US4582788A (en) 1982-01-22 1986-04-15 Cetus Corporation HLA typing method and cDNA probes used therein
EP0120516A2 (en) 1983-02-24 1984-10-03 Rijksuniversiteit Leiden A process for the incorporation of foreign DNA into the genome of dicotyledonous plants; Agrobacterium tumefaciens bacteria and a process for the production thereof
EP0154204A2 (en) 1984-03-06 1985-09-11 Mgi Pharma, Inc. Herbicide resistance in plants
US4683194A (en) 1984-05-29 1987-07-28 Cetus Corporation Method for detection of polymorphic restriction sites and nucleic acid sequences
US4683202B1 (en) 1985-03-28 1990-11-27 Cetus Corp
EP0201184A2 (en) 1985-03-28 1986-11-12 F. Hoffmann-La Roche Ag Process for amplifying nucleic acid sequences
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US6147278A (en) 1985-10-25 2000-11-14 Monsanto Company Plant vectors
EP0237362A1 (en) 1986-03-13 1987-09-16 F. Hoffmann-La Roche Ag Process for detecting specific nucleotide variations and genetic polymorphisms present in nucleic acids and kits therefor
US5107065A (en) 1986-03-28 1992-04-21 Calgene, Inc. Anti-sense regulation of gene expression in plant cells
EP0258017A2 (en) 1986-08-22 1988-03-02 F. Hoffmann-La Roche Ag Purified thermostable enzyme and process for amplifying, detecting, and/or cloning nucleic acid sequences using said enzyme
US5231020A (en) 1989-03-30 1993-07-27 Dna Plant Technology Corporation Genetic engineering of novel plant phenotypes
US5641876A (en) 1990-01-05 1997-06-24 Cornell Research Foundation, Inc. Rice actin gene and promoter
US5859347A (en) 1992-03-19 1999-01-12 Monsanto Company Enhanced expression in plants
US5689042A (en) 1995-03-29 1997-11-18 Wisconsin Alumni Research Foundation Transgenic plants with altered senescence characteristics

Non-Patent Citations (66)

* Cited by examiner, † Cited by third party
Title
"Current Protocols in Molecular Biology", 1989, JOHN WILEY & SONS, pages: 6.3.1 - 6.3.6
"Fromm,.UCLA Symposium on Molecular Strategies for Crop Improvement", 1990, KEYSTONE
ALTSCHUL ET AL., JMOI. BIOL., vol. 215, 1993, pages 403 - 410
ALTSCHUL, STEPHEN F. ET AL., NUCLEIC ACIDS RES., vol. 25, 1997, pages 3389 - 3402
ALTSCHUL, STEPHEN F. ET AL.: "Nucleic Acids Res.", vol. 25, 1997, WEBB MILLER, AND DAVID J. LIPMAN, pages: 3389 - 3402
AMMIRATO ET AL.: "Handbook of Plant Cell Culture - Crop Species", 1984, MACMILLAN PUBL. CO.
AUSUBEL ET AL.: "Current Protocols in Molecular Biology", 1989, JOHN WILEY & SONS
BACK ET AL., PLANT MOL. BIOL., vol. 17, 1991, pages 9
BEVAN, NUCLEIC ACIDS RES., vol. 12, 1984, pages 8711 - 8721
BI ET AL., PLANT J., vol. 8, 1995, pages 235 - 245
BINDING: "Regeneration of Plants, Plant Protoplasts", 1985, CRC PRESS, pages: 21 - 73
BUSK ET AL., PLANT J., vol. 11, 1997, pages 1285 - 1295
CHEN ET AL., PLANT J., vol. 10, 1996, pages 955 - 966
DATTA ET AL., BIO-TECHNOLOGY, vol. 8, 1990, pages 736 - 740
DE VEYLDER ET AL., PLANT CELL
DELLAPORTA ET AL., STADLER SYMPOSIUM, vol. 11, 1988, pages 263 - 282
EVANS ET AL.: "Handbook of Plant Cell Culture", 1983, MACMILLILAN PUBLISHING COMPANY, article "Protoplasts Isolation and Culture", pages: 124 - 176
FRALEY ET AL., PROC. NATL. ACAD. SCI. USA, vol. 80, 1983, pages 4803 - 4807
FROHMAN ET AL., PROC. NATL. ACAD. SCI. USA
GAN S.; AMASINO R.M., PLANT PHYSIOL., vol. 113, 1997, pages 313 - 319
GAN S.; AMASINO R.M., SCIENCE, vol. 270, 1995, pages 1986 - 1988
GAN; AMASINO, SCIENCE, vol. 270, 1995, pages 1986 - 1988
GUARENTE L.; RUVKUN G.; AMASINO R.M., PROC. NATL ACAD. SCI USA, vol. 95, 1998, pages 1034 - 1036
HAYASHIMOTO, PLANT PHYSIOL., vol. 93, 1990, pages 857 - 863
HENSEL L.L ET AL., THE PLANT CELL, vol. 5, 1993, pages 553 - 564
HERRERA-ESTRELLA ET AL., NATURE, vol. 303, 1983, pages 209
HINCHEE ET AL., BIO/TECHNOLOGY, vol. 6, 1988, pages 915 - 922
HUANG ET AL., PLANT MOL. BIOL., vol. 33, 1997, pages 125 - 139
IKATU ET AL., BIO/TECHNOL., vol. 8, 1990, pages 241 - 2 42
J. J. DOYLE; B. S. GAUT, PLANT MOLECULAR BIOLOGY, vol. 42, 2000, pages 1 - 23
JEFFERSON ET AL., BIOCHEM. SOC. TRANS., vol. 15, 1987, pages 17 - 19
JEFFERSON ET AL., EMBO J., vol. 6, 1987, pages 3901 - 3907
JEFFERSON, PLANT MOL. BIOL, REP., vol. 5, 1987, pages 387 - 405
K. WISING ET AL., ANN. REV. GENETICS, vol. 22, 1988, pages 421
KATZ ET AL., J. GEN. MICROBIOL., vol. 129, 1983, pages 2703 - 2714
KIRCH, PLANT MOL. BIOL., vol. 33, 1997, pages 897 - 909
KLEE ET AL., ANN. REV. PLANT PHYS., vol. 38, 1987, pages 467 - 486
KLEE ET AL., BIO-TECHNOLOGY, vol. 3, no. 7, 1985, pages 637 - 642
LIU ET AL., PLANT PHYSIOL., vol. 115, 1997, pages 397 - 407
LOHMAN K.M. ET AL., PHYSIOLOGIA PLANTARUM, vol. 92, 1994, pages 322 - 328
MULLIS ET AL.: "ymp. Quant. Biol.", vol. 51, 1986, COLD SPRING HARBOR, pages: 263 - 273
NEEDLEMAN; WUNSCH, J. MOL. BIOL., vol. 48, 1970, pages 443 - 453
OW ET AL., SCIENCE, vol. 234, 1986, pages 856 - 859
POTRYKUS ET AL., MOL. GEN. GENET., vol. 199, 1985, pages 183 - 188
RATTAN ET AL.: "Protein Synthesis: Post-translational Modifications and Aging", ANN. N.Y ACAD SCI., vol. 663, 1992, pages 48 - 62
S. HENIKOFF; J. G. HENIKOFF, PROC. NATL. ACAD. SCI. U.S.A., vol. 89, 1992, pages 10915 - 10919
SAKAI ET AL., PLANT CELL PHYSIOL., vol. 37, 1996, pages 906 - 913
SAMBROOK ET AL.: "Molecular Cloning - A Laboratory Manual", 1989, COLD SPRING HARBOR LABORATORY
SAMBROOK ET AL.: "Molecular Cloning, A Laboratory Manual", 1989, COLD SPRING HARBOR PRESS
SEIFTER ET AL., METH. ENZYMOL., vol. 182, 1990, pages 626 - M
SHEEN ET AL., SCIENCE, vol. 274, 1996, pages 1900 - 1902
SHIMAMOTO ET AL.: "Nature", vol. 338, 1989, pages: 274 - 276
STALKER ET AL., J. BIOL. CHEM., vol. 263, 1988, pages 6310 - 6314
STREIT ET AL., MOL. PLANT MICROBE INTERACT., vol. 10, 1997, pages 933 - 937
SUTCLIFFE ET AL., PROC. NATL. ACAD SCI., vol. 75, 1978, pages 3737 - 3741
T. E. CREIGHTON; W. H. FREEMAN; COMPANY: "Proteins - Structure and Molecular Properties", 1993
T. F. SMITH; M. S. WATERMAN, J. MOL.BIOL, vol. 147, 1981, pages 195 - 197
THILLET ET AL., J. BIOL. CHEM., vol. 263, 1988, pages 12500 - 12508
THOMAS H.; STODDART J. L., ANN. REV. PLANT PHYSIOL, vol. 31, 1980, pages 83 - 111
UKNES ET AL., PLANT. CELL, vol. 5, 1993, pages 159 - 169
VASIL ET AL., BIO/TECHNOLOGY, vol. 10, 1992, pages 667 - 674
VASIL ET AL., BIO/TECHNOLOGY, vol. 8, 1990, pages 429 - 434
W.R. PEARSON, GENOMICS, vol. 11, 1991, pages 635 - 650
WEAVER L.M. ET AL., PLANT MOL. BIOL., 1998, pages 455 - 469
WOLD, F.: "Post-translational Covalent Modification of Proteins", 1983, ACADEMIC PRESS, article "Post-translational Protein Modifications. Perspectives and Prospects", pages: 1 - 12
ZUKOWSKY ET AL., PROC. NATL. ACAD SCI., vol. 80, 1983, pages 1101 - 1105

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2353387A1 (en) 2010-02-05 2011-08-10 Bayer CropScience AG Use of succinate dehydrogenase (SDH) inhibitors in the treatment of plant types in the sweet grass family
WO2011095496A2 (en) 2010-02-05 2011-08-11 Bayer Cropscience Ag Use of succinate dehydrogenase (sdh) inhibitors in treating plant species of the family of true grasses
WO2014161908A1 (en) 2013-04-05 2014-10-09 Bayer Cropscience Nv Brassica plants comprising mutant da1 alleles
US10287603B2 (en) 2013-04-05 2019-05-14 Bayer Cropscience Nv Brassica plants comprising mutant DA1 alleles

Also Published As

Publication number Publication date
US20090133162A1 (en) 2009-05-21
EP1504106A2 (en) 2005-02-09
EP1504106B1 (en) 2010-07-07
EP1504106A4 (en) 2006-06-21
DK1504106T3 (en) 2010-10-11
AU2003237834A8 (en) 2003-12-02
AR040020A1 (en) 2005-03-09
ES2346863T3 (en) 2010-10-21
AR073039A2 (en) 2010-10-06
WO2003096797A3 (en) 2004-11-18
ATE473281T1 (en) 2010-07-15
US20060168693A1 (en) 2006-07-27
US7994402B2 (en) 2011-08-09
US7335812B2 (en) 2008-02-26
DE60333280D1 (en) 2010-08-19
AU2003237834A1 (en) 2003-12-02

Similar Documents

Publication Publication Date Title
US6307126B1 (en) Nucleic acid encoding GAI gene of Arabidopsis thaliana
EP2241624A2 (en) Nucleic acid molecules and their use in plant sterility
US6781035B1 (en) Leafy cotyledon1 genes and their uses
PL189711B1 (en) Method of improving seeds
US20080209585A1 (en) Genes which produce staygreen characteristics in maize and their uses
US7994402B2 (en) Method of increasing plant organ and seed size in a plant
AU2001241600B2 (en) Leafy cotyledon1 genes and their uses
US7612258B2 (en) Nucleic acid molecules associated with plant cell proliferation and growth and uses thereof
US20100281580A1 (en) Use of a gene encoding a histidine protein kinase to create drought resistant plants
US20040006796A1 (en) Methods of modulating cytokinin related processes in a plant using B3 domain proteins
US7932434B2 (en) Late blight resistance gene from wild potato
EP1699926B1 (en) Stress tolerant plants and methods thereof
EP1377666A2 (en) Plant growth regulating genes, proteins and uses thereof
WO2005017114A2 (en) Method of increasing metabolism and stress tolerance in a plant
ZA200601187B (en) Materials and methods for the modulation of cyclin-dependent kinase inhibitor-like polypeptides in maize
AU2005253642B2 (en) Nucleic acid molecules and their use in plant male sterility
AU2002304741A1 (en) Plant growth regulating genes, proteins and uses thereof

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NI NO NZ OM PH PL PT RO RU SC SD SE SG SK SL TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2003736593

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2006168693

Country of ref document: US

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 10513167

Country of ref document: US

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWP Wipo information: published in national office

Ref document number: 2003736593

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP

WWP Wipo information: published in national office

Ref document number: 10513167

Country of ref document: US