WO2013138309A1 - Genetic reduction of male fertility in plants - Google Patents

Genetic reduction of male fertility in plants Download PDF

Info

Publication number
WO2013138309A1
WO2013138309A1 PCT/US2013/030455 US2013030455W WO2013138309A1 WO 2013138309 A1 WO2013138309 A1 WO 2013138309A1 US 2013030455 W US2013030455 W US 2013030455W WO 2013138309 A1 WO2013138309 A1 WO 2013138309A1
Authority
WO
WIPO (PCT)
Prior art keywords
plant
male
plants
seq
nucleic acid
Prior art date
Application number
PCT/US2013/030455
Other languages
French (fr)
Inventor
Tim W. Fox
Original Assignee
Pioneer Hi-Bred International, Inc.
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 Pioneer Hi-Bred International, Inc. filed Critical Pioneer Hi-Bred International, Inc.
Publication of WO2013138309A1 publication Critical patent/WO2013138309A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/82Vectors or expression systems specially adapted for eukaryotic hosts for plant cells, e.g. plant artificial chromosomes (PACs)
    • C12N15/8241Phenotypically and genetically modified plants via recombinant DNA technology
    • C12N15/8261Phenotypically and genetically modified plants via recombinant DNA technology with agronomic (input) traits, e.g. crop yield
    • C12N15/8287Phenotypically and genetically modified plants via recombinant DNA technology with agronomic (input) traits, e.g. crop yield for fertility modification, e.g. apomixis
    • C12N15/8289Male sterility

Definitions

  • the disclosure relates generally to the field of molecular biology, specifically the modulation of plant fertility to improve plant stress tolerance.
  • Plants allocate photosynthates, minerals, and other growth components among various plant tissues during the developmental life cycle.
  • ear and tassel are specific female and male inflorescence structures that share certain developmental processes and compete with each other for required nutrients.
  • tassel apical dominance may limit ear growth and grain yield potential in maize plants.
  • Genetic male sterility in hybrid maize could reduce tassel dominance and divert more resources to ear growth, kernel number and/or size, ultimately leading to increased grain yield. The advantage from this shift in allocation of resources could be particularly advantageous under conditions of environmental stress.
  • Nitrogen utilization efficiency (NUE) genes affect yield and have utility for improving the use of nitrogen in crop plants, especially maize. Increased nitrogen use efficiency can result from enhanced uptake and assimilation of nitrogen fertilizer and/or the subsequent remobilization and reutilization of accumulated nitrogen reserves, as well as increased tolerance of plants to stress situations such as low nitrogen environments.
  • the genes can be used to alter the genetic composition of the plants, rendering them more productive with current fertilizer application standards or maintaining their productive rates with significantly reduced fertilizer or reduced nitrogen availability. Improving NUE in corn would increase corn harvestable yield per unit of input nitrogen fertilizer, both in developing nations where access to nitrogen fertilizer is limited and in developed nations where the level of nitrogen use remains high. Nitrogen utilization improvement also allows decreases in on-farm input costs, decreased use and dependence on the nonrenewable energy sources required for nitrogen fertilizer production and reduces the environmental impact of nitrogen fertilizer manufacturing and agricultural use.
  • plant yield is improved under stress, particularly abiotic stress, such as nitrogen limiting conditions.
  • Methods of improving plant yield include inhibiting the fertility of the plant.
  • the male fertility of a plant can be inhibited using any method known in the art, including but not limited to the disruption of a tassel development gene, or a decrease in the expression of a gene, for example through the use of co-suppression, antisense or RNA silencing or interference. Inhibition of fertility may be partial reduction or complete inhibition.
  • Male sterile plants can also be achieved by using genetic male sterile mutants.
  • Inhibiting the male fertility in a plant can improve the nitrogen stress tolerance of the plant and such plants can maintain their productive rates with significantly less nitrogen fertilizer input and/or exhibit enhanced uptake and assimilation of nitrogen fertilizer and/or remobilization and reutilization of accumulated nitrogen reserves.
  • the improvement of nitrogen stress tolerance through the reduction in male fertility can also result in increased root mass and/or length, increased ear, leaf, seed and/or endosperm size, and/or improved standability.
  • the methods further comprise growing said plants under nitrogen limiting conditions and optionally selecting those plants exhibiting greater tolerance to the low nitrogen levels.
  • compositions for improving yield under abiotic stress, which include evaluating the environmental conditions of an area of cultivation for abiotic stressors (e.g., low nitrogen levels in the soil) and planting seeds or plants having reduced male fertility, in stressful environments.
  • abiotic stressors e.g., low nitrogen levels in the soil
  • Additional methods include but are not limited to:
  • a method of increasing yield by increasing one or more yield components in a plant includes reducing male fertility by affecting the expression or activity of a nuclear encoded component in the plant, and growing the plant under plant growing conditions, wherein the component exhibits a dominant phenotype.
  • the nuclear encoded component is a male fertility gene or a male sterility gene that has a dominant phenotype.
  • the male fertility gene or the male sterility gene is a transgene.
  • the male fertility gene encodes a protein of SEQ ID NO: 10.
  • the male fertility gene includes a nucleotide sequence of SEQ ID NO: 13.
  • the male fertility gene encodes a polypeptide of SEQ ID NO: 14.
  • the reduction of male fertility or rendering the plant male sterile is effected by a single nucleotide substitution resulting in an amino acid change at amino acid 37, from Alanine to Threonine in the predicted protein (SEQ ID NO: 153).
  • the reduction of male fertility or rendering the plant male sterile is effected by an amino acid change at amino acid 37, from Alanine to Valine in the predicted protein.
  • the dominant male fertility gene is operably linked to a promoter selected from the group consisting of: inducible promoter, tissue preferred promoter, temporally regulated promoter or an element thereof.
  • the promoter preferentially drives expression in male reproductive tissue.
  • the male fertility is reduced in the female plant (e.g., a female inbred line) of a breeding pair.
  • a plant or a cell or a seed or a progeny thereof that includes the reduced male fertility sequence encoding amino acid sequence 43 - 101 of SEQ ID NO: 10 in its genome and wherein the expression of the male fertility gene confers the dominant male sterility trait.
  • An isolated nucleic acid molecule includes a polynucleotide capable of initiating transcription in a plant cell and includes a sequence selected from the group consisting of: SEQ ID NO: 15; at least 100 contiguous nucleotides of SEQ ID NO: 15 and a sequence having at least 70% sequence identity to the full length of SEQ ID NO: 15.
  • an expression cassette or a vector includes SEQ ID NO: 15 disclosed herein operably linked to a polynucleotide of interest.
  • Suitable plants for certain embodiments disclosed herein include e.g., maize
  • corn (corn), sorghum, canola, wheat, barley, rye, triticale, rice, sugar cane, turfgrass, pearl millet, soybeans, cotton, and sunflower.
  • a plant with reduced fertility or any other trait disclosed herein optionally comprises one or more polynucleotides conferring the following phenotype or trait of interest: improved nutrient uptake, nitrogen use efficiency, drought tolerance, root strength, root lodging resistance, soil pest management, corn root worm resistance, herbicide tolerance, disease resistance, or insect resistance, or altered carbohydrate metabolism, protein metabolism, fatty acid metabolism or phytohormone biosynthesis.
  • a method of increasing yield or maintaining yield stability in plants includes reducing male reproductive tissue development by expressing a transgene under the control of a male reproductive tissue preferred promoter; and increasing nutrient allocation to female reproductive tissue during concurrent male and female tissue development.
  • the male reproductive tissue is tassel.
  • the male reproductive tissue development is decreased by the expression of a gene operably linked to a promoter comprising at least 100 contiguous nucleotides of a sequence selected from the list SEQ ID NO: 64 - 106.
  • a promoter comprising at least 100 contiguous nucleotides of a sequence selected from the list SEQ ID NO: 64 - 106.
  • Subsets of the promoter sequences disclosed herein e.g., SEQ ID NOS: 64-70; 70-75; 75-80; 85-90; 90-95; 100-106 are also suitable for driving tissue-preferred expression of the polynucleotides of interest disclosed herein.
  • a plant or a plant cell or a seed that transgenically expresses a polynucleotide of interest e.g., Ms44 having the dominant male sterility mutation
  • a polynucleotide of interest e.g., Ms44 having the dominant male sterility mutation
  • a tassel-preferred promoter disclosed herein exhibits improved agronomic parameters such as increased nutrient allocation to ears during reproductive development.
  • a plant or a plant cell or a seed that transgenically expresses a polynucleotide of interest e.g., RNAi suppression sequence targeting a polynucleotide involved in tassel development
  • a polynucleotide of interest e.g., RNAi suppression sequence targeting a polynucleotide involved in tassel development
  • exhibits increased agronomic parameters such as improved nutrient allocation to ears during reproductive development and/or increased grain yield relative to a control.
  • a method of increasing yield or maintaining yield stability in plants includes reducing male fertility and increasing nutrient allocation to female reproductive tissue during concurrent male and female tissue development.
  • the male fertility is reduced in a plant by altering expression of a genetic male fertility gene.
  • the plant is grown under stress.
  • the plant is grown under nutrient limiting conditions, e.g., reduced available nitrogen.
  • the plants with reduced male fertility and wherein the nutrient is allocated more to female reproductive tissue during concurrent male and female tissue development exhibits one or more of the following agronomically relevant parameters: increased SPAD value (chlorophyll measurement); increased silk emergence rate or number or degree; increased ear length; increased ear width; increased seed number per ear; increased seed weight per ear and increased embryo size.
  • the plants with reduced male fertility and wherein the nutrient is allocated more to female reproductive tissue during concurrent male and female tissue are grown under drought stress. In an embodiment, drought tolerance of the plants is improved by male sterility.
  • An isolated nucleic acid molecule comprising a polynucleotide which initiates transcription in a plant cell in a tissue preferred manner and includes a sequence selected from the group consisting of:
  • a method of increasing yield stability in plants under stress includes expressing an element that affects male fertility operably linked to a tassel preferred promoter disclosed herein and thereby reducing the competition for nutrients during the reproductive development phase of the plant and wherein the yield is increased.
  • a method of increasing yield or maintaining yield stability in plants under nitrogen limiting conditions and/or normal nitrogen conditions includes reducing male reproductive tissue development and increasing nutrient allocation to female reproductive tissue during concurrent male and female tissue development.
  • the male reproductive tissue is tassel and the male reproductive tissue development is decreased by reducing the expression of a NIP3-1 or a Nl P3-1 -like protein.
  • NIP3-1 protein has an amino acid sequence of SEQ ID NO: 107. The male reproductive tissue development is decreased by increasing the expression of SEQ ID NO: 63.
  • the male reproductive tissue development is decreased by affecting the function of a gene involved in tassel formation, e.g., tassel-less gene.
  • the male reproductive tissue development is decreased in a plant transformed with an expression cassette that targets for suppression a gene encoding amino acid sequence of SEQ ID NO: 107 or a sequence that is at least 70% or 80% or 85% or 90% or 95% identical to SEQ ID NO: 107.
  • Plants with native mutations in the Tls1 allele are also disclosed herein.
  • a tissue-preferred promoter preferentially drives expression of a gene of interest in male reproductive tissue.
  • the promoter is a tissue-specific promoter, a constitutive promoter or an inducible promoter.
  • the tissue-preferred promoter is a tassel-preferredpromoter.
  • An isolated nucleic acid molecule comprising a polynucleotide that includes a sequence selected from the group consisting of: SEQ ID NO: 63; at least 100 contiguous nucleotides of SEQ ID NO: 63 and a sequence having at least 70% sequence identity to the full length of SEQ ID NO: 63.
  • An isolated nucleic acid molecule comprising a polynucleotide that encodes the TLS1 protein comprising an amino acid sequence of SEQ ID NO: 107 or a sequence that is at least 70% or 80% or 85% or 90% or 95% identical to SEQ ID NO: 107.
  • a method for producing male sterile hybrid seeds includes transforming an inbred line that is heterozygous for dominant male sterility with a gene construct that includes a first element that suppresses the dominant male sterility phenotype, a second element that disrupts pollen function, and optionally a marker, which may be a selectable or screenable marker, wherein expressing the construct in the inbred line renders the line male fertile; this line is referred to as the transgenic maintainer line.
  • this method further includes self-pollinating plants of the transgenic maintainer line to produce progeny that are homozygous dominant male sterile.
  • the method further includes identifying those seeds having the homozygous dominant male sterility genotypes as the female inbred line.
  • the female inbred line is; optionally increased by pollinating it with the transgenic maintainer line, resulting in 100% homozygous dominant male sterile seed lacking the construct.
  • the method further includes pollinating plants grown from the dominant male sterile seed with a male parent, to produce hybrids that are heterozygous for dominant male sterility and display the dominant male sterile phenotype.
  • the dominant male sterility phenotype is conferred by a polynucleotide sequence that includes at least 100 consecutive nucleotides of SEQ ID NO: 15 and further encodes a Threonine instead of an Alanine at position 37 of SEQ ID NO: 14 (the amino acid sequence encoded by SEQ ID NO: 15).
  • the suppression element includes a promoter inverted repeat (pIR) sequence specific to SEQ ID NO: 9 or SEQ ID NO: 13, or to a promoter operably linked to SEQ ID NO: 15 or SEQ ID NO: 152.
  • the inverted repeat sequence includes a fragment of at least 100 consecutive nucleotides of SEQ ID NO: 9 or SEQ ID NO: 13.
  • the suppression element is a RNAi construct designed to suppress the expression of the dominant Ms44 gene in the male sterile female inbred line and may target SEQ ID NO: 15 or SEQ ID NO: 152.
  • the suppression element is a genetic suppressor that acts in a dominant fashion to suppress the dominant sterile phenotype of Ms44 mutation in a plant.
  • the construct may include an element that restores the normal function of the ms44 gene, e.g., ms44 gene under the control of its own promoter or a heterologous promoter.
  • a plant or a plant cell or a seed or a progeny of the plant derived from the methods disclosed herein is disclosed.
  • a method for producing hybrid seeds includes expressing in an inbred a dominant male sterility gene operably linked to a heterologous promoter amenable to inverted-repeat inactivation; and pollinating this male sterile plant with pollen from a male fertile plant containing an inverted repeat specific to the heterologous promoter.
  • the pollen comprises the inverted repeat specific to the heterologous promoter with inverted repeat inactivation specificity.
  • the dominant male sterility gene is linked to a rice 5126 promoter.
  • the dominant male sterility gene used in the context of hybrid seed production is any gene that acts in a dominant manner to achieve male sterility and optionally is amenable to suppression to maintain the male-sterile female inbred line.
  • the dominant male sterility gene is selected from the group comprising: barnase, DAM methylase, streptavidin, MS41 and MS42.
  • Figure 1 Diagram of Genetic Dominant Male Sterility system to produce a male- sterile hybrid plant.
  • Genetic reduction of male fertility in a plant which may utilize one or more of a dominant nuclear male-sterile gene, a tassel-specific or tassel-preferred promoter, and a tassel-specific or tassel-preferred gene, has been found to increase ear tissue development, improve nutrient utilization in the growing plant, increase stress tolerance, and/or increase seed metrics, ultimately leading to improved yield.
  • Figure 2 Alignment of MS44 related sequences ( Figure 2 A - C). The identical residues are in bold and all similar residues are underlined and italicized.
  • Figure 3 Diagram of method to produce a male-sterile hybrid plant using a recessive male-sterile gene. Both the female parent and the male parent have the homozygous recessive alleles which confer sterility. However, the male parent carries the restorer allele within a construct which prevents transmission of the restorer allele through pollen. Resulting hybrid seed produce a male-sterile hybrid plant.
  • Figure 4 Diagram of method for producing male sterile hybrid plant using a dominant male-sterility gene:
  • SAM suppression, ablation, marker
  • 4C and 4D - Seeds or progeny plants are phenotyped and/or genotyped to identify those which are homozygous for dominant male sterility and are further characterized as carrying or lacking the SAM construct
  • the male-sterile inbred line can be increased by pollinating it with the transgenic maintainer line, resulting in seed which are all homozygous dominant male sterile and lack the SAM construct.
  • 4F Dominant male sterile inbred plants are pollinated by a second inbred to produce hybrids that are heterozygous for dominant male sterility and exhibit the dominant male sterile phenotype.
  • Male-sterile hybrid seed are seed that when planted will produce a hybrid plant which is male-sterile.
  • Figure 5 - Figure 5A shows MS44 hybrid yield response to N fertility - Trial 1 .
  • Figure 5B shows MS44 hybrid yield response to N fertility - Trial 2.
  • Figure 6 shows MS44 hybrid ear dry weight (R1 stage) as compared to wild-type.
  • Figure 7 - Figure 7A shows MS44 hybrid yield response to plant population- Trial 1.
  • Figure 7B shows MS44 hybrid yield response to plant population - Trial 2.
  • Figure 8 shows the tlsl mutant phenotype.
  • F Range of leaf phenotypes.
  • WT homozygous wild type plant
  • ST homozygous tlsl plant with a small tassel
  • NT homozygous tlsl plant with no tassel.
  • Figure 9 shows the map-based cloning of tlsl .
  • Figure 10 shows tlsl candidate gene validation. Knockout of ZmNIP3.1 results in tlsl phenotype.
  • Figure 10A Wild type plant with intact ZmNIP3;1 .
  • Figure 10B Plant with Mu-insertion in ZmNIP3.1 exhibits tlsl phenotype.
  • Figure 1 1 shows the tassel branch number in mutant, wild-type and mutant sprayed with boron.
  • Figure 12 shows the tassel branch length in mutant, wild-type and mutant sprayed with boron.
  • Figure 13 shows the ear length in mutant, wild-type and mutant sprayed with boron.
  • Figure 14 shows that tlsl plants are less susceptible to boron-toxic conditions of
  • Figure 14A Side-by-side of homozygous tlsl and wild type plants with mutant plants appearing taller and larger.
  • Figure 14B In wild type plants, the node of the second youngest fully expanded leaf extends above the node of the youngest fully expanded leaf, whereas mutant plants appear normal.
  • Figure 14 C Youngest fully expanded leaf of mutant is broader than wild type.
  • Figure 15 shows ZmNIP3.1 is similar to boron channel proteins.
  • Figure 15 A Phylogenetic tree shows ZmNIP3.1 is closely related to OsNIP3.1 and AtNIP5.1 (highlighted), which have been characterized as boron channel proteins.
  • Figure 15B Alignment of protein sequences highlighted in 15A where ZmNIP3.1 is 84.4 and 67.3 percent identical to OsNIP3.1 and AtNIP5.1 respectively.
  • Figure 16 Ms44 sequences from selected species.
  • the amino acid mutation for the Ms44 Dominant polypeptide sequence is indicated in bold and underlined in position 42, as T in the MS44dom allele (SEQ ID NO: 14) or V in the Ms44- 2629 allele (SEQ ID NO: 153), where all other sequences have A at that position.
  • the developing female reproductive structure competes with male reproductive structures for nitrogen, carbon and other nutrients during development of these reproductive structures. This is demonstrated in quantifying the nitrogen budget of developing maize ears and tassels when the plants are grown in increasing levels of nitrogen fertilizer.
  • the nitrogen budget of the ear is negative, or during development the ear loses nitrogen to other parts of the plant when nitrogen is limiting.
  • the nitrogen budget of the ear improves as the amount of nitrogen fertilizer provided to the plant increases until the ear maintains a positive increase in nitrogen through to silk emergence.
  • the tassel maintains a positive nitrogen budget irrespective of the level of fertility in which the plant is grown.
  • the tassel and ear compete for nitrogen during reproductive development and the developing tassel dominates over the developing ear.
  • the ear and tassel likely compete for a number of nutrients during development and the competition becomes more severe under stress conditions.
  • the ear is in competition with the tassel during reproductive development prior to anthesis, reducing the ability of the developing ear to accumulate nutrients under stress, resulting in a smaller, less developed ear with fewer kernels. More severe, extended stress can result in failure of the ear to exert silks and produce grain. Genetic reduction in male fertility would reduce the nutrient requirement for tassel development, resulting in improved ear development at anthesis.
  • MS44 a dominant genetic male sterile
  • MS44 only needs to be backcrossed into the female parent to produce hybrid seed segregating 1 :1 for male sterility.
  • Dominant male sterility is especially useful in polyploid plants such as wheat, where maintenance of homozygous recessive sterility is more complex.
  • CMS-derived male sterility is not determined until 10 days prior to anthesis, as judged by the environmental interactions associated with CMS stability (Weider, et al., (2009) Crop Sci. 49:77-84). The bulk of ear development would have already occurred prior to 10 days before anthesis, thus CMS-derived sterility would provide little relief from tassel competition during ear development. In contrast, early failure of male reproductive tissue development in genetic male sterility reduces competition between developing ear and developing tassel for nutrients when the ear is in early stages of development. Yield improvements associated with male sterile hybrids vectored through improved ear development are consistent with the reduction in competition of ear development with tassel development.
  • CMS cytoplasmic male sterile
  • Relieving competition between developing tassel and ear could also be achieved by chemically induced male sterility. A combination of chemicals and genetic manipulation could also induce male sterility.
  • Herbicide tolerance modulated by promoters with less efficacy in male reproductive tissue or the use of pro-gametocides ((Dotson, et al., (1996) The Plant Journal 10:383-392) and (Mayer and Jefferson, (2004) Molecular Methods for Hybrid Rice Production. A report for the rural Industries Research and Development Corporation)) to release male inhibitors in a tissue specific manner would also be effective means of practicing this disclosure.
  • a particular plant trait is expressed by maintenance of a homozygous recessive condition. Difficulties arise in maintaining the homozygous condition when a restoration gene must be used for maintenance.
  • the MS45 gene in maize (US Patent Number 5,478,369) has been shown to be critical to male fertility. Plants heterozygous or hemizygous for the dominant MS45 allele are fully fertile due to the sporophytic nature of the MS45 fertility trait. A natural mutation in the MS45 gene, designated ms45, imparts a male sterility phenotype to plants when this mutant allele is in the homozygous state.
  • This sterility can be reversed (i.e., fertility restored) when the non-mutant form of the gene is introduced into the plant, either through normal crossing or transgenic complementation methods.
  • restoration of fertility by crossing removes the desired homozygous recessive condition, and both methods restore full male fertility and prevent maintenance of pure male sterile maternal lines.
  • a method to maintain the desired homozygous recessive condition is described in US Patent Numbers 7,696,405 and7,893,317, where a maintainer line is used to cross onto homozygous recessive male sterile siblings.
  • the maintainer line is in the desired homozygous recessive condition for male sterility but also contains a hemizygous transgenic construct consisting of a dominant male fertility gene to complement the male sterility condition; a pollen ablation gene, which results in disruption of the formation, function, or dispersal of pollen and thereby prevents the transfer through pollen of the transgenic construct to the male sterile sibling but allows for the transfer of the recessive male sterile allele through the non-transgenic pollen grains; and a seed marker gene which allows for the sorting of transgenic maintainer seeds or plants and transgenic-null male sterile seeds or plants.
  • SPT Seed Production Technology
  • SPT provides methods to maintain the homozygous recessive condition of a male-sterility gene in a plant. See, for example, US Patent Number 7,696,405.
  • SPT utilizes a maintainer line that is the pollen source for fertilization of its homozygous-recessive male-sterile siblings.
  • the maintainer line is in the desired homozygous recessive condition for male sterility but also contains a hemizygous transgenic construct (the "SPT construct").
  • the SPT construct comprises the following three elements: (1 ) a dominant male-fertility gene to complement the male-sterile recessive condition; (2) a gene encoding a product which interferes with the formation, function, or dispersal of male gametes and (3) a marker gene which allows for the sorting of transgenic maintainer seeds/plants from those which lack the transgene. Interference with pollen formation, function or dispersal prevents the transfer through pollen of the transgenic construct; functional pollen lacks the transgene. Resulting seeds produce plants which are male-sterile.
  • male-sterile inbred plants are then used in hybrid production by pollinating with a male parent, which may be an unrelated inbred line homozygous for the dominant allele of the male-fertility gene. Resulting hybrid seeds produce plants which are male-fertile.
  • the male parent would serve as the maintainer line to cross onto male sterile female inbreds, (increased using a separate maintainer line), to give fully male sterile hybrid plants. See, for example, Figure 3.
  • a dominant male sterility approach has advantages over the use of recessive male sterility because only a single copy of the dominant gene is required for full sterility.
  • methods are not available to create a homozygous dominant male sterile line, then resulting progeny will segregate 50% for male sterility. Practically, this situation can be alleviated by transgenically linking a screenable or selectable marker to the dominant male sterility gene and screening or selecting progeny seeds or plants carrying the marker.
  • linked genetic markers or a linked phenotype could be employed to sort progeny.
  • An auto splicing protein sequence is a segment of a protein that is able to excise itself and rejoin the remaining portion/s with a peptide bond. Auto splicing protein sequences can self splice and re-ligate the remaining portions in both cis and trans states.
  • a dominant male sterile gene could be modified such that the regions coding for the N and C protein regions are separated into different transgenic constructs, coupled with a sequence coding for an auto splicing protein sequence.
  • a plant containing a single construct would be male fertile since the protein is truncated and non-functional, which allows for self fertilization to create a homozygous plant. Plants homozygous for the N-DMS-N-auto splicing protein sequence can then be crossed with plants homozygous for the C-auto splicing protein sequence-C-DMS protein. All of the progeny from this cross would be male sterile through the excision of each auto splicing protein sequence and the re-ligation of the N and C sequences to create a functional dominant male sterile protein.
  • Maize ear and tassel are both inflorescence structures that share common development processes and are controlled by a common set of genes. The tissues compete with each other for the required nutrients. Tassel however has the advantage of apical dominance over the ear, which is unfavorable to ear growth and yield potential in the maize plants. Reducing the tassel apical dominance could be used to divert more resource to the ear growth, kernel number or size and ultimately can lead to increased grain yield.
  • tassel elimination a tasseless maize plant
  • genetic mutations (mutants) of genes such as male sterility genes can be used to reduce the competition of the tassel with ear
  • transgenic manipulation offers alternatives or enabling tools for this purpose.
  • genes that are involved in tassel development are often involved in ear development, reducing tassel development by interrupting these genes may also affect the ear development.
  • the tasseless gene (Tsl1 ) mutation is an example, in which the tasseless plant is also earless.
  • a tassel-specific promoter is needed to target the gene disruption in the tassel tissues only.
  • nucleic acids are written left to right in 5' to 3' orientation; amino acid sequences are written left to right in amino to carboxy orientation, respectively. Numeric ranges are inclusive of the numbers defining the range. Amino acids may be referred to herein by either their commonly known three letter symbols or by the one-letter symbols recommended by the lUPAC-IUB Biochemical Nomenclature Commission. Nucleotides, likewise, may be referred to by their commonly accepted single-letter codes. The terms defined below are more fully defined by reference to the specification as a whole.
  • microbe any microorganism (including both eukaryotic and prokaryotic microorganisms), such as fungi, yeast, bacteria, actinomycetes, algae and protozoa, as well as other unicellular structures.
  • amplified is meant the construction of multiple copies of a nucleic acid sequence or multiple copies complementary to the nucleic acid sequence using at least one of the nucleic acid sequences as a template.
  • Amplification systems include the polymerase chain reaction (PCR) system, ligase chain reaction (LCR) system, nucleic acid sequence based amplification (NASBA, Cangene, Mississauga, Ontario), Q-Beta Replicase systems, transcription-based amplification system (TAS) and strand displacement amplification (SDA). See, e.g., Diagnostic Molecular Microbiology: Principles and Applications, Persing, et al., eds., American Society for Microbiology, Washington, DC (1993). The product of amplification is termed an amplicon.
  • conservatively modified variants refer to those nucleic acids that encode identical or conservatively modified variants of the amino acid sequences. Because of the degeneracy of the genetic code, a large number of functionally identical nucleic acids encode any given protein. For instance, the codons GCA, GCC, GCG and GCU all encode the amino acid alanine. Thus, at every position where an alanine is specified by a codon, the codon can be altered to any of the corresponding codons described without altering the encoded polypeptide. Such nucleic acid variations are "silent variations" and represent one species of conservatively modified variation.
  • Every nucleic acid sequence herein that encodes a polypeptide also describes every possible silent variation of the nucleic acid.
  • AUG which is ordinarily the only codon for methionine; one exception is Micrococcus rubens, for which GTG is the methionine codon (Ishizuka, et al. , (1993) J. Gen. Microbiol. 139:425- 32)
  • GTG is the methionine codon
  • amino acid sequences one of skill will recognize that individual substitution, deletion or addition to a nucleic acid, peptide, polypeptide or protein sequence which alters, adds or deletes a single amino acid or a small percentage of amino acids in the encoded sequence is a "conservatively modified variant" when the alteration results in the substitution of an amino acid with a chemically similar amino acid.
  • any number of amino acid residues selected from the group of integers consisting of from 1 to 15 can be so altered.
  • 1 , 2, 3, 4, 5, 7 or 10 alterations can be made.
  • Conservatively modified variants typically provide similar biological activity as the unmodified polypeptide sequence from which they are derived.
  • substrate specificity, enzyme activity or ligand/receptor binding is generally at least 30%, 40%, 50%, 60%, 70%, 80% or 90%, preferably 60-90% of the native protein for its native substrate.
  • Conservative substitution tables providing functionally similar amino acids are well known in the art.
  • construct is used to refer generally to an artificial combination of polynucleotide sequences, i.e. a combination which does not occur in nature, normally comprising one or more regulatory elements and one or more coding sequences.
  • the term may include reference to expression cassettes and/or vector sequences, as is appropriate for the context.
  • a "control” or “control plant” or “control plant cell” provides a reference point for measuring changes in phenotype of a subject plant or plant cell in which genetic alteration, such as transformation, has been effected as to a gene of interest.
  • a subject plant or plant cell may be descended from a plant or cell so altered and will comprise the alteration.
  • a control plant or plant cell may comprise, for example: (a) a wild-type plant or cell, i.e., of the same genotype as the starting material for the genetic alteration which resulted in the subject plant or cell; (b) a plant or plant cell of the same genotype as the starting material but which has been transformed with a null construct (i.e., with a construct which has no known effect on the trait of interest, such as a construct comprising a marker gene); (c) a plant or plant cell which is a non-transformed segregant among progeny of a subject plant or plant cell; (d) a plant or plant cell genetically identical to the subject plant or plant cell but which is not exposed to conditions or stimuli that would induce expression of the gene of interest; or (e) the subject plant or plant cell itself, under conditions in which the gene of interest is not expressed.
  • a control plant may also be a plant transformed with an alternative construct.
  • nucleic acid encoding a protein may comprise non-translated sequences (e.g., introns) within translated regions of the nucleic acid or may lack such intervening non-translated sequences (e.g., as in cDNA).
  • the information by which a protein is encoded is specified by the use of codons.
  • amino acid sequence is encoded by the nucleic acid using the "universal" genetic code.
  • variants of the universal code such as is present in some plant, animal and fungal mitochondria, the bacterium Mycoplasma capricolum (Yamao, et al., (1985) Proc. Natl. Acad. Sci. USA 82:2306-9) or the ciliate Macronucleus, may be used when the nucleic acid is expressed using these organisms.
  • nucleic acid sequences of the present disclosure may be expressed in both monocotyledonous and dicotyledonous plant species, sequences can be modified to account for the specific codon preferences and GC content preferences of monocotyledonous plants or dicotyledonous plants as these preferences have been shown to differ (Murray, et al., (1989) Nucleic Acids Res. 17:477-98 and herein incorporated by reference).
  • the maize preferred codon for a particular amino acid might be derived from known gene sequences from maize.
  • Maize codon usage for 28 genes from maize plants is listed in Table 4 of Murray, et al., supra.
  • endogenous when used in reference to a gene, means a gene that is normally present in the genome of cells of a specified organism and is present in its normal state in the cells (i.e., present in the genome in the state in which it normally is present in nature).
  • exogenous is used herein to refer to any material that is introduced into a cell.
  • exogenous nucleic acid molecule or “transgene” refers to any nucleic acid molecule that either is not normally present in a cell genome or is introduced into a cell.
  • exogenous nucleic acid molecules generally are recombinant nucleic acid molecules, which are generated using recombinant DNA methods as disclosed herein or otherwise known in the art.
  • a transgenic non-human organism as disclosed herein can contain, for example, a first transgene and a second transgene.
  • Such first and second transgenes can be introduced into a cell, for example, a progenitor cell of a transgenic organism, either as individual nucleic acid molecules or as a single unit (e.g., contained in different vectors or contained in a single vector, respectively).
  • a cell from which the transgenic organism is to be derived contains both of the transgenes using routine and well-known methods such as expression of marker genes or nucleic acid hybridization or PCR analysis.
  • confirmation of the presence of transgenes may occur later, for example, after regeneration of a plant from a putatively transformed cell.
  • heterologous in reference to a nucleic acid is a nucleic acid that originates from a foreign species, or, if from the same species, is substantially modified from its native form in composition and/or genomic locus by deliberate human intervention.
  • a promoter operably linked to a heterologous structural gene is from a species different from that from which the structural gene was derived or, if from the same species, one or both are substantially modified from their original form.
  • a heterologous protein may originate from a foreign species or, if from the same species, is substantially modified from its original form by deliberate human intervention.
  • host cell is meant a cell which comprises a heterologous nucleic acid sequence of the disclosure, which contains a vector and supports the replication and/or expression of the expression vector.
  • Host cells may be prokaryotic cells such as E. coli, or eukaryotic cells such as yeast, insect, plant, amphibian or mammalian cells.
  • host cells are monocotyledonous or dicotyledonous plant cells, including but not limited to maize, sorghum, sunflower, soybean, wheat, alfalfa, rice, cotton, canola, barley, millet and tomato.
  • a particularly preferred monocotyledonous host cell is a maize host cell.
  • hybridization complex includes reference to a duplex nucleic acid structure formed by two single-stranded nucleic acid sequences selectively hybridized with each other.
  • the term "introduced” in the context of inserting a nucleic acid into a cell means “transfection” or “transformation” or “transduction” and includes reference to the incorporation of a nucleic acid into a eukaryotic or prokaryotic cell where the nucleic acid may be incorporated into the genome of the cell (e.g., chromosome, plasmid, plastid or mitochondrial DNA), converted into an autonomous replicon or transiently expressed (e.g., transfected mRNA).
  • isolated refers to material, such as a nucleic acid or a protein, which is substantially or essentially free from components which normally accompany or interact with it as found in its naturally occurring environment.
  • non-naturally occurring ; “mutated”, “recombinant”; “recombinantly expressed”; “heterologous” or “heterologously expressed” are representative biological materials that are not present in its naturally occurring environment.
  • a "line” is a collection of genetically identical plants and encompasses the seeds that produce such plants.
  • An inbred line is typically homozygous at most or all loci.
  • NUE nucleic acid means a nucleic acid comprising a polynucleotide
  • NUE polynucleotide encoding a full length or partial length polypeptide.
  • nucleic acid includes reference to a deoxyribonucleotide or ribonucleotide polymer in either single- or double-stranded form, and unless otherwise limited, encompasses known analogues having the essential nature of natural nucleotides in that they hybridize to single-stranded nucleic acids in a manner similar to naturally occurring nucleotides (e.g., peptide nucleic acids).
  • nucleic acid library is meant a collection of isolated DNA or RNA molecules, which comprise and substantially represent the entire transcribed fraction of a genome of a specified organism. Construction of exemplary nucleic acid libraries, such as genomic and cDNA libraries, is taught in standard molecular biology references such as Berger and Kimmel, (1987) Guide To Molecular Cloning Techniques, from the series Methods in
  • operably linked includes reference to a functional linkage between a first sequence, such as a promoter, and a second sequence, wherein the promoter sequence initiates and mediates transcription of the DNA corresponding to the second sequence.
  • operably linked means that the nucleic acid sequences being linked are contiguous and, where necessary to join two protein coding regions, contiguous and in the same reading frame.
  • plant includes reference to whole plants, plant organs
  • Plant cell includes, without limitation, a cell present in or isolated from plant tissues including seeds, suspension cultures, embryos, meristematic regions, callus tissue, leaves, roots, shoots, gametophytes, sporophytes, pollen and microspores.
  • the class of plants which can be used in the methods of the disclosure is generally as broad as the class of higher plants amenable to transformation techniques, including both monocotyledonous and dicotyledonous plants including species from the genera: Cucurbita, Rosa, Vitis, Juglans, Fragaria, Lotus, Medicago, Onobrychis, Trifolium, Trigonella, Vigna, Citrus, Linum, Geranium, Manihot, Daucus, Arabidopsis, Brassica, Raphanus, Sinapis, Atropa, Capsicum, Datura, Hyoscyamus, Lycopersicon, Nicotiana, Solarium, Petunia, Digitalis, Majorana, Ciahorium, Helianthus, Lactuca, Bromus, Asparagus, Antirrhinum, Heterocallis, Nemesis, Pelargonium, Panieum, Pennisetum, Ranunculus, Senecio, Salpiglossis, Cucumis, Brow
  • yield may include reference to bushels per acre of a grain crop at harvest, as adjusted for grain moisture (15% typically for maize, for example) and/or the volume of biomass generated (for forage crops such as alfalfa and plant root size for multiple crops). Grain moisture is measured in the grain at harvest. The adjusted test weight of grain is determined to be the weight in pounds per bushel, adjusted for grain moisture level at harvest. Biomass is measured as the weight of harvestable plant material generated.
  • polynucleotide includes reference to a deoxyribopolynucleotide, ribopolynucleotide or analogs thereof that have the essential nature of a natural ribonucleotide in that they hybridize, under stringent hybridization conditions, to substantially the same nucleotide sequence as naturally occurring nucleotides and/or allow translation into the same amino acid(s) as the naturally occurring nucleotide(s).
  • a polynucleotide can be full-length or a subsequence of a native or heterologous structural or regulatory gene. Unless otherwise indicated, the term may include reference to the specified sequence as well as the complementary sequence thereof.
  • polypeptide peptide
  • protein protein
  • 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 naturally occurring amino acid polymers.
  • promoter includes reference to a region of DNA upstream from the start of transcription and involved in recognition and binding of RNA polymerase and other proteins to initiate transcription.
  • a "plant promoter” is a promoter capable of initiating transcription in plant cells. Exemplary plant promoters include, but are not limited to, those that are obtained from plants, plant viruses and bacteria which comprise genes expressed in plant cells such as Agrobacterium or Rhizobium. Examples are promoters that preferentially initiate transcription in certain tissues, such as leaves, roots, seeds, fibres, xylem vessels, tracheids or sclerenchyma.
  • tissue preferred Such promoters are referred to as "tissue preferred.”
  • a "cell type” specific promoter primarily drives expression in certain cell types in one or more organs, for example, vascular cells in roots or leaves.
  • An “inducible” or “regulatable” promoter is a promoter which is under environmental control. Examples of environmental conditions that may effect transcription by inducible promoters include anaerobic conditions or the presence of light.
  • Another type of promoter is a developmental ⁇ regulated promoter, for example, a promoter that drives expression during pollen development.
  • Tissue preferred, cell type specific, developmental ⁇ regulated and inducible promoters are members of the class of "non-constitutive" promoters.
  • a “constitutive” promoter is a promoter which is active in essentially all tissues of a plant, under most environmental conditions and states of development or cell differentiation.
  • polypeptide refers to one or more amino acid sequences. The term is also inclusive of fragments, variants, homologs, alleles or precursors (e.g., preproproteins or proproteins) thereof.
  • a “NUE protein” comprises a polypeptide.
  • NUE nucleic acid means a nucleic acid comprising a polynucleotide (“NUE polynucleotide”) encoding a polypeptide.
  • recombinant includes reference to a cell or vector, that has been modified by the introduction of a heterologous nucleic acid or that the cell is derived from a cell so modified.
  • recombinant cells express genes that are not found in identical form within the native (non-recombinant) form of the cell or express native genes that are otherwise abnormally expressed, under expressed or not expressed at all as a result of deliberate human intervention or may have reduced or eliminated expression of a native gene.
  • the term “recombinant” as used herein does not encompass the alteration of the cell or vector by naturally occurring events (e.g., spontaneous mutation, natural transformation/transduction/transposition) such as those occurring without deliberate human intervention.
  • a "recombinant expression cassette” is a nucleic acid construct, generated recombinantly or synthetically, with a series of specified nucleic acid elements, which permit transcription of a particular nucleic acid in a target cell.
  • the recombinant expression cassette can be incorporated into a plasmid, chromosome, mitochondrial DNA, plastid DNA, virus or nucleic acid fragment.
  • the recombinant expression cassette portion of an expression vector includes, among other sequences, a nucleic acid to be transcribed and a promoter.
  • sequences include reference to hybridization, under stringent hybridization conditions, of a nucleic acid sequence to a specified nucleic acid target sequence to a detectably greater degree (e.g., at least 2-fold over background) than its hybridization to non-target nucleic acid sequences and to the substantial exclusion of non-target nucleic acids.
  • Selectively hybridizing sequences typically have about at least 40% sequence identity, preferably 60-90% sequence identity and most preferably 100% sequence identity (i.e., complementary) with each other.
  • stringent conditions or “stringent hybridization conditions” include reference to conditions under which a probe will hybridize to its target sequence, to a detectably greater degree than other sequences (e.g., at least 2-fold over background). Stringent conditions are sequence-dependent and will be different in different circumstances. By controlling the stringency of the hybridization and/or washing conditions, target sequences can be identified which can be up to 100% complementary to the probe (homologous probing). Alternatively, stringency conditions can be adjusted to allow some mismatching in sequences so that lower degrees of similarity are detected (heterologous probing). Optimally, the probe is approximately 500 nucleotides in length, but can vary greatly in length from less than 500 nucleotides to equal to the entire length of the target sequence.
  • stringent conditions will be those in which the salt concentration is less than about 1.5 M Na ion, typically about 0.01 to 1 .0 M Na ion concentration (or other salts) at pH 7.0 to 8.3 and the temperature is at least about 30°C for short probes (e.g., 10 to 50 nucleotides) and at least about 60°C for long probes (e.g., greater than 50 nucleotides).
  • Stringent conditions may also be achieved with the addition of destabilizing agents such as formamide or Denhardt's.
  • Exemplary moderate stringency conditions include hybridization in 40 to 45% formamide, 1 M NaCI, 1 % SDS at 37°C and a wash in 0.5X to 1X SSC at 55 to 60°C.
  • Exemplary high stringency conditions include hybridization in 50% formamide, 1 M NaCI, 1 % SDS at 37°C and a wash in 0.1 X SSC at 60 to 65°C.
  • T m 81.5°C + 16.6 (log M) + 0.41 (%GC) - 0.61 (% form) - 500/L; where M is the molarity of monovalent cations, %GC is the percentage of guanosine and cytosine nucleotides in the DNA, % form is the percentage of formamide in the hybridization solution, and L is the length of the hybrid in base pairs.
  • the T m is the temperature (under defined ionic strength and pH) at which 50% of a complementary target sequence hybridizes to a perfectly matched probe. T m is reduced by about 1 °C for each 1 % of mismatching; thus, T m , hybridization and/or wash conditions can be adjusted to hybridize to sequences of the desired identity. For example, if sequences with >90% identity are sought, the T m can be decreased 10°C.
  • stringent conditions are selected to be about 5°C lower than the thermal melting point (T m ) for the specific sequence and its complement at a defined ionic strength and pH.
  • high stringency is defined as hybridization in 4X SSC, 5X Denhardt's (5 g Ficoll, 5 g polyvinypyrrolidone, 5 g bovine serum albumin in 500ml of water), 0.1 mg/ml boiled salmon sperm DNA, and 25 mM Na phosphate at 65°C and a wash in 0.1 X SSC, 0.1 % SDS at 65°C.
  • transgenic plant includes reference to a plant which comprises within its genome a heterologous polynucleotide.
  • the heterologous polynucleotide is stably integrated within the genome such that the polynucleotide is passed on to successive generations.
  • the heterologous polynucleotide may be integrated into the genome alone or as part of a recombinant expression cassette.
  • Transgenic is used herein to include any cell, cell line, callus, tissue, plant part or plant, the genotype of which has been altered by the presence of heterologous nucleic acid including those transgenics initially so altered as well as those created by sexual crosses or asexual propagation from the initial transgenic.
  • transgenic does not encompass the alteration of the genome (chromosomal or extra-chromosomal) by conventional plant breeding methods or by naturally occurring events such as random cross-fertilization, non-recombinant viral infection, non-recombinant bacterial transformation, non-recombinant transposition or spontaneous mutation.
  • vector includes reference to a nucleic acid used in transfection of a host cell and into which can be inserted a polynucleotide. Vectors are often replicons. Expression vectors permit transcription of a nucleic acid inserted therein.
  • sequence relationships between two or more nucleic acids or polynucleotides or polypeptides are used to describe the sequence relationships between two or more nucleic acids or polynucleotides or polypeptides: (a) “reference sequence,” (b) “comparison window,” (c) “sequence identity,” (d) “percentage of sequence identity” and (e) “substantial identity.”
  • reference sequence is a defined sequence used as a basis for sequence comparison.
  • a reference sequence may be a subset or the entirety of a specified sequence; for example, as a segment of a full-length cDNA or gene sequence or the complete cDNA or gene sequence.
  • comparison window means includes reference to a contiguous and specified segment of a polynucleotide sequence, wherein the polynucleotide sequence may be compared to a reference sequence and wherein the portion of the polynucleotide sequence in the comparison window may comprise additions or deletions (i.e., gaps) compared to the reference sequence (which does not comprise additions or deletions) for optimal alignment of the two sequences.
  • the comparison window is at least 20 contiguous nucleotides in length, and optionally can be 30, 40, 50, 100 or longer.
  • the BLAST family of programs which can be used for database similarity searches includes: BLASTN for nucleotide query sequences against nucleotide database sequences; BLASTX for nucleotide query sequences against protein database sequences; BLASTP for protein query sequences against protein database sequences; TBLASTN for protein query sequences against nucleotide database sequences and TBLASTX for nucleotide query sequences against nucleotide database sequences.
  • GAP uses the algorithm of Needleman and Wunsch, supra, to find the alignment of two complete sequences that maximizes the number of matches and minimizes the number of gaps. GAP considers all possible alignments and gap positions and creates the alignment with the largest number of matched bases and the fewest gaps. It allows for the provision of a gap creation penalty and a gap extension penalty in units of matched bases. GAP must make a profit of gap creation penalty number of matches for each gap it inserts. If a gap extension penalty greater than zero is chosen, GAP must, in addition, make a profit for each gap inserted of the length of the gap times the gap extension penalty. Default gap creation penalty values and gap extension penalty values in Version 10 of the Wisconsin Genetics Software Package are 8 and 2, respectively.
  • the gap creation and gap extension penalties can be expressed as an integer selected from the group of integers consisting of from 0 to 100.
  • the gap creation and gap extension penalties can be 0, 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, 50 or greater.
  • GAP presents one member of the family of best alignments. There may be many members of this family, but no other member has a better quality. GAP displays four figures of merit for alignments: Quality, Ratio, Identity and Similarity.
  • the Quality is the metric maximized in order to align the sequences. Ratio is the quality divided by the number of bases in the shorter segment.
  • Percent Identity is the percent of the symbols that actually match.
  • Percent Similarity is the percent of the symbols that are similar. Symbols that are across from gaps are ignored.
  • a similarity is scored when the scoring matrix value for a pair of symbols is greater than or equal to 0.50, the similarity threshold.
  • the scoring matrix used in Version 10 of the Wisconsin Genetics Software Package is BLOSUM62 (see, Henikoff and Henikoff, (1989) Proc. Natl. Acad. Sci. USA 89:10915).
  • sequence identity/similarity values refer to the value obtained using the BLAST 2.0 suite of programs using default parameters (Altschul, et ai, (1997) Nucleic Acids Res. 25:3389-402).
  • BLAST searches assume that proteins can be modeled as random sequences. However, many real proteins comprise regions of nonrandom sequences, which may be homopolymeric tracts, short-period repeats, or regions enriched in one or more amino acids. Such low-complexity regions may be aligned between unrelated proteins even though other regions of the protein are entirely dissimilar.
  • a number of low-complexity filter programs can be employed to reduce such low-complexity alignments. For example, the SEG (Wooten and Federhen, (1993) Comput. Chem. 17:149-63) and XNU (Claverie and States, (1993) Comput. Chem. 17:191-201 ) low-complexity filters can be employed alone or in combination.
  • sequence identity in the context of two nucleic acid or polypeptide sequences includes reference to the residues in the two sequences which are the same when aligned for maximum correspondence over a specified comparison window.
  • sequence identity or “identity” in the context of two nucleic acid or polypeptide sequences includes reference to the residues in the two sequences which are the same when aligned for maximum correspondence over a specified comparison window.
  • Sequences which differ by such conservative substitutions are said to have "sequence similarity" or "similarity.” Means for making this adjustment are well known to those of skill in the art. Typically this involves scoring a conservative substitution as a partial rather than a full mismatch, thereby increasing the percentage sequence identity. Thus, for example, where an identical amino acid is given a score of 1 and a non-conservative substitution is given a score of zero, a conservative substitution is given a score between zero and 1. The scoring of conservative substitutions is calculated, e.g., according to the algorithm of Meyers and Miller, (1988) Computer Applic. Biol. Sci. 4:1 1 -17, e.g., as implemented in the program PC/GENE (Intelligenetics, Mountain View, California, USA).
  • percentage of sequence identity means the value determined by comparing two optimally aligned sequences over a comparison window, wherein the portion of the polynucleotide 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.
  • substantially identical of polynucleotide sequences means that a polynucleotide comprises a sequence that has between 50-100% sequence identity, optionally at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or at least 95% sequence identity, compared to a reference sequence using one of the alignment programs described using standard parameters.
  • sequence identity optionally at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or at least 95% sequence identity, compared to a reference sequence using one of the alignment programs described using standard parameters.
  • Substantial identity of amino acid sequences for these purposes normally means sequence identity of between 55-100%, such as at least 55%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, up to 100% identity.
  • substantially identical in the context of a peptide indicates that a peptide comprises a sequence with between 55-100% sequence identity to a reference sequence, such as at least 55%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, up to 100% sequence identity to the reference sequence over a specified comparison window.
  • optimal alignment is conducted using the homology alignment algorithm of Needleman and Wunsch, supra.
  • An indication that two peptide sequences are substantially identical is that one peptide is immunologically reactive with antibodies raised against the second peptide.
  • a peptide is substantially identical to a second peptide, for example, where the two peptides differ only by a conservative substitution.
  • a peptide can be substantially identical to a second peptide when they differ by a non-conservative change if the epitope that the antibody recognizes is substantially identical.
  • Peptides which are "substantially similar" share sequences as noted above, except that residue positions, which are not identical, may differ by conservative amino acid changes.
  • SEQ ID NO: 16 Polypeptide Arabidopsis thaliana
  • SEQ ID NO: 17 Polypeptide Oryza sativa
  • SEQ ID NO: 18 Polypeptide Lilium longiflorum
  • SEQ ID NO: 20 Polypeptide Hordeum vulgare
  • SEQ ID NO: 22 Polypeptide Zea mays anther specific
  • SEQ ID NO: 26 Polypeptide Brassica rapa
  • SEQ ID NO: 70 Polynucleotide Tassel specific promoter
  • SEQ ID NO: 80 Polynucleotide Tassel specific promoter
  • the isolated nucleic acids of the present disclosure can be made using (a) standard recombinant methods, (b) synthetic techniques or combinations thereof.
  • the polynucleotides of the present disclosure will be cloned, amplified or otherwise constructed from a fungus or bacteria.
  • RNA Ribonucleic Acids Res. 13:7375.
  • Positive sequence motifs include translational initiation consensus sequences (Kozak, (1987) Nucleic Acids Res.15:8125) and the 5 ⁇ G> 7 methyl GpppG RNA cap structure (Drummond, et al. , (1985) Nucleic Acids Res. 13:7375).
  • Negative elements include stable intramolecular 5' UTR stem-loop structures (Muesing, et al.
  • the present disclosure provides 5' and/or 3' UTR regions for modulation of translation of heterologous coding sequences.
  • the polypeptide-encoding segments of the polynucleotides of the present disclosure can be modified to alter codon usage. Altered codon usage can be employed to alter translational efficiency and/or to optimize the coding sequence for expression in a desired host or to optimize the codon usage in a heterologous sequence for expression in maize.
  • Codon usage in the coding regions of the polynucleotides of the present disclosure can be analyzed statistically using commercially available software packages such as "Codon Preference” available from the University of Wisconsin Genetics Computer Group. See, Devereaux, et al., (1984) Nucleic Acids Res. 12:387-395) or MacVector 4.1 (Eastman Kodak Co., New Haven, CN).
  • the present disclosure provides a codon usage frequency characteristic of the coding region of at least one of the polynucleotides of the present disclosure.
  • the number of polynucleotides (3 nucleotides per amino acid) that can be used to determine a codon usage frequency can be any integer from 3 to the number of polynucleotides of the present disclosure as provided herein.
  • the polynucleotides will be full-length sequences.
  • An exemplary number of sequences for statistical analysis can be at least 1 , 5, 10, 20, 50 or 100.
  • sequence shuffling provides methods for sequence shuffling using polynucleotides of the present disclosure, and compositions resulting therefrom. Sequence shuffling is described in PCT Publication Number 1996/19256. See also, Zhang, et al. , (1997) Proc. Natl. Acad. Sci. USA 94:4504-9 and Zhao, et al. , (1998) Nature Biotech 16:258-61. Generally, sequence shuffling provides a means for generating libraries of polynucleotides having a desired characteristic, which can be selected or screened for.
  • Libraries of recombinant polynucleotides are generated from a population of related sequence polynucleotides, which comprise sequence regions, which have substantial sequence identity and can be homologously recombined in vitro or in vivo.
  • the population of sequence-recombined polynucleotides comprises a subpopulation of polynucleotides which possess desired or advantageous characteristics and which can be selected by a suitable selection or screening method.
  • the characteristics can be any property or attribute capable of being selected for or detected in a screening system, and may include properties of: an encoded protein, a transcriptional element, a sequence controlling transcription, RNA processing, RNA stability, chromatin conformation, translation or other expression property of a gene or transgene, a replicative element, a protein-binding element or the like, such as any feature which confers a selectable or detectable property.
  • the selected characteristic will be an altered K m and/or K cat over the wild-type protein as provided herein.
  • a protein or polynucleotide generated from sequence shuffling will have a ligand binding affinity greater than the non-shuffled wild-type polynucleotide.
  • a protein or polynucleotide generated from sequence shuffling will have an altered pH optimum as compared to the non-shuffled wild-type polynucleotide.
  • the increase in such properties can be at least 1 10%, 120%, 130%, 140% or greater than 150% of the wild- type value.
  • the present disclosure further provides recombinant expression cassettes comprising a nucleic acid of the present disclosure.
  • a nucleic acid sequence coding for the desired polynucleotide of the present disclosure for example a cDNA or a genomic sequence encoding a polypeptide long enough to code for an active protein of the present disclosure, can be used to construct a recombinant expression cassette which can be introduced into the desired host cell.
  • a recombinant expression cassette will typically comprise a polynucleotide of the present disclosure operably linked to transcriptional initiation regulatory sequences which will direct the transcription of the polynucleotide in the intended host cell, such as tissues of a transformed plant.
  • plant expression vectors may include (1 ) a cloned plant gene under the transcriptional control of 5' and 3' regulatory sequences and (2) a dominant selectable marker.
  • plant expression vectors may also contain, if desired, a promoter regulatory region (e.g., one conferring inducible or constitutive, environmentally- or developmentally- regulated, or cell- or tissue-specific/selective expression), a transcription initiation start site, a ribosome binding site, an RNA processing signal, a transcription termination site and/or a polyadenylation signal.
  • a plant promoter fragment can be employed which will direct expression of a polynucleotide of the present disclosure in essentially all tissues of a regenerated plant.
  • Such promoters are referred to herein as “constitutive” promoters and are active under most environmental conditions and states of development or cell differentiation.
  • constitutive promoters include the V- or 2'- promoter derived from T-DNA of Agrobacterium tumefaciens, the Smas promoter, the cinnamyl alcohol dehydrogenase promoter (US Patent Number 5,683,439), the Nos promoter, the rubisco promoter, the GRP1 -8 promoter, the 35S promoter from cauliflower mosaic virus (CaMV), as described in Odell, et al., (1985) Nature 313:810-2; rice actin (McElroy, et al., (1990) Plant Cell 163- 171 ); ubiquitin (Christensen, et al., (1992) Plant Mol. Biol.
  • ALS promoter as described in PCT Application Number WO 1996/30530 and other transcription initiation regions from various plant genes known to those of skill.
  • ubiquitin is the preferred promoter for expression in monocot plants.
  • the plant promoter can direct expression of a polynucleotide of the present disclosure in a specific tissue or may be otherwise under more precise environmental or developmental control.
  • Such promoters may be "inducible" promoters.
  • Environmental conditions that may effect transcription by inducible promoters include pathogen attack, anaerobic conditions or the presence of light.
  • inducible promoters are the Adh1 promoter, which is inducible by hypoxia or cold stress, the Hsp70 promoter, which is inducible by heat stress and the PPDK promoter, which is inducible by light.
  • Diurnal promoters that are active at different times during the circadian rhythm are also known (US Patent Application Publication Number 201 1/0167517, incorporated herein by reference).
  • promoters under developmental control include promoters that initiate transcription only, or preferentially, in certain tissues, such as leaves, roots, fruit, seeds or flowers.
  • the operation of a promoter may also vary depending on its location in the genome. Thus, an inducible promoter may become fully or partially constitutive in certain locations.
  • polypeptide expression it is generally desirable to include a polyadenylation region at the 3'-end of a polynucleotide coding region.
  • the polyadenylation region can be derived from a variety of plant genes, or from T-DNA.
  • the 3' end sequence to be added can be derived from, for example, the nopaline synthase or octopine synthase genes or alternatively from another plant gene or less preferably from any other eukaryotic gene.
  • regulatory elements include, but are not limited to, 3' termination and/or polyadenylation regions such as those of the Agrobacterium tumefaciens nopaline synthase (nos) gene (Bevan, et al., (1983) Nucleic Acids Res. 12:369-85); the potato proteinase inhibitor II (PINII) gene (Keil, et al., (1986) Nucleic Acids Res. 14:5641 -50 and An, et al., (1989) Plant Cell 1 : 1 15-22) and the CaMV 19S gene (Mogen, et al., (1990) Plant Cell 2: 1261-72).
  • PINII potato proteinase inhibitor II
  • An intron sequence can be added to the 5' untranslated region or the coding sequence of the partial coding sequence to increase the amount of the mature message that accumulates in the cytosol.
  • Inclusion of a spliceable intron in the transcription unit in both plant and animal expression constructs has been shown to increase gene expression at both the mRNA and protein levels up to 1000-fold (Buchman and Berg, (1988) Mol. Cell Biol. 8:4395-4405; Callis, et al., (1987) Genes Dev. 1 :1183-200).
  • Such intron enhancement of gene expression is typically greatest when placed near the 5' end of the transcription unit.
  • Use of maize introns Adh1 -S intron 1 , 2 and 6, the Bronze-1 intron are known in the art. See generally, The Maize Handbook, Chapter 1 16, Freeling and Walbot, eds., Springer, New York (1994).
  • Plant signal sequences including, but not limited to, signal-peptide encoding
  • DNA/RNA sequences which target proteins to the extracellular matrix of the plant cell such as the Nicotiana plumbaginifolia extension gene (DeLoose, et al., (1991 ) Gene 99:95-100); signal peptides which target proteins to the vacuole, such as the sweet potato sporamin gene (Matsuka, et al., (1991 ) Proc. Natl. Acad. Sci.
  • the vector comprising the sequences from a polynucleotide of the present disclosure will typically comprise a marker gene, which confers a selectable phenotype on plant cells.
  • the selectable marker gene may encode antibiotic resistance, with suitable genes including genes coding for resistance to the antibiotic spectinomycin (e.g., the aada gene), the streptomycin phosphotransferase (SPT) gene coding for streptomycin resistance, the neomycin phosphotransferase (NPTII) gene encoding kanamycin or geneticin resistance, the hygromycin phosphotransferase (HPT) gene coding for hygromycin resistance.
  • SPT streptomycin phosphotransferase
  • NPTII neomycin phosphotransferase
  • HPT hygromycin phosphotransferase
  • the bar gene encodes resistance to the herbicide basta and the ALS gene encodes resistance to the herbicide chlorsulfuron.
  • Constructs described herein may comprise a polynucleotide of interest encoding a reporter or marker product.
  • suitable reporter polynucleotides known in the art can be found in, for example, Jefferson et al. (1991 ) in Plant Molecular Biology Manual, ed. Gelvin et al. (Kluwer Academic Publishers), pp. 1-33; DeWet et al. Mol. Cell. Biol. 7:725-737 (1987); Goff et al. EMBO J. 9:2517-2522 (1990); Kain et al. BioTechniques 19:650-655 (1995); and Chiu et al. Current Biology 6:325-330 (1996).
  • the polynucleotide of interest encodes a selectable reporter. These can include polynucleotides that confer antibiotic resistance or resistance to herbicides.
  • Suitable selectable marker polynucleotides include, but are not limited to, genes encoding resistance to chloramphenicol, methotrexate, hygromycin, streptomycin, spectinomycin, bleomycin, sulfonamide, bromoxynil, glyphosate, and phosphinothricin.
  • the expression cassettes disclosed herein comprise a polynucleotide of interest encoding scorable or screenable markers, where presence of the polynucleotide produces a measurable product.
  • a polynucleotide of interest encoding scorable or screenable markers, where presence of the polynucleotide produces a measurable product.
  • examples include a ⁇ -glucuronidase, or uidA gene (GUS), which encodes an enzyme for which various chromogenic substrates are known (for example, U.S. Pat. Nos. 5,268,463 and 5,599,670); chloramphenicol acetyl transferase, and alkaline phosphatase.
  • Other screenable markers include the
  • anthocyanin/flavonoid polynucleotides including, for example, a R-locus polynucleotide, which encodes a product that regulates the production of anthocyanin pigments (red color) in plant tissues, the genes which control biosynthesis of flavonoid pigments, such as the maize C1 and C2 , the B gene, the p1 gene, and the bronze locus genes, among others.
  • suitable markers encoded by polynucleotides of interest include the cyan fluorescent protein (CYP) gene, the yellow fluorescent protein gene, a lux gene, which encodes a luciferase, the presence of which may be detected using, for example, X-ray film, scintillation counting, fluorescent spectrophotometry, low-light video cameras, photon counting cameras or multiwell luminometry, a green fluorescent protein (GFP), and DsRed2 (Clontechniques, 2001 ) where plant cells transformed with the marker gene are red in color, and thus visually selectable.
  • CYP cyan fluorescent protein
  • lux gene which encodes a luciferase
  • Additional examples include a p-lactamase gene encoding an enzyme for which various chromogenic substrates are known (e.g., PADAC, a chromogenic cephalosporin), a xylE gene encoding a catechol dioxygenase that can convert chromogenic catechols, an oamylase gene, and a tyrosinase gene encoding an enzyme capable of oxidizing tyrosine to DOPA and dopaquinone, which in turn condenses to form the easily detectable compound melanin.
  • PADAC chromogenic cephalosporin
  • the expression cassette can also comprise a selectable marker gene for the selection of transformed cells.
  • Selectable marker genes are utilized for the selection of transformed cells or tissues. Marker genes include genes encoding antibiotic resistance, such as those encoding neomycin phosphotransferase II (NEO) and hygromycin
  • HPT phosphotransferase
  • genes conferring resistance to herbicidal compounds such as glufosinate ammonium, bromoxynil, imidazolinones, and 2,4- dichlorophenoxyacetate (2,4-D).
  • Additional selectable markers include phenotypic markers such as ⁇ -galactosidase and fluorescent proteins such as green fluorescent protein (GFP) (Su et al. (2004) Biotechnol Bioeng 85:610-9 and Fetter et al. (2004) Plant Cell 16:215- 28), cyan florescent protein (CYP) (Bolte et al. (2004) J. Cell Science 117:943-54 and
  • Typical vectors useful for expression of genes in higher plants are well known in the art and include vectors derived from the tumor-inducing (Ti) plasmid of Agrobacterium tumefaciens described by Rogers, et al., (1987) Meth. Enzymol. 153:253-77. These vectors are plant integrating vectors in that on transformation, the vectors integrate a portion of vector DNA into the genome of the host plant.
  • Exemplary A. tumefaciens vectors useful herein are plasmids pKYLX6 and pKYLX7 of Schardl, et al., (1987) Gene 61 :1-1 1 and Berger, et al., (1989) Proc. Natl. Acad. Sci. USA, 86:8402-6.
  • Another useful vector herein is plasmid pBI 101.2 that is available from CLONTECH Laboratories, Inc. (Palo Alto, CA).
  • nucleic acids of the present disclosure may express a protein of the present disclosure in a recombinantly engineered cell such as bacteria, yeast, insect, mammalian or preferably plant cells.
  • a recombinantly engineered cell such as bacteria, yeast, insect, mammalian or preferably plant cells.
  • the cells produce the protein in a non-natural condition (e.g., in quantity, composition, location and/or time), because they have been genetically altered through human intervention to do so.
  • the expression of isolated nucleic acids encoding a protein of the present disclosure will typically be achieved by operably linking, for example, the DNA or cDNA to a promoter, followed by incorporation into an expression vector.
  • the vectors can be suitable for replication and integration in either prokaryotes or eukaryotes.
  • Typical expression vectors contain transcription and translation terminators, initiation sequences and promoters useful for regulation of the expression of the DNA of the present disclosure.
  • it is desirable to construct expression vectors which contain, at the minimum, a strong promoter, such as ubiquitin, to direct transcription, a ribosome binding site for translational initiation and a transcription/translation terminator.
  • Constitutive promoters are classified as providing for a range of constitutive expression. Thus, some are weak constitutive promoters and others are strong constitutive promoters. Generally, by “weak promoter” is intended a promoter that drives expression of a coding sequence at a low level. By “low level” is intended at levels of about 1/10,000 transcripts to about 1/100,000 transcripts to about 1/500,000 transcripts. Conversely, a “strong promoter” drives expression of a coding sequence at a "high level,” or about 1/10 transcripts to about 1/100 transcripts to about 1/1 ,000 transcripts.
  • modifications could be made to a protein of the present disclosure without diminishing its biological activity. Some modifications may be made to facilitate the cloning, expression or incorporation of the targeting molecule into a fusion protein. Such modifications are well known to those of skill in the art and include, for example, a methionine added at the amino terminus to provide an initiation site or additional amino acids (e.g., poly His) placed on either terminus to create conveniently located restriction sites or termination codons or purification sequences.
  • a methionine added at the amino terminus to provide an initiation site or additional amino acids (e.g., poly His) placed on either terminus to create conveniently located restriction sites or termination codons or purification sequences.
  • Prokaryotic cells may be used as hosts for expression. Prokaryotes most frequently are represented by various strains of £ coir, however, other microbial strains may also be used.
  • Commonly used prokaryotic control sequences which are defined herein to include promoters for transcription initiation, optionally with an operator, along with ribosome binding site sequences, include such commonly used promoters as the beta lactamase (penicillinase) and lactose (lac) promoter systems (Chang, et al., (1977) Nature 198:1056), the tryptophan (trp) promoter system (Goeddel, et al., (1980) Nucleic Acids Res.
  • Bacterial vectors are typically of plasmid or phage origin. Appropriate bacterial cells are infected with phage vector particles or transfected with naked phage vector DNA. If a plasmid vector is used, the bacterial cells are transfected with the plasmid vector DNA.
  • Expression systems for expressing a protein of the present disclosure are available using Bacillus sp. and Salmonella (Palva, et al., (1983) Gene 22:229-35; Mosbach, et al., (1983) Nature 302:543-5).
  • the pGEX-4T-1 plasmid vector from Pharmacia is the preferred £. coli expression vector for the present disclosure. Expression in Eukaryotes
  • eukaryotic expression systems such as yeast, insect cell lines, plant and mammalian cells, are known to those of skill in the art. As explained briefly below, the present disclosure can be expressed in these eukaryotic systems. In some embodiments, transformed/transfected plant cells, as discussed infra, are employed as expression systems for production of the proteins of the instant disclosure.
  • yeasts for production of eukaryotic proteins are Saccharomyces cerevisiae and Pichia pastoris.
  • Vectors, strains and protocols for expression in Saccharomyces and Pichia are known in the art and available from commercial suppliers (e.g., Invitrogen).
  • Suitable vectors usually have expression control sequences, such as promoters, including 3-phosphoglycerate kinase or alcohol oxidase and an origin of replication, termination sequences and the like as desired.
  • a protein of the present disclosure once expressed, can be isolated from yeast by lysing the cells and applying standard protein isolation techniques to the lysates or the pellets.
  • the monitoring of the purification process can be accomplished by using Western blot techniques or radioimmunoassay of other standard immunoassay techniques.
  • sequences encoding proteins of the present disclosure can also be ligated to various expression vectors for use in transfecting cell cultures of, for instance, mammalian, insect or plant origin.
  • Mammalian cell systems often will be in the form of monolayers of cells although mammalian cell suspensions may also be used.
  • a number of suitable host cell lines capable of expressing intact proteins have been developed in the art, and include the HEK293, BHK21 and CHO cell lines.
  • Expression vectors for these cells can include expression control sequences, such as an origin of replication, a promoter (e.g., the CMV promoter, a HSV tk promoter or pgk (phosphoglycerate kinase) promoter), an enhancer (Queen, et al., (1986) Immunol. Rev. 89:49) and necessary processing information sites, such as ribosome binding sites, RNA splice sites, polyadenylation sites (e.g., an SV40 large T Ag poly A addition site) and transcriptional terminator sequences.
  • a promoter e.g., the CMV promoter, a HSV tk promoter or pgk (phosphoglycerate kinase) promoter
  • an enhancer Queen, et al., (1986) Immunol. Rev. 89:49
  • necessary processing information sites such as ribosome binding sites, RNA splice sites, polyadenylation sites (e.
  • Appropriate vectors for expressing proteins of the present disclosure in insect cells are usually derived from the SF9 baculovirus.
  • suitable insect cell lines include mosquito larvae, silkworm, armyworm, moth and Drosophila cell lines such as a Schneider cell line (see, e.g., Schneider, (1987) J. Embryol. Exp. Morphol. 27:353-65).
  • polyadenlyation or transcription terminator sequences are typically incorporated into the vector.
  • An example of a terminator sequence is the polyadenylation sequence from the bovine growth hormone gene. Sequences for accurate splicing of the transcript may also be included.
  • An example of a splicing sequence is the VP1 intron from SV40 (Sprague, et al. , (1983) J. Virol. 45:773-81 ).
  • gene sequences to control replication in the host cell may be incorporated into the vector such as those found in bovine papilloma virus type-vectors (Saveria-Campo, "Bovine Papilloma Virus DNA a Eukaryotic Cloning Vector," in DNA Cloning: A Practical Approach, vol. II, Glover, ed., IRL Press, Arlington, VA, pp. 213-38 (1985)).
  • the gene of interest placed in the appropriate plant expression vector can be used to transform plant cells.
  • the polypeptide can then be isolated from plant callus or the transformed cells can be used to regenerate transgenic plants.
  • Such transgenic plants can be harvested, and the appropriate tissues (seed or leaves, for example) can be subjected to large scale protein extraction and purification techniques.
  • the methods chosen vary with the host plant and include chemical transfection methods such as calcium phosphate, microorganism-mediated gene transfer such as Agrobacterium (Horsch, et al., (1985) Science 227:1229-31 ), electroporation, micro-injection and biolistic bombardment.
  • the isolated polynucleotides or polypeptides may be introduced into the plant by one or more techniques typically used for direct delivery into cells. Such protocols may vary depending on the type of organism, cell, plant or plant cell, i.e., monocot or dicot, targeted for gene modification. Suitable methods of transforming plant cells include microinjection (Crossway, et al., (1986) Biotechniques 4:320-334 and US Patent Number 6,300,543), electroporation (Riggs, et al., (1986) Proc. Natl. Acad. Sci. USA 83:5602- 5606, direct gene transfer (Paszkowski et al., (1984) EMBO J.
  • A. tumefaciens and A. rhizogenes are plant pathogenic soil bacteria which genetically transform plant cells.
  • the Ti and Ri plasmids of A. tumefaciens and A. rhizogenes, respectively, carry genes responsible for genetic transformation of plants. See, e.g., Kado, (1991 ) Crit. Rev. Plant Sci. 10:1 . Descriptions of the Agrobacterium vector systems and methods for gene transfer are provided in Gruber, et al., supra; Miki, et al., supra and Moloney, et al., (1989) Plant Cell Reports 8:238.
  • the gene can be inserted into the T-DNA region of a Ti or Ri plasmid derived from A. tumefaciens or A. rhizogenes, respectively.
  • expression cassettes can be constructed as above, using these plasmids.
  • Many control sequences are known which when coupled to a heterologous coding sequence and transformed into a host organism show fidelity in gene expression with respect to tissue/organ specificity of the original coding sequence. See, e.g., Benfey and Chua, (1989) Science 244:174-81.
  • Particularly suitable control sequences for use in these plasmids are promoters for constitutive or tissue-preferred expression of the gene in the various target plants.
  • NOS nopaline synthase gene
  • these plasmids can be placed into A. rhizogenes or A. tumefaciens and these vectors used to transform cells of plant species which are ordinarily susceptible to Fusarium or Alternaria infection.
  • transgenic plants include but not limited to soybean, corn, sorghum, alfalfa, rice, clover, cabbage, banana, coffee, celery, tobacco, cowpea, cotton, melon and pepper.
  • the selection of either A. tumefaciens or A. rhizogenes will depend on the plant being transformed thereby. In general A. tumefaciens is the preferred organism for transformation.
  • A. rhizogenes also has a wide host range, embracing most dicots and some gymnosperms, which includes members of the Leguminosae, Compositae, and Chenopodiaceae. Monocot plants can also be transformed.
  • EP Patent Application Number 604 662 A1 discloses a method for transforming monocots using Agrobacterium.
  • EP Patent Application Number 672 752 A1 discloses a method for transforming monocots with Agrobacterium using the scutellum of immature embryos. Ishida, et al., discuss a method for transforming maize by exposing immature embryos to A. tumefaciens (Nature Biotechnology 14:745-50 (1996)).
  • these cells can be used to regenerate transgenic plants.
  • whole plants can be infected with these vectors by wounding the plant and then introducing the vector into the wound site. Any part of the plant can be wounded, including leaves, stems and roots.
  • plant tissue in the form of an explant such as cotyledonary tissue or leaf disks, can be inoculated with these vectors, and cultured under conditions which promote plant regeneration. Examples of such methods for regenerating plant tissue are disclosed in Shahin, (1985) Theor. Appl. Genet.
  • a generally applicable method of plant transformation is microprojectile-mediated transformation, where DNA is carried on the surface of microprojectiles measuring about 1 to 4 ⁇ .
  • the expression vector is introduced into plant tissues with a biolistic device that accelerates the microprojectiles to speeds of 300 to 600 m/s which is sufficient to penetrate the plant cell walls and membranes (Sanford, et al., (1987) Part. Sci. Technol. 5:27; Sanford, (1988) Trends Biotech 6:299; Sanford, (1990) Physiol. Plant 79:206 and Klein, et al., (1992) Biotechnology 10:268).
  • Another method for physical delivery of DNA to plants is sonication of target cells as described in Zang, et al., (1991 ) BioTechnology 9:996.
  • liposome or spheroplast fusions have been used to introduce expression vectors into plants. See, e.g., Deshayes, et al., (1985) EMBO J. 4:2731 and Christou, et al., (1987) Proc. Natl. Acad. Sci. USA 84:3962.
  • Direct uptake of DNA into protoplasts using CaCI 2 precipitation, polyvinyl alcohol, or poly-L-ornithine has also been reported. See, e.g., Hain, et al., (1985) Mol. Gen. Genet. 199:161 and Draper, et al., (1982) Plant Cell Physiol. 23:451 .
  • Methods are provided to reduce or eliminate the activity of a polypeptide of the disclosure by transforming a plant cell with an expression cassette that expresses a polynucleotide that inhibits the expression of the polypeptide.
  • the polynucleotide may inhibit the expression of the polypeptide directly, by preventing transcription or translation of the messenger RNA, or indirectly, by encoding a polypeptide that inhibits the transcription or translation of a gene encoding polypeptide.
  • Methods for inhibiting or eliminating the expression of a gene in a plant are well known in the art and any such method may be used in the present disclosure to inhibit the expression of polypeptide.
  • the expression of a polypeptide may be inhibited so that the protein level of the polypeptide is, for example, less than 70% of the protein level of the same polypeptide in a plant that has not been genetically modified or mutagenized to inhibit the expression of that polypeptide.
  • the protein level of the polypeptide in a modified plant according to the disclosure is less than 60%, less than 50%, less than 40%, less than 30%, less than 20%, less than 10%, less than 5% or less than 2% of the protein level of the same polypeptide in a plant that is not a mutant or that has not been genetically modified to inhibit the expression of that polypeptide.
  • the expression level of the polypeptide may be measured directly, for example, by assaying for the level of polypeptide expressed in the plant cell or plant, or indirectly, for example, by measuring the nitrogen uptake activity of the polypeptide in the plant cell or plant or by measuring the phenotypic changes in the plant. Methods for performing such assays are described elsewhere herein.
  • the activity of the polypeptide is reduced or eliminated by transforming a plant cell with an expression cassette comprising a polynucleotide encoding a polypeptide that inhibits the activity of a polypeptide.
  • the activity of a polypeptide is inhibited according to the present disclosure if the activity of the polypeptide is, for example, less than 70% of the activity of the same polypeptide in a plant that has not been modified to inhibit the activity of that polypeptide.
  • the activity of the polypeptide in a modified plant according to the disclosure is less than 60%, less than 50%, less than 40%, less than 30%, less than 20%, less than 10% or less than 5% of the activity of the same polypeptide in a plant that that has not been modified to inhibit the expression of that polypeptide.
  • the activity of a polypeptide is "eliminated" according to the disclosure when it is not detectable by the assay methods described elsewhere herein. Methods of determining the alteration of activity of a polypeptide are described elsewhere herein.
  • the activity of a polypeptide may be reduced or eliminated by disrupting the gene encoding the polypeptide.
  • the disclosure encompasses mutagenized plants that carry mutations in genes, where the mutations reduce expression of the gene or inhibit the activity of the encoded polypeptide.
  • many methods may be used to reduce or eliminate the activity of a polypeptide.
  • more than one method may be used to reduce the activity of a single polypeptide.
  • a plant is transformed with an expression cassette that is capable of expressing a polynucleotide that inhibits the expression of a polypeptide of the disclosure.
  • expression refers to the biosynthesis of a gene product, including the transcription and/or translation of said gene product.
  • an expression cassette capable of expressing a polynucleotide that inhibits the expression of at least one polypeptide is an expression cassette capable of producing an RNA molecule that inhibits the transcription and/or translation of at least one polypeptide of the disclosure.
  • the "expression” or “production” of a protein or polypeptide from a DNA molecule refers to the transcription and translation of the coding sequence to produce the protein or polypeptide
  • the "expression” or “production” of a protein or polypeptide from an RNA molecule refers to the translation of the RNA coding sequence to produce the protein or polypeptide.
  • inhibition of the expression of a polypeptide may be obtained by sense suppression or cosuppression.
  • an expression cassette is designed to express an RNA molecule corresponding to all or part of a messenger RNA encoding a polypeptide in the "sense" orientation. Over expression of the RNA molecule can result in reduced expression of the native gene. Accordingly, multiple plant lines transformed with the cosuppression expression cassette are screened to identify those that show the desired degree of inhibition of polypeptide expression.
  • the polynucleotide used for cosuppression may correspond to all or part of the sequence encoding the polypeptide, all or part of the 5' and/or 3' untranslated region of a polypeptide transcript or all or part of both the coding sequence and the untranslated regions of a transcript encoding a polypeptide.
  • the expression cassette is designed to eliminate the start codon of the polynucleotide so that no protein product will be translated.
  • Cosuppression may be used to inhibit the expression of plant genes to produce plants having undetectable protein levels for the proteins encoded by these genes. See, for example, Broin, et al., (2002) Plant Cell 14:1417-1432. Cosuppression may also be used to inhibit the expression of multiple proteins in the same plant. See, for example, US Patent Number 5,942,657. Methods for using cosuppression to inhibit the expression of endogenous genes in plants are described in Flavell, et al., (1994) Proc. Natl. Acad. Sci. USA 91 :3490-3496; Jorgensen, et al., (1996) Plant Mol. Biol. 31 :957-973; Johansen and Carrington, (2001 ) Plant Physiol.
  • nucleotide sequence has substantial sequence identity to the sequence of the transcript of the endogenous gene, optimally greater than about 65% sequence identity, more optimally greater than about 85% sequence identity, most optimally greater than about 95% sequence identity. See US Patent Numbers 5,283,184 and 5,034,323, herein incorporated by reference.
  • inhibition of the expression of the polypeptide may be obtained by antisense suppression.
  • the expression cassette is designed to express an RNA molecule complementary to all or part of a messenger RNA encoding the polypeptide. Over expression of the antisense RNA molecule can result in reduced expression of the target gene. Accordingly, multiple plant lines transformed with the antisense suppression expression cassette are screened to identify those that show the desired degree of inhibition of polypeptide expression.
  • the polynucleotide for use in antisense suppression may correspond to all or part of the complement of the sequence encoding the polypeptide, all or part of the complement of the 5' and/or 3' untranslated region of the target transcript or all or part of the complement of both the coding sequence and the untranslated regions of a transcript encoding the polypeptide.
  • the antisense polynucleotide may be fully complementary (i.e., 100% identical to the complement of the target sequence) or partially complementary (i.e., less than 100% identical to the complement of the target sequence) to the target sequence.
  • Antisense suppression may be used to inhibit the expression of multiple proteins in the same plant. See, for example, US Patent Number 5,942,657.
  • portions of the antisense nucleotides may be used to disrupt the expression of the target gene.
  • sequences of at least 50 nucleotides, 100 nucleotides, 200 nucleotides, 300, 400, 450, 500, 550 or greater may be used.
  • Methods for using antisense suppression to inhibit the expression of endogenous genes in plants are described, for example, in Liu, et al., (2002) Plant Physiol. 129:1732-1743 and US Patent Numbers 5,759,829 and 5,942,657, each of which is herein incorporated by reference.
  • Efficiency of antisense suppression may be increased by including a poly-dT region in the expression cassette at a position 3' to the antisense sequence and 5' of the polyadenylation signal. See, US Patent Application Publication Number 2002/0048814, herein incorporated by reference. / ' / ' / ' . Double-Stranded RNA Interference
  • inhibition of the expression of a polypeptide may be obtained by double-stranded RNA (dsRNA) interference.
  • dsRNA interference a sense RNA molecule like that described above for cosuppression and an antisense RNA molecule that is fully or partially complementary to the sense RNA molecule are expressed in the same cell, resulting in inhibition of the expression of the corresponding endogenous messenger RNA.
  • Expression of the sense and antisense molecules can be accomplished by designing the expression cassette to comprise both a sense sequence and an antisense sequence. Alternatively, separate expression cassettes may be used for the sense and antisense sequences. Multiple plant lines transformed with the dsRNA interference expression cassette or expression cassettes are then screened to identify plant lines that show the desired degree of inhibition of polypeptide expression. Methods for using dsRNA interference to inhibit the expression of endogenous plant genes are described in Waterhouse, et al., (1998) Proc. Natl. Acad. Sci. USA 95:13959-13964, Liu, et al., (2002) Plant Physiol.
  • inhibition of the expression of a polypeptide may be obtained by hairpin RNA (hpRNA) interference or intron-containing hairpin RNA (ihpRNA) interference.
  • hpRNA hairpin RNA
  • ihpRNA intron-containing hairpin RNA
  • the expression cassette is designed to express an RNA molecule that hybridizes with itself to form a hairpin structure that comprises a single- stranded loop region and a base-paired stem.
  • the base-paired stem region comprises a sense sequence corresponding to all or part of the endogenous messenger RNA encoded by the gene whose expression is to be inhibited, and an antisense sequence that is fully or partially complementary to the sense sequence.
  • the base-paired stem region may correspond to a portion of a promoter sequence controlling expression of the gene whose expression is to be inhibited.
  • the base-paired stem region of the molecule generally determines the specificity of the RNA interference.
  • hpRNA molecules are highly efficient at inhibiting the expression of endogenous genes and the RNA interference they induce is inherited by subsequent generations of plants. See, for example, Chuang and Meyerowitz, (2000) Proc. Natl. Acad. Sci. USA 97:4985-4990; Stoutjesdijk, et al., (2002) Plant Physiol. 129:1723-1731 and Waterhouse and Helliwell, (2003) Nat. Rev. Genet. 4:29-38. Methods for using hpRNA interference to inhibit or silence the expression of genes are described, for example, in Chuang and Meyerowitz,
  • the interfering molecules have the same general structure as for hpRNA, but the RNA molecule additionally comprises an intron that is capable of being spliced in the cell in which the ihpRNA is expressed.
  • the use of an intron minimizes the size of the loop in the hairpin RNA molecule following splicing, and this increases the efficiency of interference.
  • Smith, et al. show 100% suppression of endogenous gene expression using ihpRNA-mediated interference.
  • Methods for using ihpRNA interference to inhibit the expression of endogenous plant genes are described, for example, in Smith, et al. , (2000) Nature 407:319-320; Wesley, et al., (2001 ) Plant J. 27:581-590; Wang and Waterhouse,
  • the expression cassette for hpRNA interference may also be designed such that the sense sequence and the antisense sequence do not correspond to an endogenous RNA.
  • the sense and antisense sequence flank a loop sequence that comprises a nucleotide sequence corresponding to all or part of the endogenous messenger RNA of the target gene.
  • it is the loop region that determines the specificity of the RNA interference.
  • Amplicon expression cassettes comprise a plant-virus-derived sequence that contains all or part of the target gene but generally not all of the genes of the native virus.
  • the viral sequences present in the transcription product of the expression cassette allow the transcription product to direct its own replication.
  • the transcripts produced by the amplicon may be either sense or antisense relative to the target sequence (i.e., the messenger RNA for the polypeptide).
  • Methods of using amplicons to inhibit the expression of endogenous plant genes are described, for example, in Angell and Baulcombe, (1997) EMBO J. 16:3675-3684, Angell and Baulcombe, (1999) Plant J. 20:357-362 and US Patent Number 6,646,805, each of which is herein incorporated by reference.
  • the polynucleotide expressed by the expression cassette of the disclosure is catalytic RNA or has ribozyme activity specific for the messenger RNA of the polypeptide.
  • the polynucleotide causes the degradation of the endogenous messenger RNA, resulting in reduced expression of the polypeptide. This method is described, for example, in US Patent Number 4,987,071 , herein incorporated by reference. vii. Small Interfering RNA or Micro RNA
  • inhibition of the expression of a polypeptide may be obtained by RNA interference by expression of a polynucleotide encoding a micro RNA (miRNA).
  • miRNAs are regulatory agents consisting of about 22 ribonucleotides. miRNA are highly efficient at inhibiting the expression of endogenous genes. See, for example Javier, et al., (2003) Nature 425:257-263, herein incorporated by reference.
  • the expression cassette is designed to express an RNA molecule that is modeled on an endogenous miRNA gene.
  • the miRNA gene encodes an RNA that forms a hairpin structure containing a 22-nucleotide sequence that is complementary to an endogenous gene target sequence.
  • the 22-nucleotide sequence is selected from a NUE transcript sequence and contains 22 nucleotides of said NUE sequence in sense orientation and 21 nucleotides of a corresponding antisense sequence that is complementary to the sense sequence.
  • a fertility gene may be an miRNA target. miRNA molecules are highly efficient at inhibiting the expression of endogenous genes, and the RNA interference they induce is inherited by subsequent generations of plants.
  • the polynucleotide encodes a zinc finger protein that binds to a gene encoding a polypeptide, resulting in reduced expression of the gene.
  • the zinc finger protein binds to a regulatory region of a gene.
  • the zinc finger protein binds to a messenger RNA encoding a polypeptide and prevents its translation.
  • the polynucleotide encodes an antibody that binds to at least one polypeptide and reduces the activity of the polypeptide.
  • the binding of the antibody results in increased turnover of the antibody-polypeptide complex by cellular quality control mechanisms.
  • the activity of a polypeptide is reduced or eliminated by disrupting the gene encoding the polypeptide.
  • the gene encoding the polypeptide may be disrupted by any method known in the art. For example, in one embodiment, the gene is disrupted by transposon tagging. In another embodiment, the gene is disrupted by mutagenizing plants using random or targeted mutagenesis and selecting for plants that have reduced nitrogen utilization activity.
  • transposon tagging is used to reduce or eliminate the activity of one or more polypeptide.
  • Transposon tagging comprises inserting a transposon within an endogenous gene to reduce or eliminate expression of the polypeptide.
  • the expression of one or more polypeptides is reduced or eliminated by inserting a transposon within a regulatory region or coding region of the gene encoding the polypeptide.
  • a transposon that is within an exon, intron, 5' or 3' untranslated sequence, a promoter or any other regulatory sequence of a gene may be used to reduce or eliminate the expression and/or activity of the encoded polypeptide.
  • mutagenesis such as ethyl methanesulfonate- induced mutagenesis, deletion mutagenesis and fast neutron deletion mutagenesis used in a reverse genetics sense (with PCR) to identify plant lines in which the endogenous gene has been deleted.
  • Mutations that impact gene expression or that interfere with the function of the encoded protein are well known in the art. Insertional mutations in gene exons usually result in null-mutants. Mutations in conserved residues are particularly effective in inhibiting the activity of the encoded protein. conserveed residues of plant polypeptides suitable for mutagenesis with the goal to eliminate activity have been described. Such mutants can be isolated according to well-known procedures and mutations in different loci can be stacked by genetic crossing. See, for example, Gruis, et al., (2002) Plant Cell 14:2863-2882.
  • dominant mutants can be used to trigger
  • RNA silencing due to gene inversion and recombination of a duplicated gene locus See, for example, Kusaba, et al. , (2003) Plant Cell 15:1455-1467.
  • the disclosure encompasses additional methods for reducing or eliminating the activity of one or more polypeptide.
  • methods for altering or mutating a genomic nucleotide sequence in a plant include, but are not limited to, the use of RNA:DNA vectors, RNA:DNA mutational vectors, RNA:DNA repair vectors, mixed-duplex oligonucleotides, self-complementary RNA:DNA oligonucleotides and recombinogenic oligonucleobases.
  • Such vectors and methods of use are known in the art.
  • the level and/or activity of a NUE regulator in a plant is decreased by increasing the level or activity of the polypeptide in the plant.
  • the increased expression of a negative regulatory molecule may decrease the level of expression of downstream one or more genes responsible for an improved NUE phenotype.
  • a NUE nucleotide sequence encoding a polypeptide can be provided by introducing into the plant a polynucleotide comprising a NUE nucleotide sequence of the disclosure, expressing the NUE sequence, increasing the activity of the polypeptide and thereby decreasing the number of tissue cells in the plant or plant part.
  • the NUE nucleotide construct introduced into the plant is stably incorporated into the genome of the plant.
  • the growth of a plant tissue is increased by decreasing the level and/or activity of the polypeptide in the plant.
  • a NUE nucleotide sequence is introduced into the plant and expression of said NUE nucleotide sequence decreases the activity of the polypeptide and thereby increasing the tissue growth in the plant or plant part.
  • the NUE nucleotide construct introduced into the plant is stably incorporated into the genome of the plant.
  • such plants have stably incorporated into their genome a nucleic acid molecule comprising a NUE nucleotide sequence of the disclosure operably linked to a promoter that drives expression in the plant cell.
  • modulating root development is intended any alteration in the development of the plant root when compared to a control plant.
  • Such alterations in root development include, but are not limited to, alterations in the growth rate of the primary root, the fresh root weight, the extent of lateral and adventitious root formation, the vasculature system, meristem development or radial expansion.
  • Methods for modulating root development in a plant comprise modulating the level and/or activity of the polypeptide in the plant.
  • a sequence of the disclosure is provided to the plant.
  • the nucleotide sequence is provided by introducing into the plant a polynucleotide comprising a nucleotide sequence of the disclosure, expressing the sequence and thereby modifying root development.
  • the nucleotide construct introduced into the plant is stably incorporated into the genome of the plant.
  • root development is modulated by altering the level or activity of the polypeptide in the plant.
  • a change in activity can result in at least one or more of the following alterations to root development, including, but not limited to, alterations in root biomass and length.
  • root growth encompasses all aspects of growth of the different parts that make up the root system at different stages of its development in both monocotyledonous and dicotyledonous plants. It is to be understood that enhanced root growth can result from enhanced growth of one or more of its parts including the primary root, lateral roots, adventitious roots, etc.
  • exemplary promoters for this embodiment include constitutive promoters and root-preferred promoters. Exemplary root-preferred promoters have been disclosed elsewhere herein.
  • Stimulating root growth and increasing root mass by decreasing the activity and/or level of the polypeptide also finds use in improving the standability of a plant.
  • the term "resistance to lodging” or “standability” refers to the ability of a plant to fix itself to the soil. For plants with an erect or semi-erect growth habit, this term also refers to the ability to maintain an upright position under adverse environmental conditions. This trait relates to the size, depth and morphology of the root system.
  • stimulating root growth and increasing root mass by altering the level and/or activity of the polypeptide finds use in promoting in vitro propagation of explants.
  • root biomass production has a direct effect on the yield and an indirect effect of production of compounds produced by root cells or transgenic root cells or cell cultures of said transgenic root cells.
  • An interesting compound produced in root cultures is shikonin, the yield of which can be advantageously enhanced by said methods.
  • the present disclosure further provides plants having modulated root development when compared to the root development of a control plant.
  • the plant of the disclosure has an increased level/activity of a polypeptide of the disclosure and has enhanced root growth and/or root biomass.
  • such plants have stably incorporated into their genome a nucleic acid molecule comprising a nucleotide sequence of the disclosure operably linked to a promoter that drives expression in the plant cell.
  • Methods are also provided for modulating shoot and leaf development in a plant.
  • modulating shoot and/or leaf development is intended any alteration in the development of the plant shoot and/or leaf.
  • Such alterations in shoot and/or leaf development include, but are not limited to, alterations in shoot meristem development, in leaf number, leaf size, leaf and stem vasculature, internode length and leaf senescence.
  • leaf development andshoot development encompasses all aspects of growth of the different parts that make up the leaf system and the shoot system, respectively, at different stages of their development, both in monocotyledonous and dicotyledonous plants. Methods for measuring such developmental alterations in the shoot and leaf system are known in the art. See, for example, Werner, et al., (2001 ) PNAS 98:10487-10492 and US Patent Application Publication Number 2003/0074698, each of which is herein incorporated by reference.
  • the method for modulating shoot and/or leaf development in a plant comprises modulating the activity and/or level of a polypeptide of the disclosure.
  • a sequence of the disclosure is provided.
  • the nucleotide sequence can be provided by introducing into the plant a polynucleotide comprising a nucleotide sequence of the disclosure, expressing the sequence and thereby modifying shoot and/or leaf development.
  • the nucleotide construct introduced into the plant is stably incorporated into the genome of the plant.
  • shoot or leaf development is modulated by altering the level and/or activity of the polypeptide in the plant.
  • a change in activity can result in at least one or more of the following alterations in shoot and/or leaf development, including, but not limited to, changes in leaf number, altered leaf surface, altered vasculature, internodes and plant growth and alterations in leaf senescence when compared to a control plant.
  • promoters for this embodiment include constitutive promoters, shoot-preferred promoters, shoot meristem- preferred promoters and leaf-preferred promoters. Exemplary promoters have been disclosed elsewhere herein.
  • Increasing activity and/or level of a polypeptide of the disclosure in a plant may result in altered internodes and growth.
  • the methods of the disclosure find use in producing modified plants.
  • activity in the plant modulates both root and shoot growth.
  • the present disclosure further provides methods for altering the root/shoot ratio.
  • Shoot or leaf development can further be modulated by altering the level and/or activity of the polypeptide in the plant.
  • the present disclosure further provides plants having modulated shoot and/or leaf development when compared to a control plant.
  • the plant of the disclosure has an increased level/activity of a polypeptide of the disclosure.
  • a plant of the disclosure has a decreased level/activity of a polypeptide of the disclosure.
  • Methods for modulating reproductive tissue development are provided.
  • methods are provided to modulate floral development in a plant.
  • modulating floral development is intended any alteration in a structure of a plant's reproductive tissue as compared to a control plant in which the activity or level of the polypeptide has not been modulated.
  • Modulating floral development further includes any alteration in the timing of the development of a plant's reproductive tissue (e.g., a delayed or an accelerated timing of floral development) when compared to a control plant in which the activity or level of the polypeptide has not been modulated. Changes in timing of reproductive development may result in altered synchronization of development of male and female reproductive tissues.
  • Macroscopic alterations may include changes in size, shape, number or location of reproductive organs, the developmental time period that these structures form or the ability to maintain or proceed through the flowering process in times of environmental stress. Microscopic alterations may include changes to the types or shapes of cells that make up the reproductive organs.
  • the method for modulating floral development in a plant comprises modulating activity in a plant.
  • a sequence of the disclosure is provided.
  • a nucleotide sequence can be provided by introducing into the plant a polynucleotide comprising a nucleotide sequence of the disclosure, expressing the sequence and thereby modifying floral development.
  • the nucleotide construct introduced into the plant is stably incorporated into the genome of the plant.
  • floral development is modulated by increasing the level or activity of the polypeptide in the plant.
  • a change in activity can result in at least one or more of the following alterations in floral development, including, but not limited to, altered flowering, changed number of flowers, modified male sterility and altered seed set, when compared to a control plant.
  • Inducing delayed flowering or inhibiting flowering can be used to enhance yield in forage crops such as alfalfa.
  • Methods for measuring such developmental alterations in floral development are known in the art. See, for example, Mouradov, et al., (2002) The Plant Cell S1 1 1-S130, herein incorporated by reference.
  • promoters for this embodiment include constitutive promoters, inducible promoters, shoot-preferred promoters and inflorescence-preferred promoters.
  • floral development is modulated by altering the level and/or activity of a sequence of the disclosure.
  • Such methods can comprise introducing a nucleotide sequence into the plant and changing the activity of the polypeptide.
  • the nucleotide construct introduced into the plant is stably incorporated into the genome of the plant.
  • Altering expression of the sequence of the disclosure can modulate floral development during periods of stress.
  • the present disclosure further provides plants having modulated floral development when compared to the floral development of a control plant.
  • Compositions include plants having an altered level/activity of the polypeptide of the disclosure and having an altered floral development.
  • Compositions also include plants having a modified level/activity of the polypeptide of the disclosure wherein the plant maintains or proceeds through the flowering process in times of stress.
  • Methods are also provided for the use of the sequences of the disclosure to increase seed size and/or weight.
  • the method comprises increasing the activity of the sequences in a plant or plant part, such as the seed.
  • An increase in seed size and/or weight comprises an increased size or weight of the seed and/or an increase in the size or weight of one or more seed part including, for example, the embryo, endosperm, seed coat, aleurone or cotyledon.
  • promoters of this embodiment include constitutive promoters, inducible promoters, seed-preferred promoters, embryo-preferred promoters and endosperm-preferred promoters.
  • a method for altering seed size and/or seed weight in a plant may increasing activity in the plant.
  • the NUE nucleotide sequence can be provided by introducing into the plant a polynucleotide comprising a NUE nucleotide sequence of the disclosure, expressing the NUE sequence and thereby decreasing seed weight and/or size.
  • the NUE nucleotide construct introduced into the plant is stably incorporated into the genome of the plant.
  • increasing seed size and/or weight can also be accompanied by an increase in the speed of growth of seedlings or an increase in early vigor.
  • early vigor refers to the ability of a plant to grow rapidly during early development, and relates to the successful establishment, after germination, of a well-developed root system and a well-developed photosynthetic apparatus.
  • an increase in seed size and/or weight can also result in an increase in plant yield when compared to a control.
  • the present disclosure further provides plants having an increased seed weight and/or seed size when compared to a control plant.
  • plants having an increased vigor and plant yield are also provided.
  • the plant of the disclosure has a modified level/activity of the polypeptide of the disclosure and has an increased seed weight and/or seed size.
  • such plants have stably incorporated into their genome a nucleic acid molecule comprising a NUE nucleotide sequence of the disclosure operably linked to a promoter that drives expression in the plant cell.
  • nucleotides, expression cassettes and methods disclosed herein are useful in regulating expression of any heterologous nucleotide sequence in a host plant in order to vary the phenotype of a plant.
  • Various changes in phenotype are of interest including modifying the fatty acid composition in a plant, altering the amino acid content of a plant, altering a plant's pathogen defense mechanism and the like.
  • These results can be achieved by providing expression of heterologous products or increased expression of endogenous products in plants.
  • the results can be achieved by providing for a reduction of expression of one or more endogenous products, particularly enzymes or cofactors in the plant.
  • genes of interest are reflective of the commercial markets and interests of those involved in the development of the crop. Crops and markets of interest change, and as developing nations open up world markets, new crops and technologies will emerge also. In addition, as our understanding of agronomic traits and characteristics such as yield and heterosis increases, the choice of genes for transformation will change accordingly.
  • General categories of genes of interest include, for example, those genes involved in information, such as zinc fingers, those involved in communication, such as kinases, and those involved in housekeeping, such as heat shock proteins. More specific categories of transgenes, for example, include genes encoding important traits for agronomics, insect resistance, disease resistance, herbicide resistance, sterility, grain characteristics and commercial products. Genes of interest include, generally, those involved in oil, starch, carbohydrate or nutrient metabolism as well as those affecting kernel size, sucrose loading and the like.
  • nucleic acid sequences of the present disclosure can be used in combination ("stacked") with other polynucleotide sequences of interest in order to create plants with a desired phenotype.
  • the combinations generated can include multiple copies of any one or more of the polynucleotides of interest.
  • the promoter which is operably linked to a polynucleotide sequence of interest can be any promoter that is active in plant cells. In some embodiments it is particularly advantageous to use a promoter that is active (or can be activated) in reproductive tissues of a plant (e.g., stamens or ovaries).
  • the promoter can be, for example, a constitutively active promoter, an inducible promoter, a tissue-specific promoter or a developmental stage specific promoter.
  • the promoter of the a exogenous nucleic acid molecule can be the same as or different from the promoter of a second exogenous nucleic acid molecule.
  • the polynucleotides of the present disclosure may be stacked with any gene or combination of genes to produce plants with a variety of desired trait combinations, including but not limited to traits desirable for animal feed such as high oil genes (e.g., US Patent Number 6,232,529); balanced amino acids (e.g., hordothionins (US Patent Numbers 5,990,389; 5,885,801 ; 5,885,802 and 5,703,409); barley high lysine (Williamson, et al., (1987) Eur. J. Biochem. 165:99-106 and WO 1998/20122) and high methionine proteins (Pedersen, et al., (1986) J. Biol. Chem.
  • high oil genes e.g., US Patent Number 6,232,529)
  • balanced amino acids e.g., hordothionins (US Patent Numbers 5,990,389; 5,885,801 ; 5,885,802 and 5,703,409)
  • polynucleotides of the present disclosure can also be stacked with traits desirable for insect, disease or herbicide resistance (e.g., Bacillus thuringiensis toxic proteins (US Patent Numbers 5,366,892; 5,747,450; 5,737,514; 5723,756; 5,593,881 ; Geiser, et al., (1986) Gene 48:109); lectins (Van Damme, et al. , (1994) Plant Mol. Biol.
  • traits desirable for insect, disease or herbicide resistance e.g., Bacillus thuringiensis toxic proteins (US Patent Numbers 5,366,892; 5,747,450; 5,737,514; 5723,756; 5,593,881 ; Geiser, et al., (1986) Gene 48:109); lectins (Van Damme, et al. , (1994) Plant Mol. Biol.
  • PHAs polyhydroxyalkanoates
  • agronomic traits such as male sterility (e.g., see, US Patent Number 5.583,210), stalk strength, flowering time or transformation technology traits such as cell cycle regulation or gene targeting (e.g., WO 1999/61619; WO 2000/17364; WO 1999/25821 ), the disclosures of which are herein incorporated by reference.
  • Transgenic plants comprising or derived from plant cells or native plants with reduced male fertility of this disclosure can be further enhanced with stacked traits, e.g., a crop plant having an enhanced trait resulting from expression of DNA disclosed herein in combination with herbicide tolerance and/or pest resistance traits.
  • plants with reduced male fertility can be stacked with other traits of agronomic interest, such as a trait providing herbicide resistance and/or insect resistance, such as using a gene from Bacillus thuringensis to provide resistance against one or more of lepidopteran, coleopteran, homopteran, hemiopteran and other insects.
  • genes that confer tolerance to herbicides such as e.g., auxin, HPPD, glyphosate, dicamba, glufosinate, sulfonylurea, bromoxynil and norflurazon herbicides can be stacked either as a molecular stack or a breeding stack with plants expressing the traits disclosed herein.
  • Polynucleotide molecules encoding proteins involved in herbicide tolerance include, but are not limited to, a polynucleotide molecule encoding 5-enolpyruvylshikimate-3- phosphate synthase (EPSPS) disclosed in US Patent Numbers 39,247; 6,566,587 and for imparting glyphosate tolerance; polynucleotide molecules encoding a glyphosate oxidoreductase (GOX) disclosed in US Patent Number 5,463,175 and a glyphosate-N- acetyl transferase (GAT) disclosed in US Patent Numbers 7,622,641 ; 7,462,481 ; 7,531 ,339; 7,527,955; 7,709,709; 7,714,188 and 7,666,643, also for providing glyphosate tolerance; dicamba monooxygenase disclosed in US Patent Number 7,022,896 and WO 2007/146706 A2 for providing dicamba tolerance;
  • herbicide-tolerance traits that could be combined with the traits disclosed herein include those conferred by polynucleotides encoding an exogenous phosphinothricin acetyltransferase, as described in US Patent Numbers 5,969,213; 5,489,520; 5,550,318; 5,874,265; 5,919,675; 5,561 ,236; 5,648,477; 5,646,024; 6,177,616 and 5,879,903. Plants containing an exogenous phosphinothricin acetyltransferase can exhibit improved tolerance to glufosinate herbicides, which inhibit the enzyme glutamine synthase.
  • herbicide-tolerance traits include those conferred by polynucleotides conferring altered protoporphyrinogen oxidase (protox) activity, as described in US Patent Numbers 6,288,306 B1 ; 6,282,837 B1 and 5,767,373 and international publication WO 2001/12825. Plants containing such polynucleotides can exhibit improved tolerance to any of a variety of herbicides which target the protox enzyme (also referred to as "protox inhibitors”)
  • sequences of interest improve plant growth and/or crop yields.
  • sequences of interest include agronomically important genes that result in improved primary or lateral root systems.
  • genes include, but are not limited to, nutrient/water transporters and growth inducers.
  • genes include, but are not limited to, maize plasma membrane H + -ATPase (MHA2) (Frias, et al., (1996) Plant Cell 8:1533-44); AKT1 , a component of the potassium uptake apparatus in Arabidopsis, (Spalding, et al., (1999) J Gen Physiol 1 13:909-18); RML genes which activate cell division cycle in the root apical cells (Cheng, et al., (1995) Plant Physiol 108:881 ); maize glutamine synthetase genes (Sukanya, et al., (1994) Plant Mol Biol 26:1935-46) and hemoglobin (Duff, et al., (1997) J.
  • MHA2 maize plasma membrane H + -ATPase
  • AKT1 a component of the potassium uptake apparatus in Arabidopsis
  • RML genes which activate cell division cycle in the root apical cells
  • Additional, agronomically important traits such as oil, starch and protein content can be genetically altered in addition to using traditional breeding methods. Modifications include increasing content of oleic acid, saturated and unsaturated oils, increasing levels of lysine and sulfur, providing essential amino acids and also modification of starch. Hordothionin protein modifications are described in US Patent Numbers 5,703,049, 5,885,801 , 5,885,802 and 5,990,389, herein incorporated by reference. Another example is lysine and/or sulfur rich seed protein encoded by the soybean 2S albumin described in US Patent Number 5,850,016 and the chymotrypsin inhibitor from barley described in Williamson, et al., (1987) Eur. J. Biochem.
  • the disclosures of which are herein incorporated by reference Derivatives of the coding sequences can be made by site-directed mutagenesis to increase the level of preselected amino acids in the encoded polypeptide.
  • the gene encoding the barley high lysine polypeptide (BHL) is derived from barley chymotrypsin inhibitor, US Patent Application Serial Number 08/740,682, filed November 1 , 1996, and WO 1998/20133, the disclosures of which are herein incorporated by reference.
  • Other proteins include methionine-rich plant proteins such as from sunflower seed (Lilley, et al., (1989) Proceedings of the World Congress on Vegetable Protein Utilization in Human Foods and Animal Feedstuff s, ed. Applewhite (American Oil Chemists Society, Champaign, Illinois), pp. 497-502; herein incorporated by reference); corn (Pedersen, et al., (1986) J. Biol. Chem. 261 :6279; Kirihara, et al., (1988) Gene 71 :359, both of which are herein incorporated by reference) and rice (Musumura, et al., (1989) Plant Mol. Biol. 12:123, herein incorporated by reference).
  • Other agronomically important genes encode latex, Floury 2, growth factors, seed storage factors and transcription factors.
  • Insect resistance genes may encode resistance to pests that have great yield drag such as rootworm, cutworm, European Corn Borer and the like.
  • Such genes include, for example, Bacillus thuringiensis toxic protein genes (US Patent Numbers 5,366,892; 5,747,450; 5,736,514; 5,723,756; 5,593,881 and Geiser, et al., (1986) Gene 48:109) and the like.
  • Genes encoding disease resistance traits include detoxification genes, such as against fumonosin (US Patent Number 5,792,931 ); avirulence (avr) and disease resistance (R) genes (Jones, et al., (1994) Science 266:789; Martin, et al., (1993) Science 262:1432 and Mindrinos, et ai, (1994) Cell 78:1089) and the like.
  • Herbicide resistance traits may include genes coding for resistance to herbicides that act to inhibit the action of acetolactate synthase (ALS), in particular the sulfonylurea- type herbicides (e.g., the acetolactate synthase (ALS) gene containing mutations leading to such resistance, in particular the S4 and/or Hra mutations), genes coding for resistance to herbicides that act to inhibit action of glutamine synthase, such as phosphinothricin or basta (e.g., the bar gene) or other such genes known in the art.
  • the bar gene encodes resistance to the herbicide basta
  • the nptll gene encodes resistance to the antibiotics kanamycin and geneticin
  • the ALS-gene mutants encode resistance to the herbicide chlorsulfuron.
  • Sterility genes can also be encoded in an expression cassette and provide an alternative to physical emasculation. Examples of genes used in such ways include male tissue-preferred genes and genes with male sterility phenotypes such as QM, described in US Patent Number 5,583,210. Other genes include kinases and those encoding compounds toxic to either male or female gametophytic development.
  • Exogenous products include plant enzymes and products as well as those from other sources including procaryotes and other eukaryotes. Such products include enzymes, cofactors, hormones and the like.
  • the level of proteins, particularly modified proteins having improved amino acid distribution to improve the nutrient value of the plant, can be increased. This is achieved by the expression of such proteins having enhanced amino acid content.
  • methods to modify or alter the host endogenous genomic DNA are available. This includes altering the host native DNA sequence or a pre-existing transgenic sequence including regulatory elements, coding and non-coding sequences. These methods are also useful in targeting nucleic acids to pre-engineered target recognition sequences in the genome.
  • the genetically modified cell or plant described herein is generated using "custom" meganucleases produced to modify plant genomes (see, e.g., WO 2009/1 14321 ; Gao, et al., (2010) Plant Journal 1 : 176-187).
  • Other s/ie-directed engineering is through the use of zinc finger domain recognition coupled with the restriction properties of restriction enzyme. See, e.g., Urnov, et al., (2010) Nat Rev Genet. 11 (9):636-46; Shukla, et al., (2009) Nature 459(7245):437-41.
  • TILLING or “Targeting Induced Local Lesions IN Genomics” refers to a mutagenesis technology useful to generate and/or identify and to eventually isolate mutagenised variants of a particular nucleic acid with modulated expression and/or activity (McCallum, et al., (2000), Plant Physiology 123:439-442; McCallum, et al., (2000) Nature Biotechnology 18:455-457 and Colbert, et al., (2001 ) Plant Physiology 126:480- 484).
  • TILLING combines high density point mutations with rapid sensitive detection of the mutations.
  • EMS ethylmethanesulfonate
  • M1 ethylmethanesulfonate
  • M2 ethylmethanesulfonate
  • TILLING also allows selection of plants carrying mutant variants. These mutant variants may exhibit modified expression, either in strength or in location or in timing (if the mutations affect the promoter, for example). These mutant variants may exhibit higher or lower MS44 activity than that exhibited by the gene in its natural form.
  • TILLING combines high-density mutagenesis with high-throughput screening methods. The steps typically followed in TILLING are: (a) EMS mutagenesis (Redei and Koncz, (1992) In Methods in Arabidopsis Research, Koncz, et al. , eds. Singapore, World Scientific Publishing Co, pp. 16-82; Feldmann, et al., (1994) In Arabidopsis.
  • mutagenic methods can also be employed to introduce mutations in the MS44 gene.
  • Methods for introducing genetic mutations into plant genes and selecting plants with desired traits are well known.
  • seeds or other plant material can be treated with a mutagenic chemical substance, according to standard techniques.
  • chemical substances include, but are not limited to, the following: diethyl sulfate, ethylene imine, and N-nitroso-N-ethylurea.
  • ionizing radiation from sources such as X- rays or gamma rays can be used.
  • the present disclosure is exemplified with respect to plant fertility and more particularly with respect to plant male fertility.
  • Hybrid seed production requires elimination or inactivation of pollen produced by the female parent. Incomplete removal or inactivation of the pollen provides the potential for selfing, raising the risk that inadvertently self-pollinated seed will unintentionally be harvested and packaged with hybrid seed.
  • the selfed plants can be identified and selected; the selfed plants are genetically equivalent to the female inbred line used to produce the hybrid.
  • the selfed plants are identified and selected based on their decreased vigor relative to the hybrid plants. For example, female selfed plants of maize are identified by their less vigorous appearance for vegetative and/or reproductive characteristics, including shorter plant height, small ear size, ear and kernel shape, cob color or other characteristics.
  • Selfed lines also can be identified using molecular marker analyses (see, e.g., Smith and Wych, (1995) Seed Sci. Technol. 14:1-8). Using such methods, the homozygosity of the self-pollinated line can be verified by analyzing allelic composition at various loci in the genome.
  • hybrid plants are important and valuable field crops, plant breeders are continually working to develop high-yielding hybrids that are agronomically sound based on stable inbred lines.
  • the availability of such hybrids allows a maximum amount of crop to be produced with the inputs used, while minimizing susceptibility to pests and environmental stresses.
  • the plant breeder must develop superior inbred parental lines for producing hybrids by identifying and selecting genetically unique individuals that occur in a segregating population.
  • the present disclosure contributes to this goal, for example by providing plants that, when crossed, generate male sterile progeny, which can be used as female parental plants for generating hybrid plants.
  • Promoters useful for expressing a nucleic acid molecule of interest can be any of a range of naturally-occurring promoters known to be operative in plants or animals, as desired. Promoters that direct expression in cells of male or female reproductive organs of a plant are useful for generating a transgenic plant or breeding pair of plants of the disclosure.
  • the promoters useful in the present disclosure can include constitutive promoters, which generally are active in most or all tissues of a plant; inducible promoters, which generally are inactive or exhibit a low basal level of expression and can be induced to a relatively high activity upon contact of cells with an appropriate inducing agent; tissue-specific (or tissue-preferred) promoters, which generally are expressed in only one or a few particular cell types (e.g., plant anther cells) and developmental- or stage-specific promoters, which are active only during a defined period during the growth or development of a plant. Often promoters can be modified, if necessary, to vary the expression level. Certain embodiments comprise promoters exogenous to the species being manipulated.
  • the Ms45 gene introduced into ms45ms45 maize germplasm may be driven by a promoter isolated from another plant species; a hairpin construct may then be designed to target the exogenous plant promoter, reducing the possibility of hairpin interaction with non-target, endogenous maize promoters.
  • Exemplary constitutive promoters include the 35S cauliflower mosaic virus (CaMV) promoter promoter (Odell, et al., (1985) Nature 313:810-812), the maize ubiquitin promoter (Christensen, et al. , (1989) Plant Mol. Biol. 12:619-632 and Christensen, et al., (1992) Plant Mol. Biol.
  • CaMV cauliflower mosaic virus
  • CaMV cauliflower mosaic virus
  • ALS promoter US Patent Number 5,659,026
  • rice actin promoter US Patent Number 5,641 ,876; WO 2000/70067
  • maize histone promoter Bosset promoter
  • Other constitutive promoters include, for example, those described in US Patent Numbers 5,608,144 and 6,177,61 1 and PCT Publication Number WO 2003/102198.
  • Tissue-specific, tissue-preferred or stage-specific regulatory elements further include, for example, the AGL8/FRUITFULL regulatory element, which is activated upon floral induction (Hempel, et al., (1997) Development 124:3845-3853); root-specific regulatory elements such as the regulatory elements from the RCP1 gene and the LRP1 gene (Tsugeki and Fedoroff, (1999) Proc. Natl.
  • tissue-specific or stage-specific regulatory elements include the Zn13 promoter, which is a pollen-specific promoter (Hamilton, et al., (1992) Plant Mol. Biol. 18:21 1-218); the UNUSUAL FLORAL ORGANS ⁇ UFO) promoter, which is active in apical shoot meristem; the promoter active in shoot meristems (Atanassova, et al., (1992) Plant J. 2:291 ), the cdc2 promoter and cyc07 promoter (see, for example, Ito, et al., (1994) Plant Mol. Biol. 24:863-878; Martinez, et al., (1992) Proc. Natl.
  • tissue-specific promoters can be isolated using well known methods (see, e.g., US Patent Number 5,589,379).
  • Shoot-preferred promoters include shoot meristem-preferred promoters such as promoters disclosed in Weigel, et al. , (1992) Cell 69:843-859 (Accession Number M91208); Accession Number AJ131822; Accession Number Z71981 ; Accession Number AF049870 and shoot-preferred promoters disclosed in McAvoy, et al., (2003) Acta Hort. (ISHS) 625:379-385.
  • Inflorescence-preferred promoters include the promoter of chalcone synthase (Van der Meer, et al., (1992) Plant J.
  • tapetum-specific promoters such as the TA29 gene promoter (Mariani, et al., (1990) Nature 347:737; US Patent Number 6,372,967) and other stamen-specific promoters such as the MS45 gene promoter, 5126 gene promoter, BS7 gene promoter, PG47 gene promoter (US Patent Number 5,412,085; US Patent Number 5,545,546; Plant J 3(2):261-271 (1993)), SGB6 gene promoter (US Patent Number 5,470,359), G9 gene promoter (US Patent Number 5,8937,850; US Patent Number 5,589,610), SB200 gene promoter (WO 2002/26789), or the like.
  • TA29 gene promoter Menot al., (1990) Nature 347:737; US Patent Number 6,372,967
  • other stamen-specific promoters such as the MS45 gene promoter, 5126 gene promoter, BS7 gene promoter, PG47 gene promoter (US Patent Number 5,412,085; US Patent Number 5,545,
  • Tissue-preferred promoters of interest further include a sunflower pollen-expressed gene SF3 (Baltz, et al., (1992) The Plant Journal 2:713-721 ), B. napus pollen specific genes (Arnoldo, et al., (1992) J. Cell. Biochem, Abstract Number Y101204).
  • Tissue-preferred promoters further include those reported by Yamamoto, et al., (1997) Plant J. 12(2):255-265 (psaDb); Kawamata, et al. , (1997) Plant Cell Physiol. 38(7):792-803 (PsPAU ); Hansen, et al., (1997) Mol. Gen Genet.
  • a tissue-preferred promoter that is active in cells of male or female reproductive organs can be particularly useful in certain aspects of the present disclosure.
  • seed-developing promoters include both “seed-developing” promoters (those promoters active during seed development such as promoters of seed storage proteins) as well as “seed-germinating” promoters (those promoters active during seed germination). See, Thompson, et al., (1989) BioEssays 10:108.
  • seed-preferred promoters include, but are not limited to, Cim1 (cytokinin-induced message), cZ19B1 (maize 19 kDa zein), mil ps (myo-inositol-1 -phosphate synthase); see, WO 2000/1 1 177 and US Patent Number 6,225,529.
  • Gamma-zein is an endosperm-specific promoter.
  • Globulin-1 Glob-1
  • seed-specific promoters include, but are not limited to, bean ⁇ -phaseolin, napin, ⁇ -conglycinin, soybean lectin, cruciferin, and the like.
  • seed-specific promoters include, but are not limited to, maize 15 kDa zein, 22 kDa zein, 27 kDa zein, gamma-zein, waxy, shrunken 1 , shrunken 2, globulin 1 , etc.
  • An inducible regulatory element is one that is capable of directly or indirectly activating transcription of one or more DNA sequences or genes in response to an inducer.
  • the inducer can be a chemical agent such as a protein, metabolite, growth regulator, herbicide or phenolic compound or a physiological stress, such as that imposed directly by heat, cold, salt, or toxic elements or indirectly through the action of a pathogen or disease agent such as a virus or other biological or physical agent or environmental condition.
  • a plant cell containing an inducible regulatory element may be exposed to an inducer by externally applying the inducer to the cell or plant such as by spraying, watering, heating or similar methods.
  • An inducing agent useful for inducing expression from an inducible promoter is selected based on the particular inducible regulatory element.
  • transcription from the inducible regulatory element In response to exposure to an inducing agent, transcription from the inducible regulatory element generally is initiated de novo or is increased above a basal or constitutive level of expression.
  • the protein factor that binds specifically to an inducible regulatory element to activate transcription is present in an inactive form which is then directly or indirectly converted to the active form by the inducer.
  • Any inducible promoter can be used in the instant disclosure (See, Ward, et al. , (1993) Plant Mol. Biol. 22:361-366).
  • inducible regulatory elements include a metallothionein regulatory element, a copper-inducible regulatory element or a tetracycline-inducible regulatory element, the transcription from which can be effected in response to divalent metal ions, copper or tetracycline, respectively (Furst, et al., (1988) Cell 55:705-717; Mett, et al., (1993) Proc. Natl. Acad. Sci., USA 90:4567-4571 ; Gatz, et al., (1992) Plant J. 2:397-404; Roder, et al., (1994) Mol. Gen. Genet. 243:32-38).
  • inducible regulatory elements include a metallothionein regulatory element, a copper-inducible regulatory element or a tetracycline-inducible regulatory element, the transcription from which can be effected in response to divalent metal ions, copper or tetracycline, respectively (Furst, e
  • Inducible regulatory elements also include an ecdysone regulatory element or a glucocorticoid regulatory element, the transcription from which can be effected in response to ecdysone or other steroid (Christopherson, et al., (1992) Proc. Natl. Acad. Sci., USA 89:6314-6318; Schena, et al., (1991 ) Proc. Natl. Acad. Sci. USA 88:10421-10425; US Patent Number 6,504,082); a cold responsive regulatory element or a heat shock regulatory element, the transcription of which can be effected in response to exposure to cold or heat, respectively (Takahashi, et al. , (1992) Plant Physiol.
  • An inducible regulatory element also can be the promoter of the maize In2-1 or ln2-2 gene, which responds to benzenesulfonamide herbicide safeners (Hershey, et al., (1991 ) Mol. Gen. Gene. 227:229-237; Gatz, et al., (1994) Mol. Gen. Genet. 243:32-38) and the Tet repressor of transposon Tn10 (Gatz, et al., (1991 ) Mol. Gen. Genet. 227:229-237).
  • Stress inducible promoters include salt/water stress-inducible promoters such as P5CS (Zang, et al.
  • cold-inducible promoters such as, cor15a (Hajela, et al. , (1990) Plant Physiol. 93:1246-1252), cor15b (Wlihelm, et al., (1993) Plant Mol Biol 23:1073-1077), wsc120 (Ouellet, et al. , (1998) FEBS Lett. 423:324-328), ci7 (Kirch, et al., (1997) Plant Mol Biol. 33:897-909), ci21A (Schneider, et al. , (1997) Plant Physiol.
  • promoters include rip2 (US Patent Number 5,332,808 and US Patent Application Publication Number 2003/0217393) and rd29a (Yamaguchi-Shinozaki, et al., (1993) Mol. Gen. Genetics 236:331-340).
  • Certain promoters are inducible by wounding, including the Agrobacterium pmas promoter (Guevara-Garcia, et al. , (1993) Plant J. 4(3):495-505) and the Agrobacterium 0RF13 promoter (Hansen, et al. , (1997) Mol. Gen. Genet. 254(3):337-343).
  • a promoter is selected based, for example, on whether male fertility or female fertility is to be impacted
  • the promoter may be, for example, an MS45 gene promoter (US Patent Number 6,037,523), a 5126 gene promoter (US Patent Number 5,837,851 ), a BS7 gene promoter (WO 2002/063021 ), an SB200 gene promoter (WO 2002/26789), a TA29 gene promoter ⁇ Nature 347:737 (1990)), a PG47 gene promoter (US Patent Number 5,412,085; US Patent Number 5,545,546; Plant J 3(2):261- 271 (1993)) an SGB6 gene promoter (US Patent Number 5,470,359) a G9 gene promoter (US Patent Numbers 5,837,850 and 5,589,610) or the like.
  • the promoter can target female reproductive genes, for example an ovary specific promoter.
  • any promoter can be used that directs expression in the tissue of interest, including, for example, a constitutively active promoter such as an ubiquitin promoter, which generally effects transcription in most or all plant cells.
  • Additional regulatory elements active in plant cells and useful in the methods or compositions of the disclosure include, for example, the spinach nitrite reductase gene regulatory element (Back, et al. , (1991 ) Plant Mol. Biol. 17:9); a gamma zein promoter, an oleosin ole16 promoter, a globulin I promoter, an actin I promoter, an actin cl promoter, a sucrose synthetase promoter, an INOPS promoter, an EXM5 promoter, a globulin2 promoter, a b-32, ADPG-pyrophosphorylase promoter, an Ltpl promoter, an Ltp2 promoter, an oleosin ole17 promoter, an oleosin ole18 promoter, an actin 2 promoter, a pollen-specific protein promoter, a pollen-specific pectate lyase gene promoter or PG47 gene promoter
  • Plants suitable for purposes of the present disclosure can be monocots or dicots and include, but are not limited to, maize, wheat, barley, rye, sweet potato, bean, pea, chicory, lettuce, cabbage, cauliflower, broccoli, turnip, radish, spinach, asparagus, onion, garlic, pepper, celery, squash, pumpkin, hemp, zucchini, apple, pear, quince, melon, plum, cherry, peach, nectarine, apricot, strawberry, grape, raspberry, blackberry, pineapple, avocado, papaya, mango, banana, soybean, tomato, sorghum, sugarcane, sugar beet, sunflower, rapeseed, clover, tobacco, carrot, cotton, alfalfa, rice, potato, eggplant, cucumber, Arabidopsis thaliana and woody plants such as coniferous and deciduous trees.
  • a transgenic plant or genetically modified plant cell of the disclosure can be an angiosperm or gymnosperm.
  • Angiosperms are divided into two broad classes based on the number of cotyledons, which are seed leaves that generally store or absorb food; a monocotyledonous angiosperm has a single cotyledon and a dicotyledonous angiosperm has two cotyledons.
  • Angiosperms produce a variety of useful products including materials such as lumber, rubber and paper; fibers such as cotton and linen; herbs and medicines such as quinine and vinblastine; ornamental flowers such as roses and where included within the scope of the present disclosure, orchids and foodstuffs such as grains, oils, fruits and vegetables.
  • Angiosperms encompass a variety of flowering plants, including, for example, cereal plants, leguminous plants, oilseed plants, hardwood trees, fruit-bearing plants and ornamental flowers, which general classes are not necessarily exclusive.
  • Cereal plants which produce an edible grain, include, for example, corn, rice, wheat, barley, oat, rye, orchardgrass, guinea grass and sorghum.
  • Leguminous plants include members of the pea family (Fabaceae) and produce a characteristic fruit known as a legume.
  • leguminous plants include, for example, soybean, pea, chickpea, moth bean, broad bean, kidney bean, lima bean, lentil, cowpea, dry bean and peanut, as well as alfalfa, birdsfoot trefoil, clover and sainfoin.
  • Oilseed plants which have seeds that are useful as a source of oil, include soybean, sunflower, rapeseed (canola) and cottonseed.
  • Angiosperms also include hardwood trees, which are perennial woody plants that generally have a single stem (trunk).
  • trees examples include alder, ash, aspen, basswood (linden), beech, birch, cherry, cottonwood, elm, eucalyptus, hickory, locust, maple, oak, persimmon, poplar, sycamore, walnut, sequoia and willow. Trees are useful, for example, as a source of pulp, paper, structural material and fuel.
  • Angiosperms produce seeds enclosed within a mature, ripened ovary.
  • An angiosperm fruit can be suitable for human or animal consumption or for collection of seeds to propagate the species.
  • hops are a member of the mulberry family that are prized for their flavoring in malt liquor.
  • Fruit-bearing angiosperms also include grape, orange, lemon, grapefruit, avocado, date, peach, cherry, olive, plum, coconut, apple and pear trees and blackberry, blueberry, raspberry, strawberry, pineapple, tomato, cucumber and eggplant plants.
  • An ornamental flower is an angiosperm cultivated for its decorative flower.
  • Examples of commercially important ornamental flowers include rose, lily, tulip and chrysanthemum, snapdragon, camellia, carnation and petunia plants and can include orchids. It will be recognized that the present disclosure also can be practiced using gymnosperms, which do not produce seeds in a fruit.
  • Homozygosity is a genetic condition existing when identical alleles reside at corresponding loci on homologous chromosomes.
  • Heterozygosity is a genetic condition existing when different alleles reside at corresponding loci on homologous chromosomes.
  • Hemizygosity is a genetic condition existing when there is only one copy of a gene (or set of genes) with no allelic counterpart on the sister chromosome.
  • Backcrossing methods may be used to introduce a gene into the plants. This technique has been used for decades to introduce traits into a plant. An example of a description of this and other plant breeding methodologies that are well known can be found in references such as Plant Breeding Methodology, edit. Neal Jensen, John Wiley & Sons, Inc. (1988). In a typical backcross protocol, the original variety of interest (recurrent parent) is crossed to a second variety (nonrecurrent parent) that carries the single gene of interest to be transferred.
  • transgene any nucleic acid sequence which has been introduced into the genome of a cell by genetic engineering techniques.
  • a transgene may be a native DNA sequence or a heterologous DNA sequence.
  • the term native DNA sequence can refer to a nucleotide sequence which is naturally found in the cell but that may have been modified from its original form.
  • Certain constructs described herein comprise an element which interferes with formation, function, or dispersal of male gametes.
  • this can include use of genes which express a product cytotoxic to male gametes (See for example, US Patent Numbers 5,792,853; 5,689,049; PCT/EP89/00495); inhibit formation of a gene product important to male gamete function or formation (see, US Patent Numbers 5,859,341 ; 6,297,426); combine with another gene product to produce a substance preventing gene formation or function (see, US Patent Numbers 6,162,964; 6,013,859; 6,281 ,348; 6,399,856; 6,248,935; 6,750,868; 5,792,853); are antisense to or cause co-suppression of a gene critical to male gamete function or formation (see, US Patent Numbers 6,184,439; 5,728,926; 6,191 ,343; 5,728,558; 5,741 ,684)
  • cytotoxic genes can include, but are not limited to pectate lyase gene pelE, from Erwinia chrysanthermi (Kenn, et al. , (1986) J. Bacteroil 168:595); T-urf13 gene from cms-T maize mitochondrial genomes (Braun, et al., (1990) Plant Cell 2: 153; Dewey, et al., (1987) PNAS 84:5374); CytA toxin gene from Bacillus thuringiensis Israeliensis that causes cell membrane disruption (McLean, et al., (1987) J.
  • a suitable gene may also encode a protein involved in inhibiting pistil development, pollen stigma interactions, pollen tube growth or fertilization or a combination thereof.
  • genes that encode proteins that either interfere with the normal accumulation of starch in pollen or affect osmotic balance within pollen may also be suitable. These may include, for example, the maize alpha-amylase gene, maize beta- amylase gene, debranching enzymes such as Sugaryl and pullulanase, glucanase and SacB.
  • the DAM-methylase gene is used, discussed supra and at US Patent Numbers 5,792,852 and 5,689,049, the expression product of which catalyzes methylation of adenine residues in the DNA of the plant.
  • an .alpha. -amylase gene can be used with a male tissue-preferred promoter.
  • the aleurone layer cells will synthesize .alpha. -amylase, which participates in hydrolyzing starch to form glucose and maltose, so as to provide the nutrients needed for the growth of the germ (Rogers and Milliman, (1984) J. Biol. Chem.
  • the .alpha. -amylase gene used can be the Zea mays . alpha. -amylase- 1 gene. See, for example, Young, et al., Plant Physiol. 105(2):759-760 and GenBank Accession Numbers L25805, Gl:426481. See, also, U.S. Patent 8,013,218. Sequences encoding .alpha. -amylase are not typically found in pollen cells and when expression is directed to male tissue, the result is a breakdown of the energy source for the pollen grains and repression of pollen function.
  • the disclosure contemplates the use of promoters providing tissue-preferred expression, including promoters which preferentially express to the gamete tissue, male or female, of the plant.
  • the disclosure does not require that any particular gamete tissue- preferred promoter be used in the process, and any of the many such promoters known to one skilled in the art may be employed.
  • one such promoter is the 5126 promoter, which preferentially directs expression of the gene to which it is linked to male tissue of the plants, as described in US Patent Numbers 5,837,851 and 5,689,051.
  • promoter sequences may be isolated based on their sequence homology. In these techniques, all or part of a known promoter sequence is used as a probe which selectively hybridizes to other sequences present in a population of cloned genomic DNA fragments (i.e. genomic libraries) from a chosen organism.
  • nucleic acid sequences may be used to obtain sequences which correspond to these promoter sequences in species including, but not limited to, maize (corn; Zea mays), canola (Brassica napus, Brassica rapa ssp.), alfalfa ⁇ Medicago sativa), rice ⁇ Oryza sativa), rye (Secale cereale), sorghum (Sorghum bicolor, Sorghum vulgare), sunflower (Helianthus annuus), wheat (Triticum aestivum), soybean (Glycine max), tobacco (Nicotiana tabacum), potato (Solanum tuberosum), peanuts (Arachis hypogaea), cotton (Gossypium hirsutum), sweet potato (Ipomoea batatus), cassava (Manihot esculenta), coffee (Cofea spp.), coconut (Cocos nucifera), pineapple (Anana
  • the entire promoter sequence or portions thereof can be used as a probe capable of specifically hybridizing to corresponding promoter sequences.
  • probes include sequences that are unique and are preferably at least about 10 nucleotides in length and most preferably at least about 20 nucleotides in length.
  • Such probes can be used to amplify corresponding promoter sequences from a chosen organism by the well-known process of polymerase chain reaction (PCR). This technique can be used to isolate additional promoter sequences from a desired organism or as a diagnostic assay to determine the presence of the promoter sequence in an organism.
  • PCR polymerase chain reaction
  • sequences that correspond to a promoter sequence of the present disclosure and hybridize to a promoter sequence disclosed herein will be at least 50% homologous, 55% homologous, 60% homologous, 65% homologous, 70% homologous, 75% homologous, 80% homologous, 85% homologous, 90% homologous, 95% homologous and even 98% homologous or more with the disclosed sequence.
  • Fragments of a particular promoter sequence disclosed herein may operate to promote the pollen-preferred expression of an operably-linked isolated nucleotide sequence. These fragments will comprise at least about 20 contiguous nucleotides, preferably at least about 50 contiguous nucleotides, more preferably at least about 75 contiguous nucleotides, even more preferably at least about 100 contiguous nucleotides of the particular promoter nucleotide sequences disclosed herein. The nucleotides of such fragments will usually comprise the TATA recognition sequence of the particular promoter sequence.
  • Such fragments can be obtained by use of restriction enzymes to cleave the naturally-occurring promoter sequences disclosed herein; by synthesizing a nucleotide sequence from the naturally-occurring DNA sequence or through the use of PCR technology. See particularly, Mullis, et al., (1987) Methods Enzymol. 155:335-350 and Erlich, ed. (1989) PCR Technology (Stockton Press, New York). Again, variants of these fragments, such as those resulting from site-directed mutagenesis, are encompassed by the compositions of the present disclosure.
  • nucleotide sequences comprising at least about 20 contiguous nucleotides of the sequences set forth in SEQ ID NO: 64 - 106 are encompassed. These sequences can be isolated by hybridization, PCR, and the like. Such sequences encompass fragments capable of driving pollen-preferred expression, fragments useful as probes to identify similar sequences, as well as elements responsible for temporal or tissue specificity.
  • a regulatory "variant" is a modified form of a promoter wherein one or more bases have been modified, removed or added.
  • a routine way to remove part of a DNA sequence is to use an exonuclease in combination with DNA amplification to produce unidirectional nested deletions of double- stranded DNA clones.
  • a commercial kit for this purpose is sold under the trade name Exo-SizeTM (New England Biolabs, Beverly, Mass.).
  • this procedure entails incubating exonuclease III with DNA to progressively remove nucleotides in the 3' to 5' direction at 5' overhangs, blunt ends or nicks in the DNA template.
  • exonuclease III is unable to remove nucleotides at 3', 4-base overhangs.
  • Timed digests of a clone with this enzyme produce unidirectional nested deletions.
  • a regulatory sequence variant is a promoter formed by causing one or more deletions in a larger promoter. Deletion of the 5' portion of a promoter up to the TATA box near the transcription start site may be accomplished without abolishing promoter activity, as described by Zhu, et al., (1995) The Plant Cell 7:1681 -89. Such variants should retain promoter activity, particularly the ability to drive expression in specific tissues.
  • Biologically active variants include, for example, the native regulatory sequences of the disclosure having one or more nucleotide substitutions, deletions or insertions. Activity can be measured by Northern blot analysis, reporter activity measurements when using transcriptional fusions, and the like. See, for example, Sambrook, et al., (1989) Molecular Cloning: A Laboratory Manual (2nd ed. Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y.), herein incorporated by reference.
  • nucleotide sequences for the pollen-preferred promoters disclosed in the present disclosure are useful in the genetic manipulation of any plant when operably linked with an isolated nucleotide sequence whose expression is to be controlled to achieve a desired phenotypic response.
  • the nucleotide sequence operably linked to the regulatory elements disclosed herein can be an antisense sequence for a targeted gene.
  • antisense DNA nucleotide sequence is intended a sequence that is in inverse orientation to the 5'-to-3' normal orientation of that nucleotide sequence.
  • expression of the antisense DNA sequence prevents normal expression of the DNA nucleotide sequence for the targeted gene.
  • the antisense nucleotide sequence encodes an RNA transcript that is complementary to and capable of hybridizing with the endogenous messenger RNA (mRNA) produced by transcription of the DNA nucleotide sequence for the targeted gene. In this case, production of the native protein encoded by the targeted gene is inhibited to achieve a desired phenotypic response.
  • mRNA messenger RNA
  • Regulation of gene expression may be measured in terms of its effect on individual cells. Successful modulation of a trait may be accomplished with high stringency, for example impacting expression in all or nearly all cells of a particular cell type, or with lower stringency. Within a particular tissue, for example, modulation of expression in 98%, 95%, 90%, 80% or fewer cells may result in the desired phenotype. Further, for modification of assimilate partitioning and/or reduced competition for nitrogen between male and female reproductive structures, suppression of male fertility by 50% or even less may be effective and desirable.
  • the dominant male sterile allele of the Ms44 gene arose through a seed based EMS mutagenesis treatment of the W23 maize line and was found to be tightly linked to the C2 locus on chromosome 4 (Linkage between Ms44 and C2, Albertsen and Trimnell (1992) MNL 66:49).
  • a map-based cloning approach was undertaken to identify the Ms44 gene. An initial population of 414 individuals was used to rough map Ms44 to chromosome 4. An additional population of 2686 individuals was used for fine mapping. Marker Lab genotyping narrowed the region of the mutation to a 0.43cM interval on chromosome 4.
  • the Ms44 mutation was mapped to ⁇ 80kb region between markers made from the sequences AZM5_9212 (five recombinants) and AZM5_2221 (2 recombinants).
  • AZM5_9212 For4 SEQ ID NO: 1
  • AZM5_9212 Rev4 SEQ ID NO: 2
  • AZM5_9212 ForNest4 SEQ ID NO: 3
  • AZM5_9212 RevNest4 SEQ ID NO: 4
  • Primers AZM5_2221 For3 (SEQ ID NO: 5) and AZM5_2221 Rev3 (SEQ ID NO: 6) were used for an initial round of PCR followed by a second round of PCR using the primers AZM5_2221 ForNest3 (SEQ ID NO: 7) and AZM5_2221 RevNest3 (SEQ ID NO: 8).
  • the PCR product was digested with Bsgl and the banding pattern was analyzed to determine the genotypes at this locus..
  • a sequencing gap between BACs was present.
  • the gap was sequenced and, within this region, a gene, pco641570, was identified.
  • the first Met codon is found at nucleotide 1201 , with a 101 bp intron at nucleotides 1505-1605 and the stop codon ending at nucleotide 1613 (SEQ I D NO: 9).
  • the gene has an open reading frame of 312 bp which codes for a predicted protein of 104 amino acids (including the stop codon) (SEQ ID NO: 10).
  • the predicted protein has homology to a variety of proteins and contains the InterProscan accession domain IPR003612, a domain found in plant lipid transfer protein/seed storage/trypsin-alpha amylase inhibitors.
  • a secretory signal sequence (SSS) cleavage site was predicted, using SigCleave analysis, at amino acid 23. (von Heijne, G. "A new method for predicting signal sequence cleavage sites” Nucleic Acids Res.: 14:4683 (1986). Improved prediction of signal peptides: SignalP 3.0. , Bendtsen JD, Nielsen H, von Heijne G, Brunak S., J Mol Biol. 2004 Jul 16;340(4):783-95. Von Heijne, G. "Sequence Analysis in Molecular Biology: Treasure Trove or Trivial Pursuit” Acad. Press (1987) 1 13-1 17. See also the SI GCLEAVE program in the EMBOSS (European Molecular Biology Open Software Site) suite of applications online.)
  • SigCleave analysis of ms44 orthologs in related moncot species reveals another potential cleavage site between amino acids 37 and 38.
  • the protein is cysteine rich and BlastP analysis shows the highest homology to plant anther or tapetum specific genes such as the Lims or A9 genes.
  • RT-PCR analysis was performed on developing anther and leaf cDNAs to assess the expression of the ms44 gene.
  • Ms44-specific primers pco641570-5' (SEQ ID NO: 1 1 ) and pco641570-3'-2 (SEQ ID NO: 12) were used in an RT-PCR reaction with cDNA template from 0.5mm, 1.0mm, 1.5mm and 2.0mm anthers; anthers at pollen mother cell (PMC), quartet, early uninucleate and binucleate stages of microspore/pollen development; and leaf. Genomic DNA was also used as a template. Expression of ms44 begins early at the PMC stage and continues through quartet and early nucleate microspore stages but is absent by the binucleate stage of pollen development. No expression was detected in leaves.
  • the pco641570 gene was sequenced from the Ms44 mutant.
  • the first Met codon is found at nucleotide 1222, with a 101 bp intron at nucleotides 1526-1626 and the stop codon ends at nucleotide 1634 (SEQ ID NO: 13).
  • the sequence analysis revealed a nucleotide change which results in a translational change from an Alanine to a Threonine residue at amino acid 37 in the predicted protein (SEQ ID NO: 14).
  • This nucleotide change also created a BsmF1 restriction site in the mutant allele which is not found in the wildtype, which allows for distinguishing the two alleles by amplification of both Ms44 alleles by PCR and subsequent digestion of the products by BsmF1.
  • MsD-2629 is another dominant male sterile mutant found in maize and was also generated through EMS mutagenesis. This mutant was mapped and found to reside on chromosome 4 very near the Ms44 gene.
  • the Ms44 gene was PCR amplified and sequenced from MsD-2629 male sterile plants. Two different alleles were found through sequencing. One was a wild-type allele and the second allele had a single nucleotide change (SEQ ID NO: 152) which results in a translational change from the same Alanine residue as Ms44, but to a Valine at amino acid 37 in the predicted protein (SEQ ID NO: 153). This allele was found in all MsD-2629 male sterile plants tested and was not present in male fertile siblings. The MsD-2629 mutant represents a second Ms44 allele and was designated Ms44-2629.
  • TnT transcription/translation
  • genomic region was cloned for this allele, containing approximately 1.2Kb of upstream sequence (comprising putative promoter) and about 0.75 KB of sequence downstream of the stop codon.
  • This genomic sequence was sub-cloned into a transformation vector and designated PHP42163. The vector was used to transform maize plants through transformation. Thirty-six TO plants were grown to maturity and tassels were phenotyped for the presence or absence of pollen. Thirty four of the thirty six plants were completely male sterile.
  • transgenic plants were genotyped using primers pco641570-5' (SEQ ID NO: 1 1 ) and pco641570-3'-2 (SEQ ID NO: 12) in a PCR reaction and then digested with BsmF1 and run on a 1 % agarose gel. All thirty-four of the male sterile plants contained the mutant Ms44 allele as evidenced by the presence of two smaller bands produced by BsmF1 digestion. Genotyping revealed that the remaining two male fertile plants did not contain an intact mutant Ms44 allele. This confirms that the single nucleotide change in the Ms44 allele results in a dominant male sterile phenotype.
  • the point mutation in the Ms44 gene changes a codon from an Ala to a Thr, with a second allele having an Ala to Val change.
  • the affected amino acid is proposed to be at the -1 position of the SS cleavage site and the two mutations abolish SS cleavage of MS44 as shown by in vitro TnT assays.
  • the dominance of the mutation may be due to a defect in protein processing through the endoplasmic reticulum (ER) and not due to a functional role of the ms44 gene product as a lipid transfer protein. Since the MS44 protein is cysteine rich, an ER-tethered Ms44 protein may cross-link through disulfide bridges and inhibit overall protein processing in the anther that is ultimately required for male fertility.
  • transgenics In transgenics, one can stack a vector of tassel-specific-promoter-driven negative genes, or male sterility mutants, with other vectors that enhance vegetative or ear growth.
  • the combination of tassel reduction and enhancement of other organs can be particularly effective in diverting nutrients to the ear to achieve yield gain.
  • Tassel-specific promoters can be used to target silencing of the Tls1 gene in the tassel to knock down or knock-out the function of the gene in this tissue. This will reduce or eliminate tassel development, while the gene function in the ear remains intact.
  • Use of the tassel-specific promoters is not limited to Tls1 gene, but can be applied to driving any gene expression in tassel tissues to deliver a negative effect on tissue growth, for example to affect anther, pollen, or any cells that eventually impact male fertility.
  • Tassel- specific promoter candidates are identified based upon their native expression patterns, and are cloned and tested in transgenic plants to confirm their tassel-specificity.
  • EXAMPLE 3 tls1 mutant identification and characterization
  • the tassel-less (f/s7) mutant was described and mapped on the long arm of chromosome 1 (Albertsen, et al., (1993) Maize Genetics Newsletter 67:51-52).
  • the mutation was found to be located between two SNP markers, MZA5484-22 and MZA10765-46. These markers were used to screen for recombinants in a larger F2 population of 2985 individuals. All the recombinants were selected for self-pollination and 177 F3 ears were harvested. 177 F3 families were grown in rows in the field.
  • Phenotypes for all the individuals in rows were taken to determine each F2 line as homozygous wild- type, heterozygous or homozygous tlsl.
  • Leaf punches from 8 individuals of each F3 family were pooled together for genotyping. Using these lines, tlsl was confirmed to be between markers MZA5484 and MZA10765, which were converted to CAPS markers.
  • markers were used to fine map the tlsl mutation with the 177 F3 families. These markers were developed from DuPont proprietary sequences of known map positions, BAC-end sequences, and other low copy regions. The tlsl mutation was eventually mapped between markers c0375b06_10 and c0260e13_35.
  • the physical interval between the flanking markers c0375b06_10 and c0260e13_35 contained approximately four sequenced BAC clones based on the B73 physical map. Sequencing low copy regions within this interval revealed a very low level of polymorphism and the few markers available co-segregated with the tlsl phenotype.
  • NIP3-1 (hereafter known as NIP3-1 ) (SEQ ID NO: 62 - Genomic Sequence from B73; SEQ ID NO: 63 - CDS from B73, SEQ ID NO: 107 NIP3-1 protein), was unable to be amplified in homozygous tlsl individuals but could be amplified in homozygous wild-type and heterozygous lines.
  • PCR products from these reactions were used as templates for second reactions using the corresponding primer pairs: c0297o12_75-ForNest (SEQ ID NO: 42) and c0297o12_75-RevNest (SEQ ID NO: 43), c0297o12_76-ForNest (SEQ ID NO: 46) and c0297o12_76-RevNest (SEQ ID NO: 47), c0297o12_77-ForNest (SEQ ID NO: 50) and c0297o12_77-RevNest (SEQ ID NO: 51 ), c0297o12_78-ForNest (SEQ ID NO: 54) and c0297o12_78-RevNest (SEQ ID NO: 55).
  • a BAC library was constructed from homozygous tlsl plants in order to determine the nature of the mutation. Sequencing BAC clones covering the tlsl locus revealed a deletion of approximately 6.6kb in comparison to the B73 reference genome, corresponding to the NIP3-1 region. In addition, approximately 9kb of repetitive sequence was present in its place. Therefore, the tlsl phenotype is likely due to the deletion of NIP3-1 in homozygous mutant plants.
  • TUSC lines with Mutator (Mu) insertions in the NIP3.1 were identified to validate the candidate gene.
  • NIP3-1 specific primers D0143578 (SEQ ID NO: 56), D0143579 (SEQ ID NO: 57), D0143584 (SEQ ID NO: 58), or D0143583 (SEQ ID NO: 59) were used in combination with the Mu-specific primer, MuExt22D (SEQ ID NO: 60) to amplify the NIP3- 1 and Mutator junction regions.
  • PCR products from these reactions were used as templates for second reactions using the same NIP3-1 specific primers in combination with another Mu-specific primer, Mulnt19 (SEQ ID NO: 61 ).
  • the PCR product was run on a gel, the major bands excised, DNA extracted using a Gel Purification Kit (Qiagen) and sequenced. Sequencing results were BLASTed to confirm the Mu insertion in NIP3-1.
  • the TUCS lines which were heterozygous for the Mu insertion were used to pollinate heterozygous F3 plants at the tlsl locus.
  • the resulting progenies were phenotyped and genotyped. Plants were genotyped as described below:
  • c0297o12_75-Rev SEQ ID NO: 41
  • c0297o12_76-For SEQ ID NO: 44
  • c0297o12_76-Rev SEQ ID NO: 45
  • c0297o12_77-For SEQ ID NO: 48
  • c0297o12_77- Rev SEQ ID NO: 49
  • D0143583 SEQ ID NO: 59
  • D0143584 SEQ ID NO: 58
  • PCR products from these reactions were used as templates for second reactions using c0297o12_75-RevNest (SEQ ID NO: 43), c0297o12_76-ForNest (SEQ ID NO: 46), c0297o12_76-RevNest (SEQ ID NO: 47), c0297o12_77-ForNest (SEQ ID NO: 50), c0297o12_77-RevNest (SEQ ID NO: 51 ), D0143583 (SEQ ID NO: 59) and D0143584 (SEQ ID NO: 58) respectively in combination with the Mu-specific primer, Mulnt19 (SEQ ID NO: 61 ).
  • a positive PCR product indicated the presence of a Mu insertion.
  • c0297o12_75-For (SEQ ID NO: 40) was used in combination with c0297o12_75-Rev (SEQ ID NO: 41 ) and c0297o12_77-For (SEQ ID NO: 48) was used in combination with c0297o12_77-Rev (SEQ ID NO: 49).
  • PCR products from these reactions were used as templates for second reactions using c0297o12_75-ForNest (SEQ ID NO: 42) in combination with c0297o12_75-RevNest (SEQ ID NO: 43) and c0297o12_77-ForNest (SEQ ID NO: 50) in combination with c0297o12_77-RevNest (SEQ ID NO: 51 ), respectively.
  • the phenotyping results from the allelism test were compared with the genotyping results. Individuals without a Mu insertion were wild-type. Of the individuals that contained a Mu insertion, those that contained the wild-type allele of NIP3-1 had a wild- type phenotype while those that had the mutant allele of NIP3-1 mostly had a tlsl phenotype. The few aberrations were attributed to the incomplete penetrance of the tsl1 phenotype, which has been observed in the original description of the tlsl mutant (MNL 67:51 -52) and in the current study.
  • transgenic plants are separated into transgene (heterozygous) and null seed using a seed color marker.
  • Two different random assignments of treatments are made to each block of 54 pots, arranged as 6 rows of 9 columns and using 9 replicates of all treatments.
  • null seed of 5 events of the same construct are mixed and used as control for comparison of the 5 positive events in this block, making up 6 treatment combinations in each block.
  • 3 transgenic positive treatments and their corresponding nulls are randomly assigned to the 54 pots of the block, making 6 treatment combinations for each block, containing 9 replicates of all treatment combinations.
  • transgenic parameters are compared to a bulked construct null; in the second case, transgenic parameters are compared to the corresponding event null.
  • the events are assigned in groups of 5 events, the variances calculated for each block of 54 pots, but the block null means are pooled across blocks before mean comparisons are made.
  • the plants After emergence the plants are thinned to one seed per pot. Treatments routinely are planted on a Monday, emerge the following Friday and are harvested 18 days after planting. At harvest, plants are removed from the pots and the Turface® washed from the roots. The roots are separated from the shoot, placed in a paper bag and dried at 70°C for 70 hr. The dried plant parts (roots and shoots) are weighed and placed in a 50 ml conical tube with approximately 20 5/32 inch steel balls and ground by shaking in a paint shaker. Approximately, 30 mg of the ground tissue (weight recorded for later adjustment) is hydrolyzed in 2ml of 20% H 2 0 2 and 6M H 2 S0 4 for 30 min at 170°C.
  • OPA solution 5ul Mercaptoethanol + 1 ml OPA stock solution (make fresh, daily)
  • OPA stock 50mg o-phthadialdehyde (OPA - Sigma #P0657) dissolved in 1.5ml methanol + 4.4ml 1 M Borate buffer pH9.5 (3.09g H 3 B0 4 + 1 g NaOH in 50ml water) + 0.55ml 20% SDS (make fresh weekly)
  • Variance is calculated within each block using a nearest neighbor calculation as well as by Analysis of Variance (ANOV) using a completely random design (CRD) model.
  • An overall treatment effect for each block was calculated using an F statistic by dividing overall block treatment mean square by the overall block error mean square.
  • Transgenic plants will contain two or three doses of Gaspe Flint-3 with one dose of GS3 (GS3/(Gaspe-3)2X or GS3/(Gaspe-3)3X) and will segregate 1 :1 for a dominant transgene.
  • Plants will be planted in TURFACE®, a commercial potting medium and watered four times each day with 1 mM KN0 3 growth medium and with 2 mM KN0 3 or higher, growth medium. Control plants grown in 1 mM KN0 3 medium will be less green, produce less biomass and have a smaller ear at anthesis. Results are analyzed for statistical significance.
  • transgene will result in plants with improved plant growth in 1 mM KN0 3 when compared to a transgenic null. Thus biomass and greenness will be monitored during growth and compared to a transgenic null. Improvements in growth, greenness and ear size at anthesis will be indications of increased nitrogen utilization efficiency.
  • Transgenic maize plants are assayed for changes in root architecture at seedling stage, flowering time or maturity.
  • Assays to measure alterations of root architecture of maize plants include, but are not limited to the methods outlined below. To facilitate manual or automated assays of root architecture alterations, corn plants can be grown in clear pots.
  • Root mass dry weights. Plants are grown in Turface®, a growth medium that allows easy separation of roots. Oven-dried shoot and root tissues are weighed and a root/shoot ratio calculated.
  • Levels of lateral root branching The extent of lateral root branching (e.g., lateral root number, lateral root length) is determined by sub-sampling a complete root system, imaging with a flat-bed scanner or a digital camera and analyzing with WinRHIZOTM software (Regent Instruments Inc.).
  • Root band width measurements The root band is the band or mass of roots that forms at the bottom of greenhouse pots as the plants mature. The thickness of the root band is measured in mm at maturity as a rough estimate of root mass.
  • Nodal root count The number of crown roots coming off the upper nodes can be determined after separating the root from the support medium (e.g., potting mix). In addition the angle of crown roots and/or brace roots can be measured. Digital analysis of the nodal roots and amount of branching of nodal roots form another extension to the aforementioned manual method.
  • Seeds of Arabidopsis thaliana (control and transgenic line), ecotype Columbia, are surface sterilized (Sanchez, et al. , 2002) and then plated on to Murashige and Skoog (MS) medium containing 0.8% (w/v) BactoTM-Agar (Difco). Plates are incubated for 3 days in darkness at 4°C to break dormancy (stratification) and transferred thereafter to growth chambers (Conviron, Manitoba, Canada) at a temperature of 20°C under a 16-h light/8-h dark cycle. The average light intensity is 120 ⁇ / ⁇ 2/8. Seedling are grown for 12 days and then transferred to soil based pots.
  • Potted plants are grown on a nutrient- free soil LB2 Metro-Mix® 200 (Scott's Sierra Horticultural Products, Marysville, OH, USA) in individual 1.5-in pots (Arabidopsis system; Lehle Seeds, Round Rock, TX, USA) in growth chambers, as described above. Plants are watered with 0.6 or 6.5 mM potassium nitrate in the nutrient solution based on Murashige and Skoog (MS free Nitrogen) medium. The relative humidity is maintained around 70%. 16-18 days later plant shoots are collected for evaluation of biomass and SPAD readings.
  • EXAMPLE 8 Agrobacterium mediated transformation into maize
  • Maize plants can be transformed to overexpress a nucleic acid sequence of interest in order to examine the resulting phenotype.
  • Immature embryos are dissected from caryopses and placed in a 2 mL microtube containing 2 mL PHI-A medium.
  • PHI-A medium is removed with 1 mL micropipettor and 1 mL Agrobacterium suspension is added. Tube is gently inverted to mix. The mixture is incubated for 5 min at room temperature. 2.2 Co-Culture Step
  • the Agrobacterium suspension is removed from the infection step with a 1 mL micropipettor. Using a sterile spatula the embryos are scraped from the tube and transferred to a plate of PHI-B medium in a 100x15 mm Petri dish. The embryos are oriented with the embryonic axis down on the surface of the medium. Plates with the embryos are cultured at 20°C, in darkness, for 3 days. L-Cysteine can be used in the co- cultivation phase. With the standard binary vector, the co-cultivation medium supplied with 100-400 mg/L L-cysteine is critical for recovering stable transgenic events.
  • Embryonic tissue propagated on PHI-D medium is subcultured to PHI-E medium
  • somatic embryo maturation medium in 100x25 mm Petri dishes and incubated at 28°C, in darkness, until somatic embryos mature, for about 10-18 days.
  • Individual, matured somatic embryos with well-defined scutellum and coleoptile are transferred to PHI-F embryo germination medium and incubated at 28°C in the light (about 80 ⁇ from cool white or equivalent fluorescent lamps).
  • regenerated plants about 10 cm tall, are potted in horticultural mix and hardened-off using standard horticultural methods.
  • PHI-A 4g/L CHU basal salts, 1.0 mL/L 1000X Eriksson's vitamin mix, 0.5mg/L thiamin HCL, 1 .5 mg/L 2,4-D, 0.69 g/L L-proline, 68.5 g/L sucrose, 36g/L glucose, pH 5.2. Add 100 ⁇ acetosyringone, filter-sterilized before using.
  • PHI-B PHI-A without glucose, increased 2,4-D to 2mg/L, reduced sucrose to 30 g/L and supplemented with 0.85 mg/L silver nitrate (filter-sterilized), 3.0 g/L Gelrite®, 100 ⁇ acetosyringone (filter-sterilized), pH 5.8.
  • PHI-C PHI-B without Gelrite® and acetosyringone, reduced 2,4-D to 1 .5 mg/L and supplemented with 8.0 g/L agar, 0.5 g/L Ms-morpholino ethane sulfonic acid (MES) buffer, 100mg/L carbenicillin (filter-sterilized).
  • MES Ms-morpholino ethane sulfonic acid
  • PHI-D PHI-C supplemented with 3mg/L bialaphos (filter-sterilized).
  • PHI-E 4.3 g/L of Murashige and Skoog (MS) salts, (Gibco, BRL 1 1 1 17- 074), 0.5 mg/L nicotinic acid, 0.1 mg/L thiamine HCI, 0.5mg/L pyridoxine HCI, 2.0 mg/L glycine, 0.1 g/L myo-inositol, 0.5 mg/L zeatin (Sigma, cat.no.
  • MS Murashige and Skoog
  • IAA indole acetic acid
  • ABA abscisic acid
  • 60 g/L sucrose 60 g/L sucrose
  • 3 mg/L bialaphos filter-sterilized
  • 100 mg/L carbenicillin filter-sterilized
  • 8g/L agar pH 5.6.
  • PHI-F PHI-E without zeatin, IAA, ABA; sucrose reduced to 40 g/L; replacing agar with 1 .5 g/L Gelrite®; pH 5.6.
  • Plants can be regenerated from the transgenic callus by first transferring clusters of tissue to N6 medium supplemented with 0.2 mg per liter of 2,4-D. After two weeks the tissue can be transferred to regeneration medium (Fromm, et al., (1990) Bio/Technology 8:833-839).
  • T1 plants can be analyzed for phenotypic changes.
  • image analysis T1 plants can be analyzed for phenotypical changes in plant area, volume, growth rate and color analysis at multiple times during growth of the plants. Alteration in root architecture can be assayed as described herein.
  • Subsequent analysis of alterations in agronomic characteristics can be done to determine whether plants containing the nucleic acid sequence of interest have an improvement of at least one agronomic characteristic, when compared to the control (or reference) plants that have not been so transformed.
  • the alterations may also be studied under various environmental conditions.
  • nucleic acid sequence of interest functions in maize to alter nitrogen use efficiency.
  • Electroporation competent cells 40 ⁇
  • Agrobacterium tumefaciens LBA4404 containing PHP10523
  • PHP10523 contains VI R genes for T-DNA transfer, an Agrobacterium low copy number plasmid origin of replication, a tetracycline resistance gene and a cos site for in vivo DNA biomolecular recombination.
  • the electroporation cuvette is chilled on ice.
  • the electroporator settings are adjusted to 2.1 kV.
  • a DNA aliquot (0.5 ⁇ _ JT (US Patent Number 7,087,812) parental DNA at a concentration of 0.2 ⁇ g -1.0 ⁇ g in low salt buffer or twice distilled H 2 0) is mixed with the thawed Agrobacterium cells while still on ice. The mix is transferred to the bottom of electroporation cuvette and kept at rest on ice for 1-2 min. The cells are electroporated (Eppendorf electroporator 2510) by pushing "Pulse" button twice (ideally achieving a 4.0 msec pulse). Subsequently 0.5 ml 2xYT medium (or SOCmedium) are added to cuvette and transferred to a 15 ml Falcon tube. The cells are incubated at 28-30°C, 200-250 rpm for 3 h.
  • Option 1 Overlay plates with 30 ⁇ of 15 mg/ml Rifampicin.
  • LBA4404 has a chromosomal resistance gene for Rifampicin. This additional selection eliminates some contaminating colonies observed when using poorer preparations of LBA4404 competent cells.
  • Option 2 Perform two replicates of the electroporation to compensate for poorer electrocompetent cells.
  • a single colony for each putative co-integrate is picked and inoculated with 4 ml #60A with 50 mg/l Spectinomycin. The mix is incubated for 24 h at 28°C with shaking. Plasmid DNA from 4 ml of culture is isolated using Qiagen Miniprep + optional PB wash. The DNA is eluted in 30 ⁇ . Aliquots of 2 ⁇ are used to electroporate 20 ⁇ of DH10b + 20 ⁇ of dd H 2 0 as per above.
  • a 15 ⁇ aliquot can be used to transform 75-100 ⁇ of InvitrogenTM Library Efficiency DH5a.
  • the cells are spread on LB medium plus 50mg/mL Spectinomycin plates (#34T medium) and incubated at 37°C overnight.
  • the plasmid DNA is isolated from 4 ml of culture using QIAprep® Miniprep with optional PB wash (elute in 50 ⁇ ) and 8 ⁇ are used for digestion with Sail (using JT parent and PHP10523 as controls).
  • a vector can be transformed into embryogenic maize callus by particle bombardment, generally as described by Tomes, et al. , Plant Cell, Tissue and Organ Culture: Fundamental Methods, Eds. Gamborg and Phillips, Chapter 8, pgs. 197-213 (1995) and as briefly outlined below.
  • Transgenic maize plants can be produced by bombardment of embryogenically responsive immature embryos with tungsten particles associated with DNA plasmids.
  • the plasmids typically comprise or consist of a selectable marker and an unselected structural gene, or a selectable marker and a polynucleotide sequence or subsequence, or the like.
  • tungsten particles General Electric
  • 0.5 to 1.8 ⁇ , preferably 1 to 1.8 ⁇ , and most preferably 1 ⁇ are added to 2 ml of concentrated nitric acid.
  • This suspension is sonicated at 0°C. for 20 minutes (Branson Sonifier Model 450, 40% output, constant duty cycle).
  • Tungsten particles are pelleted by centrifugation at 10000 rpm (Biofuge) for one minute and the supernatant is removed. Two milliliters of sterile distilled water are added to the pellet and brief sonication is used to resuspend the particles.
  • the suspension is pelleted, one milliliter of absolute ethanol is added to the pellet and brief sonication is used to resuspend the particles.
  • Rinsing, pelleting and resuspending of the particles are performed two more times with sterile distilled water and finally the particles are resuspended in two milliliters of sterile distilled water.
  • the particles are subdivided into 250- ⁇ aliquots and stored frozen.
  • the stock of tungsten particles are sonicated briefly in a water bath sonicator
  • the vectors are typically cis: that is, the selectable marker and the gene (or other polynucleotide sequence) of interest are on the same plasmid.
  • Plasmid DNA is added to the particles for a final DNA amount of 0.1 to 10 ⁇ 9 in 10 ⁇ _ total volume and briefly sonicated.
  • 10 ⁇ 9 (1 ⁇ g/ ⁇ L in TE buffer) total DNA is used to mix DNA and particles for bombardment.
  • Fifty microliters (50 ⁇ _) of sterile aqueous 2.5 M CaCI 2 are added and the mixture is briefly sonicated and vortexed.
  • Twenty microliters (20 ⁇ _) of sterile aqueous 0.1 M spermidine are added and the mixture is briefly sonicated and vortexed.
  • the mixture is incubated at room temperature for 20 minutes with intermittent brief sonication.
  • the particle suspension is centrifuged and the supernatant is removed.
  • Immature embryos of maize are the target for particle bombardment-mediated transformation. Ears from F1 plants are selfed or sibbed and embryos are aseptically dissected from developing caryopses when the scutellum first becomes opaque. This stage occurs about 9 13 days post-pollination and most generally about 10 days post- pollination, depending on growth conditions. The embryos are about 0.75 to 1.5 millimeters long. Ears are surface sterilized with 20 50% Clorox® for 30 minutes, followed by three rinses with sterile distilled water.
  • Immature embryos are cultured with the scutellum oriented upward, on embryogenic induction medium comprised of N6 basal salts, Eriksson vitamins, 0.5 mg/l thiamine HCI, 30 gm/l sucrose, 2.88 gm/l L-proline, 1 mg/l 2,4-dichlorophenoxyacetic acid, 2 gm/l Gelrite® and 8.5 mg/l AgN0 3 , Chu, et al., (1975) Sci. Sin. 18:659; Eriksson, (1965) Physiol. Plant 18:976.
  • the medium is sterilized by autoclaving at 121 °C for 15 minutes and dispensed into 100x25 mm Petri dishes.
  • AgN0 3 is filter-sterilized and added to the medium after autoclaving.
  • the tissues are cultured in complete darkness at 28°C. After about 3 to 7 days, most usually about 4 days, the scutellum of the embryo swells to about double its original size and the protuberances at the coleorhizal surface of the scutellum indicate the inception of embryogenic tissue. Up to 100% of the embryos display this response, but most commonly, the embryogenic response frequency is about 80%.
  • the embryos are transferred to a medium comprised of induction medium modified to contain 120 gm/l sucrose.
  • the embryos are oriented with the coleorhizal pole, the embryogenically responsive tissue, upwards from the culture medium.
  • Ten embryos per Petri dish are located in the center of a Petri dish in an area about 2 cm in diameter.
  • the embryos are maintained on this medium for 3 to 16 hours, preferably 4 hours, in complete darkness at 28°C just prior to bombardment with particles associated with plasmid DNAs containing the selectable and unselectable marker genes.
  • the particle-DNA agglomerates are accelerated using a DuPont PDS-1000 particle acceleration device.
  • the particle-DNA agglomeration is briefly sonicated and 10 ⁇ are deposited on macrocarriers and the ethanol is allowed to evaporate.
  • the macrocarrier is accelerated onto a stainless-steel stopping screen by the rupture of a polymer diaphragm (rupture disk).
  • Rupture is affected by pressurized helium.
  • the velocity of particle-DNA acceleration is determined based on the rupture disk breaking pressure. Rupture disk pressures of 200 to 1800 psi are used, with 650 to 1 100 psi being preferred and about 900 psi being most highly preferred. Multiple disks are used to affect a range of rupture pressures.
  • the shelf containing the plate with embryos is placed 5.1 cm below the bottom of the macrocarrier platform (shelf #3).
  • a rupture disk and a macrocarrier with dried particle-DNA agglomerates are installed in the device.
  • the He pressure delivered to the device is adjusted to 200 psi above the rupture disk breaking pressure.
  • a Petri dish with the target embryos is placed into the vacuum chamber and located in the projected path of accelerated particles.
  • a vacuum is created in the chamber, preferably about 28 in Hg. After operation of the device, the vacuum is released and the Petri dish is removed.
  • Bombarded embryos remain on the osmotically-adjusted medium during bombardment, and 1 to 4 days subsequently.
  • the embryos are transferred to selection medium comprised of N6 basal salts, Eriksson vitamins, 0.5 mg/l thiamine HCI, 30 gm/l sucrose, 1 mg/l 2,4-dichlorophenoxyacetic acid, 2 gm/l Gelrite®, 0.85 mg/l Ag N0 3 and 3 mg/l bialaphos (Herbiace, Meiji). Bialaphos is added filter-sterilized.
  • the embryos are subcultured to fresh selection medium at 10 to 14 day intervals.
  • embryogenic tissue After about 7 weeks, embryogenic tissue, putatively transformed for both selectable and unselected marker genes, proliferates from a fraction of the bombarded embryos. Putative transgenic tissue is rescued and that tissue derived from individual embryos is considered to be an event and is propagated independently on selection medium. Two cycles of clonal propagation are achieved by visual selection for the smallest contiguous fragments of organized embryogenic tissue.
  • a sample of tissue from each event is processed to recover DNA.
  • the DNA is restricted with a restriction endonuclease and probed with primer sequences designed to amplify DNA sequences overlapping the coding and non-coding portion of the plasmid.
  • Embryogenic tissue with amplifiable sequence is advanced to plant regeneration.
  • embryogenic tissue is subcultured to a medium comprising MS salts and vitamins (Murashige and Skoog, (1962) Physiol. Plant 15:473), 100 mg/l myo-inositol, 60 gm/l sucrose, 3 gm/l Gelrite®, 0.5 mg/l zeatin, 1 mg/l indole-3-acetic acid, 26.4 ng/l cis-trans-abscissic acid and 3 mg/l bialaphos in 100X25 mm Petri dishes and is incubated in darkness at 28°C until the development of well-formed, matured somatic embryos is seen. This requires about 14 days.
  • Well-formed somatic embryos are opaque and cream-colored and are comprised of an identifiable scutellum and coleoptile.
  • the embryos are individually subcultured to a germination medium comprising MS salts and vitamins, 100 mg/l myo-inositol, 40 gm/l sucrose and 1.5 gm/l Gelrite® in 100x25 mm Petri dishes and incubated under a 16 hour light:8 hour dark photoperiod and 40 meinsteinsm sec from cool-white fluorescent tubes.
  • the somatic embryos germinate and produce a well-defined shoot and root.
  • the individual plants are subcultured to germination medium in 125x25 mm glass tubes to allow further plant development.
  • the plants are maintained under a 16 hour light:8 hour dark photoperiod and 40 meinsteinsm sec from cool-white fluorescent tubes. After about 7 days, the plants are well-established and are transplanted to horticultural soil, hardened off and potted into commercial greenhouse soil mixture and grown to sexual maturity in a greenhouse. An elite inbred line is used as a male to pollinate regenerated transgenic plants.
  • Soybean embryos are bombarded with a plasmid comprising a preferred promoter operably linked to a heterologous nucleotide sequence comprising a polynucleotide sequence or subsequence, as follows.
  • cotyledons of 3 5 mm in length are dissected from surface-sterilized, immature seeds of the soybean cultivar A2872, then cultured in the light or dark at 26°C on an appropriate agar medium for six to ten weeks. Somatic embryos producing secondary embryos are then excised and placed into a suitable liquid medium. After repeated selection for clusters of somatic embryos that multiply as early, globular-staged embryos, the suspensions are maintained as described below.
  • Soybean embryogenic suspension cultures can be maintained in 35 ml liquid media on a rotary shaker, 150 rpm, at 26°C with fluorescent lights on a 16:8 hour day/night schedule. Cultures are sub-cultured every two weeks by inoculating approximately 35 mg of tissue into 35 ml of liquid medium.
  • Soybean embryogenic suspension cultures may then be transformed by the method of particle gun bombardment (Klein, et al., (1987) Nature (London) 327:70-73, US Patent Number 4,945,050).
  • a DuPont BiolisticTM PDS1000/HE instrument helium retrofit
  • a selectable marker gene that can be used to facilitate soybean transformation is a transgene composed of the 35S promoter from Cauliflower Mosaic Virus (Odell, et al., (1985) Nature 313:810-812), the hygromycin phosphotransferase gene from plasmid pJR225 (from E. coli; Gritz, et al., (1983) Gene 25:179-188) and the 3' region of the nopaline synthase gene from the T-DNA of the Ti plasmid of Agrobacterium tumefaciens.
  • the expression cassette of interest comprising the preferred promoter and a heterologous polynucleotide, can be isolated as a restriction fragment. This fragment can then be inserted into a unique restriction site of the vector carrying the marker gene.
  • Approximately 300 400 mg of a two-week-old suspension culture is placed in an empty 60X5 mm petri dish and the residual liquid removed from the tissue with a pipette.
  • approximately 5-10 plates of tissue are normally bombarded.
  • Membrane rupture pressure is set at 1 100 psi, and the chamber is evacuated to a vacuum of 28 inches mercury.
  • the tissue is placed approximately 3.5 inches away from the retaining screen and bombarded three times. Following bombardment, the tissue can be divided in half and placed back into liquid and cultured as described above.
  • the liquid media may be exchanged with fresh media and eleven to twelve days post-bombardment with fresh media containing 50 mg/ml hygromycin. This selective media can be refreshed weekly.
  • Green, transformed tissue may be observed growing from untransformed, necrotic embryogenic clusters. Isolated green tissue is removed and inoculated into individual flasks to generate new, clonally propagated, transformed embryogenic suspension cultures. Each new line may be treated as an independent transformation event. These suspensions can then be subcultured and maintained as clusters of immature embryos or regenerated into whole plants by maturation and germination of individual somatic embryos.
  • EXAMPLE 12 Ear development of sterile or fertile plants at varying Nitrogen levels
  • Male sterility reduces the nutrient requirement for tassel development, resulting in improved ear development at anthesis.
  • male sterile and male fertile sibs were grown in varying levels of nitrogen fertility and sampled at -50% pollen shed.
  • Male sterile plants produced larger ears in each nitrogen fertility level, relative to their male fertile sibs.
  • the proportion of male sterile plants with emerged silks was also greater than for the fertile sib plants.
  • biomass total above ground plant minus the ear dry weight
  • was greater for plants grown in the higher nitrogen fertility compared to plants grown in lower nitrogen fertility plots there was no effect of male sterility on biomass. This shows the positive effect of male sterility on the ability of the plant to produce a heavier, more fully developed ear and improve silking without affecting overall vegetative growth.
  • the plant population in the nitrogen multiple rate experiment was 32,000 plants per acre (PPA) whereas 32,000, 48,000 and 64,000 PPA densities were used in the plant population study.
  • the N fertility regime in the population study was 180 units N pre-plant for all populations, followed by 95 units N side dressed at V6 (275 total units N) in all plots.
  • the 48,000 PPA plots were supplemented with an additional 50 units of N 10 days prior to flowering (325 total units N) and the 64,000 PPA plots were supplemented with an additional 100 units of N 10 days prior to flowering (375 total units N).
  • Silk number and kernels per ear also had significant treatment interactions and were likely due to a steeper rate of increase in silk number with N fertility in the male sterile plants than in the male fertile plants.
  • the difference in yield between male fertile and male sterile plants was much greater at low N than at higher N levels.
  • the difference between male sterile and male fertile plants was 84% whereas the difference in yield between male sterile and male fertile plants was 15% at 150 lb/acre N rate.
  • an average increase of about 37 bu/acre was observed.
  • the average increase was about 13 bu/acre.
  • SPAD was significantly different in response to N fertility and in response to male sterility but the response to N fertility of male sterile and male fertile plants was parallel, suggesting SPAD could not account for the difference in yield between male sterile and male fertile plants in response to N fertility.
  • Kernel number of male-sterile and male-fertile plants in response to N fertility showed slopes similar to the male-sterile and male-fertile yield response to N fertility, which suggests the increase in yield of male-sterile plants might be related to increased kernel number. Differences in yield between male-sterile and male-fertile hybrids across N fertilities could nearly be accounted for by the sum of the differences in ears per plot and kernels per ear between male-sterile and male-fertile hybrids across N fertilities. These data are in agreement with the hypothesis that ear development is less encumbered by tassel development in male-sterile plants, resulting in more fully developed ears (kernels/ear) with a greater success rate of ear production (ear/plot) under low N. In one of the hybrid trials, the ear dry weight of male-sterile plants increased about 62% compared to the ear dry weight from male- fertile plants.
  • the genetic male sterile hybrid also responded better than the male fertile hybrid in the population stress experiment. Though there was no effect of population on grain yield, the genetic male-sterile hybrid outperformed the male-fertile hybrid by 40% (59 bu/a) in all populations tested (see, Figure 7A). In addition, in a separate trial, an average increase of about 8 bu/acre was observed (see, Figure 7B).
  • Phenotype of the tlsl mutant is shown in Figure 8.
  • a positional cloning approach was undertaken to clone tlsl (Figure 9).
  • the tlsl region was roughly mapped on Chr1 using 75 individuals from a tlsl x Mo17 F2 population.
  • the first round of fine mapping is indicated by the red font, tlsl was narrowed to a 15cM region using 2985 F2 individuals.
  • the resulting 177 recombinants were selfed and the progeny from each line were pooled together for further fine mapping, indicated by the green font.
  • the 177 F3 families were used to narrow the tlsl interval to a four BAC region, containing no additional informative markers.
  • NIP3-1 Sequence analysis of NIP3-1 from maize revealed a high level of similarity to NIP5;1 from Arabidopsis (AtNIP5;1 ) and NIP3;1 from rice (OsNIP3; 1 ) and phylogenetic studies showed that they are closely related proteins in the NIP II subgroup (Liu, et al. , (2009) BCM Genomics 10:1471-2164). ( Figure 15). These results suggest that NIP3-1 in maize is involved in boron uptake, and boron is needed for reproductive development. Studies can be performed which manipulate the expression of tlsl in the development of hybrid maize for yield improvement under normal and stress conditions (e.g., nitrogen and water stress).
  • normal and stress conditions e.g., nitrogen and water stress
  • NIP3-1 would be down-regulated in a tissue-specific manner (i.e., in the tassel), resulting in plants with no tassels that do not exhibit any of the other pleotropic effects associated with boron deficiency (e.g., underdeveloped ears).
  • the resources that would be needed for tassel development may be allocated to the ear and shading effects from tassels would be minimized, resulting in an increased yield over other male sterility techniques in which a tassel is present. This same approach may be applied to any genes involved in the transport of boron.
  • Wild type and mutant plants from the F2 mapping population of tlsl x Mo17 were planted. Half of the mutant and wild type plants were treated once a week from ⁇ V2 to ⁇ V6 stage with a foliar boron spray consisting of 0.0792% B 2 0 2 and 0.0246% elemental Boron. It was observed that the mutant plants treated with the boron spray exhibited an increased number of tassel branches, which were longer and reminiscent of wild type in comparison to the untreated mutant plants. In addition, ears of the treated mutant plants appeared to be recovered as well. Wild type plants treated with boron had no discernable difference from untreated wild type plants. Recovered mutant plants were self-pollinated for a progeny test.
  • Progeny from selfing the recovered mutant plants were planted along with wild type for a control. Half the mutant progeny was treated with the boron spray as described above and half were left untreated. Tassel branch number ( Figure 1 1 ), branch length ( Figure 12) and ear length ( Figure 13) were measured from 24 wild type plants, 26 mutant plants treated with the boron spray and 29 untreated mutant plants. In comparison to the untreated mutant plants, mutant plants treated with the boron spray exhibited an increased number of tassel branches, increased tassel branch length, and an increased ear length similar to wild type plants ( Figures 1 1-13).
  • Homozygous tlsl plants have reduced tassel growth or substantially lack functional tassel for normal ear development. Therefore, the quantity of seeds from tlsl mutant plants or plants with reduced tassel development due to a deficiency in boron uptake are not to the levels needed for large-scale seed production. Because exogenous boron application rescues tassle development and growth in the tlsl mutant background, boron application is an option to increase seed production from tlsl plants.
  • exogenous boron e.g., as a foliar spray
  • can be applied at various stages of reproductive growth e.g., V2-V12 or V2-V8
  • varying levels of boron e.g., 10-1000 ppm
  • boron application can coincide with the transition from vegetative to reproductive state, e.g., V4-V5 depending on plant growing conditions.
  • additional weaker or stronger alleles of tlsl are obtained by performing available screens, e.g., through Targeting Induced Local Lesions in Genomes (TILLING), McCallum, et al., (2000) Nat Biotechnol 18:455-457.
  • Additional alleles of Tlsl can include those variants that completely block boron transport resulting in substantial loss of tassel growth and development and those variants that result in for example, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or 95% reduction in tassel development as evidenced by the reduced pollen production or other suitable parameter known to those or ordinary skill in the art.
  • the effect of reduced male fertility on yield of maize grown under drought stress conditions is evaluated in a field study.
  • the field study is conducted in a managed stress field environment.
  • the field location receives little or no rainfall during the growing season, allowing for the controlled imposition of drought stress by removing the irrigation at various stages of development. This field location has no insect or disease pressure to interfere with the interpretation of hybrid performance under drought.
  • Maize is extremely sensitive to drought stress during the flowering period. Typically, development of the ears, exsertion of the silks and pollination of the ovaries are all inhibited by drought stress. The sensitivity of these processes is a major factor in reducing yield under drought stress. Alleviation of this sensitivity is an effective method of improving drought stress in maize. Male-sterile plants will partition more assimilates to the ear during this critical period than will male-fertile plants, thus making male-sterile plants more tolerant to this stress. The male-sterile plants will exsert silks more rapidly, resulting in more efficient pollination of those ovaries and a higher final kernel number/plant. The improvement of this critical reproductive process results in greater yield at harvest.
  • a method for production of male-sterile hybrid plants is provided.
  • female parent (male-sterile) plants of inbred A homozygous recessive for a male-fertility gene
  • inbred B is similarly homozygous recessive for the male-fertility gene; however, Inbred B is hemizygous for a heterologous construct.
  • This construct comprises (a) the dominant allele of the male-fertility gene, which complements the recessive genotype and restores fertility to inbred B; (b) a genetic element which results in disruption of the formation, function, or dispersal of pollen; (c) optionally, a marker gene, which may be a marker expressed in seed.
  • a marker gene which may be a marker expressed in seed.
  • the pollinator can be planted in rows or plots separate from the male-sterile- hybrid seed, or can be interspersed in an orderly or random way among the male-sterile- hybrid plants. For planting of these hybrid seed in a grain-production field, it is practical to blend the hybrid seed with pollinator seed.
  • the pollinator seed will be present in the minimum amount necessary to achieve adequate pollination of a substantial portion of the plants produced from the blended seed. Preferably, at least 1 % to 50%, more preferably less than 25%, most preferably less than 15% of the blend (by weight) will be pollinator seed.
  • the pollinator seed is present in an amount of about 1 % to 10% by weight. A substantial portion would be about 90% of the plants produced, more preferably about 95%, most preferably about 98% or more of the plants produced by the blend.
  • the pollinator may be non- isogenic to the sterile hybrid plants.
  • the cloned dominant male-sterile gene Ms44 is used to produce male-sterile hybrid plants. See, Figure 4, for example.
  • a male-sterile inbred containing Ms44 in the heterozygous state is transformed with a heterologous SAM construct that comprises (1 ) a Suppression element, for example an inverted repeat (IR) engineered to the Ms44 promoter or Ms44 coding region; (2) a pollen Ablation gene which results in disruption of the formation, function, or dispersal of pollen; (3) a Marker gene, which may be a seed color gene.
  • the suppression element disrupts expression of the dominant Ms44 allele, such that the otherwise male-sterile plant is male-fertile and can be selfed.
  • the resulting progeny on the ear will segregate 50:50 with respect to the hemizygous SAM construct and 25% of all the progeny will be homozygous for the Ms44 dominant allele.
  • Seeds comprising the SAM construct can be identified by presence of the marker.
  • Progeny from these seed can be genotyped to identify homozygous Ms44 progeny with the SAM construct; these plants are collectively referred to as the maintainer line (or "transgenic maintainer" line).
  • the maintainer line or "transgenic maintainer” line
  • Homozygous Ms44 progeny without the SAM construct are collectively referred to as the male sterile female inbred (or "male-sterile inbred" line).
  • Male-sterile inbred seed can be increased by crossing the maintainer line onto male sterile inbred lines.
  • the resulting progeny are male-sterile homozygous Ms44 female inbreds, because the SAM construct is not passed through pollen to progeny. In this way the transgenic maintainer line is used to maintain, propagate, or increase the male sterile plants.
  • the male inbred crosses normally onto this male- sterile inbred line, and no detasseling is required.
  • the Ms44 gene is a dominant male-sterile gene and is homozygous in the female inbred, 100% of the hybrid seed will contain one dominant Ms44 allele, and plants produced from those seed will be male-sterile.
  • the pollinator seed is present in the minimum necessary amount sufficient to permit adequate pollination of the plants produced from the blend.
  • at least 1 % to 50%, more preferably less than 25%, most preferably less than 15%, of the blend (by weight) will be pollinator seed.
  • the pollinator seed should be present in the blend only in an amount sufficient to pollinate a substantial portion of the plants produced by the blend. A substantial portion would be about 90% of the plants produced, more preferably about 95%, most preferably about 98% or more of the plants produced by the blend.
  • the dominant Ms44 gene could be introduced transgenically, operably linked to a heterologous promoter that is amenable to inverted-repeat inactivation and expresses, such that dominant male sterility is achieved.
  • a heterologous promoter that is amenable to inverted-repeat inactivation and expresses, such that dominant male sterility is achieved.
  • Use of the heterologous promoter would ensure that the native ms44 expression is not inhibited by the pIR (promoter inverted repeat).
  • the 5126 promoter from another species, such as rice, may be particularly appropriate, since the 5126 promoter has an expression pattern that is similar to that of the ms44 gene and has been successfully inactivated by pIR methods.
  • This approach has applications not only for yield gain during stress but is also useful for any crop that can outcross to weedy species, such as sorghum, by reducing the propensity for outcrossing and minimizing the risk of adventitious presence.
  • weedy species such as sorghum
  • the biofuels industry is utilizing enzymes transgenically to aid in the digestibility of substrates (i.e. cellulose) used in ethanol production. Linking these types of transgenes to the Ms44 gene would prevent outcrossing through pollen in a production field.
  • One or more dominant traits could be linked to Ms44 to prevent an unintentional outcross to weedy species.
  • the dominant male sterility (DMS) gene Ms44 is introgressed into an inbred maize line. Since this gene acts dominantly, selfing of these lines is not possible and the mutation will segregate 50:50 in resulting outcrossed progeny.
  • Linked genetic markers may be employed to identify those seeds or plants containing the DMS gene so they can be selected for pollination by a maize male inbred line to create F1 hybrid seed. Again this hybrid seed will segregate 50% for male sterility.
  • Ms41 and Ms42 are other known DMS mutants that are dominant in maize. (Liu and Cande, (1992) MNL 66:25-26; and
  • This gene would be linked to a seed marker gene and transformed into an inbred line.
  • Seed from this line could then be sorted based on the presence of the seed marker gene to ensure a pure population of Ms44 male-sterile progeny. These progeny would then be crossed with a male inbred in a hybrid production field to yield 50% male sterility in the resultant hybrid progeny.
  • MS44 mutant sequences can be generated by known means including but not limited to truncations and point mutations. These variants can be assessed for their impact on male fertility by using standard transformation, regeneration, and evaluation protocols.
  • the disclosed nucleotide sequences are used to generate variant nucleotide sequences having the nucleotide sequence of the open reading frame with about 70%, 75%, 80%, 85%, 90% and 95% nucleotide sequence identity when compared to the starting unaltered ORF nucleotide sequence of the corresponding SEQ ID NO.
  • These functional variants are generated using a standard codon table. While the nucleotide sequence of the variants is altered, the amino acid sequence encoded by the open reading frames does not change.
  • These variants are associated with component traits that determine biomass production and quality. The ones that show association are then used as markers to select for each component traits.
  • the disclosed nucleotide sequences are used to generate variant nucleotide sequences having the nucleotide sequence of the 5'-untranslated region, 3'-untranslated region or promoter region that is approximately 70%, 75%, 80%, 85%, 90% and 95% identical to the original nucleotide sequence of the corresponding SEQ ID NO. These variants are then associated with natural variation in the germplasm for component traits related to biomass production and quality. The associated variants are used as marker haplotypes to select for the desirable traits.
  • Variant amino acid sequences of the disclosed polypeptides are generated.
  • one amino acid is altered.
  • the open reading frames are reviewed to determine the appropriate amino acid alteration.
  • the selection of the amino acid to change is made by consulting the protein alignment (with the other orthologs and other gene family members from various species).
  • An amino acid is selected that is deemed not to be under high selection pressure (not highly conserved) and which is rather easily substituted by an amino acid with similar chemical characteristics (i.e., similar functional side-chain).
  • an appropriate amino acid can be changed.
  • the procedure outlined in the following section C is followed.
  • Variants having about 70%, 75%, 80%, 85%, 90% and 95% nucleic acid sequence identity are generated using this method. These variants are then associated with natural variation in the germplasm for component traits related to biomass production and quality. The associated variants are used as marker haplotypes to select for the desirable traits.
  • H, C and P are not changed in any circumstance.
  • the changes will occur with isoleucine first, sweeping N-terminal to C-terminal. Then leucine, and so on down the list until the desired target it reached. Interim number substitutions can be made so as not to cause reversal of changes.
  • the list is ordered 1 -17, so start with as many isoleucine changes as needed before leucine, and so on down to methionine. Clearly many amino acids will in this manner not need to be changed.
  • L, I and V will involve a 50:50 substitution of the two alternate optimal substitutions.
  • variant amino acid sequences are written as output. Perl script is used to calculate the percent identities. Using this procedure, variants of the disclosed polypeptides are generated having about 80%, 85%, 90% and 95% amino acid identity to the starting unaltered ORF nucleotide sequence.
  • Variant amino acid sequences of the disclosed polypeptides are generated.
  • one or more amino acids are altered.
  • the N-terminal secretory signal sequence (SS) is reviewed to determine the possible amino acid(s) alteration.
  • the selection of the amino acid to change is made by predicting the SS cleavage site using available prediction programs such as SignalP (von Heijne, G. "A new method for predicting signal sequence cleavage sites” Nucleic Acids Res.: 14:4683 (1986).
  • SignalP von Heijne, G. "A new method for predicting signal sequence cleavage sites” Nucleic Acids Res.: 14:4683 (1986).
  • Improved prediction of signal peptides SignalP 3.0. , Bendtsen JD, Nielsen H, von Heijne G, Brunak S., J Mol Biol. 2004 Jul 16;340(4):783-95.
  • SRP signal-recognition particle
  • the SRP directs the binding of the ribosome to the ER membrane, as well as threading the protein through the transmembrane channel, called the translocon, where it is processed into its mature form by signal peptidase cleavage of the SS, producing a mature protein.
  • An amino acid change that disrupts SRP binding or signal peptidase cleavage could inhibit the normal processing and secretion of the protein.
  • these types of amino acid substitutions would lead to a dominant male sterility phenotype.
  • the present disclosure includes nucleic acids having a polynucleotide sequence that encodes a non-functional signal peptide (SP). Also included are polypeptides or proteins having a non-functional SP. A non-functional SP will result in the protein having the non-functional SP being retained and/or associated in the ER. A non-functional SP may disrupt the processing, such as cleavage, and/or secretion of the polypeptide or protein containing the non-functional SP.
  • SP signal peptide
  • Disruption of the processing and/or secretion of the polypeptide or protein containing the non-functional SP herein may confer a dominant phenotype to the plant.
  • the dominant phenotype is reduced fertility.
  • the reduced fertility is reduced male fertility.
  • the reduced fertility is reduced female fertility.
  • non-functional signal peptide Various techniques can be used to create a non-functional signal peptide. This may involve modifying an existing SP so that it is non-functional or fusing a non-functional SP to a protein with a functional SP. Accordingly the present disclosure includes fusion proteins containing a non-functional SP as described herein.
  • Signal peptide sequences in polypeptides or proteins can be predicted or identified in a number of ways as is known in the art, including through the use of prediction software.
  • SignallP predicts the presence and location of signal peptide cleavage sites in amino acid sequences, von Heijne, G. "A new method for predicting signal sequence cleavage sites” Nucleic Acids Res.: 14:4683 (1986).
  • Improved prediction of signal peptides SignalP 3.O., Bendtsen JD., Nielsen H., von Heijne G., Brunak S., J Mol Biol. 2004 Jul 16;340(4):783-95. See also the sigcleave program in the EMBOSS (European Molecular Biology Open Software Site) suite of applications online.
  • the SP or predicted SP sequence can be rendered non-functional by inserting, deleting or substituting one or more nucleotides in a polynucleotide encoding an amino acid residue that is deemed to be necessary for proper protein processing and/or secretion of the protein or the polypeptide.
  • the insertion, deletion or substitution of one or more nucleotides in a polynucleotide encoding an amino acid residue may be in the SP or in the encoded protein associated with the SP.
  • the one or more insertions, deletions or substitutions may alter an amino acid at position -10, -9, -8, -7, -6, -5, -4, -3, -2 -1 , +1 , +2, +3, +4, +5, +6, +7, +8, +9, +10 with respect to SP cleavage site, e.g., a predicted SP cleavage site, where the cleavage site is located between positions +1 and -1.
  • the one or more insertions, deletions or substitutions may be alone or combinations thereof.
  • any amino acid insertion, deletion, or substitution at position +2 relative to the SP cleavage site that causes the polypeptide or protein to be retained in the ER may be used in the nucleic acids, proteins, polypeptides, and/or methods of the present disclosure.
  • the amino acid inserted or substituted at position +2 relative to the SP cleavage site is an aromatic amino acid residue, such as phenylalanine, histidine, tryptophan, or tyrosine.
  • the amino acid inserted or substituted at position +2 relative to the SP cleavage site is a charged amino acid residue such as aspartic acid, glutamic acid, lysine, or arginine.
  • the amino acid inserted or substituted at position +2 relative to the SP cleavage site is a large polar amino acid residue such as asparagine or glutamine.
  • the amino acid inserted or substituted at position +2 relative to the SP cleavage site is a hydrophobic amino acid residue such as phenylalanine, isoleucine, leucine, methionine, or valine.
  • the amino acid inserted or substituted at position +2 relative to the SP cleavage site is a small neutral amino acid residue such as alanine, cysteine, serine, or threonine.
  • the amino acid inserted or substituted at position +2 relative to the SP cleavage site is a glycine or proline amino acid.
  • any amino acid insertion, deletion, or substitution at position +1 relative to the SP cleavage that causes the polypeptide or protein to be retained in the ER may be used in the nucleic acids, proteins, polypeptides, and/or the methods of the present disclosure.
  • an amino acid residue is inserted, deleted or substituted for a a charged amino acid residue such as aspartic acid, glutamic acid, lysine, or arginine at position +1 relative to the SP cleavage site.
  • the amino acid inserted or substituted at position +1 relative to the SP cleavage site is an aromatic amino acid residue, such as phenylalanine, histidine, tryptophan, or tyrosine.
  • the amino acid inserted or substituted at position +1 relative to the SP cleavage site is a large polar amino acid residue such as asparagine or glutamine.
  • the amino acid inserted or substituted at position +1 relative to the SP cleavage site is a hydrophobic amino acid residue such as phenylalanine, isoleucine, leucine, methionine, or valine.
  • the amino acid inserted or substituted at position +1 relative to the SP cleavage site is a glycine or proline amino acid.
  • any amino acid insertion, deletion, or substitution at position -1 relative to the SP cleavage that causes the polypeptide or protein to be retained in the ER may be used in the nucleic acids, proteins, polypeptides, and/or the methods of the present disclosure.
  • an amino acid residue is inserted, deleted or substituted for an alanine at position -1 relative to the SP cleavage site.
  • an amino acid residue is inserted, deleted or substituted for a small neutral amino acid, such as alanine, cysteine, serine, or threonine.
  • an amino acid residue is inserted, deleted or substituted for glycine or proline at position -1 relative to the SP cleavage site.
  • the amino acid inserted or substituted at position -1 relative to the SP cleavage site is an aromatic amino acid residue, such as phenylalanine, histidine, tryptophan, or tyrosine.
  • the amino acid inserted or substituted at position -1 relative to the SP cleavage site is a charged amino acid residue such as aspartic acid, glutamic acid, lysine, or arginine.
  • the amino acid inserted or substituted at position -1 relative to the SP cleavage site is a large polar amino acid residue such as asparagine or glutamine.
  • the amino acid inserted or substituted at position -1 relative to the SP cleavage site is a hydrophobic amino acid residue such as phenylalanine, isoleucine, leucine, methionine, or valine. In one embodiment, the amino acid inserted or substituted at position -1 relative to the SP cleavage site is a proline amino acid.
  • any amino acid insertion, deletion, or substitution at position -2 relative to the SP cleavage that causes the polypeptide or protein to be retained in the ER may be used in the nucleic acids, proteins, polypeptides, and/or the methods of the present disclosure.
  • an amino acid residue is inserted, deleted or substituted for an aromatic amino acid residue, such as phenylalanine, histidine, tryptophan, or tyrosine, at position - 2 relative to the SP cleavage site.
  • an amino acid residue is inserted, deleted or substituted for an hydrophobic amino acid residue, such as phenylalanine, isoleucine, leucine, methionine, or valine, at position -2 relative to the SP cleavage site.
  • an amino acid residue is inserted, deleted or substituted for a small neutral amino acid, such as alanine, cysteine, serine, threonine.
  • an amino acid residue is inserted, deleted or substituted for glycine or proline, at position -2 relative to the SP cleavage site.
  • the amino acid inserted or substituted at position -2 relative to the SP cleavage site is a charged amino acid residue such as aspartic acid, glutamic acid, lysine, or arginine.
  • the amino acid inserted or substituted at position -2 relative to the SP cleavage site is a small neutral amino acid residue such as alanine, cysteine, serine, threonine.
  • the amino acid inserted or substituted at position -2 relative to the SP cleavage site is glycine or proline.
  • any amino acid insertion, deletion, or substitution at position -3 relative to the SP cleavage that causes the polypeptide or protein to be retained in the ER may be used in the nucleic acids, proteins, polypeptides, and/or the methods of the present disclosure.
  • an amino acid residue is inserted, deleted or substituted for an alanine at position -3 relative to the SP cleavage site.
  • an amino acid residue is inserted, deleted or substituted for a hydrophobic amino acid residue, such as phenylalanine, isoleucine, leucine, methionine, or valine, at position -3 relative to the SP cleavage site.
  • an amino acid residue is inserted, deleted or substituted for a small neutral amino acid residue such as alanine, cysteine, serine, threonine.
  • an amino acid residue is inserted, deleted or substituted for glycine or proline at position -3 relative to the SP cleavage site.
  • the amino acid inserted or substituted at position -3 relative to the SP cleavage site is an aromatic amino acid residue, such as phenylalanine, histidine, tryptophan, or tyrosine.
  • the amino acid inserted or substituted at position -3 relative to the SP cleavage site is a charged amino acid residue such as aspartic acid, glutamic acid, lysine, or arginine.
  • the amino acid inserted or substituted at position -3 relative to the SP cleavage site is a large polar amino acid residue such as asparagine or glutamine.
  • the amino acid inserted or substituted at position -3 relative to the SP cleavage site is a proline or glycine amino acid.
  • any amino acid insertion, deletion, or substitution at position -4 relative to the SP cleavage that causes the polypeptide or protein to be retained in the ER may be used in the nucleic acids, proteins, polypeptides, and/or the methods of the present disclosure.
  • an amino acid residue is inserted, deleted or substituted for an alanine at position -4 relative to the SP cleavage site.
  • an amino acid residue is inserted, deleted or substituted for a hydrophobic amino acid residue, such as henylalanine, isoleucine, leucine, methionine, or valine, at position -4 relative to the SP cleavage site.
  • an amino acid residue is inserted, deleted or substituted for a small neutral amino acid residue such as alanine, cysteine, serine, threonine.
  • an amino acid residue is inserted, deleted or substituted for glycine or proline at position -4 relative to the SP cleavage site.
  • the amino acid inserted or substituted at position -4 relative to the SP cleavage site is an aromatic amino acid residue, such as phenylalanine, histidine, tryptophan, or tyrosine.
  • the amino acid inserted or substituted at position -4 relative to the SP cleavage site is a glycine.
  • the amino acid inserted or substituted at position -4 relative to the SP cleavage site is an aromatic amino acid residue, such as phenylalanine, histidine, tryptophan, or tyrosine.
  • the amino acid inserted or substituted at position -4 relative to the SP cleavage site is a charged amino acid residue such as aspartic acid, glutamic acid, lysine, or arginine.
  • the amino acid inserted or substituted at position -4 relative to the SP cleavage site is a large polar amino acid residue such as asparagine or glutamine.
  • the amino acid inserted or substituted at position -4 relative to the SP cleavage site is a glycine amino acid.
  • any amino acid insertion, deletion, or substitution at position -5 relative to the SP cleavage that causes the polypeptide or protein to be retained in the ER may be used in the nucleic acids, proteins, polypeptides, and/or the methods of the present disclosure.
  • an amino acid residue is inserted, deleted or substituted for an alanine at position -5 relative to the SP cleavage site.
  • an amino acid residue is inserted, deleted or substituted for a small neutral amino acid residue such as alanine, cysteine, serine, or threonine.
  • an amino acid residue is inserted, deleted or substituted for glycine or proline, at position -5 relative to the SP cleavage site.
  • the amino acid inserted or substituted at position -5 relative to the SP cleavage site is an aromatic amino acid residue, such as phenylalanine, histidine, tryptophan, or tyrosine.
  • the amino acid inserted or substituted at position -5 relative to the SP cleavage site is a charged amino acid residue such as aspartic acid, glutamic acid, lysine, or arginine.
  • the amino acid inserted or substituted at position -5 relative to the SP cleavage site is a large polar amino acid residue such as asparagine or glutamine.
  • the amino acid inserted or substituted at position -5 relative to the SP cleavage site is a hydrophobic amino acid residue such as phenylalanine, isoleucine, leucine, methionine, or valine. In one embodiment, the amino acid inserted or substituted at position -5 relative to the SP cleavage site is a glycine amino acid.
  • any amino acid insertion, deletion, or substitution at position -6 relative to the SP cleavage that causes the polypeptide or protein to be retained in the ER may be used in the nucleic acids, proteins, polypeptides, and/or the methods of the present disclosure.
  • an amino acid residue is inserted, deleted or substituted for an alanine at position -6 relative to the SP cleavage site.
  • an amino acid residue is inserted, deleted or substituted for a hydrophobic amino acid residue, such as phenylalanine, isoleucine, leucine, methionine, or valine, at position -6 relative to the SP cleavage site.
  • an amino acid residue is inserted, deleted or substituted for a small neutral amino acid residue such as alanine, cysteine, serine, or threonine.
  • an amino acid residue is inserted, deleted or substituted for glycine or proline, at position -6 relative to the SP cleavage site.
  • the amino acid inserted or substituted at position -6 relative to the SP cleavage site is an aromatic amino acid residue, such as phenylalanine, histidine, tryptophan, or tyrosine.
  • the amino acid inserted or substituted at position -6 relative to the SP cleavage site is a charged amino acid residue such as aspartic acid, glutamic acid, lysine, or arginine.
  • the amino acid inserted or substituted at position -6 relative to the SP cleavage site is a large polar amino acid residue such as asparagine or glutamine.
  • the amino acid inserted or substituted at position -6 relative to the SP cleavage site is a proline or glycine amino acid.
  • the nucleic acid can include the nucleotide sequence of SEQ ID NO:13, 15 or 152 encoding the SP. In other embodiments the nucleic acid includes one of the sequences set forth in SEQ ID NO: 13, 15 or 152 or fragments thereof that includes sequences encoding the SP.
  • the present disclosure also includes polypeptides or proteins that have the SP of SEQ ID NO:14 or 153. In other embodiments the nucleic acid includes the sequences encoding the amino acid sequences set forth in SEQ ID NO: 14 or 153 or fragments thereof that includes sequences encoding the SP.
  • Disruption of secretion and/or processing of the polypeptide or protein can be determined using any number of routine assays, including but not limited to in vitro transcription/translation assays reactions (EasyXpress Insect Kit II, Qiagen, (Carlsbad, CA) Cat# 32561 ).
  • a polypeptide or protein with a non-functional SP of the present disclosure may be retained in the ER as compared to a polypeptide or protein with a functional signal peptide that is secreted extracellularly.
  • Retention in the ER and lack of protein processing can be determined using any number of routine assays, including but not limited to in vitro transcription/translation assays reactions (EasyXpress Insect Kit II, Qiagen, (Carlsbad, CA), Cat# 32561 ) or using ultrastructural microscopy.
  • the disclosure furthermore relates to vectors or constructs containing the nucleic acid molecules of the disclosure.
  • the nucleic acid molecule of the disclosure can be operatively linked to any suitable regulatory element or promoter as described elsewhere herein.
  • the present disclosure relates to host cells transiently or stably containing the nucleic acid molecules or vectors/constructs of the disclosure.
  • polypeptide having a non-functional signal peptide SP
  • SP non-functional signal peptide
  • the isolated nucleic acid of embodiment 3, wherein the polypeptide having the nonfunctional signal peptide has one or more amino acid insertions, deletions or substitutions at position -10, -9, -8, -7, -6, -5, -4, -3, -2 -1 , +1 , 2, +3, +4, +5, +6, +7, +8, +9, +10 with respect to a SP cleavage site in the SP, wherein the SP cleavage site is located between positions -1 and +1.
  • nucleic acid of embodiment 1 wherein nucleic acid comprises the non-functional SP of SEQ ID NO.:13, 15 or 152.
  • nucleic acid of embodiment 1 wherein nucleic acid comprises the sequence of SEQ ID NO. 13, 15 or 152.
  • the isolated nucleic acid of embodiment 1 wherein the polypeptide comprises the non-functional SP of SEQ ID NO: 14 or 153.
  • the isolated nucleic acid of embodiment 1 wherein the non-functional SP comprises the amino acid sequence corresponding to position 1-37 of SEQ ID NO.:14 and fragments thereof.
  • the isolated nucleic acid of embodiment 1 wherein the non-functional SP comprises the amino acid sequence corresponding to position 1-37 of SEQ ID NO.:153 and fragments thereof.
  • the isolated nucleic acid of embodiment 1 wherein the non-functional signal peptide has a threonine or valine at position -1 relative to the SP cleavage site.
  • the isolated nucleic acid of embodiment 1 wherein the non-functional signal peptide has a proline at position +1 relative to the SP cleavage site.
  • the isolated nucleic acid of embodiment 1 wherein the polypeptide is a tassel-less (f/s7) mutant.
  • the isolated nucleic acid of embodiment 1 wherein the polynucleotide encoding a polypeptide having a non-functional signal peptide is operably linked to promoter.
  • the isolated nucleic acid of embodiment 1 wherein expression of the nucleic acid in a plant increases the yield of a plant.
  • An isolated construct/vector comprising the nucleic acid of any of the embodiments of 1-28.
  • An isolated polypeptide having a non-functional signal peptide SP
  • the non-functional signal peptide is not able to be processed and wherein expression of the polypeptide in a plant confers a dominant phenotype to the plant.
  • polypeptide of embodiment 33 wherein the polypeptide having a nonfunctional signal peptide is retained in the ER.
  • polypeptide of embodiment 33 wherein the non-functional signal peptide is rendered non-functional due to one or more insertions, deletions or substitutions of an amino acid residue in the SP.
  • the polypeptide of embodiment 3, wherein the polypeptide comprises the nonfunctional SP of SEQ ID NO: 14 or 153.
  • polypeptide of embodiment 33 wherein the non-functional SP comprises the amino acid sequence corresponding to position 1 -37 of SEQ I D NO.:14 and fragments thereof.
  • polypeptide of embodiment 33 wherein the non-functional SP comprises the amino acid sequence corresponding to position 1 -37 of SEQ ID NO.:153 and fragments thereof.
  • polypeptide of embodiment 33 wherein the polypeptide comprises the sequence of SEQ ID NO: 14 or 153.
  • polypeptide of embodiment 33 wherein the non-functional signal peptide has a threonine or valine at position -1 relative to the SP cleavage site.
  • polypeptide of embodiment 33 wherein plant is a crop plant.
  • polypeptide of embodiment 33 wherein plant is a monocot or dicot plant.
  • polypeptide of embodiment 33 wherein the polypeptide is a tassel-less ⁇ tls1) mutant.
  • polypeptide of embodiment 33 wherein the polynucleotide encoding the polypeptide having a non-functional signal peptide is operably linked to promoter.
  • the polypeptide of embodiment 47 wherein the promoter is an inducible promoter, a constituitive promoter, a tissue preferred promoter, a temporally regulated promoter or an element thereof.
  • polypeptide of embodiment 47 wherein the promoter preferentially drives expression in male reproductive tissue.
  • polypeptide of embodiment 47 wherein the promoter is a tassel-preferred promoter.
  • polypeptide of embodiment 47 wherein the promoter is selected from the group consisting of:
  • polypeptide is ms45 or ms26.
  • 57. A plant expressing any of the polypeptides of embodiments of 33-55, wherein the plant has increased yield compared to the yield of a plant that does not express any of the polypeptides of embodiments of 1-55.
  • a method of conferring a dominant phenotype of reduced male fertility to a plant comprising:
  • a method of conferring a dominant phenotype of reduced male fertility to a plant comprising:
  • a method of retaining a polypeptide to the endoplasmic reticulum (ER) in a plant cell comprising:
  • a method of retaining a polypeptide to the ER in a plant cell comprising:
  • a method of conferring a dominant phenotype to a plant comprising:
  • a method of conferring a dominant phenotype to a plant comprising:
  • a fusion protein comprising a protein operably linked to a non-functional peptide signal, wherein the non-functional signal peptide is not processed, and wherein expression of the protein in a plant confers a dominant phenotype to the plant.
  • fusion protein of embodiment 65, wherein the non-functional SP comprises any of the polypeptides of the embodiments of 33-55.
  • 75. A plant expressing any of the polypeptides of embodiments of 65-67, wherein the plant has reduced male fertility compared to the male fertility of a plant that does not express any of the polypeptides of embodiments of 65-67.
  • a method of method of conferring a dominant phenotype to a plant comprising: a. expressing a nucleic acid in a plant cell, wherein the nucleic acid is the nucleic acid of embodiment 68, thereby conferring a dominant phenotype to a plant.
  • a method of method of conferring a dominant phenotype to a plant comprising: a. expressing in the plant a fusion protein of any of the embodiments of 65- 67, thereby conferring the dominant phenotype of reduced male fertility to the plant.
  • a method of retaining a polypeptide to the ER in a in plant cell comprising:
  • nucleic acid within a plant cell, wherein the nucleic acid is the nucleic acid of embodiment 68, thereby retaining the polypeptide in the ER.
  • a method of retaining a polypeptide to the ER in a plant cell comprising:
  • a. expressing in the plant a fusion protein of any of the embodiments of 65- 67, thereby retaining the fusion protein in the ER.
  • a method of conferring a dominant phenotype to a plant comprising:
  • a. expressing in a non-anther plant tissue in the plant a nucleic acid of any of the embodiments of 1-28, thereby conferring a dominant phenotype to the plant.
  • a method of conferring a dominant phenotype to a plant comprising:
  • a method of conferring a dominant phenotype to a plant comprising:
  • embodiment 68 thereby conferring a dominant phenotype to the plant.
  • a method of conferring a dominant phenotype to a plant comprising:
  • Example 21 Dominant sterility using a non-functional signal peptide (SP), and restoration of fertility.
  • SP signal peptide
  • the present disclosure also relates to another breeding pair of plants, including a first transgenic plant that is a male-sterile plant, and a second transgenic plant that is a restorer plant.
  • the first transgenic male-sterile plant contains an expressible exogenous nucleic acid molecule comprising a polynucleotide sequence that encodes a polypeptide comprising or fused to a non-functional SP, wherein expression of the exogenous nucleic acid molecule confers a dominant phenotype of male sterility to the plant.
  • Non-functional SP for use in individual plants, breeding pair of plants, and methods of the present disclosure are described elsewhere herein.
  • polynucleotide sequence that encodes a polypeptide comprising or fused to a non-functional SP of the first plant may be exogenous to the plant and can be operably linked to any suitable promoter for expression in male reproductive tissues, including but not limited to MS45, MS26, MS22, tassel-preferred, or 5126 gene promoters. Other suitable promoters are described elsewhere herein. In one embodiment, the
  • polynucleotide sequence that encodes a polypeptide comprising or fused to a nonfunctional SP includes but is not limited to a mutant MS44 gene, including SEQ ID NOs: 13, 15, and 152.
  • the restorer plant includes an expressible exogenous nucleic acid molecule having a polynucleotide that when expressed suppresses the expression of the polynucleotide sequence that encodes the polypeptide comprising or fused to a nonfunctional SP. Such suppression may be directed to said polynucleotide sequence or to its operably-linked promoter.
  • Useful expressible nucleic acid suppression molecules for this embodiment include but are not limited to antisense, RNAi, and hairpin elements as described elsewhere herein and known to one of ordinary skill in the art.
  • the second plant has the expressible exogenous nucleic acid molecule integrated in its genome.
  • the suppression element in the second plant is operably linked to a promoter. Any promoter that expresses in the male reproductive tissues, including a constitutive promoter, may be used to drive the suppression element. Suitable promoters for expressing the suppression element include but are not limited to MS45, MS26, MS22, tassel-less or 5126 gene promoters.
  • the first and second plants are further characterized in that, when bred with each other, progeny are produced in which expression of the suppression element of the second plant suppresses expression of the polynucleotide sequence that encodes the polypeptide comprising or fused to the non-functional SP of the first plant, such that the progeny are male fertile.
  • the present disclosure also provides methods of producing a male-fertile plant by crossing this pair of breeding plants.
  • a breeding pair of plants comprising: a first plant and a second plant, wherein the first plant expresses an exogenous nucleic acid molecule comprising a
  • polynucleotide sequence that encodes a polypeptide with a non-functional SP, wherein expression of the exogenous nucleic acid molecule confers a dominant phenotype of male sterility to the first plant so that the first plant is male-sterile
  • the second plant comprises an expressible exogenous nucleic acid molecule comprising a polynucleotide that when expressed suppresses the expression of the polynucleotide sequence that encodes the polypeptide with the non-functional SP or the promoter operatively linked to the polynucleotide sequence encoding the polypeptide with the non-functional SP of the first plant.
  • the pair of plants of embodiment 5, wherein the male reproductive tissue preferred promoter is the MS45, MS26, MS22, or 5126 promoter.
  • the pair of plants of embodiment 1 wherein the exogenous nucleic acid molecule in the second plant is an RNAi, an antisense or hairpin suppression element.
  • the pair of plants of embodiment 1 wherein the polynucleotide in the second plant that suppresses the expression of the exogenous nucleic acid molecule in the first plant is operably linked to a promoter.
  • the pair of plants of embodiment 10, wherein the male reproductive tissue-preferred promoter is MS45, MS26, MS22, or 5126.
  • the pair of plants of embodiment 16, wherein the MS44 mutant gene in the first plant is selected from the group consisting of SEQ ID NOs: 13, 15, and 152.
  • exogenous nucleic acid molecule in the second plant is a hairpin suppression element that targets the promoter driving expression of a MS44 polynucleotide encoding a polypeptide with a non-functional SP in the first plant.
  • the pair of plants of embodiment 1 wherein the first and second plants are wheat, maize, rice, sorghum, barley, rye, or sunflower.
  • a method of restoring male fertility in a plant exhibiting male sterility comprising crossing the breeding pair of plants of embodiments 1-20.
  • Seed or progeny of the plant of embodiment 21 Seed or progeny of the plant of embodiment 21 .
  • a method of producing hybrid plant seed comprising crossing the breeding pair of plants of embodiments 1-20, wherein the first and second plants are plants of different inbred lines.
  • a method of obtaining a hybrid plant comprising growing the hybrid seed of embodiment 25
  • a hybrid plant produced by the method of embodiment 26 is A hybrid plant produced by the method of embodiment 26.
  • a breeding pair of plants comprising: a first plant and a second plant, wherein the first plant expresses an exogenous nucleic acid molecule comprising a
  • polynucleotide sequence that encodes a polypeptide fused to a non-functional SP wherein expression of the exogenous nucleic acid molecule confers a dominant phenotype of male sterility to the first plant so that the first plant is male-sterile
  • the second plant comprises an expressible exogenous nucleic acid molecule comprising a polynucleotide that when expressed suppresses the expression of nucleic acid molecule comprising a polynucleotide sequence that encodes a polypeptide fused to a non-functional SP or the promoter operatively linked to the nucleic acid molecule comprising a polynucleotide sequence that encodes a polypeptide fused to a non-functional SP of the first plant.
  • the pair of plants of embodiment 31 wherein the promoter is an inducible promoter, a constitutive promoter, a tissue preferred promoter, a temporally regulated promoter or an element thereof.
  • the pair of plants of embodiment 31 wherein the promoter preferentially drives expression in male reproductive tissue.
  • the pair of plants of embodiment 31 wherein the male reproductive tissue-preferred promoter is MS45, MS26, MS22, or 5126.
  • the pair of plants of embodiment 31 wherein the promoter is a tassel-preferred promoter.
  • the pair of plants of embodiment 29, wherein the exogenous nucleic acid molecule in the second plant is an RNAi, an antisense or hairpin suppression element.
  • the pair of plants of embodiment 39, wherein the male reproductive tissue-preferred is MS45, MS26, MS22, or 5126.
  • the pair of plants of embodiment 45, wherein the MS44 mutant gene in the first plant is selected from the group consisting of SEQ ID NOs: 13, 15, and 152.
  • exogenous nucleic acid molecule in the second plant is a hairpin suppression element that targets the promoter driving expression of a MS44 polynucleotide encoding a polypeptide fused to a nonfunctional SP in the first plant.
  • the pair of plants of embodiment 29, wherein the first and second plants are wheat, maize, rice, sorghum, barley, rye, or sunflower.
  • a method of restoring male fertility in a plant exhibiting male sterility comprising crossing the breeding pair of plants of embodiments 29-48.
  • a method of producing hybrid plant seed comprising crossing the breeding pair of plants of embodiments 29-48, wherein the first and second plants are plants of different inbred lines.
  • a method of obtaining a hybrid plant comprising growing the hybrid seed of

Abstract

A nuclear maize gene is mutated to result in dominant male sterility. Two mutants are provided which impact signal peptide processing. Restoration constructs and methods are provided for use of the dominant male-sterile mutants in producing hybrid seed for male-sterile hybrid plants. Other signal-peptide modifications are provided. Orthologs of the wild type or mutated gene in various species are provided.

Description

GENETIC REDUCTION OF MALE FERTILITY IN PLANTS
FIELD OF THE DISCLOSURE
The disclosure relates generally to the field of molecular biology, specifically the modulation of plant fertility to improve plant stress tolerance.
BACKGROUND INFORMATION
Plants allocate photosynthates, minerals, and other growth components among various plant tissues during the developmental life cycle. In maize, for example, ear and tassel are specific female and male inflorescence structures that share certain developmental processes and compete with each other for required nutrients. For example, tassel apical dominance may limit ear growth and grain yield potential in maize plants. Genetic male sterility in hybrid maize could reduce tassel dominance and divert more resources to ear growth, kernel number and/or size, ultimately leading to increased grain yield. The advantage from this shift in allocation of resources could be particularly advantageous under conditions of environmental stress.
SUMMARY OF THE DISCLOSURE
Nitrogen utilization efficiency (NUE) genes affect yield and have utility for improving the use of nitrogen in crop plants, especially maize. Increased nitrogen use efficiency can result from enhanced uptake and assimilation of nitrogen fertilizer and/or the subsequent remobilization and reutilization of accumulated nitrogen reserves, as well as increased tolerance of plants to stress situations such as low nitrogen environments. The genes can be used to alter the genetic composition of the plants, rendering them more productive with current fertilizer application standards or maintaining their productive rates with significantly reduced fertilizer or reduced nitrogen availability. Improving NUE in corn would increase corn harvestable yield per unit of input nitrogen fertilizer, both in developing nations where access to nitrogen fertilizer is limited and in developed nations where the level of nitrogen use remains high. Nitrogen utilization improvement also allows decreases in on-farm input costs, decreased use and dependence on the nonrenewable energy sources required for nitrogen fertilizer production and reduces the environmental impact of nitrogen fertilizer manufacturing and agricultural use.
Methods and compositions for improving plant yield, particularly plant grain yield, are provided. In some embodiments, plant yield is improved under stress, particularly abiotic stress, such as nitrogen limiting conditions. Methods of improving plant yield include inhibiting the fertility of the plant. The male fertility of a plant can be inhibited using any method known in the art, including but not limited to the disruption of a tassel development gene, or a decrease in the expression of a gene, for example through the use of co-suppression, antisense or RNA silencing or interference. Inhibition of fertility may be partial reduction or complete inhibition. Male sterile plants can also be achieved by using genetic male sterile mutants.
Inhibiting the male fertility in a plant can improve the nitrogen stress tolerance of the plant and such plants can maintain their productive rates with significantly less nitrogen fertilizer input and/or exhibit enhanced uptake and assimilation of nitrogen fertilizer and/or remobilization and reutilization of accumulated nitrogen reserves. In addition to an overall increase in yield, the improvement of nitrogen stress tolerance through the reduction in male fertility can also result in increased root mass and/or length, increased ear, leaf, seed and/or endosperm size, and/or improved standability. Accordingly, in some embodiments, the methods further comprise growing said plants under nitrogen limiting conditions and optionally selecting those plants exhibiting greater tolerance to the low nitrogen levels.
Further, methods and compositions are provided for improving yield under abiotic stress, which include evaluating the environmental conditions of an area of cultivation for abiotic stressors (e.g., low nitrogen levels in the soil) and planting seeds or plants having reduced male fertility, in stressful environments.
Constructs and expression cassettes comprising nucleotide sequences that can efficiently reduce male fertility are also provided herein.
Additional methods include but are not limited to:
A method of increasing yield by increasing one or more yield components in a plant includes reducing male fertility by affecting the expression or activity of a nuclear encoded component in the plant, and growing the plant under plant growing conditions, wherein the component exhibits a dominant phenotype. In an embodiment, the nuclear encoded component is a male fertility gene or a male sterility gene that has a dominant phenotype. Optionally, the male fertility gene or the male sterility gene is a transgene.
In an embodiment, the male fertility gene encodes a protein of SEQ ID NO: 10. In an embodiment, the male fertility gene includes a nucleotide sequence of SEQ ID NO: 13. In an embodiment, the male fertility gene encodes a polypeptide of SEQ ID NO: 14.
In an embodiment, the reduction of male fertility or rendering the plant male sterile is effected by a single nucleotide substitution resulting in an amino acid change at amino acid 37, from Alanine to Threonine in the predicted protein (SEQ ID NO: 153). In an embodiment, the reduction of male fertility or rendering the plant male sterile is effected by an amino acid change at amino acid 37, from Alanine to Valine in the predicted protein. In an embodiment, the dominant male fertility gene is operably linked to a promoter selected from the group consisting of: inducible promoter, tissue preferred promoter, temporally regulated promoter or an element thereof. For example, the promoter preferentially drives expression in male reproductive tissue.
In an embodiment, the male fertility is reduced in the female plant (e.g., a female inbred line) of a breeding pair.
In an embodiment, a plant or a cell or a seed or a progeny thereof that includes the reduced male fertility sequence encoding amino acid sequence 43 - 101 of SEQ ID NO: 10 in its genome and wherein the expression of the male fertility gene confers the dominant male sterility trait.
An isolated nucleic acid molecule includes a polynucleotide capable of initiating transcription in a plant cell and includes a sequence selected from the group consisting of: SEQ ID NO: 15; at least 100 contiguous nucleotides of SEQ ID NO: 15 and a sequence having at least 70% sequence identity to the full length of SEQ ID NO: 15. In an embodiment, an expression cassette or a vector includes SEQ ID NO: 15 disclosed herein operably linked to a polynucleotide of interest.
Suitable plants for certain embodiments disclosed herein include e.g., maize
(corn), sorghum, canola, wheat, barley, rye, triticale, rice, sugar cane, turfgrass, pearl millet, soybeans, cotton, and sunflower.
In an embodiment, a plant with reduced fertility or any other trait disclosed herein optionally comprises one or more polynucleotides conferring the following phenotype or trait of interest: improved nutrient uptake, nitrogen use efficiency, drought tolerance, root strength, root lodging resistance, soil pest management, corn root worm resistance, herbicide tolerance, disease resistance, or insect resistance, or altered carbohydrate metabolism, protein metabolism, fatty acid metabolism or phytohormone biosynthesis.
A method of increasing yield or maintaining yield stability in plants includes reducing male reproductive tissue development by expressing a transgene under the control of a male reproductive tissue preferred promoter; and increasing nutrient allocation to female reproductive tissue during concurrent male and female tissue development.
In an embodiment, the male reproductive tissue is tassel. In an embodiment, the male reproductive tissue development is decreased by the expression of a gene operably linked to a promoter comprising at least 100 contiguous nucleotides of a sequence selected from the list SEQ ID NO: 64 - 106. Subsets of the promoter sequences disclosed herein e.g., SEQ ID NOS: 64-70; 70-75; 75-80; 85-90; 90-95; 100-106 are also suitable for driving tissue-preferred expression of the polynucleotides of interest disclosed herein.
In an embodiment, a plant or a plant cell or a seed that transgenically expresses a polynucleotide of interest (e.g., Ms44 having the dominant male sterility mutation) under the control of a tassel-preferred promoter disclosed herein exhibits improved agronomic parameters such as increased nutrient allocation to ears during reproductive development.
Certain embodiments comprise an isolated nucleic acid molecule comprising a polynucleotide which initiates transcription in a plant cell and comprises a sequence selected from the group consisting of:
a sequence selected from SEQ ID NO: 64 - 106;
at least 100 contiguous nucleotides of a sequence selected from SEQ ID NO: 64 - 106 and
a sequence having at least 70% to about 95% sequence identity to the full length of a sequence selected from SEQ ID NO: 64 -106 or to sub-promoter regions thereof.
In an embodiment, a plant or a plant cell or a seed that transgenically expresses a polynucleotide of interest (e.g., RNAi suppression sequence targeting a polynucleotide involved in tassel development) under the control of a tassel-preferred promoter disclosed herein exhibits increased agronomic parameters such as improved nutrient allocation to ears during reproductive development and/or increased grain yield relative to a control.
A method of increasing yield or maintaining yield stability in plants includes reducing male fertility and increasing nutrient allocation to female reproductive tissue during concurrent male and female tissue development. In an embodiment, the male fertility is reduced in a plant by altering expression of a genetic male fertility gene. In an embodiment, the plant is grown under stress. In an embodiment, the plant is grown under nutrient limiting conditions, e.g., reduced available nitrogen.
In an embodiment, the plants with reduced male fertility and wherein the nutrient is allocated more to female reproductive tissue during concurrent male and female tissue development exhibits one or more of the following agronomically relevant parameters: increased SPAD value (chlorophyll measurement); increased silk emergence rate or number or degree; increased ear length; increased ear width; increased seed number per ear; increased seed weight per ear and increased embryo size. In an embodiment, the plants with reduced male fertility and wherein the nutrient is allocated more to female reproductive tissue during concurrent male and female tissue are grown under drought stress. In an embodiment, drought tolerance of the plants is improved by male sterility.
An isolated nucleic acid molecule comprising a polynucleotide which initiates transcription in a plant cell in a tissue preferred manner and includes a sequence selected from the group consisting of:
The regulatory region of SEQ ID NO: 9 or 13;
SEQ ID NOS: 62 and 64-106;
at least 100 contiguous nucleotides of the regulatory region of SEQ ID NO: 9 or
13;
at least 100 contiguous nucleotides of SEQ ID NOS: 62 and 64-106;
a sequence having at least 70% sequence identity to the full length of the regulatory region of SEQ ID NO: 9 or 13;
a sequence having at least 70% sequence identity to the full length of SEQ ID
NOS: 62 and 64-106.
In an embodiment, a method of increasing yield stability in plants under stress includes expressing an element that affects male fertility operably linked to a tassel preferred promoter disclosed herein and thereby reducing the competition for nutrients during the reproductive development phase of the plant and wherein the yield is increased.
A method of increasing yield or maintaining yield stability in plants under nitrogen limiting conditions and/or normal nitrogen conditions includes reducing male reproductive tissue development and increasing nutrient allocation to female reproductive tissue during concurrent male and female tissue development.
In an embodiment, the male reproductive tissue is tassel and the male reproductive tissue development is decreased by reducing the expression of a NIP3-1 or a Nl P3-1 -like protein. In an embodiment, NIP3-1 protein has an amino acid sequence of SEQ ID NO: 107. The male reproductive tissue development is decreased by increasing the expression of SEQ ID NO: 63.
In an embodiment, the male reproductive tissue development is decreased by affecting the function of a gene involved in tassel formation, e.g., tassel-less gene.
In an embodiment, the male reproductive tissue development is decreased in a plant transformed with an expression cassette that targets for suppression a gene encoding amino acid sequence of SEQ ID NO: 107 or a sequence that is at least 70% or 80% or 85% or 90% or 95% identical to SEQ ID NO: 107. Plants with native mutations in the Tls1 allele are also disclosed herein.
In an embodiment, a tissue-preferred promoter preferentially drives expression of a gene of interest in male reproductive tissue. In an embodiment, the promoter is a tissue-specific promoter, a constitutive promoter or an inducible promoter. In an embodiment, the tissue-preferred promoter is a tassel-preferredpromoter.
An isolated nucleic acid molecule comprising a polynucleotide that includes a sequence selected from the group consisting of: SEQ ID NO: 63; at least 100 contiguous nucleotides of SEQ ID NO: 63 and a sequence having at least 70% sequence identity to the full length of SEQ ID NO: 63. An isolated nucleic acid molecule comprising a polynucleotide that encodes the TLS1 protein comprising an amino acid sequence of SEQ ID NO: 107 or a sequence that is at least 70% or 80% or 85% or 90% or 95% identical to SEQ ID NO: 107.
A method for producing male sterile hybrid seeds includes transforming an inbred line that is heterozygous for dominant male sterility with a gene construct that includes a first element that suppresses the dominant male sterility phenotype, a second element that disrupts pollen function, and optionally a marker, which may be a selectable or screenable marker, wherein expressing the construct in the inbred line renders the line male fertile; this line is referred to as the transgenic maintainer line. In an embodiment, this method further includes self-pollinating plants of the transgenic maintainer line to produce progeny that are homozygous dominant male sterile.
The method further includes identifying those seeds having the homozygous dominant male sterility genotypes as the female inbred line. The female inbred line is; optionally increased by pollinating it with the transgenic maintainer line, resulting in 100% homozygous dominant male sterile seed lacking the construct.
The method further includes pollinating plants grown from the dominant male sterile seed with a male parent, to produce hybrids that are heterozygous for dominant male sterility and display the dominant male sterile phenotype.
In an embodiment, the dominant male sterility phenotype is conferred by a polynucleotide sequence that includes at least 100 consecutive nucleotides of SEQ ID NO: 15 and further encodes a Threonine instead of an Alanine at position 37 of SEQ ID NO: 14 (the amino acid sequence encoded by SEQ ID NO: 15).
In an embodiment, the suppression element includes a promoter inverted repeat (pIR) sequence specific to SEQ ID NO: 9 or SEQ ID NO: 13, or to a promoter operably linked to SEQ ID NO: 15 or SEQ ID NO: 152. In an embodiment, the inverted repeat sequence includes a fragment of at least 100 consecutive nucleotides of SEQ ID NO: 9 or SEQ ID NO: 13. In an embodiment, the suppression element is a RNAi construct designed to suppress the expression of the dominant Ms44 gene in the male sterile female inbred line and may target SEQ ID NO: 15 or SEQ ID NO: 152. In an embodiment, the suppression element is a genetic suppressor that acts in a dominant fashion to suppress the dominant sterile phenotype of Ms44 mutation in a plant. Optionally, if the endogenous normal ms44 is also suppressed by the suppression element, the construct may include an element that restores the normal function of the ms44 gene, e.g., ms44 gene under the control of its own promoter or a heterologous promoter.
In an embodiment, a plant or a plant cell or a seed or a progeny of the plant derived from the methods disclosed herein is disclosed.
In an embodiment, a method for producing hybrid seeds includes expressing in an inbred a dominant male sterility gene operably linked to a heterologous promoter amenable to inverted-repeat inactivation; and pollinating this male sterile plant with pollen from a male fertile plant containing an inverted repeat specific to the heterologous promoter. In an embodiment, the pollen comprises the inverted repeat specific to the heterologous promoter with inverted repeat inactivation specificity. In an embodiment, the dominant male sterility gene is linked to a rice 5126 promoter.
In an embodiment, the dominant male sterility gene used in the context of hybrid seed production is any gene that acts in a dominant manner to achieve male sterility and optionally is amenable to suppression to maintain the male-sterile female inbred line. In an embodiment, the dominant male sterility gene is selected from the group comprising: barnase, DAM methylase, streptavidin, MS41 and MS42.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 - Diagram of Genetic Dominant Male Sterility system to produce a male- sterile hybrid plant. Genetic reduction of male fertility in a plant, which may utilize one or more of a dominant nuclear male-sterile gene, a tassel-specific or tassel-preferred promoter, and a tassel-specific or tassel-preferred gene, has been found to increase ear tissue development, improve nutrient utilization in the growing plant, increase stress tolerance, and/or increase seed metrics, ultimately leading to improved yield.
Figure 2 - Alignment of MS44 related sequences (Figure 2 A - C). The identical residues are in bold and all similar residues are underlined and italicized. Figure 3 - Diagram of method to produce a male-sterile hybrid plant using a recessive male-sterile gene. Both the female parent and the male parent have the homozygous recessive alleles which confer sterility. However, the male parent carries the restorer allele within a construct which prevents transmission of the restorer allele through pollen. Resulting hybrid seed produce a male-sterile hybrid plant.
Figure 4 - Diagram of method for producing male sterile hybrid plant using a dominant male-sterility gene:
4A - An inbred line heterozygous for dominant male sterility (DMS) is transformed with a gene construct (SAM; suppression, ablation, marker) that comprises an element that suppresses the dominant male sterility, a second element that disrupts pollen function, and optionally a selectable marker.
Expression of this construct in the inbred line renders the plants male fertile.
4B - The plants are self-pollinated to produce seed.
4C and 4D - Seeds or progeny plants are phenotyped and/or genotyped to identify those which are homozygous for dominant male sterility and are further characterized as carrying or lacking the SAM construct
4E - The male-sterile inbred line can be increased by pollinating it with the transgenic maintainer line, resulting in seed which are all homozygous dominant male sterile and lack the SAM construct.
4F - Dominant male sterile inbred plants are pollinated by a second inbred to produce hybrids that are heterozygous for dominant male sterility and exhibit the dominant male sterile phenotype. Male-sterile hybrid seed are seed that when planted will produce a hybrid plant which is male-sterile.
Figure 5 - Figure 5A shows MS44 hybrid yield response to N fertility - Trial 1 . Figure 5B shows MS44 hybrid yield response to N fertility - Trial 2.
Figure 6 shows MS44 hybrid ear dry weight (R1 stage) as compared to wild-type.
Figure 7 - Figure 7A shows MS44 hybrid yield response to plant population- Trial 1. Figure 7B shows MS44 hybrid yield response to plant population - Trial 2.
Figure 8 shows the tlsl mutant phenotype. A) Tassel from a wild type plant. B) Homozygous tlsl plant with a small-tassel phenotype. C) Homozygous tlsl plant with no tassel. D) Plants with most severe phenotypes tend to have multiple ears with long husks and no silk emergence (arrows). E) Range of ear phenotypes. F) Range of leaf phenotypes. WT = homozygous wild type plant; ST = homozygous tlsl plant with a small tassel; NT = homozygous tlsl plant with no tassel.
Figure 9 shows the map-based cloning of tlsl . Figure 10 shows tlsl candidate gene validation. Knockout of ZmNIP3.1 results in tlsl phenotype. Figure 10A - Wild type plant with intact ZmNIP3;1 . Figure 10B- Plant with Mu-insertion in ZmNIP3.1 exhibits tlsl phenotype.
Figure 1 1 shows the tassel branch number in mutant, wild-type and mutant sprayed with boron.
Figure 12 shows the tassel branch length in mutant, wild-type and mutant sprayed with boron.
Figure 13 shows the ear length in mutant, wild-type and mutant sprayed with boron.
Figure 14 shows that tlsl plants are less susceptible to boron-toxic conditions of
50ppm boron. Figure 14A - Side-by-side of homozygous tlsl and wild type plants with mutant plants appearing taller and larger. Figure 14B - In wild type plants, the node of the second youngest fully expanded leaf extends above the node of the youngest fully expanded leaf, whereas mutant plants appear normal. Figure 14 C -Youngest fully expanded leaf of mutant is broader than wild type.
Figure 15 shows ZmNIP3.1 is similar to boron channel proteins. Figure 15 A - Phylogenetic tree shows ZmNIP3.1 is closely related to OsNIP3.1 and AtNIP5.1 (highlighted), which have been characterized as boron channel proteins. Figure 15B - Alignment of protein sequences highlighted in 15A where ZmNIP3.1 is 84.4 and 67.3 percent identical to OsNIP3.1 and AtNIP5.1 respectively.
Figure 16 - Ms44 sequences from selected species. In this alignment, the amino acid mutation for the Ms44 Dominant polypeptide sequence is indicated in bold and underlined in position 42, as T in the MS44dom allele (SEQ ID NO: 14) or V in the Ms44- 2629 allele (SEQ ID NO: 153), where all other sequences have A at that position.
DETAILED DESCRIPTION
The developing female reproductive structure competes with male reproductive structures for nitrogen, carbon and other nutrients during development of these reproductive structures. This is demonstrated in quantifying the nitrogen budget of developing maize ears and tassels when the plants are grown in increasing levels of nitrogen fertilizer. When maize is grown under lower nitrogen fertility levels the nitrogen budget of the ear is negative, or during development the ear loses nitrogen to other parts of the plant when nitrogen is limiting. The nitrogen budget of the ear improves as the amount of nitrogen fertilizer provided to the plant increases until the ear maintains a positive increase in nitrogen through to silk emergence. In contrast, the tassel maintains a positive nitrogen budget irrespective of the level of fertility in which the plant is grown. The tassel and ear compete for nitrogen during reproductive development and the developing tassel dominates over the developing ear. The ear and tassel likely compete for a number of nutrients during development and the competition becomes more severe under stress conditions. The ear is in competition with the tassel during reproductive development prior to anthesis, reducing the ability of the developing ear to accumulate nutrients under stress, resulting in a smaller, less developed ear with fewer kernels. More severe, extended stress can result in failure of the ear to exert silks and produce grain. Genetic reduction in male fertility would reduce the nutrient requirement for tassel development, resulting in improved ear development at anthesis.
Genetic male-sterile and male-fertile sibs were grown in varying levels of nitrogen fertility and sampled at -50% pollen shed. Male sterile plants produced larger ears than their male-sterile sibs under both nitrogen fertility levels. The proportion of male sterile plants with emerged silks was also greater than in the fertile sib plants. Though the biomass (total above ground plant dry weight minus the ear dry weight) was greater in the higher-nitrogenfertility grown plants, there was no effect of male sterility on biomass. This shows the positive effect of male sterility is specifically on the ability of the plant to produce a heavier more fully developed ear without affecting overall vegetative growth.
Yield experiments with genetic male sterile derived hybrids have not previously been done because, until recently, there has been no reasonable method of producing hybrid seed using this source of male sterility. Since most genetic male steriles are recessive, producing male sterile hybrids would require the source of male sterility to be backcrossed into both parents of the hybrid. The female parent would have to be homozygous recessive (male sterile) and the male parent would have to be heterozygous (male fertile) for the hybrid to segregate 1 :1 for male sterility.
In contrast, MS44, a dominant genetic male sterile, only needs to be backcrossed into the female parent to produce hybrid seed segregating 1 :1 for male sterility. Dominant male sterility is especially useful in polyploid plants such as wheat, where maintenance of homozygous recessive sterility is more complex.
The process of expressing a dominant genetic male sterile gene in a plant, optionally combined with tassel-tissue specific or tassel-tissue-preferred promoters and tassel-specific or tassel-preferred genes, has been found to increase ear tissue development, improve nutrient utilization in the growing plant and increase seed metrics, ultimately leading to improved yield. (Figure 1 ) Compared to cytoplasmic male sterility (CMS), genetic male sterility is much more likely to produce a yield response because pollen development fails much earlier in genetic male sterile mutants than in CMS-derived sterility. Most genetic male sterile mutants fail shortly after pollen tetrad release (Albertsen and Phillips, (1981 ) Can. J. Genet. Cytol. 23:195-208) which occurs during very early stages of female (ear) development. CMS-derived male sterility is not determined until 10 days prior to anthesis, as judged by the environmental interactions associated with CMS stability (Weider, et al., (2009) Crop Sci. 49:77-84). The bulk of ear development would have already occurred prior to 10 days before anthesis, thus CMS-derived sterility would provide little relief from tassel competition during ear development. In contrast, early failure of male reproductive tissue development in genetic male sterility reduces competition between developing ear and developing tassel for nutrients when the ear is in early stages of development. Yield improvements associated with male sterile hybrids vectored through improved ear development are consistent with the reduction in competition of ear development with tassel development.
The yield response to N fertility was tested in restored (male fertile) and non- restored (male sterile) cytoplasmic male sterile (CMS) hybrids. One hybrid became male fertile due to environmental conditions during flowering and the other hybrid showed no significant yield effects due to male sterility. These results would not be unexpected since male sterility determined via cytoplasmic genes is not established until very late in tassel and ear development, as judged by the environmental interactions associated with CMS stability. The bulk of ear development has already occurred before CMS male sterility is irrefutably set (10 days before anthesis) providing little relief from tassel competition during ear development. Most genetic male sterile mutants fail shortly after pollen tetrad release (Albertsen and Phillips, 1981 ) which is during very early stages of female (ear) development. Thus tassel development in a genetic male sterile would be reduced during the entire ear developmental timeframe and compete less with ear development. Genetic male sterile mutants are little affected by environmental conditions.
Relieving competition between developing tassel and ear could also be achieved by chemically induced male sterility. A combination of chemicals and genetic manipulation could also induce male sterility. Herbicide tolerance modulated by promoters with less efficacy in male reproductive tissue or the use of pro-gametocides ((Dotson, et al., (1996) The Plant Journal 10:383-392) and (Mayer and Jefferson, (2004) Molecular Methods for Hybrid Rice Production. A report for the Rural Industries Research and Development Corporation)) to release male inhibitors in a tissue specific manner would also be effective means of practicing this disclosure.
In a number of circumstances, a particular plant trait is expressed by maintenance of a homozygous recessive condition. Difficulties arise in maintaining the homozygous condition when a restoration gene must be used for maintenance. For example, the MS45 gene in maize (US Patent Number 5,478,369) has been shown to be critical to male fertility. Plants heterozygous or hemizygous for the dominant MS45 allele are fully fertile due to the sporophytic nature of the MS45 fertility trait. A natural mutation in the MS45 gene, designated ms45, imparts a male sterility phenotype to plants when this mutant allele is in the homozygous state. This sterility can be reversed (i.e., fertility restored) when the non-mutant form of the gene is introduced into the plant, either through normal crossing or transgenic complementation methods. However, restoration of fertility by crossing removes the desired homozygous recessive condition, and both methods restore full male fertility and prevent maintenance of pure male sterile maternal lines.
A method to maintain the desired homozygous recessive condition is described in US Patent Numbers 7,696,405 and7,893,317, where a maintainer line is used to cross onto homozygous recessive male sterile siblings. The maintainer line is in the desired homozygous recessive condition for male sterility but also contains a hemizygous transgenic construct consisting of a dominant male fertility gene to complement the male sterility condition; a pollen ablation gene, which results in disruption of the formation, function, or dispersal of pollen and thereby prevents the transfer through pollen of the transgenic construct to the male sterile sibling but allows for the transfer of the recessive male sterile allele through the non-transgenic pollen grains; and a seed marker gene which allows for the sorting of transgenic maintainer seeds or plants and transgenic-null male sterile seeds or plants.
Seed Production Technology (SPT) provides methods to maintain the homozygous recessive condition of a male-sterility gene in a plant. See, for example, US Patent Number 7,696,405. SPT utilizes a maintainer line that is the pollen source for fertilization of its homozygous-recessive male-sterile siblings. The maintainer line is in the desired homozygous recessive condition for male sterility but also contains a hemizygous transgenic construct (the "SPT construct"). In certain embodiments the SPT construct comprises the following three elements: (1 ) a dominant male-fertility gene to complement the male-sterile recessive condition; (2) a gene encoding a product which interferes with the formation, function, or dispersal of male gametes and (3) a marker gene which allows for the sorting of transgenic maintainer seeds/plants from those which lack the transgene. Interference with pollen formation, function or dispersal prevents the transfer through pollen of the transgenic construct; functional pollen lacks the transgene. Resulting seeds produce plants which are male-sterile. These male-sterile inbred plants are then used in hybrid production by pollinating with a male parent, which may be an unrelated inbred line homozygous for the dominant allele of the male-fertility gene. Resulting hybrid seeds produce plants which are male-fertile.
To create hybrid male sterile progeny, the male parent would serve as the maintainer line to cross onto male sterile female inbreds, (increased using a separate maintainer line), to give fully male sterile hybrid plants. See, for example, Figure 3.
The use of a dominant approach is another method to achieve male sterility. In many regards a dominant male sterility approach has advantages over the use of recessive male sterility because only a single copy of the dominant gene is required for full sterility. However, if methods are not available to create a homozygous dominant male sterile line, then resulting progeny will segregate 50% for male sterility. Practically, this situation can be alleviated by transgenically linking a screenable or selectable marker to the dominant male sterility gene and screening or selecting progeny seeds or plants carrying the marker. For a dominant male sterile allele, linked genetic markers or a linked phenotype could be employed to sort progeny. Methods describing a reversible dominant male sterility system are described in US Patent Number 5,962,769 where a chemical is applied to dominant male sterile plants, which reverses the phenotype and results in male fertility, allowing for self pollinations so that homozygous dominant male sterile plants can be obtained. Other methods for creating a homozygous dominant male sterile plant could be envisioned using an inducible promoter controlling a gene that represses or interferes with function of the dominant male sterile gene. The plant is constitutively sterile, becoming fertile only when the promoter is induced, allowing for expression of the repressor which disrupts the dominant male sterile gene function. A repressor might be an antisense gene, RNAi, an inverted repeat that targets either the dominant male sterile gene itself or its promoter, or a gene product that is capable of binding or inactivating the dominant male sterile gene product.
Another approach to produce 100% male sterility in progeny from dominant male sterility would use auto splicing protein sequences. An auto splicing protein sequence is a segment of a protein that is able to excise itself and rejoin the remaining portion/s with a peptide bond. Auto splicing protein sequences can self splice and re-ligate the remaining portions in both cis and trans states. A dominant male sterile gene could be modified such that the regions coding for the N and C protein regions are separated into different transgenic constructs, coupled with a sequence coding for an auto splicing protein sequence. A plant containing a single construct would be male fertile since the protein is truncated and non-functional, which allows for self fertilization to create a homozygous plant. Plants homozygous for the N-DMS-N-auto splicing protein sequence can then be crossed with plants homozygous for the C-auto splicing protein sequence-C-DMS protein. All of the progeny from this cross would be male sterile through the excision of each auto splicing protein sequence and the re-ligation of the N and C sequences to create a functional dominant male sterile protein.
A series of field experiments were used to quantify the yield response of genetic male sterility under a variety of environmental variables. There were two variables used: nitrogen fertilizer rate, and plant density, to subject the plants to various degrees of stress. This continuum of stress treatments allowed for clear separation of plant performance due to greater assimilate partitioning to ears of the genetic male sterile plants. These methods were used to quantify and demonstrate positive yield effects in a representative crop canopy environment in the field. These data validated earlier individual plant responses measured in greenhouse studies.
Male sterility is manifested in the changes in development of specific plant tissues. Maize ear and tassel are both inflorescence structures that share common development processes and are controlled by a common set of genes. The tissues compete with each other for the required nutrients. Tassel however has the advantage of apical dominance over the ear, which is unfavorable to ear growth and yield potential in the maize plants. Reducing the tassel apical dominance could be used to divert more resource to the ear growth, kernel number or size and ultimately can lead to increased grain yield.
There are multiple approaches to reducing the competition of the tassel, such as male sterility, tassel size reduction, or tassel elimination (a tasseless maize plant). While genetic mutations (mutants) of genes such as male sterility genes can be used to reduce the competition of the tassel with ear, transgenic manipulation offers alternatives or enabling tools for this purpose. As genes that are involved in tassel development are often involved in ear development, reducing tassel development by interrupting these genes may also affect the ear development. The tasseless gene (Tsl1 ) mutation is an example, in which the tasseless plant is also earless. To enable tassel growth reduction without interfering with the ear development, a tassel-specific promoter is needed to target the gene disruption in the tassel tissues only.
All references referred to are incorporated herein by reference. Unless specifically defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. Unless mentioned otherwise, the techniques employed or contemplated herein are standard methodologies well known to one of ordinary skill in the art. The materials, methods and examples are illustrative only and not limiting. The following is presented by way of illustration and is not intended to limit the scope of the disclosure.
Many modifications and other embodiments of the disclosures set forth herein will come to mind to one skilled in the art to which these disclosures pertain having the benefit of the teachings presented in the foregoing descriptions and the associated drawings. Therefore, it is to be understood that the disclosures are not to be limited to the specific embodiments disclosed and that modifications and other embodiments are intended to be included within the scope of the appended claims. Although specific terms are employed herein, they are used in a generic and descriptive sense only and not for purposes of limitation.
The practice of the present disclosure will employ, unless otherwise indicated, conventional techniques of botany, microbiology, tissue culture, molecular biology, chemistry, biochemistry and recombinant DNA technology, which are within the skill of the art.
Units, prefixes and symbols may be denoted in their SI accepted form. Unless otherwise indicated, nucleic acids are written left to right in 5' to 3' orientation; amino acid sequences are written left to right in amino to carboxy orientation, respectively. Numeric ranges are inclusive of the numbers defining the range. Amino acids may be referred to herein by either their commonly known three letter symbols or by the one-letter symbols recommended by the lUPAC-IUB Biochemical Nomenclature Commission. Nucleotides, likewise, may be referred to by their commonly accepted single-letter codes. The terms defined below are more fully defined by reference to the specification as a whole.
In describing the present disclosure, the following terms will be employed and are intended to be defined as indicated below.
By "microbe" is meant any microorganism (including both eukaryotic and prokaryotic microorganisms), such as fungi, yeast, bacteria, actinomycetes, algae and protozoa, as well as other unicellular structures.
By "amplified" is meant the construction of multiple copies of a nucleic acid sequence or multiple copies complementary to the nucleic acid sequence using at least one of the nucleic acid sequences as a template. Amplification systems include the polymerase chain reaction (PCR) system, ligase chain reaction (LCR) system, nucleic acid sequence based amplification (NASBA, Cangene, Mississauga, Ontario), Q-Beta Replicase systems, transcription-based amplification system (TAS) and strand displacement amplification (SDA). See, e.g., Diagnostic Molecular Microbiology: Principles and Applications, Persing, et al., eds., American Society for Microbiology, Washington, DC (1993). The product of amplification is termed an amplicon.
The term "conservatively modified variants" applies to both amino acid and nucleic acid sequences. With respect to particular nucleic acid sequences, conservatively modified variants refer to those nucleic acids that encode identical or conservatively modified variants of the amino acid sequences. Because of the degeneracy of the genetic code, a large number of functionally identical nucleic acids encode any given protein. For instance, the codons GCA, GCC, GCG and GCU all encode the amino acid alanine. Thus, at every position where an alanine is specified by a codon, the codon can be altered to any of the corresponding codons described without altering the encoded polypeptide. Such nucleic acid variations are "silent variations" and represent one species of conservatively modified variation. Every nucleic acid sequence herein that encodes a polypeptide also describes every possible silent variation of the nucleic acid. One of ordinary skill will recognize that each codon in a nucleic acid (except AUG, which is ordinarily the only codon for methionine; one exception is Micrococcus rubens, for which GTG is the methionine codon (Ishizuka, et al. , (1993) J. Gen. Microbiol. 139:425- 32)) can be modified to yield a functionally identical molecule. Accordingly, each silent variation of a nucleic acid, which encodes a polypeptide of the present disclosure, is implicit in each described polypeptide sequence and incorporated herein by reference.
As to amino acid sequences, one of skill will recognize that individual substitution, deletion or addition to a nucleic acid, peptide, polypeptide or protein sequence which alters, adds or deletes a single amino acid or a small percentage of amino acids in the encoded sequence is a "conservatively modified variant" when the alteration results in the substitution of an amino acid with a chemically similar amino acid. Thus, any number of amino acid residues selected from the group of integers consisting of from 1 to 15 can be so altered. Thus, for example, 1 , 2, 3, 4, 5, 7 or 10 alterations can be made. Conservatively modified variants typically provide similar biological activity as the unmodified polypeptide sequence from which they are derived. For example, substrate specificity, enzyme activity or ligand/receptor binding is generally at least 30%, 40%, 50%, 60%, 70%, 80% or 90%, preferably 60-90% of the native protein for its native substrate. Conservative substitution tables providing functionally similar amino acids are well known in the art.
The following six groups each contain amino acids that are conservative substitutions for one another:
1 ) Alanine (A), Serine (S), Threonine (T);
2) Aspartic acid (D), Glutamic acid (E);
3) Asparagine (N), Glutamine (Q);
4) Arginine (R), Lysine (K);
5) Isoleucine (I), Leucine (L), Methionine (M), Valine (V) and
6) Phenylalanine (F), Tyrosine (Y), Tryptophan (W).
See also, Creighton, Proteins, W.H. Freeman and Co. (1984).
As used herein, "consisting essentially of" means the inclusion of additional sequences to an object polynucleotide or polypeptide where the additional sequences do not materially affect the basic function of the claimed polynucleotide or polypeptide sequences.
The term "construct" is used to refer generally to an artificial combination of polynucleotide sequences, i.e. a combination which does not occur in nature, normally comprising one or more regulatory elements and one or more coding sequences. The term may include reference to expression cassettes and/or vector sequences, as is appropriate for the context.
A "control" or "control plant" or "control plant cell" provides a reference point for measuring changes in phenotype of a subject plant or plant cell in which genetic alteration, such as transformation, has been effected as to a gene of interest. A subject plant or plant cell may be descended from a plant or cell so altered and will comprise the alteration.
A control plant or plant cell may comprise, for example: (a) a wild-type plant or cell, i.e., of the same genotype as the starting material for the genetic alteration which resulted in the subject plant or cell; (b) a plant or plant cell of the same genotype as the starting material but which has been transformed with a null construct (i.e., with a construct which has no known effect on the trait of interest, such as a construct comprising a marker gene); (c) a plant or plant cell which is a non-transformed segregant among progeny of a subject plant or plant cell; (d) a plant or plant cell genetically identical to the subject plant or plant cell but which is not exposed to conditions or stimuli that would induce expression of the gene of interest; or (e) the subject plant or plant cell itself, under conditions in which the gene of interest is not expressed. A control plant may also be a plant transformed with an alternative construct.
By "encoding" or "encoded," with respect to a specified nucleic acid, is meant comprising the information for translation into the specified protein. A nucleic acid encoding a protein may comprise non-translated sequences (e.g., introns) within translated regions of the nucleic acid or may lack such intervening non-translated sequences (e.g., as in cDNA). The information by which a protein is encoded is specified by the use of codons. Typically, the amino acid sequence is encoded by the nucleic acid using the "universal" genetic code. However, variants of the universal code, such as is present in some plant, animal and fungal mitochondria, the bacterium Mycoplasma capricolum (Yamao, et al., (1985) Proc. Natl. Acad. Sci. USA 82:2306-9) or the ciliate Macronucleus, may be used when the nucleic acid is expressed using these organisms.
When the nucleic acid is prepared or altered synthetically, advantage can be taken of known codon preferences of the intended host where the nucleic acid is to be expressed. For example, although nucleic acid sequences of the present disclosure may be expressed in both monocotyledonous and dicotyledonous plant species, sequences can be modified to account for the specific codon preferences and GC content preferences of monocotyledonous plants or dicotyledonous plants as these preferences have been shown to differ (Murray, et al., (1989) Nucleic Acids Res. 17:477-98 and herein incorporated by reference). Thus, the maize preferred codon for a particular amino acid might be derived from known gene sequences from maize. Maize codon usage for 28 genes from maize plants is listed in Table 4 of Murray, et al., supra.
As used herein, the term "endogenous", when used in reference to a gene, means a gene that is normally present in the genome of cells of a specified organism and is present in its normal state in the cells (i.e., present in the genome in the state in which it normally is present in nature).
The term "exogenous" is used herein to refer to any material that is introduced into a cell. The term "exogenous nucleic acid molecule" or "transgene" refers to any nucleic acid molecule that either is not normally present in a cell genome or is introduced into a cell. Such exogenous nucleic acid molecules generally are recombinant nucleic acid molecules, which are generated using recombinant DNA methods as disclosed herein or otherwise known in the art. In various embodiments, a transgenic non-human organism as disclosed herein, can contain, for example, a first transgene and a second transgene. Such first and second transgenes can be introduced into a cell, for example, a progenitor cell of a transgenic organism, either as individual nucleic acid molecules or as a single unit (e.g., contained in different vectors or contained in a single vector, respectively). In either case, confirmation may be made that a cell from which the transgenic organism is to be derived contains both of the transgenes using routine and well-known methods such as expression of marker genes or nucleic acid hybridization or PCR analysis. Alternatively, or additionally, confirmation of the presence of transgenes may occur later, for example, after regeneration of a plant from a putatively transformed cell.
As used herein, "heterologous" in reference to a nucleic acid is a nucleic acid that originates from a foreign species, or, if from the same species, is substantially modified from its native form in composition and/or genomic locus by deliberate human intervention. For example, a promoter operably linked to a heterologous structural gene is from a species different from that from which the structural gene was derived or, if from the same species, one or both are substantially modified from their original form. A heterologous protein may originate from a foreign species or, if from the same species, is substantially modified from its original form by deliberate human intervention.
By "host cell" is meant a cell which comprises a heterologous nucleic acid sequence of the disclosure, which contains a vector and supports the replication and/or expression of the expression vector. Host cells may be prokaryotic cells such as E. coli, or eukaryotic cells such as yeast, insect, plant, amphibian or mammalian cells. Preferably, host cells are monocotyledonous or dicotyledonous plant cells, including but not limited to maize, sorghum, sunflower, soybean, wheat, alfalfa, rice, cotton, canola, barley, millet and tomato. A particularly preferred monocotyledonous host cell is a maize host cell.
The term "hybridization complex" includes reference to a duplex nucleic acid structure formed by two single-stranded nucleic acid sequences selectively hybridized with each other.
The term "introduced" in the context of inserting a nucleic acid into a cell, means "transfection" or "transformation" or "transduction" and includes reference to the incorporation of a nucleic acid into a eukaryotic or prokaryotic cell where the nucleic acid may be incorporated into the genome of the cell (e.g., chromosome, plasmid, plastid or mitochondrial DNA), converted into an autonomous replicon or transiently expressed (e.g., transfected mRNA).
The terms "isolated" refers to material, such as a nucleic acid or a protein, which is substantially or essentially free from components which normally accompany or interact with it as found in its naturally occurring environment. The terms "non-naturally occurring"; "mutated", "recombinant"; "recombinantly expressed"; "heterologous" or "heterologously expressed" are representative biological materials that are not present in its naturally occurring environment.
With reference to plants, a "line" is a collection of genetically identical plants and encompasses the seeds that produce such plants. An inbred line is typically homozygous at most or all loci.
The term "NUE nucleic acid" means a nucleic acid comprising a polynucleotide
("NUE polynucleotide") encoding a full length or partial length polypeptide.
As used herein, "nucleic acid" includes reference to a deoxyribonucleotide or ribonucleotide polymer in either single- or double-stranded form, and unless otherwise limited, encompasses known analogues having the essential nature of natural nucleotides in that they hybridize to single-stranded nucleic acids in a manner similar to naturally occurring nucleotides (e.g., peptide nucleic acids).
By "nucleic acid library" is meant a collection of isolated DNA or RNA molecules, which comprise and substantially represent the entire transcribed fraction of a genome of a specified organism. Construction of exemplary nucleic acid libraries, such as genomic and cDNA libraries, is taught in standard molecular biology references such as Berger and Kimmel, (1987) Guide To Molecular Cloning Techniques, from the series Methods in
Enzymology, vol. 152, Academic Press, Inc., San Diego, CA; Sambrook, et al., (1989)
Molecular Cloning: A Laboratory Manual, 2nd ed., vols. 1 -3; and Current Protocols in Molecular Biology, Ausubel, et al., eds, Current Protocols, a joint venture between Greene
Publishing Associates, Inc. and John Wiley & Sons, Inc. (1994 Supplement).
As used herein "operably linked" includes reference to a functional linkage between a first sequence, such as a promoter, and a second sequence, wherein the promoter sequence initiates and mediates transcription of the DNA corresponding to the second sequence. Generally, operably linked means that the nucleic acid sequences being linked are contiguous and, where necessary to join two protein coding regions, contiguous and in the same reading frame.
As used herein, the term "plant" includes reference to whole plants, plant organs
(e.g., leaves, stems, roots, etc.), seeds and plant cells and progeny of same. Plant cell, as used herein includes, without limitation, a cell present in or isolated from plant tissues including seeds, suspension cultures, embryos, meristematic regions, callus tissue, leaves, roots, shoots, gametophytes, sporophytes, pollen and microspores. The class of plants which can be used in the methods of the disclosure is generally as broad as the class of higher plants amenable to transformation techniques, including both monocotyledonous and dicotyledonous plants including species from the genera: Cucurbita, Rosa, Vitis, Juglans, Fragaria, Lotus, Medicago, Onobrychis, Trifolium, Trigonella, Vigna, Citrus, Linum, Geranium, Manihot, Daucus, Arabidopsis, Brassica, Raphanus, Sinapis, Atropa, Capsicum, Datura, Hyoscyamus, Lycopersicon, Nicotiana, Solarium, Petunia, Digitalis, Majorana, Ciahorium, Helianthus, Lactuca, Bromus, Asparagus, Antirrhinum, Heterocallis, Nemesis, Pelargonium, Panieum, Pennisetum, Ranunculus, Senecio, Salpiglossis, Cucumis, Browaalia, Glycine, Pisum, Phaseolus, Lolium, Oryza, Avena, Hordeum, Secale, Allium and Triticum. A particularly preferred plant is Zea mays.
As used herein, "yield" may include reference to bushels per acre of a grain crop at harvest, as adjusted for grain moisture (15% typically for maize, for example) and/or the volume of biomass generated (for forage crops such as alfalfa and plant root size for multiple crops). Grain moisture is measured in the grain at harvest. The adjusted test weight of grain is determined to be the weight in pounds per bushel, adjusted for grain moisture level at harvest. Biomass is measured as the weight of harvestable plant material generated.
As used herein, "polynucleotide" includes reference to a deoxyribopolynucleotide, ribopolynucleotide or analogs thereof that have the essential nature of a natural ribonucleotide in that they hybridize, under stringent hybridization conditions, to substantially the same nucleotide sequence as naturally occurring nucleotides and/or allow translation into the same amino acid(s) as the naturally occurring nucleotide(s). A polynucleotide can be full-length or a subsequence of a native or heterologous structural or regulatory gene. Unless otherwise indicated, the term may include reference to the specified sequence as well as the complementary sequence thereof.
The terms "polypeptide," "peptide" and "protein" are used interchangeably herein to refer to a polymer of amino acid residues. The terms apply to 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 naturally occurring amino acid polymers.
As used herein "promoter" includes reference to a region of DNA upstream from the start of transcription and involved in recognition and binding of RNA polymerase and other proteins to initiate transcription. A "plant promoter" is a promoter capable of initiating transcription in plant cells. Exemplary plant promoters include, but are not limited to, those that are obtained from plants, plant viruses and bacteria which comprise genes expressed in plant cells such as Agrobacterium or Rhizobium. Examples are promoters that preferentially initiate transcription in certain tissues, such as leaves, roots, seeds, fibres, xylem vessels, tracheids or sclerenchyma. Such promoters are referred to as "tissue preferred." A "cell type" specific promoter primarily drives expression in certain cell types in one or more organs, for example, vascular cells in roots or leaves. An "inducible" or "regulatable" promoter is a promoter which is under environmental control. Examples of environmental conditions that may effect transcription by inducible promoters include anaerobic conditions or the presence of light. Another type of promoter is a developmental^ regulated promoter, for example, a promoter that drives expression during pollen development. Tissue preferred, cell type specific, developmental^ regulated and inducible promoters are members of the class of "non-constitutive" promoters. A "constitutive" promoter is a promoter which is active in essentially all tissues of a plant, under most environmental conditions and states of development or cell differentiation.
The term "polypeptide" refers to one or more amino acid sequences. The term is also inclusive of fragments, variants, homologs, alleles or precursors (e.g., preproproteins or proproteins) thereof. A "NUE protein" comprises a polypeptide. Unless otherwise stated, the term "NUE nucleic acid" means a nucleic acid comprising a polynucleotide ("NUE polynucleotide") encoding a polypeptide.
As used herein "recombinant" includes reference to a cell or vector, that has been modified by the introduction of a heterologous nucleic acid or that the cell is derived from a cell so modified. Thus, for example, recombinant cells express genes that are not found in identical form within the native (non-recombinant) form of the cell or express native genes that are otherwise abnormally expressed, under expressed or not expressed at all as a result of deliberate human intervention or may have reduced or eliminated expression of a native gene. The term "recombinant" as used herein does not encompass the alteration of the cell or vector by naturally occurring events (e.g., spontaneous mutation, natural transformation/transduction/transposition) such as those occurring without deliberate human intervention.
As used herein, a "recombinant expression cassette" is a nucleic acid construct, generated recombinantly or synthetically, with a series of specified nucleic acid elements, which permit transcription of a particular nucleic acid in a target cell. The recombinant expression cassette can be incorporated into a plasmid, chromosome, mitochondrial DNA, plastid DNA, virus or nucleic acid fragment. Typically, the recombinant expression cassette portion of an expression vector includes, among other sequences, a nucleic acid to be transcribed and a promoter. The term "selectively hybridizes" includes reference to hybridization, under stringent hybridization conditions, of a nucleic acid sequence to a specified nucleic acid target sequence to a detectably greater degree (e.g., at least 2-fold over background) than its hybridization to non-target nucleic acid sequences and to the substantial exclusion of non-target nucleic acids. Selectively hybridizing sequences typically have about at least 40% sequence identity, preferably 60-90% sequence identity and most preferably 100% sequence identity (i.e., complementary) with each other.
The terms "stringent conditions" or "stringent hybridization conditions" include reference to conditions under which a probe will hybridize to its target sequence, to a detectably greater degree than other sequences (e.g., at least 2-fold over background). Stringent conditions are sequence-dependent and will be different in different circumstances. By controlling the stringency of the hybridization and/or washing conditions, target sequences can be identified which can be up to 100% complementary to the probe (homologous probing). Alternatively, stringency conditions can be adjusted to allow some mismatching in sequences so that lower degrees of similarity are detected (heterologous probing). Optimally, the probe is approximately 500 nucleotides in length, but can vary greatly in length from less than 500 nucleotides to equal to the entire length of the target sequence.
Typically, stringent conditions will be those in which the salt concentration is less than about 1.5 M Na ion, typically about 0.01 to 1 .0 M Na ion concentration (or other salts) at pH 7.0 to 8.3 and the temperature is at least about 30°C for short probes (e.g., 10 to 50 nucleotides) and at least about 60°C for long probes (e.g., greater than 50 nucleotides). Stringent conditions may also be achieved with the addition of destabilizing agents such as formamide or Denhardt's. Exemplary low stringency conditions include hybridization with a buffer solution of 30 to 35% formamide, 1 M NaCI, 1 % SDS (sodium dodecyl sulphate) at 37°C and a wash in 1X to 2X SSC (20X SSC = 3.0 M NaCI/0.3 M trisodium citrate) at 50 to 55°C. Exemplary moderate stringency conditions include hybridization in 40 to 45% formamide, 1 M NaCI, 1 % SDS at 37°C and a wash in 0.5X to 1X SSC at 55 to 60°C. Exemplary high stringency conditions include hybridization in 50% formamide, 1 M NaCI, 1 % SDS at 37°C and a wash in 0.1 X SSC at 60 to 65°C. Specificity is typically the function of post-hybridization washes, the critical factors being the ionic strength and temperature of the final wash solution. For DNA-DNA hybrids, the Tm can be approximated from the equation of Meinkoth and Wahl, (1984) Anal. Biochem., 138:267- 84: Tm = 81.5°C + 16.6 (log M) + 0.41 (%GC) - 0.61 (% form) - 500/L; where M is the molarity of monovalent cations, %GC is the percentage of guanosine and cytosine nucleotides in the DNA, % form is the percentage of formamide in the hybridization solution, and L is the length of the hybrid in base pairs. The Tm is the temperature (under defined ionic strength and pH) at which 50% of a complementary target sequence hybridizes to a perfectly matched probe. Tm is reduced by about 1 °C for each 1 % of mismatching; thus, Tm, hybridization and/or wash conditions can be adjusted to hybridize to sequences of the desired identity. For example, if sequences with >90% identity are sought, the Tm can be decreased 10°C. Generally, stringent conditions are selected to be about 5°C lower than the thermal melting point (Tm) for the specific sequence and its complement at a defined ionic strength and pH. However, severely stringent conditions can utilize a hybridization and/or wash at 1 , 2, 3 or 4°C lower than the thermal melting point (Tm); moderately stringent conditions can utilize a hybridization and/or wash at 6, 7, 8, 9 or 10°C lower than the thermal melting point (Tm); low stringency conditions can utilize a hybridization and/or wash at 1 1 , 12, 13, 14, 15 or 20°C lower than the thermal melting point (Tm). Using the equation, hybridization and wash compositions, and desired Tm, those of ordinary skill will understand that variations in the stringency of hybridization and/or wash solutions are inherently described. If the desired degree of mismatching results in a Tm of less than 45°C (aqueous solution) or 32°C (formamide solution) it is preferred to increase the SSC concentration so that a higher temperature can be used. An extensive guide to the hybridization of nucleic acids is found in Tijssen, Laboratory Techniques in Biochemistry and Molecular Biology - Hybridization with Nucleic Acid Probes, part I, chapter 2, Overview of principles of hybridization and the strategy of nucleic acid probe assays," Elsevier, New York (1993); and Current Protocols in Molecular Biology, chapter 2, Ausubel, et al., eds, Greene Publishing and Wiley- Interscience, New York (1995). Unless otherwise stated, in the present application high stringency is defined as hybridization in 4X SSC, 5X Denhardt's (5 g Ficoll, 5 g polyvinypyrrolidone, 5 g bovine serum albumin in 500ml of water), 0.1 mg/ml boiled salmon sperm DNA, and 25 mM Na phosphate at 65°C and a wash in 0.1 X SSC, 0.1 % SDS at 65°C.
As used herein, "transgenic plant" includes reference to a plant which comprises within its genome a heterologous polynucleotide. Generally, the heterologous polynucleotide is stably integrated within the genome such that the polynucleotide is passed on to successive generations. The heterologous polynucleotide may be integrated into the genome alone or as part of a recombinant expression cassette. "Transgenic" is used herein to include any cell, cell line, callus, tissue, plant part or plant, the genotype of which has been altered by the presence of heterologous nucleic acid including those transgenics initially so altered as well as those created by sexual crosses or asexual propagation from the initial transgenic. The term "transgenic" as used herein does not encompass the alteration of the genome (chromosomal or extra-chromosomal) by conventional plant breeding methods or by naturally occurring events such as random cross-fertilization, non-recombinant viral infection, non-recombinant bacterial transformation, non-recombinant transposition or spontaneous mutation.
As used herein, "vector" includes reference to a nucleic acid used in transfection of a host cell and into which can be inserted a polynucleotide. Vectors are often replicons. Expression vectors permit transcription of a nucleic acid inserted therein.
The following terms are used to describe the sequence relationships between two or more nucleic acids or polynucleotides or polypeptides: (a) "reference sequence," (b) "comparison window," (c) "sequence identity," (d) "percentage of sequence identity" and (e) "substantial identity."
As used herein, "reference sequence" is a defined sequence used as a basis for sequence comparison. A reference sequence may be a subset or the entirety of a specified sequence; for example, as a segment of a full-length cDNA or gene sequence or the complete cDNA or gene sequence.
As used herein, "comparison window" means includes reference to a contiguous and specified segment of a polynucleotide sequence, wherein the polynucleotide sequence may be compared to a reference sequence and wherein the portion of the polynucleotide sequence in the comparison window may comprise additions or deletions (i.e., gaps) compared to the reference sequence (which does not comprise additions or deletions) for optimal alignment of the two sequences. Generally, the comparison window is at least 20 contiguous nucleotides in length, and optionally can be 30, 40, 50, 100 or longer. Those of skill in the art understand that to avoid a high similarity to a reference sequence due to inclusion of gaps in the polynucleotide sequence a gap penalty is typically introduced and is subtracted from the number of matches.
Methods of alignment of nucleotide and amino acid sequences for comparison are well known in the art. The local homology algorithm (BESTFIT) of Smith and Waterman, (1981 ) Adv. Appl. Math 2:482, may conduct optimal alignment of sequences for comparison; by the homology alignment algorithm (GAP) of Needleman and Wunsch, (1970) J. Mol. Biol. 48:443-53; by the search for similarity method (Tfasta and Fasta) of Pearson and Lipman, (1988) Proc. Natl. Acad. Sci. USA 85:2444; by computerized implementations of these algorithms, including, but not limited to: CLUSTAL in the PC/Gene program by Intelligenetics, Mountain View, California, GAP, BESTFIT, BLAST, FASTA and TFASTA in the Wisconsin Genetics Software Package, Version 8 (available from Genetics Computer Group (GCG® programs (Accelrys, Inc., San Diego, CA).). The CLUSTAL program is well described by Higgins and Sharp, (1988) Gene 73:237-44; Higgins and Sharp, (1989) CABIOS 5:151-3; Corpet, et al., (1988) Nucleic Acids Res. 16:10881-90; Huang, et al., (1992) Computer Applications in the Biosciences 8:155-65 and Pearson, et al., (1994) Meth. Mol. Biol. 24:307-31. The preferred program to use for optimal global alignment of multiple sequences is PileUp (Feng and Doolittle, (1987) J. Mol. Evol. , 25:351-60 which is similar to the method described by Higgins and Sharp, (1989) CABIOS 5:151 -53 and hereby incorporated by reference). The BLAST family of programs which can be used for database similarity searches includes: BLASTN for nucleotide query sequences against nucleotide database sequences; BLASTX for nucleotide query sequences against protein database sequences; BLASTP for protein query sequences against protein database sequences; TBLASTN for protein query sequences against nucleotide database sequences and TBLASTX for nucleotide query sequences against nucleotide database sequences. See, Current Protocols in Molecular Biology, Chapter 19, Ausubel et al., eds., Greene Publishing and Wiley-lnterscience, New York (1995).
GAP uses the algorithm of Needleman and Wunsch, supra, to find the alignment of two complete sequences that maximizes the number of matches and minimizes the number of gaps. GAP considers all possible alignments and gap positions and creates the alignment with the largest number of matched bases and the fewest gaps. It allows for the provision of a gap creation penalty and a gap extension penalty in units of matched bases. GAP must make a profit of gap creation penalty number of matches for each gap it inserts. If a gap extension penalty greater than zero is chosen, GAP must, in addition, make a profit for each gap inserted of the length of the gap times the gap extension penalty. Default gap creation penalty values and gap extension penalty values in Version 10 of the Wisconsin Genetics Software Package are 8 and 2, respectively. The gap creation and gap extension penalties can be expressed as an integer selected from the group of integers consisting of from 0 to 100. Thus, for example, the gap creation and gap extension penalties can be 0, 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, 50 or greater.
GAP presents one member of the family of best alignments. There may be many members of this family, but no other member has a better quality. GAP displays four figures of merit for alignments: Quality, Ratio, Identity and Similarity. The Quality is the metric maximized in order to align the sequences. Ratio is the quality divided by the number of bases in the shorter segment. Percent Identity is the percent of the symbols that actually match. Percent Similarity is the percent of the symbols that are similar. Symbols that are across from gaps are ignored. A similarity is scored when the scoring matrix value for a pair of symbols is greater than or equal to 0.50, the similarity threshold. The scoring matrix used in Version 10 of the Wisconsin Genetics Software Package is BLOSUM62 (see, Henikoff and Henikoff, (1989) Proc. Natl. Acad. Sci. USA 89:10915).
Unless otherwise stated, sequence identity/similarity values provided herein refer to the value obtained using the BLAST 2.0 suite of programs using default parameters (Altschul, et ai, (1997) Nucleic Acids Res. 25:3389-402).
As those of ordinary skill in the art will understand, BLAST searches assume that proteins can be modeled as random sequences. However, many real proteins comprise regions of nonrandom sequences, which may be homopolymeric tracts, short-period repeats, or regions enriched in one or more amino acids. Such low-complexity regions may be aligned between unrelated proteins even though other regions of the protein are entirely dissimilar. A number of low-complexity filter programs can be employed to reduce such low-complexity alignments. For example, the SEG (Wooten and Federhen, (1993) Comput. Chem. 17:149-63) and XNU (Claverie and States, (1993) Comput. Chem. 17:191-201 ) low-complexity filters can be employed alone or in combination.
As used herein, "sequence identity" or "identity" in the context of two nucleic acid or polypeptide sequences includes reference to the residues in the two sequences which are the same when aligned for maximum correspondence over a specified comparison window. When percentage of sequence identity is used in reference to proteins it is recognized that residue positions which are not identical often differ by conservative amino acid substitutions, where amino acid residues are substituted for other amino acid residues with similar chemical properties (e.g., charge or hydrophobicity) and therefore do not change the functional properties of the molecule. Where sequences differ in conservative substitutions, the percent sequence identity may be adjusted upwards to correct for the conservative nature of the substitution. Sequences which differ by such conservative substitutions are said to have "sequence similarity" or "similarity." Means for making this adjustment are well known to those of skill in the art. Typically this involves scoring a conservative substitution as a partial rather than a full mismatch, thereby increasing the percentage sequence identity. Thus, for example, where an identical amino acid is given a score of 1 and a non-conservative substitution is given a score of zero, a conservative substitution is given a score between zero and 1. The scoring of conservative substitutions is calculated, e.g., according to the algorithm of Meyers and Miller, (1988) Computer Applic. Biol. Sci. 4:1 1 -17, e.g., as implemented in the program PC/GENE (Intelligenetics, Mountain View, California, USA).
As used herein, "percentage of sequence identity" means the value determined by comparing two optimally aligned sequences over a comparison window, wherein the portion of the polynucleotide 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 "substantial identity" of polynucleotide sequences means that a polynucleotide comprises a sequence that has between 50-100% sequence identity, optionally at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or at least 95% sequence identity, compared to a reference sequence using one of the alignment programs described using standard parameters. One of skill will recognize that these values can be appropriately adjusted to determine corresponding identity of proteins encoded by two nucleotide sequences by taking into account codon degeneracy, amino acid similarity, reading frame positioning and the like. Substantial identity of amino acid sequences for these purposes normally means sequence identity of between 55-100%, such as at least 55%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, up to 100% identity.
The terms "substantial identity" in the context of a peptide indicates that a peptide comprises a sequence with between 55-100% sequence identity to a reference sequence, such as at least 55%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, up to 100% sequence identity to the reference sequence over a specified comparison window. Preferably, optimal alignment is conducted using the homology alignment algorithm of Needleman and Wunsch, supra. An indication that two peptide sequences are substantially identical is that one peptide is immunologically reactive with antibodies raised against the second peptide. Thus, a peptide is substantially identical to a second peptide, for example, where the two peptides differ only by a conservative substitution. In addition, a peptide can be substantially identical to a second peptide when they differ by a non-conservative change if the epitope that the antibody recognizes is substantially identical. Peptides which are "substantially similar" share sequences as noted above, except that residue positions, which are not identical, may differ by conservative amino acid changes.
TABLE 1
SEQ ID POLYNUCLEOTIDE/POLYPEPTIDE IDENTITY
NUMBER
SEQ ID NO: 1 Polynucleotide Primer
SEQ ID NO: 2 Polynucleotide Primer
SEQ ID NO: 3 Polynucleotide Primer
SEQ ID NO: 4 Polynucleotide Primer
SEQ ID NO: 5 Polynucleotide Primer
SEQ ID NO: 6 Polynucleotide Primer
SEQ ID NO: 7 Polynucleotide Primer
SEQ ID NO: 8 Polynucleotide Primer
SEQ ID NO: 9 Polynucleotide ms44 wildtype genomic
SEQ ID NO: 10 Polypeptide ms44 wildtype protein
SEQ ID NO: 1 1 Polynucleotide Primer
SEQ ID NO: 12 Polynucleotide Primer
SEQ ID NO: 13 Polynucleotide MS44 mutant allele dominant genomic seq
SEQ ID NO: 14 Polypeptide MS44 dominant protein
SEQ ID NO: 15 Polynucleotide MS44 dom CDS
SEQ ID NO: 16 Polypeptide Arabidopsis thaliana
SEQ ID NO: 17 Polypeptide Oryza sativa
SEQ ID NO: 18 Polypeptide Lilium longiflorum
SEQ ID NO: 19 Polypeptide Zea mays YY1
SEQ ID NO: 20 Polypeptide Hordeum vulgare
SEQ ID NO: 21 Polypeptide Oryza brachyantha
SEQ ID NO: 22 Polypeptide Zea mays anther specific
SEQ ID NO: 23 Polypeptide Sorghum bicolor
SEQ ID NO: 24 Polypeptide Lilium longiflorum
SEQ ID NO: 25 Polypeptide Lilium longiflorum
SEQ ID NO: 26 Polypeptide Brassica rapa
SEQ ID NO: 27 Polypeptide Silene latiflia
SEQ ID NO: 28 Polynucleotide Primer
SEQ ID NO: 29 Polynucleotide Primer
SEQ ID NO: 30 Polynucleotide Primer
SEQ ID NO: 31 Polynucleotide Primer
SEQ ID NO: 32 Polynucleotide Primer
SEQ ID NO: 33 Polynucleotide Primer
SEQ ID NO: 34 Polynucleotide Primer
SEQ ID NO: 35 Polynucleotide Primer
SEQ ID NO: 36 Polynucleotide Primer
SEQ ID NO: 37 Polynucleotide Primer
SEQ ID NO: 38 Polynucleotide Primer
SEQ ID NO: 39 Polynucleotide Primer
SEQ ID NO: 40 Polynucleotide Primer
SEQ ID NO: 41 Polynucleotide Primer
SEQ ID NO: 42 Polynucleotide Primer
SEQ ID NO: 43 Polynucleotide Primer
SEQ ID NO: 44 Polynucleotide Primer SEQ ID NO: 45 Polynucleotide Primer
SEQ ID NO: 46 Polynucleotide Primer
SEQ ID NO: 47 Polynucleotide Primer
SEQ ID NO: 48 Polynucleotide Primer
SEQ ID NO: 49 Polynucleotide Primer
SEQ ID NO: 50 Polynucleotide Primer
SEQ ID NO: 51 Polynucleotide Primer
SEQ ID NO: 52 Polynucleotide Primer
SEQ ID NO: 53 Polynucleotide Primer
SEQ ID NO: 54 Polynucleotide Primer
SEQ ID NO: 55 Polynucleotide Primer
SEQ ID NO: 56 Polynucleotide Primer
SEQ ID NO: 57 Polynucleotide Primer
SEQ ID NO: 58 Polynucleotide Primer
SEQ ID NO: 59 Polynucleotide Primer
SEQ ID NO: 60 Polynucleotide Primer
SEQ ID NO: 61 Polynucleotide Primer
SEQ ID NO: 62 Polynucleotide tlsl mutant genomic
SEQ ID NO: 63 Polynucleotide tlsl mutant CDS
SEQ ID NO: 64 Polynucleotide Tassel specific promoter
(variant of SEQ ID NO: 136 from base pairs 1 to 1227)
SEQ ID NO: 65 Polynucleotide Tassel specific promoter
SEQ ID NO: 66 Polynucleotide Tassel specific promoter
SEQ ID NO: 67 Polynucleotide Tassel specific promoter
SEQ ID NO: 68 Polynucleotide Tassel specific promoter
SEQ ID NO: 69 Polynucleotide Tassel specific promoter
SEQ ID NO: 70 Polynucleotide Tassel specific promoter
SEQ ID NO: 71 Polynucleotide Tassel specific promoter
SEQ ID NO: 72 Polynucleotide Tassel specific promoter
SEQ ID NO: 73 Polynucleotide Tassel specific promoter
SEQ ID NO: 74 Polynucleotide Tassel specific promoter
SEQ ID NO: 75 Polynucleotide Tassel specific promoter
SEQ ID NO: 76 Polynucleotide Tassel specific promoter
SEQ ID NO: 77 Polynucleotide Tassel specific promoter
SEQ ID NO: 78 Polynucleotide Tassel specific promoter
SEQ ID NO: 79 Polynucleotide Tassel specific promoter
SEQ ID NO: 80 Polynucleotide Tassel specific promoter
SEQ ID NO: 81 Polynucleotide Tassel specific promoter
SEQ ID NO: 82 Polynucleotide Tassel specific promoter
SEQ ID NO: 83 Polynucleotide Tassel specific promoter
SEQ ID NO: 84 Polynucleotide Tassel specific promoter
SEQ ID NO: 85 Polynucleotide Tassel specific promoter
SEQ ID NO: 86 Polynucleotide Tassel specific promoter
SEQ ID NO: 87 Polynucleotide Tassel specific promoter
SEQ ID NO: 88 Polynucleotide Tassel specific promoter
SEQ ID NO: 89 Polynucleotide Tassel specific promoter
SEQ ID NO: 90 Polynucleotide Tassel specific promoter
SEQ ID NO: 91 Polynucleotide Tassel specific promoter
SEQ ID NO: 92 Polynucleotide Tassel specific promoter
SEQ ID NO: 93 Polynucleotide Tassel specific promoter
Figure imgf000032_0001
Figure imgf000033_0001
Construction of Nucleic Acids
The isolated nucleic acids of the present disclosure can be made using (a) standard recombinant methods, (b) synthetic techniques or combinations thereof. In some embodiments, the polynucleotides of the present disclosure will be cloned, amplified or otherwise constructed from a fungus or bacteria.
UTRs and Codon Preference
In general, translational efficiency has been found to be regulated by specific sequence elements in the 5' non-coding or untranslated region (5' UTR) of the RNA. Positive sequence motifs include translational initiation consensus sequences (Kozak, (1987) Nucleic Acids Res.15:8125) and the 5<G> 7 methyl GpppG RNA cap structure (Drummond, et al. , (1985) Nucleic Acids Res. 13:7375). Negative elements include stable intramolecular 5' UTR stem-loop structures (Muesing, et al. , (1987) Cell 48:691 ) and AUG sequences or short open reading frames preceded by an appropriate AUG in the 5' UTR (Kozak, supra, Rao, et al. , (1988) Mol. and Cell. Biol. 8:284). Accordingly, the present disclosure provides 5' and/or 3' UTR regions for modulation of translation of heterologous coding sequences. Further, the polypeptide-encoding segments of the polynucleotides of the present disclosure can be modified to alter codon usage. Altered codon usage can be employed to alter translational efficiency and/or to optimize the coding sequence for expression in a desired host or to optimize the codon usage in a heterologous sequence for expression in maize. Codon usage in the coding regions of the polynucleotides of the present disclosure can be analyzed statistically using commercially available software packages such as "Codon Preference" available from the University of Wisconsin Genetics Computer Group. See, Devereaux, et al., (1984) Nucleic Acids Res. 12:387-395) or MacVector 4.1 (Eastman Kodak Co., New Haven, CN). Thus, the present disclosure provides a codon usage frequency characteristic of the coding region of at least one of the polynucleotides of the present disclosure. The number of polynucleotides (3 nucleotides per amino acid) that can be used to determine a codon usage frequency can be any integer from 3 to the number of polynucleotides of the present disclosure as provided herein. Optionally, the polynucleotides will be full-length sequences. An exemplary number of sequences for statistical analysis can be at least 1 , 5, 10, 20, 50 or 100.
Sequence Shuffling
The present disclosure provides methods for sequence shuffling using polynucleotides of the present disclosure, and compositions resulting therefrom. Sequence shuffling is described in PCT Publication Number 1996/19256. See also, Zhang, et al. , (1997) Proc. Natl. Acad. Sci. USA 94:4504-9 and Zhao, et al. , (1998) Nature Biotech 16:258-61. Generally, sequence shuffling provides a means for generating libraries of polynucleotides having a desired characteristic, which can be selected or screened for. Libraries of recombinant polynucleotides are generated from a population of related sequence polynucleotides, which comprise sequence regions, which have substantial sequence identity and can be homologously recombined in vitro or in vivo. The population of sequence-recombined polynucleotides comprises a subpopulation of polynucleotides which possess desired or advantageous characteristics and which can be selected by a suitable selection or screening method. The characteristics can be any property or attribute capable of being selected for or detected in a screening system, and may include properties of: an encoded protein, a transcriptional element, a sequence controlling transcription, RNA processing, RNA stability, chromatin conformation, translation or other expression property of a gene or transgene, a replicative element, a protein-binding element or the like, such as any feature which confers a selectable or detectable property. In some embodiments, the selected characteristic will be an altered Km and/or Kcat over the wild-type protein as provided herein. In other embodiments, a protein or polynucleotide generated from sequence shuffling will have a ligand binding affinity greater than the non-shuffled wild-type polynucleotide. In yet other embodiments, a protein or polynucleotide generated from sequence shuffling will have an altered pH optimum as compared to the non-shuffled wild-type polynucleotide. The increase in such properties can be at least 1 10%, 120%, 130%, 140% or greater than 150% of the wild- type value. Recombinant Expression Cassettes
The present disclosure further provides recombinant expression cassettes comprising a nucleic acid of the present disclosure. A nucleic acid sequence coding for the desired polynucleotide of the present disclosure, for example a cDNA or a genomic sequence encoding a polypeptide long enough to code for an active protein of the present disclosure, can be used to construct a recombinant expression cassette which can be introduced into the desired host cell. A recombinant expression cassette will typically comprise a polynucleotide of the present disclosure operably linked to transcriptional initiation regulatory sequences which will direct the transcription of the polynucleotide in the intended host cell, such as tissues of a transformed plant.
For example, plant expression vectors may include (1 ) a cloned plant gene under the transcriptional control of 5' and 3' regulatory sequences and (2) a dominant selectable marker. Such plant expression vectors may also contain, if desired, a promoter regulatory region (e.g., one conferring inducible or constitutive, environmentally- or developmentally- regulated, or cell- or tissue-specific/selective expression), a transcription initiation start site, a ribosome binding site, an RNA processing signal, a transcription termination site and/or a polyadenylation signal.
Promoters, Terminators, Introns
A plant promoter fragment can be employed which will direct expression of a polynucleotide of the present disclosure in essentially all tissues of a regenerated plant. Such promoters are referred to herein as "constitutive" promoters and are active under most environmental conditions and states of development or cell differentiation. Examples of constitutive promoters include the V- or 2'- promoter derived from T-DNA of Agrobacterium tumefaciens, the Smas promoter, the cinnamyl alcohol dehydrogenase promoter (US Patent Number 5,683,439), the Nos promoter, the rubisco promoter, the GRP1 -8 promoter, the 35S promoter from cauliflower mosaic virus (CaMV), as described in Odell, et al., (1985) Nature 313:810-2; rice actin (McElroy, et al., (1990) Plant Cell 163- 171 ); ubiquitin (Christensen, et al., (1992) Plant Mol. Biol. 12:619-632 and Christensen, et al., (1992) Plant Mol. Biol. 18:675-89); pEMU (Last, et al., (1991 ) Theor. Appl. Genet. 81 :581-8); MAS (Velten, et ai, (1984) EMBO J. 3:2723-30) and maize H3 histone (Lepetit, et al., (1992) Mol. Gen. Genet. 231 :276-85 and Atanassvoa, et al., (1992) Plant Journal 2(3):291 -300); ALS promoter, as described in PCT Application Number WO 1996/30530 and other transcription initiation regions from various plant genes known to those of skill. For the present disclosure ubiquitin is the preferred promoter for expression in monocot plants.
Alternatively, the plant promoter can direct expression of a polynucleotide of the present disclosure in a specific tissue or may be otherwise under more precise environmental or developmental control. Such promoters may be "inducible" promoters. Environmental conditions that may effect transcription by inducible promoters include pathogen attack, anaerobic conditions or the presence of light. Examples of inducible promoters are the Adh1 promoter, which is inducible by hypoxia or cold stress, the Hsp70 promoter, which is inducible by heat stress and the PPDK promoter, which is inducible by light. Diurnal promoters that are active at different times during the circadian rhythm are also known (US Patent Application Publication Number 201 1/0167517, incorporated herein by reference).
Examples of promoters under developmental control include promoters that initiate transcription only, or preferentially, in certain tissues, such as leaves, roots, fruit, seeds or flowers. The operation of a promoter may also vary depending on its location in the genome. Thus, an inducible promoter may become fully or partially constitutive in certain locations.
If polypeptide expression is desired, it is generally desirable to include a polyadenylation region at the 3'-end of a polynucleotide coding region. The polyadenylation region can be derived from a variety of plant genes, or from T-DNA. The 3' end sequence to be added can be derived from, for example, the nopaline synthase or octopine synthase genes or alternatively from another plant gene or less preferably from any other eukaryotic gene. Examples of such regulatory elements include, but are not limited to, 3' termination and/or polyadenylation regions such as those of the Agrobacterium tumefaciens nopaline synthase (nos) gene (Bevan, et al., (1983) Nucleic Acids Res. 12:369-85); the potato proteinase inhibitor II (PINII) gene (Keil, et al., (1986) Nucleic Acids Res. 14:5641 -50 and An, et al., (1989) Plant Cell 1 : 1 15-22) and the CaMV 19S gene (Mogen, et al., (1990) Plant Cell 2: 1261-72).
An intron sequence can be added to the 5' untranslated region or the coding sequence of the partial coding sequence to increase the amount of the mature message that accumulates in the cytosol. Inclusion of a spliceable intron in the transcription unit in both plant and animal expression constructs has been shown to increase gene expression at both the mRNA and protein levels up to 1000-fold (Buchman and Berg, (1988) Mol. Cell Biol. 8:4395-4405; Callis, et al., (1987) Genes Dev. 1 :1183-200). Such intron enhancement of gene expression is typically greatest when placed near the 5' end of the transcription unit. Use of maize introns Adh1 -S intron 1 , 2 and 6, the Bronze-1 intron are known in the art. See generally, The Maize Handbook, Chapter 1 16, Freeling and Walbot, eds., Springer, New York (1994).
Signal Peptide Sequences
Plant signal sequences, including, but not limited to, signal-peptide encoding
DNA/RNA sequences which target proteins to the extracellular matrix of the plant cell (Dratewka-Kos, et al., (1989) J. Biol. Chem. 264:4896-900), such as the Nicotiana plumbaginifolia extension gene (DeLoose, et al., (1991 ) Gene 99:95-100); signal peptides which target proteins to the vacuole, such as the sweet potato sporamin gene (Matsuka, et al., (1991 ) Proc. Natl. Acad. Sci. USA 88:834) and the barley lectin gene (Wilkins, et al., (1990) Plant Cell, 2:301 -13); signal peptides which cause proteins to be secreted, such as that of PRIb (Lind, et al., (1992) Plant Mol. Biol. 18:47-53) or the barley alpha amylase (BAA) (Rahmatullah, et al. , (1989) Plant Mol. Biol. 12:1 19) or signal peptides which target proteins to the plastids such as that of rapeseed enoyl-Acp reductase (Verwaert, et al., (1994) Plant Mol. Biol. 26:189-202) are useful in the disclosure.
Markers
The vector comprising the sequences from a polynucleotide of the present disclosure will typically comprise a marker gene, which confers a selectable phenotype on plant cells. The selectable marker gene may encode antibiotic resistance, with suitable genes including genes coding for resistance to the antibiotic spectinomycin (e.g., the aada gene), the streptomycin phosphotransferase (SPT) gene coding for streptomycin resistance, the neomycin phosphotransferase (NPTII) gene encoding kanamycin or geneticin resistance, the hygromycin phosphotransferase (HPT) gene coding for hygromycin resistance. Also useful are genes coding for resistance to herbicides which act to inhibit the action of acetolactate synthase (ALS), in particular the sulfonylurea-type herbicides (e.g., the acetolactate synthase (ALS) gene containing mutations leading to such resistance in particular the S4 and/or Hra mutations), genes coding for resistance to herbicides which act to inhibit action of glutamine synthase, such as phosphinothricin or basta (e.g., the bar gene), or other such genes known in the art. The bar gene encodes resistance to the herbicide basta and the ALS gene encodes resistance to the herbicide chlorsulfuron.
Constructs described herein may comprise a polynucleotide of interest encoding a reporter or marker product. Examples of suitable reporter polynucleotides known in the art can be found in, for example, Jefferson et al. (1991 ) in Plant Molecular Biology Manual, ed. Gelvin et al. (Kluwer Academic Publishers), pp. 1-33; DeWet et al. Mol. Cell. Biol. 7:725-737 (1987); Goff et al. EMBO J. 9:2517-2522 (1990); Kain et al. BioTechniques 19:650-655 (1995); and Chiu et al. Current Biology 6:325-330 (1996). In certain embodiments, the polynucleotide of interest encodes a selectable reporter. These can include polynucleotides that confer antibiotic resistance or resistance to herbicides.
Examples of suitable selectable marker polynucleotides include, but are not limited to, genes encoding resistance to chloramphenicol, methotrexate, hygromycin, streptomycin, spectinomycin, bleomycin, sulfonamide, bromoxynil, glyphosate, and phosphinothricin.
In some embodiments, the expression cassettes disclosed herein comprise a polynucleotide of interest encoding scorable or screenable markers, where presence of the polynucleotide produces a measurable product. Examples include a β-glucuronidase, or uidA gene (GUS), which encodes an enzyme for which various chromogenic substrates are known (for example, U.S. Pat. Nos. 5,268,463 and 5,599,670); chloramphenicol acetyl transferase, and alkaline phosphatase. Other screenable markers include the
anthocyanin/flavonoid polynucleotides including, for example, a R-locus polynucleotide, which encodes a product that regulates the production of anthocyanin pigments (red color) in plant tissues, the genes which control biosynthesis of flavonoid pigments, such as the maize C1 and C2 , the B gene, the p1 gene, and the bronze locus genes, among others. Further examples of suitable markers encoded by polynucleotides of interest include the cyan fluorescent protein (CYP) gene, the yellow fluorescent protein gene, a lux gene, which encodes a luciferase, the presence of which may be detected using, for example, X-ray film, scintillation counting, fluorescent spectrophotometry, low-light video cameras, photon counting cameras or multiwell luminometry, a green fluorescent protein (GFP), and DsRed2 (Clontechniques, 2001 ) where plant cells transformed with the marker gene are red in color, and thus visually selectable. Additional examples include a p-lactamase gene encoding an enzyme for which various chromogenic substrates are known (e.g., PADAC, a chromogenic cephalosporin), a xylE gene encoding a catechol dioxygenase that can convert chromogenic catechols, an oamylase gene, and a tyrosinase gene encoding an enzyme capable of oxidizing tyrosine to DOPA and dopaquinone, which in turn condenses to form the easily detectable compound melanin.
The expression cassette can also comprise a selectable marker gene for the selection of transformed cells. Selectable marker genes are utilized for the selection of transformed cells or tissues. Marker genes include genes encoding antibiotic resistance, such as those encoding neomycin phosphotransferase II (NEO) and hygromycin
phosphotransferase (HPT), as well as genes conferring resistance to herbicidal compounds, such as glufosinate ammonium, bromoxynil, imidazolinones, and 2,4- dichlorophenoxyacetate (2,4-D). Additional selectable markers include phenotypic markers such as β-galactosidase and fluorescent proteins such as green fluorescent protein (GFP) (Su et al. (2004) Biotechnol Bioeng 85:610-9 and Fetter et al. (2004) Plant Cell 16:215- 28), cyan florescent protein (CYP) (Bolte et al. (2004) J. Cell Science 117:943-54 and
Kato et al. (2002) Plant Physiol 729:913-42), and yellow florescent protein (PhiYFP™from Evrogen, see, Bolte et al. (2004) J. Cell Science 117:943-54). For additional selectable markers, see generally, Yarranton (1992) Curr. Opin. Biotech. 3:506-511 ; Christopherson et al. (1992) Proc. Natl. Acad. Sci. USA 89:6314-6318; Yao et al. (1992) Cell 71 :63-72;
Reznikoff (1992) Mol. Microbiol. 6:2419-2422; Barkley et al. (1980) in The Operon, pp. 177- 220; Hu et al. (1987) Cell 48:555-566; Brown et al. (1987) Cell 49:603-612; Figge et al. (1988) Cell 52:713-722; Deuschle et al. (1989) Proc. Natl. Acad. Aci. USA 86:5400-5404; Fuerst et al. (1989) Proc. Natl. Acad. Sci. USA 86:2549-2553; Deuschle et al. (1990) Science 248:480-483; Gossen (1993) Ph.D. Thesis, University of Heidelberg; Reines et al. (1993) Proc. Natl. Acad. Sci. USA 90:1917-1921 ; Labow et al. (1990) Mol. Cell. Biol.
10:3343-3356; Zambretti et al. (1992) Proc. Natl. Acad. Sci. USA 89:3952-3956; Bairn et al. (1991 ) Proc. Natl. Acad. Sci. USA 88:5072-5076; Wyborski et al. (1991 ) Nucleic Acids Res. 19:4647-4653; Hillenand-Wissman (1989) Topics Mol. Struc. Biol. 10:143-162; Degenkolb ei al. (1991 ) Antimicrob. Agents Chemother. 35:1591-1595; Kleinschnidt et al. (1988)
Biochemistry 27:1094-1 104; Bonin (1993) Ph.D. Thesis, University of Heidelberg; Gossen et al. (1992) Proc. Natl. Acad. Sci. USA 89:5547-5551 ; Oliva et al. (1992) Antimicrob. Agents Chemother. 36:913-919; Hlavka et al. (1985) Handbook of Experimental Pharmacology, Vol. 78 ( Springer-Verlag, Berlin); Gill et al. (1988) Nature 334:721-724. Such disclosures are herein incorporated by reference. The above list of selectable marker genes is not meant to be limiting. Any selectable marker gene can be used in the compositions and methods disclosed herein.
Typical vectors useful for expression of genes in higher plants are well known in the art and include vectors derived from the tumor-inducing (Ti) plasmid of Agrobacterium tumefaciens described by Rogers, et al., (1987) Meth. Enzymol. 153:253-77. These vectors are plant integrating vectors in that on transformation, the vectors integrate a portion of vector DNA into the genome of the host plant. Exemplary A. tumefaciens vectors useful herein are plasmids pKYLX6 and pKYLX7 of Schardl, et al., (1987) Gene 61 :1-1 1 and Berger, et al., (1989) Proc. Natl. Acad. Sci. USA, 86:8402-6. Another useful vector herein is plasmid pBI 101.2 that is available from CLONTECH Laboratories, Inc. (Palo Alto, CA).
Expression of Proteins in Host Cells
Using the nucleic acids of the present disclosure, one may express a protein of the present disclosure in a recombinantly engineered cell such as bacteria, yeast, insect, mammalian or preferably plant cells. The cells produce the protein in a non-natural condition (e.g., in quantity, composition, location and/or time), because they have been genetically altered through human intervention to do so.
It is expected that those of skill in the art are knowledgeable in the numerous expression systems available for expression of a nucleic acid encoding a protein of the present disclosure. No attempt to describe in detail the various methods known for the expression of proteins in prokaryotes or eukaryotes will be made.
In brief summary, the expression of isolated nucleic acids encoding a protein of the present disclosure will typically be achieved by operably linking, for example, the DNA or cDNA to a promoter, followed by incorporation into an expression vector. The vectors can be suitable for replication and integration in either prokaryotes or eukaryotes. Typical expression vectors contain transcription and translation terminators, initiation sequences and promoters useful for regulation of the expression of the DNA of the present disclosure. To obtain high level expression of a cloned gene, it is desirable to construct expression vectors which contain, at the minimum, a strong promoter, such as ubiquitin, to direct transcription, a ribosome binding site for translational initiation and a transcription/translation terminator. Constitutive promoters are classified as providing for a range of constitutive expression. Thus, some are weak constitutive promoters and others are strong constitutive promoters. Generally, by "weak promoter" is intended a promoter that drives expression of a coding sequence at a low level. By "low level" is intended at levels of about 1/10,000 transcripts to about 1/100,000 transcripts to about 1/500,000 transcripts. Conversely, a "strong promoter" drives expression of a coding sequence at a "high level," or about 1/10 transcripts to about 1/100 transcripts to about 1/1 ,000 transcripts.
One of skill would recognize that modifications could be made to a protein of the present disclosure without diminishing its biological activity. Some modifications may be made to facilitate the cloning, expression or incorporation of the targeting molecule into a fusion protein. Such modifications are well known to those of skill in the art and include, for example, a methionine added at the amino terminus to provide an initiation site or additional amino acids (e.g., poly His) placed on either terminus to create conveniently located restriction sites or termination codons or purification sequences.
Expression in Prokaryotes
Prokaryotic cells may be used as hosts for expression. Prokaryotes most frequently are represented by various strains of £ coir, however, other microbial strains may also be used. Commonly used prokaryotic control sequences which are defined herein to include promoters for transcription initiation, optionally with an operator, along with ribosome binding site sequences, include such commonly used promoters as the beta lactamase (penicillinase) and lactose (lac) promoter systems (Chang, et al., (1977) Nature 198:1056), the tryptophan (trp) promoter system (Goeddel, et al., (1980) Nucleic Acids Res. 8:4057) and the lambda derived P L promoter and N-gene ribosome binding site (Shimatake, et al., (1981 ) Nature 292:128). The inclusion of selection markers in DNA vectors transfected in £ coli is also useful. Examples of such markers include genes specifying resistance to ampicillin, tetracycline or chloramphenicol.
The vector is selected to allow introduction of the gene of interest into the appropriate host cell. Bacterial vectors are typically of plasmid or phage origin. Appropriate bacterial cells are infected with phage vector particles or transfected with naked phage vector DNA. If a plasmid vector is used, the bacterial cells are transfected with the plasmid vector DNA. Expression systems for expressing a protein of the present disclosure are available using Bacillus sp. and Salmonella (Palva, et al., (1983) Gene 22:229-35; Mosbach, et al., (1983) Nature 302:543-5). The pGEX-4T-1 plasmid vector from Pharmacia is the preferred £. coli expression vector for the present disclosure. Expression in Eukaryotes
A variety of eukaryotic expression systems such as yeast, insect cell lines, plant and mammalian cells, are known to those of skill in the art. As explained briefly below, the present disclosure can be expressed in these eukaryotic systems. In some embodiments, transformed/transfected plant cells, as discussed infra, are employed as expression systems for production of the proteins of the instant disclosure.
Synthesis of heterologous proteins in yeast is well known. Sherman, et al., (1982) Methods in Yeast Genetics, Cold Spring Harbor Laboratory is a well recognized work describing the various methods available to produce the protein in yeast. Two widely utilized yeasts for production of eukaryotic proteins are Saccharomyces cerevisiae and Pichia pastoris. Vectors, strains and protocols for expression in Saccharomyces and Pichia are known in the art and available from commercial suppliers (e.g., Invitrogen). Suitable vectors usually have expression control sequences, such as promoters, including 3-phosphoglycerate kinase or alcohol oxidase and an origin of replication, termination sequences and the like as desired.
A protein of the present disclosure, once expressed, can be isolated from yeast by lysing the cells and applying standard protein isolation techniques to the lysates or the pellets. The monitoring of the purification process can be accomplished by using Western blot techniques or radioimmunoassay of other standard immunoassay techniques.
The sequences encoding proteins of the present disclosure can also be ligated to various expression vectors for use in transfecting cell cultures of, for instance, mammalian, insect or plant origin. Mammalian cell systems often will be in the form of monolayers of cells although mammalian cell suspensions may also be used. A number of suitable host cell lines capable of expressing intact proteins have been developed in the art, and include the HEK293, BHK21 and CHO cell lines. Expression vectors for these cells can include expression control sequences, such as an origin of replication, a promoter (e.g., the CMV promoter, a HSV tk promoter or pgk (phosphoglycerate kinase) promoter), an enhancer (Queen, et al., (1986) Immunol. Rev. 89:49) and necessary processing information sites, such as ribosome binding sites, RNA splice sites, polyadenylation sites (e.g., an SV40 large T Ag poly A addition site) and transcriptional terminator sequences. Other animal cells useful for production of proteins of the present disclosure are available, for instance, from the American Type Culture Collection Catalogue of Cell Lines and Hybridomas (7th ed., 1992).
Appropriate vectors for expressing proteins of the present disclosure in insect cells are usually derived from the SF9 baculovirus. Suitable insect cell lines include mosquito larvae, silkworm, armyworm, moth and Drosophila cell lines such as a Schneider cell line (see, e.g., Schneider, (1987) J. Embryol. Exp. Morphol. 27:353-65).
As with yeast, when higher animal or plant host cells are employed, polyadenlyation or transcription terminator sequences are typically incorporated into the vector. An example of a terminator sequence is the polyadenylation sequence from the bovine growth hormone gene. Sequences for accurate splicing of the transcript may also be included. An example of a splicing sequence is the VP1 intron from SV40 (Sprague, et al. , (1983) J. Virol. 45:773-81 ). Additionally, gene sequences to control replication in the host cell may be incorporated into the vector such as those found in bovine papilloma virus type-vectors (Saveria-Campo, "Bovine Papilloma Virus DNA a Eukaryotic Cloning Vector," in DNA Cloning: A Practical Approach, vol. II, Glover, ed., IRL Press, Arlington, VA, pp. 213-38 (1985)).
In addition, the gene of interest placed in the appropriate plant expression vector can be used to transform plant cells. The polypeptide can then be isolated from plant callus or the transformed cells can be used to regenerate transgenic plants. Such transgenic plants can be harvested, and the appropriate tissues (seed or leaves, for example) can be subjected to large scale protein extraction and purification techniques.
Plant Transformation Methods
Numerous methods for introducing heterologous genes into plants are known and can be used to insert a polynucleotide into a plant host, including biological and physical plant transformation protocols. See, e.g., Miki et al., "Procedure for Introducing Foreign DNA into Plants," in Methods in Plant Molecular Biology and Biotechnology, Glick and Thompson, eds., CRC Press, Inc., Boca Raton, pp. 67-88 (1993). The methods chosen vary with the host plant and include chemical transfection methods such as calcium phosphate, microorganism-mediated gene transfer such as Agrobacterium (Horsch, et al., (1985) Science 227:1229-31 ), electroporation, micro-injection and biolistic bombardment.
Expression cassettes and vectors and in vitro culture methods for plant cell or tissue transformation and regeneration of plants are known and available. See, e.g., Gruber, et al., "Vectors for Plant Transformation," in Methods in Plant Molecular Biology and Biotechnology, supra, pp. 89-1 19.
The isolated polynucleotides or polypeptides may be introduced into the plant by one or more techniques typically used for direct delivery into cells. Such protocols may vary depending on the type of organism, cell, plant or plant cell, i.e., monocot or dicot, targeted for gene modification. Suitable methods of transforming plant cells include microinjection (Crossway, et al., (1986) Biotechniques 4:320-334 and US Patent Number 6,300,543), electroporation (Riggs, et al., (1986) Proc. Natl. Acad. Sci. USA 83:5602- 5606, direct gene transfer (Paszkowski et al., (1984) EMBO J. 3:2717-2722) and ballistic particle acceleration (see, for example, Sanford, et al., US Patent Number 4,945,050; WO 1991/10725 and McCabe, et al., (1988) Biotechnology 6:923-926). Also see, Tomes, et al. , "Direct DNA Transfer into Intact Plant Cells Via Microprojectile Bombardment", pp. 197-213 in Plant Cell, Tissue and Organ Culture, Fundamental Methods, eds. Gamborg and Phillips. Springer-Verlag Berlin Heidelberg New York, 1995; US Patent Number 5,736,369 (meristem); Weissinger, et al., (1988) Ann. Rev. Genet. 22:421 -477; Sanford, et al. , (1987) Particulate Science and Technology 5:27-37 (onion); Christou, et al., (1988) Plant Physiol. 87:671 -674 (soybean); Datta, et al., (1990) Biotechnology 8:736-740 (rice); Klein, et al., (1988) Proc. Natl. Acad. Sci. USA 85:4305-4309 (maize); Klein, et al., (1988) Biotechnology 6:559-563 (maize); WO 91/10725 (maize); Klein, et al., (1988) Plant Physiol. 91 :440-444 (maize); Fromm, et al., (1990) Biotechnology 8:833-839 and Gordon- Kamm, et al., (1990) Plant Cell 2:603-618 (maize); Hooydaas-Van Slogteren and Hooykaas, (1984) Nature (London) 31 1 :763-764; Bytebierm, et al. , (1987) Proc. Natl. Acad. Sci. USA 84:5345-5349 (Liliaceae); De Wet, et al. , (1985) In The Experimental Manipulation of Ovule Tissues, ed. G.P. Chapman, et al., pp. 197-209. Longman, NY (pollen); Kaeppler, et al., (1990) Plant Cell Reports 9:415-418 and Kaeppler, et al., (1992) Theor. Appl. Genet. 84:560-566 (whisker-mediated transformation); US Patent Number 5,693,512 (sonication); D'Halluin, et al. , (1992) Plant Cell 4:1495-1505 (electroporation); Li, et al., (1993) Plant Cell Reports 12:250-255 and Christou and Ford, (1995) Annals of Botany 75:407-413 (rice); Osjoda, et al., (1996) Nature Biotech. 14:745-750; Agrobacterium mediated maize transformation (US Patent Number 5,981 ,840); silicon carbide whisker methods (Frame, et al. , (1994) Plant J. 6:941-948); laser methods (Guo, et al., (1995) Physiologia Plantarum 93:19-24); sonication methods (Bao, et al., (1997) Ultrasound in Medicine & Biology 23:953-959; Finer and Finer, (2000) Lett Appl Microbiol. 30:406-10; Amoah, et al., (2001 ) J Exp Bot 52:1 135-42); polyethylene glycol methods (Krens, et al., (1982) Nature 296:72-77); protoplasts of monocot and dicot cells can be transformed using electroporation (Fromm, et al., (1985) Proc. Natl. Acad. Sci. USA 82:5824-5828) and microinjection (Crossway, et al., (1986) Mol. Gen. Genet. 202:179- 185), all of which are herein incorporated by reference.
Figure imgf000045_0001
Transformation
The most widely utilized method for introducing an expression vector into plants is based on the natural transformation system of Agrobacterium. A. tumefaciens and A. rhizogenes are plant pathogenic soil bacteria which genetically transform plant cells. The Ti and Ri plasmids of A. tumefaciens and A. rhizogenes, respectively, carry genes responsible for genetic transformation of plants. See, e.g., Kado, (1991 ) Crit. Rev. Plant Sci. 10:1 . Descriptions of the Agrobacterium vector systems and methods for
Figure imgf000045_0002
gene transfer are provided in Gruber, et al., supra; Miki, et al., supra and Moloney, et al., (1989) Plant Cell Reports 8:238.
Similarly, the gene can be inserted into the T-DNA region of a Ti or Ri plasmid derived from A. tumefaciens or A. rhizogenes, respectively. Thus, expression cassettes can be constructed as above, using these plasmids. Many control sequences are known which when coupled to a heterologous coding sequence and transformed into a host organism show fidelity in gene expression with respect to tissue/organ specificity of the original coding sequence. See, e.g., Benfey and Chua, (1989) Science 244:174-81. Particularly suitable control sequences for use in these plasmids are promoters for constitutive or tissue-preferred expression of the gene in the various target plants. Other useful control sequences include a promoter and terminator from the nopaline synthase gene (NOS). The NOS promoter and terminator are present in the plasmid pARC2, available from the American Type Culture Collection and designated ATCC 67238. If such a system is used, the virulence (vir) gene from either the Ti or Ri plasmid must also be present, either along with the T-DNA portion, or via a binary system where the vir gene is present on a separate vector. Such systems, vectors for use therein, and methods of transforming plant cells are described in US Patent Number 4,658,082; US Patent Application Serial Number 913,914, filed October 1 , 1986, as referenced in US Patent Number 5,262,306, issued November 16, 1993 and Simpson, et al., (1986) Plant Mol. Biol. 6:403-15 (also referenced in the '306 patent), all incorporated by reference in their entirety.
Once constructed, these plasmids can be placed into A. rhizogenes or A. tumefaciens and these vectors used to transform cells of plant species which are ordinarily susceptible to Fusarium or Alternaria infection. Several other transgenic plants are also contemplated by the present disclosure including but not limited to soybean, corn, sorghum, alfalfa, rice, clover, cabbage, banana, coffee, celery, tobacco, cowpea, cotton, melon and pepper. The selection of either A. tumefaciens or A. rhizogenes will depend on the plant being transformed thereby. In general A. tumefaciens is the preferred organism for transformation. Most dicotyledonous plants, some gymnosperms and a few monocotyledonous plants (e.g., certain members of the Liliales and Arales) are susceptible to infection with A. tumefaciens. A. rhizogenes also has a wide host range, embracing most dicots and some gymnosperms, which includes members of the Leguminosae, Compositae, and Chenopodiaceae. Monocot plants can also be transformed. EP Patent Application Number 604 662 A1 discloses a method for transforming monocots using Agrobacterium. EP Patent Application Number 672 752 A1 discloses a method for transforming monocots with Agrobacterium using the scutellum of immature embryos. Ishida, et al., discuss a method for transforming maize by exposing immature embryos to A. tumefaciens (Nature Biotechnology 14:745-50 (1996)).
Once transformed, these cells can be used to regenerate transgenic plants. For example, whole plants can be infected with these vectors by wounding the plant and then introducing the vector into the wound site. Any part of the plant can be wounded, including leaves, stems and roots. Alternatively, plant tissue in the form of an explant, such as cotyledonary tissue or leaf disks, can be inoculated with these vectors, and cultured under conditions which promote plant regeneration. Examples of such methods for regenerating plant tissue are disclosed in Shahin, (1985) Theor. Appl. Genet. 69:235- 40; US Patent Number 4,658,082; Simpson, et al., supra and US Patent Application Serial Numbers 913,913 and 913,914, both filed October 1 , 1986, as referenced in US Patent Number 5,262,306, issued November 16, 1993, the entire disclosures therein incorporated herein by reference.
Direct Gene Transfer
Despite the fact that the host range for
Figure imgf000046_0001
transformation is broad, some major cereal crop species and gymnosperms have generally been recalcitrant to this mode of gene transfer, even though some success has recently been achieved in rice (Hiei, et al., (1994) The Plant Journal 6:271 -82). Several methods of plant transformation, collectively referred to as direct gene transfer, have been developed as an alternative to ^grobacfer/i/m-mediated transformation.
A generally applicable method of plant transformation is microprojectile-mediated transformation, where DNA is carried on the surface of microprojectiles measuring about 1 to 4 μηη. The expression vector is introduced into plant tissues with a biolistic device that accelerates the microprojectiles to speeds of 300 to 600 m/s which is sufficient to penetrate the plant cell walls and membranes (Sanford, et al., (1987) Part. Sci. Technol. 5:27; Sanford, (1988) Trends Biotech 6:299; Sanford, (1990) Physiol. Plant 79:206 and Klein, et al., (1992) Biotechnology 10:268).
Another method for physical delivery of DNA to plants is sonication of target cells as described in Zang, et al., (1991 ) BioTechnology 9:996. Alternatively, liposome or spheroplast fusions have been used to introduce expression vectors into plants. See, e.g., Deshayes, et al., (1985) EMBO J. 4:2731 and Christou, et al., (1987) Proc. Natl. Acad. Sci. USA 84:3962. Direct uptake of DNA into protoplasts using CaCI2 precipitation, polyvinyl alcohol, or poly-L-ornithine has also been reported. See, e.g., Hain, et al., (1985) Mol. Gen. Genet. 199:161 and Draper, et al., (1982) Plant Cell Physiol. 23:451 .
Electroporation of protoplasts and whole cells and tissues has also been described. See, e.g., Donn, et al., (1990) Abstracts of the Vllth Int'l. Congress on Plant Cell and Tissue Culture IAPTC, A2-38, p. 53; D'Halluin, et al., (1992) Plant Cell 4: 1495- 505 and Spencer, et al., (1994) Plant Mol. Biol. 24:51 -61. Reducing the Activity and/or Level of a Polypeptide
Methods are provided to reduce or eliminate the activity of a polypeptide of the disclosure by transforming a plant cell with an expression cassette that expresses a polynucleotide that inhibits the expression of the polypeptide. The polynucleotide may inhibit the expression of the polypeptide directly, by preventing transcription or translation of the messenger RNA, or indirectly, by encoding a polypeptide that inhibits the transcription or translation of a gene encoding polypeptide. Methods for inhibiting or eliminating the expression of a gene in a plant are well known in the art and any such method may be used in the present disclosure to inhibit the expression of polypeptide.
In accordance with the present disclosure, the expression of a polypeptide may be inhibited so that the protein level of the polypeptide is, for example, less than 70% of the protein level of the same polypeptide in a plant that has not been genetically modified or mutagenized to inhibit the expression of that polypeptide. In particular embodiments of the disclosure, the protein level of the polypeptide in a modified plant according to the disclosure is less than 60%, less than 50%, less than 40%, less than 30%, less than 20%, less than 10%, less than 5% or less than 2% of the protein level of the same polypeptide in a plant that is not a mutant or that has not been genetically modified to inhibit the expression of that polypeptide. The expression level of the polypeptide may be measured directly, for example, by assaying for the level of polypeptide expressed in the plant cell or plant, or indirectly, for example, by measuring the nitrogen uptake activity of the polypeptide in the plant cell or plant or by measuring the phenotypic changes in the plant. Methods for performing such assays are described elsewhere herein.
In other embodiments of the disclosure, the activity of the polypeptide is reduced or eliminated by transforming a plant cell with an expression cassette comprising a polynucleotide encoding a polypeptide that inhibits the activity of a polypeptide. The activity of a polypeptide is inhibited according to the present disclosure if the activity of the polypeptide is, for example, less than 70% of the activity of the same polypeptide in a plant that has not been modified to inhibit the activity of that polypeptide. In particular embodiments of the disclosure, the activity of the polypeptide in a modified plant according to the disclosure is less than 60%, less than 50%, less than 40%, less than 30%, less than 20%, less than 10% or less than 5% of the activity of the same polypeptide in a plant that that has not been modified to inhibit the expression of that polypeptide. The activity of a polypeptide is "eliminated" according to the disclosure when it is not detectable by the assay methods described elsewhere herein. Methods of determining the alteration of activity of a polypeptide are described elsewhere herein.
In other embodiments, the activity of a polypeptide may be reduced or eliminated by disrupting the gene encoding the polypeptide. The disclosure encompasses mutagenized plants that carry mutations in genes, where the mutations reduce expression of the gene or inhibit the activity of the encoded polypeptide.
Thus, many methods may be used to reduce or eliminate the activity of a polypeptide. In addition, more than one method may be used to reduce the activity of a single polypeptide.
1. Polynucleotide-Based Methods:
In some embodiments of the present disclosure, a plant is transformed with an expression cassette that is capable of expressing a polynucleotide that inhibits the expression of a polypeptide of the disclosure. The term "expression" as used herein refers to the biosynthesis of a gene product, including the transcription and/or translation of said gene product. For example, for the purposes of the present disclosure, an expression cassette capable of expressing a polynucleotide that inhibits the expression of at least one polypeptide is an expression cassette capable of producing an RNA molecule that inhibits the transcription and/or translation of at least one polypeptide of the disclosure. The "expression" or "production" of a protein or polypeptide from a DNA molecule refers to the transcription and translation of the coding sequence to produce the protein or polypeptide, while the "expression" or "production" of a protein or polypeptide from an RNA molecule refers to the translation of the RNA coding sequence to produce the protein or polypeptide.
Examples of polynucleotides that inhibit the expression of a polypeptide are given below.
/'. Sense Suppression/Cosuppression
In some embodiments of the disclosure, inhibition of the expression of a polypeptide may be obtained by sense suppression or cosuppression. For cosuppression, an expression cassette is designed to express an RNA molecule corresponding to all or part of a messenger RNA encoding a polypeptide in the "sense" orientation. Over expression of the RNA molecule can result in reduced expression of the native gene. Accordingly, multiple plant lines transformed with the cosuppression expression cassette are screened to identify those that show the desired degree of inhibition of polypeptide expression.
The polynucleotide used for cosuppression may correspond to all or part of the sequence encoding the polypeptide, all or part of the 5' and/or 3' untranslated region of a polypeptide transcript or all or part of both the coding sequence and the untranslated regions of a transcript encoding a polypeptide. In some embodiments where the polynucleotide comprises all or part of the coding region for the polypeptide, the expression cassette is designed to eliminate the start codon of the polynucleotide so that no protein product will be translated.
Cosuppression may be used to inhibit the expression of plant genes to produce plants having undetectable protein levels for the proteins encoded by these genes. See, for example, Broin, et al., (2002) Plant Cell 14:1417-1432. Cosuppression may also be used to inhibit the expression of multiple proteins in the same plant. See, for example, US Patent Number 5,942,657. Methods for using cosuppression to inhibit the expression of endogenous genes in plants are described in Flavell, et al., (1994) Proc. Natl. Acad. Sci. USA 91 :3490-3496; Jorgensen, et al., (1996) Plant Mol. Biol. 31 :957-973; Johansen and Carrington, (2001 ) Plant Physiol. 126:930-938; Broin, et al., (2002) Plant Cell 14:1417-1432; Stoutjesdijk, et al., (2002) Plant Physiol. 129:1723-1731 ; Yu, et al., (2003) Phytochemistry 63:753-763 and US Patent Numbers 5,034,323, 5,283,184 and 5,942,657, each of which is herein incorporated by reference. The efficiency of cosuppression may be increased by including a poly-dT region in the expression cassette at a position 3' to the sense sequence and 5' of the polyadenylation signal. See, US Patent Application Publication Number 2002/0048814, herein incorporated by reference. Typically, such a nucleotide sequence has substantial sequence identity to the sequence of the transcript of the endogenous gene, optimally greater than about 65% sequence identity, more optimally greater than about 85% sequence identity, most optimally greater than about 95% sequence identity. See US Patent Numbers 5,283,184 and 5,034,323, herein incorporated by reference.
/'/'. Antisense Suppression
In some embodiments of the disclosure, inhibition of the expression of the polypeptide may be obtained by antisense suppression. For antisense suppression, the expression cassette is designed to express an RNA molecule complementary to all or part of a messenger RNA encoding the polypeptide. Over expression of the antisense RNA molecule can result in reduced expression of the target gene. Accordingly, multiple plant lines transformed with the antisense suppression expression cassette are screened to identify those that show the desired degree of inhibition of polypeptide expression.
The polynucleotide for use in antisense suppression may correspond to all or part of the complement of the sequence encoding the polypeptide, all or part of the complement of the 5' and/or 3' untranslated region of the target transcript or all or part of the complement of both the coding sequence and the untranslated regions of a transcript encoding the polypeptide. In addition, the antisense polynucleotide may be fully complementary (i.e., 100% identical to the complement of the target sequence) or partially complementary (i.e., less than 100% identical to the complement of the target sequence) to the target sequence. Antisense suppression may be used to inhibit the expression of multiple proteins in the same plant. See, for example, US Patent Number 5,942,657. Furthermore, portions of the antisense nucleotides may be used to disrupt the expression of the target gene. Generally, sequences of at least 50 nucleotides, 100 nucleotides, 200 nucleotides, 300, 400, 450, 500, 550 or greater may be used. Methods for using antisense suppression to inhibit the expression of endogenous genes in plants are described, for example, in Liu, et al., (2002) Plant Physiol. 129:1732-1743 and US Patent Numbers 5,759,829 and 5,942,657, each of which is herein incorporated by reference. Efficiency of antisense suppression may be increased by including a poly-dT region in the expression cassette at a position 3' to the antisense sequence and 5' of the polyadenylation signal. See, US Patent Application Publication Number 2002/0048814, herein incorporated by reference. /'/'/'. Double-Stranded RNA Interference
In some embodiments of the disclosure, inhibition of the expression of a polypeptide may be obtained by double-stranded RNA (dsRNA) interference. For dsRNA interference, a sense RNA molecule like that described above for cosuppression and an antisense RNA molecule that is fully or partially complementary to the sense RNA molecule are expressed in the same cell, resulting in inhibition of the expression of the corresponding endogenous messenger RNA.
Expression of the sense and antisense molecules can be accomplished by designing the expression cassette to comprise both a sense sequence and an antisense sequence. Alternatively, separate expression cassettes may be used for the sense and antisense sequences. Multiple plant lines transformed with the dsRNA interference expression cassette or expression cassettes are then screened to identify plant lines that show the desired degree of inhibition of polypeptide expression. Methods for using dsRNA interference to inhibit the expression of endogenous plant genes are described in Waterhouse, et al., (1998) Proc. Natl. Acad. Sci. USA 95:13959-13964, Liu, et al., (2002) Plant Physiol. 129:1732-1743 and WO 1999/49029, WO 1999/53050, WO 1999/61631 and WO 2000/49035, each of which is herein incorporated by reference. iv. Hairpin RNA Interference and Intron-Containing Hairpin RNA Interference
In some embodiments of the disclosure, inhibition of the expression of a polypeptide may be obtained by hairpin RNA (hpRNA) interference or intron-containing hairpin RNA (ihpRNA) interference. These methods are highly efficient at inhibiting the expression of endogenous genes. See, Waterhouse and Helliwell, (2003) Nat. Rev. Genet. 4:29-38 and the references cited therein.
For hpRNA interference, the expression cassette is designed to express an RNA molecule that hybridizes with itself to form a hairpin structure that comprises a single- stranded loop region and a base-paired stem. The base-paired stem region comprises a sense sequence corresponding to all or part of the endogenous messenger RNA encoded by the gene whose expression is to be inhibited, and an antisense sequence that is fully or partially complementary to the sense sequence. Alternatively, the base-paired stem region may correspond to a portion of a promoter sequence controlling expression of the gene whose expression is to be inhibited. Thus, the base-paired stem region of the molecule generally determines the specificity of the RNA interference. hpRNA molecules are highly efficient at inhibiting the expression of endogenous genes and the RNA interference they induce is inherited by subsequent generations of plants. See, for example, Chuang and Meyerowitz, (2000) Proc. Natl. Acad. Sci. USA 97:4985-4990; Stoutjesdijk, et al., (2002) Plant Physiol. 129:1723-1731 and Waterhouse and Helliwell, (2003) Nat. Rev. Genet. 4:29-38. Methods for using hpRNA interference to inhibit or silence the expression of genes are described, for example, in Chuang and Meyerowitz,
(2000) Proc. Natl. Acad. Sci. USA 97:4985-4990; Stoutjesdijk, et al., (2002) Plant Physiol. 129:1723-1731 ; Waterhouse and Helliwell, (2003) Nat. Rev. Genet. 4:29-38; Pandolfini et al. , BMC Biotechnology 3:7 and US Patent Application Publication Number 2003/0175965, each of which is herein incorporated by reference. A transient assay for the efficiency of hpRNA constructs to silence gene expression in vivo has been described by Panstruga, et al., (2003) Mol. Biol. Rep. 30:135-140, herein incorporated by reference.
For ihpRNA, the interfering molecules have the same general structure as for hpRNA, but the RNA molecule additionally comprises an intron that is capable of being spliced in the cell in which the ihpRNA is expressed. The use of an intron minimizes the size of the loop in the hairpin RNA molecule following splicing, and this increases the efficiency of interference. See, for example, Smith, et al., (2000) Nature 407:319-320. In fact, Smith, et al. , show 100% suppression of endogenous gene expression using ihpRNA-mediated interference. Methods for using ihpRNA interference to inhibit the expression of endogenous plant genes are described, for example, in Smith, et al. , (2000) Nature 407:319-320; Wesley, et al., (2001 ) Plant J. 27:581-590; Wang and Waterhouse,
(2001 ) Curr. Opin. Plant Biol. 5:146-150; Waterhouse and Helliwell, (2003) Nat. Rev. Genet. 4:29-38; Helliwell and Waterhouse, (2003) Methods 30:289-295 and US Patent Application Publication Number 2003/0180945, each of which is herein incorporated by reference.
The expression cassette for hpRNA interference may also be designed such that the sense sequence and the antisense sequence do not correspond to an endogenous RNA. In this embodiment, the sense and antisense sequence flank a loop sequence that comprises a nucleotide sequence corresponding to all or part of the endogenous messenger RNA of the target gene. Thus, it is the loop region that determines the specificity of the RNA interference. See, for example, WO 2002/00904; Mette, et al., (2000) EMBO J 19:5194-5201 ; Matzke, et al., (2001 ) Curr. Opin. Genet. Devel. 11 :221 - 227; Scheid, et al., (2002) Proc. Natl. Acad. Sci., USA 99:13659-13662; Aufsaftz, et al.,
(2002) Proc. Nat'l. Acad. Sci. 99(4): 16499-16506; Sijen, et al., Curr. Biol. (2001 ) 11 :436- 440), herein incorporated by reference. v. Amplicon-Mediated Interference
Amplicon expression cassettes comprise a plant-virus-derived sequence that contains all or part of the target gene but generally not all of the genes of the native virus. The viral sequences present in the transcription product of the expression cassette allow the transcription product to direct its own replication. The transcripts produced by the amplicon may be either sense or antisense relative to the target sequence (i.e., the messenger RNA for the polypeptide). Methods of using amplicons to inhibit the expression of endogenous plant genes are described, for example, in Angell and Baulcombe, (1997) EMBO J. 16:3675-3684, Angell and Baulcombe, (1999) Plant J. 20:357-362 and US Patent Number 6,646,805, each of which is herein incorporated by reference. vi. Ribozymes
In some embodiments, the polynucleotide expressed by the expression cassette of the disclosure is catalytic RNA or has ribozyme activity specific for the messenger RNA of the polypeptide. Thus, the polynucleotide causes the degradation of the endogenous messenger RNA, resulting in reduced expression of the polypeptide. This method is described, for example, in US Patent Number 4,987,071 , herein incorporated by reference. vii. Small Interfering RNA or Micro RNA
In some embodiments of the disclosure, inhibition of the expression of a polypeptide may be obtained by RNA interference by expression of a polynucleotide encoding a micro RNA (miRNA). miRNAs are regulatory agents consisting of about 22 ribonucleotides. miRNA are highly efficient at inhibiting the expression of endogenous genes. See, for example Javier, et al., (2003) Nature 425:257-263, herein incorporated by reference.
For miRNA interference, the expression cassette is designed to express an RNA molecule that is modeled on an endogenous miRNA gene. For example, the miRNA gene encodes an RNA that forms a hairpin structure containing a 22-nucleotide sequence that is complementary to an endogenous gene target sequence. For suppression of NUE expression, the 22-nucleotide sequence is selected from a NUE transcript sequence and contains 22 nucleotides of said NUE sequence in sense orientation and 21 nucleotides of a corresponding antisense sequence that is complementary to the sense sequence. A fertility gene, whether endogenous or exogenous, may be an miRNA target. miRNA molecules are highly efficient at inhibiting the expression of endogenous genes, and the RNA interference they induce is inherited by subsequent generations of plants.
2. Polypeptide-Based Inhibition of Gene Expression
In one embodiment, the polynucleotide encodes a zinc finger protein that binds to a gene encoding a polypeptide, resulting in reduced expression of the gene. In particular embodiments, the zinc finger protein binds to a regulatory region of a gene. In other embodiments, the zinc finger protein binds to a messenger RNA encoding a polypeptide and prevents its translation. Methods of selecting sites for targeting by zinc finger proteins have been described, for example, in US Patent Number 6,453,242, and methods for using zinc finger proteins to inhibit the expression of genes in plants are described, for example, in US Patent Application Publication Number 2003/0037355, each of which is herein incorporated by reference. 3. Polypeptide-Based Inhibition of Protein Activity
In some embodiments of the disclosure, the polynucleotide encodes an antibody that binds to at least one polypeptide and reduces the activity of the polypeptide. In another embodiment, the binding of the antibody results in increased turnover of the antibody-polypeptide complex by cellular quality control mechanisms. The expression of antibodies in plant cells and the inhibition of molecular pathways by expression and binding of antibodies to proteins in plant cells are well known in the art. See, for example, Conrad and Sonnewald, (2003) Nature Biotech. 21 :35-36, incorporated herein by reference. 4. Gene Disruption
In some embodiments of the present disclosure, the activity of a polypeptide is reduced or eliminated by disrupting the gene encoding the polypeptide. The gene encoding the polypeptide may be disrupted by any method known in the art. For example, in one embodiment, the gene is disrupted by transposon tagging. In another embodiment, the gene is disrupted by mutagenizing plants using random or targeted mutagenesis and selecting for plants that have reduced nitrogen utilization activity.
/'. Transposon Tagging
In one embodiment of the disclosure, transposon tagging is used to reduce or eliminate the activity of one or more polypeptide. Transposon tagging comprises inserting a transposon within an endogenous gene to reduce or eliminate expression of the polypeptide.
In this embodiment, the expression of one or more polypeptides is reduced or eliminated by inserting a transposon within a regulatory region or coding region of the gene encoding the polypeptide. A transposon that is within an exon, intron, 5' or 3' untranslated sequence, a promoter or any other regulatory sequence of a gene may be used to reduce or eliminate the expression and/or activity of the encoded polypeptide.
Methods for the transposon tagging of specific genes in plants are well known in the art. See, for example, Maes, et al. , (1999) Trends Plant Sci. 4:90-96; Dharmapuri and Sonti, (1999) FEMS Microbiol. Lett. 179:53-59; Meissner, et al., (2000) Plant J. 22:265- 274; Phogat, et al., (2000) J. Biosci. 25:57-63; Walbot, (2000) Curr. Opin. Plant Biol. 2:103-107; Gai, et al., (2000) Nucleic Acids Res. 28:94-96; Fitzmaurice, et al., (1999) Genetics 153:1919-1928). In addition, the TUSC process for selecting Mu insertions in selected genes has been described in Bensen, et al. , (1995) Plant Cell 7:75-84; Mena, et al. , (1996) Science 274:1537-1540 and US Patent Number 5,962,764, each of which is herein incorporated by reference.
/'/'. Mutant Plants with Reduced Activity
Additional methods for decreasing or eliminating the expression of endogenous genes in plants are known in the art and can be similarly applied to the instant disclosure. These methods include other forms of mutagenesis, such as ethyl methanesulfonate- induced mutagenesis, deletion mutagenesis and fast neutron deletion mutagenesis used in a reverse genetics sense (with PCR) to identify plant lines in which the endogenous gene has been deleted. For examples of these methods see, Ohshima, et al., (1998) Virology 243:472-481 ; Okubara, et al., (1994) Genetics 137:867-874 and Quesada, et al., (2000) Genetics 154:421 -436, each of which is herein incorporated by reference. In addition, a fast and automatable method for screening for chemically induced mutations, TILLING (Targeting Induced Local Lesions In Genomes), using denaturing HPLC or selective endonuclease digestion of selected PCR products is also applicable to the instant disclosure. See, McCallum, et al. , (2000) Nat. Biotechnol. 18:455-457, herein incorporated by reference.
Mutations that impact gene expression or that interfere with the function of the encoded protein are well known in the art. Insertional mutations in gene exons usually result in null-mutants. Mutations in conserved residues are particularly effective in inhibiting the activity of the encoded protein. Conserved residues of plant polypeptides suitable for mutagenesis with the goal to eliminate activity have been described. Such mutants can be isolated according to well-known procedures and mutations in different loci can be stacked by genetic crossing. See, for example, Gruis, et al., (2002) Plant Cell 14:2863-2882.
In another embodiment of this disclosure, dominant mutants can be used to trigger
RNA silencing due to gene inversion and recombination of a duplicated gene locus. See, for example, Kusaba, et al. , (2003) Plant Cell 15:1455-1467.
The disclosure encompasses additional methods for reducing or eliminating the activity of one or more polypeptide. Examples of other methods for altering or mutating a genomic nucleotide sequence in a plant are known in the art and include, but are not limited to, the use of RNA:DNA vectors, RNA:DNA mutational vectors, RNA:DNA repair vectors, mixed-duplex oligonucleotides, self-complementary RNA:DNA oligonucleotides and recombinogenic oligonucleobases. Such vectors and methods of use are known in the art. See, for example, US Patent Numbers 5,565,350; 5,731 ,181 ; 5,756,325; 5,760,012; 5,795,972 and 5,871 ,984, each of which are herein incorporated by reference. See also, WO 1998/49350, WO 1999/07865, WO 1999/25821 and Beetham, et al., (1999) Proc. Natl. Acad. Sci. USA 96:8774-8778, each of which is herein incorporated by reference. /'/'/'. Modulating nitrogen utilization activity
In specific methods, the level and/or activity of a NUE regulator in a plant is decreased by increasing the level or activity of the polypeptide in the plant. The increased expression of a negative regulatory molecule may decrease the level of expression of downstream one or more genes responsible for an improved NUE phenotype.
Methods for increasing the level and/or activity of polypeptides in a plant are discussed elsewhere herein. Briefly, such methods comprise providing a polypeptide of the disclosure to a plant and thereby increasing the level and/or activity of the polypeptide. In other embodiments, a NUE nucleotide sequence encoding a polypeptide can be provided by introducing into the plant a polynucleotide comprising a NUE nucleotide sequence of the disclosure, expressing the NUE sequence, increasing the activity of the polypeptide and thereby decreasing the number of tissue cells in the plant or plant part. In other embodiments, the NUE nucleotide construct introduced into the plant is stably incorporated into the genome of the plant. In other methods, the growth of a plant tissue is increased by decreasing the level and/or activity of the polypeptide in the plant. Such methods are disclosed in detail elsewhere herein. In one such method, a NUE nucleotide sequence is introduced into the plant and expression of said NUE nucleotide sequence decreases the activity of the polypeptide and thereby increasing the tissue growth in the plant or plant part. In other embodiments, the NUE nucleotide construct introduced into the plant is stably incorporated into the genome of the plant.
As discussed above, one of skill will recognize the appropriate promoter to use to modulate the level/activity of a NUE in the plant. Exemplary promoters for this embodiment have been disclosed elsewhere herein.
In other embodiments, such plants have stably incorporated into their genome a nucleic acid molecule comprising a NUE nucleotide sequence of the disclosure operably linked to a promoter that drives expression in the plant cell. iv. Modulating Root Development
Methods for modulating root development in a plant are provided. By "modulating root development" is intended any alteration in the development of the plant root when compared to a control plant. Such alterations in root development include, but are not limited to, alterations in the growth rate of the primary root, the fresh root weight, the extent of lateral and adventitious root formation, the vasculature system, meristem development or radial expansion.
Methods for modulating root development in a plant are provided. The methods comprise modulating the level and/or activity of the polypeptide in the plant. In one method, a sequence of the disclosure is provided to the plant. In another method, the nucleotide sequence is provided by introducing into the plant a polynucleotide comprising a nucleotide sequence of the disclosure, expressing the sequence and thereby modifying root development. In still other methods, the nucleotide construct introduced into the plant is stably incorporated into the genome of the plant.
In other methods, root development is modulated by altering the level or activity of the polypeptide in the plant. A change in activity can result in at least one or more of the following alterations to root development, including, but not limited to, alterations in root biomass and length.
As used herein, "root growth" encompasses all aspects of growth of the different parts that make up the root system at different stages of its development in both monocotyledonous and dicotyledonous plants. It is to be understood that enhanced root growth can result from enhanced growth of one or more of its parts including the primary root, lateral roots, adventitious roots, etc.
Methods of measuring such developmental alterations in the root system are known in the art. See, for example, US Patent Application Publication Number 2003/0074698 and Werner, et al. , (2001 ) PNAS 18:10487-10492, both of which are herein incorporated by reference.
As discussed above, one of skill will recognize the appropriate promoter to use to modulate root development in the plant. Exemplary promoters for this embodiment include constitutive promoters and root-preferred promoters. Exemplary root-preferred promoters have been disclosed elsewhere herein.
Stimulating root growth and increasing root mass by decreasing the activity and/or level of the polypeptide also finds use in improving the standability of a plant. The term "resistance to lodging" or "standability" refers to the ability of a plant to fix itself to the soil. For plants with an erect or semi-erect growth habit, this term also refers to the ability to maintain an upright position under adverse environmental conditions. This trait relates to the size, depth and morphology of the root system. In addition, stimulating root growth and increasing root mass by altering the level and/or activity of the polypeptide finds use in promoting in vitro propagation of explants.
Furthermore, higher root biomass production has a direct effect on the yield and an indirect effect of production of compounds produced by root cells or transgenic root cells or cell cultures of said transgenic root cells. One example of an interesting compound produced in root cultures is shikonin, the yield of which can be advantageously enhanced by said methods.
Accordingly, the present disclosure further provides plants having modulated root development when compared to the root development of a control plant. In some embodiments, the plant of the disclosure has an increased level/activity of a polypeptide of the disclosure and has enhanced root growth and/or root biomass. In other embodiments, such plants have stably incorporated into their genome a nucleic acid molecule comprising a nucleotide sequence of the disclosure operably linked to a promoter that drives expression in the plant cell. v. Modulating Shoot and Leaf Development
Methods are also provided for modulating shoot and leaf development in a plant. By "modulating shoot and/or leaf development" is intended any alteration in the development of the plant shoot and/or leaf. Such alterations in shoot and/or leaf development include, but are not limited to, alterations in shoot meristem development, in leaf number, leaf size, leaf and stem vasculature, internode length and leaf senescence. As used herein, "leaf development" and "shoot development" encompasses all aspects of growth of the different parts that make up the leaf system and the shoot system, respectively, at different stages of their development, both in monocotyledonous and dicotyledonous plants. Methods for measuring such developmental alterations in the shoot and leaf system are known in the art. See, for example, Werner, et al., (2001 ) PNAS 98:10487-10492 and US Patent Application Publication Number 2003/0074698, each of which is herein incorporated by reference.
The method for modulating shoot and/or leaf development in a plant comprises modulating the activity and/or level of a polypeptide of the disclosure. In one embodiment, a sequence of the disclosure is provided. In other embodiments, the nucleotide sequence can be provided by introducing into the plant a polynucleotide comprising a nucleotide sequence of the disclosure, expressing the sequence and thereby modifying shoot and/or leaf development. In other embodiments, the nucleotide construct introduced into the plant is stably incorporated into the genome of the plant.
In specific embodiments, shoot or leaf development is modulated by altering the level and/or activity of the polypeptide in the plant. A change in activity can result in at least one or more of the following alterations in shoot and/or leaf development, including, but not limited to, changes in leaf number, altered leaf surface, altered vasculature, internodes and plant growth and alterations in leaf senescence when compared to a control plant.
As discussed above, one of skill will recognize the appropriate promoter to use to modulate shoot and leaf development of the plant. Exemplary promoters for this embodiment include constitutive promoters, shoot-preferred promoters, shoot meristem- preferred promoters and leaf-preferred promoters. Exemplary promoters have been disclosed elsewhere herein.
Increasing activity and/or level of a polypeptide of the disclosure in a plant may result in altered internodes and growth. Thus, the methods of the disclosure find use in producing modified plants. In addition, as discussed above, activity in the plant modulates both root and shoot growth. Thus, the present disclosure further provides methods for altering the root/shoot ratio. Shoot or leaf development can further be modulated by altering the level and/or activity of the polypeptide in the plant.
Accordingly, the present disclosure further provides plants having modulated shoot and/or leaf development when compared to a control plant. In some embodiments, the plant of the disclosure has an increased level/activity of a polypeptide of the disclosure. In other embodiments, a plant of the disclosure has a decreased level/activity of a polypeptide of the disclosure. vi. Modulating Reproductive Tissue Development
Methods for modulating reproductive tissue development are provided. In one embodiment, methods are provided to modulate floral development in a plant. By "modulating floral development" is intended any alteration in a structure of a plant's reproductive tissue as compared to a control plant in which the activity or level of the polypeptide has not been modulated. "Modulating floral development" further includes any alteration in the timing of the development of a plant's reproductive tissue (e.g., a delayed or an accelerated timing of floral development) when compared to a control plant in which the activity or level of the polypeptide has not been modulated. Changes in timing of reproductive development may result in altered synchronization of development of male and female reproductive tissues. Macroscopic alterations may include changes in size, shape, number or location of reproductive organs, the developmental time period that these structures form or the ability to maintain or proceed through the flowering process in times of environmental stress. Microscopic alterations may include changes to the types or shapes of cells that make up the reproductive organs.
The method for modulating floral development in a plant comprises modulating activity in a plant. In one method, a sequence of the disclosure is provided. A nucleotide sequence can be provided by introducing into the plant a polynucleotide comprising a nucleotide sequence of the disclosure, expressing the sequence and thereby modifying floral development. In other embodiments, the nucleotide construct introduced into the plant is stably incorporated into the genome of the plant.
In specific methods, floral development is modulated by increasing the level or activity of the polypeptide in the plant. A change in activity can result in at least one or more of the following alterations in floral development, including, but not limited to, altered flowering, changed number of flowers, modified male sterility and altered seed set, when compared to a control plant. Inducing delayed flowering or inhibiting flowering can be used to enhance yield in forage crops such as alfalfa. Methods for measuring such developmental alterations in floral development are known in the art. See, for example, Mouradov, et al., (2002) The Plant Cell S1 1 1-S130, herein incorporated by reference.
As discussed above, one of skill will recognize the appropriate promoter to use to modulate floral development of the plant. Exemplary promoters for this embodiment include constitutive promoters, inducible promoters, shoot-preferred promoters and inflorescence-preferred promoters.
In other methods, floral development is modulated by altering the level and/or activity of a sequence of the disclosure. Such methods can comprise introducing a nucleotide sequence into the plant and changing the activity of the polypeptide. In other methods, the nucleotide construct introduced into the plant is stably incorporated into the genome of the plant. Altering expression of the sequence of the disclosure can modulate floral development during periods of stress. Such methods are described elsewhere herein. Accordingly, the present disclosure further provides plants having modulated floral development when compared to the floral development of a control plant. Compositions include plants having an altered level/activity of the polypeptide of the disclosure and having an altered floral development. Compositions also include plants having a modified level/activity of the polypeptide of the disclosure wherein the plant maintains or proceeds through the flowering process in times of stress.
Methods are also provided for the use of the sequences of the disclosure to increase seed size and/or weight. The method comprises increasing the activity of the sequences in a plant or plant part, such as the seed. An increase in seed size and/or weight comprises an increased size or weight of the seed and/or an increase in the size or weight of one or more seed part including, for example, the embryo, endosperm, seed coat, aleurone or cotyledon.
As discussed above, one of skill will recognize the appropriate promoter to use to increase seed size and/or seed weight. Exemplary promoters of this embodiment include constitutive promoters, inducible promoters, seed-preferred promoters, embryo-preferred promoters and endosperm-preferred promoters.
A method for altering seed size and/or seed weight in a plant may increasing activity in the plant. In one embodiment, the NUE nucleotide sequence can be provided by introducing into the plant a polynucleotide comprising a NUE nucleotide sequence of the disclosure, expressing the NUE sequence and thereby decreasing seed weight and/or size. In other embodiments, the NUE nucleotide construct introduced into the plant is stably incorporated into the genome of the plant.
It is further recognized that increasing seed size and/or weight can also be accompanied by an increase in the speed of growth of seedlings or an increase in early vigor. As used herein, the term "early vigor" refers to the ability of a plant to grow rapidly during early development, and relates to the successful establishment, after germination, of a well-developed root system and a well-developed photosynthetic apparatus. In addition, an increase in seed size and/or weight can also result in an increase in plant yield when compared to a control.
Accordingly, the present disclosure further provides plants having an increased seed weight and/or seed size when compared to a control plant. In other embodiments, plants having an increased vigor and plant yield are also provided. In some embodiments, the plant of the disclosure has a modified level/activity of the polypeptide of the disclosure and has an increased seed weight and/or seed size. In other embodiments, such plants have stably incorporated into their genome a nucleic acid molecule comprising a NUE nucleotide sequence of the disclosure operably linked to a promoter that drives expression in the plant cell. vii. Method of Use for polynucleotide, expression cassettes, and additional polynucleotides
The nucleotides, expression cassettes and methods disclosed herein are useful in regulating expression of any heterologous nucleotide sequence in a host plant in order to vary the phenotype of a plant. Various changes in phenotype are of interest including modifying the fatty acid composition in a plant, altering the amino acid content of a plant, altering a plant's pathogen defense mechanism and the like. These results can be achieved by providing expression of heterologous products or increased expression of endogenous products in plants. Alternatively, the results can be achieved by providing for a reduction of expression of one or more endogenous products, particularly enzymes or cofactors in the plant. These changes result in a change in phenotype of the transformed plant.
Genes of interest are reflective of the commercial markets and interests of those involved in the development of the crop. Crops and markets of interest change, and as developing nations open up world markets, new crops and technologies will emerge also. In addition, as our understanding of agronomic traits and characteristics such as yield and heterosis increases, the choice of genes for transformation will change accordingly. General categories of genes of interest include, for example, those genes involved in information, such as zinc fingers, those involved in communication, such as kinases, and those involved in housekeeping, such as heat shock proteins. More specific categories of transgenes, for example, include genes encoding important traits for agronomics, insect resistance, disease resistance, herbicide resistance, sterility, grain characteristics and commercial products. Genes of interest include, generally, those involved in oil, starch, carbohydrate or nutrient metabolism as well as those affecting kernel size, sucrose loading and the like.
In certain embodiments the nucleic acid sequences of the present disclosure can be used in combination ("stacked") with other polynucleotide sequences of interest in order to create plants with a desired phenotype. The combinations generated can include multiple copies of any one or more of the polynucleotides of interest. The promoter which is operably linked to a polynucleotide sequence of interest can be any promoter that is active in plant cells. In some embodiments it is particularly advantageous to use a promoter that is active (or can be activated) in reproductive tissues of a plant (e.g., stamens or ovaries). As such, the promoter can be, for example, a constitutively active promoter, an inducible promoter, a tissue-specific promoter or a developmental stage specific promoter. Also, the promoter of the a exogenous nucleic acid molecule can be the same as or different from the promoter of a second exogenous nucleic acid molecule.
The polynucleotides of the present disclosure may be stacked with any gene or combination of genes to produce plants with a variety of desired trait combinations, including but not limited to traits desirable for animal feed such as high oil genes (e.g., US Patent Number 6,232,529); balanced amino acids (e.g., hordothionins (US Patent Numbers 5,990,389; 5,885,801 ; 5,885,802 and 5,703,409); barley high lysine (Williamson, et al., (1987) Eur. J. Biochem. 165:99-106 and WO 1998/20122) and high methionine proteins (Pedersen, et al., (1986) J. Biol. Chem. 261 :6279; Kirihara, et al. , (1988) Gene 71 :359 and Musumura, et al. , (1989) Plant Mol. Biol. 12:123)); increased digestibility (e.g., modified storage proteins (US Patent Application Serial Number 10/053,410, filed November 7, 2001 ) and thioredoxins (US Patent Application Serial Number 10/005,429, filed December 3, 2001 )), the disclosures of which are herein incorporated by reference. The polynucleotides of the present disclosure can also be stacked with traits desirable for insect, disease or herbicide resistance (e.g., Bacillus thuringiensis toxic proteins (US Patent Numbers 5,366,892; 5,747,450; 5,737,514; 5723,756; 5,593,881 ; Geiser, et al., (1986) Gene 48:109); lectins (Van Damme, et al. , (1994) Plant Mol. Biol. 24:825); fumonisin detoxification genes (US Patent Number 5,792,931 ); avirulence and disease resistance genes (Jones, et al., (1994) Science 266:789; Martin, et al., (1993) Science 262:1432; Mindrinos, et al., (1994) Cell 78:1089); acetolactate synthase (ALS) mutants that lead to herbicide resistance such as the S4 and/or Hra mutations; inhibitors of glutamine synthase such as phosphinothricin or basta (e.g., bar gene); and glyphosate resistance (EPSPS gene)) and traits desirable for processing or process products such as high oil (e.g., US Patent Number 6,232,529 ); modified oils (e.g., fatty acid desaturase genes (US Patent Number 5,952,544; WO 1994/1 1516)); modified starches (e.g., ADPG pyrophosphorylases (AGPase), starch synthases (SS), starch branching enzymes (SBE) and starch debranching enzymes (SDBE)) and polymers or bioplastics (e.g., US Patent Number 5,602,321 ; beta-ketothiolase, polyhydroxybutyrate synthase, and acetoacetyl- CoA reductase (Schubert, et ai, (1988) J. Bacteriol. 170:5837-5847) facilitate expression of polyhydroxyalkanoates (PHAs)), the disclosures of which are herein incorporated by reference. One could also combine the polynucleotides of the present disclosure with polynucleotides affecting agronomic traits such as male sterility (e.g., see, US Patent Number 5.583,210), stalk strength, flowering time or transformation technology traits such as cell cycle regulation or gene targeting (e.g., WO 1999/61619; WO 2000/17364; WO 1999/25821 ), the disclosures of which are herein incorporated by reference.
Transgenic plants comprising or derived from plant cells or native plants with reduced male fertility of this disclosure can be further enhanced with stacked traits, e.g., a crop plant having an enhanced trait resulting from expression of DNA disclosed herein in combination with herbicide tolerance and/or pest resistance traits. For example, plants with reduced male fertility can be stacked with other traits of agronomic interest, such as a trait providing herbicide resistance and/or insect resistance, such as using a gene from Bacillus thuringensis to provide resistance against one or more of lepidopteran, coleopteran, homopteran, hemiopteran and other insects. Known genes that confer tolerance to herbicides such as e.g., auxin, HPPD, glyphosate, dicamba, glufosinate, sulfonylurea, bromoxynil and norflurazon herbicides can be stacked either as a molecular stack or a breeding stack with plants expressing the traits disclosed herein. Polynucleotide molecules encoding proteins involved in herbicide tolerance include, but are not limited to, a polynucleotide molecule encoding 5-enolpyruvylshikimate-3- phosphate synthase (EPSPS) disclosed in US Patent Numbers 39,247; 6,566,587 and for imparting glyphosate tolerance; polynucleotide molecules encoding a glyphosate oxidoreductase (GOX) disclosed in US Patent Number 5,463,175 and a glyphosate-N- acetyl transferase (GAT) disclosed in US Patent Numbers 7,622,641 ; 7,462,481 ; 7,531 ,339; 7,527,955; 7,709,709; 7,714,188 and 7,666,643, also for providing glyphosate tolerance; dicamba monooxygenase disclosed in US Patent Number 7,022,896 and WO 2007/146706 A2 for providing dicamba tolerance; a polynucleotide molecule encoding AAD12 disclosed in US Patent Application Publication Number 2005/731044 or WO 2007/053482 A2 or encoding AAD1 disclosed in US Patent Application Publication Number 201 1/0124503 A1 or US Patent Number 7,838,733 for providing tolerance to auxin herbicides (2,4-D); a polynucleotide molecule encoding hydroxyphenylpyruvate dioxygenase (HPPD) for providing tolerance to HPPD inhibitors (e.g., hydroxyphenylpyruvate dioxygenase) disclosed in e.g., US Patent Number 7,935,869; US Patent Application Publication Numbers 2009/0055976 A1 and 201 1/0023180 A1 ; each publication is herein incorporated by reference in its entirety.
Other examples of herbicide-tolerance traits that could be combined with the traits disclosed herein include those conferred by polynucleotides encoding an exogenous phosphinothricin acetyltransferase, as described in US Patent Numbers 5,969,213; 5,489,520; 5,550,318; 5,874,265; 5,919,675; 5,561 ,236; 5,648,477; 5,646,024; 6,177,616 and 5,879,903. Plants containing an exogenous phosphinothricin acetyltransferase can exhibit improved tolerance to glufosinate herbicides, which inhibit the enzyme glutamine synthase. Other examples of herbicide-tolerance traits include those conferred by polynucleotides conferring altered protoporphyrinogen oxidase (protox) activity, as described in US Patent Numbers 6,288,306 B1 ; 6,282,837 B1 and 5,767,373 and international publication WO 2001/12825. Plants containing such polynucleotides can exhibit improved tolerance to any of a variety of herbicides which target the protox enzyme (also referred to as "protox inhibitors")
In one embodiment, sequences of interest improve plant growth and/or crop yields. For example, sequences of interest include agronomically important genes that result in improved primary or lateral root systems. Such genes include, but are not limited to, nutrient/water transporters and growth inducers. Examples of such genes include, but are not limited to, maize plasma membrane H+-ATPase (MHA2) (Frias, et al., (1996) Plant Cell 8:1533-44); AKT1 , a component of the potassium uptake apparatus in Arabidopsis, (Spalding, et al., (1999) J Gen Physiol 1 13:909-18); RML genes which activate cell division cycle in the root apical cells (Cheng, et al., (1995) Plant Physiol 108:881 ); maize glutamine synthetase genes (Sukanya, et al., (1994) Plant Mol Biol 26:1935-46) and hemoglobin (Duff, et al., (1997) J. Biol. Chem 27:16749-16752, Arredondo-Peter, et al., (1997) Plant Physiol. 1 15:1259-1266; Arredondo-Peter, et al., (1997) Plant Physiol 1 14:493-500 and references sited therein). The sequence of interest may also be useful in expressing antisense nucleotide sequences of genes that negatively affect root development.
Additional, agronomically important traits such as oil, starch and protein content can be genetically altered in addition to using traditional breeding methods. Modifications include increasing content of oleic acid, saturated and unsaturated oils, increasing levels of lysine and sulfur, providing essential amino acids and also modification of starch. Hordothionin protein modifications are described in US Patent Numbers 5,703,049, 5,885,801 , 5,885,802 and 5,990,389, herein incorporated by reference. Another example is lysine and/or sulfur rich seed protein encoded by the soybean 2S albumin described in US Patent Number 5,850,016 and the chymotrypsin inhibitor from barley described in Williamson, et al., (1987) Eur. J. Biochem. 165:99-106, the disclosures of which are herein incorporated by reference. Derivatives of the coding sequences can be made by site-directed mutagenesis to increase the level of preselected amino acids in the encoded polypeptide. For example, the gene encoding the barley high lysine polypeptide (BHL) is derived from barley chymotrypsin inhibitor, US Patent Application Serial Number 08/740,682, filed November 1 , 1996, and WO 1998/20133, the disclosures of which are herein incorporated by reference. Other proteins include methionine-rich plant proteins such as from sunflower seed (Lilley, et al., (1989) Proceedings of the World Congress on Vegetable Protein Utilization in Human Foods and Animal Feedstuff s, ed. Applewhite (American Oil Chemists Society, Champaign, Illinois), pp. 497-502; herein incorporated by reference); corn (Pedersen, et al., (1986) J. Biol. Chem. 261 :6279; Kirihara, et al., (1988) Gene 71 :359, both of which are herein incorporated by reference) and rice (Musumura, et al., (1989) Plant Mol. Biol. 12:123, herein incorporated by reference). Other agronomically important genes encode latex, Floury 2, growth factors, seed storage factors and transcription factors.
Insect resistance genes may encode resistance to pests that have great yield drag such as rootworm, cutworm, European Corn Borer and the like. Such genes include, for example, Bacillus thuringiensis toxic protein genes (US Patent Numbers 5,366,892; 5,747,450; 5,736,514; 5,723,756; 5,593,881 and Geiser, et al., (1986) Gene 48:109) and the like.
Genes encoding disease resistance traits include detoxification genes, such as against fumonosin (US Patent Number 5,792,931 ); avirulence (avr) and disease resistance (R) genes (Jones, et al., (1994) Science 266:789; Martin, et al., (1993) Science 262:1432 and Mindrinos, et ai, (1994) Cell 78:1089) and the like.
Herbicide resistance traits may include genes coding for resistance to herbicides that act to inhibit the action of acetolactate synthase (ALS), in particular the sulfonylurea- type herbicides (e.g., the acetolactate synthase (ALS) gene containing mutations leading to such resistance, in particular the S4 and/or Hra mutations), genes coding for resistance to herbicides that act to inhibit action of glutamine synthase, such as phosphinothricin or basta (e.g., the bar gene) or other such genes known in the art. The bar gene encodes resistance to the herbicide basta, the nptll gene encodes resistance to the antibiotics kanamycin and geneticin and the ALS-gene mutants encode resistance to the herbicide chlorsulfuron.
Sterility genes can also be encoded in an expression cassette and provide an alternative to physical emasculation. Examples of genes used in such ways include male tissue-preferred genes and genes with male sterility phenotypes such as QM, described in US Patent Number 5,583,210. Other genes include kinases and those encoding compounds toxic to either male or female gametophytic development.
The quality of grain is reflected in traits such as levels and types of oils, saturated and unsaturated, quality and quantity of essential amino acids, and levels of cellulose. In corn, modified hordothionin proteins are described in US Patent Numbers 5,703,049, 5,885,801 , 5,885,802 and 5,990,389.
Commercial traits can also be encoded on a gene or genes that could increase, for example, starch for ethanol production or provide expression of proteins. Another important commercial use of transformed plants is the production of polymers and bioplastics such as described in US Patent Number 5,602,321. Genes such as β- Ketothiolase, PHBase (polyhydroxyburyrate synthase) and acetoacetyl-CoA reductase (see, Schubert, et a/., (1988) J. Bacterial. 170:5837-5847) facilitate expression of polyhyroxyalkanoates (PHAs).
Exogenous products include plant enzymes and products as well as those from other sources including procaryotes and other eukaryotes. Such products include enzymes, cofactors, hormones and the like. The level of proteins, particularly modified proteins having improved amino acid distribution to improve the nutrient value of the plant, can be increased. This is achieved by the expression of such proteins having enhanced amino acid content.
Genome Editing and Induced Mutagenesis
In general, methods to modify or alter the host endogenous genomic DNA are available. This includes altering the host native DNA sequence or a pre-existing transgenic sequence including regulatory elements, coding and non-coding sequences. These methods are also useful in targeting nucleic acids to pre-engineered target recognition sequences in the genome. As an example, the genetically modified cell or plant described herein is generated using "custom" meganucleases produced to modify plant genomes (see, e.g., WO 2009/1 14321 ; Gao, et al., (2010) Plant Journal 1 : 176-187). Other s/ie-directed engineering is through the use of zinc finger domain recognition coupled with the restriction properties of restriction enzyme. See, e.g., Urnov, et al., (2010) Nat Rev Genet. 11 (9):636-46; Shukla, et al., (2009) Nature 459(7245):437-41.
"TILLING" or "Targeting Induced Local Lesions IN Genomics" refers to a mutagenesis technology useful to generate and/or identify and to eventually isolate mutagenised variants of a particular nucleic acid with modulated expression and/or activity (McCallum, et al., (2000), Plant Physiology 123:439-442; McCallum, et al., (2000) Nature Biotechnology 18:455-457 and Colbert, et al., (2001 ) Plant Physiology 126:480- 484).
TILLING combines high density point mutations with rapid sensitive detection of the mutations. Typically, ethylmethanesulfonate (EMS) is used to mutagenize plant seed. EMS alkylates guanine, which typically leads to mispairing. For example, seeds are soaked in an about 10-20 mM solution of EMS for about 10 to 20 hours; the seeds are washed and then sown. The plants of this generation are known as M1. M1 plants are then self-fertilized. Mutations that are present in cells that form the reproductive tissues are inherited by the next generation (M2). Typically, M2 plants are screened for mutation in the desired gene and/or for specific phenotypes.
TILLING also allows selection of plants carrying mutant variants. These mutant variants may exhibit modified expression, either in strength or in location or in timing (if the mutations affect the promoter, for example). These mutant variants may exhibit higher or lower MS44 activity than that exhibited by the gene in its natural form. TILLING combines high-density mutagenesis with high-throughput screening methods. The steps typically followed in TILLING are: (a) EMS mutagenesis (Redei and Koncz, (1992) In Methods in Arabidopsis Research, Koncz, et al. , eds. Singapore, World Scientific Publishing Co, pp. 16-82; Feldmann, et al., (1994) In Arabidopsis. Meyerowitz and Somerville eds, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., pp 137- 172; Lightner and Caspar (1998) In Methods on Molecular Biology 82:91 -104; Martinez- Zapater and Salinas, eds, Humana Press, Totowa, N.J.); (b) DNA preparation and pooling of individuals; (c) PCR amplification of a region of interest; (d) denaturation and annealing to allow formation of heteroduplexes; (e) DHPLC, where the presence of a heteroduplex in a pool is detected as an extra peak in the chromatogram; (f) identification of the mutant individual; and (g) sequencing of the mutant PCR product. Methods for TILLING are well known in the art (US Patent Number 8,071 ,840).
Other mutagenic methods can also be employed to introduce mutations in the MS44 gene. Methods for introducing genetic mutations into plant genes and selecting plants with desired traits 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, and N-nitroso-N-ethylurea. Alternatively, ionizing radiation from sources such as X- rays or gamma rays can be used.
In some embodiments, the present disclosure is exemplified with respect to plant fertility and more particularly with respect to plant male fertility.
Hybrid seed production requires elimination or inactivation of pollen produced by the female parent. Incomplete removal or inactivation of the pollen provides the potential for selfing, raising the risk that inadvertently self-pollinated seed will unintentionally be harvested and packaged with hybrid seed. Once the seed is planted, the selfed plants can be identified and selected; the selfed plants are genetically equivalent to the female inbred line used to produce the hybrid. Typically, the selfed plants are identified and selected based on their decreased vigor relative to the hybrid plants. For example, female selfed plants of maize are identified by their less vigorous appearance for vegetative and/or reproductive characteristics, including shorter plant height, small ear size, ear and kernel shape, cob color or other characteristics. Selfed lines also can be identified using molecular marker analyses (see, e.g., Smith and Wych, (1995) Seed Sci. Technol. 14:1-8). Using such methods, the homozygosity of the self-pollinated line can be verified by analyzing allelic composition at various loci in the genome.
Because hybrid plants are important and valuable field crops, plant breeders are continually working to develop high-yielding hybrids that are agronomically sound based on stable inbred lines. The availability of such hybrids allows a maximum amount of crop to be produced with the inputs used, while minimizing susceptibility to pests and environmental stresses. To accomplish this goal, the plant breeder must develop superior inbred parental lines for producing hybrids by identifying and selecting genetically unique individuals that occur in a segregating population. The present disclosure contributes to this goal, for example by providing plants that, when crossed, generate male sterile progeny, which can be used as female parental plants for generating hybrid plants.
A large number of genes have been identified as being tassel preferred in their expression pattern using traditional methods and more recent high-throughput methods. The correlation of function of these genes with important biochemical or developmental processes that ultimately lead to functional pollen is arduous when approaches are limited to classical forward or reverse genetic mutational analysis. As disclosed herein, suppression approaches in maize provide an alternative rapid means to identify genes that are directly related to pollen development in maize. Promoters useful for expressing a nucleic acid molecule of interest can be any of a range of naturally-occurring promoters known to be operative in plants or animals, as desired. Promoters that direct expression in cells of male or female reproductive organs of a plant are useful for generating a transgenic plant or breeding pair of plants of the disclosure. The promoters useful in the present disclosure can include constitutive promoters, which generally are active in most or all tissues of a plant; inducible promoters, which generally are inactive or exhibit a low basal level of expression and can be induced to a relatively high activity upon contact of cells with an appropriate inducing agent; tissue-specific (or tissue-preferred) promoters, which generally are expressed in only one or a few particular cell types (e.g., plant anther cells) and developmental- or stage-specific promoters, which are active only during a defined period during the growth or development of a plant. Often promoters can be modified, if necessary, to vary the expression level. Certain embodiments comprise promoters exogenous to the species being manipulated. For example, the Ms45 gene introduced into ms45ms45 maize germplasm may be driven by a promoter isolated from another plant species; a hairpin construct may then be designed to target the exogenous plant promoter, reducing the possibility of hairpin interaction with non-target, endogenous maize promoters.
Exemplary constitutive promoters include the 35S cauliflower mosaic virus (CaMV) promoter promoter (Odell, et al., (1985) Nature 313:810-812), the maize ubiquitin promoter (Christensen, et al. , (1989) Plant Mol. Biol. 12:619-632 and Christensen, et al., (1992) Plant Mol. Biol. 18:675-689); the core promoter of the Rsyn7 promoter and other constitutive promoters disclosed in WO 1999/43838 and US Patent Number 6,072,050; rice actin (McElroy, et al., (1990) Plant Cell 2: 163-171 ); pEMU (Last, et al., (1991 ) Theor. Appl. Genet. 81 :581 -588); MAS (Velten, et al., (1984) EMBO J. 3:2723-2730); ALS promoter (US Patent Number 5,659,026); rice actin promoter (US Patent Number 5,641 ,876; WO 2000/70067), maize histone promoter (Brignon, et al., (1993) Plant Mol Bio 22(6):1007-1015; Rasco-Gaunt, et al., (2003) Plant Cell Rep. 21 (6):569-576) and the like. Other constitutive promoters include, for example, those described in US Patent Numbers 5,608,144 and 6,177,61 1 and PCT Publication Number WO 2003/102198.
Tissue-specific, tissue-preferred or stage-specific regulatory elements further include, for example, the AGL8/FRUITFULL regulatory element, which is activated upon floral induction (Hempel, et al., (1997) Development 124:3845-3853); root-specific regulatory elements such as the regulatory elements from the RCP1 gene and the LRP1 gene (Tsugeki and Fedoroff, (1999) Proc. Natl. Acad., USA 96:12941-12946; Smith and Fedoroff, (1995) Plant Cell 7:735-745); flower-specific regulatory elements such as the regulatory elements from the LEAFY gene and the APETALA 1 gene (Blazquez, et al., (1997) Development 124:3835-3844; Hempel, et al., supra, 1997); seed-specific regulatory elements such as the regulatory element from the oleosin gene (Plant, et al., (1994) Plant Mol. Biol. 25:193-205) and dehiscence zone specific regulatory element. Additional tissue-specific or stage-specific regulatory elements include the Zn13 promoter, which is a pollen-specific promoter (Hamilton, et al., (1992) Plant Mol. Biol. 18:21 1-218); the UNUSUAL FLORAL ORGANS {UFO) promoter, which is active in apical shoot meristem; the promoter active in shoot meristems (Atanassova, et al., (1992) Plant J. 2:291 ), the cdc2 promoter and cyc07 promoter (see, for example, Ito, et al., (1994) Plant Mol. Biol. 24:863-878; Martinez, et al., (1992) Proc. Natl. Acad. Sci., USA 89:7360); the meristematic-preferred meri-5 and H3 promoters (Medford, et al. , (1991 ) Plant Cell 3:359; Terada, et al., (1993) Plant J. 3:241 ); meristematic and phloem-preferred promoters of Myb-related genes in barley (Wissenbach, et al., (1993) Plant J. 4:41 1 ); Arabidopsis cyc3aAt and cydAt (Shaul, et al., (1996) Proc. Natl. Acad. Sci. 93:4868-4872); C. roseus cyclins CYS and CYM (Ito, et al., (1997) Plant J. 1 1 :983-992); and Nicotiana CyclinBI (Trehin, et al. , (1997) Plant Mol. Biol. 35:667-672); the promoter of the APETALA3 gene, which is active in floral meristems (Jack, et al., (1994) Cell 76:703; Hempel, et al., supra, 1997); a promoter of an agamous-like (AGL) family member, for example, AGL8, which is active in shoot meristem upon the transition to flowering (Hempel, et al. , supra, 1997); floral abscission zone promoters; L1 -specific promoters; the ripening-enhanced tomato polygalacturonase promoter (Nicholass, et al., (1995) Plant Mol. Biol. 28:423-435), the E8 promoter (Deikman, et al., (1992) Plant Physiol. 100:2013-2017) and the fruit-specific 2A1 promoter, U2 and U5 snRNA promoters from maize, the Z4 promoter from a gene encoding the Z4 22 kD zein protein, the Z10 promoter from a gene encoding a 10 kD zein protein, a Z27 promoter from a gene encoding a 27 kD zein protein, the A20 promoter from the gene encoding a 19 kD zein protein, and the like. Additional tissue-specific promoters can be isolated using well known methods (see, e.g., US Patent Number 5,589,379). Shoot-preferred promoters include shoot meristem-preferred promoters such as promoters disclosed in Weigel, et al. , (1992) Cell 69:843-859 (Accession Number M91208); Accession Number AJ131822; Accession Number Z71981 ; Accession Number AF049870 and shoot-preferred promoters disclosed in McAvoy, et al., (2003) Acta Hort. (ISHS) 625:379-385. Inflorescence-preferred promoters include the promoter of chalcone synthase (Van der Meer, et al., (1992) Plant J. 2(4):525-535), anther-specific LAT52 (Twell, et al., (1989) Mol. Gen. Genet. 217:240-245), pollen-specific Bp4 (Albani, et al., (1990) Plant Mol Biol. 15:605, maize pollen-specific gene Zm13 (Hamilton, et al., (1992) Plant Mol. Biol. 18:21 1-218; Guerrero, et al. , (1993) Mol. Gen. Genet. 224:161 -168), microspore-specific promoters such as the apg gene promoter (Twell, et al. , (1993) Sex. Plant Reprod. 6:217-224) and tapetum-specific promoters such as the TA29 gene promoter (Mariani, et al., (1990) Nature 347:737; US Patent Number 6,372,967) and other stamen-specific promoters such as the MS45 gene promoter, 5126 gene promoter, BS7 gene promoter, PG47 gene promoter (US Patent Number 5,412,085; US Patent Number 5,545,546; Plant J 3(2):261-271 (1993)), SGB6 gene promoter (US Patent Number 5,470,359), G9 gene promoter (US Patent Number 5,8937,850; US Patent Number 5,589,610), SB200 gene promoter (WO 2002/26789), or the like. Tissue-preferred promoters of interest further include a sunflower pollen-expressed gene SF3 (Baltz, et al., (1992) The Plant Journal 2:713-721 ), B. napus pollen specific genes (Arnoldo, et al., (1992) J. Cell. Biochem, Abstract Number Y101204). Tissue-preferred promoters further include those reported by Yamamoto, et al., (1997) Plant J. 12(2):255-265 (psaDb); Kawamata, et al. , (1997) Plant Cell Physiol. 38(7):792-803 (PsPAU ); Hansen, et al., (1997) Mol. Gen Genet. 254(3):337-343 (ORF13); Russell, et al., (1997) Transgenic Res. 6(2):157-168 (waxy or ZmGBS; 27kDa zein, ZmZ27; osAGP; osGT1 ); Rinehart, et al., (1996) Plant Physiol. 112(3): 1331-1341 (Fbl2A from cotton); Van Camp, et al., (1996) Plant Physiol. 1 12(2):525-535 {Nicotiana SodA1 and SodA2); Canevascini, et al., (1996) Plant Physiol. 1 12(2):513-524 {Nicotiana Itp1 ); Yamamoto, et al. , (1994) Plant Cell Physiol. 35(5):773-778 {Pinus cab-6 promoter); Lam, (1994) Results Probl. Cell Differ. 20:181-196; Orozco, et al. , (1993) Plant Mol Biol. 23(6):1 129-1 138 (spinach rubisco activase (Rca)); Matsuoka, et al., (1993) Proc Natl. Acad. Sci. USA 90(20):9586-9590 (PPDK promoter) and Guevara-Garcia, et al., (1993) Plant J. 4(3):495-505 (Agrobacterium pmas promoter). A tissue-preferred promoter that is active in cells of male or female reproductive organs can be particularly useful in certain aspects of the present disclosure.
"Seed-preferred" promoters include both "seed-developing" promoters (those promoters active during seed development such as promoters of seed storage proteins) as well as "seed-germinating" promoters (those promoters active during seed germination). See, Thompson, et al., (1989) BioEssays 10:108. Such seed-preferred promoters include, but are not limited to, Cim1 (cytokinin-induced message), cZ19B1 (maize 19 kDa zein), mil ps (myo-inositol-1 -phosphate synthase); see, WO 2000/1 1 177 and US Patent Number 6,225,529. Gamma-zein is an endosperm-specific promoter. Globulin-1 (Glob-1 ) is a representative embryo-specific promoter. For dicots, seed- specific promoters include, but are not limited to, bean β-phaseolin, napin, β-conglycinin, soybean lectin, cruciferin, and the like. For monocots, seed-specific promoters include, but are not limited to, maize 15 kDa zein, 22 kDa zein, 27 kDa zein, gamma-zein, waxy, shrunken 1 , shrunken 2, globulin 1 , etc. See also, WO 2000/12733 and US Patent Number 6,528,704, where seed-preferred promoters from endl and end2 genes are disclosed. Additional embryo specific promoters are disclosed in Sato, et al., (1996) Proc. Natl. Acad. Sci. 93:81 17-8122 (rice homeobox, OSH1 ) and Postma-Haarsma, et al., (1999) Plant Mol. Biol. 39:257-71 (rice KNOX genes). Additional endosperm specific promoters are disclosed in Albani, et al., (1984) EMBO 3: 1405-15; Albani, et al. , (1999) Theor. Appl. Gen. 98:1253-62; Albani, et al., (1993) Plant J. 4:343-55; Mena, et al. , (1998) The Plant Journal 1 16:53-62 (barley DOF); Opsahl-Ferstad, et al., (1997) Plant J 12:235- 46 (maize Esr) and Wu, et al., (1998) Plant Cell Physiology 39:885-889 (rice GluA-3, GluB-1 , NRP33, RAG-1 ).
An inducible regulatory element is one that is capable of directly or indirectly activating transcription of one or more DNA sequences or genes in response to an inducer. The inducer can be a chemical agent such as a protein, metabolite, growth regulator, herbicide or phenolic compound or a physiological stress, such as that imposed directly by heat, cold, salt, or toxic elements or indirectly through the action of a pathogen or disease agent such as a virus or other biological or physical agent or environmental condition. A plant cell containing an inducible regulatory element may be exposed to an inducer by externally applying the inducer to the cell or plant such as by spraying, watering, heating or similar methods. An inducing agent useful for inducing expression from an inducible promoter is selected based on the particular inducible regulatory element. In response to exposure to an inducing agent, transcription from the inducible regulatory element generally is initiated de novo or is increased above a basal or constitutive level of expression. Typically the protein factor that binds specifically to an inducible regulatory element to activate transcription is present in an inactive form which is then directly or indirectly converted to the active form by the inducer. Any inducible promoter can be used in the instant disclosure (See, Ward, et al. , (1993) Plant Mol. Biol. 22:361-366).
Examples of inducible regulatory elements include a metallothionein regulatory element, a copper-inducible regulatory element or a tetracycline-inducible regulatory element, the transcription from which can be effected in response to divalent metal ions, copper or tetracycline, respectively (Furst, et al., (1988) Cell 55:705-717; Mett, et al., (1993) Proc. Natl. Acad. Sci., USA 90:4567-4571 ; Gatz, et al., (1992) Plant J. 2:397-404; Roder, et al., (1994) Mol. Gen. Genet. 243:32-38). Inducible regulatory elements also include an ecdysone regulatory element or a glucocorticoid regulatory element, the transcription from which can be effected in response to ecdysone or other steroid (Christopherson, et al., (1992) Proc. Natl. Acad. Sci., USA 89:6314-6318; Schena, et al., (1991 ) Proc. Natl. Acad. Sci. USA 88:10421-10425; US Patent Number 6,504,082); a cold responsive regulatory element or a heat shock regulatory element, the transcription of which can be effected in response to exposure to cold or heat, respectively (Takahashi, et al. , (1992) Plant Physiol. 99:383-390); the promoter of the alcohol dehydrogenase gene (Gerlach, et al. , (1982) PNAS USA 79:2981 -2985; Walker, et al., (1987) PNAS 84(19):6624-6628), inducible by anaerobic conditions; and the light-inducible promoter derived from the pea rbcS gene or pea psaDb gene (Yamamoto, et al., (1997) Plant J. 12(2):255-265); a light-inducible regulatory element (Feinbaum, et al., (1991 ) Mol. Gen. Genet. 226:449; Lam and Chua, (1990) Science 248:471 ; Matsuoka, et al., (1993) Proc. Natl. Acad. Sci. USA 90(20):9586-9590; Orozco, et al.,. (1993) Plant Mol. Bio. 23(6):1129- 1138), a plant hormone inducible regulatory element (Yamaguchi-Shinozaki, et al., (1990) Plant Mol. Biol. 15:905; Kares, et al., (1990) Plant Mol. Biol. 15:225), and the like. An inducible regulatory element also can be the promoter of the maize In2-1 or ln2-2 gene, which responds to benzenesulfonamide herbicide safeners (Hershey, et al., (1991 ) Mol. Gen. Gene. 227:229-237; Gatz, et al., (1994) Mol. Gen. Genet. 243:32-38) and the Tet repressor of transposon Tn10 (Gatz, et al., (1991 ) Mol. Gen. Genet. 227:229-237). Stress inducible promoters include salt/water stress-inducible promoters such as P5CS (Zang, et al. , (1997) Plant Sciences 129:81-89); cold-inducible promoters, such as, cor15a (Hajela, et al. , (1990) Plant Physiol. 93:1246-1252), cor15b (Wlihelm, et al., (1993) Plant Mol Biol 23:1073-1077), wsc120 (Ouellet, et al. , (1998) FEBS Lett. 423:324-328), ci7 (Kirch, et al., (1997) Plant Mol Biol. 33:897-909), ci21A (Schneider, et al. , (1997) Plant Physiol. 1 13:335-45); drought-inducible promoters, such as, Trg-31 (Chaudhary, et al. , (1996) Plant Mol. Biol. 30:1247-57), rd29 (Kasuga, et al. , (1999) Nature Biotechnology 18:287- 291 ); osmotic inducible promoters, such as Rab17 (Vilardell, et al., (1991 ) Plant Mol. Biol. 17:985-93) and osmotin (Raghothama, et al., (1993) Plant Mol Biol 23:1 1 17-28) and heat inducible promoters, such as heat shock proteins (Barros, et al., (1992) Plant Mol. 19:665- 75; Marrs, et al., (1993) Dev. Genet. 14:27-41 ), smHSP (Waters, et al., (1996) J. Experimental Botany 47:325-338) and the heat-shock inducible element from the parsley ubiquitin promoter (WO 03/102198). Other stress-inducible promoters include rip2 (US Patent Number 5,332,808 and US Patent Application Publication Number 2003/0217393) and rd29a (Yamaguchi-Shinozaki, et al., (1993) Mol. Gen. Genetics 236:331-340). Certain promoters are inducible by wounding, including the Agrobacterium pmas promoter (Guevara-Garcia, et al. , (1993) Plant J. 4(3):495-505) and the Agrobacterium 0RF13 promoter (Hansen, et al. , (1997) Mol. Gen. Genet. 254(3):337-343).
In certain embodiments, a promoter is selected based, for example, on whether male fertility or female fertility is to be impacted Thus, where the male fertility is to be impacted, (e.g., a BS7 gene and an SB200 gene), the promoter may be, for example, an MS45 gene promoter (US Patent Number 6,037,523), a 5126 gene promoter (US Patent Number 5,837,851 ), a BS7 gene promoter (WO 2002/063021 ), an SB200 gene promoter (WO 2002/26789), a TA29 gene promoter {Nature 347:737 (1990)), a PG47 gene promoter (US Patent Number 5,412,085; US Patent Number 5,545,546; Plant J 3(2):261- 271 (1993)) an SGB6 gene promoter (US Patent Number 5,470,359) a G9 gene promoter (US Patent Numbers 5,837,850 and 5,589,610) or the like. Where female fertility is to be impacted, the promoter can target female reproductive genes, for example an ovary specific promoter. In certain embodiments, any promoter can be used that directs expression in the tissue of interest, including, for example, a constitutively active promoter such as an ubiquitin promoter, which generally effects transcription in most or all plant cells.
Additional regulatory elements active in plant cells and useful in the methods or compositions of the disclosure include, for example, the spinach nitrite reductase gene regulatory element (Back, et al. , (1991 ) Plant Mol. Biol. 17:9); a gamma zein promoter, an oleosin ole16 promoter, a globulin I promoter, an actin I promoter, an actin cl promoter, a sucrose synthetase promoter, an INOPS promoter, an EXM5 promoter, a globulin2 promoter, a b-32, ADPG-pyrophosphorylase promoter, an Ltpl promoter, an Ltp2 promoter, an oleosin ole17 promoter, an oleosin ole18 promoter, an actin 2 promoter, a pollen-specific protein promoter, a pollen-specific pectate lyase gene promoter or PG47 gene promoter, an anther specific RTS2 gene promoter, SGB6 gene promoter, or G9 gene promoter, a tapetum specific RAB24 gene promoter, an anthranilate synthase alpha subunit promoter, an alpha zein promoter, an anthranilate synthase beta subunit promoter, a dihydrodipicolinate synthase promoter, a Thi I promoter, an alcohol dehydrogenase promoter, a cab binding protein promoter, an H3C4 promoter, a RUBISCO SS starch branching enzyme promoter, an actin3 promoter, an actin7 promoter, a regulatory protein GF14-12 promoter, a ribosomal protein L9 promoter, a cellulose biosynthetic enzyme promoter, an S-adenosyl-L-homocysteine hydrolase promoter, a superoxide dismutase promoter, a C-kinase receptor promoter, a phosphoglycerate mutase promoter, a root-specific RCc3 mRNA promoter, a glucose-6 phosphate isomerase promoter, a pyrophosphate-fructose 6-phosphate- l-phosphotransferase promoter, a beta-ketoacyl-ACP synthase promoter, a 33 kDa photosystem 1 1 promoter, an oxygen evolving protein promoter, a 69 kDa vacuolar ATPase subunit promoter, a glyceraldehyde-3-phosphate dehydrogenase promoter, an ABA- and ripening- inducible-like protein promoter, a phenylalanine ammonia lyase promoter, an adenosine triphosphatase S-adenosyl-L-homocysteine hydrolase promoter, a chalcone synthase promoter, a zein promoter, a globulin-1 promoter, an auxin-binding protein promoter, a UDP glucose flavonoid glycosyl-transferase gene promoter, an NTI promoter, an actin promoter and an opaque 2 promoter.
Plants suitable for purposes of the present disclosure can be monocots or dicots and include, but are not limited to, maize, wheat, barley, rye, sweet potato, bean, pea, chicory, lettuce, cabbage, cauliflower, broccoli, turnip, radish, spinach, asparagus, onion, garlic, pepper, celery, squash, pumpkin, hemp, zucchini, apple, pear, quince, melon, plum, cherry, peach, nectarine, apricot, strawberry, grape, raspberry, blackberry, pineapple, avocado, papaya, mango, banana, soybean, tomato, sorghum, sugarcane, sugar beet, sunflower, rapeseed, clover, tobacco, carrot, cotton, alfalfa, rice, potato, eggplant, cucumber, Arabidopsis thaliana and woody plants such as coniferous and deciduous trees. Thus, a transgenic plant or genetically modified plant cell of the disclosure can be an angiosperm or gymnosperm.
Angiosperms are divided into two broad classes based on the number of cotyledons, which are seed leaves that generally store or absorb food; a monocotyledonous angiosperm has a single cotyledon and a dicotyledonous angiosperm has two cotyledons. Angiosperms produce a variety of useful products including materials such as lumber, rubber and paper; fibers such as cotton and linen; herbs and medicines such as quinine and vinblastine; ornamental flowers such as roses and where included within the scope of the present disclosure, orchids and foodstuffs such as grains, oils, fruits and vegetables. Angiosperms encompass a variety of flowering plants, including, for example, cereal plants, leguminous plants, oilseed plants, hardwood trees, fruit-bearing plants and ornamental flowers, which general classes are not necessarily exclusive. Cereal plants, which produce an edible grain, include, for example, corn, rice, wheat, barley, oat, rye, orchardgrass, guinea grass and sorghum. Leguminous plants include members of the pea family (Fabaceae) and produce a characteristic fruit known as a legume. Examples of leguminous plants include, for example, soybean, pea, chickpea, moth bean, broad bean, kidney bean, lima bean, lentil, cowpea, dry bean and peanut, as well as alfalfa, birdsfoot trefoil, clover and sainfoin. Oilseed plants, which have seeds that are useful as a source of oil, include soybean, sunflower, rapeseed (canola) and cottonseed. Angiosperms also include hardwood trees, which are perennial woody plants that generally have a single stem (trunk). Examples of such trees include alder, ash, aspen, basswood (linden), beech, birch, cherry, cottonwood, elm, eucalyptus, hickory, locust, maple, oak, persimmon, poplar, sycamore, walnut, sequoia and willow. Trees are useful, for example, as a source of pulp, paper, structural material and fuel.
Angiosperms produce seeds enclosed within a mature, ripened ovary. An angiosperm fruit can be suitable for human or animal consumption or for collection of seeds to propagate the species. For example, hops are a member of the mulberry family that are prized for their flavoring in malt liquor. Fruit-bearing angiosperms also include grape, orange, lemon, grapefruit, avocado, date, peach, cherry, olive, plum, coconut, apple and pear trees and blackberry, blueberry, raspberry, strawberry, pineapple, tomato, cucumber and eggplant plants. An ornamental flower is an angiosperm cultivated for its decorative flower. Examples of commercially important ornamental flowers include rose, lily, tulip and chrysanthemum, snapdragon, camellia, carnation and petunia plants and can include orchids. It will be recognized that the present disclosure also can be practiced using gymnosperms, which do not produce seeds in a fruit.
Homozygosity is a genetic condition existing when identical alleles reside at corresponding loci on homologous chromosomes. Heterozygosity is a genetic condition existing when different alleles reside at corresponding loci on homologous chromosomes. Hemizygosity is a genetic condition existing when there is only one copy of a gene (or set of genes) with no allelic counterpart on the sister chromosome.
The plant breeding methods used herein are well known to one skilled in the art. For a discussion of plant breeding techniques, see, Poehlman, (1987) Breeding Field Crops AVI Publication Co., Westport Conn. Many of the plants which would be most preferred in this method are bred through techniques that take advantage of the plant's method of pollination.
Backcrossing methods may be used to introduce a gene into the plants. This technique has been used for decades to introduce traits into a plant. An example of a description of this and other plant breeding methodologies that are well known can be found in references such as Plant Breeding Methodology, edit. Neal Jensen, John Wiley & Sons, Inc. (1988). In a typical backcross protocol, the original variety of interest (recurrent parent) is crossed to a second variety (nonrecurrent parent) that carries the single gene of interest to be transferred. The resulting progeny from this cross are then crossed again to the recurrent parent and the process is repeated until a plant is obtained wherein essentially all of the desired morphological and physiological characteristics of the recurrent parent are recovered in the converted plant, in addition to the single transferred gene from the nonrecurrent parent.
By transgene is meant any nucleic acid sequence which has been introduced into the genome of a cell by genetic engineering techniques. A transgene may be a native DNA sequence or a heterologous DNA sequence. The term native DNA sequence can refer to a nucleotide sequence which is naturally found in the cell but that may have been modified from its original form.
Certain constructs described herein comprise an element which interferes with formation, function, or dispersal of male gametes. By way of example but not limitation, this can include use of genes which express a product cytotoxic to male gametes (See for example, US Patent Numbers 5,792,853; 5,689,049; PCT/EP89/00495); inhibit formation of a gene product important to male gamete function or formation (see, US Patent Numbers 5,859,341 ; 6,297,426); combine with another gene product to produce a substance preventing gene formation or function (see, US Patent Numbers 6,162,964; 6,013,859; 6,281 ,348; 6,399,856; 6,248,935; 6,750,868; 5,792,853); are antisense to or cause co-suppression of a gene critical to male gamete function or formation (see, US Patent Numbers 6,184,439; 5,728,926; 6,191 ,343; 5,728,558; 5,741 ,684); interfere with expression through use of hairpin formations (Smith, et al. , (2000) Nature 407:319-320; WO 1999/53050 and WO 1998/53083) or the like. Many nucleotide sequences are known which inhibit pollen formation or function and any sequences which accomplish this function will suffice. A discussion of genes which can impact proper development or function is included at US Patent Number 6,399,856 and includes dominant negative genes such as cytotoxin genes, methylase genes and growth-inhibiting genes. Dominant negative genes include diphtheria toxin A-chain gene (Czako and An, (1991 ) Plant Physiol. 95:687-692. and Greenfield, et al., (1983) PNAS 80:6853, Palmiter, et al., (1987) Cell 50:435); cell cycle division mutants such as CDC in maize (Colasanti, et al., (1991 ) PNAS 88:3377-3381 ); the WT gene (Farmer, et al., (1994) Hum. Mol. Genet. 3:723-728) and P68 (Chen, et al. , (1991 ) PAWS 88:315-319).
Further examples of so-called "cytotoxic" genes are discussed supra and can include, but are not limited to pectate lyase gene pelE, from Erwinia chrysanthermi (Kenn, et al. , (1986) J. Bacteroil 168:595); T-urf13 gene from cms-T maize mitochondrial genomes (Braun, et al., (1990) Plant Cell 2: 153; Dewey, et al., (1987) PNAS 84:5374); CytA toxin gene from Bacillus thuringiensis Israeliensis that causes cell membrane disruption (McLean, et al., (1987) J. Bacteriol 169:1017, US Patent Number 4,918,006); DNAses, RNAses, (US Patent Number 5,633,441 ); proteases or genes expressing anti- sense RNA. A suitable gene may also encode a protein involved in inhibiting pistil development, pollen stigma interactions, pollen tube growth or fertilization or a combination thereof. In addition genes that encode proteins that either interfere with the normal accumulation of starch in pollen or affect osmotic balance within pollen may also be suitable. These may include, for example, the maize alpha-amylase gene, maize beta- amylase gene, debranching enzymes such as Sugaryl and pullulanase, glucanase and SacB.
In an illustrative embodiment, the DAM-methylase gene is used, discussed supra and at US Patent Numbers 5,792,852 and 5,689,049, the expression product of which catalyzes methylation of adenine residues in the DNA of the plant. In another embodiment, an .alpha. -amylase gene can be used with a male tissue-preferred promoter. During the initial germinating period of cereal seeds, the aleurone layer cells will synthesize .alpha. -amylase, which participates in hydrolyzing starch to form glucose and maltose, so as to provide the nutrients needed for the growth of the germ (Rogers and Milliman, (1984) J. Biol. Chem. 259(19):12234-12240; Rogers, (1985) J. Biol. Chem. 260:3731-3738). In an embodiment, the .alpha. -amylase gene used can be the Zea mays . alpha. -amylase- 1 gene. See, for example, Young, et al., Plant Physiol. 105(2):759-760 and GenBank Accession Numbers L25805, Gl:426481. See, also, U.S. Patent 8,013,218. Sequences encoding .alpha. -amylase are not typically found in pollen cells and when expression is directed to male tissue, the result is a breakdown of the energy source for the pollen grains and repression of pollen function.
One skilled in this area readily appreciates the methods described herein are particularly applicable to any other crops which have the potential to outcross. By way of example, but not limitation it can include maize, soybean, sorghum, rice, sunflower, or any plant with the capacity to outcross.
The disclosure contemplates the use of promoters providing tissue-preferred expression, including promoters which preferentially express to the gamete tissue, male or female, of the plant. The disclosure does not require that any particular gamete tissue- preferred promoter be used in the process, and any of the many such promoters known to one skilled in the art may be employed. By way of example, but not limitation, one such promoter is the 5126 promoter, which preferentially directs expression of the gene to which it is linked to male tissue of the plants, as described in US Patent Numbers 5,837,851 and 5,689,051. Other examples include the MS45 promoter described at US Patent Number 6,037,523, SF3 promoter described at US Patent Number 6,452,069, the BS92-7 or BS7 promoter described at WO 2002/063021 , the SBMu200 promoter described at WO 2002/26789, a SGB6 regulatory element described at US Patent Number 5,470,359 and TA39 (Koltunow, et al., (1990) Plant Cell 2: 1201-1224; Goldberg, et ai, (1993) Plant Cell 5: 1217-1229 and US Patent Number 6,399,856. See, also, Nadeau, et ai, (1996) Plant Cell 8(2):213-39 and Lu, et al., (1996) Plant Cell 8(12):2155- 68.
Using well-known techniques, additional promoter sequences may be isolated based on their sequence homology. In these techniques, all or part of a known promoter sequence is used as a probe which selectively hybridizes to other sequences present in a population of cloned genomic DNA fragments (i.e. genomic libraries) from a chosen organism. Methods that are readily available in the art for the hybridization of nucleic acid sequences may be used to obtain sequences which correspond to these promoter sequences in species including, but not limited to, maize (corn; Zea mays), canola (Brassica napus, Brassica rapa ssp.), alfalfa {Medicago sativa), rice {Oryza sativa), rye (Secale cereale), sorghum (Sorghum bicolor, Sorghum vulgare), sunflower (Helianthus annuus), wheat (Triticum aestivum), soybean (Glycine max), tobacco (Nicotiana tabacum), potato (Solanum tuberosum), peanuts (Arachis hypogaea), cotton (Gossypium hirsutum), sweet potato (Ipomoea batatus), cassava (Manihot esculenta), coffee (Cofea spp.), coconut (Cocos nucifera), pineapple (Ananas comosus), citrus trees (Citrus spp.), cocoa (Theobroma cacao), tea (Camellia sinensis), banana (Musa spp.), avocado (Persea americana), fig (Ficus casica), guava (Psidium guajava), mango (Mangifera indica), olive (Olea europaea), oats, barley, vegetables, ornamentals and conifers. Preferably, plants include maize, soybean, sunflower, safflower, canola, wheat, barley, rye, alfalfa and sorghum.
The entire promoter sequence or portions thereof can be used as a probe capable of specifically hybridizing to corresponding promoter sequences. To achieve specific hybridization under a variety of conditions, such probes include sequences that are unique and are preferably at least about 10 nucleotides in length and most preferably at least about 20 nucleotides in length. Such probes can be used to amplify corresponding promoter sequences from a chosen organism by the well-known process of polymerase chain reaction (PCR). This technique can be used to isolate additional promoter sequences from a desired organism or as a diagnostic assay to determine the presence of the promoter sequence in an organism. Examples include hybridization screening of plated DNA libraries (either plaques or colonies; see e.g., Innis, et al., (1990,) PCR Protocols, A Guide to Methods and Applications, eds., Academic Press). In general, sequences that correspond to a promoter sequence of the present disclosure and hybridize to a promoter sequence disclosed herein will be at least 50% homologous, 55% homologous, 60% homologous, 65% homologous, 70% homologous, 75% homologous, 80% homologous, 85% homologous, 90% homologous, 95% homologous and even 98% homologous or more with the disclosed sequence.
Fragments of a particular promoter sequence disclosed herein may operate to promote the pollen-preferred expression of an operably-linked isolated nucleotide sequence. These fragments will comprise at least about 20 contiguous nucleotides, preferably at least about 50 contiguous nucleotides, more preferably at least about 75 contiguous nucleotides, even more preferably at least about 100 contiguous nucleotides of the particular promoter nucleotide sequences disclosed herein. The nucleotides of such fragments will usually comprise the TATA recognition sequence of the particular promoter sequence. Such fragments can be obtained by use of restriction enzymes to cleave the naturally-occurring promoter sequences disclosed herein; by synthesizing a nucleotide sequence from the naturally-occurring DNA sequence or through the use of PCR technology. See particularly, Mullis, et al., (1987) Methods Enzymol. 155:335-350 and Erlich, ed. (1989) PCR Technology (Stockton Press, New York). Again, variants of these fragments, such as those resulting from site-directed mutagenesis, are encompassed by the compositions of the present disclosure.
Thus, nucleotide sequences comprising at least about 20 contiguous nucleotides of the sequences set forth in SEQ ID NO: 64 - 106 are encompassed. These sequences can be isolated by hybridization, PCR, and the like. Such sequences encompass fragments capable of driving pollen-preferred expression, fragments useful as probes to identify similar sequences, as well as elements responsible for temporal or tissue specificity.
Biologically active variants of the promoter sequence are also encompassed by the compositions of the present disclosure. A regulatory "variant" is a modified form of a promoter wherein one or more bases have been modified, removed or added. For example, a routine way to remove part of a DNA sequence is to use an exonuclease in combination with DNA amplification to produce unidirectional nested deletions of double- stranded DNA clones. A commercial kit for this purpose is sold under the trade name Exo-Size™ (New England Biolabs, Beverly, Mass.). Briefly, this procedure entails incubating exonuclease III with DNA to progressively remove nucleotides in the 3' to 5' direction at 5' overhangs, blunt ends or nicks in the DNA template. However, exonuclease III is unable to remove nucleotides at 3', 4-base overhangs. Timed digests of a clone with this enzyme produce unidirectional nested deletions.
One example of a regulatory sequence variant is a promoter formed by causing one or more deletions in a larger promoter. Deletion of the 5' portion of a promoter up to the TATA box near the transcription start site may be accomplished without abolishing promoter activity, as described by Zhu, et al., (1995) The Plant Cell 7:1681 -89. Such variants should retain promoter activity, particularly the ability to drive expression in specific tissues. Biologically active variants include, for example, the native regulatory sequences of the disclosure having one or more nucleotide substitutions, deletions or insertions. Activity can be measured by Northern blot analysis, reporter activity measurements when using transcriptional fusions, and the like. See, for example, Sambrook, et al., (1989) Molecular Cloning: A Laboratory Manual (2nd ed. Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y.), herein incorporated by reference.
The nucleotide sequences for the pollen-preferred promoters disclosed in the present disclosure, as well as variants and fragments thereof, are useful in the genetic manipulation of any plant when operably linked with an isolated nucleotide sequence whose expression is to be controlled to achieve a desired phenotypic response.
The nucleotide sequence operably linked to the regulatory elements disclosed herein can be an antisense sequence for a targeted gene. By "antisense DNA nucleotide sequence" is intended a sequence that is in inverse orientation to the 5'-to-3' normal orientation of that nucleotide sequence. When delivered into a plant cell, expression of the antisense DNA sequence prevents normal expression of the DNA nucleotide sequence for the targeted gene. The antisense nucleotide sequence encodes an RNA transcript that is complementary to and capable of hybridizing with the endogenous messenger RNA (mRNA) produced by transcription of the DNA nucleotide sequence for the targeted gene. In this case, production of the native protein encoded by the targeted gene is inhibited to achieve a desired phenotypic response. Thus the regulatory sequences claimed herein can be operably linked to antisense DNA sequences to reduce or inhibit expression of a native or exogenous protein in the plant.
Regulation of gene expression may be measured in terms of its effect on individual cells. Successful modulation of a trait may be accomplished with high stringency, for example impacting expression in all or nearly all cells of a particular cell type, or with lower stringency. Within a particular tissue, for example, modulation of expression in 98%, 95%, 90%, 80% or fewer cells may result in the desired phenotype. Further, for modification of assimilate partitioning and/or reduced competition for nitrogen between male and female reproductive structures, suppression of male fertility by 50% or even less may be effective and desirable.
EXAMPLES EXAMPLE 1 : Ms44 isolation and characterization
The dominant male sterile allele of the Ms44 gene arose through a seed based EMS mutagenesis treatment of the W23 maize line and was found to be tightly linked to the C2 locus on chromosome 4 (Linkage between Ms44 and C2, Albertsen and Trimnell (1992) MNL 66:49). A map-based cloning approach was undertaken to identify the Ms44 gene. An initial population of 414 individuals was used to rough map Ms44 to chromosome 4. An additional population of 2686 individuals was used for fine mapping. Marker Lab genotyping narrowed the region of the mutation to a 0.43cM interval on chromosome 4.
Additional markers were developed for fine mapping using the 39 recombinants. The Ms44 mutation was mapped to ~80kb region between markers made from the sequences AZM5_9212 (five recombinants) and AZM5_2221 (2 recombinants).
Primers AZM5_9212 For4 (SEQ ID NO: 1 ) and AZM5_9212 Rev4 (SEQ ID NO: 2) were used for an initial round of PCR followed by a second round of PCR using the primers AZM5_9212 ForNest4 (SEQ ID NO: 3) and AZM5_9212 RevNest4 (SEQ ID NO: 4). The PCR product was digested with Mspl and the banding pattern was analyzed to determine the genotypes at this locus.
Primers AZM5_2221 For3 (SEQ ID NO: 5) and AZM5_2221 Rev3 (SEQ ID NO: 6) were used for an initial round of PCR followed by a second round of PCR using the primers AZM5_2221 ForNest3 (SEQ ID NO: 7) and AZM5_2221 RevNest3 (SEQ ID NO: 8). The PCR product was digested with Bsgl and the banding pattern was analyzed to determine the genotypes at this locus..
Within the ~80kb Ms44 interval, a sequencing gap between BACs was present. The gap was sequenced and, within this region, a gene, pco641570, was identified. The first Met codon is found at nucleotide 1201 , with a 101 bp intron at nucleotides 1505-1605 and the stop codon ending at nucleotide 1613 (SEQ I D NO: 9). The gene has an open reading frame of 312 bp which codes for a predicted protein of 104 amino acids (including the stop codon) (SEQ ID NO: 10). The predicted protein has homology to a variety of proteins and contains the InterProscan accession domain IPR003612, a domain found in plant lipid transfer protein/seed storage/trypsin-alpha amylase inhibitors. A secretory signal sequence (SSS) cleavage site was predicted, using SigCleave analysis, at amino acid 23. (von Heijne, G. "A new method for predicting signal sequence cleavage sites" Nucleic Acids Res.: 14:4683 (1986). Improved prediction of signal peptides: SignalP 3.0. , Bendtsen JD, Nielsen H, von Heijne G, Brunak S., J Mol Biol. 2004 Jul 16;340(4):783-95. Von Heijne, G. "Sequence Analysis in Molecular Biology: Treasure Trove or Trivial Pursuit" Acad. Press (1987) 1 13-1 17. See also the SI GCLEAVE program in the EMBOSS (European Molecular Biology Open Software Site) suite of applications online.)
However, SigCleave analysis of ms44 orthologs in related moncot species reveals another potential cleavage site between amino acids 37 and 38. The protein is cysteine rich and BlastP analysis shows the highest homology to plant anther or tapetum specific genes such as the Lims or A9 genes. (The characterization of tapetum-specific cDNAs isolated from a Lilium henryi L. meiocyte subtractive cDNA library. Crossley, et al. , (1995) Planta. 196(3):523-529. The isolation and characterization of the tapetum-specific Arabidopsis thaliana A9 gene. Paul, et al., (1992) Plant Mol Biol. 19(4):61 1 -22.).
RT-PCR analysis was performed on developing anther and leaf cDNAs to assess the expression of the ms44 gene. Ms44-specific primers pco641570-5' (SEQ ID NO: 1 1 ) and pco641570-3'-2 (SEQ ID NO: 12) were used in an RT-PCR reaction with cDNA template from 0.5mm, 1.0mm, 1.5mm and 2.0mm anthers; anthers at pollen mother cell (PMC), Quartet, early uninucleate and binucleate stages of microspore/pollen development; and leaf. Genomic DNA was also used as a template. Expression of ms44 begins early at the PMC stage and continues through quartet and early nucleate microspore stages but is absent by the binucleate stage of pollen development. No expression was detected in leaves.
The pco641570 gene was sequenced from the Ms44 mutant. The first Met codon is found at nucleotide 1222, with a 101 bp intron at nucleotides 1526-1626 and the stop codon ends at nucleotide 1634 (SEQ ID NO: 13). The sequence analysis revealed a nucleotide change which results in a translational change from an Alanine to a Threonine residue at amino acid 37 in the predicted protein (SEQ ID NO: 14). This nucleotide change also created a BsmF1 restriction site in the mutant allele which is not found in the wildtype, which allows for distinguishing the two alleles by amplification of both Ms44 alleles by PCR and subsequent digestion of the products by BsmF1.
MsD-2629 is another dominant male sterile mutant found in maize and was also generated through EMS mutagenesis. This mutant was mapped and found to reside on chromosome 4 very near the Ms44 gene. To determine whether MsD-2629 was an allele of Ms44, the Ms44 gene was PCR amplified and sequenced from MsD-2629 male sterile plants. Two different alleles were found through sequencing. One was a wild-type allele and the second allele had a single nucleotide change (SEQ ID NO: 152) which results in a translational change from the same Alanine residue as Ms44, but to a Valine at amino acid 37 in the predicted protein (SEQ ID NO: 153). This allele was found in all MsD-2629 male sterile plants tested and was not present in male fertile siblings. The MsD-2629 mutant represents a second Ms44 allele and was designated Ms44-2629.
Both Ms44 mutations affect the same Alanine residue at position 37 and that amino acid is implicated through SigCleave analysis as being the possible -1 signal sequence (SS; may also be referred to as SSS) cleavage site. In vitro
transcription/translation (TnT) reactions (EasyXpress™ Insect Kit II, Qiagen, Cat# 32561 ) were performed to assess cleavage of Ms44 protein variants that had been engineered with various amino acid substitutions based on conservation of amino acids around SS cleavage sites. (Patterns of Amino Acids near Signal-Sequence Cleavage Sites. Gunnar Von Heijne (1983) Eur. J. Bioc em. 133,17-21 .) (Sequence Analysis in Molecular Biology: Treasure Trove or Trivial Pursuit. Von Heijne, G. (1987) Acad. Press 1 13-1 17.) The in vitro TnT assay showed that the wild-type ms44 protein (-1 Ala) is processed to a smaller mature form, whereas the mutant Ms44 (-1 Thr) is not. The Ms44-2629 protein (-1 Val) is not processed, nor is a +1 Pro, but a control -1 Gly protein is processed normally as determined by Western blot analysis. These data confirm that the SS cleavage site is between amino acids 37 and 38.
To confirm that this mutation was responsible for the dominant male sterile phenotype, the genomic region was cloned for this allele, containing approximately 1.2Kb of upstream sequence (comprising putative promoter) and about 0.75 KB of sequence downstream of the stop codon. This genomic sequence was sub-cloned into a transformation vector and designated PHP42163. The vector was used to transform maize plants through
Figure imgf000085_0001
transformation. Thirty-six TO plants were grown to maturity and tassels were phenotyped for the presence or absence of pollen. Thirty four of the thirty six plants were completely male sterile. These transgenic plants were genotyped using primers pco641570-5' (SEQ ID NO: 1 1 ) and pco641570-3'-2 (SEQ ID NO: 12) in a PCR reaction and then digested with BsmF1 and run on a 1 % agarose gel. All thirty-four of the male sterile plants contained the mutant Ms44 allele as evidenced by the presence of two smaller bands produced by BsmF1 digestion. Genotyping revealed that the remaining two male fertile plants did not contain an intact mutant Ms44 allele. This confirms that the single nucleotide change in the Ms44 allele results in a dominant male sterile phenotype. The point mutation in the Ms44 gene changes a codon from an Ala to a Thr, with a second allele having an Ala to Val change. The affected amino acid is proposed to be at the -1 position of the SS cleavage site and the two mutations abolish SS cleavage of MS44 as shown by in vitro TnT assays. Without being bound to any theory, the dominance of the mutation may be due to a defect in protein processing through the endoplasmic reticulum (ER) and not due to a functional role of the ms44 gene product as a lipid transfer protein. Since the MS44 protein is cysteine rich, an ER-tethered Ms44 protein may cross-link through disulfide bridges and inhibit overall protein processing in the anther that is ultimately required for male fertility.
EXAMPLE 2: Tassel preferred promoter identification
In transgenics, one can stack a vector of tassel-specific-promoter-driven negative genes, or male sterility mutants, with other vectors that enhance vegetative or ear growth. The combination of tassel reduction and enhancement of other organs can be particularly effective in diverting nutrients to the ear to achieve yield gain.
Tassel-specific promoters can be used to target silencing of the Tls1 gene in the tassel to knock down or knock-out the function of the gene in this tissue. This will reduce or eliminate tassel development, while the gene function in the ear remains intact. Use of the tassel-specific promoters is not limited to Tls1 gene, but can be applied to driving any gene expression in tassel tissues to deliver a negative effect on tissue growth, for example to affect anther, pollen, or any cells that eventually impact male fertility. Tassel- specific promoter candidates are identified based upon their native expression patterns, and are cloned and tested in transgenic plants to confirm their tassel-specificity. EXAMPLE 3: tls1 mutant identification and characterization
The tassel-less (f/s7) mutant was described and mapped on the long arm of chromosome 1 (Albertsen, et al., (1993) Maize Genetics Newsletter 67:51-52). A small F2 population of 75 individuals, generated by crossing homozygous tlsl plants (background unknown) to Mo17, was genotyped to confirm the previously identified tlsl position. The mutation was found to be located between two SNP markers, MZA5484-22 and MZA10765-46. These markers were used to screen for recombinants in a larger F2 population of 2985 individuals. All the recombinants were selected for self-pollination and 177 F3 ears were harvested. 177 F3 families were grown in rows in the field. Phenotypes for all the individuals in rows were taken to determine each F2 line as homozygous wild- type, heterozygous or homozygous tlsl. Leaf punches from 8 individuals of each F3 family were pooled together for genotyping. Using these lines, tlsl was confirmed to be between markers MZA5484 and MZA10765, which were converted to CAPS markers.
Primers MZA5484-F768 (SEQ ID NO: 28) and MZA5484-R (SEQ ID NO: 29) were used to amplify the MZA5484 locus. The PCR product was digested with Mwol and the banding pattern was analyzed to determine the genotypes at this locus.
Primers MZA10765-F429 (SEQ ID NO: 30) and MZA10765-R1062 (SEQ ID NO: 31 ) were used to amplify the MZA10765 locus. The PCR product was digested with Bsll and the banding pattern was analyzed to determine the genotypes at this locus.
Additional markers were used to fine map the tlsl mutation with the 177 F3 families. These markers were developed from DuPont proprietary sequences of known map positions, BAC-end sequences, and other low copy regions. The tlsl mutation was eventually mapped between markers c0375b06_10 and c0260e13_35.
Primers c0375b06_10-For (SEQ ID NO: 32) and c0375b16_10-Rev (SEQ ID NO:
33) were used to amplify the c0375b06_10 locus. PCR product for this reaction was used as template for a second reaction using the primers c0375b06_10-ForNest (SEQ ID NO:
34) and c0375b06_10-RevNest (SEQ ID NO: 35). This PCR product was digested with Mboll and the banding pattern was analyzed to determine the genotypes at this locus.
Primers c0260e13_35-For (SEQ ID NO: 36) and c0260e13_35-Rev (SEQ ID NO:
37) were used to amplify the c0260e13_35 locus. PCR product for this reaction was used as template for a second reaction using the primers c0260e13_35-ForNest (SEQ ID NO:
38) and c0260e13_35-RevNest (SEQ ID NO: 39). This PCR product was digested with Hphl and the banding pattern was analyzed to determine the genotypes at this locus.
The physical interval between the flanking markers c0375b06_10 and c0260e13_35 contained approximately four sequenced BAC clones based on the B73 physical map. Sequencing low copy regions within this interval revealed a very low level of polymorphism and the few markers available co-segregated with the tlsl phenotype.
All the annotated genes in this interval were sequenced to identify the causative mutation.
One gene, annotated as NOD26-like integral membrane protein/aquaporin/ZmNIP3-1
(hereafter known as NIP3-1 ) (SEQ ID NO: 62 - Genomic Sequence from B73; SEQ ID NO: 63 - CDS from B73, SEQ ID NO: 107 NIP3-1 protein), was unable to be amplified in homozygous tlsl individuals but could be amplified in homozygous wild-type and heterozygous lines.
Primer pairs c0297o12_75-For (SEQ ID NO: 40) and c0297o12_75-Rev (SEQ ID NO: 41 ), c0297o12_76-For (SEQ ID NO: 44) and c0297o12_76-Rev (SEQ ID NO: 45), c0297o12_77-For (SEQ ID NO: 48) and c0297o12_77-Rev (SEQ ID NO: 49), c0297o12_78-For (SEQ ID NO: 52) and c0297o12_78-Rev (SEQ ID NO: 53) were used to amplify the genomic region spanning NIP3-1 . PCR products from these reactions were used as templates for second reactions using the corresponding primer pairs: c0297o12_75-ForNest (SEQ ID NO: 42) and c0297o12_75-RevNest (SEQ ID NO: 43), c0297o12_76-ForNest (SEQ ID NO: 46) and c0297o12_76-RevNest (SEQ ID NO: 47), c0297o12_77-ForNest (SEQ ID NO: 50) and c0297o12_77-RevNest (SEQ ID NO: 51 ), c0297o12_78-ForNest (SEQ ID NO: 54) and c0297o12_78-RevNest (SEQ ID NO: 55).
A BAC library was constructed from homozygous tlsl plants in order to determine the nature of the mutation. Sequencing BAC clones covering the tlsl locus revealed a deletion of approximately 6.6kb in comparison to the B73 reference genome, corresponding to the NIP3-1 region. In addition, approximately 9kb of repetitive sequence was present in its place. Therefore, the tlsl phenotype is likely due to the deletion of NIP3-1 in homozygous mutant plants. Candidate Gene Validation
TUSC lines with Mutator (Mu) insertions in the NIP3.1 were identified to validate the candidate gene. Two independent TUSC lines, put-tls1 -P30D5 and put-tls1-P177F10, were confirmed by PCR and sequencing to have Mu insertions within NIP3-1 .
NIP3-1 specific primers D0143578 (SEQ ID NO: 56), D0143579 (SEQ ID NO: 57), D0143584 (SEQ ID NO: 58), or D0143583 (SEQ ID NO: 59) were used in combination with the Mu-specific primer, MuExt22D (SEQ ID NO: 60) to amplify the NIP3- 1 and Mutator junction regions. PCR products from these reactions were used as templates for second reactions using the same NIP3-1 specific primers in combination with another Mu-specific primer, Mulnt19 (SEQ ID NO: 61 ). The PCR product was run on a gel, the major bands excised, DNA extracted using a Gel Purification Kit (Qiagen) and sequenced. Sequencing results were BLASTed to confirm the Mu insertion in NIP3-1.
The TUSC lines mentioned above, which contained a Mu insertion in NIP3-1 , were used in an allelism test. The TUCS lines which were heterozygous for the Mu insertion were used to pollinate heterozygous F3 plants at the tlsl locus. The resulting progenies were phenotyped and genotyped. Plants were genotyped as described below:
To confirm that a progeny from the allelism test contained a Mu insertion in NIP3- 1 , c0297o12_75-Rev (SEQ ID NO: 41 ), c0297o12_76-For (SEQ ID NO: 44), c0297o12_76-Rev (SEQ ID NO: 45), c0297o12_77-For (SEQ ID NO: 48), c0297o12_77- Rev (SEQ ID NO: 49), D0143583 (SEQ ID NO: 59) and D0143584 (SEQ ID NO: 58) were used in combination with the Mu-specific primer, MuExt22D (SEQ ID NO: 60). PCR products from these reactions were used as templates for second reactions using c0297o12_75-RevNest (SEQ ID NO: 43), c0297o12_76-ForNest (SEQ ID NO: 46), c0297o12_76-RevNest (SEQ ID NO: 47), c0297o12_77-ForNest (SEQ ID NO: 50), c0297o12_77-RevNest (SEQ ID NO: 51 ), D0143583 (SEQ ID NO: 59) and D0143584 (SEQ ID NO: 58) respectively in combination with the Mu-specific primer, Mulnt19 (SEQ ID NO: 61 ). A positive PCR product indicated the presence of a Mu insertion.
To determine if a progeny from the allelism test inherited the wild-type or the reference tlsl allele, c0297o12_75-For (SEQ ID NO: 40) was used in combination with c0297o12_75-Rev (SEQ ID NO: 41 ) and c0297o12_77-For (SEQ ID NO: 48) was used in combination with c0297o12_77-Rev (SEQ ID NO: 49). PCR products from these reactions were used as templates for second reactions using c0297o12_75-ForNest (SEQ ID NO: 42) in combination with c0297o12_75-RevNest (SEQ ID NO: 43) and c0297o12_77-ForNest (SEQ ID NO: 50) in combination with c0297o12_77-RevNest (SEQ ID NO: 51 ), respectively.
The phenotyping results from the allelism test were compared with the genotyping results. Individuals without a Mu insertion were wild-type. Of the individuals that contained a Mu insertion, those that contained the wild-type allele of NIP3-1 had a wild- type phenotype while those that had the mutant allele of NIP3-1 mostly had a tlsl phenotype. The few aberrations were attributed to the incomplete penetrance of the tsl1 phenotype, which has been observed in the original description of the tlsl mutant (MNL 67:51 -52) and in the current study.
EXAMPLE 4: Low Nitrogen Seedling Assay Protocol
Seeds produced by transgenic plants are separated into transgene (heterozygous) and null seed using a seed color marker. Two different random assignments of treatments are made to each block of 54 pots, arranged as 6 rows of 9 columns and using 9 replicates of all treatments. In one case, null seed of 5 events of the same construct are mixed and used as control for comparison of the 5 positive events in this block, making up 6 treatment combinations in each block. In the second case, 3 transgenic positive treatments and their corresponding nulls are randomly assigned to the 54 pots of the block, making 6 treatment combinations for each block, containing 9 replicates of all treatment combinations. In the first case transgenic parameters are compared to a bulked construct null; in the second case, transgenic parameters are compared to the corresponding event null. In cases where there are 10, 15 or 20 events in a construct, the events are assigned in groups of 5 events, the variances calculated for each block of 54 pots, but the block null means are pooled across blocks before mean comparisons are made.
Two seeds of each treatment are planted in 4-inch-square pots containing TURFACE® -MVP on 8-inch, staggered centers and watered four times each day with a solution containing the following nutrients:
1 mM CaCI2 2mM MgS04 0.5mM KH2P04 83ppm
Sprint330
3mM KCI 1 mM KN03 1 uM ZnS04 1 uM MnCI2
3uM H3B04 1 uM MnCI2 0.1 uM CuSO4 0.1 uM
NaMo04
After emergence the plants are thinned to one seed per pot. Treatments routinely are planted on a Monday, emerge the following Friday and are harvested 18 days after planting. At harvest, plants are removed from the pots and the Turface® washed from the roots. The roots are separated from the shoot, placed in a paper bag and dried at 70°C for 70 hr. The dried plant parts (roots and shoots) are weighed and placed in a 50 ml conical tube with approximately 20 5/32 inch steel balls and ground by shaking in a paint shaker. Approximately, 30 mg of the ground tissue (weight recorded for later adjustment) is hydrolyzed in 2ml of 20% H202 and 6M H2S04 for 30 min at 170°C. After cooling, water is added to 20 ml, mixed thoroughly and a 50 μΙ aliquot removed and added to 950 μΙ 1 M Na2C03. The ammonia in this solution is used to estimate total reduced plant nitrogen by placing 100 μΙ of this solution in individual wells of a 96 well plate followed by adding 50 μΙ of OPA solution. Fluorescence, excitation = 360nM / emission = 530nM, is determined and compared to NH4CI standards dissolved in a similar solution and treated with OPA solution.
OPA solution - 5ul Mercaptoethanol + 1 ml OPA stock solution (make fresh, daily) OPA stock - 50mg o-phthadialdehyde (OPA - Sigma #P0657) dissolved in 1.5ml methanol + 4.4ml 1 M Borate buffer pH9.5 (3.09g H3B04 + 1 g NaOH in 50ml water) + 0.55ml 20% SDS (make fresh weekly)
Using these data the following parameters are measured and means are compared to null mean parameters using a Student's t test:
Total Plant Biomass Root Biomass
Shoot Biomass
Root/Shoot Ratio
Plant N concentration
Total Plant N
Variance is calculated within each block using a nearest neighbor calculation as well as by Analysis of Variance (ANOV) using a completely random design (CRD) model. An overall treatment effect for each block was calculated using an F statistic by dividing overall block treatment mean square by the overall block error mean square.
EXAMPLE 5: Screening of Gaspe Bay Flint Derived Maize Lines Under Nitrogen Limiting Conditions
Transgenic plants will contain two or three doses of Gaspe Flint-3 with one dose of GS3 (GS3/(Gaspe-3)2X or GS3/(Gaspe-3)3X) and will segregate 1 :1 for a dominant transgene. Plants will be planted in TURFACE®, a commercial potting medium and watered four times each day with 1 mM KN03 growth medium and with 2 mM KN03 or higher, growth medium. Control plants grown in 1 mM KN03 medium will be less green, produce less biomass and have a smaller ear at anthesis. Results are analyzed for statistical significance.
Expression of a transgene will result in plants with improved plant growth in 1 mM KN03 when compared to a transgenic null. Thus biomass and greenness will be monitored during growth and compared to a transgenic null. Improvements in growth, greenness and ear size at anthesis will be indications of increased nitrogen utilization efficiency.
EXAMPLE 6: Assays to Determine Alterations of Root Architecture in Maize
Transgenic maize plants are assayed for changes in root architecture at seedling stage, flowering time or maturity. Assays to measure alterations of root architecture of maize plants include, but are not limited to the methods outlined below. To facilitate manual or automated assays of root architecture alterations, corn plants can be grown in clear pots.
1 ) Root mass (dry weights). Plants are grown in Turface®, a growth medium that allows easy separation of roots. Oven-dried shoot and root tissues are weighed and a root/shoot ratio calculated. 2) Levels of lateral root branching. The extent of lateral root branching (e.g., lateral root number, lateral root length) is determined by sub-sampling a complete root system, imaging with a flat-bed scanner or a digital camera and analyzing with WinRHIZO™ software (Regent Instruments Inc.).
3) Root band width measurements. The root band is the band or mass of roots that forms at the bottom of greenhouse pots as the plants mature. The thickness of the root band is measured in mm at maturity as a rough estimate of root mass.
4) Nodal root count. The number of crown roots coming off the upper nodes can be determined after separating the root from the support medium (e.g., potting mix). In addition the angle of crown roots and/or brace roots can be measured. Digital analysis of the nodal roots and amount of branching of nodal roots form another extension to the aforementioned manual method.
All data taken on root phenotype are subjected to statistical analysis, normally a t- test to compare the transgenic roots with those of non-transgenic sibling plants. One-way ANOVA may also be used in cases where multiple events and/or constructs are involved in the analysis.
EXAMPLE 7: NUE assay of plant growth
Seeds of Arabidopsis thaliana (control and transgenic line), ecotype Columbia, are surface sterilized (Sanchez, et al. , 2002) and then plated on to Murashige and Skoog (MS) medium containing 0.8% (w/v) Bacto™-Agar (Difco). Plates are incubated for 3 days in darkness at 4°C to break dormancy (stratification) and transferred thereafter to growth chambers (Conviron, Manitoba, Canada) at a temperature of 20°C under a 16-h light/8-h dark cycle. The average light intensity is 120 μΕ/Γη2/8. Seedling are grown for 12 days and then transferred to soil based pots. Potted plants are grown on a nutrient- free soil LB2 Metro-Mix® 200 (Scott's Sierra Horticultural Products, Marysville, OH, USA) in individual 1.5-in pots (Arabidopsis system; Lehle Seeds, Round Rock, TX, USA) in growth chambers, as described above. Plants are watered with 0.6 or 6.5 mM potassium nitrate in the nutrient solution based on Murashige and Skoog (MS free Nitrogen) medium. The relative humidity is maintained around 70%. 16-18 days later plant shoots are collected for evaluation of biomass and SPAD readings. EXAMPLE 8: Agrobacterium mediated transformation into maize
Maize plants can be transformed to overexpress a nucleic acid sequence of interest in order to examine the resulting phenotype.
Figure imgf000093_0001
transformation of maize is performed essentially as described by Zhao, et al. , (2006) Met . Mol. Biol. 318:315-323 (see, also, Zhao, et al., (2001 ) Mol. Breed. 8:323-333 and US Patent Number 5,981 ,840 issued November 9, 1999, incorporated herein by reference). The transformation process involves bacterium inoculation, co-cultivation, resting, selection and plant regeneration. 1. Immature Embryo Preparation
Immature embryos are dissected from caryopses and placed in a 2 mL microtube containing 2 mL PHI-A medium.
2. Agrobacterium Infection and Co-Cultivation of Embryos
2.1 Infection Step
PHI-A medium is removed with 1 mL micropipettor and 1 mL Agrobacterium suspension is added. Tube is gently inverted to mix. The mixture is incubated for 5 min at room temperature. 2.2 Co-Culture Step
The Agrobacterium suspension is removed from the infection step with a 1 mL micropipettor. Using a sterile spatula the embryos are scraped from the tube and transferred to a plate of PHI-B medium in a 100x15 mm Petri dish. The embryos are oriented with the embryonic axis down on the surface of the medium. Plates with the embryos are cultured at 20°C, in darkness, for 3 days. L-Cysteine can be used in the co- cultivation phase. With the standard binary vector, the co-cultivation medium supplied with 100-400 mg/L L-cysteine is critical for recovering stable transgenic events.
3. Selection of Putative Transgenic Events
To each plate of PHI-D medium in a 100x15 mm Petri dish, 10 embryos are transferred, maintaining orientation, and the dishes are sealed with Parafilm®. The plates are incubated in darkness at 28°C. Actively growing putative events, as pale yellow embryonic tissue are expected to be visible in 6-8 weeks. Embryos that produce no events may be brown and necrotic, and little friable tissue growth is evident. Putative transgenic embryonic tissue is subcultured to fresh PHI-D plates at 2-3 week intervals, depending on growth rate. The events are recorded.
4. Regeneration of TO plants
Embryonic tissue propagated on PHI-D medium is subcultured to PHI-E medium
(somatic embryo maturation medium); in 100x25 mm Petri dishes and incubated at 28°C, in darkness, until somatic embryos mature, for about 10-18 days. Individual, matured somatic embryos with well-defined scutellum and coleoptile are transferred to PHI-F embryo germination medium and incubated at 28°C in the light (about 80 μΕ from cool white or equivalent fluorescent lamps). In 7-10 days, regenerated plants, about 10 cm tall, are potted in horticultural mix and hardened-off using standard horticultural methods.
Media for Plant Transformation
1. PHI-A: 4g/L CHU basal salts, 1.0 mL/L 1000X Eriksson's vitamin mix, 0.5mg/L thiamin HCL, 1 .5 mg/L 2,4-D, 0.69 g/L L-proline, 68.5 g/L sucrose, 36g/L glucose, pH 5.2. Add 100μΜ acetosyringone, filter-sterilized before using.
2. PHI-B: PHI-A without glucose, increased 2,4-D to 2mg/L, reduced sucrose to 30 g/L and supplemented with 0.85 mg/L silver nitrate (filter-sterilized), 3.0 g/L Gelrite®, 100μΜ acetosyringone (filter-sterilized), pH 5.8.
3. PHI-C: PHI-B without Gelrite® and acetosyringone, reduced 2,4-D to 1 .5 mg/L and supplemented with 8.0 g/L agar, 0.5 g/L Ms-morpholino ethane sulfonic acid (MES) buffer, 100mg/L carbenicillin (filter-sterilized).
4. PHI-D: PHI-C supplemented with 3mg/L bialaphos (filter-sterilized).
5. PHI-E: 4.3 g/L of Murashige and Skoog (MS) salts, (Gibco, BRL 1 1 1 17- 074), 0.5 mg/L nicotinic acid, 0.1 mg/L thiamine HCI, 0.5mg/L pyridoxine HCI, 2.0 mg/L glycine, 0.1 g/L myo-inositol, 0.5 mg/L zeatin (Sigma, cat.no. Z-0164), 1 mg/L indole acetic acid (IAA), 26.4 μg/L abscisic acid (ABA), 60 g/L sucrose, 3 mg/L bialaphos (filter-sterilized), 100 mg/L carbenicillin (filter-sterilized), 8g/L agar, pH 5.6.
6. PHI-F: PHI-E without zeatin, IAA, ABA; sucrose reduced to 40 g/L; replacing agar with 1 .5 g/L Gelrite®; pH 5.6.
Plants can be regenerated from the transgenic callus by first transferring clusters of tissue to N6 medium supplemented with 0.2 mg per liter of 2,4-D. After two weeks the tissue can be transferred to regeneration medium (Fromm, et al., (1990) Bio/Technology 8:833-839).
Phenotypic analysis of transgenic TO plants and T1 plants can be performed. T1 plants can be analyzed for phenotypic changes. Using image analysis T1 plants can be analyzed for phenotypical changes in plant area, volume, growth rate and color analysis at multiple times during growth of the plants. Alteration in root architecture can be assayed as described herein.
Subsequent analysis of alterations in agronomic characteristics can be done to determine whether plants containing the nucleic acid sequence of interest have an improvement of at least one agronomic characteristic, when compared to the control (or reference) plants that have not been so transformed. The alterations may also be studied under various environmental conditions.
Expression constructs containing the nucleic acid sequence of interest that result in a significant alteration in root and/or shoot biomass, improved green color, larger ear at anthesis or yield will be considered evidence that the nucleic acid sequence of interest functions in maize to alter nitrogen use efficiency.
EXAMPLE 9: Electroporation of Agrobacterium tumefaciens LBA4404
Electroporation competent cells (40 μΙ), such as Agrobacterium tumefaciens LBA4404 (containing PHP10523), are thawed on ice (20-30 min). PHP10523 contains VI R genes for T-DNA transfer, an Agrobacterium low copy number plasmid origin of replication, a tetracycline resistance gene and a cos site for in vivo DNA biomolecular recombination. Meanwhile the electroporation cuvette is chilled on ice. The electroporator settings are adjusted to 2.1 kV.
A DNA aliquot (0.5 μΙ_ JT (US Patent Number 7,087,812) parental DNA at a concentration of 0.2 μg -1.0 μg in low salt buffer or twice distilled H20) is mixed with the thawed Agrobacterium cells while still on ice. The mix is transferred to the bottom of electroporation cuvette and kept at rest on ice for 1-2 min. The cells are electroporated (Eppendorf electroporator 2510) by pushing "Pulse" button twice (ideally achieving a 4.0 msec pulse). Subsequently 0.5 ml 2xYT medium (or SOCmedium) are added to cuvette and transferred to a 15 ml Falcon tube. The cells are incubated at 28-30°C, 200-250 rpm for 3 h.
Aliquots of 250 μΙ are spread onto #30B (YM + 50μg/mL Spectinomycin) plates and incubated 3 days at 28-30°C. To increase the number of transformants one of two optional steps can be performed: Option 1: Overlay plates with 30 μΙ of 15 mg/ml Rifampicin. LBA4404 has a chromosomal resistance gene for Rifampicin. This additional selection eliminates some contaminating colonies observed when using poorer preparations of LBA4404 competent cells.
Option 2: Perform two replicates of the electroporation to compensate for poorer electrocompetent cells.
Identification of transformants:
Four independent colonies are picked and streaked on AB minimal medium plus 50mg/ml_ Spectinomycin plates (#12S medium) for isolation of single colonies. The plates are incubated at 28°C for 2-3 days.
A single colony for each putative co-integrate is picked and inoculated with 4 ml #60A with 50 mg/l Spectinomycin. The mix is incubated for 24 h at 28°C with shaking. Plasmid DNA from 4 ml of culture is isolated using Qiagen Miniprep + optional PB wash. The DNA is eluted in 30 μΙ. Aliquots of 2 μΙ are used to electroporate 20 μΙ of DH10b + 20 μΙ of dd H20 as per above.
Optionally a 15 μΙ aliquot can be used to transform 75-100 μΙ of Invitrogen™ Library Efficiency DH5a. The cells are spread on LB medium plus 50mg/mL Spectinomycin plates (#34T medium) and incubated at 37°C overnight.
Three to four independent colonies are picked for each putative co-integrate and inoculated 4 ml of 2xYT (#60A) with 50 μg/ml Spectinomycin. The cells are incubated at 37°C overnight with shaking.
The plasmid DNA is isolated from 4 ml of culture using QIAprep® Miniprep with optional PB wash (elute in 50 μΙ) and 8 μΙ are used for digestion with Sail (using JT parent and PHP10523 as controls).
Three more digestions using restriction enzymes BamHI, EcoRI and Hind 111 are performed for 4 plasmids that represent 2 putative co-integrates with correct Sail digestion pattern (using parental DNA and PHP10523 as controls). Electronic gels are recommended for comparison.
EXAMPLE 10: Particle-mediated bombardment for Transformation of Maize
A vector can be transformed into embryogenic maize callus by particle bombardment, generally as described by Tomes, et al. , Plant Cell, Tissue and Organ Culture: Fundamental Methods, Eds. Gamborg and Phillips, Chapter 8, pgs. 197-213 (1995) and as briefly outlined below. Transgenic maize plants can be produced by bombardment of embryogenically responsive immature embryos with tungsten particles associated with DNA plasmids. The plasmids typically comprise or consist of a selectable marker and an unselected structural gene, or a selectable marker and a polynucleotide sequence or subsequence, or the like.
Preparation of Particles
Fifteen mg of tungsten particles (General Electric), 0.5 to 1.8μ, preferably 1 to 1.8μ, and most preferably 1 μ, are added to 2 ml of concentrated nitric acid. This suspension is sonicated at 0°C. for 20 minutes (Branson Sonifier Model 450, 40% output, constant duty cycle). Tungsten particles are pelleted by centrifugation at 10000 rpm (Biofuge) for one minute and the supernatant is removed. Two milliliters of sterile distilled water are added to the pellet and brief sonication is used to resuspend the particles. The suspension is pelleted, one milliliter of absolute ethanol is added to the pellet and brief sonication is used to resuspend the particles. Rinsing, pelleting and resuspending of the particles are performed two more times with sterile distilled water and finally the particles are resuspended in two milliliters of sterile distilled water. The particles are subdivided into 250-μΙ aliquots and stored frozen.
Preparation of Particle-Plasmid DNA Association
The stock of tungsten particles are sonicated briefly in a water bath sonicator
(Branson Sonifier Model 450, 20% output, constant duty cycle) and 50 μΙ is transferred to a microfuge tube. The vectors are typically cis: that is, the selectable marker and the gene (or other polynucleotide sequence) of interest are on the same plasmid.
Plasmid DNA is added to the particles for a final DNA amount of 0.1 to 10 μ9 in 10 μΙ_ total volume and briefly sonicated. Preferably, 10 μ9 (1 μg/μL in TE buffer) total DNA is used to mix DNA and particles for bombardment. Fifty microliters (50 μΙ_) of sterile aqueous 2.5 M CaCI2 are added and the mixture is briefly sonicated and vortexed. Twenty microliters (20 μΙ_) of sterile aqueous 0.1 M spermidine are added and the mixture is briefly sonicated and vortexed. The mixture is incubated at room temperature for 20 minutes with intermittent brief sonication. The particle suspension is centrifuged and the supernatant is removed. Two hundred fifty microliters (250 μΙ_) of absolute ethanol are added to the pellet, followed by brief sonication. The suspension is pelleted, the supernatant is removed and 60 μΙ of absolute ethanol are added. The suspension is sonicated briefly before loading the particle-DNA agglomeration onto macrocarriers. Preparation of Tissue
Immature embryos of maize are the target for particle bombardment-mediated transformation. Ears from F1 plants are selfed or sibbed and embryos are aseptically dissected from developing caryopses when the scutellum first becomes opaque. This stage occurs about 9 13 days post-pollination and most generally about 10 days post- pollination, depending on growth conditions. The embryos are about 0.75 to 1.5 millimeters long. Ears are surface sterilized with 20 50% Clorox® for 30 minutes, followed by three rinses with sterile distilled water.
Immature embryos are cultured with the scutellum oriented upward, on embryogenic induction medium comprised of N6 basal salts, Eriksson vitamins, 0.5 mg/l thiamine HCI, 30 gm/l sucrose, 2.88 gm/l L-proline, 1 mg/l 2,4-dichlorophenoxyacetic acid, 2 gm/l Gelrite® and 8.5 mg/l AgN03, Chu, et al., (1975) Sci. Sin. 18:659; Eriksson, (1965) Physiol. Plant 18:976. The medium is sterilized by autoclaving at 121 °C for 15 minutes and dispensed into 100x25 mm Petri dishes. AgN03 is filter-sterilized and added to the medium after autoclaving. The tissues are cultured in complete darkness at 28°C. After about 3 to 7 days, most usually about 4 days, the scutellum of the embryo swells to about double its original size and the protuberances at the coleorhizal surface of the scutellum indicate the inception of embryogenic tissue. Up to 100% of the embryos display this response, but most commonly, the embryogenic response frequency is about 80%.
When the embryogenic response is observed, the embryos are transferred to a medium comprised of induction medium modified to contain 120 gm/l sucrose. The embryos are oriented with the coleorhizal pole, the embryogenically responsive tissue, upwards from the culture medium. Ten embryos per Petri dish are located in the center of a Petri dish in an area about 2 cm in diameter. The embryos are maintained on this medium for 3 to 16 hours, preferably 4 hours, in complete darkness at 28°C just prior to bombardment with particles associated with plasmid DNAs containing the selectable and unselectable marker genes.
To effect particle bombardment of embryos, the particle-DNA agglomerates are accelerated using a DuPont PDS-1000 particle acceleration device. The particle-DNA agglomeration is briefly sonicated and 10 μΙ are deposited on macrocarriers and the ethanol is allowed to evaporate. The macrocarrier is accelerated onto a stainless-steel stopping screen by the rupture of a polymer diaphragm (rupture disk). Rupture is affected by pressurized helium. The velocity of particle-DNA acceleration is determined based on the rupture disk breaking pressure. Rupture disk pressures of 200 to 1800 psi are used, with 650 to 1 100 psi being preferred and about 900 psi being most highly preferred. Multiple disks are used to affect a range of rupture pressures.
The shelf containing the plate with embryos is placed 5.1 cm below the bottom of the macrocarrier platform (shelf #3). To effect particle bombardment of cultured immature embryos, a rupture disk and a macrocarrier with dried particle-DNA agglomerates are installed in the device. The He pressure delivered to the device is adjusted to 200 psi above the rupture disk breaking pressure. A Petri dish with the target embryos is placed into the vacuum chamber and located in the projected path of accelerated particles. A vacuum is created in the chamber, preferably about 28 in Hg. After operation of the device, the vacuum is released and the Petri dish is removed.
Bombarded embryos remain on the osmotically-adjusted medium during bombardment, and 1 to 4 days subsequently. The embryos are transferred to selection medium comprised of N6 basal salts, Eriksson vitamins, 0.5 mg/l thiamine HCI, 30 gm/l sucrose, 1 mg/l 2,4-dichlorophenoxyacetic acid, 2 gm/l Gelrite®, 0.85 mg/l Ag N03 and 3 mg/l bialaphos (Herbiace, Meiji). Bialaphos is added filter-sterilized. The embryos are subcultured to fresh selection medium at 10 to 14 day intervals. After about 7 weeks, embryogenic tissue, putatively transformed for both selectable and unselected marker genes, proliferates from a fraction of the bombarded embryos. Putative transgenic tissue is rescued and that tissue derived from individual embryos is considered to be an event and is propagated independently on selection medium. Two cycles of clonal propagation are achieved by visual selection for the smallest contiguous fragments of organized embryogenic tissue.
A sample of tissue from each event is processed to recover DNA. The DNA is restricted with a restriction endonuclease and probed with primer sequences designed to amplify DNA sequences overlapping the coding and non-coding portion of the plasmid. Embryogenic tissue with amplifiable sequence is advanced to plant regeneration.
For regeneration of transgenic plants, embryogenic tissue is subcultured to a medium comprising MS salts and vitamins (Murashige and Skoog, (1962) Physiol. Plant 15:473), 100 mg/l myo-inositol, 60 gm/l sucrose, 3 gm/l Gelrite®, 0.5 mg/l zeatin, 1 mg/l indole-3-acetic acid, 26.4 ng/l cis-trans-abscissic acid and 3 mg/l bialaphos in 100X25 mm Petri dishes and is incubated in darkness at 28°C until the development of well-formed, matured somatic embryos is seen. This requires about 14 days. Well-formed somatic embryos are opaque and cream-colored and are comprised of an identifiable scutellum and coleoptile. The embryos are individually subcultured to a germination medium comprising MS salts and vitamins, 100 mg/l myo-inositol, 40 gm/l sucrose and 1.5 gm/l Gelrite® in 100x25 mm Petri dishes and incubated under a 16 hour light:8 hour dark photoperiod and 40 meinsteinsm sec from cool-white fluorescent tubes. After about 7 days, the somatic embryos germinate and produce a well-defined shoot and root. The individual plants are subcultured to germination medium in 125x25 mm glass tubes to allow further plant development. The plants are maintained under a 16 hour light:8 hour dark photoperiod and 40 meinsteinsm sec from cool-white fluorescent tubes. After about 7 days, the plants are well-established and are transplanted to horticultural soil, hardened off and potted into commercial greenhouse soil mixture and grown to sexual maturity in a greenhouse. An elite inbred line is used as a male to pollinate regenerated transgenic plants.
EXAMPLE 1 1 : Soybean embryo transformation
Soybean embryos are bombarded with a plasmid comprising a preferred promoter operably linked to a heterologous nucleotide sequence comprising a polynucleotide sequence or subsequence, as follows. To induce somatic embryos, cotyledons of 3 5 mm in length are dissected from surface-sterilized, immature seeds of the soybean cultivar A2872, then cultured in the light or dark at 26°C on an appropriate agar medium for six to ten weeks. Somatic embryos producing secondary embryos are then excised and placed into a suitable liquid medium. After repeated selection for clusters of somatic embryos that multiply as early, globular-staged embryos, the suspensions are maintained as described below.
Soybean embryogenic suspension cultures can be maintained in 35 ml liquid media on a rotary shaker, 150 rpm, at 26°C with fluorescent lights on a 16:8 hour day/night schedule. Cultures are sub-cultured every two weeks by inoculating approximately 35 mg of tissue into 35 ml of liquid medium.
Soybean embryogenic suspension cultures may then be transformed by the method of particle gun bombardment (Klein, et al., (1987) Nature (London) 327:70-73, US Patent Number 4,945,050). A DuPont Biolistic™ PDS1000/HE instrument (helium retrofit) can be used for these transformations.
A selectable marker gene that can be used to facilitate soybean transformation is a transgene composed of the 35S promoter from Cauliflower Mosaic Virus (Odell, et al., (1985) Nature 313:810-812), the hygromycin phosphotransferase gene from plasmid pJR225 (from E. coli; Gritz, et al., (1983) Gene 25:179-188) and the 3' region of the nopaline synthase gene from the T-DNA of the Ti plasmid of Agrobacterium tumefaciens. The expression cassette of interest, comprising the preferred promoter and a heterologous polynucleotide, can be isolated as a restriction fragment. This fragment can then be inserted into a unique restriction site of the vector carrying the marker gene.
To 50 μΙ of a 60 mg/ml 1 μηη gold particle suspension is added (in order): 5 μΙ DNA (1 Mg/μΙ), 20 μΙ spermidine (0.1 M) and 50 μΙ CaCI2 (2.5 M). The particle preparation is then agitated for three minutes, spun in a microfuge for 10 seconds and the supernatant removed. The DNA-coated particles are then washed once in 400 μΙ 70% ethanol and resuspended in 40 μΙ of anhydrous ethanol. The DNA/particle suspension can be sonicated three times for one second each. Five microliters of the DNA-coated gold particles are then loaded on each macro carrier disk.
Approximately 300 400 mg of a two-week-old suspension culture is placed in an empty 60X5 mm petri dish and the residual liquid removed from the tissue with a pipette. For each transformation experiment, approximately 5-10 plates of tissue are normally bombarded. Membrane rupture pressure is set at 1 100 psi, and the chamber is evacuated to a vacuum of 28 inches mercury. The tissue is placed approximately 3.5 inches away from the retaining screen and bombarded three times. Following bombardment, the tissue can be divided in half and placed back into liquid and cultured as described above.
Five to seven days post bombardment, the liquid media may be exchanged with fresh media and eleven to twelve days post-bombardment with fresh media containing 50 mg/ml hygromycin. This selective media can be refreshed weekly. Seven to eight weeks post-bombardment, green, transformed tissue may be observed growing from untransformed, necrotic embryogenic clusters. Isolated green tissue is removed and inoculated into individual flasks to generate new, clonally propagated, transformed embryogenic suspension cultures. Each new line may be treated as an independent transformation event. These suspensions can then be subcultured and maintained as clusters of immature embryos or regenerated into whole plants by maturation and germination of individual somatic embryos.
EXAMPLE 12: Ear development of sterile or fertile plants at varying Nitrogen levels
Male sterility reduces the nutrient requirement for tassel development, resulting in improved ear development at anthesis. In this experiment, male sterile and male fertile sibs were grown in varying levels of nitrogen fertility and sampled at -50% pollen shed. Male sterile plants produced larger ears in each nitrogen fertility level, relative to their male fertile sibs. The proportion of male sterile plants with emerged silks was also greater than for the fertile sib plants. Although the biomass (total above ground plant minus the ear dry weight) was greater for plants grown in the higher nitrogen fertility compared to plants grown in lower nitrogen fertility plots, there was no effect of male sterility on biomass. This shows the positive effect of male sterility on the ability of the plant to produce a heavier, more fully developed ear and improve silking without affecting overall vegetative growth.
EXAMPLE 13: Nitrogen Budget study
A study was undertaken to quantify the nitrogen budget of developing maize ears and tassels when the plants are grown in increasing levels of nitrogen fertilizer. When maize is grown under lower nitrogen fertility levels, the nitrogen budget of the ear is negative; that is, during development the ear loses nitrogen to other parts of the plant when nitrogen is limiting. The nitrogen budget of the ear improves as the amount of nitrogen fertilizer provided to the plant increases until the ear maintains a positive increase in nitrogen through to silk emergence. In contrast, the tassel maintains a positive nitrogen budget irrespective of the level of fertility in which the plant is grown. This result clearly shows that the tassel and ear compete for nitrogen during reproductive development and that the developing tassel dominates over the developing ear. Yield improvements associated with male sterile hybrids vectored through improved ear development are consistent with the reduction in competition of ear development with tassel development.
EXAMPLE 14: Yield Improvement with Male Sterile plants
Genetic male sterile hybrids also perform better in field experiments for grain yield. Two field experiments were performed. In one experiment, nitrogen fertilizer was varied, with male-sterile and male-fertile hybrids segregating within each nitrogen fertility level. Plant population density was varied in the second experiment, with male-sterile and male- fertile hybrids segregating within plant population densities. The experimental design of each experiment was a split plot. Nitrogen fertilizer rate was the main plot in the multiple- rate nitrogen experiment, and male-sterile or male-fertile was the sub plot. In the population experiment, plant population was the main plot, and male-sterility or male- fertility was the sub plot. The nitrogen fertilizer rates used in the multiple N experiment were 0, 30, 60, 90, 120 and 150 units (lbs per acre) applied at V3 stage of development. The plant population in the nitrogen multiple rate experiment was 32,000 plants per acre (PPA) whereas 32,000, 48,000 and 64,000 PPA densities were used in the plant population study. The N fertility regime in the population study was 180 units N pre-plant for all populations, followed by 95 units N side dressed at V6 (275 total units N) in all plots. The 48,000 PPA plots were supplemented with an additional 50 units of N 10 days prior to flowering (325 total units N) and the 64,000 PPA plots were supplemented with an additional 100 units of N 10 days prior to flowering (375 total units N).
Significant effects of male sterility were observed in both experiments. A significant effect of nitrogen fertility on yield was also observed but there was no significant effect of population density on yield. Results are presented below for each experiment. Multiple N Experiment
The overall significance level (P > F) of each parameter was analyzed. Overall male sterile plants had statistically significantly (P > F <0.001 ) greater grain yield, number of ears per plot, higher SPAD, more silks, longer and wider ears and more kernels per ear. These parameters also varied significantly with N fertility. There was a significant N fertility x male sterile/fertile interaction in ears per plot and kernels per ear. This was due to the fact that for fertile plants, ear number per plot increased with increased N fertility; the sterile plants had a constant number of ears per plot across all of the N fertility levels. Silk number and kernels per ear also had significant treatment interactions and were likely due to a steeper rate of increase in silk number with N fertility in the male sterile plants than in the male fertile plants. The difference in yield between male fertile and male sterile plants was much greater at low N than at higher N levels. At 0 N the difference between male sterile and male fertile plants was 84% whereas the difference in yield between male sterile and male fertile plants was 15% at 150 lb/acre N rate. In a hybrid trial involving MS44 mutants, an average increase of about 37 bu/acre was observed. In another hybrid trial, the average increase was about 13 bu/acre. (Figures 5A-5B).
SPAD was significantly different in response to N fertility and in response to male sterility but the response to N fertility of male sterile and male fertile plants was parallel, suggesting SPAD could not account for the difference in yield between male sterile and male fertile plants in response to N fertility.
Kernel number of male-sterile and male-fertile plants in response to N fertility showed slopes similar to the male-sterile and male-fertile yield response to N fertility, which suggests the increase in yield of male-sterile plants might be related to increased kernel number. Differences in yield between male-sterile and male-fertile hybrids across N fertilities could nearly be accounted for by the sum of the differences in ears per plot and kernels per ear between male-sterile and male-fertile hybrids across N fertilities. These data are in agreement with the hypothesis that ear development is less encumbered by tassel development in male-sterile plants, resulting in more fully developed ears (kernels/ear) with a greater success rate of ear production (ear/plot) under low N. In one of the hybrid trials, the ear dry weight of male-sterile plants increased about 62% compared to the ear dry weight from male- fertile plants.
Population/Male Sterility Experiment
The genetic male sterile hybrid also responded better than the male fertile hybrid in the population stress experiment. Though there was no effect of population on grain yield, the genetic male-sterile hybrid outperformed the male-fertile hybrid by 40% (59 bu/a) in all populations tested (see, Figure 7A). In addition, in a separate trial, an average increase of about 8 bu/acre was observed (see, Figure 7B).
EXAMPLE 15: Characterization of tls1 gene and utilization for yield enhancement
Phenotype of the tlsl mutant is shown in Figure 8. A positional cloning approach was undertaken to clone tlsl (Figure 9). The tlsl region was roughly mapped on Chr1 using 75 individuals from a tlsl x Mo17 F2 population. A) The first round of fine mapping is indicated by the red font, tlsl was narrowed to a 15cM region using 2985 F2 individuals. The resulting 177 recombinants were selfed and the progeny from each line were pooled together for further fine mapping, indicated by the green font. The 177 F3 families were used to narrow the tlsl interval to a four BAC region, containing no additional informative markers. The genes in the four BAC interval were sequenced and the only obvious difference was that ZmNIP3;1 could not be PCR amplified in the mutant. A BAC library from homozygous tlsl plants was created and BACs spanning the ZmNI P3;1 gene were sequenced to determine the nature of the mutation. B) BAC sequencing results. A yellow line indicates sequence that could be aligned to the B73 reference sequence. A blue line indicates repetitive sequence that could not be aligned to the B73 reference sequence. ZmNIP3;1 is missing in the mutant and in its place is ~9kb of repetitive sequence. The closest neighboring genes, cytochrome P450 and IMP dehydrogenase, are indicated. Figures 2A and 2B are not drawn to scale. Sequence analysis of NIP3-1 from maize revealed a high level of similarity to NIP5;1 from Arabidopsis (AtNIP5;1 ) and NIP3;1 from rice (OsNIP3; 1 ) and phylogenetic studies showed that they are closely related proteins in the NIP II subgroup (Liu, et al. , (2009) BCM Genomics 10:1471-2164). (Figure 15). These results suggest that NIP3-1 in maize is involved in boron uptake, and boron is needed for reproductive development. Studies can be performed which manipulate the expression of tlsl in the development of hybrid maize for yield improvement under normal and stress conditions (e.g., nitrogen and water stress). NIP3-1 would be down-regulated in a tissue-specific manner (i.e., in the tassel), resulting in plants with no tassels that do not exhibit any of the other pleotropic effects associated with boron deficiency (e.g., underdeveloped ears). In this case, the resources that would be needed for tassel development may be allocated to the ear and shading effects from tassels would be minimized, resulting in an increased yield over other male sterility techniques in which a tassel is present. This same approach may be applied to any genes involved in the transport of boron. tlsl Mutant Phenotype Rescued with Boron Application
Wild type and mutant plants from the F2 mapping population of tlsl x Mo17 were planted. Half of the mutant and wild type plants were treated once a week from ~V2 to ~V6 stage with a foliar boron spray consisting of 0.0792% B202 and 0.0246% elemental Boron. It was observed that the mutant plants treated with the boron spray exhibited an increased number of tassel branches, which were longer and reminiscent of wild type in comparison to the untreated mutant plants. In addition, ears of the treated mutant plants appeared to be recovered as well. Wild type plants treated with boron had no discernable difference from untreated wild type plants. Recovered mutant plants were self-pollinated for a progeny test.
Progeny from selfing the recovered mutant plants were planted along with wild type for a control. Half the mutant progeny was treated with the boron spray as described above and half were left untreated. Tassel branch number (Figure 1 1 ), branch length (Figure 12) and ear length (Figure 13) were measured from 24 wild type plants, 26 mutant plants treated with the boron spray and 29 untreated mutant plants. In comparison to the untreated mutant plants, mutant plants treated with the boron spray exhibited an increased number of tassel branches, increased tassel branch length, and an increased ear length similar to wild type plants (Figures 1 1-13). In addition, the observation that the progeny of recovered mutant plants still display the tlsl phenotype when left untreated indicates that the effects of treating with the boron spray are not transmitted to subsequent generations. tlsl Mutant are More Tolerant to Boron Toxicity
Preliminary results suggest that the tlsl mutant may be more tolerant of boron toxic conditions than wild type plants. Wild type and mutant plants were grown hydroponically using Hoagland media containing either a normal Boron concentration (0.5ppm) or 50ppm of Boron. At ~V7 stage, mutant and wild type plants grown under normal Boron conditions were indistinguishable (Figure 14). However, when grown in 50ppm of Boron, mutant plants appeared larger overall and had wider leaves. In addition, in wild type plants grown in 50ppm Boron, the node of the second youngest fully expanded leaf extended above the node of the youngest fully expanded leaf, while the mutant plants appeared normal.
Mutant rescue and seed production by boron application
Homozygous tlsl plants have reduced tassel growth or substantially lack functional tassel for normal ear development. Therefore, the quantity of seeds from tlsl mutant plants or plants with reduced tassel development due to a deficiency in boron uptake are not to the levels needed for large-scale seed production. Because exogenous boron application rescues tassle development and growth in the tlsl mutant background, boron application is an option to increase seed production from tlsl plants. Depending on the need and the mode of application, exogenous boron (e.g., as a foliar spray) can be applied at various stages of reproductive growth (e.g., V2-V12 or V2-V8) and with varying levels of boron (e.g., 10-1000 ppm). In an embodiment, boron application can coincide with the transition from vegetative to reproductive state, e.g., V4-V5 depending on plant growing conditions.
Alleles of tlsl
Based on the disclosure and guidance provided herein, additional weaker or stronger alleles of tlsl are obtained by performing available screens, e.g., through Targeting Induced Local Lesions in Genomes (TILLING), McCallum, et al., (2000) Nat Biotechnol 18:455-457. Additional alleles of Tlsl can include those variants that completely block boron transport resulting in substantial loss of tassel growth and development and those variants that result in for example, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or 95% reduction in tassel development as evidenced by the reduced pollen production or other suitable parameter known to those or ordinary skill in the art.
EXAMPLE 16: Field Experiments on Reduced Male Fertilitv plants with Drought Stress Treatments
The effect of reduced male fertility on yield of maize grown under drought stress conditions is evaluated in a field study. The field study is conducted in a managed stress field environment. The field location receives little or no rainfall during the growing season, allowing for the controlled imposition of drought stress by removing the irrigation at various stages of development. This field location has no insect or disease pressure to interfere with the interpretation of hybrid performance under drought.
Male sterile and male-fertile versions of a single hybrid are planted in 10 replicates of a split plot design using standard planting practices. Plants are thinned to a standard density so that plant water use perplot should be uniform. Half of the research plots receive full irrigation to meet the water demands of the plant throughout development. This Full Irrigation treatment serves as the control environment. The other half of the research plots receive the drought stress treatment. At approximately the V8 stage of development, irrigation is withheld from these plots and the plants will continue to utilize water that is remaining in the soil profile. After approximately 3 weeks, plant water deficits will occur, as indicated by leaf rolling and decreased plant growth. Plants will remain under this water deficit condition until approximately 2 weeks after flowering, when the stress replicates are fully rewatered. Thus the total duration of the stress treatment is about 5-6 weeks, bracketing the flowering period of development.
Maize is extremely sensitive to drought stress during the flowering period. Typically, development of the ears, exsertion of the silks and pollination of the ovaries are all inhibited by drought stress. The sensitivity of these processes is a major factor in reducing yield under drought stress. Alleviation of this sensitivity is an effective method of improving drought stress in maize. Male-sterile plants will partition more assimilates to the ear during this critical period than will male-fertile plants, thus making male-sterile plants more tolerant to this stress. The male-sterile plants will exsert silks more rapidly, resulting in more efficient pollination of those ovaries and a higher final kernel number/plant. The improvement of this critical reproductive process results in greater yield at harvest.
Thus drought tolerance is improved by reduced male fertility. Yields of both male- sterile and male-fertile hybrids in this stress treatment are compared to the yields under full irrigation. The reduction in yield under stress, compared to the full irrigation treatment, is less in the male-sterile hybrid, confirming the improvement in yield stability that is expected through the use of male sterility.
EXAMPLE 17: Creation of male-sterile hybrid progeny
A method for production of male-sterile hybrid plants is provided. In the hybrid production field, in one embodiment, female parent (male-sterile) plants of inbred A, homozygous recessive for a male-fertility gene, are fertilized by plants of inbred B. Inbred B is similarly homozygous recessive for the male-fertility gene; however, Inbred B is hemizygous for a heterologous construct. This construct comprises (a) the dominant allele of the male-fertility gene, which complements the recessive genotype and restores fertility to inbred B; (b) a genetic element which results in disruption of the formation, function, or dispersal of pollen; (c) optionally, a marker gene, which may be a marker expressed in seed. As a result, seed produced on Inbred A are homozygous recessive for the male-fertility gene and will produce male-sterile progeny. These progeny are non- transgenic with respect to the described construct, because element "b" prevents transmission of the construct through pollen. See, for example, Figure 3.
Because these hybrid plants are male-sterile, it is necessary to provide a pollinator. The pollinator can be planted in rows or plots separate from the male-sterile- hybrid seed, or can be interspersed in an orderly or random way among the male-sterile- hybrid plants. For planting of these hybrid seed in a grain-production field, it is practical to blend the hybrid seed with pollinator seed. The pollinator seed will be present in the minimum amount necessary to achieve adequate pollination of a substantial portion of the plants produced from the blended seed. Preferably, at least 1 % to 50%, more preferably less than 25%, most preferably less than 15% of the blend (by weight) will be pollinator seed. Especially preferred is a blend wherein the pollinator seed is present in an amount of about 1 % to 10% by weight. A substantial portion would be about 90% of the plants produced, more preferably about 95%, most preferably about 98% or more of the plants produced by the blend. To take advantage of effects of xenia, the pollinator may be non- isogenic to the sterile hybrid plants.
EXAMPLE 18: Creation of hybrid male-sterile progeny using dominant Ms44
In this example, the cloned dominant male-sterile gene Ms44 is used to produce male-sterile hybrid plants. See, Figure 4, for example. A male-sterile inbred containing Ms44 in the heterozygous state is transformed with a heterologous SAM construct that comprises (1 ) a Suppression element, for example an inverted repeat (IR) engineered to the Ms44 promoter or Ms44 coding region; (2) a pollen Ablation gene which results in disruption of the formation, function, or dispersal of pollen; (3) a Marker gene, which may be a seed color gene. The suppression element disrupts expression of the dominant Ms44 allele, such that the otherwise male-sterile plant is male-fertile and can be selfed. Because element 2 prevents transgene transmission through pollen, the resulting progeny on the ear will segregate 50:50 with respect to the hemizygous SAM construct and 25% of all the progeny will be homozygous for the Ms44 dominant allele. Seeds comprising the SAM construct can be identified by presence of the marker. Progeny from these seed can be genotyped to identify homozygous Ms44 progeny with the SAM construct; these plants are collectively referred to as the maintainer line (or "transgenic maintainer" line). Homozygous Ms44 progeny without the SAM construct are collectively referred to as the male sterile female inbred (or "male-sterile inbred" line).
Male-sterile inbred seed can be increased by crossing the maintainer line onto male sterile inbred lines. The resulting progeny are male-sterile homozygous Ms44 female inbreds, because the SAM construct is not passed through pollen to progeny. In this way the transgenic maintainer line is used to maintain, propagate, or increase the male sterile plants.
In a hybrid production cross, the male inbred crosses normally onto this male- sterile inbred line, and no detasseling is required. However, because the Ms44 gene is a dominant male-sterile gene and is homozygous in the female inbred, 100% of the hybrid seed will contain one dominant Ms44 allele, and plants produced from those seed will be male-sterile.
When this hybrid seed is planted in a grain-production field, it is practical to blend it with seed of a pollinator. The pollinator seed is present in the minimum necessary amount sufficient to permit adequate pollination of the plants produced from the blend. Preferably, at least 1 % to 50%, more preferably less than 25%, most preferably less than 15%, of the blend (by weight) will be pollinator seed. Especially preferred is a blend wherein the pollinator seed is present in an amount of about 1 -10% by weight. The pollinator seed should be present in the blend only in an amount sufficient to pollinate a substantial portion of the plants produced by the blend. A substantial portion would be about 90% of the plants produced, more preferably about 95%, most preferably about 98% or more of the plants produced by the blend.
Alternatively, the dominant Ms44 gene could be introduced transgenically, operably linked to a heterologous promoter that is amenable to inverted-repeat inactivation and expresses, such that dominant male sterility is achieved. Use of the heterologous promoter would ensure that the native ms44 expression is not inhibited by the pIR (promoter inverted repeat). The 5126 promoter from another species, such as rice, may be particularly appropriate, since the 5126 promoter has an expression pattern that is similar to that of the ms44 gene and has been successfully inactivated by pIR methods.
This approach has applications not only for yield gain during stress but is also useful for any crop that can outcross to weedy species, such as sorghum, by reducing the propensity for outcrossing and minimizing the risk of adventitious presence. For example, the biofuels industry is utilizing enzymes transgenically to aid in the digestibility of substrates (i.e. cellulose) used in ethanol production. Linking these types of transgenes to the Ms44 gene would prevent outcrossing through pollen in a production field. One or more dominant traits could be linked to Ms44 to prevent an unintentional outcross to weedy species.
EXAMPLE 19: Dominant male sterility in hybrids
The dominant male sterility (DMS) gene Ms44 is introgressed into an inbred maize line. Since this gene acts dominantly, selfing of these lines is not possible and the mutation will segregate 50:50 in resulting outcrossed progeny. Linked genetic markers may be employed to identify those seeds or plants containing the DMS gene so they can be selected for pollination by a maize male inbred line to create F1 hybrid seed. Again this hybrid seed will segregate 50% for male sterility. Ms41 and Ms42 are other known DMS mutants that are dominant in maize. (Liu and Cande, (1992) MNL 66:25-26; and
Albertsen, et al., (1993) MNL 67:64)
An alternative approach is to use a transgenic Ms44 gene for dominant sterility.
This gene would be linked to a seed marker gene and transformed into an inbred line.
Seed from this line could then be sorted based on the presence of the seed marker gene to ensure a pure population of Ms44 male-sterile progeny. These progeny would then be crossed with a male inbred in a hybrid production field to yield 50% male sterility in the resultant hybrid progeny.
EXAMPLE 20: Variants of Disclosed Sequences
Additional MS44 mutant sequences can be generated by known means including but not limited to truncations and point mutations. These variants can be assessed for their impact on male fertility by using standard transformation, regeneration, and evaluation protocols. A. Variant Nucleotide Sequences That Do Not Alter the Encoded Amino Acid
Sequence
The disclosed nucleotide sequences are used to generate variant nucleotide sequences having the nucleotide sequence of the open reading frame with about 70%, 75%, 80%, 85%, 90% and 95% nucleotide sequence identity when compared to the starting unaltered ORF nucleotide sequence of the corresponding SEQ ID NO. These functional variants are generated using a standard codon table. While the nucleotide sequence of the variants is altered, the amino acid sequence encoded by the open reading frames does not change. These variants are associated with component traits that determine biomass production and quality. The ones that show association are then used as markers to select for each component traits.
B. Variant Nucleotide Sequences in the non-coding regions
The disclosed nucleotide sequences are used to generate variant nucleotide sequences having the nucleotide sequence of the 5'-untranslated region, 3'-untranslated region or promoter region that is approximately 70%, 75%, 80%, 85%, 90% and 95% identical to the original nucleotide sequence of the corresponding SEQ ID NO. These variants are then associated with natural variation in the germplasm for component traits related to biomass production and quality. The associated variants are used as marker haplotypes to select for the desirable traits.
C. Variant Amino Acid Sequences of Disclosed Polypeptides
Variant amino acid sequences of the disclosed polypeptides are generated. In this example, one amino acid is altered. Specifically, the open reading frames are reviewed to determine the appropriate amino acid alteration. The selection of the amino acid to change is made by consulting the protein alignment (with the other orthologs and other gene family members from various species). An amino acid is selected that is deemed not to be under high selection pressure (not highly conserved) and which is rather easily substituted by an amino acid with similar chemical characteristics (i.e., similar functional side-chain). Using a protein alignment, an appropriate amino acid can be changed. Once the targeted amino acid is identified, the procedure outlined in the following section C is followed. Variants having about 70%, 75%, 80%, 85%, 90% and 95% nucleic acid sequence identity are generated using this method. These variants are then associated with natural variation in the germplasm for component traits related to biomass production and quality. The associated variants are used as marker haplotypes to select for the desirable traits.
D. Additional Variant Amino Acid Sequences of Disclosed Polypeptides
In this example, artificial protein sequences are created having 80%, 85%, 90% and 95% identity relative to the reference protein sequence. This latter effort requires identifying conserved and variable regions from an alignment and then the judicious application of an amino acid substitutions table. These parts will be discussed in more detail below.
Largely, the determination of which amino acid sequences are altered is made based on the conserved regions among disclosed protein or among the other disclosed polypeptides. Based on the sequence alignment, the various regions of the disclosed polypeptide that can likely be altered are represented in lower case letters, while the conserved regions are represented by capital letters. It is recognized that conservative substitutions can be made in the conserved regions below without altering function. In addition, one of skill will understand that functional variants of the disclosed sequence of the disclosure can have minor non-conserved amino acid alterations in the conserved domain.
Artificial protein sequences are then created that are different from the original in the intervals of 80-85%, 85-90%, 90-95% and 95-100% identity. Midpoints of these intervals are targeted, with liberal latitude of plus or minus 1 %, for example. The amino acids substitutions will be effected by a custom Perl script. The substitution table is provided below in Table 2.
Table 2. Substitution Table
Figure imgf000112_0001
c Na No good substitutes
P Na No good substitutes
First, any conserved amino acids in the protein that should not be changed is identified and "marked off" for insulation from the substitution. The start methionine will of course be added to this list automatically. Next, the changes are made.
H, C and P are not changed in any circumstance. The changes will occur with isoleucine first, sweeping N-terminal to C-terminal. Then leucine, and so on down the list until the desired target it reached. Interim number substitutions can be made so as not to cause reversal of changes. The list is ordered 1 -17, so start with as many isoleucine changes as needed before leucine, and so on down to methionine. Clearly many amino acids will in this manner not need to be changed. L, I and V will involve a 50:50 substitution of the two alternate optimal substitutions.
The variant amino acid sequences are written as output. Perl script is used to calculate the percent identities. Using this procedure, variants of the disclosed polypeptides are generated having about 80%, 85%, 90% and 95% amino acid identity to the starting unaltered ORF nucleotide sequence.
E. Variant Amino Acid Sequences of Disclosed Polypeptides that Interfere with Signal Peptide Processing
Variant amino acid sequences of the disclosed polypeptides are generated. In this example, one or more amino acids are altered. Specifically, the N-terminal secretory signal sequence (SS) is reviewed to determine the possible amino acid(s) alteration. The selection of the amino acid to change is made by predicting the SS cleavage site using available prediction programs such as SignalP (von Heijne, G. "A new method for predicting signal sequence cleavage sites" Nucleic Acids Res.: 14:4683 (1986). Improved prediction of signal peptides: SignalP 3.0. , Bendtsen JD, Nielsen H, von Heijne G, Brunak S., J Mol Biol. 2004 Jul 16;340(4):783-95. See also the sigcleave program in the EMBOSS (European Molecular Biology Open Software Site) suite of applications online.) An amino acid is selected that is deemed to be necessary for proper protein processing and secretion. Secretory proteins are synthesized on ribosomes bound to the rough ER. In the plant cell, the signal sequence, a sequence of hydrophobic amino acids usually at the N-terminus, is bound by a signal-recognition particle (SRP), which in turn is bound by an SRP receptor on the rough ER membrane. The SRP directs the binding of the ribosome to the ER membrane, as well as threading the protein through the transmembrane channel, called the translocon, where it is processed into its mature form by signal peptidase cleavage of the SS, producing a mature protein. An amino acid change that disrupts SRP binding or signal peptidase cleavage could inhibit the normal processing and secretion of the protein. For the Ms44 protein these types of amino acid substitutions would lead to a dominant male sterility phenotype.
In an embodiment, the present disclosure includes nucleic acids having a polynucleotide sequence that encodes a non-functional signal peptide (SP). Also included are polypeptides or proteins having a non-functional SP. A non-functional SP will result in the protein having the non-functional SP being retained and/or associated in the ER. A non-functional SP may disrupt the processing, such as cleavage, and/or secretion of the polypeptide or protein containing the non-functional SP.
Disruption of the processing and/or secretion of the polypeptide or protein containing the non-functional SP herein may confer a dominant phenotype to the plant. In one embodiment the dominant phenotype is reduced fertility. In one aspect, the reduced fertility is reduced male fertility. In one aspect, the reduced fertility is reduced female fertility.
Various techniques can be used to create a non-functional signal peptide. This may involve modifying an existing SP so that it is non-functional or fusing a non-functional SP to a protein with a functional SP. Accordingly the present disclosure includes fusion proteins containing a non-functional SP as described herein.
Signal peptide sequences in polypeptides or proteins can be predicted or identified in a number of ways as is known in the art, including through the use of prediction software. For example, SignallP predicts the presence and location of signal peptide cleavage sites in amino acid sequences, von Heijne, G. "A new method for predicting signal sequence cleavage sites" Nucleic Acids Res.: 14:4683 (1986). Improved prediction of signal peptides: SignalP 3.O., Bendtsen JD., Nielsen H., von Heijne G., Brunak S., J Mol Biol. 2004 Jul 16;340(4):783-95. See also the sigcleave program in the EMBOSS (European Molecular Biology Open Software Site) suite of applications online.
The SP or predicted SP sequence can be rendered non-functional by inserting, deleting or substituting one or more nucleotides in a polynucleotide encoding an amino acid residue that is deemed to be necessary for proper protein processing and/or secretion of the protein or the polypeptide. The insertion, deletion or substitution of one or more nucleotides in a polynucleotide encoding an amino acid residue may be in the SP or in the encoded protein associated with the SP. The one or more insertions, deletions or substitutions may alter an amino acid at position -10, -9, -8, -7, -6, -5, -4, -3, -2 -1 , +1 , +2, +3, +4, +5, +6, +7, +8, +9, +10 with respect to SP cleavage site, e.g., a predicted SP cleavage site, where the cleavage site is located between positions +1 and -1. The one or more insertions, deletions or substitutions may be alone or combinations thereof.
Any amino acid insertion, deletion, or substitution at position +2 relative to the SP cleavage site that causes the polypeptide or protein to be retained in the ER may be used in the nucleic acids, proteins, polypeptides, and/or methods of the present disclosure. In one embodiment, the amino acid inserted or substituted at position +2 relative to the SP cleavage site is an aromatic amino acid residue, such as phenylalanine, histidine, tryptophan, or tyrosine. In one embodiment, the amino acid inserted or substituted at position +2 relative to the SP cleavage site is a charged amino acid residue such as aspartic acid, glutamic acid, lysine, or arginine. In one embodiment, the amino acid inserted or substituted at position +2 relative to the SP cleavage site is a large polar amino acid residue such as asparagine or glutamine. In one embodiment, the amino acid inserted or substituted at position +2 relative to the SP cleavage site is a hydrophobic amino acid residue such as phenylalanine, isoleucine, leucine, methionine, or valine. In one embodiment, the amino acid inserted or substituted at position +2 relative to the SP cleavage site is a small neutral amino acid residue such as alanine, cysteine, serine, or threonine. In one embodiment, the amino acid inserted or substituted at position +2 relative to the SP cleavage site is a glycine or proline amino acid.
Any amino acid insertion, deletion, or substitution at position +1 relative to the SP cleavage that causes the polypeptide or protein to be retained in the ER may be used in the nucleic acids, proteins, polypeptides, and/or the methods of the present disclosure. In one embodiment, an amino acid residue is inserted, deleted or substituted for a a charged amino acid residue such as aspartic acid, glutamic acid, lysine, or arginine at position +1 relative to the SP cleavage site. In one embodiment, the amino acid inserted or substituted at position +1 relative to the SP cleavage site is an aromatic amino acid residue, such as phenylalanine, histidine, tryptophan, or tyrosine. In one embodiment, the amino acid inserted or substituted at position +1 relative to the SP cleavage site is a large polar amino acid residue such as asparagine or glutamine. In one embodiment, the amino acid inserted or substituted at position +1 relative to the SP cleavage site is a hydrophobic amino acid residue such as phenylalanine, isoleucine, leucine, methionine, or valine. In one embodiment, the amino acid inserted or substituted at position +1 relative to the SP cleavage site is a glycine or proline amino acid.
Any amino acid insertion, deletion, or substitution at position -1 relative to the SP cleavage that causes the polypeptide or protein to be retained in the ER may be used in the nucleic acids, proteins, polypeptides, and/or the methods of the present disclosure. In one embodiment, an amino acid residue is inserted, deleted or substituted for an alanine at position -1 relative to the SP cleavage site. In one embodiment, an amino acid residue is inserted, deleted or substituted for a small neutral amino acid, such as alanine, cysteine, serine, or threonine. In one embodiment, an amino acid residue is inserted, deleted or substituted for glycine or proline at position -1 relative to the SP cleavage site. In one embodiment, the amino acid inserted or substituted at position -1 relative to the SP cleavage site is an aromatic amino acid residue, such as phenylalanine, histidine, tryptophan, or tyrosine. In one embodiment, the amino acid inserted or substituted at position -1 relative to the SP cleavage site is a charged amino acid residue such as aspartic acid, glutamic acid, lysine, or arginine. In one embodiment, the amino acid inserted or substituted at position -1 relative to the SP cleavage site is a large polar amino acid residue such as asparagine or glutamine. In one embodiment, the amino acid inserted or substituted at position -1 relative to the SP cleavage site is a hydrophobic amino acid residue such as phenylalanine, isoleucine, leucine, methionine, or valine. In one embodiment, the amino acid inserted or substituted at position -1 relative to the SP cleavage site is a proline amino acid.
Any amino acid insertion, deletion, or substitution at position -2 relative to the SP cleavage that causes the polypeptide or protein to be retained in the ER may be used in the nucleic acids, proteins, polypeptides, and/or the methods of the present disclosure. In one embodiment, an amino acid residue is inserted, deleted or substituted for an aromatic amino acid residue, such as phenylalanine, histidine, tryptophan, or tyrosine, at position - 2 relative to the SP cleavage site.
In one embodiment, an amino acid residue is inserted, deleted or substituted for an hydrophobic amino acid residue, such as phenylalanine, isoleucine, leucine, methionine, or valine, at position -2 relative to the SP cleavage site. In one embodiment, an amino acid residue is inserted, deleted or substituted for a small neutral amino acid, such as alanine, cysteine, serine, threonine. In one embodiment, an amino acid residue is inserted, deleted or substituted for glycine or proline, at position -2 relative to the SP cleavage site. In one embodiment, the amino acid inserted or substituted at position -2 relative to the SP cleavage site is a charged amino acid residue such as aspartic acid, glutamic acid, lysine, or arginine. In one embodiment, the amino acid inserted or substituted at position -2 relative to the SP cleavage site is a small neutral amino acid residue such as alanine, cysteine, serine, threonine. In one embodiment, the amino acid inserted or substituted at position -2 relative to the SP cleavage site is glycine or proline. Any amino acid insertion, deletion, or substitution at position -3 relative to the SP cleavage that causes the polypeptide or protein to be retained in the ER may be used in the nucleic acids, proteins, polypeptides, and/or the methods of the present disclosure. In one embodiment, an amino acid residue is inserted, deleted or substituted for an alanine at position -3 relative to the SP cleavage site. In one embodiment, an amino acid residue is inserted, deleted or substituted for a hydrophobic amino acid residue, such as phenylalanine, isoleucine, leucine, methionine, or valine, at position -3 relative to the SP cleavage site. In one embodiment, an amino acid residue is inserted, deleted or substituted for a small neutral amino acid residue such as alanine, cysteine, serine, threonine. In one embodiment, an amino acid residue is inserted, deleted or substituted for glycine or proline at position -3 relative to the SP cleavage site. In one embodiment, the amino acid inserted or substituted at position -3 relative to the SP cleavage site is an aromatic amino acid residue, such as phenylalanine, histidine, tryptophan, or tyrosine. In one embodiment, the amino acid inserted or substituted at position -3 relative to the SP cleavage site is a charged amino acid residue such as aspartic acid, glutamic acid, lysine, or arginine. In one embodiment, the amino acid inserted or substituted at position -3 relative to the SP cleavage site is a large polar amino acid residue such as asparagine or glutamine. In one embodiment, the amino acid inserted or substituted at position -3 relative to the SP cleavage site is a proline or glycine amino acid.
Any amino acid insertion, deletion, or substitution at position -4 relative to the SP cleavage that causes the polypeptide or protein to be retained in the ER may be used in the nucleic acids, proteins, polypeptides, and/or the methods of the present disclosure. In one embodiment, an amino acid residue is inserted, deleted or substituted for an alanine at position -4 relative to the SP cleavage site. In one embodiment, an amino acid residue is inserted, deleted or substituted for a hydrophobic amino acid residue, such as henylalanine, isoleucine, leucine, methionine, or valine, at position -4 relative to the SP cleavage site. In one embodiment, an amino acid residue is inserted, deleted or substituted for a small neutral amino acid residue such as alanine, cysteine, serine, threonine. In one embodiment, an amino acid residue is inserted, deleted or substituted for glycine or proline at position -4 relative to the SP cleavage site. In one embodiment, the amino acid inserted or substituted at position -4 relative to the SP cleavage site is an aromatic amino acid residue, such as phenylalanine, histidine, tryptophan, or tyrosine. In one embodiment, the amino acid inserted or substituted at position -4 relative to the SP cleavage site is a glycine. In one embodiment, the amino acid inserted or substituted at position -4 relative to the SP cleavage site is an aromatic amino acid residue, such as phenylalanine, histidine, tryptophan, or tyrosine. In one embodiment, the amino acid inserted or substituted at position -4 relative to the SP cleavage site is a charged amino acid residue such as aspartic acid, glutamic acid, lysine, or arginine. In one embodiment, the amino acid inserted or substituted at position -4 relative to the SP cleavage site is a large polar amino acid residue such as asparagine or glutamine. In one embodiment, the amino acid inserted or substituted at position -4 relative to the SP cleavage site is a glycine amino acid.
Any amino acid insertion, deletion, or substitution at position -5 relative to the SP cleavage that causes the polypeptide or protein to be retained in the ER may be used in the nucleic acids, proteins, polypeptides, and/or the methods of the present disclosure. In one embodiment, an amino acid residue is inserted, deleted or substituted for an alanine at position -5 relative to the SP cleavage site. In one embodiment, an amino acid residue is inserted, deleted or substituted for a small neutral amino acid residue such as alanine, cysteine, serine, or threonine. In one embodiment, an amino acid residue is inserted, deleted or substituted for glycine or proline, at position -5 relative to the SP cleavage site. In one embodiment, the amino acid inserted or substituted at position -5 relative to the SP cleavage site is an aromatic amino acid residue, such as phenylalanine, histidine, tryptophan, or tyrosine. In one embodiment, the amino acid inserted or substituted at position -5 relative to the SP cleavage site is a charged amino acid residue such as aspartic acid, glutamic acid, lysine, or arginine. In one embodiment, the amino acid inserted or substituted at position -5 relative to the SP cleavage site is a large polar amino acid residue such as asparagine or glutamine. In one embodiment, the amino acid inserted or substituted at position -5 relative to the SP cleavage site is a hydrophobic amino acid residue such as phenylalanine, isoleucine, leucine, methionine, or valine. In one embodiment, the amino acid inserted or substituted at position -5 relative to the SP cleavage site is a glycine amino acid.
Any amino acid insertion, deletion, or substitution at position -6 relative to the SP cleavage that causes the polypeptide or protein to be retained in the ER may be used in the nucleic acids, proteins, polypeptides, and/or the methods of the present disclosure. In one embodiment, an amino acid residue is inserted, deleted or substituted for an alanine at position -6 relative to the SP cleavage site. In one embodiment, an amino acid residue is inserted, deleted or substituted for a hydrophobic amino acid residue, such as phenylalanine, isoleucine, leucine, methionine, or valine, at position -6 relative to the SP cleavage site. In one embodiment, an amino acid residue is inserted, deleted or substituted for a small neutral amino acid residue such as alanine, cysteine, serine, or threonine. In one embodiment, an amino acid residue is inserted, deleted or substituted for glycine or proline, at position -6 relative to the SP cleavage site. In one embodiment, the amino acid inserted or substituted at position -6 relative to the SP cleavage site is an aromatic amino acid residue, such as phenylalanine, histidine, tryptophan, or tyrosine. In one embodiment, the amino acid inserted or substituted at position -6 relative to the SP cleavage site is a charged amino acid residue such as aspartic acid, glutamic acid, lysine, or arginine. In one embodiment, the amino acid inserted or substituted at position -6 relative to the SP cleavage site is a large polar amino acid residue such as asparagine or glutamine. In one embodiment, the amino acid inserted or substituted at position -6 relative to the SP cleavage site is a proline or glycine amino acid.
The nucleic acid can include the nucleotide sequence of SEQ ID NO:13, 15 or 152 encoding the SP. In other embodiments the nucleic acid includes one of the sequences set forth in SEQ ID NO: 13, 15 or 152 or fragments thereof that includes sequences encoding the SP. The present disclosure also includes polypeptides or proteins that have the SP of SEQ ID NO:14 or 153. In other embodiments the nucleic acid includes the sequences encoding the amino acid sequences set forth in SEQ ID NO: 14 or 153 or fragments thereof that includes sequences encoding the SP.
Disruption of secretion and/or processing of the polypeptide or protein can be determined using any number of routine assays, including but not limited to in vitro transcription/translation assays reactions (EasyXpress Insect Kit II, Qiagen, (Carlsbad, CA) Cat# 32561 ). For example, a polypeptide or protein with a non-functional SP of the present disclosure may be retained in the ER as compared to a polypeptide or protein with a functional signal peptide that is secreted extracellularly. Retention in the ER and lack of protein processing can be determined using any number of routine assays, including but not limited to in vitro transcription/translation assays reactions (EasyXpress Insect Kit II, Qiagen, (Carlsbad, CA), Cat# 32561 ) or using ultrastructural microscopy. The disclosure furthermore relates to vectors or constructs containing the nucleic acid molecules of the disclosure. The nucleic acid molecule of the disclosure can be operatively linked to any suitable regulatory element or promoter as described elsewhere herein.
In a further embodiment, the present disclosure relates to host cells transiently or stably containing the nucleic acid molecules or vectors/constructs of the disclosure.
Certain embodiments are listed herein. 1 . An isolated nucleic acid comprising a polynucleotide sequence encoding a
polypeptide having a non-functional signal peptide (SP), wherein the non-functional signal peptide is not able to be processed and wherein expression of the nucleic acid in a plant confers a dominant phenotype to the plant.
The isolated nucleic acid of embodiment 1 , wherein the polypeptide having a nonfunctional signal peptide is retained in the ER.
The isolated nucleic acid of embodiment 1 , wherein the non-functional signal peptide is rendered non-functional due to one or more insertions, deletions or substitutions of an amino acid residue in the SP.
The isolated nucleic acid of embodiment 3, wherein the polypeptide having the nonfunctional signal peptide has one or more amino acid insertions, deletions or substitutions at position -10, -9, -8, -7, -6, -5, -4, -3, -2 -1 , +1 , =2, +3, +4, +5, +6, +7, +8, +9, +10 with respect to a SP cleavage site in the SP, wherein the SP cleavage site is located between positions -1 and +1.
The isolated nucleic acid of embodiment 1 , wherein nucleic acid comprises the non-functional SP of SEQ ID NO.:13, 15 or 152.
The isolated nucleic acid of embodiment 1 , wherein the non-functional SP is encoded by the polynucleotide sequence corresponding to position 1222-1332 of SEQ ID NO.:13 and fragments thereof.
The isolated nucleic acid of embodiment 1 , wherein the non-functional SP is encoded by the polynucleotide sequence corresponding to position 1 -1 1 1 of SEQ I D NO.:15 and fragments thereof.
The isolated nucleic acid of embodiment 1 , wherein the non-functional SP is encoded by the polynucleotide sequence corresponding to position 1 -1 1 1 of SEQ I D NO.:152 and fragments thereof.
The isolated nucleic acid of embodiment 1 , wherein nucleic acid comprises the sequence of SEQ ID NO. 13, 15 or 152.
The isolated nucleic acid of embodiment 1 , wherein the polypeptide comprises the non-functional SP of SEQ ID NO: 14 or 153.
The isolated nucleic acid of embodiment 1 , wherein the non-functional SP comprises the amino acid sequence corresponding to position 1-37 of SEQ ID NO.:14 and fragments thereof.
The isolated nucleic acid of embodiment 1 , wherein the non-functional SP comprises the amino acid sequence corresponding to position 1-37 of SEQ ID NO.:153 and fragments thereof.
The isolated nucleic acid of embodiment 1 , wherein the polypeptide comprises the sequence of SEQ ID NO: 14 or 153.
The isolated nucleic acid of embodiment 1 , wherein the non-functional signal peptide has a threonine or valine at position -1 relative to the SP cleavage site. The isolated nucleic acid of embodiment 1 , wherein the non-functional signal peptide has a proline at position +1 relative to the SP cleavage site.
The isolated nucleic acid of embodiment 1 , wherein the dominant phenotype is reduced male fertility.
The isolated nucleic acid of embodiment 1 , wherein plant is a crop plant.
The isolated nucleic acid of embodiment 1 , wherein plant is a monocot or dicot plant.
The isolated nucleic acid of embodiment 1 , wherein the polypeptide is a tassel-less (f/s7) mutant. The isolated nucleic acid of embodiment 1 , wherein the polynucleotide encoding a polypeptide having a non-functional signal peptide is operably linked to promoter. The isolated nucleic acid of embodiment 20, wherein the promoter is an inducible promoter, a constituitive promoter, a tissue preferred promoter, a temporally regulated promoter or an element thereof.
The isolated nucleic acid of embodiment 20, wherein the promoter preferentially drives expression in male reproductive tissue.
The isolated nucleic acid of embodiment 20, wherein the promoter is a tassel- preferred promoter.
The isolated nucleic acid of embodiment 20, wherein the promoter is selected from the group consisting of:
a. SEQ ID NOS: 62 and 64-106, and the regulatory regions of SEQ ID NO: 9 and 13;
b. at least 100 contiguous nucleotides of SEQ ID NOS: 62 and 64-106, and of the regulatory regions of SEQ ID NO: 9 and 13; and
c. a sequence having at least 70% sequence identity to the full length of SEQ ID NOS: 62 and 64-106, and of the regulatory regions of SEQ ID NO: 9 and 13.
The isolated nucleic acid of embodiment 1 , wherein the polynucleotude encodes an anther specific polypeptide.
The isolated nucleic acid of embodiment 25, wherein the anther specific polypeptide is ms45 or ms26.
The isolated nucleic acid of embodiment 16, wherein the dominant reduced male fertility phenotype results from cell ablation.
The isolated nucleic acid of embodiment 1 , wherein expression of the nucleic acid in a plant increases the yield of a plant.
An isolated construct/vector comprising the nucleic acid of any of the embodiments of 1-28.
A cell transformed with the construct or vector of embodiment of any of the embodiments of 1-28.
A plant having the nucleic acid of any of the embodiments of 1 -28, wherein the plant has increased yield compared to the yield of a plant that does not express the nucleic acid of any of the embodiments of 1-28.
A plant having the nucleic acid of any of the embodiments of 1 -28, wherein the plant has reduced male fertility compared to the male fertility of a plant that does not express the nucleic acid of any of the embodiments of 1-28.
An isolated polypeptide having a non-functional signal peptide (SP), wherein the non-functional signal peptide is not able to be processed and wherein expression of the polypeptide in a plant confers a dominant phenotype to the plant.
The polypeptide of embodiment 33, wherein the polypeptide having a nonfunctional signal peptide is retained in the ER.
The polypeptide of embodiment 33, wherein the non-functional signal peptide is rendered non-functional due to one or more insertions, deletions or substitutions of an amino acid residue in the SP.
The polypeptide of embodiment 35, wherein the non-functional signal peptide has one or more amino acid insertions, deletions or substitutions at position -10, -9, -8, - 7, -6, -5, -4, -3, -2 -1 , +1 , =2, +3, +4, +5, +6, +7, +8, +9, +10 with respect to a SP cleavage site in the SP, wherein the SP cleavage site is located between position - 1 and +1. The polypeptide of embodiment 3, wherein the polypeptide comprises the nonfunctional SP of SEQ ID NO: 14 or 153.
The polypeptide of embodiment 33, wherein the non-functional SP comprises the amino acid sequence corresponding to position 1 -37 of SEQ I D NO.:14 and fragments thereof.
The polypeptide of embodiment 33, wherein the non-functional SP comprises the amino acid sequence corresponding to position 1 -37 of SEQ ID NO.:153 and fragments thereof.
The polypeptide of embodiment 33, wherein the polypeptide comprises the sequence of SEQ ID NO: 14 or 153.
The polypeptide of embodiment 33, wherein the non-functional signal peptide has a threonine or valine at position -1 relative to the SP cleavage site.
The polypeptide of embodiment 33, wherein the non-functional signal peptide has a proline at position +1 relative to the SP cleavage site.
The polypeptide of embodiment 33, wherein the dominant phenotype is reduced male fertility.
The polypeptide of embodiment 33, wherein plant is a crop plant.
The polypeptide of embodiment 33, wherein plant is a monocot or dicot plant.
The polypeptide of embodiment 33, wherein the polypeptide is a tassel-less {tls1) mutant.
The polypeptide of embodiment 33, wherein the polynucleotide encoding the polypeptide having a non-functional signal peptide is operably linked to promoter. The polypeptide of embodiment 47, wherein the promoter is an inducible promoter, a constituitive promoter, a tissue preferred promoter, a temporally regulated promoter or an element thereof.
The polypeptide of embodiment 47, wherein the promoter preferentially drives expression in male reproductive tissue.
The polypeptide of embodiment 47, wherein the promoter is a tassel-preferred promoter.
The polypeptide of embodiment 47, wherein the promoter is selected from the group consisting of:
a. SEQ ID NOS: 62 and 64-106, and the regulatory regions of SEQ ID NOs: 9 and 13;
b. at least 100 contiguous nucleotides of SEQ ID NOS: 62 and 64-106, and of the regulatory regions of SEQ ID NO: 9 and 13; and
c. a sequence having at least 70% sequence identity to any one of the full length of SEQ ID NOS: 62 and 64-106 or to the full length of the regulatory regions of SEQ ID NO: 9 or 13.
The isolated polypeptide of embodiment 33, wherein the polynucleotide encodes an anther specific polypeptide.
The isolated polypeptide of embodiment 52, wherein the anther specific
polypeptide is ms45 or ms26.
The isolated polypeptide of embodiment 43, wherein the dominant reduced male fertility phenotype results from cell ablation.
The isolated polypeptide of embodiment 33, wherein expression of the polypeptide in a plant increases the yield of the plant
A cell expressing any of the polypeptides of embodiments of 33-55. 57. A plant expressing any of the polypeptides of embodiments of 33-55, wherein the plant has increased yield compared to the yield of a plant that does not express any of the polypeptides of embodiments of 1-55.
58. A plant expressing any of the polypeptides of embodiments of 33-55, wherein the plant has reduced male fertility compared to the male fertility of a plant that does not express any of the polypeptides of embodiments of 33-55.
59. A method of conferring a dominant phenotype of reduced male fertility to a plant comprising:
a. expressing in the plant a nucleic acid of any of the embodiments of 1-28, thereby conferring the dominant phenotype of reduced male fertility to the plant.
60. A method of conferring a dominant phenotype of reduced male fertility to a plant comprising:
a. expressing in the plant a polypeptide of any of the embodiments of 33-55, thereby conferring the dominant phenotype of reduced male fertility to the plant.
61. A method of retaining a polypeptide to the endoplasmic reticulum (ER) in a plant cell, comprising:
a. expressing in the plant cell a nucleic acid of any of the embodiments of 1-
28, thereby retaining a polypeptide to the ER.
62. A method of retaining a polypeptide to the ER in a plant cell, comprising:
a. expressing in the plant a polypeptide of any of the embodiments of 33-55, thereby retaining a polypeptide to the ER.
63. A method of conferring a dominant phenotype to a plant comprising:
a. expressing in the plant a nucleic acid of any of the embodiments of 1-28, thereby conferring a dominant phenotype to the plant.
64. A method of conferring a dominant phenotype to a plant comprising:
a. expressing in the plant a polypeptide of any of the embodiments of 33-55, thereby conferring the dominant phenotype to the plant.
65. A fusion protein comprising a protein operably linked to a non-functional peptide signal, wherein the non-functional signal peptide is not processed, and wherein expression of the protein in a plant confers a dominant phenotype to the plant.
66. The fusion protein of embodiment 65, wherein the non-functional SP is encoded by any of the nucleic acids of embodiments of 1-28.
67. The fusion protein of embodiment 65, wherein the non-functional SP comprises any of the polypeptides of the embodiments of 33-55.
68. A nucleic acid encoding the fusion protein of any of the embodiments of 65-67.
69. An isolated construct or vector comprising the nucleic acid of any of the
embodiments of 68.
70. A cell transformed with the construct or vector of any of the embodiments of 69.
71. A plant having the nucleic acid of any of the embodiments of 68, wherein the plant has increased yield compared to the yield of a plant that does not express the nucleic acid of embodiment 68.
72. A plant having the nucleic acid of embodiment 68, wherein the plant has reduced male fertility compared to the male fertility of a plant that does not express the nucleic acid of embodiment 68.
73. A cell expressing any of the polypeptides of embodiments of 65-67.
74. A plant expressing any of the polypeptides of embodiments of 65-67, wherein the plant has increased yield compared to the yield of a plant that does not express any of the polypeptides of embodiments of 65-67. 75. A plant expressing any of the polypeptides of embodiments of 65-67, wherein the plant has reduced male fertility compared to the male fertility of a plant that does not express any of the polypeptides of embodiments of 65-67.
76. A method of method of conferring a dominant phenotype to a plant comprising: a. expressing a nucleic acid in a plant cell, wherein the nucleic acid is the nucleic acid of embodiment 68, thereby conferring a dominant phenotype to a plant.
77. A method of method of conferring a dominant phenotype to a plant comprising: a. expressing in the plant a fusion protein of any of the embodiments of 65- 67, thereby conferring the dominant phenotype of reduced male fertility to the plant.
78. A method of retaining a polypeptide to the ER in a in plant cell, comprising:
a. expressing a nucleic acid within a plant cell, wherein the nucleic acid is the nucleic acid of embodiment 68, thereby retaining the polypeptide in the ER.
79. A method of retaining a polypeptide to the ER in a plant cell, comprising:
a. expressing in the plant a fusion protein of any of the embodiments of 65- 67, thereby retaining the fusion protein in the ER.
80. A method of conferring a dominant phenotype to a plant comprising:
a. expressing in a non-anther plant tissue in the plant a nucleic acid of any of the embodiments of 1-28, thereby conferring a dominant phenotype to the plant.
81. A method of conferring a dominant phenotype to a plant comprising:
a. expressing in a non-anther plant tissue in the plant a polypeptide of any of the embodiments of 33-55, thereby conferring the dominant phenotype to the plant.
82. A method of conferring a dominant phenotype to a plant comprising:
a. expressing in a non-anther plant tissue in the plant a nucleic acid of
embodiment 68, thereby conferring a dominant phenotype to the plant.
83. A method of conferring a dominant phenotype to a plant comprising:
a. expressing in a non-anther plant tissue in the plant a polypeptide of any of the embodiments of 65-67, thereby conferring the dominant phenotype to the plant.
Example 21 . Dominant sterility using a non-functional signal peptide (SP), and restoration of fertility.
In another embodiment, the present disclosure also relates to another breeding pair of plants, including a first transgenic plant that is a male-sterile plant, and a second transgenic plant that is a restorer plant. The first transgenic male-sterile plant contains an expressible exogenous nucleic acid molecule comprising a polynucleotide sequence that encodes a polypeptide comprising or fused to a non-functional SP, wherein expression of the exogenous nucleic acid molecule confers a dominant phenotype of male sterility to the plant. Non-functional SP for use in individual plants, breeding pair of plants, and methods of the present disclosure are described elsewhere herein. The polynucleotide sequence that encodes a polypeptide comprising or fused to a non-functional SP of the first plant may be exogenous to the plant and can be operably linked to any suitable promoter for expression in male reproductive tissues, including but not limited to MS45, MS26, MS22, tassel-preferred, or 5126 gene promoters. Other suitable promoters are described elsewhere herein. In one embodiment, the
polynucleotide sequence that encodes a polypeptide comprising or fused to a nonfunctional SP includes but is not limited to a mutant MS44 gene, including SEQ ID NOs: 13, 15, and 152.
The restorer plant includes an expressible exogenous nucleic acid molecule having a polynucleotide that when expressed suppresses the expression of the polynucleotide sequence that encodes the polypeptide comprising or fused to a nonfunctional SP. Such suppression may be directed to said polynucleotide sequence or to its operably-linked promoter.
Useful expressible nucleic acid suppression molecules for this embodiment, include but are not limited to antisense, RNAi, and hairpin elements as described elsewhere herein and known to one of ordinary skill in the art.
In one embodiment, the second plant has the expressible exogenous nucleic acid molecule integrated in its genome. In another aspect, the suppression element in the second plant is operably linked to a promoter. Any promoter that expresses in the male reproductive tissues, including a constitutive promoter, may be used to drive the suppression element. Suitable promoters for expressing the suppression element include but are not limited to MS45, MS26, MS22, tassel-less or 5126 gene promoters.
The first and second plants are further characterized in that, when bred with each other, progeny are produced in which expression of the suppression element of the second plant suppresses expression of the polynucleotide sequence that encodes the polypeptide comprising or fused to the non-functional SP of the first plant, such that the progeny are male fertile. The present disclosure also provides methods of producing a male-fertile plant by crossing this pair of breeding plants.
Embodiments of this example include:
1. A breeding pair of plants, comprising: a first plant and a second plant, wherein the first plant expresses an exogenous nucleic acid molecule comprising a
polynucleotide sequence that encodes a polypeptide with a non-functional SP, wherein expression of the exogenous nucleic acid molecule confers a dominant phenotype of male sterility to the first plant so that the first plant is male-sterile, and (b) a second plant, wherein the second plant comprises an expressible exogenous nucleic acid molecule comprising a polynucleotide that when expressed suppresses the expression of the polynucleotide sequence that encodes the polypeptide with the non-functional SP or the promoter operatively linked to the polynucleotide sequence encoding the polypeptide with the non-functional SP of the first plant.
The pair of plants of embodiment 1 , wherein the non-functional SP is any of the nonfunctional SP's in embodiments 4 - 15 of Example 20.
The pair of plants of embodiment 1 , wherein, in the first plant, the polynucleotide sequence that encodes the polypeptide with a non-functional SP is operatively linked to a promoter.
The pair of plants of embodiment 3, wherein the promoter is an inducible promoter, a constitutive promoter, a tissue preferred promoter, a temporally regulated promoter or an element thereof.
The pair of plants of embodiment 3 wherein the promoter preferentially drives expression in male reproductive tissue.
The pair of plants of embodiment 5, wherein the male reproductive tissue preferred promoter is the MS45, MS26, MS22, or 5126 promoter.
The pair of plants of embodiment 3, wherein the promoter is a tassel-preferred promoter.
The pair of plants of embodiment 3, wherein the promoter is selected from the group consisting of:
a. SEQ ID NOS: 62 and 64-106, and the regulatory regions of SEQ ID NO: 9, and 13;
b. at least 100 contiguous nucleotides of SEQ ID NOS: 62 and 64-106, and the regulatory regions of SEQ ID NOS: 9 and 13; and
c. a sequence having at least 70% sequence identity to the full length of SEQ ID NOS: 9, 13, 62 and 64-106.
The pair of plants of embodiment 1 , wherein the exogenous nucleic acid molecule in the second plant is an RNAi, an antisense or hairpin suppression element.
The pair of plants of embodiment 1 , wherein the polynucleotide in the second plant that suppresses the expression of the exogenous nucleic acid molecule in the first plant is operably linked to a promoter.
The pair of plants of embodiment 10, wherein the promoter s an inducible promoter, a constitutive promoter, a tissue preferred promoter, a temporally regulated promoter or an element thereof.
The pair of plants of embodiment 10 wherein the promoter preferentially drives expression in male reproductive tissue.
The pair of plants of embodiment 10, wherein the male reproductive tissue-preferred promoter is MS45, MS26, MS22, or 5126.
The pair of plants of embodiment 10, wherein the promoter is a tassel-preferred promoter.
The pair of plants of embodiment 10, wherein the promoter is selected from the group consisting of:
d. SEQ ID NOS: 62 and 64-106, and the regulatory regions of SEQ ID NO: 9 and 13;
e. at least 100 contiguous nucleotides of SEQ ID NOS: 62 and 64-106, and of the regulatory regions of SEQ ID NO: 9 and 13; and
f. a sequence having at least 70% sequence identity to the full length of SEQ ID NOS: 62 and 64-106, and to the full length of the regulatory regions of SEQ ID NO: 9 and 13. The pair of plants of embodiment 1 , wherein the exogenous nucleic acid molecule in the second plant suppresses expression of the polynucleotide sequence of a MS44 mutant gene in the first plant.
The pair of plants of embodiment 16, wherein the MS44 mutant gene in the first plant is selected from the group consisting of SEQ ID NOs: 13, 15, and 152.
The pair of plants of embodiment 1 , wherein the exogenous nucleic acid molecule in the second plant is a hairpin suppression element that targets the promoter driving expression of a MS44 polynucleotide encoding a polypeptide with a non-functional SP in the first plant.
The pair of plants of embodiment 1 , wherein the first and second plants are wheat, maize, rice, sorghum, barley, rye, or sunflower.
A method of restoring male fertility in a plant exhibiting male sterility, comprising crossing the breeding pair of plants of embodiments 1-20.
A plant produced by the method of embodiment 20, wherein the plant is male-fertile Cells of the plant of embodiment 21.
Seed or progeny of the plant of embodiment 21 .
A method of producing hybrid plant seed, comprising crossing the breeding pair of plants of embodiments 1-20, wherein the first and second plants are plants of different inbred lines.
Seed produced by the method of embodiment 24.
A method of obtaining a hybrid plant, comprising growing the hybrid seed of embodiment 25
A hybrid plant produced by the method of embodiment 26.
The hybrid plant of claim 27, wherein said plant is male-sterile.
A breeding pair of plants, comprising: a first plant and a second plant, wherein the first plant expresses an exogenous nucleic acid molecule comprising a
polynucleotide sequence that encodes a polypeptide fused to a non-functional SP, wherein expression of the exogenous nucleic acid molecule confers a dominant phenotype of male sterility to the first plant so that the first plant is male-sterile, and (b) a second plant, wherein the second plant comprises an expressible exogenous nucleic acid molecule comprising a polynucleotide that when expressed suppresses the expression of nucleic acid molecule comprising a polynucleotide sequence that encodes a polypeptide fused to a non-functional SP or the promoter operatively linked to the nucleic acid molecule comprising a polynucleotide sequence that encodes a polypeptide fused to a non-functional SP of the first plant.
The pair of plants of embodiment 29, wherein the non-functional SP is any of the non-functional SP's in embodiments 4-15 of Example 20.
The pair of plants of embodiment 29, wherein, in the first plant, the polynucleotide sequence encoding the polypeptide fused to the non-functional SP is operatively linked to a promoter.
The pair of plants of embodiment 31 , wherein the promoter is an inducible promoter, a constitutive promoter, a tissue preferred promoter, a temporally regulated promoter or an element thereof.
The pair of plants of embodiment 31 , wherein the promoter preferentially drives expression in male reproductive tissue. The pair of plants of embodiment 31 , wherein the male reproductive tissue-preferred promoter is MS45, MS26, MS22, or 5126.
The pair of plants of embodiment 3, wherein the promoter is a regulatory region from SEQ ID: 9 or 13.
The pair of plants of embodiment 31 , wherein the promoter is a tassel-preferred promoter.
The pair of plants of embodiment 31 , wherein the promoter is selected from the group consisting of:
a. SEQ ID NOS: 62 and 64-106;
b. at least 100 contiguous nucleotides of SEQ ID NOS: 62 and 64-106 and of the regulatory regions of SEQ ID NO: 9 and 13; and
c. a sequence having at least 70% sequence identity to the full length of SEQ ID NOS: 62 and 64-106, and of the regulatory regions of SEQ ID NO: 9 and 13.
The pair of plants of embodiment 29, wherein the exogenous nucleic acid molecule in the second plant is an RNAi, an antisense or hairpin suppression element.
The pair of plants of embodiment 29, wherein the polynucleotide in the second plant that suppresses the expression of the exogenous nucleic acid molecule in the first plant is operably linked to a promoter.
The pair of plants of embodiment 39, wherein the promoter is an inducible promoter, a constitutive promoter, a tissue preferred promoter, a temporally regulated promoter or an element thereof.
The pair of plants of embodiment 39, wherein the promoter preferentially drives expression in male reproductive tissue.
The pair of plants of embodiment 39, wherein the male reproductive tissue-preferred is MS45, MS26, MS22, or 5126.
The pair of plants of embodiment 39, wherein the promoter is a tassel-preferred promoter.
The pair of plants of embodiment 39, wherein the promoter is selected from the group consisting of:
a. SEQ ID NOS: 62 and 64-106, and the regulatory regions of SEQ ID NO: 9 and 13;
b. at least 100 contiguous nucleotides of SEQ ID NOS: 62 and 64-106, and of the regulatory regions of SEQ ID NO: 9 and 13; and
c. a sequence having at least 70% sequence identity to the full length of SEQ ID NOS: 62 and 64-106, and of the regulatory regions of SEQ ID NO: 9 and 13.
The pair of plants of embodiment 29, wherein the exogenous nucleic acid molecule in the second plant suppresses expression of the polynucleotide sequence of a MS44 mutant gene in the first plant.
The pair of plants of embodiment 45, wherein the MS44 mutant gene in the first plant is selected from the group consisting of SEQ ID NOs: 13, 15, and 152.
The pair of plants of embodiment 29, wherein the exogenous nucleic acid molecule in the second plant is a hairpin suppression element that targets the promoter driving expression of a MS44 polynucleotide encoding a polypeptide fused to a nonfunctional SP in the first plant.
The pair of plants of embodiment 29, wherein the first and second plants are wheat, maize, rice, sorghum, barley, rye, or sunflower. 49. A method of restoring male fertility in a plant exhibiting male sterility, comprising crossing the breeding pair of plants of embodiments 29-48.
50. A plant produced by the method of embodiment 49, wherein said plant is male- fertile.
51. Cells of the plant of embodiment 50.
52. Seed or progeny of the plant of embodiment 50.
53. A method of producing hybrid plant seed, comprising crossing the breeding pair of plants of embodiments 29-48, wherein the first and second plants are plants of different inbred lines.
54. Seed produced by the method of embodiment 53.
55. A method of obtaining a hybrid plant, comprising growing the hybrid seed of
embodiment 54.
56. A hybrid plant produced by the method of embodiment 55.
All publications and patent applications in this specification are indicative of the level of ordinary skill in the art to which this disclosure pertains. All publications and patent applications are herein incorporated by reference to the same extent as if each individual publication or patent application was specifically and individually indicated by reference.
The disclosure has been described with reference to various specific and preferred embodiments and techniques. However, it should be understood that many variations and modifications may be made while remaining within the spirit and scope of the disclosure.

Claims

1. An isolated nucleic acid comprising a polynucleotide sequence encoding a polypeptide having a non-functional signal peptide (SP), wherein the non-functional signal peptide is not able to be processed and wherein expression of the nucleic acid in a plant confers a dominant phenotype to the plant.
2. The isolated nucleic acid of claim 1, wherein the polypeptide having a non-functional signal peptide is retained in the E .
3. The isolated nucleic acid of claim 1, wherein the non-functional signal peptide is rendered non-functional due to one or more insertions, deletions or substitutions of an amino acid residue in or near the signal peptide.
4. The isolated nucleic acid of claim 3, wherein the polypeptide having the non-functional signal peptide has one or more amino acid insertions, deletions or substitutions at position -10, -9, -8, -7, -6, -5, -4, -3, -2 -1, +1, +2, +3, +4, +5, +6, +7, +8, +9, +10 with respect to a signal peptide cleavage site, wherein the signal peptide cleavage site is located between positions -1 and +1.
5. The isolated nucleic acid of claim 1, wherein the nucleic acid comprises the nonfunctional signal peptide coding region of SEQ ID NO.:13, 15 or 152.
6. The isolated nucleic acid of claim 1, wherein the non-functional signal peptide is encoded by the polynucleotide sequence corresponding to positions 1222-1332 of SEQ ID NO.:13 and fragments thereof.
7. The isolated nucleic acid of claim 1, wherein the non-functional signal peptide is encoded by the polynucleotide sequence corresponding to positions 1-111 of SEQ ID NO.:15 and fragments thereof.
8. The isolated nucleic acid of claim 1, wherein the non-functional signal peptide is encoded by the polynucleotide sequence corresponding to positions 1-111 of SEQ ID NO.:152 and fragments thereof.
9. The isolated nucleic acid of claim 1, wherein nucleic acid comprises the sequence of SEQ
ID NO. 13, 15 or 152.
10. The isolated nucleic acid of claim 1, wherein the polypeptide comprises the nonfunctional signal peptide of SEQ ID NO: 14 or 153.
11. The isolated nucleic acid of claim 1, wherein the non-functional signal peptide comprises the amino acid sequence corresponding to positions 1-37 of SEQ ID NO.:14 and fragments thereof.
12. The isolated nucleic acid of claim 1, wherein the non-functional signal peptide comprises the amino acid sequence corresponding to positions 1-37 of SEQ ID NO.: 153 and fragments thereof.
13. The isolated nucleic acid of claim 1, wherein the polypeptide comprises the sequence of SEQ ID NO: 14 or 153.
14. The isolated nucleic acid of claim 1, wherein the non-functional signal peptide has a threonine or valine at position -1 relative to the signal peptide cleavage site.
15. The isolated nucleic acid of claim 1, wherein the non-functional signal peptide has a proline at position +1 relative to the signal peptide cleavage site.
16. The isolated nucleic acid of claim 1, wherein the dominant phenotype is reduced male fertility.
17. The isolated nucleic acid of claim 1, wherein plant is a crop plant.
18. The isolated nucleic acid of claim 1, wherein plant is a monocot or dicot plant.
19. The isolated nucleic acid of claim 1, wherein the polypeptide is a tassel-less (tlsl )
mutant.
20. The isolated nucleic acid of claim 1, wherein the polynucleotide encoding a polypeptide having a non-functional signal peptide is operably linked to a promoter.
21. The isolated nucleic acid of claim 20, wherein the promoter is an inducible promoter, a constitutive promoter, a tissue preferred promoter, a temporally regulated promoter, or an element thereof.
22. The isolated nucleic acid of claim 20, wherein the promoter preferentially drives
expression in male reproductive tissue.
23. The isolated nucleic acid of claim 20, wherein the promoter is a tassel-preferred
promoter.
24. The isolated nucleic acid of claim 20, wherein the promoter is selected from the group consisting of:
a. The regulatory region of SEQ ID NO: 9 or 13;
b. SEQ ID NOS: 62 and 64-106;
c. At least 100 contiguous nucleotides of the regulatory region of SEQ ID NO: 9 or 13;
d. At least 100 contiguous nucleotides of any of SEQ ID NOS: 62 and 64-106;
e. a sequence having at least 70% sequence identity to the full length of the regulatory region of SEQ I D NO: 9 or 13; and f. A sequence having at least 70% sequence identity to the full length of any of SEQ ID NOS: 62 and 64-106.
25. An isolated polypeptide having a non-functional signal peptide, wherein the nonfunctional signal peptide is not able to be processed and wherein expression of the polypeptide in a plant confers a dominant phenotype to the plant.
26. The polypeptide of claim 25, wherein the non-functional signal peptide is rendered nonfunctional due to one or more insertions, deletions or substitutions of an amino acid residue in or near the signal peptide.
27. The polypeptide of claim 26, wherein the non-functional signal peptide has one or more amino acid insertions, deletions or substitutions at position -10, -9, -8, -7, -6, -5, -4, -3, -2 -1, +1, +2, +3, +4, +5, +6, +7, +8, +9, +10 with respect to a signal peptide cleavage site in the signal peptide, wherein the signal peptide cleavage site is located between position -1 and +1.
28. The polypeptide of claim 25, wherein the polypeptide comprises the non-functional signal peptide of SEQ ID NO: 14 or 153.
29. The polypeptide of claim 25, wherein the non-functional SP comprises the amino acid sequence corresponding to positions 1-37 of SEQ ID NO.:14 and fragments thereof.
30. The polypeptide of claim 25, wherein the non-functional SP comprises the amino acid sequence corresponding to positions 1-37 of SEQ ID NO.:153 and fragments thereof.
31. The polypeptide of claim 25, wherein the polypeptide comprises the sequence of SEQ ID NO: 14 or 153.
32. The polypeptide of claim 25, wherein the non-functional signal peptide has a threonine or valine at position -1 relative to the signal peptide cleavage site.
33. The polypeptide of claim 25, wherein the non-functional signal peptide has a proline at position +1 relative to the signal peptide cleavage site.
34. The polypeptide of claim 25, wherein the dominant phenotype is reduced male fertility.
35. The polypeptide of claim 25, wherein plant is a crop plant.
36. The polypeptide of claim 25, wherein plant is a monocot or dicot plant.
37. A cell expressing the polypeptide of any of claims 25-33.
38. A plant expressing the polypeptide of any of claims 25-33, wherein the plant has
increased yield relative to a control.
39. A plant expressing the polypeptide of any of claims 25-33, wherein the plant has reduced male fertility compared to a control.
40. A method of conferring a dominant phenotype of reduced male fertility to a plant comprising expressing in the plant a nucleic acid of any of Claims 1-24, thereby conferring the dominant phenotype of reduced male fertility to the plant.
41. A method of conferring a dominant phenotype of reduced male fertility to a plant
comprising expressing in the plant a polypeptide of any of Claims 25-36, thereby conferring the dominant phenotype of reduced male fertility to the plant.
42. A method of retaining a polypeptide to the endoplasmic reticulum in a plant cell,
comprising expressing in the plant cell a nucleic acid of any of Claims 1-24, thereby retaining a polypeptide to the endoplasmic reticulum.
43. A method of conferring a dominant phenotype to a plant comprising expressing in the plant a nucleic acid of any of claims 1-28, thereby conferring a dominant phenotype to the plant.
44. A method of conferring a dominant phenotype to a plant comprising expressing in the plant a polypeptide of any of claims 25-33, thereby conferring the dominant phenotype to the plant.
45. A fusion protein comprising a protein operably linked to a non-functional peptide signal, wherein the non-functional signal peptide is not processed, and wherein expression of the protein in a plant confers a dominant phenotype to the plant.
46. The fusion protein of claim 45, wherein the non-functional signal peptide is encoded by any of the nucleic acids of claims 1-24.
47. The fusion protein of claim 45, wherein the non-functional signal peptide comprises any of the polypeptides of claims 25-36.
48. A nucleic acid encoding the fusion protein of any of claims 45-47.
49. A plant expressing any of the fusion proteins of claims 45-47, wherein the plant has reduced male fertility and/or increased yield relative to a control.
50. A method of conferring a dominant phenotype to a plant, comprising expressing in the plant a nucleic acid of claim 48.
51. An isolated polynucleotide comprising a nucleic acid selected from the group consisting of:
a. a nucleic acid having at least 90% identity to the full length of a nucleic acid of SEQ ID NO: 9, 13, 15, or 152;
b. a nucleic acid having at least 95% identity to the full length of a nucleic acid of SEQ ID NO: 9, 13, 15, or 152; c. a nucleic acid encoding a polypeptide at least 90% identical to the full length of a polypeptide of SEQ ID NO: 10, 14, 153, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, or 130; d. a nucleic acid encoding a polypeptide at least 95% identical to the full length of a polypeptide of SEQ ID NO: 10, 14, 153, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, or 130; e. a nucleic acid of SEQ ID NO: 9, 13, 15, or 152;
f. a nucleic acid having at least 90% identity to the full length of a nucleic acid of SEQ ID NO: 9, 13, 15, or 152, and encoding an amino acid other than alanine or glycine at the position corresponding to amino acid 37 of SEQ ID NO: 10, when said nucleic acid is aligned with SEQ ID NO: 10;
g. a nucleic acid having at least 90% identity to the full length of a nucleic acid of SEQ ID NO: 9, 13, 15, or 152, and encoding an amino acid of threonine or valine at the position corresponding to amino acid 37 of SEQ ID NO: 10, when said nucleic acid is aligned with SEQ ID NO: 10; and
h. the complement of any nucleic acid of a, b, c, d, e, f, or g.
52. An expression cassette comprising a polynucleotide of Claim 51 operably linked to a promoter.
53. An expression cassette of Claim 52 wherein the promoter is a constitutive, tissue- preferred or inducible promoter.
54. The expression cassette of Claim 53 wherein the tissue-preferred promoter preferentially drives expression in male reproductive tissue.
55. The expression cassette of Claim 51 wherein the promoter is the MS45 or 5126 or MS44 promoter isolated from maize, wheat, or rice.
56. A plant comprising the expression cassette of any of Claims 52-55.
57. A plant cell comprising the expression cassette of any of Claims 52-55.
58. A seed from a plant of Claim 56, wherein the seed comprises the expression cassette.
59. A plant of Claim 56, wherein the plant is maize.
60. An isolated polypeptide comprising an amino acid sequence selected from the group consisting of:
a. An amino acid sequence comprising SEQ ID NO: 10, 14, 153, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, or 130; b. An amino acid sequence at least 90% identical to the full length of a polypeptide of SEQ ID NO: 10, 14, 153, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, or 130;
c. An amino acid sequence at least 95% identical to the full length of a polypeptide of SEQ ID NO: 10, 14, 153, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, or 130;
d. An amino acid sequence comprising the mature protein encoded by SEQ ID NO:
10, 14, 153, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, or 130;
e. An amino acid sequence at least 90% identical to the full length of the mature protein encoded by SEQ ID NO: 10, 14, 153, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, or 130; and
f. An amino acid sequence at least 95% identical to the full length of the mature protein encoded by SEQ ID NO: 10, 14, 153, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, or 130.
61. An isolated polypeptide of Claim 60 wherein the amino acid corresponding to position 37 of SEQ ID NO: 10 is threonine or valine.
62. An isolated polypeptide of Claim 60 wherein the amino acid corresponding to position 37 of SEQ ID NO: 10 is not alanine or glycine.
63. A transgenic maintainer line for propagating plants which are homozygous for a nuclear gene or construct conferring dominant male sterility, wherein the transgenic maintainer line is homozygous for said nuclear gene or construct conferring dominant male sterility, and wherein said transgenic maintainer line is homozygous for a SAM construct, wherein the SAM construct comprises an element that suppresses the dominant male sterility gene or construct, a second element that disrupts pollen function, and optionally a third element which is a marker, wherein functional pollen produced by the transgenic maintainer line lack said SAM construct.
64. A method for creating a transgenic maintainer line for propagating plants which are homozygous for a nuclear gene or construct conferring dominant male sterility, the method comprising: a. transforming a plant heterozygous for said nuclear gene or construct with a SAM construct, wherein said SAM construct comprises:
i. an element that suppresses the dominant male sterility gene or construct,
ii. a second element that disrupts pollen function,
iii. and optionally a third element which is a marker;
b. self-pollinating said transformed plant to produce seed;
c. selecting those seed homozygous for the dominant male sterility gene and which comprise the SAM construct; and
d. planting the selected seed to produce a transgenic maintainer line.
65. A method for creating a male-sterile line of plants homozygous for a nuclear gene or construct conferring dominant male-sterility, the method comprising:
a. Self-pollinating the transgenic maintainer line of claim 63; and
b. Genotyping the seed produced in part a, or plants grown therefrom, to select those seeds or plants which are homozygous for the gene or construct conferring dominant male sterility and which lack the SAM construct of claim 63;
wherein the selected seeds or plants comprise the male-sterile line.
66. A method for increasing a male-sterile line of plants homozygous for a nuclear gene conferring dominant male sterility, the method comprising:
a. Growing the selected seeds or plants of Claim 65, to produce male-sterile plants; b. Pollinating said male-sterile plants with the transgenic maintainer line of Claim 63; and
c. Harvesting seed from the male-sterile plants
wherein the harvested seed is homozygous for the nuclear gene or construct conferring dominant male sterility.
67. A method for creating hybrid plants heterozygous for a nuclear gene or construct conferring dominant male sterility, the method comprising
a. Planting the harvested seed of claim 66 to grow male-sterile plants, wherein the plants are a male-sterile inbred line;
b. Pollinating said male-sterile inbred plants with a second inbred which is male- fertile;
c. Harvesting hybrid seed from said male-sterile inbred plants; d. Planting said harvested seed to produce hybrid plants which are heterozygous for the gene or construct conferring dominant male sterility.
68. Hybrid seed produced by steps a through c of the method of Claim 67.
69. A method for producing male-sterile hybrid seed, the method comprising
a. Transforming a male-sterile line heterozygous for dominant male-sterility with a gene construct comprising:
i. An element that suppresses the dominant male sterility; ii. A second element that disrupts pollen function;
iii. An optional third element which is a marker
and expressing said elements in the inbred line to render the plants male fertile;
b. Self-pollinating the male-fertile plants;
c. Producing homozygous progeny for dominant male sterility;
d. Identifying those seeds having the homozygous dominant male sterility genotype and comprising the construct as the maintainer line;
e. Identifying those seeds having the homozygous dominant male sterility genotype and lacking the construct as the dominant-male-sterile inbred line;
f. Pollinating the male-sterile inbred line with pollen of the maintainer line to increase the dominant-male-sterile inbred line;
g. Pollinating the male-sterile inbred line with pollen of a second inbred line, to produce male-sterile hybrid seed heterozygous for dominant male sterility.
70. The method of Claim 64 or Claim 69 wherein the element that suppresses the dominant male sterility is selected from the group consisting of antisense, NAi, and hairpin molecules directed to the nuclear gene or construct conferring dominant male sterility.
71. The transgenic maintainer line of Claim 63 wherein the element that suppresses the dominant male sterility is selected from the group consisting of antisense, RNAi, and hairpin molecules directed to the nuclear gene or construct conferring male sterility.
72. The transgenic maintainer line of Claim 71, wherein the suppression element is a promoter inverted repeat targeting the promoter driving the nuclear gene conferring male sterility.
73. The transgenic maintainer line of Claim 72, wherein the suppression element is a promoter inverted repeat targeting the MS44 promoter.
74. The transgenic maintainer line of any of Claims 63 and 71-73 wherein the element that disrupts pollen function encodes a protein which interferes with the normal accumulation of starch in pollen or affects osmotic balance within pollen.
75. The transgenic maintainer line of Claim 74 wherein the element that disrupts pollen function encodes an alpha-amylase, beta-amylase, or debranching enzyme.
76. The transgenic maintainer line of any of Claims 63 and 71-73 wherein the element that disrupts pollen function is selected from the group consisting of genes which express a product cytotoxic to male gametes or otherwise inhibit male gamete development.
77. The transgenic maintainer line of Clam 76, wherein the element that disrupts pollen function is DAM-methylase, barnase, or streptavidin.
78. The transgenic maintainer line of any of Claims 63 and 71-77 wherein the SAM construct comprises a marker expressed in seed tissue.
79. The transgenic maintainer line of any of Claims 63 and 71-78 wherein the marker is expressed in endosperm, aleurone, or pericarp tissue.
80. The transgenic maintainer line of any of Claims 63 and 71-79 wherein the marker is a fluorescent protein.
81. The transgenic maintainer line of any of Claims 63 and 71-80 wherein the marker is a fluorescent red protein.
82. A method of producing grain on male-sterile hybrid plants, the method comprising
a. growing male-sterile hybrid plants from the seed produced by the method of
Claim 68 or 69;
b. providing pollinator plants to fertilize the male-sterile hybrid plants;
c. Allowing pollen from the male-fertile plants to pollinate the male-sterile hybrid plants; and
d. Harvesting grain from the male-sterile hybrid plants.
83. The method of claim 82, further comprising
a. mixing with the hybrid seed produced by the method of Claim 68 or 69, seed which will grow into pollinator plants;
b. Planting the seed mixture;
c. Allowing pollen from the male-fertile plants to pollinate the male-sterile hybrid plants; and
d. Harvesting grain from the male-sterile plants.
84. The method of any of Claims 64, 65, 66, 67, 69, 82 or 83, wherein said plants are maize plants.
85. The method of Claim 82 wherein the method is practiced under conditions of environmental stress.
86. The transgenic maintainer line of any of Claims 63 and 71-81, wherein said nuclear gene or construct conferring dominant male sterility is selected from the group consisting of: a. An isolated nucleic acid comprising a polynucleotide sequence encoding a polypeptide comprising a non-functional signal peptide, wherein expression of the nucleic acid in a plant confers a dominant male-sterile phenotype to the plant;
b. An isolated nucleic acid of SEQ I D NO: 13, 15, or 152;
c. Barnase, streptavidin, DAM, MS41, or MS42.
87. A breeding pair of plants, comprising: a first plant and a second plant, wherein the first plant expresses an exogenous nucleic acid molecule comprising a polynucleotide sequence that encodes a polypeptide with a non-functional signal peptide sequence, wherein expression of the exogenous nucleic acid molecule confers a dominant phenotype of male sterility to the first plant so that the first plant is male-sterile, and (b) a second plant, wherein the second plant comprises an expressible exogenous nucleic acid molecule comprising a polynucleotide that when expressed suppresses the expression of the polynucleotide sequence that encodes the polypeptide with the nonfunctional signal peptide sequence or the promoter operatively linked to the polynucleotide sequence encoding the polypeptide with the non-functional signal peptide of the first plant.
88. The pair of plants of claim 87, wherein, in the first plant, the polynucleotide sequence that encodes the polypeptide with a non-functional signal peptide sequence is operatively linked to a promoter.
89. The pair of plants of claim 88 wherein the promoter is an inducible promoter, a
constitutive promoter, a tissue preferred promoter, a temporally regulated promoter or an element thereof.
90. The pair of plants of claim 89 wherein the promoter preferentially drives expression in male reproductive tissue.
91. The pair of plants of claim 90, wherein the male reproductive tissue preferred promoter is the MS45, MS26, MS22, or 5126 promoter.
92. The pair of plants of claim 88, wherein the promoter is selected from the group consisting of:
g. SEQ ID NOS: 62 and 64-106, and the regulatory regions of SEQ ID NO: 9 and 13; h. at least 100 contiguous nucleotides of SEQ ID NOS: 9, 13, 62 and 64-106; and i. a sequence having at least 70% sequence identity to the full length of SEQ ID NOS: 9, 13, 62 and 64-106.
93. The pair of plants of claim 87, wherein the exogenous nucleic acid molecule in the
second plant suppresses expression of the polynucleotide sequence of a MS44 mutant gene in the first plant.
94. The pair of plants of claim 93, wherein the MS44 mutant gene in the first plant is
selected from the group consisting of SEQ ID NOs: 13, 15, and 152.
95. The pair of plants of claim 87, wherein the exogenous nucleic acid molecule in the
second plant is a hairpin suppression element that targets the promoter driving expression of a MS44 polynucleotide encoding a polypeptide with a non-functional SP in the first plant.
96. A breeding pair of plants, comprising: a first plant and a second plant, wherein the first plant expresses an exogenous nucleic acid molecule comprising a polynucleotide sequence that encodes a polypeptide fused to a non-functional SP, wherein expression of the exogenous nucleic acid molecule confers a dominant phenotype of male sterility to the first plant so that the first plant is male-sterile, and (b) a second plant, wherein the second plant comprises an expressible exogenous nucleic acid molecule comprising a polynucleotide that when expressed suppresses the expression of nucleic acid molecule comprising a polynucleotide sequence that encodes a polypeptide fused to a nonfunctional SP or the promoter operatively linked to the nucleic acid molecule comprising a polynucleotide sequence that encodes a polypeptide fused to a non-functional SP of the first plant.
97. The pair of plants of claim 96, wherein, in the first plant, the polynucleotide encoding the polypeptide fused to the non-functional signal peptide is operably linked to an inducible promoter, a constitutive promoter, a tissue preferred promoter, a temporally regulated promoter or an element thereof.
98. The pair of plants of claim 97, wherein the tissue-preferred promoter is MS45, MS26, MS22, or 5126.
99. The pair of plants of claim 96, wherein the exogenous nucleic acid molecule in the second plant suppresses expression of the polynucleotide sequence of a MS44 mutant gene in the first plant.
100. The pair of plants of claim 99, wherein the MS44 mutant gene in the first plant is selected from the group consisting of SEQ I D NOs: 13, 15, and 152.
101. A method of reducing pollen produced by a crop species in a grain-production field, the method comprising growing in said field a blend of male-fertility phenotypes, wherein the majority of the plants are male-sterile, wherein total pollen flow for the field is less than total pollen flow for a field in which the majority of the plants are male- fertile.
102. The method of claim 101 wherein the male-sterile plants are hybrids.
103. The method of claim 101 or 102 wherein the male-sterility is conferred by a
dominant gene or construct.
104. The method of claim 103 wherein the dominant male-sterility gene is an allele of MS44.
105. The method of any of claims 101-104 wherein the crop species is maize or
sorghum.
106. The method of any of Claims 101-105, wherein outcrossing to weedy species is reduced, relative to a field in which the majority of the plants are male-fertile.
107. A method for reducing the risk of adventitious presence of a transgene in a
product of a crop species, wherein the method comprises maintaining the transgene in a male-sterile line of the crop.
108. The method of claim 107 wherein the transgene is present in a male-sterile
hybrid of the crop species.
109. The method of Claim 107 or 108 wherein the product is grain.
110. The method of any of Claims 107-109 wherein the crop species is maize or
sorghum or sunflower.
111. The method of any of Claims 107-109 wherein the transgene is linked to a gene or construct conferring dominant male sterility.
112. The method of claim 111 wherein the gene conferring dominant male sterility is an allele of MS44.
113. The method of Claim 111 or 112 wherein the transgene encodes an enzyme
which improves digestibility of cellulose.
PCT/US2013/030455 2012-03-13 2013-03-12 Genetic reduction of male fertility in plants WO2013138309A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201261610274P 2012-03-13 2012-03-13
US61/610,274 2012-03-13

Publications (1)

Publication Number Publication Date
WO2013138309A1 true WO2013138309A1 (en) 2013-09-19

Family

ID=47902383

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/030455 WO2013138309A1 (en) 2012-03-13 2013-03-12 Genetic reduction of male fertility in plants

Country Status (1)

Country Link
WO (1) WO2013138309A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE102016106656A1 (en) 2016-04-12 2017-10-12 Kws Saat Se Nuclear encoded male sterility by mutation in cytochrome P450 oxidase
DE102016015741A1 (en) 2016-04-12 2017-11-30 Kws Saat Se Nuclear encoded male sterility by mutation in cytochrome P450 oxidase
CN107667853A (en) * 2017-06-07 2018-02-09 湖南杂交水稻研究中心 The method for creating of the common line with genic sterile of rice and application
US10113181B2 (en) 2013-03-12 2018-10-30 Pioneer Hi-Bred International, Inc. Manipulation of dominant male sterility
US11124797B1 (en) * 2015-06-04 2021-09-21 Shandong Agricultural University Wheat male-sterility gene WMS and its anther-specific expression promoter and uses thereof
CN116724886A (en) * 2023-08-08 2023-09-12 四川天能璟秀生物科技有限公司 Method for propagating corn cell nucleus male sterile line by color marking

Citations (127)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US39247A (en) 1863-07-14 Improvement in skates
US897360A (en) 1908-03-04 1908-09-01 Nathan Fein Buckle.
US4658082A (en) 1984-07-25 1987-04-14 Atlantic Richfield Company Method for producing intact plants containing foreign DNA
US4918006A (en) 1985-07-01 1990-04-17 E. I. Du Pont De Nemours And Company Gene coding for insecticidal crystal protein
US4945050A (en) 1984-11-13 1990-07-31 Cornell Research Foundation, Inc. Method for transporting substances into living cells and tissues and apparatus therefor
US4987071A (en) 1986-12-03 1991-01-22 University Patents, Inc. RNA ribozyme polymerases, dephosphorylases, restriction endoribonucleases and methods
US5034323A (en) 1989-03-30 1991-07-23 Dna Plant Technology Corporation Genetic engineering of novel plant phenotypes
WO1991010725A1 (en) 1990-01-22 1991-07-25 Dekalb Plant Genetics Fertile transgenic corn plants
EP0465024A1 (en) * 1990-06-12 1992-01-08 Pioneer Hi-Bred International, Inc. Control of microsporogenesis using externally inducible promoter sequences
WO1992018625A1 (en) * 1991-04-16 1992-10-29 Mogen International N.V. Male-sterile plants, method for obtaining male-sterile plants and recombinant dna for use therein
WO1993002197A1 (en) * 1991-07-23 1993-02-04 Nickerson Biocem Limited CALLASE-RELATED DNAs AND THEIR USE IN ARTIFICIAL MALE STERILITY
US5262306A (en) 1989-09-26 1993-11-16 Robeson David J Methods for identifying cercosporin-degrading microorganisms
US5268463A (en) 1986-11-11 1993-12-07 Jefferson Richard A Plant promoter α-glucuronidase gene construct
US5283184A (en) 1989-03-30 1994-02-01 Dna Plant Technology Corporation Genetic engineering of novel plant phenotypes
WO1994011516A1 (en) 1992-11-17 1994-05-26 E.I. Du Pont De Nemours And Company Genes for microsomal delta-12 fatty acid desaturases and related enzymes from plants
EP0604662A1 (en) 1992-07-07 1994-07-06 Japan Tobacco Inc. Method of transforming monocotyledon
US5332808A (en) 1992-09-08 1994-07-26 North Carolina State University DNA encoding a ribosome inactivating protein
US5341001A (en) 1992-02-13 1994-08-23 Matsushita Electric Industrial Co., Ltd. Sulfide-selenide manganese-zinc mixed crystal photo semiconductor and laser diode
US5366892A (en) 1991-01-16 1994-11-22 Mycogen Corporation Gene encoding a coleopteran-active toxin
US5412085A (en) 1992-07-09 1995-05-02 Pioneer Hi-Bred International Inc. Pollen-specific promoter from maize
EP0672752A1 (en) 1993-09-03 1995-09-20 Japan Tobacco Inc. Method of transforming monocotyledon by using scutellum of immature embryo
US5463175A (en) 1990-06-25 1995-10-31 Monsanto Company Glyphosate tolerant plants
US5470359A (en) 1994-04-21 1995-11-28 Pioneer Hi-Bred Internation, Inc. Regulatory element conferring tapetum specificity
US5478369A (en) 1990-06-12 1995-12-26 Pioneer Hi-Bred International, Inc. Nucleotide sequences mediating male fertility and method of using same
US5489520A (en) 1990-04-17 1996-02-06 Dekalb Genetics Corporation Process of producing fertile transgenic zea mays plants and progeny comprising a gene encoding phosphinothricin acetyl transferase
US5561236A (en) 1986-03-11 1996-10-01 Plant Genetic Systems Genetically engineered plant cells and plants exhibiting resistance to glutamine synthetase inhibitors, DNA fragments and recombinants for use in the production of said cells and plants
WO1996030530A1 (en) 1995-03-24 1996-10-03 Pioneer Hi-Bred International, Inc. Als3 promoter
US5565350A (en) 1993-12-09 1996-10-15 Thomas Jefferson University Compounds and methods for site directed mutations in eukaryotic cells
US5583210A (en) 1993-03-18 1996-12-10 Pioneer Hi-Bred International, Inc. Methods and compositions for controlling plant development
US5589610A (en) 1991-02-07 1996-12-31 Plant Genetic Systems, N.V. Stamen-specific promoters from corn
US5589379A (en) 1993-02-26 1996-12-31 Calgene, Inc. Geminivirus-based gene expression system
US5593881A (en) 1994-05-06 1997-01-14 Mycogen Corporation Bacillus thuringiensis delta-endotoxin
US5602321A (en) 1992-11-20 1997-02-11 Monsanto Company Transgenic cotton plants producing heterologous polyhydroxy(e) butyrate bioplastic
US5608144A (en) 1994-08-12 1997-03-04 Dna Plant Technology Corp. Plant group 2 promoters and uses thereof
US5633441A (en) 1989-08-04 1997-05-27 Plant Genetic Systems, N.V. Plants with genetic female sterility
US5641876A (en) 1990-01-05 1997-06-24 Cornell Research Foundation, Inc. Rice actin gene and promoter
US5683439A (en) 1993-10-20 1997-11-04 Hollister Incorporated Post-operative thermal blanket
US5689049A (en) 1994-12-08 1997-11-18 Pioneer Hi-Bred International, Inc. Transgenic plant and method for producing male sterility using anther specific promoter 5126
US5693512A (en) 1996-03-01 1997-12-02 The Ohio State Research Foundation Method for transforming plant tissue by sonication
US5703409A (en) 1993-12-21 1997-12-30 Fujitsu Limited Error counting circuit
US5703049A (en) 1996-02-29 1997-12-30 Pioneer Hi-Bred Int'l, Inc. High methionine derivatives of α-hordothionin for pathogen-control
US5723756A (en) 1990-04-26 1998-03-03 Plant Genetic Systems, N.V. Bacillus thuringiensis strains and their genes encoding insecticidal toxins
US5728558A (en) 1988-02-03 1998-03-17 Pioneer Hi-Bred International, Inc. Molecular methods of hybrid seed production
US5728926A (en) 1988-02-03 1998-03-17 Pioneer Hi-Bred International, Inc. Antisense gene systems of pollination control for hybrid seed production
US5731181A (en) 1996-06-17 1998-03-24 Thomas Jefferson University Chimeric mutational vectors having non-natural nucleotides
US5736514A (en) 1994-10-14 1998-04-07 Nissan Chemical Industries, Ltd. Bacillus strain and harmful organism controlling agents
US5737514A (en) 1995-11-29 1998-04-07 Texas Micro, Inc. Remote checkpoint memory system and protocol for fault-tolerant computer system
US5736369A (en) 1994-07-29 1998-04-07 Pioneer Hi-Bred International, Inc. Method for producing transgenic cereal plants
US5747450A (en) 1991-08-02 1998-05-05 Kubota Corporation Microorganism and insecticide
WO1998020133A2 (en) 1996-11-01 1998-05-14 Pioneer Hi-Bred International, Inc. Proteins with enhanced levels of essential amino acids
WO1998020122A1 (en) 1996-11-01 1998-05-14 The Institute Of Physical And Chemical Research METHOD FOR FORMING FULL-LENGTH cDNA LIBRARY
US5759829A (en) 1986-03-28 1998-06-02 Calgene, Inc. Antisense regulation of gene expression in plant cells
US5760012A (en) 1996-05-01 1998-06-02 Thomas Jefferson University Methods and compounds for curing diseases caused by mutations
US5767373A (en) 1994-06-16 1998-06-16 Novartis Finance Corporation Manipulation of protoporphyrinogen oxidase enzyme activity in eukaryotic organisms
US5792931A (en) 1994-08-12 1998-08-11 Pioneer Hi-Bred International, Inc. Fumonisin detoxification compositions and methods
US5792852A (en) 1992-11-16 1998-08-11 Cancer Research Fund Of Contra Costa Polynucleotides encoding modified antibodies with human milk fat globule specificity
WO1998049350A1 (en) 1997-04-30 1998-11-05 Regents Of The University Of Minnesota In vivo use of recombinagenic oligonucleobases to correct genetic lesions in hepatocytes
US5837850A (en) 1994-04-21 1998-11-17 Pioneer Hi-Bred International, Inc. Regulatory element conferring tapetum specificity
US5837851A (en) 1994-12-08 1998-11-17 Pioneer Hi-Bred International, Inc. DNA promoter 5126 and constructs useful in a reversible nuclear genetic system for male sterility in transgenic plants
WO1998053083A1 (en) 1997-05-21 1998-11-26 Zeneca Limited Gene silencing
US5850016A (en) 1996-03-20 1998-12-15 Pioneer Hi-Bred International, Inc. Alteration of amino acid compositions in seeds
WO1999007865A1 (en) 1997-08-05 1999-02-18 Kimeragen, Inc. The use of mixed duplex oligonucleotides to effect localized genetic changes in plants
US5879903A (en) 1986-08-23 1999-03-09 Hoechst Aktiengesellschaft Phosphinothricin-resistance gene, and its use
US5885801A (en) 1995-06-02 1999-03-23 Pioneer Hi-Bred International, Inc. High threonine derivatives of α-hordothionin
US5885802A (en) 1995-06-02 1999-03-23 Pioneer Hi-Bred International, Inc. High methionine derivatives of α-hordothionin
WO1999025821A1 (en) 1997-11-18 1999-05-27 Pioneer Hi-Bred International, Inc. Compositions and methods for genetic modification of plants
US5942657A (en) 1992-05-13 1999-08-24 Zeneca Limited Co-ordinated inhibition of plant gene expression
WO1999043838A1 (en) 1998-02-24 1999-09-02 Pioneer Hi-Bred International, Inc. Synthetic promoters
US5952544A (en) 1991-12-04 1999-09-14 E. I. Du Pont De Nemours And Company Fatty acid desaturase genes from plants
WO1999049029A1 (en) 1998-03-20 1999-09-30 Benitec Australia Ltd Control of gene expression
US5962769A (en) 1995-06-07 1999-10-05 Pioneer Hi-Bred International, Inc. Induction of male sterility in plants by expression of high levels of avidin
US5962764A (en) 1994-06-17 1999-10-05 Pioneer Hi-Bred International, Inc. Functional characterization of genes
WO1999053050A1 (en) 1998-04-08 1999-10-21 Commonwealth Scientific And Industrial Research Organisation Methods and means for obtaining modified phenotypes
US5981840A (en) 1997-01-24 1999-11-09 Pioneer Hi-Bred International, Inc. Methods for agrobacterium-mediated transformation
US5990389A (en) 1993-01-13 1999-11-23 Pioneer Hi-Bred International, Inc. High lysine derivatives of α-hordothionin
WO1999061631A1 (en) 1998-05-26 1999-12-02 Novartis Ag Dsrna-mediated regulation of gene expression in plants
WO1999061619A2 (en) 1998-05-22 1999-12-02 Pioneer Hi-Bred International, Inc. Cell cycle genes, proteins and uses thereof
US6013859A (en) 1994-07-14 2000-01-11 Pioneer Hi-Bred International, Inc. Molecular methods of hybrid seed production
WO2000011177A1 (en) 1998-08-20 2000-03-02 Pioneer Hi-Bred International, Inc. Seed-preferred promoters
WO2000012733A1 (en) 1998-08-28 2000-03-09 Pioneer Hi-Bred International, Inc. Seed-preferred promoters from end genes
US6037523A (en) 1997-06-23 2000-03-14 Pioneer Hi-Bred International Male tissue-preferred regulatory region and method of using same
WO2000017364A2 (en) 1998-09-23 2000-03-30 Pioneer Hi-Bred International, Inc. Cyclin d polynucleotides, polypeptides and uses thereof
WO2000049035A1 (en) 1999-02-19 2000-08-24 The General Hospital Corporation Gene silencing
WO2000070067A1 (en) 1999-05-14 2000-11-23 Dekalb Genetics Corporation The rice actin 2 promoter and intron and methods for use thereof
US6162964A (en) 1988-02-03 2000-12-19 Pioneer Hi-Bred International, Inc. Molecular methods of hybrid seed production
WO2001000834A1 (en) * 1999-06-29 2001-01-04 Pioneer Hi-Bred International, Inc. Gene affecting male fertility in plants
US6177616B1 (en) 1996-12-16 2001-01-23 Hoecsht Schering Agrevo Gmbh Genes coding for amino acid deacetylases with specificity for N-acetyl-L-phosphinothricin, their isolation and their use
US6177611B1 (en) 1998-02-26 2001-01-23 Pioneer Hi-Bred International, Inc. Maize promoters
US6184439B1 (en) 1988-02-03 2001-02-06 Pioneer Hi Bred International, Inc. Antisense gene systems of pollination control for hybrid seed production
WO2001012825A1 (en) 1999-08-13 2001-02-22 Syngenta Participations Ag Herbicide-tolerant protoporphyrinogen oxidase
US6232529B1 (en) 1996-11-20 2001-05-15 Pioneer Hi-Bred International, Inc. Methods of producing high-oil seed by modification of starch levels
US6297426B1 (en) 1990-06-12 2001-10-02 Pioneer Hi-Bred International, Inc. Methods of mediating female fertility in plants
US6300543B1 (en) 1996-07-08 2001-10-09 Pioneer Hi-Bred International, Inc. Transformation of zygote, egg or sperm cells and recovery of transformed plants from isolated embryo sacs
WO2002000904A2 (en) 2000-06-23 2002-01-03 E. I. Du Pont De Nemours And Company Recombinant constructs and their use in reducing gene expression
WO2002026789A2 (en) 2000-09-26 2002-04-04 Pioneer Hi-Bred International, Inc. Nucleotide sequences mediating male fertility and method of using same
US6372967B1 (en) 1988-04-28 2002-04-16 Aventis Cropscience N.V. Plants with modified stamen cells
US20020048814A1 (en) 2000-08-15 2002-04-25 Dna Plant Technology Corporation Methods of gene silencing using poly-dT sequences
WO2002063021A2 (en) 2001-02-08 2002-08-15 Pioneer Hi-Bred International, Inc. Nucleotide sequence mediating male fertility and method of using same
US6453242B1 (en) 1999-01-12 2002-09-17 Sangamo Biosciences, Inc. Selection of sites for targeting by zinc finger proteins and methods of designing zinc finger proteins to bind to preselected sites
US6452069B1 (en) 1999-03-16 2002-09-17 Pioneer Hi-Bred International, Inc. SF3 promoter and methods of use
US6504082B1 (en) 1998-09-10 2003-01-07 Pioneer Hi-Bred International, Inc. Ecdysone receptors and methods for their use
US20030037355A1 (en) 2000-01-21 2003-02-20 Barbas Carlos F. Methods and compositions to modulate expression in plants
US20030074698A1 (en) 2000-06-16 2003-04-17 Thomas Schmulling Method for modifying plant morphology, biochemistry and physiology
US6566587B1 (en) 1995-07-19 2003-05-20 Bayer Cropscience S.A. Mutated 5-enolpyruvylshikimate-3-phosphate synthase, gene coding for said protein and transformed plants containing said gene
US20030180945A1 (en) 2002-03-14 2003-09-25 Ming-Bo Wang Modified gene-silencing RNA and uses thereof
US6646805B2 (en) 2001-03-02 2003-11-11 Fujitsu Limited Apparatus for variable wavelength dispersion and wavelength dispersion slope
US20030217393A1 (en) 2002-02-15 2003-11-20 Pioneer Hi-Bred International, Inc. Development of a stress-responsive promoter from maize
WO2003102198A1 (en) 2002-06-04 2003-12-11 Metanomics Gmbh & Co. Kgaa Method for the stable expression of nucleic acids in transgenic plants, controlled by a parsley-ubiquitin promoter
US6750868B2 (en) 2001-09-03 2004-06-15 Via Technologies, Inc. Universal accelerated graphic port system and method for operating the same
US7022896B1 (en) 1997-04-04 2006-04-04 Board Of Regents Of University Of Nebraska Methods and materials for making and using transgenic dicamba-degrading organisms
US7087812B1 (en) 1999-09-30 2006-08-08 Japan Tobacco Inc. Vectors for transforming plants
WO2007053482A2 (en) 2005-10-28 2007-05-10 Dow Agrosciences Llc Novel herbicide resistance genes
US20070277269A1 (en) * 2006-04-17 2007-11-29 Ceres, Inc. Nucleotide sequences and polypeptides encoded thereby useful for modifying plant characteristics
WO2007146706A2 (en) 2006-06-06 2007-12-21 Monsanto Technology Llc Modified dicamba monooxygenase enzyme and methods of its use
US7462481B2 (en) 2000-10-30 2008-12-09 Verdia, Inc. Glyphosate N-acetyltransferase (GAT) genes
US20090055976A1 (en) 2000-12-07 2009-02-26 Syngenta Limited Methods for the production of plants resistant to HPPD herbicides
WO2009114321A2 (en) 2008-03-11 2009-09-17 Precision Biosciencs, Inc. Rationally-designed meganucleases for maize genome engineering
US7622641B2 (en) 2005-08-24 2009-11-24 Pioneer Hi-Bred Int'l., Inc. Methods and compositions for providing tolerance to multiple herbicides
US7696405B2 (en) 2003-12-16 2010-04-13 Pioneer Hi-Bred International, Inc. Dominant gene suppression transgenes and methods of using same
US7709709B1 (en) 2009-03-25 2010-05-04 Monsanto Technology Llc Plants and seeds of corn variety CV334995
US7838733B2 (en) 2004-04-30 2010-11-23 Dow Agrosciences Llc Herbicide resistance genes
US20110023180A1 (en) 2009-01-22 2011-01-27 Syngenta Participations Ag Novel hydroxyphenylpyruvate dioxygenase polypeptides and methods of use
US7893317B2 (en) 2000-09-26 2011-02-22 Pioneer Hi-Bred International, Inc Nucleotide sequences mediating plant male fertility and method of using same
US7935869B2 (en) 1996-07-16 2011-05-03 Bayer S.A.S. Chimeric gene with several herbicide tolerance genes, plant cell and plant resistant to several herbicides
US20110167517A1 (en) 2010-01-06 2011-07-07 Pioneer Hi-Bred International, Inc. Identification of diurnal rhythms in photosynthetic and non-photsynthetic tissues from zea mays and use in improving crop plants
WO2011090752A1 (en) * 2009-12-29 2011-07-28 The Board Of Governors For Higher Education, State Of Rhode Island And Providence Plantations Male and female sterility lines used to make hybrids in genetically modified plants
US8071840B2 (en) 2005-09-15 2011-12-06 Cropdesign N.V. Plants having increase yield and method for making the same

Patent Citations (159)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US39247A (en) 1863-07-14 Improvement in skates
US897360A (en) 1908-03-04 1908-09-01 Nathan Fein Buckle.
US4658082A (en) 1984-07-25 1987-04-14 Atlantic Richfield Company Method for producing intact plants containing foreign DNA
US4945050A (en) 1984-11-13 1990-07-31 Cornell Research Foundation, Inc. Method for transporting substances into living cells and tissues and apparatus therefor
US4918006A (en) 1985-07-01 1990-04-17 E. I. Du Pont De Nemours And Company Gene coding for insecticidal crystal protein
US5561236A (en) 1986-03-11 1996-10-01 Plant Genetic Systems Genetically engineered plant cells and plants exhibiting resistance to glutamine synthetase inhibitors, DNA fragments and recombinants for use in the production of said cells and plants
US5648477A (en) 1986-03-11 1997-07-15 Plant Genetic Systems, N.V. Genetically engineered plant cells and plants exhibiting resistance to glutamine synthetase inhibitors, DNA fragments and recombinants for use in the production of said cells and plants
US5646024A (en) 1986-03-11 1997-07-08 Plant Genetic Systems, N.V. Genetically engineered plant cells and plants exhibiting resistance to glutamine synthetase inhibitors, DNA fragments and recombinants for use in the production of said cells and plants
US5759829A (en) 1986-03-28 1998-06-02 Calgene, Inc. Antisense regulation of gene expression in plant cells
US5879903A (en) 1986-08-23 1999-03-09 Hoechst Aktiengesellschaft Phosphinothricin-resistance gene, and its use
US5268463A (en) 1986-11-11 1993-12-07 Jefferson Richard A Plant promoter α-glucuronidase gene construct
US5599670A (en) 1986-11-11 1997-02-04 Cambia Biosystems. L.L.C. β-glucuronidase and glucuronide permease gene system
US4987071A (en) 1986-12-03 1991-01-22 University Patents, Inc. RNA ribozyme polymerases, dephosphorylases, restriction endoribonucleases and methods
US5741684A (en) 1988-02-03 1998-04-21 Pioneer Hi-Bred International, Inc. Molecular methods of hybrid seed production
US5728926A (en) 1988-02-03 1998-03-17 Pioneer Hi-Bred International, Inc. Antisense gene systems of pollination control for hybrid seed production
US6191343B1 (en) 1988-02-03 2001-02-20 Pioneer Hi-Bred International Molecular methods of hybrid seed production
US6184439B1 (en) 1988-02-03 2001-02-06 Pioneer Hi Bred International, Inc. Antisense gene systems of pollination control for hybrid seed production
US5728558A (en) 1988-02-03 1998-03-17 Pioneer Hi-Bred International, Inc. Molecular methods of hybrid seed production
US6162964A (en) 1988-02-03 2000-12-19 Pioneer Hi-Bred International, Inc. Molecular methods of hybrid seed production
US6372967B1 (en) 1988-04-28 2002-04-16 Aventis Cropscience N.V. Plants with modified stamen cells
US5283184A (en) 1989-03-30 1994-02-01 Dna Plant Technology Corporation Genetic engineering of novel plant phenotypes
US5034323A (en) 1989-03-30 1991-07-23 Dna Plant Technology Corporation Genetic engineering of novel plant phenotypes
US5633441A (en) 1989-08-04 1997-05-27 Plant Genetic Systems, N.V. Plants with genetic female sterility
US5262306A (en) 1989-09-26 1993-11-16 Robeson David J Methods for identifying cercosporin-degrading microorganisms
US5641876A (en) 1990-01-05 1997-06-24 Cornell Research Foundation, Inc. Rice actin gene and promoter
WO1991010725A1 (en) 1990-01-22 1991-07-25 Dekalb Plant Genetics Fertile transgenic corn plants
US5489520A (en) 1990-04-17 1996-02-06 Dekalb Genetics Corporation Process of producing fertile transgenic zea mays plants and progeny comprising a gene encoding phosphinothricin acetyl transferase
US5550318A (en) 1990-04-17 1996-08-27 Dekalb Genetics Corporation Methods and compositions for the production of stably transformed, fertile monocot plants and cells thereof
US5969213A (en) 1990-04-17 1999-10-19 Dekalb Genetics Corporation Methods and compositions for the production of stably transformed fertile monocot plants and cells thereof
US5874265A (en) 1990-04-17 1999-02-23 Dekalb Genetics Corporation Methods and compositions for the production of stably transformed fertile monocot plants and cells thereof
US5919675A (en) 1990-04-17 1999-07-06 Dekalb Genetics Corporation Methods and compositions for the production of stably transformed, fertile monocot plants and cells thereof
US5723756A (en) 1990-04-26 1998-03-03 Plant Genetic Systems, N.V. Bacillus thuringiensis strains and their genes encoding insecticidal toxins
US6297426B1 (en) 1990-06-12 2001-10-02 Pioneer Hi-Bred International, Inc. Methods of mediating female fertility in plants
EP0465024A1 (en) * 1990-06-12 1992-01-08 Pioneer Hi-Bred International, Inc. Control of microsporogenesis using externally inducible promoter sequences
US5859341A (en) 1990-06-12 1999-01-12 Pioneer Hi-Bred International, Inc. Nucleotide sequences mediating fertility and method of using same
US5478369A (en) 1990-06-12 1995-12-26 Pioneer Hi-Bred International, Inc. Nucleotide sequences mediating male fertility and method of using same
US5463175A (en) 1990-06-25 1995-10-31 Monsanto Company Glyphosate tolerant plants
US5366892A (en) 1991-01-16 1994-11-22 Mycogen Corporation Gene encoding a coleopteran-active toxin
US5589610A (en) 1991-02-07 1996-12-31 Plant Genetic Systems, N.V. Stamen-specific promoters from corn
WO1992018625A1 (en) * 1991-04-16 1992-10-29 Mogen International N.V. Male-sterile plants, method for obtaining male-sterile plants and recombinant dna for use therein
WO1993002197A1 (en) * 1991-07-23 1993-02-04 Nickerson Biocem Limited CALLASE-RELATED DNAs AND THEIR USE IN ARTIFICIAL MALE STERILITY
US5747450A (en) 1991-08-02 1998-05-05 Kubota Corporation Microorganism and insecticide
US5952544A (en) 1991-12-04 1999-09-14 E. I. Du Pont De Nemours And Company Fatty acid desaturase genes from plants
US5341001A (en) 1992-02-13 1994-08-23 Matsushita Electric Industrial Co., Ltd. Sulfide-selenide manganese-zinc mixed crystal photo semiconductor and laser diode
US5942657A (en) 1992-05-13 1999-08-24 Zeneca Limited Co-ordinated inhibition of plant gene expression
EP0604662A1 (en) 1992-07-07 1994-07-06 Japan Tobacco Inc. Method of transforming monocotyledon
US5412085A (en) 1992-07-09 1995-05-02 Pioneer Hi-Bred International Inc. Pollen-specific promoter from maize
US5545546A (en) 1992-07-09 1996-08-13 Pioneer Hi-Bred International, Inc. Pollen-specific promoter from maize
US5332808A (en) 1992-09-08 1994-07-26 North Carolina State University DNA encoding a ribosome inactivating protein
US5792852A (en) 1992-11-16 1998-08-11 Cancer Research Fund Of Contra Costa Polynucleotides encoding modified antibodies with human milk fat globule specificity
WO1994011516A1 (en) 1992-11-17 1994-05-26 E.I. Du Pont De Nemours And Company Genes for microsomal delta-12 fatty acid desaturases and related enzymes from plants
US5602321A (en) 1992-11-20 1997-02-11 Monsanto Company Transgenic cotton plants producing heterologous polyhydroxy(e) butyrate bioplastic
US5990389A (en) 1993-01-13 1999-11-23 Pioneer Hi-Bred International, Inc. High lysine derivatives of α-hordothionin
US5589379A (en) 1993-02-26 1996-12-31 Calgene, Inc. Geminivirus-based gene expression system
US5583210A (en) 1993-03-18 1996-12-10 Pioneer Hi-Bred International, Inc. Methods and compositions for controlling plant development
EP0672752A1 (en) 1993-09-03 1995-09-20 Japan Tobacco Inc. Method of transforming monocotyledon by using scutellum of immature embryo
US5683439A (en) 1993-10-20 1997-11-04 Hollister Incorporated Post-operative thermal blanket
US5756325A (en) 1993-12-09 1998-05-26 Thomas Jefferson University Compounds and methods for site directed mutations in eukaryotic cells
US5565350A (en) 1993-12-09 1996-10-15 Thomas Jefferson University Compounds and methods for site directed mutations in eukaryotic cells
US5871984A (en) 1993-12-09 1999-02-16 Thomas Jefferson University Compounds and methods for site directed mutations in eukaryotic cells
US5703409A (en) 1993-12-21 1997-12-30 Fujitsu Limited Error counting circuit
US5470359A (en) 1994-04-21 1995-11-28 Pioneer Hi-Bred Internation, Inc. Regulatory element conferring tapetum specificity
US5837850A (en) 1994-04-21 1998-11-17 Pioneer Hi-Bred International, Inc. Regulatory element conferring tapetum specificity
US5593881A (en) 1994-05-06 1997-01-14 Mycogen Corporation Bacillus thuringiensis delta-endotoxin
US6282837B1 (en) 1994-06-16 2001-09-04 Novartis Finance Corporation Methods of controlling the growth of undesired vegetation with herbicide tolerant plants or plant seeds having altered protoporphyrinogen oxidase activity
US5767373A (en) 1994-06-16 1998-06-16 Novartis Finance Corporation Manipulation of protoporphyrinogen oxidase enzyme activity in eukaryotic organisms
US6288306B1 (en) 1994-06-16 2001-09-11 Syngenta Investment Corporation Methods of selecting plants, plant tissue or plant cells resistant to a protoporphyrinogen oxidase inhibitor
US5962764A (en) 1994-06-17 1999-10-05 Pioneer Hi-Bred International, Inc. Functional characterization of genes
US6013859A (en) 1994-07-14 2000-01-11 Pioneer Hi-Bred International, Inc. Molecular methods of hybrid seed production
US5736369A (en) 1994-07-29 1998-04-07 Pioneer Hi-Bred International, Inc. Method for producing transgenic cereal plants
US5792931A (en) 1994-08-12 1998-08-11 Pioneer Hi-Bred International, Inc. Fumonisin detoxification compositions and methods
US5608144A (en) 1994-08-12 1997-03-04 Dna Plant Technology Corp. Plant group 2 promoters and uses thereof
US5736514A (en) 1994-10-14 1998-04-07 Nissan Chemical Industries, Ltd. Bacillus strain and harmful organism controlling agents
US5837851A (en) 1994-12-08 1998-11-17 Pioneer Hi-Bred International, Inc. DNA promoter 5126 and constructs useful in a reversible nuclear genetic system for male sterility in transgenic plants
US5792853A (en) 1994-12-08 1998-08-11 Pioneer Hi-Bred International, Inc. Reversible nuclear genetic system for male sterility in transgenic plants
US6248935B1 (en) 1994-12-08 2001-06-19 Pioneer Hi-Bred International, Inc. Reversible nuclear genetic system for male sterility in transgenic plants
US6281348B1 (en) 1994-12-08 2001-08-28 Pioneer Hi-Bred International, Inc. Reversible nuclear genetic system for male sterility in transgenic plants
US5689049A (en) 1994-12-08 1997-11-18 Pioneer Hi-Bred International, Inc. Transgenic plant and method for producing male sterility using anther specific promoter 5126
US5689051A (en) 1994-12-08 1997-11-18 Pioneer Hi-Bred International, Inc. Transgenic plants and DNA comprising anther specific promoter 5126 and gene to achieve male sterility
US6399856B1 (en) 1994-12-08 2002-06-04 Pioneer Hi-Bred International, Inc. Reversible nuclear genetic system for male sterility in transgenic plants
WO1996030530A1 (en) 1995-03-24 1996-10-03 Pioneer Hi-Bred International, Inc. Als3 promoter
US5659026A (en) 1995-03-24 1997-08-19 Pioneer Hi-Bred International ALS3 promoter
US5885802A (en) 1995-06-02 1999-03-23 Pioneer Hi-Bred International, Inc. High methionine derivatives of α-hordothionin
US5885801A (en) 1995-06-02 1999-03-23 Pioneer Hi-Bred International, Inc. High threonine derivatives of α-hordothionin
US5962769A (en) 1995-06-07 1999-10-05 Pioneer Hi-Bred International, Inc. Induction of male sterility in plants by expression of high levels of avidin
US6566587B1 (en) 1995-07-19 2003-05-20 Bayer Cropscience S.A. Mutated 5-enolpyruvylshikimate-3-phosphate synthase, gene coding for said protein and transformed plants containing said gene
US5737514A (en) 1995-11-29 1998-04-07 Texas Micro, Inc. Remote checkpoint memory system and protocol for fault-tolerant computer system
US5703049A (en) 1996-02-29 1997-12-30 Pioneer Hi-Bred Int'l, Inc. High methionine derivatives of α-hordothionin for pathogen-control
US5693512A (en) 1996-03-01 1997-12-02 The Ohio State Research Foundation Method for transforming plant tissue by sonication
US5850016A (en) 1996-03-20 1998-12-15 Pioneer Hi-Bred International, Inc. Alteration of amino acid compositions in seeds
US5760012A (en) 1996-05-01 1998-06-02 Thomas Jefferson University Methods and compounds for curing diseases caused by mutations
US6072050A (en) 1996-06-11 2000-06-06 Pioneer Hi-Bred International, Inc. Synthetic promoters
US5731181A (en) 1996-06-17 1998-03-24 Thomas Jefferson University Chimeric mutational vectors having non-natural nucleotides
US5795972A (en) 1996-06-17 1998-08-18 Thomas Jefferson University Chimeric mutational vectors having non-natural nucleotides
US6300543B1 (en) 1996-07-08 2001-10-09 Pioneer Hi-Bred International, Inc. Transformation of zygote, egg or sperm cells and recovery of transformed plants from isolated embryo sacs
US7935869B2 (en) 1996-07-16 2011-05-03 Bayer S.A.S. Chimeric gene with several herbicide tolerance genes, plant cell and plant resistant to several herbicides
WO1998020122A1 (en) 1996-11-01 1998-05-14 The Institute Of Physical And Chemical Research METHOD FOR FORMING FULL-LENGTH cDNA LIBRARY
WO1998020133A2 (en) 1996-11-01 1998-05-14 Pioneer Hi-Bred International, Inc. Proteins with enhanced levels of essential amino acids
US6232529B1 (en) 1996-11-20 2001-05-15 Pioneer Hi-Bred International, Inc. Methods of producing high-oil seed by modification of starch levels
US6177616B1 (en) 1996-12-16 2001-01-23 Hoecsht Schering Agrevo Gmbh Genes coding for amino acid deacetylases with specificity for N-acetyl-L-phosphinothricin, their isolation and their use
US5981840A (en) 1997-01-24 1999-11-09 Pioneer Hi-Bred International, Inc. Methods for agrobacterium-mediated transformation
US7022896B1 (en) 1997-04-04 2006-04-04 Board Of Regents Of University Of Nebraska Methods and materials for making and using transgenic dicamba-degrading organisms
WO1998049350A1 (en) 1997-04-30 1998-11-05 Regents Of The University Of Minnesota In vivo use of recombinagenic oligonucleobases to correct genetic lesions in hepatocytes
WO1998053083A1 (en) 1997-05-21 1998-11-26 Zeneca Limited Gene silencing
US20030175965A1 (en) 1997-05-21 2003-09-18 Lowe Alexandra Louise Gene silencing
US6037523A (en) 1997-06-23 2000-03-14 Pioneer Hi-Bred International Male tissue-preferred regulatory region and method of using same
WO1999007865A1 (en) 1997-08-05 1999-02-18 Kimeragen, Inc. The use of mixed duplex oligonucleotides to effect localized genetic changes in plants
WO1999025821A1 (en) 1997-11-18 1999-05-27 Pioneer Hi-Bred International, Inc. Compositions and methods for genetic modification of plants
WO1999043838A1 (en) 1998-02-24 1999-09-02 Pioneer Hi-Bred International, Inc. Synthetic promoters
US6177611B1 (en) 1998-02-26 2001-01-23 Pioneer Hi-Bred International, Inc. Maize promoters
WO1999049029A1 (en) 1998-03-20 1999-09-30 Benitec Australia Ltd Control of gene expression
WO1999053050A1 (en) 1998-04-08 1999-10-21 Commonwealth Scientific And Industrial Research Organisation Methods and means for obtaining modified phenotypes
WO1999061619A2 (en) 1998-05-22 1999-12-02 Pioneer Hi-Bred International, Inc. Cell cycle genes, proteins and uses thereof
WO1999061631A1 (en) 1998-05-26 1999-12-02 Novartis Ag Dsrna-mediated regulation of gene expression in plants
US6225529B1 (en) 1998-08-20 2001-05-01 Pioneer Hi-Bred International, Inc. Seed-preferred promoters
WO2000011177A1 (en) 1998-08-20 2000-03-02 Pioneer Hi-Bred International, Inc. Seed-preferred promoters
US6528704B1 (en) 1998-08-28 2003-03-04 Pioneer Hi-Bred International, Inc. Seed-preferred promoters from end genes
WO2000012733A1 (en) 1998-08-28 2000-03-09 Pioneer Hi-Bred International, Inc. Seed-preferred promoters from end genes
US6504082B1 (en) 1998-09-10 2003-01-07 Pioneer Hi-Bred International, Inc. Ecdysone receptors and methods for their use
WO2000017364A2 (en) 1998-09-23 2000-03-30 Pioneer Hi-Bred International, Inc. Cyclin d polynucleotides, polypeptides and uses thereof
US6453242B1 (en) 1999-01-12 2002-09-17 Sangamo Biosciences, Inc. Selection of sites for targeting by zinc finger proteins and methods of designing zinc finger proteins to bind to preselected sites
WO2000049035A1 (en) 1999-02-19 2000-08-24 The General Hospital Corporation Gene silencing
US6452069B1 (en) 1999-03-16 2002-09-17 Pioneer Hi-Bred International, Inc. SF3 promoter and methods of use
WO2000070067A1 (en) 1999-05-14 2000-11-23 Dekalb Genetics Corporation The rice actin 2 promoter and intron and methods for use thereof
WO2001000834A1 (en) * 1999-06-29 2001-01-04 Pioneer Hi-Bred International, Inc. Gene affecting male fertility in plants
WO2001012825A1 (en) 1999-08-13 2001-02-22 Syngenta Participations Ag Herbicide-tolerant protoporphyrinogen oxidase
US7087812B1 (en) 1999-09-30 2006-08-08 Japan Tobacco Inc. Vectors for transforming plants
US20030037355A1 (en) 2000-01-21 2003-02-20 Barbas Carlos F. Methods and compositions to modulate expression in plants
US20030074698A1 (en) 2000-06-16 2003-04-17 Thomas Schmulling Method for modifying plant morphology, biochemistry and physiology
WO2002000904A2 (en) 2000-06-23 2002-01-03 E. I. Du Pont De Nemours And Company Recombinant constructs and their use in reducing gene expression
US20020048814A1 (en) 2000-08-15 2002-04-25 Dna Plant Technology Corporation Methods of gene silencing using poly-dT sequences
WO2002026789A2 (en) 2000-09-26 2002-04-04 Pioneer Hi-Bred International, Inc. Nucleotide sequences mediating male fertility and method of using same
US8013218B2 (en) 2000-09-26 2011-09-06 Pioneer Hi-Bred International, Inc. Nucleotide sequences mediating plant male fertility and method of using same
US7893317B2 (en) 2000-09-26 2011-02-22 Pioneer Hi-Bred International, Inc Nucleotide sequences mediating plant male fertility and method of using same
US7666643B2 (en) 2000-10-30 2010-02-23 Pioneer Hi-Bred International, Inc. Glyphosate-N-acetyltransferase (GAT) genes
US7462481B2 (en) 2000-10-30 2008-12-09 Verdia, Inc. Glyphosate N-acetyltransferase (GAT) genes
US7531339B2 (en) 2000-10-30 2009-05-12 Verdia, Inc. Glyphosate-N-acetyltransferase (GAT) genes
US7527955B2 (en) 2000-10-30 2009-05-05 Pioneer Hi-Bred International, Inc. Glyphosate-N-acetyltransferase (GAT) genes
US7714188B2 (en) 2000-10-30 2010-05-11 Pioneer Hi-Bred Int'l., Inc. Glyphosate-N-acetyltransferase (GAT) genes
US20090055976A1 (en) 2000-12-07 2009-02-26 Syngenta Limited Methods for the production of plants resistant to HPPD herbicides
WO2002063021A2 (en) 2001-02-08 2002-08-15 Pioneer Hi-Bred International, Inc. Nucleotide sequence mediating male fertility and method of using same
US6646805B2 (en) 2001-03-02 2003-11-11 Fujitsu Limited Apparatus for variable wavelength dispersion and wavelength dispersion slope
US6750868B2 (en) 2001-09-03 2004-06-15 Via Technologies, Inc. Universal accelerated graphic port system and method for operating the same
US20030217393A1 (en) 2002-02-15 2003-11-20 Pioneer Hi-Bred International, Inc. Development of a stress-responsive promoter from maize
US20030180945A1 (en) 2002-03-14 2003-09-25 Ming-Bo Wang Modified gene-silencing RNA and uses thereof
WO2003102198A1 (en) 2002-06-04 2003-12-11 Metanomics Gmbh & Co. Kgaa Method for the stable expression of nucleic acids in transgenic plants, controlled by a parsley-ubiquitin promoter
US7696405B2 (en) 2003-12-16 2010-04-13 Pioneer Hi-Bred International, Inc. Dominant gene suppression transgenes and methods of using same
US20110124503A1 (en) 2004-04-30 2011-05-26 Dow Agrosciences Llc Novel herbicide resistance genes
US7838733B2 (en) 2004-04-30 2010-11-23 Dow Agrosciences Llc Herbicide resistance genes
US7622641B2 (en) 2005-08-24 2009-11-24 Pioneer Hi-Bred Int'l., Inc. Methods and compositions for providing tolerance to multiple herbicides
US8071840B2 (en) 2005-09-15 2011-12-06 Cropdesign N.V. Plants having increase yield and method for making the same
WO2007053482A2 (en) 2005-10-28 2007-05-10 Dow Agrosciences Llc Novel herbicide resistance genes
US20070277269A1 (en) * 2006-04-17 2007-11-29 Ceres, Inc. Nucleotide sequences and polypeptides encoded thereby useful for modifying plant characteristics
WO2007146706A2 (en) 2006-06-06 2007-12-21 Monsanto Technology Llc Modified dicamba monooxygenase enzyme and methods of its use
WO2009114321A2 (en) 2008-03-11 2009-09-17 Precision Biosciencs, Inc. Rationally-designed meganucleases for maize genome engineering
US20110023180A1 (en) 2009-01-22 2011-01-27 Syngenta Participations Ag Novel hydroxyphenylpyruvate dioxygenase polypeptides and methods of use
US7709709B1 (en) 2009-03-25 2010-05-04 Monsanto Technology Llc Plants and seeds of corn variety CV334995
WO2011090752A1 (en) * 2009-12-29 2011-07-28 The Board Of Governors For Higher Education, State Of Rhode Island And Providence Plantations Male and female sterility lines used to make hybrids in genetically modified plants
US20110167517A1 (en) 2010-01-06 2011-07-07 Pioneer Hi-Bred International, Inc. Identification of diurnal rhythms in photosynthetic and non-photsynthetic tissues from zea mays and use in improving crop plants

Non-Patent Citations (378)

* Cited by examiner, † Cited by third party
Title
"American Type Culture Collection Catalogue of Cell Lines and Hybridomas", 1992
AIMEI TIAN ET AL: "Characterization of a male sterile related gene BcMF15 from Brassica campestris ssp. chinensis", MOLECULAR BIOLOGY REPORTS ; AN INTERNATIONAL JOURNAL ON MOLECULAR AND CELLULAR BIOLOGY, KLUWER ACADEMIC PUBLISHERS, DO, vol. 36, no. 2, 22 November 2007 (2007-11-22), pages 307 - 314, XP019647213, ISSN: 1573-4978 *
ALBANI ET AL., EMBO, vol. 3, 1984, pages 1405 - 15
ALBANI ET AL., PLANT J., vol. 4, 1993, pages 343 - 55
ALBANI ET AL., PLANT MOL BIOL., vol. 15, 1990, pages 605
ALBANI, THEOR. APPL. GEN., vol. 98, 1999, pages 1253 - 62
ALBERTSEN ET AL., MAIZE GENETICS NEWSLETTER, vol. 67, 1993, pages 51 - 52
ALBERTSEN, MNL, vol. 67, 1993, pages 64
ALBERTSEN; PHILLIPS, CAN. J. GENET. CYTOL., vol. 23, 1981, pages 195 - 208
ALTSCHUL ET AL., NUCLEIC ACIDS RES., vol. 25, 1997, pages 3389 - 402
AMOAH ET AL., J EXP BOT, vol. 52, 2001, pages 1135 - 42
AN ET AL., PLANT CELL, vol. 1, 1989, pages 115 - 22
ANGELL; BAULCOMBE, EMBO J., vol. 16, 1997, pages 3675 - 3684
ANGELL; BAULCOMBE, PLANT J., vol. 20, 1999, pages 357 - 362
ARNOLDO ET AL., J. CELL. BIOCHEM, 1992
ARREDONDO-PETER ET AL., PLANT PHYSIOL, vol. 114, pages 493 - 500
ARREDONDO-PETER ET AL., PLANT PHYSIOL., vol. 115, 1997, pages 1259 - 1266
ATANASSOVA ET AL., PLANT J., vol. 2, 1992, pages 291
ATANASSVOA ET AL., PLANT JOURNAL, vol. 2, no. 3, 1992, pages 291 - 300
AUFSAFTZ ET AL., PROC. NAT'L. ACAD. SCI., vol. 99, no. 4, 2002, pages 16499 - 16506
AUSUBEL, ET AL.,: "Current Protocols in Molecular Biology", 1994, CURRENT PROTOCOLS
AUSUBEL, ET AL.: "Current Protocols in Molecular Biology", 1995, GREENE PUBLISHING AND WILEY-INTERSCIENCE
BACK ET AL., PLANT MOL. BIOL., vol. 17, 1991, pages 9
BAIM ET AL., PROC. NATL. ACAD. SCI. USA, vol. 88, 1991, pages 5072 - 5076
BALTZ ET AL., THE PLANT JOURNAL, vol. 2, 1992, pages 713 - 721
BAO, ULTRASOUND IN MEDICINE & BIOLOGY, vol. 23, 1997, pages 953 - 959
BARKLEY ET AL., THE OPERON, 1980, pages 177 - 220
BARROS ET AL., PLANT MOL., vol. 19, 1992, pages 665 - 75
BEETHAM ET AL., PROC. NATL ACAD. SCI. USA, vol. 96, 1999, pages 8774 - 8778
BENDTSEN JD; NIELSEN H; VON HEIJNE G; BRUNAK S., J MOL BIOL., vol. 340, no. 4, 16 July 2004 (2004-07-16), pages 783 - 95
BENFEY; CHUA, SCIENCE, vol. 244, 1989, pages 174 - 81
BENSEN ET AL., PLANT CE, vol. 7, 1995, pages 75 - 84
BERGER ET AL., PROC. NATL. ACAD. SCI. USA, vol. 86, 1989, pages 8402 - 6
BERGER; KIMMEL: "Methods in Enzymology", vol. 152, 1987, ACADEMIC PRESS, INC., article "Guide To Molecular Cloning Techniques"
BEVAN ET AL., NUCLEIC ACIDS RES., vol. 12, 1983, pages 369 - 85
BLAZQUEZ ET AL., DEVELOPMENT, vol. 124, 1997, pages 3835 - 3844
BOLTE ET AL., J. CELL SCIENCE, vol. 117, 2004, pages 943 - 54
BONIN, PH.D. THESIS, 1993
BRAUN ET AL., PLANT CELL, vol. 2, 1990, pages 153
BRIGNON ET AL., PLANT MOL BIO, vol. 22, no. 6, 1993, pages 1007 - 1015
BROIN ET AL., PLANT CELL, vol. 14, 2002, pages 1417 - 1432
BROWN ET AL., CELL, vol. 49, 1987, pages 603 - 612
BUCHMAN; BERG, MOL. CELL BIOL., vol. 8, 1988, pages 4395 - 4405
BYTEBIERM ET AL., PROC. NATL. ACAD. SCI. USA, vol. 84, 1987, pages 5345 - 5349
CALLIS ET AL., GENES DEV., vol. 1, 1987, pages 1183 - 200
CANEVASCINI ET AL., PLANT PHYSIOL., vol. 112, no. 2, 1996, pages 513 - 524
CHANG ET AL., NATURE, vol. 198, 1977, pages 1056
CHANGMING CHEN ET AL: "CaMF2, an anther-specific lipid transfer protein (LTP) gene, affects pollen development inL", PLANT SCIENCE, ELSEVIER IRELAND LTD, IE, vol. 181, no. 4, 12 July 2011 (2011-07-12), pages 439 - 448, XP028281969, ISSN: 0168-9452, [retrieved on 20110723], DOI: 10.1016/J.PLANTSCI.2011.07.003 *
CHAUDHARY ET AL., PLANT MOL. BIOL., vol. 30, 1996, pages 1247 - 57
CHEN ET AL., PNAS, vol. 88, 1991, pages 315 - 319
CHENG ET AL., PLANT PHYSIOL, vol. 108, 1995, pages 881
CHIU ET AL., CURRENT BIOLOGY, vol. 6, 1996, pages 325 - 330
CHRISTENSEN ET AL., PLANT MO/. BIOL., vol. 12, 1989, pages 619 - 632
CHRISTENSEN ET AL., PLANT MOL. BIOI., vol. 12, 1992, pages 619 - 632
CHRISTENSEN ET AL., PLANT MOL. BIOL., vol. 18, 1992, pages 675 - 689
CHRISTENSEN ET AL., PLANT MOL. BIOL., vol. 18, 1992, pages 675 - 89
CHRISTOPHERSON ET AL., PROC. NATL. ACAD. SCI. USA, vol. 89, 1992, pages 6314 - 6318
CHRISTOPHERSON ET AL., PROC. NATL. ACAD. SCI., USA, vol. 89, 1992, pages 6314 - 6318
CHRISTOU ET AL., PLANT PHYSIOL., vol. 87, 1988, pages 671 - 674
CHRISTOU ET AL., PROC. NATL. ACAD. SCI. USA, vol. 84, 1987, pages 3962
CHRISTOU; FORD, ANNALS OF BOTANY, vol. 75, 1995, pages 407 - 413
CHU ET AL., SCI. SIN., vol. 18, 1975, pages 659
CHUANG; MEYEROWITZ, PROC. NATL. ACAD. SCI. USA, vol. 97, 2000, pages 4985 - 4990
CLAVERIE; STATES, COMPUT. CHEM., vol. 17, 1993, pages 191 - 201
COLASANTI ET AL., PNAS, vol. 88, 1991, pages 3377 - 3381
COLBERT ET AL., PLANT PHYSIOLOGY, vol. 126, 2001, pages 480 - 484
CONRAD; SONNEWAID, NATURE BIOTECH., vol. 21, 2003, pages 35 - 36
CORPET, NUCLEIC ACIDS RES., vol. 16, 1988, pages 10881 - 90
CREIGHTON: "Proteins", 1984, W.H. FREEMAN AND CO
CROSSLEY ET AL., PLANTA, vol. 196, no. 3, 1995, pages 523 - 529
CROSSWAY ET AL., BIOTECHNIQUES, vol. 4, 1986, pages 320 - 334
CROSSWAY ET AL., MOL. GEN. GENET., vol. 202, 1986, pages 179 - 185
CZAKO; AN, PLANT PHYSIOL., vol. 95, 1991, pages 687 - 692
DATABASE UniProt [Online] 1 May 2000 (2000-05-01), Lauga et al.: "Anther specific protein (Ltp-like protein)", XP002697369, Database accession no. Q9SBW9 *
DATTA ET AL., BIOTECHNOLOGY, vol. 8, 1990, pages 736 - 740
DE WET ET AL.: "In The Experimental Manipulation of Ovule Tissues", 1985, LONGMAN, pages: 197 - 209
DEGENKOLB ET AL., ANTIMICROB. AGENTS CHEMOTHER., vol. 35, 1991, pages 1591 - 1595
DEIKMAN ET AL., PLANT PHYSIOL., vol. 100, 1992, pages 2013 - 2017
DELOOSE ET AL., GENE, vol. 99, 1991, pages 95 - 100
DESHAYES ET AL., EMBO J., vol. 4, 1985, pages 2731
DEUSCHLE ET AL., PROC. NATL. ACAD. ACI. USA, vol. 86, 1989, pages 5400 - 5404
DEUSCHLE ET AL., SCIENCE, vol. 248, 1990, pages 480 - 483
DEVEREAUX ET AL., NUCLEIC ACIDS RES., vol. 12, 1984, pages 387 - 395
DEWET ET AL., MOL. CELL. BIOL., vol. 7, 1987, pages 725 - 737
DEWEY ET AL., PNAS, vol. 84, 1987, pages 5374
D'HALLUIN ET AL., PLANT CELL, vol. 4, 1992, pages 1495 - 1505
D'HALLUIN ET AL., PLANT CELL, vol. 4, 1992, pages 1495 - 505
DHARMAPURI; SONTI; 1999, FEMS MICROBIOL. LETT., vol. 179, pages 53 - 59
DONN ET AL., ABSTRACTS OF THE VLLTH LNT'L. CONGRESS ON PLANT CELL AND TISSUE CULTURE IAPTC, vol. A2-38, 1990, pages 53
DOTSON ET AL., THE PLANT JOUMAL, vol. 10, 1996, pages 383 - 392
DRAPER ET AL., PLANT CELL PHYSIOL., vol. 23, 1982, pages 451
DRATEWKA-KOS ET AL., J. BIOL. CHEM., vol. 264, 1989, pages 4896 - 900
DRUMMOND ET AL., NUCLEIC ACIDS RES., vol. 13, 1985, pages 7375
DUFF ET AL., J. BIOL. CHEM, vol. 27, 1997, pages 16749 - 16752
ERIKSSON, PHYSIOL. PLANT, vol. 18, 1965, pages 976
ERLICH,: "PCR Technology", 1989, STOCKTON PRESS
FARMER ET AL., HUM. MOL. GENET., vol. 3, 1994, pages 723 - 728
FEINBAUM ET AL., MOL. GEN. GENET., vol. 226, 1991, pages 449
FELDMANN ET AL.: "Arabidopsis", 1994, COLD SPRING HARBOR LABORATORY PRESS, pages: 137 - 172
FENG; DOOLITTLE, J. MOL. EVOL., vol. 25, 1987, pages 351 - 60
FETTER ET AL., PLANT CELL, vol. 16, 2004, pages 215 - 28
FIGGE ET AL., CELL, vol. 52, 1988, pages 713 - 722
FINER; FINER, LETT APPL MICROBIOL., vol. 30, 2000, pages 406 - 10
FITZMAURICE ET AL., GENETICS, vol. 153, 1999, pages 1919 - 1928
FLAVELL ET AL., PROC. NATL. ACAD. SCI. USA, vol. 91, 1994, pages 3490 - 3496
FRAME ET AL., PLANT J., vol. 6, 1994, pages 941 - 948
FREELING AND WALBOT,: "The Maize Handbook", 1994, SPRINGER, article "Chapter 116,"
FRIAS ET AL., PLANT CELL, vol. 8, 1996, pages 1533 - 44
FROMM ET AL., BIOLTECHNOLOGY, vol. 8, 1990, pages 833 - 839
FROMM ET AL., BIOTECHNOLOGY, vol. 8, 1990, pages 833 - 839
FROMM ET AL., PROC. NATL. ACAD. SCI. USA, vol. 82, 1985, pages 5824 - 5828
FUERST ET AL., PROC. NATL. ACAD. SCI. USA, vol. 86, 1989, pages 2549 - 2553
FURST ET AL., CELL, vol. 55, 1988, pages 705 - 717
GAI ET AL., NUCLEIC ACIDS RES., vol. 28, 2000, pages 94 - 96
GAO ET AL., PLANT JOURNAL, vol. 1, 2010, pages 176 - 187
GATZ ET AL., MOL. GEN. GENET., vol. 227, 1991, pages 229 - 237
GATZ ET AL., MOL. GEN. GENET., vol. 243, 1994, pages 32 - 38
GATZ ET AL., PLANT J., vol. 2, 1992, pages 397 - 404
GEISER ET AL., GENE, vol. 48, 1986, pages 109
GERLACH ET AL., PNAS USA, vol. 79, 1982, pages 2981 - 2985
GILL ET AL., NATURE, vol. 334, 1988, pages 721 - 724
GOEDDEL ET AL., NUCLEIC ACIDS RES., vol. 8, 1980, pages 4057
GOFF ET AL., EMBO J., vol. 9, 1990, pages 2517 - 2522
GOLDBERG ET AL., PLANT CELL, vol. 5, 1993, pages 1217 - 1229
GORDON-KAMM ET AL., PLANT CELL, vol. 2, 1990, pages 603 - 618
GOSSEN E, PROC. NATL. ACAD. SCI. USA, vol. 89, 1992, pages 5547 - 5551
GOSSEN, PH.D. THESIS, 1993
GREENFIELD ET AL., PNAS, vol. 80, 1983, pages 6853
GRITZ ET AL., GENE, vol. 25, 1983, pages 179 - 188
GRUBER ET AL.: "Vectors for Plant Transformation", METHODS IN PLANT MOLECULAR BIOLOGY AND BIOTECHNOLOGY, pages 89 - 119
GRUIS ET AL., PLANT CELL, vol. 14, 2002, pages 2863 - 2882
GUERRERO ET AL., MOL. GEN. GENET., vol. 224, 1993, pages 161 - 168
GUEVARA-GARCIA ET AL., PLANT J., vol. 4, no. 3, 1993, pages 495 - 505
GUNNAR; VON HEIJNE, EUR. J. BIOCHEM., vol. 133, 1983, pages 17 - 21
GUO ET AL., PHYSIOLOGIA PLANTARUM, vol. 93, 1995, pages 19 - 24
HAIN ET AL., MOL. GEN. GENET., vol. 199, 1985, pages 161
HAJELA ET AL., PLANT PHYSIOL., vol. 93, 1990, pages 1246 - 1252
HAMILTON ET AL., PLANT MOL. BIOL., vol. 18, 1992, pages 211 - 218
HANSEN ET AL., MOL. GEN GENET., vol. 254, no. 3, 1997, pages 337 - 343
HANSEN ET AL., MOL. GEN. GENET., vol. 254, no. 3, 1997, pages 337 - 343
HELLIWELL; WATERHOUSE, METHODS, vol. 30, 2003, pages 289 - 295
HEMPEL ET AL., DEVELOPMENT, vol. 124, 1997, pages 3845 - 3853
HENIKOFF; HENIKOFF, PROC. NATL. ACAD. SCI. USA, vol. 89, 1989, pages 10915
HERSHEY ET AL., MOL. GEN. GENE, vol. 227, 1991, pages 229 - 237
HIEI ET AL., THE PLANT JOURNAL, vol. 6, 1994, pages 271 - 82
HIGGINS; SHARP, CABIOS, vol. 5, 1989, pages 151 - 3
HIGGINS; SHARP, CABIOS, vol. 5, 1989, pages 151 - 53
HIGGINS; SHARP, GENE, vol. 73, 1988, pages 237 - 44
HILLENAND-WISSMAN, TOPICS MOL. STRUC. BIOL., vol. 10, 1989, pages 143 - 162
HLAVKA ET AL.: "Handbook of Experimental Pharmacology", vol. 78, 1985, SPRINGER-VERLAG
HOOYDAAS-VAN SLOGTEREN; HOOYKAAS, NATURE (LONDON, vol. 311, 1984, pages 763 - 764
HORSCH ET AL., SCIENCE, vol. 227, 1985, pages 1229 - 31
HU ET AL., CELL, vol. 48, 1987, pages 555 - 566
HUANG ET AL., COMPUTER APPLICATIONS IN THE BIOSCIENCES, vol. 8, 1992, pages 155 - 65
INNIS ET AL.: "PCR Protocols, A Guide to Methods and Applications", 1990, ACADEMIC PRESS
ISHIZUKA ET AL., J. GEN. MICROBIOL., vol. 139, 1993, pages 425 - 32
ITO ET AL., PLANT J., vol. 11, 1997, pages 983 - 992
ITO ET AL., PLANT MOL. BIOL., vol. 24, 1994, pages 863 - 878
JACK ET AL., CELL, vol. 76, 1994, pages 703
JAVIER ET AL., NATURE, vol. 425, 2003, pages 257 - 263
JEFFERSON ET AL.: "Plant Molecular Biology Manual", 1991, KLUWER ACADEMIC PUBLISHERS, pages: 1 - 33
JOHANSEN; CARRINGTON, PLANT PHYSIOL., vol. 126, 2001, pages 930 - 938
JONES ET AL., SCIENCE, vol. 266, 1994, pages 789
JORGENSEN ET AL., PLANT MOL. BIOL., vol. 31, 1996, pages 957 - 973
KADO, CRIT. REV. PLANT SCI., vol. 10, 1991, pages 1
KAEPPLER ET AL., THEOR. APPL. GENET., vol. 84, 1992, pages 560 - 566
KAEPPLER, PLANT CELL REPORTS, vol. 9, 1990, pages 415 - 418
KAIN ET AL., BIOTECHNIQUES, vol. 19, 1995, pages 650 - 655
KARES, PLANT MOL. BIOL., vol. 15, 1990, pages 225
KASUGA ET AL., NATURE BIOTECHNOLOGY, vol. 18, 1999, pages 287 - 291
KATO ET AL., PLANT PHYSIOL, vol. 129, 2002, pages 913 - 42
KAWAMATA ET AL., PLANT CELL PHYSIOL., vol. 38, no. 7, 1997, pages 792 - 803
KEIL ET AL., NUCLEIC ACIDS RES., vol. 14, 1986, pages 5641 - 50
KENN ET AL., J. BACTEROIL, vol. 168, 1986, pages 595
KIRCH ET AL., PLANT MOL BIOL., vol. 33, 1997, pages 897 - 909
KIRIHARA ET AL., GENE, vol. 71, 1988, pages 359
KLEIN ET AL., BIOTECHNOLOGY, vol. 10, 1992, pages 268
KLEIN ET AL., BIOTECHNOLOGY, vol. 6, 1988, pages 559 - 563
KLEIN ET AL., NATURE (LONDON, vol. 327, 1987, pages 70 - 73
KLEIN ET AL., PLANT PHYSIOL., vol. 91, 1988, pages 440 - 444
KLEIN ET AL., PROC. NATL. ACAD. SCI. USA, vol. 85, 1988, pages 4305 - 4309
KLEINSCHNIDT ET AL., BIOCHEMISTRY, vol. 27, 1988, pages 1094 - 1104
KOLTUNOW ET AL., PLANT CELL, vol. 2, 1990, pages 1201 - 1224
KOZAK, NUCLEIC ACIDS RES., vol. 15, 1987, pages 8125
KRENS ET AL., NATURE, vol. 296, 1982, pages 72 - 77
KUSABA ET AL., PLANT CELL, vol. 15, 2003, pages 1455 - 1467
LABOW ET AL., MOL. CELL. BIOL., vol. 10, 1990, pages 3343 - 3356
LAM, RESULTS PROBL. CELL DIFFER., vol. 20, 1994, pages 181 - 196
LAM; CHUA, SCIENCE, vol. 248, 1990, pages 471
LAST ET AL., THEOR. APPL. GENET., vol. 81, 1991, pages 581 - 588
LAST ET AL., THEOR. APPL. GENET., vol. 81, 1991, pages 581 - 8
LEPETIT ET AL., MOL. GEN. GENET., vol. 231, 1992, pages 276 - 85
LI ET AL., PLANT CELL REPORTS, vol. 12, 1993, pages 250 - 255
LIGHTNER; CASPAR: "Methods on Molecular Biology", vol. 82, 1998, HUMANA PRESS, pages: 91 - 104
LILLEY ET AL.: "Proceedings of the World Congress on Vegetable Protein Utilization in Human Foods and Animal Feedstuffs", 1989, AMERICAN OIL CHEMISTS SOCIETY, CHAMPAIGN, ILLINOIS, pages: 497 - 502
LIND ET AL., PLANT MOL. BIO/., vol. 18, 1992, pages 47 - 53
LIU ET AL., BCM GENOMICS, vol. 10, 2009, pages 1471 - 2164
LIU ET AL., PLANT PHYSIOL., vol. 129, 2002, pages 1732 - 1743
LIU; CANDE, MNL, vol. 66, 1992, pages 25 - 26
LU ET AL., PLANT CELL, vol. 8, no. 12, 1996, pages 2155 - 68
MAES ET AL., TRENDS PLANT SCI., vol. 4, 1999, pages 90 - 96
MARIANI ET AL., NATURE, vol. 347, 1990, pages 737
MARRS ET AL., DEV. GENET., vol. 14, 1993, pages 27 - 41
MARTIN ET AL., SCIENCE, vol. 262, 1993, pages 1432
MARTINEZ ET AL., PROC. NATL. ACAD. SCI., 1992
MATSUKA ET AL., PROC. NATL. ACAD. SCI. USA, vol. 88, 1991, pages 834
MATSUOKA ET AL., PROC NATL. ACAD. SCI. USA, vol. 90, no. 20, 1993, pages 9586 - 9590
MATSUOKA ET AL., PROC. NATL. ACAD. SCI. USA, vol. 90, no. 20, 1993, pages 9586 - 9590
MATZKE ET AL., CURR. OPIN. GENET. DEVE/., vol. 11, 2001, pages 221 - 227
MAYER; JEFFERSON: "Molecular Methods for Hybrid Rice Production", RURAL INDUSTRIES RESEARCH AND DEVELOPMENT CORPORATION, 2004
MCAVOY ET AL., ACTA HORT. (ISHS, vol. 625, 2003, pages 379 - 385
MCCABE, BIOTECHNOLOGY, vol. 6, 1988, pages 923 - 926
MCCALLUM ET AL., NAT BIOTECHNOL, vol. 18, 2000, pages 455 - 457
MCCALLUM ET AL., NATURE BIOTECHNOLOGY, vol. 18, 2000, pages 455 - 457
MCCALLUM ET AL., PLANT PHYSIOLOGY, vol. 123, 2000, pages 439 - 442
MCCALLUM, NAT. BIOTECHNOL., vol. 18, 2000, pages 455 - 457
MCELROY ET AL., PLANT CE, 1990, pages 163 - 171
MCELROY ET AL., PLANT CELL, vol. 2, 1990, pages 163 - 171
MCLEAN ET AL., J. BACTERIOL, vol. 169, 1987, pages 1017
MEDFORD ET AL., PLANT CELL, vol. 3, 1991, pages 359
MEINKOTH; WAHL, ANAL. BIOCHEM., vol. 138, 1984, pages 267 - 84
MEISSNER ET AL., PLANT J., vol. 22, 2000, pages 265 - 274
MENA ET AL., SCIENCE, vol. 274, 1996, pages 1537 - 1540
MENA ET AL., THE PLANT JOURNAL, vol. 116, 1998, pages 53 - 62
METT ET AL., PROC. NATL. ACAD. SCI., 1993
METTE ET AL., EMBO J, vol. 19, 2000, pages 5194 - 5201
MEYERS; MILLER, COMPUTER APPLIC. BIOL. SCI., vol. 4, 1988, pages 11 - 17
MIKI ET AL.: "Methods in Plant Molecular Biology and Biotechnology", 1993, CRC PRESS, INC., article "Procedure for Introducing Foreign DNA into Plants", pages: 67 - 88
MINDRINOS ET AL., CELL, vol. 78, 1994, pages 1089
MINDRINOS, CELL, vol. 78, 1994, pages 1089
MOGEN ET AL., PLANT CELL, vol. 2, 1990, pages 1261 - 72
MOLONEY ET AL., PLANT CELL REPORTS, vol. 8, 1989, pages 238
MOSBACH ET AL., NATURE, vol. 302, 1983, pages 543 - 5
MOURADOV ET AL., THE PLANT CELL, 2002, pages S111 - S130
MUESING ET AL., CELL, vol. 48, 1987, pages 691
MULLIS ET AL., METHODS ENZYMOL., vol. 155, 1987, pages 335 - 350
MURASHIGE; SKOOG, PHYSIOL. PLANT, vol. 15, 1962, pages 473
MURRAY ET AL., NUCLEIC ACIDS RES., vol. 17, 1989, pages 477 - 98
MUSUMURA ET AL., PLANT MOL. BIOL., vol. 12, 1989, pages 123
NADEAU ET AL., PLANT CELL, vol. 8, no. 2, 1996, pages 213 - 39
NATURE BIOTECHNOLOGY, vol. 14, 1996, pages 745 - 50
NATURE, vol. 347, 1990, pages 737
NEAL JENSEN: "Plant Breeding Methodology", 1988, JOHN WILEY & SONS, INC.
NEEDLEMAN; WUNSCH, J. MOL. BIOL., vol. 48, 1970, pages 443 - 53
NICHOLASS ET AL., PLANT MOL. BIOL., vol. 28, 1995, pages 423 - 435
ODELL ET AL., NATURE, vol. 313, 1985, pages 810 - 2
ODELL ET AL., NATURE, vol. 313, 1985, pages 810 - 812
OHSHIMA ET AL., VIROLOGY, vol. 243, 1998, pages 472 - 481
OKUBARA ET AL., GENETICS, vol. 137, 1994, pages 867 - 874
OLIVA ET AL., ANTIMICROB. AGENTS CHEMOTHER., vol. 36, 1992, pages 913 - 919
OPSAHL-FERSTAD ET AL., PLANT J, vol. 12, 1997, pages 235 - 46
OROZCO ET AL., PLANT MOL BIOL., vol. 23, no. 6, 1993, pages 1129 - 1138
OROZCO ET AL., PLANT MOL. BIO., vol. 23, no. 6, 1993, pages 1129 - 1138
OSJODA ET AL., NATURE BIOTECH., vol. 14, 1996, pages 745 - 750
OUELLET ET AL., FEBS LETT., vol. 423, 1998, pages 324 - 328
PALMITER ET AL., CELL, vol. 50, 1987, pages 435
PALVA ET AL., GENE, vol. 22, 1983, pages 229 - 35
PANDOLFINI ET AL., BMC BIOTECHNOLOGY, vol. 3, pages 7
PANSTRUGA ET AL., MOL. BIOL. REP., vol. 30, 2003, pages 135 - 140
PASZKOWSKI ET AL., EMBO J., vol. 3, 1984, pages 2717 - 2722
PAUL ET AL., PLANT MOL BIOL., vol. 19, no. 4, 1992, pages 611 - 22
PEARSON ET AL., METH. MOL. BIOL., vol. 24, 1994, pages 307 - 31
PEARSON; LIPMAN, PROC. NATL. ACAD. SCI. USA, vol. 85, 1988, pages 2444
PEDERSEN ET AL., J. BIOL. CHEM., vol. 261, 1986, pages 6279
PERSING, ET AL.,: "Diagnostic Molecular Microbiology Principles and Applications", 1993
PHOGAT ET AL., J. BIOSCI., vol. 25, 2000, pages 57 - 63
PLANT ET AL., PLANT MOL. BIOL., vol. 25, 1994, pages 193 - 205
PLANT J, vol. 3, no. 2, 1993, pages 261 - 271
POEHLMAN: "Breeding Field Crops", 1987, AVI PUBLICATION CO
POSTMA-HAARSMA ET AL., PLANT MOL. BIOL., vol. 39, 1999, pages 257 - 71
QUEEN ET AL., IMMUNOL. REV., vol. 89, 1986, pages 49
QUESADA ET AL., GENETICS, vol. 154, 2000, pages 421 - 436
RAGHOTHAMA ET AL., PLANT MOL BIOL, vol. 23, 1993, pages 1117 - 28
RAHMATULLAH ET AL., PLANT MOL. BIOL., vol. 12, 1989, pages 119
RAO ET AL., MOL. AND CELL. BIOL., vol. 8, 1988, pages 284
RASCO-GAUNT ET AL., PLANT CELL REP., vol. 21, no. 6, 2003, pages 569 - 576
REDEI; KONCZ ET AL.: "Methods in Arabidopsis Research", 1992, SINGAPORE, WORLD SCIENTIFIC PUBLISHING CO, pages: 16 - 82
REINES ET AL., PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 1917 - 1921
REZNIKOFF, MOL. MICROBIOL., vol. 6, 1992, pages 2419 - 2422
RIGGS ET AL., PROC. NATL. ACAD. SCI. USA, vol. 83, 1986, pages 5602 - 5606
RINEHART, PLANT PHYSIOL., vol. 112, no. 3, 1996, pages 1331 - 1341
RODER ET AL., MOL. GEN. GENET., vol. 243, 1994, pages 32 - 38
ROGERS ET AL., METH. ENZYMOL., vol. 153, 1987, pages 253 - 77
ROGERS, J. BIOL. CHEM., vol. 260, 1985, pages 3731 - 3738
ROGERS; MILLIMAN, J. BIOL. CHEM., vol. 259, no. 19, 1984, pages 12234 - 12240
RUSSELL ET AL., TRANSGENIC RES., vol. 6, no. 2, 1997, pages 157 - 168
SAMBROOK ET AL.: "Molecular Cloning: A Laboratory Manual 2nd ed.,", vol. 1-3, 1989
SAMBROOK ET AL.: "Molecular Cloning: A Laboratory Manual(2nd ed.", 1989, COLD SPRING HARBOR LABORATORY
SANFORD ET AL., PART. SCI. TECHNOL., vol. 5, 1987, pages 27
SANFORD ET AL., PARTICULATE SCIENCE AND TECHNOLOGY, vol. 5, 1987, pages 27 - 37
SANFORD, PHYSIOL PLANT, vol. 79, 1990, pages 206
SANFORD, TRENDS BIOTECH, vol. 6, 1988, pages 299
SATO ET AL., PROC. NATL. ACAD. SCI., vol. 93, 1996, pages 8117 - 8122
SAVERIA-CAMPO: "DNA Cloning: A Practical Approach", vol. II, 1985, IRL PRESS, article "Bovine Papilloma Virus DNA a Eukaryotic Cloning Vector", pages: 213 - 38
SCHARDI ET AL., GENE, vol. 61, 1987, pages 1 - 11
SCHEID ET AL., PROC. NATL. ACAD. SCI., USA, vol. 99, 2002, pages 13659 - 13662
SCHENA ET AL., PROC. NATL. ACAD. SCI. USA, vol. 88, 1991, pages 10421 - 10425
SCHNEIDER ET AL., PLANT PHYSIOL., vol. 113, 1997, pages 335 - 45
SCHNEIDER, J. EMBRYOL. EXP. MORPHOL., vol. 27, 1987, pages 353 - 65
SCHUBERT ET AL., J. BACTERIOL, vol. 170, 1988, pages 5837 - 5847
SCHUBERT ET AL., J. BACTERIOL., vol. 170, 1988, pages 5837 - 5847
SHAHIN, THEOR. APPL. GENET., vol. 69, 1985, pages 235 - 40
SHAUL ET AL., PROC. NATL. ACAD. SCI., vol. 93, 1996, pages 4868 - 4872
SHERMAN ET AL.: "Methods in Yeast Genetics", 1982, COLD SPRING HARBOR LABORATORY
SHIMATAKE ET AL., NATURE, vol. 292, 1981, pages 128
SHUKLA ET AL., NATURE, vol. 459, no. 7245, 2009, pages 437 - 41
SIGNALP 3.0.; BENDTSEN JD.; NIELSEN H.; VON HEIJNE G.; BRUNAK S., J MOL BIOL., vol. 340, no. 4, 16 July 2004 (2004-07-16), pages 783 - 95
SIGNALP 3.0.; BENDTSEN JD; NIELSEN H; VON HEIJNE G; BRUNAK S., J MOL BIOL., vol. 340, no. 4, 16 July 2004 (2004-07-16), pages 783 - 95
SIJEN ET AL., CU/R. BIOL., vol. 11, 2001, pages 436 - 440
SIMPSON ET AL., PLANT MOL. BIOL., vol. 6, 1986, pages 403 - 15
SMITH ET AL., NATURE, vol. 407, 2000, pages 319 - 320
SMITH; FEDOROFF; 1995, PLANT CELL, vol. 7, pages 735 - 745
SMITH; WATERMAN, ADV. APPL. MATH, vol. 2, 1981, pages 482
SMITH; WYCH, SEED SCI. TECHNO/., vol. 14, 1995, pages 1 - 8
SPALDING ET AL., J GEN PHYSIOL, vol. 113, 1999, pages 909 - 18
SPENCER ET AL., PLANT MOL. BIOL., vol. 24, 1994, pages 51 - 61
SPRAGUE ET AL., J. VIROL., vol. 45, 1983, pages 773 - 81
STOUTJESDIJK ET AL., PLANT PHYSIOL., vol. 129, 2002, pages 1723 - 1731
SU ET AL., BIOTECHNOL BIOENG, vol. 85, 2004, pages 610 - 9
SUKANYA ET AL., PLANT MOL BIOL, vol. 26, 1994, pages 1935 - 46
TAKAHASHI ET AL., PLANT PHYSIOL., vol. 99, 1992, pages 383 - 390
TERADA ET AL., PLANT J., vol. 3, 1993, pages 241
THOMPSON ET AL., BIOESSAYS, vol. 10, 1989, pages 108
TIJSSEN: "Laboratory Techniques in Biochemistry and Molecular Biology - Hybridization with Nucleic Acid Probes", 1993, ELSEVIER, article "Overview of principles of hybridization and the strategy of nucleic acid probe assays"
TOMES ET AL.: "Plant Cell, Tissue and Organ Culture, Fundamental Methods", 1995, SPRINGER-VERLAG BERLIN HEIDELBERG NEW YORK, article "Direct DNA Transfer into Intact Plant Cells Via Microprojectile Bombardment", pages: 197 - 213
TOMES ET AL.: "Plant Cell, Tissue and Organ Culture: Fundamental Methods", 1995, pages: 197 - 213
TREHIN ET AL., PLANT MOL. BIOL., vol. 35, 1997, pages 667 - 672
TSUGEKI; FEDOROFF, PROC. NATL. ACAD., USA, vol. 96, 1999, pages 12941 - 12946
TWELL ET AL., MOL. GEN. GENET., vol. 217, 1989, pages 240 - 245
TWELL ET AL., SEX. PLANT REPROD., vol. 6, 1993, pages 217 - 224
URNOV ET AL., NAT REV GENET., vol. 11, no. 9, 2010, pages 636 - 46
VAN CAMP ET AL., PLANT PHYSIOL., vol. 112, no. 2, 1996, pages 525 - 535
VAN DAMME ET AL., PLANT MOL. BIOL., vol. 24, 1994, pages 825
VAN DER MEER ET AL., PLANT J., vol. 2, no. 4, 1992, pages 525 - 535
VELTEN ET AL., EMBO J., vol. 3, 1984, pages 2723 - 2730
VELTEN ET AL., EMBO J., vol. 3, 1984, pages 2723 - 30
VERWAERT ET AL., PLANT MOL. BIOL., vol. 26, 1994, pages 189 - 202
VILARDELL ET AL., PLANT MOL. BIOL., vol. 17, 1991, pages 985 - 93
VITALE ALESSANDRO ET AL: "The endoplasmic reticulum: Gateway of the secretory pathway", THE PLANT CELL, AMERICAN SOCIETY OF PLANT BIOLOGISTS, US, vol. 11, no. 4, 1 April 1999 (1999-04-01), pages 615 - 628, XP002137306, ISSN: 1040-4651, DOI: 10.1105/TPC.11.4.615 *
VON HEIJNE, G.: "A new method for predicting signal sequence cleavage sites", NUCLEIC ACIDS RES., vol. 14, 1986, pages 4683
VON HEIJNE, G.: "Sequence Analysis in Molecular Biology: Treasure Trove or Trivial Pursuit", 1987, ACAD. PRESS, pages: 113 - 117
WALBOT, CURR. OPIN. PLANT BIOL., vol. 2, 2000, pages 103 - 107
WALKER ET AL., PNAS, vol. 84, no. 19, 1987, pages 6624 - 6628
WANG; WATERHOUSE, CURR. OPIN. PLANT BIOL., vol. 5, 2001, pages 146 - 150
WARD ET AL., PLANT MOL. BIOL., vol. 22, 1993, pages 361 - 366
WATERHOUSE ET AL., PROC. NATL. ACAD. SCI. USA, vol. 95, 1998, pages 13959 - 13964
WATERHOUSE; HELLIWELL, NAT. REV. GENET., vol. 4, 2003, pages 29 - 38
WATERS ET AL., J. EXPERIMENTAL BOTANY, vol. 47, 1996, pages 325 - 338
WEIDER ET AL., CROP SCI., vol. 49, 2009, pages 77 - 84
WEIGEL ET AL., CELL, vol. 69, 1992, pages 843 - 859
WEISSINGER ET AL., ANN. REV. GENET., vol. 22, 1988, pages 421 - 477
WERNER ET AL., PNAS, vol. 18, 2001, pages 10487 - 10492
WERNER ET AL., PNAS, vol. 98, 2001, pages 10487 - 10492
WESLEY ET AL., PLANT J., vol. 27, 2001, pages 581 - 590
WILKINS ET AL., PLANT CELL, vol. 2, 1990, pages 301 - 13
WILLIAMSON ET AL., EUR. J. BIOCHEM., vol. 165, 1987, pages 99 - 106
WISSENBACH ET AL., PLANT J., vol. 4, 1993, pages 411
WLIHELM ET AL., PLANT MOL BIOL, vol. 23, 1993, pages 1073 - 1077
WOOTEN; FEDERHEN, COMPUT. CHEM., vol. 17, 1993, pages 149 - 63
WU, PLANT CELL PHYSIOLOGY, vol. 39, 1998, pages 885 - 889
WYBORSKI ET AL., NUCLEIC ACIDS RES., vol. 19, 1991, pages 4647 - 4653
YAMAGUCHI-SHINOZAKI ET AL., MOL. GEN. GENETICS, vol. 236, 1993, pages 331 - 340
YAMAGUCHI-SHINOZAKI ET AL., PLANT MOL. BIOL., vol. 15, 1990, pages 905
YAMAMOTO ET AL., PLANT CELL PHYSIOL., vol. 35, no. 5, 1994, pages 773 - 778
YAMAMOTO ET AL., PLANT J., vol. 12, no. 2, 1997, pages 255 - 265
YAMAO ET AL., PROC. NATL. ACAD. SCI. USA, vol. 82, 1985, pages 2306 - 9
YAO ET AL., CELL, vol. 71, 1992, pages 63 - 72
YARRANTON, CURR. OPIN. BIOTECH., vol. 3, 1992, pages 506 - 511
YOUNG ET AL., PLANT PHYSIOL., vol. 105, no. 2, pages 759 - 760
YU ET AL., PHYTOCHEMISTRY, vol. 63, 2003, pages 753 - 763
ZAMBRETTI ET AL., PROC. NATL. ACAD. SCI. USA, vol. 89, 1992, pages 3952 - 3956
ZANG ET AL., BIOTECHNOLOGY, vol. 9, 1991, pages 996
ZANG ET AL., PLANT SCIENCES, vol. 129, 1997, pages 81 - 89
ZHANG ET AL., PROC. NATL. ACAD. SCI. USA, vol. 94, 1997, pages 4504 - 9
ZHAO ET AL., MOL. BREED., vol. 8, 2001, pages 323 - 333
ZHAO ET AL., NATURE BIOTECH, vol. 16, 1998, pages 258 - 61
ZHAO, METH. MOL. BIOL., vol. 318, 2006, pages 315 - 323
ZHU ET AL., THE PLANT CELL, vol. 7, 1995, pages 1681 - 89

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10113181B2 (en) 2013-03-12 2018-10-30 Pioneer Hi-Bred International, Inc. Manipulation of dominant male sterility
US10822614B2 (en) 2013-03-12 2020-11-03 Pioneer Hi-Bred International, Inc. Manipulation of dominant male sterility
US11124797B1 (en) * 2015-06-04 2021-09-21 Shandong Agricultural University Wheat male-sterility gene WMS and its anther-specific expression promoter and uses thereof
US11795466B2 (en) 2015-06-04 2023-10-24 Spring Valley Agriscience Co., Ltd. Wheat-male sterility gene WMS and its anther-specific expression promoter and uses thereof
DE102016106656A1 (en) 2016-04-12 2017-10-12 Kws Saat Se Nuclear encoded male sterility by mutation in cytochrome P450 oxidase
WO2017178541A1 (en) 2016-04-12 2017-10-19 Kws Saat Se Nucleus-encoded male sterility through mutation in cytochrome p450 oxidase
DE102016015741A1 (en) 2016-04-12 2017-11-30 Kws Saat Se Nuclear encoded male sterility by mutation in cytochrome P450 oxidase
US11473103B2 (en) 2016-04-12 2022-10-18 KWS SAAT SE & Co. KGaA Nucleus-encoded male sterility through mutation in cytochrome P450 oxidase
CN107667853A (en) * 2017-06-07 2018-02-09 湖南杂交水稻研究中心 The method for creating of the common line with genic sterile of rice and application
CN107667853B (en) * 2017-06-07 2020-06-23 湖南杂交水稻研究中心 Method for creating rice common genic male sterile line and application
CN116724886A (en) * 2023-08-08 2023-09-12 四川天能璟秀生物科技有限公司 Method for propagating corn cell nucleus male sterile line by color marking
CN116724886B (en) * 2023-08-08 2023-11-10 四川天能璟秀生物科技有限公司 Method for propagating corn cell nucleus male sterile line by color marking

Similar Documents

Publication Publication Date Title
US10870860B2 (en) Genetic reduction of male fertility in plants
US9631203B2 (en) Genetic reduction of male fertility in plants
US10548276B2 (en) Genetic reduction of male fertility in plants
US20160017350A1 (en) Compositions and methods of use of acc oxidase polynucleotides and polypeptides
US20150240254A1 (en) Genetic reduction of male fertility in plants
WO2013138309A1 (en) Genetic reduction of male fertility in plants
US20140259225A1 (en) Manipulation of serine/threonine protein phosphatases for crop improvement
US20140304857A1 (en) Maize stress related transcription factor 18 and uses thereof
CA2563344C (en) Cytokinin oxidase sequences and methods of use
US20170298382A1 (en) Genetic reduction of male fertility in plants

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 13710965

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 13710965

Country of ref document: EP

Kind code of ref document: A1