US20070162999A1 - Crop Plant Cystatin Proteinase Inhibitors and Methods of Use - Google Patents

Crop Plant Cystatin Proteinase Inhibitors and Methods of Use Download PDF

Info

Publication number
US20070162999A1
US20070162999A1 US11/674,765 US67476507A US2007162999A1 US 20070162999 A1 US20070162999 A1 US 20070162999A1 US 67476507 A US67476507 A US 67476507A US 2007162999 A1 US2007162999 A1 US 2007162999A1
Authority
US
United States
Prior art keywords
plant
sequences
nucleic acid
sequence
expression
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/674,765
Inventor
Daniel Altier
Zhongmeng Bao
Guihua Lu
Pedro Navarro Acevedo
Vincent Sewalt
Carl Simmons
Nasser Yalpani
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Pioneer Hi Bred International Inc
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
Priority to US11/674,765 priority Critical patent/US20070162999A1/en
Publication of US20070162999A1 publication Critical patent/US20070162999A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/81Protease inhibitors
    • C07K14/8107Endopeptidase (E.C. 3.4.21-99) inhibitors
    • C07K14/8139Cysteine protease (E.C. 3.4.22) inhibitors, e.g. cystatin
    • 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
    • C12N15/8271Phenotypically and genetically modified plants via recombinant DNA technology with agronomic (input) traits, e.g. crop yield for stress resistance, e.g. heavy metal resistance
    • C12N15/8279Phenotypically and genetically modified plants via recombinant DNA technology with agronomic (input) traits, e.g. crop yield for stress resistance, e.g. heavy metal resistance for biotic stress resistance, pathogen resistance, disease resistance

Definitions

  • the invention relates to the field of the genetic manipulation of plants, particularly the modulation of gene activity and development in plants resulting in improvements in agronomic traits.
  • Agronomic traits such as disease resistance, nutritional quality, senescence and cell proliferation, have been subject to improvement attempts by various methods in the past. Often, improvements are attempted through plant breeding methods, which are often very expensive and of uncertain success. More recently, genetic modifications, such as those creating transgenic plants, have been used in attempts to reach these trait improvement goals. These approaches are meeting with varied success. No one strategy or gene has proven to be a panacea, although some show promise. Successful broad improvement of crop disease resistance, among other traits, will require multiple strategies. The addition of novel genes and methods is especially of value in the area of disease resistance, where pathogens are continually evolving and no single-gene method will have sustained success for long. Thus multiple genes and strategies for genetic improvement of agronomic traits are sought. This invention provides novel genes and methods of use through which agronomic traits can be improved.
  • compositions and methods relating to disease resistance and other plant agronomic traits are provided. Particularly, the nucleotide and amino acid sequences for cystatin homologs from maize, soybean, wheat and rice are provided.
  • the nucleotide sequences of the invention encode proteinase inhibitors of the cystatin cysteine proteinase inhibitor class.
  • the cystatin genes of the present invention may find use in enhancing agronomic traits of plants, including a wide variety of crop plants.
  • the compositions and methods of the invention can be used to manipulate the plant pathogen defense system, the control of senescence, the control of cell proliferation and cell death, and the nutritional quality of plant seeds intended for human and animal consumption.
  • the methods involve stably transforming a plant with a nucleotide sequence capable of modulating the production of one or more cystatins in the plant, operably linked with a promoter capable of driving expression of a gene in a plant cell.
  • the present invention is directed to an isolated polynucleotide set forth in SEQ ID NOs: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 43, 45, 47, 49, 51, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, and 75, isolated polynucleotides encoding the amino acid sequences set forth in SEQ ID NOs: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, and 76, isolated polypeptides having the sequences set forth in SEQ ID NOs: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64
  • Transformed plants, plant cells, and seeds, as well as methods for making such plants, plant cells, and seeds are additionally provided. It is recognized that a variety of promoters will be useful in the invention, the choice of which will depend in part upon the desired level of expression of the disclosed genes. It is recognized that the levels of expression can be controlled to modulate the levels of expression in the plant cell.
  • Further embodiments of the invention include methods of enhancing disease resistance of plants; methods of modulating the timing of plant maturation; methods of reducing cell death in plant tissue culture preparations; and methods of modulating protein digestibility and energy availability in plant products; wherein the preceding methods use plants transformed with the nucleotide sequences of the present invention.
  • FIG. 1 shows the chemical reaction inhibited by the cystatins of the present invention.
  • nucleic acids are written left to right in 5′ to 3′ orientation and amino acid sequences are written left to right in amino to carboxy orientation, respectively.
  • Numeric ranges recited within the specification are inclusive of the numbers defining the range and include each integer within the defined 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 IUPAC-IUB Biochemical Nomenclature Commission.
  • Nucleotides, likewise, may be referred to by their commonly accepted single-letter codes.
  • software, electrical, and electronics terms as used herein are as defined in The New IEEE Standard Dictionary of Electrical and Electronics Terms (5 th edition, 1993). The terms below are more fully defined by reference to the specification as a whole.
  • “Amplified” means 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 , D. H. Persing et al., Ed., American Society for Microbiology, Washington, D.C. (1993). The product of amplification is termed an amplicon.
  • antisense orientation includes reference to a duplex polynucleotide sequence that is operably linked to a promoter in an orientation where the antisense strand is transcribed.
  • the antisense strand is sufficiently complementary to an endogenous transcription product such that translation of the endogenous transcription product is often inhibited.
  • Encoding or “encoded”, with respect to a specified nucleic acid, means 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.
  • the amino acid sequence is encoded by the nucleic acid using the “universal” genetic code.
  • variants of the universal code such as are present in some plant, animal, and fungal mitochondria, the bacterium Mycoplasma capricolum , or the ciliate Macronucleus , may be used when the nucleic acid is expressed therein.
  • nucleic acid sequences of the present invention may be expressed in both monocotyledonous and dicotyledonous plant species, sequences can be modified to account for the specific codon preferences and GC content preferences of monocotyledons or dicotyledons as these preferences have been shown to differ (Murray et al. Nucl. Acids Res. 17: 477-498 (1989)).
  • the maize preferred codon for a particular amino acid may be derived from known gene sequences from maize. Maize codon usage for 28 genes from maize plants is listed in Table 4 of Murray et al., supra.
  • full-length sequence in reference to a specified polynucleotide or its encoded protein means having the entire amino acid sequence of, a native (non-synthetic), endogenous, biologically active form of the specified protein.
  • Methods to determine whether a sequence is full-length are well known in the art including such exemplary techniques as northern or western blots, primer extension, S1 protection, and ribonuclease protection. See, e.g., Plant Molecular Biology: A Laboratory Manual , Clark, Ed., Springer-Verlag, Berlin (1997). Comparison to known full-length homologous (orthologous and/or paralogous) sequences can also be used to identify full-length sequences of the present invention.
  • consensus sequences typically present at the 5′ and 3′ untranslated regions of mRNA aid in the identification of a polynucleotide as full-length.
  • the consensus sequence ANNNN AUG G where the underlined codon represents the N-terminal methionine, aids in determining whether the polynucleotide has a complete 5′ end.
  • Consensus sequences at the 3′ end such as polyadenylation sequences, aid in determining whether the polynucleotide has a complete 3′ end.
  • 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 nucleotide sequence can be from a species different from that from which the nucleotide sequence was derived, or, if from the same species, the promoter is not naturally found operably linked to the nucleotide sequence.
  • 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” means a cell which contains a vector and supports the replication and/or expression of the vector.
  • Host cells may be prokaryotic cells such as E. coli , or eukaryotic cells such as yeast, insect, amphibian, or mammalian cells, excluding human cells.
  • host cells are monocotyledonous or dicotyledonous plant cells.
  • a particularly preferred monocotyledonous host cell is a maize host cell.
  • 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: (1) substantially or essentially free from components that normally accompany or interact with it as it is found in its naturally occurring environment.
  • the isolated material optionally comprises material not found with it in its natural environment; or (2) if the material is in its natural environment, the material has been synthetically (non-naturally) altered by deliberate human intervention to a composition and/or placed at a location in the cell (e.g., genome or subcellular organelle) not native to a material found in that environment.
  • the alteration to yield the synthetic material can be performed on the material within or removed from its natural state.
  • a naturally occurring nucleic acid becomes an isolated nucleic acid if it is altered, or if it is transcribed from DNA which has been altered, by means of human intervention performed within the cell from which it originates. See, e.g., Compounds and Methods for Site Directed Mutagenesis in Eukaryotic Cells, Kmiec, U.S. Pat. No. 5,565,350; In Vivo Homologous Sequence Targeting in Eukaryotic Cells; Zarling et al., PCT/US93/03868.
  • a naturally occurring nucleic acid e.g., a promoter
  • Nucleic acids which are “isolated” as defined herein, are also referred to as “heterologous” nucleic acids.
  • nucleic acid and “polynucleotide” are used interchangeably and include reference to a deoxyribonucleotide or ribonucleotide polymer, or chimeras thereof, 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.
  • a polynucleotide can be full-length or a subsequence of a native or heterologous structural or regulatory gene. Unless otherwise indicated, the term includes reference to the specified sequence as well as the complementary sequence thereof.
  • DNAs or RNAs with backbones modified for stability or for other reasons are “polynucleotides” as that term is intended herein.
  • DNAs or RNAs comprising unusual bases, such as inosine, or modified bases, such as tritylated bases, to name just two examples are polynucleotides as the term is used herein. It will be appreciated that a great variety of modifications have been made to DNA and RNA that serve many useful purposes known to those of skill in the art.
  • polynucleotide as it is employed herein embraces such chemically, enzymatically or metabolically modified forms of polynucleotides, as well as the chemical forms of DNA and RNA characteristic of viruses and cells, including among other things, simple and complex cells.
  • Nucleic acid library means a collection of isolated DNA or RNA molecules which comprise and substantially represent the entire transcribed fraction of a genome of a specified organism or of a tissue from that 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, Guide to Molecular Cloning Techniques, Methods in Enzymology , Vol. 152, Academic Press, Inc., San Diego, Calif. (Berger); Sambrook et al., Molecular Cloning—A Laboratory Manual, 2nd ed., Vol. 1-3 (1989) (hereinafter Sambrook); and Current Protocols in Molecular Biology , F. M. Ausubel et al., Eds., Current Protocols, a joint venture between Greene Publishing Associates, Inc. and John Wiley & Sons, Inc. (1994) (hereinafter Ausubel).
  • operably linked includes reference to a functional linkage between a promoter and a second sequence, wherein the promoter sequence initiates and mediates transcription of the DNA sequence 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 (e.g., leaves, stems, roots, etc.), seeds and plant cells and progeny of same.
  • Plant cell as used herein includes, without limitation, seeds, suspension cultures, embryos, meristematic regions, callus tissue, leaves, roots, shoots, gametophytes, sporophytes, pollen, and microspores.
  • the classes of plants which can be used in the methods of the invention include both monocotyledonous and dicotyledonous plants.
  • a particularly preferred plant is Zea mays.
  • 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.
  • the essential nature of such analogues of naturally occurring amino acids is that, when incorporated into a protein, that protein is specifically reactive to antibodies elicited to the same protein but consisting entirely of naturally occurring amino acids.
  • polypeptide “peptide” and “protein” are also inclusive of modifications including, but not limited to, glycosylation, lipid attachment, sulfation, gamma-carboxylation of glutamic acid residues, hydroxylation and ADP-ribosylation. Further, this invention contemplates the use of both the methionine-containing and the methionine-less amino terminal variants of the protein of the invention.
  • 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 whether or not its origin is from a plant cell. 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 of promoters under developmental control include promoters that preferentially initiate transcription in certain tissues, such as leaves, roots, or seeds. Such promoters are referred to as “tissue preferred”. Promoters which initiate transcription only in certain tissues are referred to as “tissue specific”.
  • 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 “repressible” 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. Tissue specific, tissue preferred, cell type specific, and inducible promoters constitute the class of “non-constitutive” promoters.
  • a “constitutive” promoter is a promoter which is active under most environmental conditions.
  • 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.
  • 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 host 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.
  • amino acid residue amino acid residue
  • amino acid amino acid
  • 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 80% sequence identity, 90% sequence identity, 95% or 100% sequence identity (i.e., complementary) with each other.
  • stringent conditions or “stringent hybridization conditions” includes reference to conditions under which a probe will selectively hybridize to its target sequence, to a detectably greater degree than to 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 are 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). Generally, a probe is less than about 1000 nucleotides in length, and optionally less than 500 nucleotides in length.
  • 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 a destabilizing agent such as formamide.
  • Exemplary moderate stringency conditions include hybridization in 40 to 45% formamide, 1 M NaCl, 1% SDS at 37° C., and a wash in 0.5 ⁇ to 1 ⁇ SSC at 55 to 60° C.
  • Exemplary high stringency conditions include hybridization in 50% formamide, 1 M NaCl, 1% SDS at 37° C. for at least 4 hours, more preferably up to 12 hours or longer, and a final wash in 0.1 ⁇ SSC at 60 to 65° C. for 30 minutes.
  • T m thermal melting point
  • % GC percentage of guanosine and cytosine nucleotides in the DNA
  • % form percentage of formamide in the hybridization solution
  • 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 T m for the specific sequence and its complement at a defined ionic strength and pH.
  • severely stringent conditions can utilize a hybridization and/or wash at 1, 2, 3, or 4° C. lower than the T m ;
  • moderately stringent conditions can utilize a hybridization and/or wash at 6, 7, 8, 9, or 10° C. lower than the T m ;
  • low stringency conditions can utilize a hybridization and/or wash at 11, 12, 13, 14, 15, or 20° C. lower than the T m .
  • T m 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.
  • 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 a 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 the introduction of a polynucleotide of the present invention into a host cell. Vectors are often replicons. Expression vectors permit transcription of a nucleic acid inserted therein.
  • sequence relationships between a polynucleotide/polypeptide of the present invention with a reference polynucleotide/polypeptide (a) “reference sequence”, (b) “comparison window”, (c) “sequence identity”, and (d) “percentage of sequence identity”.
  • a “reference sequence” is a defined sequence used as a basis for sequence comparison with a polynucleotide/polypeptide of the present invention.
  • a reference sequence may be a subset of, 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.
  • a “comparison window” includes reference to a contiguous and specified segment of a polynucleotide/polypeptide sequence, wherein the polynucleotide/polypeptide sequence may be compared to a reference sequence and wherein the portion of the polynucleotide/polypeptide 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 nucleotide or amino acid residues in length, and optionally can be 30, 40, 50, 100, or longer.
  • Optimal alignment of sequences for comparison may be conducted by the local alignment algorithm of Smith and Waterman, Adv. Appl. Math. 2: 482 (1981); by the global alignment algorithm of Needleman and Wunsch, J. Mol. Biol. 48: 443 (1970); by the local alignment method of Pearson and Lipman, Proc. Natl. Acad. Sci.
  • 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.
  • HSPs high scoring sequence pairs
  • Cumulative scores are calculated using, for nucleotide sequences, the parameters M (reward score for a pair of matching residues; always >0) and N (penalty score for mismatching residues; always ⁇ 0).
  • M forward score for a pair of matching residues; always >0
  • N penalty score for mismatching residues; always ⁇ 0.
  • a scoring matrix is used to calculate the cumulative score. Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached.
  • the BLAST algorithm parameters W, T, and X determine the sensitivity and speed of the alignment.
  • W wordlength
  • E expectation
  • BLOSUM62 scoring matrix see Henikoff & Henikoff (1989) Proc. Natl. Acad. Sci. USA 89:10915.
  • the BLAST algorithm In addition to calculating percent sequence identity, the BLAST algorithm also performs a statistical analysis of the similarity between two sequences (see, e.g., Karlin & Altschul, Proc. Nat'l. Acad. Sci. USA 90:5873-5877, 1993).
  • One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P(N)), which provides an indication of the probability by which a match between two nucleotide or amino acid sequences would occur by chance.
  • 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, Comput. Chem., 17:149-163, 1993) and XNU (Clayerie and States, Comput. Chem., 17:191-201, 1993) low-complexity filters can be employed alone or in combination.
  • GAP can also be used to compare a polynucleotide or polypeptide of the present invention with a reference sequence.
  • GAP uses the algorithm of Needleman and Wunsch ( J. Mol. Biol. 48:443-453, 1970) 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.
  • gap extension penalty greater than zero
  • 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 for protein sequences are 8 and 2, respectively.
  • the default gap creation penalty is 50 while the default gap extension penalty is 3.
  • the gap creation and gap extension penalties can be expressed as an integer selected from the group of integers consisting of from 0 to 200.
  • the gap creation and gap extension penalties can each independently be: 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, 50, 60, 65 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 & 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 al., Nucleic Acids Res. 25:3389-3402, 1997; Altschul et al., J. Mol. Bio. 215: 403-410, 1990) or to the value obtained using the GAP program using default parameters (see the Wisconsin Genetics Software Package, (Accelrys, Inc., San Diego, Calif.)).
  • 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 when the 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, Computer Applic. Biol. Sci., 4: 11-17 (1988) e.g., as implemented in the program PC/GENE (Intelligenetics, Mountain View, Calif., 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 at least 70% sequence identity, preferably at least 80%, more preferably at least 90%, and most preferably at least 95%, compared to a reference sequence using one of the alignment programs described using standard parameters.
  • sequence identity preferably at least 80%, more preferably at least 90%, and most preferably at least 95%.
  • the present invention provides, inter alia, compositions and methods for modulating the total level of proteins of the present invention and/or altering their ratios in a plant. “Modulation” is intended to mean an increase or decrease in a particular character, quality, substance, or response.
  • compositions comprise the nucleotide and amino acid sequence for 38 homologs of cysteine proteinases from maize, wheat, rice, and soybean, as presented in Table 1. These plant cystatin genes are characterized by their cysteine proteinase inhibitory activity.
  • Plant cystatin genes is intended to mean genes that are structurally related to plant cystatins, also known as plant cysteine proteinases.
  • proteinases are also called “proteases” and “peptidases” interchangeably.
  • cystin-like activity is intended to include the activity of peptides that inhibit the activity of cysteine proteinases in plants. In addition, at least some of these peptides also retain antifungal and/or antibacterial activity.
  • the genes of the present invention are called cystatins after a structural classification of proteins (SCOP) classification system.
  • Cystatins are a group of proteins which inhibit the activity of cysteine proteinases.
  • the cystatins identified in vertebrates, insects, and plants have been classified into four groups, all belonging to a cystatin superfamily.
  • Groups 1 through 3 are primarily vertebrate cystatin molecules, while group 4 comprises all the known plant cystatins.
  • Group 1 cystatins are referred to as the stefins, single chain proteins with molecular weights of about 11 kDa, which contain no disulfide bonds or carbohydrates.
  • the second group is referred to as the cystatins, and comprise single chain proteins of about 13 kDa, with two disulfide bonds located toward the carboxyl terminus.
  • the kininogens, group 3 are the largest of the cystatins. They are characterized by having three Type 2-like domains, bound carbohydrates, and an additional polypeptide (kinin) unrelated to the cystatin segments.
  • the cysteine proteinase inhibitors of plant origin have been grouped into a fourth cystatin family, the “phytocystatins,” or “plant cystatins” based on their sequence similarity and absence of disulfide bonds or cysteine residues.
  • the phytocystatins are single polypeptide chains with molecular weights ranging from 10 to 16 kDa. Many share several reported conserved sequence motifs, including glycine residue(s) in the vicinity of the N-terminal region, a Gln-Xaa-Val-Xaa-Gly (SEQ ID NO: 77) motif in the first hairpin loop, and a Pro-Trp in the second hairpin loop.
  • Table 2 shows the sequences of the first and second hairpin loops and their locations in the cystatin sequences of the present invention.
  • the highly conserved nature of the QXVXG (SEQ ID NO: 77) motif within the first hairpin loop is evident.
  • the second hairpin loop while less highly conserved than the first, generally presents a tryptophan residue at the end of the motif.
  • Table 3 shows the conserved region of the N-terminal alpha-1-helix in each of the sequences of the present invention.
  • the protein sequences of the present invention were analyzed for percent identities and similarities using the GAP algorithm. These analyses were performed by species, such that the maize sequences were compared to the other maize sequences, and so on for each of soybean, rice, and wheat. It is evident from the tables that follow that the sequences of the present invention, although they are all cystatins, can vary markedly at the protein level while still retaining cystatin activity. In Tables 4 through 11, sequence similarities and identities among crop plant sequence groups are presented. Those SEQ ID NOs: for which activity data are provided in the instant application are shown in bold faced type (see Examples).
  • the phytocystatins play a role in a wide range of plant physiological processes, including plant defense mechanisms.
  • Plant cystatins have been found in various tissues in numerous plant species, including, but not limited to, crop plants such as rice (Abe et al. (1987) J Biol Chem 262: 16793-16797; Kondo et al. (1990) J Biol Chem 265: 15832-15837), tomato (Wu et al. (2000) Comp Biochem Phys C 127: 209-220), maize (Yamada et al. (2000) Plant Cell Physiol 42(7): 710-716), sunflower (Doi-Kawano et al.
  • Cystatins play a role in many plant physiological functions, including defense, more specifically plant defense against pathogens.
  • a range of functions performed by plant cystatins are responsible for enhancing plant defense against different pathogens.
  • the sequences and related genes of the present invention encode proteins with antimicrobial and antifungal activity. These proteins may inhibit the proteinases of the pathogen, so as to thwart their utilization of the plant tissue.
  • cystatins which are expressed around disease-induced lesions may control symptom development, as in a hypersensitive response (HR), by controlling the proteinase-mediated cell death mechanism.
  • HR hypersensitive response
  • compositions of the present invention include the sequences for maize, soybean, rice and wheat nucleotide sequences which have been identified as cystatins that are involved in plant defense response and development.
  • the present invention provides for isolated nucleic acid molecules comprising nucleotide sequences encoding the amino acid sequences shown in SEQ ID NOs: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, and 76.
  • polypeptides having an amino acid sequence encoded by a nucleic acid molecule described herein for example those nucleotide sequences set forth in SEQ ID NOs: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 43, 45, 47, 49, 51, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, and 75.
  • the nucleotide sequences of the invention are maize, soybean, rice and wheat sequences comprising plant cysteine proteinases.
  • the claimed sequences are members of the plant cystatin class of genes and polypeptides. These plant cystatins are identified herein as “Zm-Cys”, “Ta-Cys”, “Gm-Cys” and “Os-Cys” for cystatins originating from Zea mays, Triticum aestivum, Glycine max and Oryza sativa , respectively, and are numbered for easy reference (e.g. Zm-Cys10 or Gm-Cys8). These sequences represent a diverse and conserved supergene family in plants.
  • compositions of the invention can be used in a variety of methods whereby the protein products can be expressed in crop plants to function as antimicrobial proteins. Such expression results in the alteration or modulation of the level, tissue, or timing of expression to achieve enhanced disease or stress resistance.
  • the compositions of the invention may be expressed in the same species from which the particular cystatin originates, or alternatively, can be expressed in any plant of interest.
  • the coding sequence for the cystatin can be used in combination with a promoter that is introduced into a crop plant.
  • a high-level expressing constitutive promoter may be utilized and would result in high levels of expression of the cystatin.
  • the coding sequence may be operably linked to a tissue-specific promoter to direct the expression to a plant tissue known to be susceptible to a pathogen.
  • tissue-specific promoter to direct the expression to a plant tissue known to be susceptible to a pathogen.
  • manipulation of the timing of expression may be utilized. For example, by judicious choice of promoter, expression can be enhanced early in plant growth to prime the plant to be responsive to pathogen attack.
  • pathogen inducible promoters can be used wherein expression of the cystatin is turned on in the presence of the pathogen.
  • the cystatin genes of the present invention additionally find use in enhancing the plant pathogen defense system.
  • the compositions and methods of the invention can be used for enhancing resistance to plant pathogens including fungal pathogens, plant viruses, and the like.
  • the method involves stably transforming a plant with a nucleotide sequence capable of modulating the plant pathogen defense system operably linked with a promoter capable of driving expression of a gene in a plant cell.
  • “Enhancing resistance” means that the plant's tolerance to pathogens is increased. That is, the cystatin may slow or prevent pathogen infection and spread.
  • methods for increasing pathogen resistance in a plant comprise stably transforming a plant with a DNA construct comprising an anti-pathogenic nucleotide sequence of the invention operably linked to a promoter that drives expression in a plant.
  • a promoter that drives expression in a plant.
  • Such methods find use in agriculture, particularly in limiting the impact of plant pathogens on crop plants. While the choice of promoter will depend on the desired timing and location of expression of the anti-pathogenic nucleotide sequences, preferred promoters include constitutive and pathogen-inducible promoters.
  • compositions can be used in formulations used for their disease resistance activities.
  • the proteins of the invention can be formulated with an acceptable carrier into a pesticidal composition(s) that is for example, a suspension, a solution, an emulsion, a dusting powder, a dispersible granule, a wettable powder, an emulsifiable concentrate, an aerosol, an impregnated granule, an adjuvant, a coatable paste, or an encapsulation in, for example, polymer substances.
  • Transformed plants, plant cells, plant tissues and seeds thereof are additionally provided.
  • the present invention is not dependent upon a particular mechanism of defense. Rather, the genes and methods of the invention work to increase resistance of the plant to pathogens independent of how that resistance is increased or achieved.
  • cystatin sequences find use in disrupting cellular function of plant pathogens or insect pests as well as altering the defense mechanisms of a host plant to enhance resistance to disease or insect pests. While the invention is not bound by any particular mechanism of action to enhance disease resistance, the gene products, probably proteins or polypeptides, function to inhibit or prevent diseases in a plant.
  • any one of a variety of second nucleotide sequences may be utilized, embodiments of the invention encompass those second nucleotide sequences that, when expressed in a plant, help to increase the resistance of a plant to pathogens. It is recognized that such second nucleotide sequences may be used in either the sense or antisense orientation depending on the desired outcome.
  • Other plant defense proteins include those described in PCT patent publications WO 99/43823 and WO 99/43821, both of which are herein incorporated by reference.
  • Plant senescence is an important trait affecting life cycle duration or maturity, seed dry down, the disease resistance profile, and ‘stay green’, which in turn affect yield, stalk strength, appearance, and nutritional value (silage quality). All of these factors, which relate to cell death processes, are considered in maize breeding efforts. Plant proteinases have been implicated in these processes and thus comprise an area of active research.
  • cysteine proteinases are induced upon plant organ senescence, such as in tomato leaves (Drake et al. (1996) Plant Mol Biol 30(4): 755-767), sweet potato (Chen et al. (2002) Plant and Cell Phys 43(9): 984-991), and day-lily flowers (Valpuesta et al. (1995) Plant Mol Biol 28(3): 575-582).
  • the maize cysteine proteinase Seel (“Senescence enhanced”) has been linked to the stay green phenotype in maize (Griffiths et al. (1997) Plant Mol Biol 34: 815-821). Cysteine proteinase inhibitors have also been shown to delay flower senescence (Eason et al.
  • cystatins proteinsase inhibitors
  • the position of the starting breeding material relative to the desired outcome will dictate what direction one will want to push a trait. For example, one may want increase or decrease maturity time; generally decrease. To increase senescence one may want to suppress cystatin expression, and to decrease senescence one may want to increase cystatin expression. Methods for increasing or decreasing cystatin expression are outlined elsewhere in this specification. However, tissue-targeted or developmental-targeted expression may be desirable to reach these ends.
  • the proteinase promoters such as those from Seel, can be useful in conjunction with forward or antisense constructs of the proteinase inhibitor gene in question, to coordinately augment or cancel, respectively, the death-promoting capacity of the cysteine proteinase.
  • the proteinase inhibitor genes herein are useful for controlling the senescence of special crop plant tissues.
  • tissue or organs are desired to senesce. This includes controlled dropping of cotton leaves to facilitate cotton boll harvesting.
  • organs are desired not to senesce, as in the petioles of fruit; premature fruit drop can cause loss of yield.
  • delay of senescence of the pedicel/hilum region of kernels may be desirable to allow for prolonged kernel fill or delayed maturation of seed, with higher yield and/or higher digestibility as possible outcomes.
  • PIs proteinase inhibitors
  • Oryzacystatin-I is a cysteine proteinase inhibitor from rice seeds (Abe et al. (1987) Supra), while Oc-1D86 is a modified form of Oc-1 which has shown stronger inhibitory activity (Urwin et al. (1995) Plant J 8: 121-131). When expressed in tomato hairy roots both Oc-1 and Oc-ID86 had a detrimental effect on the growth and development of potato cyst nematode G. pallida (Id.).
  • compositions of the instant invention indicate that cystatins can also confer resistance to soybean cyst nematode (SCN) in soybean and other crops, by inhibiting nematode growth and development.
  • SCN soybean cyst nematode
  • these proteinase inhibitors can retard cell death by suppressing proteinase inhibitor activity, they can be used to help transformed cells survive.
  • Cysteine proteinases are known to be induced in the plant HR response, and transgenic ectopically expressed cystatins can counteract this response (Pechan et al. (2000) Plant Cell 12(7): 1031-1040; Solomon et al. (1999) Plant Cell 11 (3): 431-443).
  • the transformed cells receive a copy of one or more of these proteinase inhibitor coding region(s) driven by an appropriate promoter.
  • promoter choices are discussed elsewhere in this application, however, this embodiment can benefit from the use of a constitutive promoter, or by a transiently expressed promoter targeted to the cell culture phase or induced by plant hormones used in culture.
  • Constitutive expression may help disease resistance generally, and as such, constitutive promoters, for example the ubiquitin promoter, can be useful beyond cell culture.
  • constitutive promoters for example the ubiquitin promoter
  • a variety of promoters would be effective.
  • the resulting transformed cells would be more viable. This would effect a cleaner separation of the dying non-transformed cells and allow for cleaner and more rapid growth of the transformed line. Plant transformation techniques would be improved as a result.
  • plants can be wounded abiotically, as by drought stress, wind stress (which includes damage by wind-blown soil particles), and chemical and nutrient stress. Such stresses can precipitate cell death that can reduce plant yield. To the extent that these proteinase inhibitors may retard cell death by thwarting proteinase inhibitor activity, they can retard the symptom development of necrosis resulting from these stresses when driven by a death-induced promoter.
  • the proteinase inhibitor genes can have application in the development and implementation of herbicide resistance mechanisms in crop plants. Ectopic expression of the proteinase inhibitors, as in leaves, can result in a retardation of cell death following the application of herbicides. This would be subject to the kind of herbicide used and its mode of action, but it is an area of utility for these genes. Herbicides and herbicide resistance systems are often used as selectable markers in plant transformation experiments. Thus, in a way similar to the herbicide resistance application, these proteinase inhibitor genes can be used as selectable markers—only cells expressing the proteinase inhibitor genes (ectopically) would grow or stay alive in the face of an antibiotic/herbicide medium. This application of course bears direct overlap with the examples given above for improving plant transformation.
  • Cell death can also be a mechanism of male infertility. Consequently similar methods, probably with anther- or tapetum- or pollen-preferred expression, could be a means of enhancing or controlling male fertility. For example, expressing cystatins can suppress cell death and thus suppress sterility, rendering the plants male fertile. This could be used in a conditional situation, where the plants would be sterile until induced to be fertile.
  • Proteinase inhibitors are expressed in seeds.
  • the chief biological role of seed expression of proteinase inhibitors is to inhibit, or otherwise control, proteinase activities in the seeds. This is especially important during seed development/maturation, in order to regulate protein processing by proteinases.
  • Cereal cysteine proteinases play a chief role in the digestion of seed storage proteins, especially during germination (Gruis et al. (2002) Plant Cell 14(11):2863-2882; Debarros & Larkins (1994) Plant Sci 99(2) 189-197; Koehler & Ho (1990) Plant Physiol 94(1):251-258; Poulle & Jones (1988) Plant Physiol 88(4): 1454-1460).
  • Regulating the activity of cysteine proteinases in seeds prevents undesirable loss of seed proteins, including storage proteins, and also prevents premature germination (Corre et al (2002) Plant Mol Biol 50(4-5):687-698). Furthermore, regulating the processing of proteins can serve as an anti-nutritional/protective agent against microbes, insects, and herbivores.
  • crop plant seeds such as maize caryopses
  • crop plant seeds are mostly intended for animal consumption as feed grain, and some also for human food consumption.
  • the proteinase inhibitors from the seed can inhibit digestive proteinases in the gastrointestinal tract of livestock and humans. This can change the site and extent of digestion of protein and other grain components within, as well as elicit hyper-secretion of pancreatic enzymes.
  • the impact on overall nutritional status may either be positive or negative.
  • NPN non-protein-nitrogen
  • ruminant livestock airy and beef cattle, sheep and goats
  • protein is of high Biological Value (i.e., of balanced amino acid composition, containing especially lysine, tryptophan, threonine, and methionine).
  • silage made of various forages, such as ryegrass and alfalfa is subject to excessive proteolysis during the ensiling process. Total protein losses can amount to 50% and the dairy cow poorly utilizes the resulting NPN. Proteinase inhibitors can be employed to reduce these proteolytic losses.
  • lowering the proteolysis in the rumen is beneficial to allow otherwise easily digestible high-protein concentrates (such as fat-extracted soybean & canola meals) and high-protein forages (such as alfalfa) to bypass rumen fermentation.
  • Rapid and extensive ruminal breakdown of protein leads to decreased protein efficiency because 1) the rumen microbes do not use the degraded protein as fast as it is broken down, leading to excessive formation of ammonia, much of which will be excreted in the urine as urea, and 2) the microbial protein that is re-synthesized from ammonia is generally of lower biological value than soybean or canola protein.
  • cystatin proteinase inhibitors can alter digestion characteristics of the grain, it would be desirable to reduce the level of their expression to increase protein digestibility and energy availability for monogastric livestock and humans.
  • the different proteinase inhibitor genes have somewhat different expression profiles. Based upon the maize EST library distributions, Zm-Cys5, Zm-Cys6, Zm-Cys9, Zm-Cys10, Zm-Cys13, and Zm-Cys14 are abundant in, or specific to, maize endosperm or other kernel tissues. Generally, the best mode for this invention (in maize or cereals) is to express the sense version of the proteinase inhibitor genes under the control of a seed-preferred, especially endosperm-preferred, especially R3-R5-preferred promoter, or, in the case of alfalfa, a constitutive promoter.
  • the sense transcript when produced, it will result in either over-expression or silencing of the targeted proteinase inhibitor gene.
  • Sequences of the invention encompass coding sequences, antisense sequences, and fragments and variants thereof. Expression of the sequences of the invention can be used to modulate or regulate the expression of corresponding cystatin proteins.
  • the invention encompasses isolated or substantially purified nucleic acid or protein compositions. An “isolated” or “purified” nucleic acid molecule or protein, or biologically active portion thereof, is substantially free of other cellular material, or culture medium when produced by recombinant techniques, or substantially free of chemical precursors or other chemicals when chemically synthesized.
  • an “isolated” nucleic acid is free of sequences (preferably protein encoding sequences) that naturally flank the nucleic acid (i.e., sequences located at the 5′ and 3′ ends of the nucleic acid) in the genomic DNA of the organism from which the nucleic acid is derived.
  • the isolated nucleic acid molecule can contain less than about 5 kb, 4 kb, 3 kb, 2 kb, 1 kb, 0.5 kb, or 0.1 kb of nucleotide sequences that naturally flank the nucleic acid molecule in genomic DNA of the cell from which the nucleic acid is derived.
  • a protein that is substantially free of cellular material includes preparations of protein having less than about 30%, 20%, 10%, 5%, (by dry weight) of contaminating protein.
  • culture medium represents less than about 30%, 20%, 10%, or 5% (by dry weight) of chemical precursors or non-protein-of-interest chemicals.
  • Fragments and variants of the disclosed nucleotide sequences and proteins encoded thereby are also encompassed by the present invention.
  • “Fragment” means a portion of the nucleotide sequence or a portion of the amino acid sequence and hence protein encoded thereby.
  • Fragments of a nucleotide sequence may encode protein fragments that retain the biological activity of the native protein and hence have cystatin-like activity and thereby affect development, developmental pathways, and defense responses.
  • fragments of a nucleotide sequence that are useful as hybridization probes generally do not encode fragment proteins retaining biological activity.
  • fragments of a nucleotide sequence may range from at least about 20 nucleotides, about 50 nucleotides, about 100 nucleotides, and up to the full-length nucleotide sequence encoding the proteins of the invention.
  • a fragment of a cystatin nucleotide sequence that encodes a biologically active portion of a cystatin protein of the invention will encode at least 15, 25, 30, 50, 100, 150, 200, or 250 contiguous amino acids, or up to the total number of amino acids present in a full-length protein of the invention (for example, 135, 134, 134, 245, 176, 116, 110 or 157 amino acids for SEQ ID NO:2, 4, 6, 8, 10, 12, 14, or 16, respectively).
  • Fragments of a cystatin nucleotide sequence that are useful as hybridization probes for PCR primers generally need not encode a biologically active portion of a cystatin protein.
  • a fragment of a cystatin nucleotide sequence may encode a biologically active portion of a cystatin protein, or it may be a fragment that can be used as a hybridization probe or PCR primer using methods disclosed below.
  • a biologically active portion of a cystatin protein can be prepared by isolating a portion of one of the cystatin nucleotide sequences of the invention, expressing the encoded portion of the cystatin protein (e.g., by recombinant expression in vitro), and assessing the activity of the encoded portion of the cystatin protein.
  • Nucleic acid molecules that are fragments of a cystatin nucleotide sequence comprise at least 16, 20, 50, 75, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, or 800 nucleotides, or up to the number of nucleotides present in a full-length cystatin nucleotide sequence disclosed herein (for example, 408, 405, 405, 738, 531, 351, 333, or 474 nucleotides for SEQ ID NO:1, 3, 5, 7, 9, 11, 13, or 15, respectively).
  • “Variants” is intended to mean substantially similar sequences.
  • conservative variants include those sequences that, because of the degeneracy of the genetic code, encode the amino acid sequence of one of the cystatin polypeptides of the invention.
  • Naturally occurring allelic variants such as these can be identified with the use of well-known molecular biology techniques, as, for example, with polymerase chain reaction (PCR) and hybridization techniques as outlined below.
  • Variant nucleotide sequences also include synthetically derived nucleotide sequences, such as those generated, for example, by using site-directed mutagenesis but which still encode a cystatin protein of the invention.
  • variants of a particular nucleotide sequence of the invention will have at least about 50%, 60%, 65%, 70%, generally at least about 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to that particular nucleotide sequence as determined by sequence alignment programs described elsewhere herein using default parameters.
  • variant nucleotide sequences can also be evaluated by comparison of the percent sequence identity shared by the polypeptides they encode.
  • isolated nucleic acids which encode a polypeptide with a given percent sequence identity to the polypeptide of SEQ ID NO: 2, 4, 6, 8 and 10 are disclosed.
  • Identity can be calculated using, for example, the BLAST, CLUSTALW, or GAP algorithms under default conditions. The percentage of identity to a reference sequence is at least 50% and, rounded upwards to the nearest integer, can be expressed as an integer selected from the group of integers consisting of from 50 to 99.
  • the percentage of identity to a reference sequence can be at least 60%, 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%.
  • a “variant” protein is intended to mean a protein derived from the native protein by deletion (so-called truncation) or addition of one or more amino acids to the N-terminal and/or C-terminal end of the native protein; deletion or addition of one or more amino acids at one or more sites in the native protein; or substitution of one or more amino acids at one or more sites in the native protein.
  • Variant proteins encompassed by the present invention are biologically active, that is they continue to possess the desired biological activity of the native protein, that is, cystatin-like activity as described herein. Such variants may result from, for example, genetic polymorphism or from human manipulation.
  • Biologically active variants of a native cystatin protein of the invention will have at least about 40%, 50%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to the amino acid sequence of the native protein as determined by sequence alignment programs described elsewhere herein using default parameters.
  • a biologically active variant of a protein of the invention may differ from that protein by as few as 1-15 amino acid residues, as few as 1-10, such as 6-10, as few as 5, as few as 4, 3, 2, or even 1 amino acid residue.
  • polypeptides of the invention may be altered in various ways including amino acid substitutions, deletions, truncations, and insertions.
  • Novel proteins having properties of interest may be created by combining elements and fragments of proteins of the present invention as well as other proteins. Methods for such manipulations are generally known in the art.
  • amino acid sequence variants of the cystatin proteins can be prepared by mutations in the DNA. Methods for mutagenesis and nucleotide sequence alterations are well known in the art. See, for example, Kunkel (1985) Proc Nat Acad Sci USA 82:488-492; Kunkel et al. (1987) Method Enzymol 154:367-382; U.S. Pat. No.
  • genes and nucleotide sequences of the invention include both the naturally occurring sequences as well as mutant forms.
  • proteins of the invention encompass both naturally occurring proteins as well as variations and modified forms thereof. Such variants will continue to possess the desired developmental activity, or defense response activity.
  • mutations that will be made in the DNA encoding the variant must not place the sequence out of reading frame and preferably will not create complementary regions that could produce secondary mRNA structures. See, EP Patent Application Publication No. 0075444.
  • polypeptides are produced as complex precursors which, in addition to targeting labels such as the signal peptides discussed elsewhere in this application, also contain other fragments of peptides which are removed (processed) at some point during protein maturation, resulting in a mature form of the polypeptide that is different from the primary translation product (aside from the removal of the signal peptide).
  • “Mature protein” refers to a post-translationally processed polypeptide; i.e., one from which any pre- or propeptides present in the primary translation product have been removed.
  • Precursor protein” or “prepropeptide” or “preproprotein” all refer to the primary product of translation of mRNA; i.e., with pre- and propeptides still present.
  • Pre- and propeptides may include, but are not limited to, intracellular localization signals. “Pre” in this nomenclature generally refers to the signal peptide. The form of the translation product with only the signal peptide removed but not further processing yet is called a “propeptide” or “proprotein”. The fragments or segments to be removed may themselves also be referred to as “propeptides.” A proprotein or propeptide thus has had the signal peptide removed, but contains propeptides (here referring to propeptide segments) and the portions that will make up the mature protein.
  • the skilled artisan is able to determine, depending on the species in which the proteins are being expressed and the desired intracellular location, if higher expression levels might be obtained by using a gene construct encoding just the mature form of the protein, the mature form with a signal peptide, or the proprotein (i.e., a form including propeptides) with a signal peptide.
  • the pre- and propeptide sequences may be needed.
  • the propeptide segments may play a role in aiding correct peptide folding.
  • deletions, insertions, and substitutions of the protein sequences encompassed herein are not expected to produce radical changes in the characteristics of the protein. However, when it is difficult to predict the exact effect of the substitution, deletion, or insertion in advance of doing so, one skilled in the art will appreciate that the effect can be evaluated by routine screening assays. That is, the activity can be evaluated by cystatin activity assays. Additionally, differences in the expression of specific genes between uninfected and infected plants can be determined using gene expression profiling.
  • Variant nucleotide sequences and proteins also encompass sequences and proteins derived from a mutagenic and recombinogenic procedure such as DNA shuffling. With such a procedure, one or more different cystatin coding sequences can be manipulated to create a new cystatin protein possessing the desired properties. In this manner, libraries of recombinant polynucleotides are generated from a population of related sequence polynucleotides comprising sequence regions that have substantial sequence identity and can be homologously recombined in vitro or in vivo.
  • sequence motifs encoding a domain of interest may be shuffled between the cystatin genes and partial sequences of the invention and other known cystatin genes to obtain a new gene coding for a protein with an improved property of interest, such as an increased K m in the case of an enzyme.
  • Such shuffling of domains may also be used to assemble novel proteins having novel properties.
  • Strategies for such DNA shuffling are known in the art. See, for example, Stemmer (1994) Proc Natl Acad Sci USA 91: 10747-10751; Stemmer (1994) Nature 370: 389-391; Crameri et al. (1997) Nature Biotech 15: 436-438; Moore et al.
  • nucleotide sequences of the invention can be used to isolate corresponding sequences from other organisms, particularly other plants, more particularly other monocots. In this manner, methods such as PCR, hybridization, and the like can be used to identify such sequences based on their sequence homology to the sequences set forth herein. Sequences isolated based on their sequence identity to the entire cystatin sequences set forth herein or to fragments thereof are encompassed by the present invention. Such sequences include sequences that are orthologs of the disclosed sequences. “Orthologs” means genes derived from a common ancestral gene and which are found in different species as a result of speciation. Genes found in different species are considered orthologs when their nucleotide sequences and/or their encoded protein sequences share substantial identity as defined elsewhere herein. Functions of orthologs are often highly conserved among species.
  • oligonucleotide primers can be designed for use in PCR reactions to amplify corresponding DNA sequences from cDNA or genomic DNA extracted from any plant of interest.
  • Methods for designing PCR primers and PCR cloning are generally known in the art and are disclosed in, for example, Sambrook. See also Innis et al., eds. (1990) PCR Protocols: A Guide to Methods and Applications (Academic Press, New York); Innis and Gelfand, eds. (1995) PCR Strategies (Academic Press, New York); and Innis and Gelfand, eds. (1999) PCR Methods Manual (Academic Press, New York).
  • Known methods of PCR include, but are not limited to, methods using paired primers, nested primers, single specific primers, degenerate primers, gene-specific primers, vector-specific primers, partially-mismatched primers, and the like.
  • hybridization techniques all or part of a known nucleotide sequence is used as a probe that selectively hybridizes to other corresponding nucleotide sequences present in a population of cloned genomic DNA fragments or cDNA fragments (i.e., genomic or cDNA libraries) from a chosen organism.
  • the hybridization probes may be genomic DNA fragments, cDNA fragments, RNA fragments, or other oligonucleotides, and may be labeled with a detectable group such as 32 P, or any other detectable marker.
  • probes for hybridization can be made by labeling synthetic oligonucleotides based on the cystatin sequences of the invention.
  • an entire cystatin sequence disclosed herein, or one or more portions thereof may be used as a probe capable of specifically hybridizing to corresponding cystatin sequences and messenger RNAs.
  • probes include sequences that are unique among cystatin sequences and are preferably at least about 10 nucleotides in length, and most preferably at least about 20 nucleotides in length.
  • Such probes may be used to amplify corresponding sequences from a chosen organism by PCR. This technique may be used to isolate additional coding sequences from a desired organism or as a diagnostic assay to determine the presence of coding sequences in an organism.
  • Hybridization techniques include hybridization screening of plated DNA libraries (either plaques or colonies; see, for example, Sambrook.
  • isolated sequences that encode for a cystatin polypeptide and which hybridize under stringent conditions to the cystatin sequences disclosed herein, or to fragments thereof, are encompassed by the present invention.
  • Biological activity of the cystatin polypeptides can be assayed by any method known in the art.
  • Biological activity of the polypeptides of the present invention can be assayed by any method known in the art.
  • most published cystatin activity assays are based on inhibition of papain-mediated substrate hydrolysis.
  • a variety of synthetic papain substrates are known in the art and can be used for this purpose, such as N-benzoyl-asparaginyl-p-nitroanilide (Schlereth et al.
  • cysteine proteinase that is more relevant to the biological system studied (e.g., a Fusarium cysteine proteinase).
  • Assays to detect cystatin-like activity include, for example, assessing antifungal and/or antimicrobial activity (Soares-Costa et al. (2002) Biochem Biophys Res Comm 296: 1194-1199; Duvick et al. (1992) J Biol Chem 267(26): 18814-18820; Pernas-Monica et al. (1999) Mol Plant Microbe In 12 (7): 624-627; Blankenvoorde-Michiel et al. (1998) Biol Chem 379(11): 1371-1375, all of which are herein incorporated by reference).
  • Assays that measure antipathogenic activity are commonly known in the art, as are methods to quantitate disease resistance in plants following pathogen infection. See, for example, U.S. Pat. No. 5,614,395, herein incorporated by reference. Such techniques include, measuring over time, the average lesion diameter, the pathogen biomass, and the overall percentage of decayed plant tissues. For example, a plant either expressing an antipathogenic polypeptide or having an antipathogenic composition applied to its surface shows a decrease in tissue necrosis (i.e., lesion diameter) or a decrease in plant death following pathogen challenge when compared to a control plant that was not exposed to the antipathogenic composition. Alternatively, antipathogenic activity can be measured by a decrease in pathogen biomass.
  • a plant expressing an antipathogenic polypeptide or exposed to an antipathogenic composition is challenged with a pathogen of interest.
  • tissue samples from the pathogen-inoculated tissues are obtained and RNA is extracted.
  • the percent of a specific pathogen RNA transcript relative to the level of a plant specific transcript allows the level of pathogen biomass to be determined. See, for example, Thomma et al. (1998) Plant Biology 95:15107-15111, herein incorporated by reference.
  • in vitro antipathogenic assays include, for example, the addition of varying concentrations of the antipathogenic composition to paper disks and placing the disks on agar containing a suspension of the pathogen of interest. Following incubation, clear inhibition zones develop around the discs that contain an effective concentration of the antipathogenic polypeptide (Liu et al. (1994) Plant Biology 91:1888-1892, herein incorporated by reference). Additionally, microspectrophotometrical analysis can be used to measure the in vitro antipathogenic properties of a composition (Hu et al. (1997) Plant Mol. Biol. 34:949-959 and Cammue et al. (1992) J. Biol. Chem. 267: 2228-2233, both of which are herein incorporated by reference).
  • compositions and methods for controlling pathogenic agents are provided.
  • the anti-pathogenic compositions comprise maize, soybean, rice and wheat cystatin nucleotide and amino acid sequences. Particularly, the nucleic acid and amino acid sequences and fragments and variants thereof set forth herein. Accordingly, the compositions and methods are also useful in protecting plants against fungal pathogens, viruses, nematodes, insects and the like.
  • Plant pathogen or “plant pest” is intended to mean any microorganism that can cause harm to a plant, such as by inhibiting or slowing the growth of a plant, by damaging the tissues of a plant, by weakening the immune system of a plant or the resistance of a plant to abiotic stresses, and/or by causing the premature death of the plant, etc.
  • Plant pathogens and plant pests include microbes such as fungi, viruses, bacteria, and nematodes.
  • Disease resistance or “pathogen resistance” is intended to mean that the organisms avoid the disease symptoms which are the outcome of organism-pathogen interactions. That is, pathogens are prevented from causing diseases and the associated disease symptoms, or alternatively, the disease symptoms caused by the pathogen is minimized or lessened.
  • the methods of the invention can be utilized to protect plants from disease, particularly those diseases that are caused by plant pathogens.
  • Anti-pathogenic compositions is intended to mean that the compositions of the invention are capable of suppressing, controlling, and/or killing the invading pathogenic organism.
  • An antipathogenic composition of the invention will reduce the disease symptoms resulting from pathogen challenge by at least about 5% to about 50%, at least about 10% to about 60%, at least about 30% to about 70%, at least about 40% to about 80%, or at least about 50% to about 90% or greater.
  • the methods of the invention can be utilized to protect plants from disease, particularly those diseases that are caused by plant pathogens.
  • An “antimicrobial agent,” a “pesticidal agent,” a “cystatin,” and/or a “fungicidal agent” will act similarly to suppress, control, and/or kill the invading pathogen.
  • a defensive agent will possess defensive activity.
  • “Defensive activity” means an antipathogenic, antimicrobial, or antifungal activity.
  • Antipathogenic compositions is intended to mean that the compositions of the invention have activity against pathogens; including fungi, microorganisms, viruses, and nematodes, and thus are capable of suppressing, controlling, and/or killing the invading pathogenic organism.
  • An antipathogenic composition of the invention will reduce the disease symptoms resulting from plant pathogen challenge by at least about 5% to about 50%, at least about 10% to about 60%, at least about 30% to about 70%, at least about 40% to about 80%, or at least about 50% to about 90% or greater.
  • the methods of the invention can be utilized to protect organisms, particularly plants, from disease, particularly those diseases that are caused by invading pathogens.
  • Pathogens of the invention include, but are not limited to, viruses or viroids, bacteria, insects, nematodes, fungi, and the like.
  • Viruses include any plant virus, for example, tobacco or cucumber mosaic virus, ringspot virus, necrosis virus, maize dwarf mosaic virus, etc.
  • Specific fungal and viral pathogens for the major crops include, but are not limited to: Soybeans: Phytophthora megasperma fsp. glycinea, Macrophomina phaseolina, Rhizoctonia solani, Sclerotinia sclerotiorum, Fusarium oxysporum, Diaporthe phaseolorum var. sojae ( Phomopsis sojae ), Diaporthe phaseolorum var.
  • phaseoli Microsphaera diffusa, Fusarium semitectum, Phialophora gregata , Soybean mosaic virus, Glomerella glycines , Tobacco Ring spot virus, Tobacco Streak virus, Phakopsora pachyrhizi, Pythium aphanidermatum, Pythium ultimum, Pythium debaryanum , Tomato spotted wilt virus, Heterodera glycines, Fusarium solani ; Canola: Albugo candida, Alternaria brassicae, Leptosphaeria maculans, Rhizoctonia solani, Sclerotinia sclerotiorum, Mycosphaerella brassiccola, Pythium ultimum, Peronospora parasitica, Fusarium roseum, Alternaria alternata ; Alfalfa: Clavibater michiganese subsp.
  • Rhizoctonia spp. including but not limited to Rhizoctonia solani, Rhizoctonia oryzae and Rhizoctonia oryzae - sativae
  • Pseudomonas spp. including but not limited to Pseudomonas plantarii, Pseudomonas avenae, Pseudomonas glumae, Pseudomonas fuscovaginae, Pseudomonas alboprecipitans, Pseudomonas syringae pv.
  • Erwinia spp. including but not limited to Erwinia herbicola, Erwinia amylovaora, Erwinia chrysanthemi and Erwinia carotovora
  • Achyla spp. including but not limited to Achyla conspicua and Achyla klebsiana
  • Curvularia lunata including but not limited to Curvularia lunata, Curvularia affinis, Curvularia clavata, Curvularia eragrostidis, Curvularia fallax, Curvularia geniculata, Curvularia inaequalis, Curvularia intermedia, Curvularia oryzae, Curvularia oryzae - sativae, Curvularia pallescens, Curvularia senegalensis, Curvularia tuberculata, Curvularia uncinata and Curvularia verruculosa ), Sarocladium oryzae, Gerlachia oryzae, Fusarium spp.
  • Neovossia spp. including but not limited to Neovossia horrida ), Tilletia spp., Balansia oryzae - sativae, Phoma spp.
  • Nigrospora spp. including but not limited to Nigrospora oryzae, Nigrospora sphaerica, Nigrospora panici and Nigrospora padwickii ), Epiococcum nigrum, Phyllostica spp., Wolkia decolorans, Monascus purpureus, Aspergillus spp., Penicillium spp., Absidia spp., Mucor spp., Chaetomium spp., Dematium spp., Monilia spp., Streptomyces spp., Syncephalastrum spp., Verticillium spp., Nematospora coryli, Nakataea sigmoidea, Cladosporium spp.,
  • Nematodes include plant-parasitic nematodes such as root-knot, cyst, and lesion nematodes, including Heterodera and Globodera spp. such as Globodera rostochiensis and Globodera pailida (potato cyst nematodes); Heterodera glycines (soybean cyst nematode); Heterodera schachtii (beet cyst nematode); and Heterodera avenae (cereal cyst nematode).
  • plant-parasitic nematodes such as root-knot, cyst, and lesion nematodes, including Heterodera and Globodera spp. such as Globodera rostochiensis and Globodera pailida (potato cyst nematodes); Heterodera glycines (soybean cyst nematode); Heterodera schachtii (beet cyst
  • Insect pests include insects selected from the orders Coleoptera, Diptera, Hymenoptera, Lepidoptera, Mallophaga, Homoptera, Hemiptera, Orthoptera, Thysanoptera, Dermaptera, Isoptera, Anoplura, Siphonaptera, Trichoptera, etc., particularly Coleoptera and Lepidoptera.
  • Insect pests of the invention for the major crops include, but are not limited to: Maize: Ostrinia nubilalis , European corn borer; Agrotis ipsilon , black cutworm; Helicoverpa zea , corn earworm; Spodoptera frugiperda , fall armyworm; Diatraea grandiosella , southwestern corn borer; Elasmopalpus lignosellus , lesser cornstalk borer; Diatraea saccharalis , surgarcane borer; Diabrotica virgifera , western corn rootworm; Diabrotica longicornis barberi , northern corn rootworm; Diabrotica undecimpunctata howardi , southern corn rootworm; Melanotus spp., wireworms; Cyclocephala borealis , northern masked chafer (white grub); Cyclocephala immaculata , southern masked chafer (white grub); Popillia ja
  • nucleic acid sequences of the present invention can be expressed in a host cell such as bacteria, yeast, insect, mammalian, or preferably plant cells. 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 invention. No attempt to describe in detail the various methods known for the expression of proteins in prokaryotes or eukaryotes will be made.
  • the cystatin sequences of the invention are provided in expression cassettes or DNA constructs for expression in the plant of interest.
  • the cassette will include 5′ and 3′ regulatory sequences operably linked to a cystatin sequence of the invention.
  • the cassette may additionally contain at least one additional gene to be cotransformed into the organism. Alternatively, the additional gene(s) can be provided on multiple expression cassettes.
  • Such an expression cassette is provided with a plurality of restriction sites for insertion of the cystatin sequence to be under the transcriptional regulation of the regulatory regions.
  • the expression cassette may additionally contain selectable marker genes.
  • the expression cassette will include in the 5′-3′ direction of transcription, a transcriptional initiation region (i.e., a promoter), translational initiation region, a polynucleotide of the invention, a translational termination region and, optionally, a transcriptional termination region functional in the host organism.
  • the regulatory regions i.e., promoters, transcriptional regulatory regions, and translational termination regions
  • the polynucleotide of the invention may be native/analogous to the host cell or to each other.
  • the regulatory regions and/or the polynucleotide of the invention may be heterologous to the host cell or to each other.
  • heterologous in reference to a sequence is a sequence 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 polynucleotide is from a species different from the species from which the polynucleotide was derived, or, if from the same/analogous species, one or both are substantially modified from their original form and/or genomic locus, or the promoter is not the native promoter for the operably linked polynucleotide.
  • the native promoter sequences may be used. Such constructs would change expression levels of cystatin in the host cell (i.e., plant or plant cell). Thus, the phenotype of the host cell (i.e., plant or plant cell) is altered.
  • the termination region may be native with the transcriptional initiation region, may be native with the operably linked DNA sequence of interest, or may be derived from another source.
  • Convenient termination regions are available from the Ti-plasmid of A. tumefaciens , such as the octopine synthase and nopaline synthase termination regions. See also Guerineau et al. (1991) Mol. Gen. Genet. 262:141-144; Proudfoot (1991) Cell 64:671-674; Sanfacon et al. (1991) Genes Dev. 5:141-149; Mogen et al. (1990) Plant Cell 2:1261-1272; Munroe et al. (1990) Gene 91:151-158; Ballas et al. (1989) Nucleic Acids Res. 17:7891-7903; and Joshi et al. (1987) Nucleic Acid Res. 15:9627-9639.
  • the gene(s) may be optimized for increased expression in the transformed plant. That is, the genes can be synthesized using plant-preferred codons for improved expression. Methods are available in the art for synthesizing plant-preferred genes. See, for example, U.S. Pat. Nos. 5,380,831, and 5,436,391, and Murray et al. (1989) Nucleic Acids Res. 17:477-498, herein incorporated by reference.
  • Additional sequence modifications are known to enhance gene expression in a cellular host. These include elimination of sequences encoding spurious polyadenylation signals, exon-intron splice site signals, transposon-like repeats, and other such well-characterized sequences that may be deleterious to gene expression.
  • the G-C content of the sequence may be adjusted to levels average for a given cellular host, as calculated by reference to known genes expressed in the host cell. When possible, the sequence is modified to avoid predicted hairpin secondary mRNA structures.
  • the expression cassettes may additionally contain 5′ leader sequences in the expression cassette construct.
  • leader sequences can act to enhance translation.
  • Translation leaders are known in the art and include: picornavirus leaders, for example, EMCV leader (Encephalomyocarditis 5′ noncoding region) (Elroy-Stein et al. (1989) PNAS USA 86:6126-6130); potyvirus leaders, for example, TEV leader (Tobacco Etch Virus) (Allison et al. (1986) Virology 154:9-20); and human immunoglobulin heavy-chain binding protein (BiP), (Macejak et al.
  • the various DNA fragments may be manipulated, so as to provide for the DNA sequences in the proper orientation and, as appropriate, in the proper reading frame.
  • adapters or linkers may be employed to join the DNA fragments or other manipulations may be involved to provide for convenient restriction sites, removal of superfluous DNA, removal of restriction sites, or the like.
  • in vitro mutagenesis, primer repair, restriction, annealing, resubstitutions, e.g., transitions and transversions may be involved.
  • the expression cassette will 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, glyphosate, ammonium, bromoxynil, imidazolinones, and 2,4-dichlorophenoxyacetate (2,4-D). See generally, Yarranton (1992) Curr. Opin. Biotech. 3:506-511; Christopherson et al. (1992) Proc.
  • selectable marker genes are not meant to be limiting. Any selectable marker gene can be used in the present invention.
  • promoters can be used in the practice of the invention.
  • the promoters can be selected based on the desired outcome. That is, the nucleic acids can be combined with constitutive, tissue-preferred, or other promoters for expression in the host cell of interest.
  • constitutive promoters include, for example, the core promoter of the Rsyn7 (WO 99/48338 and U.S. Pat. No. 6,072,050); the core CaMV 35S promoter (Odell et al. (1985) Nature 313:810-812); rice actin (McElroy et al. (1990) Plant Cell 2:163-171); ubiquitin (Christensen et al. (1989) Plant Mol. Biol.
  • an inducible promoter particularly from a pathogen-inducible promoter.
  • promoters include those from pathogenesis-related proteins (PR proteins), which are induced following infection by a pathogen; e.g., PR proteins, SAR proteins, beta-1,3-glucanase, chitinase, etc.
  • PR proteins pathogenesis-related proteins
  • SAR proteins pathogenesis-related proteins
  • beta-1,3-glucanase chitinase, etc.
  • Van Loon (1985) Plant Mol. Virol. 4:111-116 See also, U.S. application Ser. No. 09/257,583 and WO 99/43819, herein incorporated by reference.
  • promoters that are expressed locally at or near the site of pathogen infection. See, for example, Marineau et al. (1987) Plant Mol. Biol. 9:335-342; Matton et al. (1989) Molecular Plant - Microbe Interactions 2:325-331; Somsisch et al. (1986) Proc. Natl. Acad. Sci. USA 83:2427-2430; Somsisch et al. (1988) Mol. Gen. Genet. 2:93-98; and Yang (1996) Proc. Natl. Acad. Sci. USA 93:14972-14977. See also, Chen et al. (1996) Plant J. 10:955-966; Zhang et al. (1994) Proc.
  • a wound-inducible promoter may be used in the constructions of the invention.
  • Such wound-inducible promoters include potato proteinase inhibitor (pin 11) gene (Ryan (1990) Ann. Rev. Phytopath. 28:425-449; Duan et al. (1996) Nature Biotechnology 14:494-498); wun1 and wun2, U.S. Pat. No. 5,428,148; win1 and win2 (Stanford et al. (1989) Mol. Gen. Genet. 215:200-208); systemin (McGurl et al. (1992) Science 225:1570-1573); WIP1 (Rohmeier et al.
  • Chemical-regulated promoters can be used to modulate the expression of a gene in a plant through the application of an exogenous chemical regulator.
  • the promoter may be a chemical-inducible promoter, where application of the chemical induces gene expression, or a chemical-repressible promoter, where application of the chemical represses gene expression.
  • Chemical-inducible promoters are known in the art and include, but are not limited to, the maize ln2-2 promoter, which is activated by benzenesulfonamide herbicide safeners, the maize GST promoter, which is activated by hydrophobic electrophilic compounds that are used as pre-emergent herbicides, and the tobacco PR-1a promoter, which is activated by salicylic acid.
  • promoters of interest include steroid-responsive promoters (see, for example, the glucocorticoid-inducible promoter in Schena et al. (1991) Proc. Natl. Acad. Sci. USA 88:10421-10425 and McNellis et al. (1998) Plant J. 14(2):247-257) and tetracycline-inducible and tetracycline-repressible promoters (see, for example, Gatz et al. (1991) Mol. Gen. Genet. 227:229-237, and U.S. Pat. Nos. 5,814,618 and 5,789,156), herein incorporated by reference.
  • Tissue-preferred promoters can be utilized to target enhanced cystatin expression within a particular plant tissue.
  • Tissue-preferred promoters include those disclosed in Yamamoto et al. (1997) Plant J. 12(2):255-265; Kawamata et al. (1997) Plant Cell Physiol. 38(7):792-803; Hansen et al. (1997) Mol. Gen. Genet. 254(3):337-343; Russell et al. (1997) Transgenic Res. 6(2):157-168; Rinehart et al. (1996) Plant Physiol. 112(3):1331-1341; Van Camp et al. (1996) Plant Physiol. 112(2):525-535; Canevascini et al. (1996) Plant Physiol.
  • Leaf-specific promoters are known in the art. See, for example, Yamamoto et al. (1997) Plant J. 12(2):255-265; Kwon et al. (1994) Plant Physiol. 105:357-67; Yamamoto et al. (1994) Plant Cell Physiol. 35(5):773-778; Gotor et al. (1993) Plant J. 3:509-18; Orozco et al. (1993) Plant Mol. Biol. 23(6):1129-1138; and Matsuoka et al. (1993) Proc. Natl. Acad. Sci. USA 90(20):9586-9590.
  • an antipathogenic polypeptide of the invention may be targeted to specific plant tissues or cell types through the use of appropriate promoters, it may also be targeted to different locations within the cell through the use of targeting information or “targeting labels”. Unlike the promoter, which acts at the transcriptional level, such targeting information is part of the initial translation product. Depending on the mode of infection of the pathogen or the metabolic function of the tissue or cell type, the location of the protein in different compartments of the cell may make it more efficacious against a given pathogen or make it interfere less with the functions of the cell. For example, one may produce a protein preceded by a signal peptide, which directs the translation product into the endoplasmic reticulum, by including in the construct (i.e. expression cassette) sequences encoding a signal peptide (such sequences may also be called the “signal sequence”). The signal sequence used could be, for example, one associated with the gene encoding the polypeptide, or it may be taken from another gene.
  • the default pathway that is, the pathway taken by the polypeptide if no other targeting labels are included, results in secretion of the polypeptide across the cell membrane (Raikhel and Chrispeels, supra) into the apoplast.
  • the apoplast is the region outside the plasma membrane system and includes cell walls, intercellular spaces, and the xylem vessels that form a continuous, permeable system through which water and solutes may move.
  • transformation/transfection is not critical to the instant invention; various methods of transformation or transfection are currently available. As newer methods are available to transform crops or other host cells they may be directly applied. Accordingly, a wide variety of methods have been developed to insert a DNA sequence into the genome of a host cell to obtain the transcription and/or translation of the sequence to effect phenotypic changes in the organism. Thus, any method, which provides for effective transformation/transfection may be employed.
  • Transformation protocols as well as protocols for introducing nucleotide sequences into plants may vary depending on the type of plant or plant cell, i.e., monocot or dicot, targeted for transformation. Suitable methods of introducing nucleotide sequences into plant cells and subsequent insertion into the plant genome include microinjection (Crossway et al. (1986) Biotechniques 4:320-334), electroporation (Riggs et al. (1986) Proc. Natl. Acad. Sci. USA 83:5602-5606), Agrobacterium -mediated transformation (Townsend et al., U.S. Pat. No. 5,563,055 and Zhao et al., U.S. Pat. No.
  • the cells that have been transformed may be grown into plants in accordance with conventional ways. See, for example, McCormick et al. (1986) Plant Cell Reports 5:81-84. These plants may then be grown, and either pollinated with the same transformed strain or different strains, and the resulting hybrid having constitutive expression of the desired phenotypic characteristic identified. Two or more generations may be grown to ensure that constitutive expression of the desired phenotypic characteristic is stably maintained and inherited and then seeds harvested to ensure constitutive expression of the desired phenotypic characteristic.
  • One of skill will recognize that after the recombinant expression cassette is stably incorporated in transgenic plants and confirmed to be operable, it can be introduced into other plants by sexual crossing. Any of number of standard breeding techniques can be used, depending upon the species to be crossed.
  • mature transgenic plants can be propagated by the taking of cuttings or by tissue culture techniques to produce multiple identical plants. Selection of desirable transgenics is made and new varieties are obtained and propagated vegetatively for commercial use.
  • mature transgenic plants can be self-crossed to produce a homozygous inbred plant. The inbred plant produces seed containing the newly introduced heterologous nucleic acid. These seeds can be grown to produce plans that would produce the selected phenotype.
  • Parts obtained from the regenerated plant such as flowers, seeds, leaves, branches, fruit, and the like are included in the invention, provided that these parts comprise cells comprising the isolated nucleic acid of the present invention. Progeny and variants, and mutants of the regenerated plants are also included within the scope of the invention, provided that these parts comprise the introduced nucleic acid sequences.
  • a preferred embodiment is a transgenic plant that is homozygous for the added heterologous nucleic acid; i.e., a transgenic plant that contains two added nucleic acid sequences, one gene at the same locus on each chromosome of a chromosome pair.
  • a homozygous transgenic plant can be obtained by sexually mating (selfing) a heterozygous transgenic plant that contains a single added heterologous nucleic acid, germinating some of the seed produced and analyzing the resulting plants produced for altered expression of a polynucleotide of the present invention relative to a control plant (i.e., native, non-transgenic). Backcrossing to a parental plant and out-crossing with a non-transgenic plant are also contemplated.
  • the present invention may be used for transformation of any plant species, including, but not limited to, monocots and dicots.
  • plants of interest include, but are not limited to, corn ( Zea mays ), Brassica sp. (e.g., B. napus, B. rapa, B.
  • juncea particularly those Brassica species useful as sources of seed oil, alfalfa ( Medicago sativa ), rice ( Oryza sativa ), rye ( Secale cereale ), sorghum ( Sorghum bicolor, Sorghum vulgare ), millet (e.g., pearl millet ( Pennisetum glaucum ), proso millet ( Panicum miliaceum ), foxtail millet ( Setaria italica ), finger millet ( Eleusine coracana )), sunflower ( Helianthus annuus ), safflower ( Carthamus tinctorius ), wheat ( Triticum aestivum ), soybean ( Glycine max ), tobacco ( Nicotiana tabacum ), potato ( Solanum tuberosum ), peanuts ( Arachis hypogaea ), cotton ( Gossypium barbadense, Gossypium hirsutum ), sweet potato ( Ipomoea batat
  • Vegetables include tomatoes ( Lycopersicon esculentum ), lettuce (e.g., Lactuca sativa ), green beans ( Phaseolus vulgaris ), lima beans ( Phaseolus limensis ), peas ( Lathyrus spp.), and members of the genus Cucumis such as cucumber ( C. sativus ), cantaloupe ( C. cantalupensis ), and musk melon ( C. melo ).
  • tomatoes Lycopersicon esculentum
  • lettuce e.g., Lactuca sativa
  • green beans Phaseolus vulgaris
  • lima beans Phaseolus limensis
  • peas Lathyrus spp.
  • members of the genus Cucumis such as cucumber ( C. sativus ), cantaloupe ( C. cantalupensis ), and musk melon ( C. melo ).
  • Ornamentals include azalea ( Rhododendron spp.), hydrangea ( Macrophylla hydrangea ), hibiscus ( Hibiscus rosasanensis ), roses ( Rosa spp.), tulips ( Tulipa spp.), daffodils ( Narcissus spp.), petunias ( Petunia hybrida ), carnation ( Dianthus caryophyllus ), poinsettia ( Euphorbia pulcherrima ), and chrysanthemum.
  • Conifers that may be employed in practicing the present invention include, for example, pines such as loblolly pine ( Pinus taeda ), slash pine ( Pinus elliotii ), ponderosa pine ( Pinus ponderosa ), lodgepole pine ( Pinus contorta ), and Monterey pine ( Pinus radiata ); Douglas-fir ( Pseudotsuga menziesii ); Western hemlock ( Tsuga canadensis ); Sitka spruce ( Picea glauca ); redwood ( Sequoia sempervirens ); true firs such as silver fir ( Abies amabilis ) and balsam fir ( Abies balsamea ); and cedars such as Western red cedar ( Thuja plicata ) and Alaska yellow-cedar ( Chamaecyparis nootkatensis ).
  • pines such as loblolly pine ( Pinus taeda ),
  • plants of the present invention are crop plants (for example, corn, alfalfa, sunflower, Brassica , soybean, cotton, safflower, peanut, sorghum, wheat, millet, tobacco, etc.), more preferably corn and soybean plants, yet more preferably corn plants.
  • crop plants for example, corn, alfalfa, sunflower, Brassica , soybean, cotton, safflower, peanut, sorghum, wheat, millet, tobacco, etc.
  • Prokaryotic cells may be used as hosts for expression. Prokaryotes most frequently are represented by various strains of E. coli , 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 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.
  • promoters for transcription initiation optionally with an operator, along with ribosome binding sequences
  • promoters include such commonly used promoters as the beta lactamase (penicillinase) and lactose (lac) promoter systems (Chang et al. (1977) Nature 198:1056), the trypto
  • E. coli examples include, for example, genes specifying resistance to ampicillin, tetracycline, or chloramphenicol.
  • 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 invention are available using Bacillus sp. and Salmonella (Palva et al. (1983) Gene 22:229-235 and Mosbach et al. (1983) Nature 302:543-545).
  • eukaryotic expression systems such as yeast, insect cell lines, plant and mammalian cells, are known to those of skill in the art.
  • a polynucleotide of the present invention can be expressed in these eukaryotic systems.
  • transformed/transfected plant cells as discussed infra, are employed as expression systems for production of the proteins of the instant invention.
  • antimicrobial proteins can be used for any application including coating surfaces to target microbes. In this manner, target microbes include human pathogens or microorganisms.
  • yeast Synthesis of heterologous nucleotide sequences in yeast is well known.
  • Sherman, F., et al. (1982) Methods in Yeast Genetics , Cold Spring Harbor Laboratory is a well recognized work describing the various methods available to produce a 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 invention once expressed, can be isolated from yeast by lysing the cells and applying standard protein isolation techniques to the lysates.
  • the monitoring of the purification process can be accomplished by using Western blot techniques, radioimmunoassay, or other standard immunoassay techniques.
  • sequences of the present invention can also be ligated to various expression vectors for use in transfecting cell cultures of, for instance, mammalian, insect, or plant origin.
  • Illustrative cell cultures useful for the production of the peptides are mammalian cells.
  • 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.
  • ribosome binding 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.
  • polyadenylation sites e.g., an SV40 large T Ag poly A addition site
  • transcriptional terminator sequences e.g., an SV40 large T Ag poly A addition site
  • Appropriate vectors for expressing proteins of the present invention 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, Schneider, J. Embryol. Exp. Morphol. 27:353-365 (1987)).
  • polyadenylation 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. 45:773-781).
  • 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.
  • Animal and lower eukaryotic (e.g., yeast) host cells are competent or rendered competent for transfection by various means.
  • eukaryotic (e.g., yeast) host cells are competent or rendered competent for transfection by various means.
  • methods of introducing DNA into animal cells include: calcium phosphate precipitation, fusion of the recipient cells with bacterial protoplasts containing the DNA, treatment of the recipient cells with liposomes containing the DNA, DEAE dextrin, electroporation, biolistics, and micro-injection of the DNA directly into the cells.
  • the transfected cells are cultured by means well known in the art. See, Kuchler, R. J. (1997) Biochemical Methods in Cell Culture and Virology , Dowden, Hutchinson and Ross, Inc.
  • antisense constructions complementary to at least a portion of the messenger RNA (mRNA) for the cystatin sequences can be constructed.
  • Antisense nucleotides are constructed to hybridize with the corresponding mRNA. Modifications of the antisense sequences may be made as long as the sequences hybridize to and interfere with expression of the corresponding mRNA. In this manner, antisense constructions having 70%, preferably 80%, more preferably 85% sequence identity to the corresponding antisensed sequences may be used. 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, or greater may be used.
  • the nucleotide sequences of the present invention may also be used in the sense orientation to suppress the expression of endogenous genes in plants.
  • Methods for suppressing gene expression in plants using nucleotide sequences in the sense orientation are known in the art.
  • the methods generally involve transforming plants with a DNA construct comprising a promoter that drives expression in a plant operably linked to at least a portion of a nucleotide sequence that corresponds to the transcript of the endogenous gene.
  • a nucleotide sequence has substantial sequence identity to the sequence of the transcript of the endogenous gene, preferably greater than about 65% sequence identity, more preferably greater than about 85% sequence identity, most preferably greater than about 95% sequence identity. See, U.S. Pat. Nos. 5,283,184 and 5,034,323; herein incorporated by reference.
  • the present invention further provides a method for modulating (i.e., increasing or decreasing) the concentration or composition of the polypeptides of the present invention in a plant or part thereof.
  • Increasing or decreasing the concentration and/or the composition of polypeptides in a plant can affect modulation.
  • increasing the ratio of polypeptides of the invention to native polypeptides can affect modulation.
  • the method comprises: introducing a polynucleotide of the present invention into a plant cell with a recombinant expression cassette as described above to obtain a transformed plant cell, culturing the transformed plant cell under appropriate growing conditions, and inducing or repressing expression of a polynucleotide of the present invention in the plant for a time sufficient to modulate the concentration and/or the composition of polypeptides in the plant or plant part.
  • the content and/or composition of polypeptides of the present invention in a plant may be modulated by altering, in vivo or in vitro, the promoter of the nucleotide sequence to up- or down-regulate expression.
  • the promoter of the nucleotide sequence may be modulated by altering, in vivo or in vitro, the promoter of the nucleotide sequence to up- or down-regulate expression.
  • an isolated nucleic acid comprising a promoter sequence is transfected into a plant cell.
  • a plant cell comprising the promoter operably linked to a polynucleotide of the present invention is selected for by means known to those of skill in the art such as, but not limited to, Southern blot, DNA sequencing, or PCR analysis using primers specific to the promoter and to the gene and detecting amplicons produced therefrom.
  • a plant or plant part altered or modified by the foregoing embodiments is grown under plant forming conditions for a time sufficient to modulate the concentration and/or composition of polypeptides of the present invention in the plant.
  • Plant forming conditions are well known in the art and discussed briefly, supra.
  • concentration or composition is increased or decreased by at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% relative to a native control plant, plant part, or cell lacking the aforementioned recombinant expression cassette.
  • Modulation in the present invention may occur during and/or subsequent to growth of the plant to the desired stage of development.
  • Modulating nucleic acid expression temporally and/or in particular tissues can be controlled by employing the appropriate promoter operably linked to a polynucleotide of the present invention in, for example, sense or antisense orientation as discussed in greater detail, supra.
  • Induction of expression of a polynucleotide of the present invention can also be controlled by exogenous administration of an effective amount of inducing compound. Inducible promoters and inducing compounds, which activate expression from these promoters, are well known in the art.
  • the polypeptides of the present invention are modulated in monocots, particularly maize.
  • the nucleic acid sequences of the present invention can be stacked with any combination of polynucleotide sequences of interest in order to create plants with a desired phenotype.
  • the polynucleotides of the present invention may be stacked with any other polynucleotides of the present invention, such as any combination of the maize, soybean, rice and wheat cystatin sequences presented (SEQ ID NOS: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 43, 45, 47, 49, 51, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, and 75), or with other genes implicated in disease resistance pathways, especially those that have antimicrobial activity like cystatins, such as: (a) other proteinase inhibitors, such as trypsin proteinase inhibitors (Chen et al.
  • subtilisin/chymotrypsin inhibitors Cordero et al. (1994) Plant J 6(2): 141-150
  • trypsin/alpha-amylase inhibitors Wen et al. (1992) Plant Mol. Biol. 18(4): 813-814
  • Bowman-Birk proteinase inhibitors Prakash et al. (1996) J Mol Evol 1996; 42 (5): 560-569
  • small cysteine-rich antimicrobial proteins such as defensins (Thomma et al.
  • the combinations generated can also include multiple copies of any one of the polynucleotides of interest.
  • the polynucleotides of the present invention can also be stacked with any other 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., U.S. Pat. No. 6,232,529); balanced amino acids (e.g. hordothionins (U.S. Pat. Nos. 5,990,389; 5,885,801; 5,885,802; and 5,703,409)); barley high lysine (Williamson et al. (1987) Eur. J. Biochem.
  • the polynucleotides of the present invention can also be stacked with traits desirable for insect, disease or herbicide resistance (e.g., Bacillus thuringiensis toxic proteins (U.S. Pat. Nos. 5,366,892; 5,747,450; 5,737,514; 5,723,756; 5,593,881; Geiser et al (1986) Gene 48:109); lectins (Van Damme et al. (1994) Plant Mol. Biol. 24:825); fumonisin detoxification genes (U.S. Pat. No. 5,792,931); avirulence and disease resistance genes (Jones et al. (1994) Science 266:789; Martin et al.
  • traits desirable for insect, disease or herbicide resistance e.g., Bacillus thuringiensis toxic proteins (U.S. Pat. Nos. 5,366,892; 5,747,450; 5,737,514; 5,723,756; 5,593,881; Geiser et
  • 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 GAT gene)
  • traits desirable for processing or process products such as high oil (U.S. Pat. No. 6,232,529); modified oils (e.g., fatty acid desaturase genes (U.S. Pat. No.
  • modified starches e.g., ADPG pyrophosphorylases (AGPase), starch synthases (SS), starch branching enzymes (SBE) and starch debranching enzymes (SDBE)
  • AGPase ADPG pyrophosphorylases
  • SS starch synthases
  • SBE starch branching enzymes
  • SDBE starch debranching enzymes
  • polymers or bioplastics U.S. Pat. No. 5,602,321; beta-ketothiolase, polyhydroxybutyrate synthase, and acetoacetyl-CoA reductase (Schubert et al. (1988) J. Bacteriol. 170:5837-5847), which facilitate expression of polyhydroxyalkanoates (PHAs)), the disclosures of which are herein incorporated by reference.
  • polynucleotides of the present invention could also combine with polynucleotides providing agronomic traits such as male sterility (see U.S. Pat. No. 5,583,210), stalk strength, flowering time, or transformation technology traits such as cell cycle regulation or gene targeting (see, WO 99/61619; WO 00/17364; WO 99/25821), the disclosures of which are herein incorporated by reference.
  • agronomic traits such as male sterility (see U.S. Pat. No. 5,583,210), stalk strength, flowering time, or transformation technology traits such as cell cycle regulation or gene targeting (see, WO 99/61619; WO 00/17364; WO 99/25821), the disclosures of which are herein incorporated by reference.
  • stacked combinations can be created by any method including, but not limited to, polynucleotide sequences of interest can be combined at any time and in any order.
  • a transgenic plant comprising one or more desired traits can be used as the target to introduce further traits by subsequent transformation.
  • the traits can be introduced simultaneously in a co-transformation protocol with the polynucleotides of interest provided by any combination of transformation cassettes.
  • the two sequences can be contained in separate transformation cassettes (trans) or contained on the same transformation cassette (cis). Expression of the sequences can be driven by the same promoter or by different promoters.
  • the present invention provides a method of genotyping a plant comprising a polynucleotide of the present invention.
  • the plant is a monocot, such as maize or sorghum.
  • Genotyping provides a means of distinguishing homologs of a chromosome pair and can be used to differentiate segregants in a plant population.
  • Molecular marker methods can be used for phylogenetic studies, characterizing genetic relationships among crop varieties, identifying crosses or somatic hybrids, localizing chromosomal segments affecting monogenic traits, map based cloning, and the study of quantitative inheritance. See, e.g., Plant Molecular Biology: A Laboratory Manual , Chapter 7, Clark, Ed., Springer-Verlag, Berlin (1997).
  • RFLPs restriction fragment length polymorphisms
  • RFLPs are the product of allelic differences between DNA restriction fragments resulting from nucleotide sequence variability.
  • RFLPs are typically detected by extraction of genomic DNA and digestion with a restriction enzyme. Generally, the resulting fragments are separated according to size and hybridized with a probe; single copy probes are preferred. Restriction fragments from homologous chromosomes are revealed. Differences in fragment size among alleles represent an RFLP.
  • the present invention further provides a means to follow segregation of a gene or nucleic acid of the present invention as well as chromosomal sequences genetically linked to these genes or nucleic acids using such techniques as RFLP analysis.
  • Linked chromosomal sequences are within 50 centiMorgans (cM), often within 40 or 30 cM, preferably within 20 or 10 cM, more preferably within 5, 3, 2, or 1 cM of a gene of the present invention.
  • the nucleic acid probes employed for molecular marker mapping of plant nuclear genomes hybridize, under selective hybridization conditions, to a gene encoding a polynucleotide of the present invention.
  • the probes are selected from polynucleotides of the present invention.
  • these probes are cDNA probes or restriction enzyme treated (e.g., PST I) genomic clones.
  • the length of the probes is typically at least 15 bases in length, more preferably at least 20, 25, 30, 35, 40, or 50 bases in length. Generally, however, the probes are less than about 1 kilobase in length.
  • the probes are single copy probes that hybridize to a unique locus in a haploid chromosome compliment.
  • Some exemplary restriction enzymes employed in RFLP mapping are EcoRI, EcoRV, and SstI.
  • restriction enzyme includes reference to a composition that recognizes and, alone or in conjunction with another composition, cleaves at a specific nucleotide sequence.
  • the method of detecting an RFLP comprises the steps of (a) digesting genomic DNA of a plant with a restriction enzyme; (b) hybridizing a nucleic acid probe, under selective hybridization conditions, to a sequence of a polynucleotide of the present invention of the genomic DNA; (c) detecting therefrom a RFLP.
  • polymorphic (allelic) variants of polynucleotides of the present invention can be had by utilizing molecular marker techniques well known to those of skill in the art including such techniques as: 1) single stranded conformation analysis (SSCA); 2) denaturing gradient gel electrophoresis (DGGE); 3) RNase protection assays; 4) allele-specific oligonucleotides (ASOs); 5) the use of proteins which recognize nucleotide mismatches, such as the E. coli mutS protein; and 6) allele-specific PCR.
  • molecular marker techniques well known to those of skill in the art including such techniques as: 1) single stranded conformation analysis (SSCA); 2) denaturing gradient gel electrophoresis (DGGE); 3) RNase protection assays; 4) allele-specific oligonucleotides (ASOs); 5) the use of proteins which recognize nucleotide mismatches, such as the E. coli mutS protein; and 6) allele
  • the present invention further provides a method of genotyping comprising the steps of contacting, under stringent hybridization conditions, a sample suspected of comprising a polynucleotide of the present invention with a nucleic acid probe.
  • a sample suspected of comprising a polynucleotide of the present invention with a nucleic acid probe.
  • the sample is a plant sample, preferably, a sample suspected of comprising a maize polynucleotide of the present invention (e.g., gene, mRNA).
  • the nucleic acid probe selectively hybridizes, under stringent conditions, to a subsequence of a polynucleotide of the present invention comprising a polymorphic marker. Selective hybridization of the nucleic acid probe to the polymorphic marker nucleic acid sequence yields a hybridization complex. Detection of the hybridization complex indicates the presence of that polymorphic marker in the sample.
  • the nucleic acid probe comprises a polynucleotide of the present invention.
  • nucleotide constructs are not intended to limit the present invention to nucleotide constructs comprising DNA.
  • nucleotide constructs particularly polynucleotides and oligonucleotides, comprised of ribonucleotides and combinations of ribonucleotides and deoxyribonucleotides may also be employed in the methods disclosed herein.
  • the nucleotide constructs of the present invention encompass all nucleotide constructs that can be employed in the methods of the present invention for transforming plants including, but not limited to, those comprised of deoxyribonucleotides, ribonucleotides, and combinations thereof.
  • nucleotide constructs of the invention also encompass all forms of nucleotide constructs including, but not limited to, single-stranded forms, double-stranded forms, hairpins, stem-and-loop structures, and the like.
  • the methods of the invention may employ a nucleotide construct that is capable of directing, in a transformed plant, the expression of at least one protein, or at least one RNA, such as, for example, an antisense RNA that is complementary to at least a portion of an mRNA.
  • a nucleotide construct is comprised of a coding sequence for a protein or an RNA operably linked to 5′ and 3′ transcriptional regulatory regions.
  • the methods of the invention may employ a nucleotide construct that is not capable of directing, in a transformed plant, the expression of a protein or an RNA.
  • methods of the present invention do not depend on the incorporation of the entire nucleotide construct into the genome, only that the plant or cell thereof is altered as a result of the introduction of the nucleotide construct into a cell.
  • the genome may be altered following the introduction of the nucleotide construct into a cell.
  • the nucleotide construct, or any part thereof may incorporate into the genome of the plant.
  • Alterations to the genome of the present invention include, but are not limited to, additions, deletions, and substitutions of nucleotides in the genome. While the methods of the present invention do not depend on additions, deletions, or substitutions of any particular number of nucleotides, it is recognized that such additions, deletions, or substitutions comprise at least one nucleotide.
  • nucleotide constructs of the invention also encompass nucleotide constructs that may be employed in methods for altering or mutating a genomic nucleotide sequence in an organism, including, but not limited to, chimeric vectors, chimeric mutational vectors, chimeric repair vectors, mixed-duplex oligonucleotides, self-complementary chimeric oligonucleotides, and recombinogenic oligonucleobases.
  • nucleotide constructs and methods of use such as, for example, chimeraplasty, are known in the art.
  • Chimeraplasty involves the use of such nucleotide constructs to introduce site-specific changes into the sequence of genomic DNA within an organism. See, U.S. Pat.
  • the methods of the invention can be used with other methods available in the art for enhancing disease resistance in plants.
  • the antimicrobial compositions described herein may be used alone or in combination with other nucleotide sequences, polypeptides, or agents to protect against plant diseases and pathogens.
  • any one of a variety of second nucleotide sequences may be utilized, specific embodiments of the invention encompass those second nucleotide sequences that, when expressed in a plant, help to increase the resistance of a plant to pathogens.
  • Proteins, peptides, and lysozymes that naturally occur in insects (Jaynes et al. (1987) Bioassays 6:263-270), plants (Broekaert et al. (1997) Critical Reviews in Plant Sciences 16:297-323), animals (Vunnam et al. (1997) J. Peptide Res. 49:59-66), and humans (Mitra and Zang (1994) Plant Physiol. 106:977-981; Nakajima et al. (1997) Plant Cell Reports 16:674-679) are also a potential source of plant disease resistance (Ko, K. (2000) www.scisoc.org/feature/BioTechnology/antimicrobial.html).
  • plant resistance-conferring sequences examples include those encoding sunflower rhoGTPase-Activating Protein (rhoGAP), lipoxygenase (LOX), Alcohol Dehydrogenase (ADH), and Sclerotinia -Inducible Protein-1 (SCIP-1) described in U.S. application Ser. No. 09/714,767, herein incorporated by reference. These nucleotide sequences enhance plant disease resistance through the modulation of development, developmental pathways, and the plant pathogen defense system. Other plant defense proteins include those described in WO 99/43823 and WO 99/43821, all of which are herein incorporated by reference. It is recognized that such second nucleotide sequences may be used in either the sense or antisense orientation depending on the desired outcome.
  • the cystatins comprise isolated polypeptides of the invention.
  • the cystatins of the invention find use in the decontamination of plant pathogens during the processing of grain for animal or human food consumption; during the processing of feedstuffs, and during the processing of plant material for silage.
  • the cystatins of the invention are presented to grain, plant material for silage, or a contaminated food crop, or during an appropriate stage of the processing procedure, in amounts effective for anti-microbial activity.
  • compositions can be applied to the environment of a plant pathogen by, for example, spraying, atomizing, dusting, scattering, coating or pouring, introducing into or on the soil, introducing into irrigation water, by seed treatment, or dusting at the time when the plant pathogen has begun to appear or before the appearance of pests as a protective measure. It is recognized that any means to bring the defensive agent polypeptides in contact with the plant pathogen can be used in the practice of the invention.
  • compositions can be used in formulations used for their antimicrobial activities.
  • Methods are provided for controlling plant pathogens comprising applying a decontaminating amount of a polypeptide or composition of the invention to the environment of the plant pathogen.
  • the polypeptides of the invention can be formulated with an acceptable carrier into a composition(s) that is, for example, a suspension, a solution, an emulsion, a dusting powder, a dispersible granule, a wettable powder, an emulsifiable concentrate, an aerosol, an impregnated granule, an adjuvant, a coatable paste, and also encapsulations in, for example, polymer substances.
  • compositions disclosed above may be obtained by the addition of a surface-active agent, an inert carrier, a preservative, a humectant, a feeding stimulant, an attractant, an encapsulating agent, a binder, an emulsifier, a dye, a UV protectant, a buffer, a flow agent or fertilizers, micronutrient donors or other preparations that influence plant growth.
  • One or more agrochemicals including, but not limited to, herbicides, insecticides, fungicides, bacteriocides, nematocides, molluscicides, acaracides, plant growth regulators, harvest aids, and fertilizers, can be combined with carriers, surfactants, or adjuvants customarily employed in the art of formulation or other components to facilitate product handling and application for particular target pathogens.
  • Suitable carriers and adjuvants can be solid or liquid and correspond to the substances ordinarily employed in formulation technology, e.g., natural or regenerated mineral substances, solvents, dispersants, wetting agents, tackifiers, binders, or fertilizers.
  • the active ingredients of the present invention are normally applied in the form of compositions and can be applied to the crop area or plant to be treated, simultaneously or in succession, with other compounds.
  • methods of applying an active ingredient of the present invention or an agrochemical composition of the present invention are foliar application, seed coating, and soil application.
  • Suitable surface-active agents include, but are not limited to, anionic compounds such as a carboxylate of, for example, a metal; a carboxylate of a long chain fatty acid; an N-acylsarcosinate; mono or di-esters of phosphoric acid with fatty alcohol ethoxylates or salts of such esters; fatty alcohol sulfates such as sodium dodecyl sulfate, sodium octadecyl sulfate, or sodium cetyl sulfate; ethoxylated fatty alcohol sulfates; ethoxylated alkylphenol sulfates; lignin sulfonates; petroleum sulfonates; alkyl aryl sulfonates such as alkyl-benzene sulfonates or lower alkylnaphtalene sulfonates, e.g., butyl-naphthalene sulfonate;
  • Non-ionic agents include condensation products of fatty acid esters, fatty alcohols, fatty acid amides or fatty-alkyl- or alkenyl-substituted phenols with ethylene oxide, fatty esters of polyhydric alcohol ethers, e.g., sorbitan fatty acid esters, condensation products of such esters with ethylene oxide, e.g. polyoxyethylene sorbitar fatty acid esters, block copolymers of ethylene oxide and propylene oxide, acetylenic glycols such as 2,4,7,9-tetraethyl-5-decyn-4,7-diol, or ethoxylated acetylenic glycols.
  • a cationic surface-active agent examples include, for instance, an aliphatic mono-, di-, or polyamine such as an acetate, naphthenate, or oleate; or oxygen-containing amine such as an amine oxide of polyoxyethylene alkylamine; an amide-linked amine prepared by the condensation of a carboxylic acid with a di- or polyamine; or a quaternary ammonium salt.
  • inert materials include, but are not limited to, inorganic minerals such as kaolin, phyllosilicates, carbonates, sulfates, phosphates, or botanical materials such as cork, powdered corncobs, peanut hulls, rice hulls, and walnut shells.
  • inorganic minerals such as kaolin, phyllosilicates, carbonates, sulfates, phosphates, or botanical materials such as cork, powdered corncobs, peanut hulls, rice hulls, and walnut shells.
  • compositions of the present invention can be in a suitable form for direct application or as concentrate of primary composition, which requires dilution with a suitable quantity of water or other diluent before application.
  • the decontaminating concentration will vary depending upon the nature of the particular formulation, specifically, whether it is a concentrate or to be used directly.
  • compositions, as well as the polypeptides of the present invention can be treated prior to formulation to prolong their activity when applied to the environment of a plant pathogen as long as the pretreatment is not deleterious to the activity.
  • Such treatment can be by chemical and/or physical means as long as the treatment does not deleteriously affect the properties of the composition(s).
  • chemical reagents include, but are not limited to, halogenating agents; aldehydes such as formaldehyde and glutaraldehyde; anti-infectives, such as zephiran chloride; alcohols, such as isopropanol and ethanol; and histological fixatives, such as Bouin's fixative and Helly's fixative (see, for example, Humason (1967) Animal Tissue Techniques (W.H. Freeman and Co.)).
  • aldehydes such as formaldehyde and glutaraldehyde
  • anti-infectives such as zephiran chloride
  • alcohols such as isopropanol and ethanol
  • histological fixatives such as Bouin's fixative and Helly's fixative (see, for example, Humason (1967) Animal Tissue Techniques (W.H. Freeman and Co.)).
  • the compositions of the invention comprise a microbe having stably integrated the nucleotide sequence of a defensive agent.
  • the resulting microbes can be processed and used as a microbial spray. Any suitable microorganism can be used for this purpose. See, for example, Gaertner et al. (1993) in Advanced Engineered Pesticides , Kim (Ed.).
  • the nucleotide sequences of the invention are introduced into microorganisms that multiply on plants (epiphytes) to deliver the cystatins to potential target crops.
  • Epiphytes can be, for example, gram-positive or gram-negative bacteria.
  • whole, i.e., unlysed, cells of the transformed microorganism can be treated with reagents that prolong the activity of the polypeptide produced in the microorganism when the microorganism is applied to the environment of a target plant.
  • a secretion signal sequence may be used in combination with the gene of interest such that the resulting enzyme is secreted outside the microorganism for presentation to the target plant.
  • a gene encoding a defensive agent of the invention may be introduced via a suitable vector into a microbial host, and said transformed host applied to the environment, plants, or animals.
  • Microorganism hosts that are known to occupy the “phytosphere” (phylloplane, phyllosphere, rhizosphere, and/or rhizoplane) of one or more crops of interest may be selected for transformation. These microorganisms are selected so as to be capable of successfully competing in the particular environment with the wild-type microorganisms, to provide for stable maintenance and expression of the gene expressing the detoxifying polypeptide, and for improved protection of the enzymes of the invention from environmental degradation and inactivation.
  • microorganisms include bacteria, algae, and fungi.
  • microorganisms such as bacteria, e.g., Pseudomonas, Erwinia, Serratia, Klebsiella, Xanthomonas, Streptomyces, Rhizobium, Rhodopseudomonas, Methylius, Agrobacterium, Acetobacter, Lactobacillus, Arthrobacter, Azotobacter, Leuconostoc , and Alcaligenes ; fungi, particularly yeast, e.g., Saccharomyces, Pichia, Cryptococcus, Kluyveromyces, Sporobolomyces, Rhodotorula, Aureobasidium , and Gliocladium .
  • phytosphere bacterial species as Pseudomonas syringae, Pseudomonas fluorescens, Serratia marcescens, Acetobacter xylinum, Agrobacteria, Rhodopseudomonas spheroides, Xanthomonas campestris, Rhizobium melioti, Alcaligenes entrophus, Clavibacter xyli , and Azotobacter vinlandii ; and phytosphere yeast species such as Rhodotorula rubra, R. glutinis, R. marina, R. aurantiaca, Cryptococcus albidus, C. diffluens, C.
  • Illustrative prokaryotes both Gram-negative and -positive, include Enterobacteriaceae, such as Escherichia, Erwinia, Shigella, Salmonella , and Proteus; Bacillaceae; Rhizobiaceae , such as Rhizobium; Spirillaceae , such as photobacterium, Zymomonas, Serratia, Aeromonas, Vibrio, Desulfovibrio, Spirillum; Lactobacillaceae; Pseudomonadaceae , such as Pseudomonas and Acetobacter; Azotobacteraceae ; and Nitrobacteraceae .
  • Enterobacteriaceae such as Escherichia, Erwinia, Shigella, Salmonella , and Proteus
  • Bacillaceae Rhizobiaceae , such as Rhizobium
  • Spirillaceae such as photobacterium, Zymomonas, Serratia, Aeromona
  • fungi such as Phycomycetes and Ascomycetes , which includes yeast, such as Saccharomyces and Schizosaccharomyces ; and Basidiomycetes yeast, such as Rhodotorula, Aureobasidium, Sporobolomyces , and the like.
  • the cystatins of the invention may be used as a pharmaceutical compound for treatment of fungal and microbial pathogens in humans and other animals.
  • Diseases and disorders caused by fungal and microbial pathogens include but are not limited to fungal meningoencephalitis, superficial fungal infections, ringworm, Athlete's foot, histoplasmosis, candidiasis, thrush, coccidioidoma, pulmonary cryptococcus, trichosporonosis, piedra, tinea nigra, fungal keratitis, onychomycosis, tinea capitis, chromomycosis, aspergillosis, endobronchial pulmonary aspergillosis, mucormycosis, chromoblastomycosis, dermatophytosis, tinea, fusariosis, pityriasis, mycetoma, pseudallescheriasis, and sporo
  • compositions of the invention may be used as pharmaceutical compounds to provide treatment for diseases and disorders associated with, but not limited to, the following fungal pathogens: Histoplasma capsulatum, Candida spp. ( C. albicans, C. tropicalis, C. parapsilosis, C. guilliermondii, C. glabratal Torulopsis glabrata, C. krusei, C. lusitaniae ), Aspergillus fumigatus, A. flavus, A.
  • compositions of the invention as anti-fungal treatments may be determined through anti-fungal assays known to one of skill in the art.
  • the cystatins may be administered to a patient through numerous means.
  • Systemic administration can also be by transmucosal or transdermal means.
  • penetrants appropriate to the barrier to be permeated are used in the formulation.
  • penetrants are generally known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives.
  • Transmucosal administration can be accomplished through the use of nasal sprays or suppositories.
  • the active compounds are formulated into ointments, salves, gels, or creams as generally known in the art.
  • the compounds can also be prepared in the form of suppositories (e.g., with conventional suppository bases such as cocoa butter and other glycerides) or retention enemas for rectal delivery.
  • the active compounds are prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems.
  • a controlled release formulation including implants and microencapsulated delivery systems.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art.
  • the materials can also be obtained commercially from Alza Corporation and Nova Pharmaceuticals, Inc.
  • Liposomal suspensions (including liposomes targeted to infected cells with monoclonal antibodies to viral antigens) can also be used as pharmaceutically acceptable carriers. These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Pat. No. 4,522,811.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the subject to be treated with each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • about 1 ⁇ g/kg to about 15 mg/kg (e.g., 0.1 to 20 mg/kg) of antibody is an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion.
  • a typical daily dosage might range from about 1 ⁇ g/kg to about 100 mg/kg or more, depending on the factors mentioned above.
  • the treatment is sustained until a desired suppression of disease symptoms occurs.
  • other dosage regimens may be useful.
  • the progress of this therapy is easily monitored by conventional techniques and assays.
  • An exemplary dosing regimen is disclosed in WO 94/04188.
  • the specification for the dosage unit forms of the invention are dictated by and directly dependent on the unique characteristics of the active compound and the particular therapeutic effect to be achieved, and the limitations inherent in the art of compounding such an active compound for the treatment of individuals.
  • Treatment is herein defined as the application or administration of a therapeutic agent to a patient, or application or administration of a therapeutic agent to an isolated tissue or cell line from a patient, who has a disease, a symptom of disease or a predisposition toward a disease, with the purpose to cure, heal, alleviate, relieve, alter, remedy, ameliorate, improve or affect the disease, the symptoms of disease or the predisposition toward disease.
  • a “therapeutic agent” includes, but is not limited to, small molecules, peptides, antibodies, ribozymes and antisense oligonucleotides.
  • the cystatins of the invention can be used for any application including coating surfaces to target microbes.
  • target microbes include human pathogens or microorganisms.
  • Surfaces that might be coated with the cystatins of the invention include carpets and sterile medical facilities.
  • Polymer bound polypeptides of the invention may be used to coat surfaces. Methods for incorporating compositions with anti-microbial properties into polymers are known in the art. See U.S. Pat. No. 5,847,047 herein incorporated by reference.
  • An isolated polypeptide of the invention can be used as an immunogen to generate antibodies that bind cystatins using standard techniques for polyclonal and monoclonal antibody preparation.
  • the full-length cystatins can be used or, alternatively, the invention provides antigenic peptide fragments of cystatins for use as immunogens.
  • the antigenic peptide of a defensive agent comprises at least 8, preferably 10, 15, 20, or 30 amino acid residues of the amino acid sequence shown in any of SEQ ID NOs: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, and 76, and encompasses an epitope of a cystatin such that an antibody raised against the peptide forms a specific immune complex with the anti-microbial polypeptides.
  • Preferred epitopes encompassed by the antigenic peptide are regions of cystatins that are located on the surface of the protein, e.g., hydrophilic regions.
  • Another aspect of the invention pertains to anti-cystatin polyclonal and monoclonal antibodies that bind a cystatin.
  • Polyclonal cystatin-like antibodies can be prepared by immunizing a suitable subject (e.g., rabbit, goat, mouse, or other mammal) with an defensive agent-like immunogen.
  • the anti-cystatin antibody titer in the immunized subject can be monitored over time by standard techniques, such as with an enzyme linked immunosorbent assay (ELISA) using immobilized anti-microbial polypeptides.
  • ELISA enzyme linked immunosorbent assay
  • antibody-producing cells can be obtained from the subject and used to prepare monoclonal antibodies by standard techniques, such as the hybridoma technique originally described by Kohler and Milstein (1975) Nature 256:495-497, the human B cell hybridoma technique (Kozbor et al. (1983) Immunol. Today 4:72), the EBV-hybridoma technique (Cole et al. (1985) in Monoclonal Antibodies and Cancer Therapy , ed. Reisfeld and Sell (Alan R. Liss, Inc., New York, N.Y.), pp. 77-96) or trioma techniques.
  • standard techniques such as the hybridoma technique originally described by Kohler and Milstein (1975) Nature 256:495-497, the human B cell hybridoma technique (Kozbor et al. (1983) Immunol. Today 4:72), the EBV-hybridoma technique (Cole et al. (1985) in Monoclonal Antibodies and Cancer Therapy ,
  • hybridomas The technology for producing hybridomas is well known (see generally Coligan et al., eds. (1994) Current Protocols in Immunology (John Wiley & Sons, Inc., New York, N.Y.); Galfre et al. (1977) Nature 266:55052; Kenneth (1980) in Monoclonal Antibodies: A New Dimension In Biological Analyses (Plenum Publishing Corp., NY; and Lerner (1981) Yale J. Biol. Med., 54:387-402).
  • a monoclonal anti-cystatin-like antibody can be identified and isolated by screening a recombinant combinatorial immunoglobulin library (e.g., an antibody phage display library) with a cystatin to thereby isolate immunoglobulin library members that bind the defensive agent.
  • Kits for generating and screening phage display libraries are commercially available (e.g., the Pharmacia Recombinant Phage Antibody System , Catalog No. 27-9400-01; and the Stratagene SurfZAPTM Phage Display Kit , Catalog No. 240612).
  • examples of methods and reagents particularly amenable for use in generating and screening an antibody display library can be found in, for example, U.S. Pat. No. 5,223,409; PCT Publication Nos. WO 92/18619; WO 91/17271; WO 92/20791; WO 92/15679; 93/01288; WO 92/01047; 92/09690; and 90/02809; Fuchs et al. (1991) Bio/Technology 9:1370-1372; Hay et al. (1992) Hum. Antibod. Hybridomas 3:81-85; Huse et al. (1989) Science 246:1275-1281; Griffiths et al. (1993) EMBO J. 12:725-734.
  • the antibodies can be used to identify homologs of the cystatins of the invention.
  • Immature maize embryos from greenhouse donor plants are bombarded with a plasmid containing the cystatin sequences of the present invention operably linked to a ubiquitin promoter and the selectable marker gene PAT (Wohlleben et al. (1988) Gene 70:25-37), which confers resistance to the herbicide Bialaphos.
  • PAT Wihlleben et al. (1988) Gene 70:25-37
  • the selectable marker gene is provided on a separate plasmid. Transformation is performed as follows. Media recipes follow below.
  • the ears are husked and surface sterilized in 30% Clorox bleach plus 0.5% Micro detergent for 20 minutes, and rinsed two times with sterile water.
  • the immature embryos are excised and placed embryo axis side down (scutellum side up), 25 embryos per plate, on 560Y medium for 4 hours and then aligned within the 2.5 cm target zone in preparation for bombardment.
  • This plasmid DNA plus plasmid DNA containing a PAT selectable marker is precipitated onto 1.1 ⁇ m (average diameter) tungsten pellets using a CaCl 2 precipitation procedure as follows:
  • Each reagent is added sequentially to the tungsten particle suspension, while maintained on the multitube vortexer.
  • the final mixture is sonicated briefly and allowed to incubate under constant vortexing for 10 minutes.
  • the tubes are centrifuged briefly, liquid removed, washed with 500 ⁇ L 100% ethanol, and centrifuged for 30 seconds. Again the liquid is removed, and 105 ⁇ l 100% ethanol is added to the final tungsten particle pellet.
  • the tungsten/DNA particles are briefly sonicated and 10 ⁇ L spotted onto the center of each macrocarrier and allowed to dry about 2 minutes before bombardment.
  • sample plates are bombarded at level #4 in particle gun #HE34-1 or #HE34-2. All samples receive a single shot at 650 PSI, with a total of ten aliquots taken from each tube of prepared particles/DNA.
  • the embryos are kept on 560Y medium for 2 days, then transferred to 560R selection medium containing 3 mg/L Bialaphos, and subcultured every 2 weeks. After approximately 10 weeks of selection, selection-resistant callus clones are transferred to 288J medium to initiate plant regeneration. Following somatic embryo maturation (2-4 weeks), well-developed somatic embryos are transferred to medium for germination and transferred to the lighted culture room. Approximately 7-10 days later, developing plantlets are transferred to 272V hormone-free medium in tubes for 7-10 days until plantlets are well established.
  • Plants are then transferred to inserts in flats (equivalent to 2.5′′ pot) containing potting soil and grown for 1 week in a growth chamber, subsequently grown an additional 1-2 weeks in the greenhouse, then transferred to classic 600 pots (1.6 gallon) and grown to maturity. Plants are monitored and scored for and altered level of expression of the cystatin sequence of the invention. Alternatively, the cysteine proteinase activity can be assayed.
  • Bombardment medium comprises 4.0 g/L N6 basal salts (SIGMA C-1416), 1.0 mL/L Eriksson's Vitamin Mix (1000X SIGMA-1511), 0.5 mg/L thiamine HCl, 120.0 g/L sucrose, 1.0 mg/L 2,4-D, and 2.88 g/L L-proline (brought to volume with D-1 H 2 0 following adjustment to pH 5.8 with KOH); 2.0 g/L Gelrite (added after bringing to volume with D-I H 2 0); and 8.5 mg/L silver nitrate (added after sterilizing the medium and cooling to room temperature).
  • Selection medium comprises 4.0 g/L N6 basal salts (SIGMA C-1416), 1.0 mL/L Eriksson's Vitamin Mix (1000X SIGMA-1511), 0.5 mg/L thiamine HCl, 30.0 g/L sucrose, and 2.0 mg/L 2,4-D (brought to volume with D-I H 2 0 following adjustment to pH 5.8 with KOH); 3.0 g/L Gelrite (added after bringing to volume with D-I H 2 0); and 0.85 mg/L silver nitrate and 3.0 mg/L bialaphos (both added after sterilizing the medium and cooling to room temperature).
  • Plant regeneration medium (288J) comprises 4.3 g/L MS salts (GIBCO 11117-074), 5.0 mL/L MS vitamins stock solution (0.100 g nicotinic acid, 0.02 g/L thiamine HCL, 0.10 g/L pyridoxine HCL, and 0.40 g/L glycine brought to volume with polished D-I H 2 0) (Murashige and Skoog (1962) Physiol. Plant.
  • Hormone-free medium comprises 4.3 g/L MS salts (GIBCO 11117-074), 5.0 mL/L MS vitamins stock solution (0.100 g/L nicotinic acid, 0.02 g/L thiamine HCL, 0.10 g/L pyridoxine HCL, and 0.40 g/L glycine brought to volume with polished D-I H 2 0), 0.1 g/L myo-inositol, and 40.0 g/L sucrose (brought to volume with polished D-I H 2 0 after adjusting pH to 5.6); and 6 g/L bacto-agar (added after bringing to volume with polished D-I H 2 0), sterilized and cooled to 60° C.
  • a cystatin nucleotide sequence of the invention operably linked to a ubiquitin promoter
  • the method of Zhao is employed (U.S. Pat. No. 5,981,840, and PCT patent publication WO98/32326; the contents of which are hereby incorporated by reference).
  • immature embryos are isolated from maize and the embryos contacted with a suspension of Agrobacterium , where the bacteria are capable of transferring the DNA construct containing the cystatin nucleotide sequence to at least one cell of at least one of the immature embryos (step 1: the infection step).
  • the immature embryos are preferably immersed in an Agrobacterium suspension for the initiation of inoculation.
  • the embryos are co-cultured for a time with the Agrobacterium (step 2: the co-cultivation step).
  • the immature embryos are cultured on solid medium following the infection step.
  • an optional “resting” step is contemplated.
  • the embryos are incubated in the presence of at least one antibiotic known to inhibit the growth of Agrobacterium without the addition of a selective agent for plant transformants (step 3: resting step).
  • the immature embryos are cultured on solid medium with antibiotic, but without a selecting agent, for elimination of Agrobacterium and for a resting phase for the infected cells.
  • inoculated embryos are cultured on medium containing a selective agent and growing transformed callus is recovered (step 4: the selection step).
  • the immature embryos are cultured on solid medium with a selective agent resulting in the selective growth of transformed cells.
  • the callus is then regenerated into plants (step 5: the regeneration step), and preferably calli grown on selective medium are cultured on solid medium to regenerate the plants.
  • BLAST Basic Local Alignment Search Tool
  • Altschul, S. F., et al. (1993) J. Mol. Biol. 215:403-410 searches under default parameters for similarity to sequences contained in the BLAST “nr” database (comprising all non-redundant GenBank CDS translations, sequences derived from the 3-dimensional structure Brookhaven Protein Data Bank, the last major release of the SWISS-PROT protein sequence database, EMBL, and DDBJ databases).
  • the cDNA sequences of the present invention were analyzed for similarity to all publicly available DNA sequences contained in the “nr” database using the BLASTN algorithm.
  • the DNA sequences were translated in all reading frames and compared for similarity to all publicly available protein sequences contained in the “nr” database using the BLASTX algorithm (Gish, W. and States, D. J. Nature Genetics 3:266-272 (1993)) provided by the NCBI.
  • the sequencing data from two or more clones containing overlapping segments of DNA were used to construct contiguous DNA sequences.
  • Soybean embryos are bombarded with a plasmid containing the cystatin sequence operably linked to a ubiquitin promoter as follows.
  • somatic embryos cotyledons, 3-5 mm in length dissected from surface-sterilized, immature seeds of the soybean cultivar A2872, are 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 multiplied as early, globular-staged embryos, the suspensions are maintained as described below.
  • Soybean embryogenic suspension cultures are maintained in 35 mL liquid media on a rotary shaker, 150 rpm, at 26° C. with florescent lights on a 16:8 hour day/night schedule. Cultures are subcultured 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, U.S. Pat. No. 4,945,050).
  • a DuPont Biolistic 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 comprising the cystatin sequence operably linked to the ubiquitin promoter 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 60 ⁇ 15 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 1100 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.
  • Sunflower meristem tissues are transformed with an expression cassette containing the cystatin sequence operably linked to a ubiquitin promoter as follows (see also European Patent Number EP 0 486233, herein incorporated by reference, and Malone-Schoneberg et al. (1994) Plant Science 103:199-207).
  • Mature sunflower seed Helianthus annuus L.
  • Seeds are surface sterilized for 30 minutes in a 20% Clorox bleach solution with the addition of two drops of Tween 20 per 50 mL of solution. The seeds are rinsed twice with sterile distilled water.
  • Split embryonic axis explants are prepared by a modification of procedures described by Schrammeijer et al. (Schrammeijer et al. (1990) Plant Cell Rep. 9: 55-60). Seeds are imbibed in distilled water for 60 minutes following the surface sterilization procedure. The cotyledons of each seed are then broken off, producing a clean fracture at the plane of the embryonic axis. Following excision of the root tip, the explants are bisected longitudinally between the primordial leaves. The two halves are placed, cut surface up, on GBA medium consisting of Murashige and Skoog mineral elements (Murashige et al. (1962) Physiol.
  • the explants are subjected to microprojectile bombardment prior to Agrobacterium treatment (Bidney et al. (1992) Plant Mol. Biol. 18: 301-313). Thirty to forty explants are placed in a circle at the center of a 60 ⁇ 20 mm plate for this treatment. Approximately 4.7 mg of 1.8 mm tungsten microprojectiles are resuspended in 25 mL of sterile TE buffer (10 mM Tris HCl, 1 mM EDTA, pH 8.0) and 1.5 mL aliquots are used per bombardment. Each plate is bombarded twice through a 150 mm nytex screen placed 2 cm above the samples in a PDS 1000® particle acceleration device.
  • Disarmed Agrobacterium tumefaciens strain EHA105 is used in all transformation experiments.
  • a binary plasmid vector comprising the expression cassette described above is introduced into Agrobacterium strain EHA105 via freeze-thawing as described by Holsters et al. (1978) Mol. Gen. Genet. 163:181-187.
  • This plasmid further comprises a kanamycin selectable marker gene (i.e, nptII).
  • Bacteria for plant transformation experiments are grown overnight (28° C.
  • liquid YEP medium (10 g/L yeast extract, 10 g/L Bactopeptone, and 5 g/L NaCl, pH 7.0) with the appropriate antibiotics required for bacterial strain and binary plasmid maintenance.
  • the suspension is used when it reaches an OD 600 of about 0.4 to 0.8.
  • the Agrobacterium cells are pelleted and resuspended at a final OD 600 of 0.5 in an inoculation medium comprised of 12.5 mM MES pH 5.7, 1 g/L NH 4 Cl, and 0.3 g/L MgSO 4 .
  • Freshly bombarded explants are placed in an Agrobacterium suspension, mixed, and left undisturbed for 30 minutes. The explants are then transferred to GBA medium and co-cultivated, cut surface down, at 26° C. and 18-hour days. After three days of co-cultivation, the explants are transferred to 374B (GBA medium lacking growth regulators and a reduced sucrose level of 1%) supplemented with 250 mg/L cefotaxime and 50 mg/L kanamycin sulfate. The explants are cultured for two to five weeks on selection and then transferred to fresh 374B medium lacking kanamycin for one to two weeks of continued development.
  • Explants with differentiating, antibiotic-resistant areas of growth that have not produced shoots suitable for excision are transferred to GBA medium containing 250 mg/L cefotaxime for a second 3-day phytohormone treatment.
  • Leaf samples from green, kanamycin-resistant shoots are assayed for the presence of NPTII by ELISA and for the presence of transgene expression by assaying for the activity of the cystatin sequences.
  • NPTII-positive shoots are grafted to Pioneer® hybrid 6440 in vitro-grown sunflower seedling rootstock.
  • Surface sterilized seeds are germinated in 48-0 medium (half-strength Murashige and Skoog salts, 0.5% sucrose, 0.3% gelrite, pH 5.6) and grown under conditions described for explant culture. The upper portion of the seedling is removed, a 1 cm vertical slice is made in the hypocotyl, and the transformed shoot inserted into the cut. The entire area is wrapped with parafilm to secure the shoot.
  • Grafted plants can be transferred to soil following one week of in vitro culture. Grafts in soil are maintained under high humidity conditions followed by a slow acclimatization to the greenhouse environment.
  • Transformed sectors of T 0 plants (parental generation) maturing in the greenhouse are identified by NPTII ELISA and/or by the analysis of the activity of the cystatin sequences in the leaf extracts while transgenic seeds harvested from NPTII-positive T 0 plants are identified by the analysis of the activity the cystatin sequences in small portions of dry seed cotyledon.
  • An alternative sunflower transformation protocol allows the recovery of transgenic progeny without the use of chemical selection pressure. Seeds are dehulled and surface-sterilized for 20 minutes in a 20% Clorox bleach solution with the addition of two to three drops of Tween 20 per 100 mL of solution, then rinsed three times with distilled water. Sterilized seeds are imbibed in the dark at 26° C. for 20 hours on filter paper moistened with water.
  • the cotyledons and root radical are removed, and the meristem explants are cultured on 374E (GBA medium consisting of MS salts, Shepard vitamins, 40 mg/L adenine sulfate, 3% sucrose, 0.5 mg/L 6-BAP, 0.25 mg/L IAA, 0.1 mg/L GA, and 0.8% Phytagar at pH 5.6) for 24 hours under the dark.
  • the primary leaves are removed to expose the apical meristem, around 40 explants are placed with the apical dome facing upward in a 2 cm circle in the center of 374M (GBA medium with 1.2% Phytagar), and then cultured on the medium for 24 hours in the dark.
  • the plasmid of interest is introduced into Agrobacterium tumefaciens strain EHA105 via freeze thawing as described previously.
  • the pellet of overnight-grown bacteria at 28° C. in a liquid YEP medium (10 g/L yeast extract, 10 g/L Bactopeptone, and 5 g/L NaCl, pH 7.0) in the presence of 50 ⁇ g/L kanamycin is resuspended in an inoculation medium (12.5 mM 2-mM 2-(N-morpholino) ethanesulfonic acid, MES, 1 g/L NH 4 Cl and 0.3 g/L MgSO 4 at pH 5.7) to reach a final concentration of 4.0 at OD 600.
  • Particle-bombarded explants are transferred to GBA medium (374E), and a droplet of bacteria suspension is placed directly onto the top of the meristem.
  • the explants are co-cultivated on the medium for 4 days, after which the explants are transferred to 374C medium (GBA with 1% sucrose and no BAP, IAA, GA3 and supplemented with 250 ⁇ g/mL cefotaxime).
  • the plantlets are cultured on the medium for about two weeks under 16-hour day and 26° C. incubation conditions.
  • Explants (around 2 cm long) from two weeks of culture in 374C medium are screened for cystatin activity using assays known in the art. After positive (i.e., for cystatin expression) explants are identified, those shoots that fail to exhibit cystatin activity are discarded, and every positive explant is subdivided into nodal explants.
  • One nodal explant contains at least one potential node.
  • the nodal segments are cultured on GBA medium for three to four days to promote the formation of auxiliary buds from each node. Then they are transferred to 374C medium and allowed to develop for an additional four weeks. Developing buds are separated and cultured for an additional four weeks on 374C medium. Pooled leaf samples from each newly recovered shoot are screened again by the appropriate protein activity assay. At this time, the positive shoots recovered from a single node will generally have been enriched in the transgenic sector detected in the initial assay prior to nodal culture.
  • Recovered shoots positive for defense-inducible expression are grafted to Pioneer hybrid 6440 in vitro-grown sunflower seedling rootstock.
  • the rootstocks are prepared in the following manner. Seeds are dehulled and surface-sterilized for 20 minutes in a 20% Clorox bleach solution with the addition of two to three drops of Tween 20 per 100 mL of solution, and are rinsed three times with distilled water. The sterilized seeds are germinated on the filter moistened with water for three days, then they are transferred into 48 medium (half-strength MS salt, 0.5% sucrose, 0.3% gelrite pH 5.0) and grown at 26° C. under the dark for three days, then incubated at 16-hour-day culture conditions.
  • the upper portion of selected seedling is removed, a vertical slice is made in each hypocotyl, and a transformed shoot is inserted into a V-cut.
  • the cut area is wrapped with parafilm. After one week of culture on the medium, grafted plants are transferred to soil. In the first two weeks, they are maintained under high humidity conditions to acclimatize to a greenhouse environment.
  • mini-prep DNA was incubated on ice with 20 ⁇ L of BL21 Star cells (Invitrogen) for 30 minutes. The DNA/cell mixtures were then heated at 42° C. for 45 sec, then iced for 2 min. 200 ⁇ L of SOC were added to each tube, and these mixtures were then incubated at 37° C. for 1 hour. 50 ⁇ L samples were spread out on LB broth plates containing 100 mg/L carbenicillin and 0.1 M MgSO 4 to incubate overnight at 37° C.
  • the OD readings for the wells were obtained after diluting 10 ⁇ L bacterial samples with 90 ⁇ L of water in a standard 96-well, flat bottomed microtiter plate, with the absorbance read at 600 nm. Based on these values, the cultures were diluted with fresh 2xYT (+carb.) to the two target induction OD 600 values of 0.1 and 0.6, to a final volume of 2 mL. Each target OD value was diluted in quadruplicate to account for two isopropyl-beta-D-thiogalactopyranoside (IPTG) induction values each and two incubation temperatures each.
  • IPTG isopropyl-beta-D-thiogalactopyranoside
  • the wells were induced with IPTG at concentrations of either 0.05 mM or 1.0 mM.
  • the plates were then incubated overnight on shakers (225 rpm) at temperatures of either 16° C. or 30° C. After approximately 20 hours of incubation, the OD values for all the wells were obtained as described supra.
  • the cells were then harvested by centrifuging the plates at 1700 rpm for 10 minutes. The supernatants were discarded and the pelleted cells frozen at ⁇ 20° C. until purification could take place.
  • the cells were allowed to thaw for several minutes, then 200 ⁇ L of B-Peril protein extraction reagent (Pierce) containing 0.2 units of benzonase (Novagen) were added to each well, and the solution was pipetted repeatedly to resuspend the pellet. The cells were incubated for 10 min in the B-Peril solution.
  • 25-30 ⁇ m MBPP 800 ⁇ L purification filter plates (Whatman) were prepared by adding 200 ⁇ L of 50% slurry of glutathione Sepharose 4B resin (Amersham) for GST-tagged proteins, or TALON resin (Clontech) for HIS-tagged proteins into each well. The plates were centrifuged briefly to remove excess buffer. The lysates were transferred directly from the growth plate to the plates with the resins, and the solutions were pipetted up and down several times to mix them well. The plates were then centrifuged at 760 rpm for 5 min, and the flow-through was discarded.
  • the resins with the bound proteins were washed twice with 200 ⁇ L of buffer A (50 mM Tris, pH 8.0, 200 mM NaCl, 10% glycerol), and each time the plates were centrifuged at 760 rpm for 5 min and the wash was discarded. Standard 96 well plates were placed underneath the Whatman filter plates, and the proteins were eluted by adding and mixing 100 ⁇ L of buffer A containing either 20 mM reduced glutathione (Sigma) for GST tags, or 500 mM imidazole (Sigma) for HIS tags. The plates were centrifuged at 1000 rpm for 5 min, and the purified proteins were collected in the standard 96 well plates placed underneath.
  • buffer A 50 mM Tris, pH 8.0, 200 mM NaCl, 10% glycerol
  • the anti-fungal and anti-bacterial activity of the cystatin polypeptides of the invention can be tested using a variety of assays.
  • BL21 cells were transformed with cystatin genes using the Lambda CE6 Induction Kit (Stratagene) according to the manufacturer's protocol.
  • the induced BL21 cells were grown and harvested, and then resuspended in the binding buffer from a His-Bind Kit (Novagen).
  • the suspended cells were sonicated to release the expressed protein from the cells.
  • the crude protein extract was denatured by dissolving it into a 6 M urea solution for 1 hour on ice in order to allow the His-tag to efficiently bind to the resin.
  • the denatured cystatins were then purified by His-Bind Resin Chromatography according to the manufacturer's protocol, except that 6M Urea was added to the binding, washing and elution buffers.
  • the purified proteins were renatured by dialysis using 1 ⁇ PBS buffer with gradually decreased concentration of urea (5M, 4M, 3M, 1.5M, and 0.75M).
  • the dialyses were performed with each concentration of urea for one hour and then with 1 ⁇ PBS buffer without urea overnight at 4° C.
  • Bradford reactions were performed to determine the protein concentrations for all the samples. The isolated polypeptide was then tested for activity.
  • Fusarium verticillioides (strain M033) was grown on 1 ⁇ 2 ⁇ potato dextrose agar plates: (For each liter, 12 grams Difco Potato Dextrose Broth (#0549-17-9) and 15 grams agar were suspended in dH 2 O, final volume was raised to 1 liter and autoclaved at 121° C. for 20 minutes. Plates were poured in sterile hood.) Spores were collected from 10 day old to 3 week old culture plates of Fusarium verticillioides by rinsing a portion of the plate with potato dextrose broth (24 grams Difco Potato Dextrose Broth (#0549-17-9) per liter+0.08% tween-20).
  • the collected spore solution was then vortexed and placed on ice.
  • the spores were counted with a hemocytometer and used within 2 hours.
  • 4 ⁇ assay medium with spores was prepared by diluting the collected spores with potato dextrose broth+0.08% tween-20 to 16,000 spores per milliliter.
  • the two purified cystatins used in this assay were supplied in PBS buffer, which was prepared by diluting a 10 ⁇ commercial stock buffer (10 ⁇ phosphate buffered saline [137 mM NaCl, 2.7 mM KCl, 10 mM phosphate pH 7.3-7.5] EM Science Catalog #6505) with water.
  • the protein concentration of each cystatin was measured using the Pierce BCA protein assay (BCA Protein Assay Reagent, Pierce catalog #23225; Smith, P. K., et al. (1985). Measurement of protein using bicinchoninic acid ( Anal. Biochem. 150, 76-85.) calibrated against BSA.
  • the purified GmCys-2 (SEQ ID NO: 30) protein was supplied at 5.7 mg/mL and the ZmCys-4 (SEQ ID NO: 6) protein was supplied at 3.2 mg/mL.
  • Antifungal activity was rated as follows:
  • Assays for bactericidal activity are performed by incubating 10 5 bacterial colony forming units in 90 ⁇ L with 10 mL of peptide (or water for control). After 60 min at 37° C. ( E. coli ) or 25° C. ( C. nebraskense ), four serial, 10-fold dilutions are made in sterile phosphate buffer. Aliquots of 100 ⁇ L are plated on LB or NBY plates, using 1 or 2 plates/dilution. Resulting colonies are counted, and the effect of the peptide is expressed as percent of initial colony count (Selsted et al. (1984) Infect. Immun. 45:150-154).
  • Assays for bacteriostatic activity are performed by incubating 10 5 bacteria with MBP-1 in 200 ⁇ L of dilute medium (1 part NBY broth to 4 parts 10 mM sodium phosphate, pH 5.8) in microtiter plate wells. Plates are covered, sealed, and incubated at 28° C. Growth is monitored spectrophotometrically at 600 nm. After 41 h controls will have grown sufficiently (optical density >0.20) to measure effect of peptide as percent of control.
  • the measurement of inhibition activity of cystatins toward papain was based on the method by Kouzuma et al. (1996) (J. Biochem. 119: 1106-1113) (see FIG. 1 ).
  • the assay was performed in a 96 well plate. Twenty ⁇ L of a cystatin solution containing either 6 ⁇ g or 0.06 ⁇ g of cystatin protein were incubated with 20 ⁇ L of papain solution (0.1 mg/mL papain (Calbiochem) stock in 100 mM phosphate buffer, pH 6.5, 0.3 M KCl, 0.1 mM EDTA, and 1 mM freshly added DTT) at 37° C. for 15 min.
  • papain solution 0.1 mg/mL papain (Calbiochem) stock in 100 mM phosphate buffer, pH 6.5, 0.3 M KCl, 0.1 mM EDTA, and 1 mM freshly added DTT
  • Cysteine proteinase inhibitory activity was indicated by smaller absorbance values for the 6 ⁇ g protein wells versus the 0.06 ⁇ g wells for each cystatin. Negative controls consisted of buffer without cystatin protein. Positive controls consisted of previously tested cystatins. TABLE 14 Impact of N-terminal tag and induction conditions on cystatin activity. Cystatin activity Gene SEQ ID NO: GST-tagged His-tagged Conditions for highest activity GmCys2 29 low/medium Low GST: 16 C.
  • Table 14 above, is a summary of the cystatin activity detected in the tested genes of the present invention. Detailed data for the assay results on each of the above cystatins is presented in tables 15 through 60.
  • cystatins did not show any proteinase inhibitor activity in the conditions of the assay as conducted above. There can be numerous reasons for these results. As is well known in the art, certain microorganisms perform better than others for expression of a given protein. Often it is difficult to predict which one will work best.
  • the cystatins of the present invention have been expressed in BL21 Star cells (Invitrogen). Other bacterial lines or expression systems may be more effective for a selection of the genes. Furthermore, the cystatin proteins were expressed with a N-terminal His- or GST-tag to facilitate cystatin purification from the bacterial culture.
  • cystatin genes of the present invention Two of the cystatin genes of the present invention, ZmCys4 (SEQ ID NO: 5) and GmCys2 (SEQ ID NO: 29), were expressed in, and their encoded proteins purified from E. coli as set forth in Example 7.
  • a control expression vector was also prepared which contained no cystatin gene. The purified cystatins and control were injected into sugar beet nematode-induced syncytia in Arabidopsis roots one week after inoculation, as per the methods outlined in Bockenhoff and Grundler ((1994), Parasitology. 109: 249-255), hereby incorporated by reference.
  • C. elegans populations are cultured on NGM agar carrying a lawn of E. coli OP50 cells as described by Wood ((1988) The nematode C. elegans , Cold Spring Harbor, N.Y. Cold Spring Harbor Laboratory Press). After populations are maintained for five days, agar plugs are removed to fresh plates. Cystatins are added to the medium to a final concentration of 2.5 mg/L just prior to pouring. In order to study the effect of the cystatins on egg laying, hermaphrodite nematodes are taken from their normal growth media and transferred individually to fresh plates containing the cystatin(s) to be used for testing. Egg laying is carefully monitored.
  • groups of larvae hatched on normal media can be transferred to plates containing the cystatin(s) to be tested at time points corresponding to larval stages L1, L2, L3 and L4.
  • the larvae for each stage should be removed, respectively, 6, 12, 24, and 30 hours post hatching. Development of the various larval stages on the supplemented media is monitored.
  • GR..LEFGRV VAAQRQ VVSG os-cys5 NKAVQS LGRF AVAEHN RRLR HGGSGGPADP VPVKLAFARV VEAQKQ VVSD ZmCys3 DLHLQE LARF AVDEHN ...K KA........ .NALLGFEKL VKAKTQ VVAG ZmCys4 DLHLQE LARF AVDEHN ...K KA........ .NALLGFEKL VKAKTQ VVAG ta-cys13 DLHLQE LARF AVDEHN ...K KA........

Abstract

Methods and compositions for modulating development and defense responses are provided. Nucleotide sequences encoding maize, soybean, wheat and rice cystatin proteins are provided. The sequences can be used in expression cassettes for modulating development, developmental pathways, and defense responses. Transformed plants, plant cells, tissues, and seed are also provided.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application claims priority to and the benefit of U.S. Provisional Application No. 60/505,948, filed Sep. 25, 2003, and U.S. Utility application Ser. No. 10/947,979, filed Sep. 23, 2004, which are hereby incorporated in their entirety by reference.
  • FIELD OF THE INVENTION
  • The invention relates to the field of the genetic manipulation of plants, particularly the modulation of gene activity and development in plants resulting in improvements in agronomic traits.
  • BACKGROUND OF THE INVENTION
  • Agronomic traits, such as disease resistance, nutritional quality, senescence and cell proliferation, have been subject to improvement attempts by various methods in the past. Often, improvements are attempted through plant breeding methods, which are often very expensive and of uncertain success. More recently, genetic modifications, such as those creating transgenic plants, have been used in attempts to reach these trait improvement goals. These approaches are meeting with varied success. No one strategy or gene has proven to be a panacea, although some show promise. Successful broad improvement of crop disease resistance, among other traits, will require multiple strategies. The addition of novel genes and methods is especially of value in the area of disease resistance, where pathogens are continually evolving and no single-gene method will have sustained success for long. Thus multiple genes and strategies for genetic improvement of agronomic traits are sought. This invention provides novel genes and methods of use through which agronomic traits can be improved.
  • SUMMARY OF THE INVENTION
  • Compositions and methods relating to disease resistance and other plant agronomic traits are provided. Particularly, the nucleotide and amino acid sequences for cystatin homologs from maize, soybean, wheat and rice are provided. The nucleotide sequences of the invention encode proteinase inhibitors of the cystatin cysteine proteinase inhibitor class.
  • The cystatin genes of the present invention may find use in enhancing agronomic traits of plants, including a wide variety of crop plants. The compositions and methods of the invention can be used to manipulate the plant pathogen defense system, the control of senescence, the control of cell proliferation and cell death, and the nutritional quality of plant seeds intended for human and animal consumption. The methods involve stably transforming a plant with a nucleotide sequence capable of modulating the production of one or more cystatins in the plant, operably linked with a promoter capable of driving expression of a gene in a plant cell.
  • Specifically, the present invention is directed to an isolated polynucleotide set forth in SEQ ID NOs: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 43, 45, 47, 49, 51, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, and 75, isolated polynucleotides encoding the amino acid sequences set forth in SEQ ID NOs: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, and 76, isolated polypeptides having the sequences set forth in SEQ ID NOs: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, and 76, variant polynucleotide and amino acid sequences, DNA constructs comprising the sequences of the present invention, and host cells having incorporated such DNA constructs. Transformed plants, plant cells, and seeds, as well as methods for making such plants, plant cells, and seeds are additionally provided. It is recognized that a variety of promoters will be useful in the invention, the choice of which will depend in part upon the desired level of expression of the disclosed genes. It is recognized that the levels of expression can be controlled to modulate the levels of expression in the plant cell.
  • Further embodiments of the invention include methods of enhancing disease resistance of plants; methods of modulating the timing of plant maturation; methods of reducing cell death in plant tissue culture preparations; and methods of modulating protein digestibility and energy availability in plant products; wherein the preceding methods use plants transformed with the nucleotide sequences of the present invention.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 shows the chemical reaction inhibited by the cystatins of the present invention.
  • DETAILED DESCRIPTION OF THE INVENTION
  • Units, prefixes, and symbols are denoted in their SI accepted form. Unless otherwise indicated, nucleic acids are written left to right in 5′ to 3′ orientation and amino acid sequences are written left to right in amino to carboxy orientation, respectively. Numeric ranges recited within the specification are inclusive of the numbers defining the range and include each integer within the defined 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 IUPAC-IUB Biochemical Nomenclature Commission. Nucleotides, likewise, may be referred to by their commonly accepted single-letter codes. Unless otherwise provided for, software, electrical, and electronics terms as used herein are as defined in The New IEEE Standard Dictionary of Electrical and Electronics Terms (5th edition, 1993). The terms below are more fully defined by reference to the specification as a whole.
  • “Amplified” means 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, D. H. Persing et al., Ed., American Society for Microbiology, Washington, D.C. (1993). The product of amplification is termed an amplicon.
  • As used herein, “antisense orientation” includes reference to a duplex polynucleotide sequence that is operably linked to a promoter in an orientation where the antisense strand is transcribed. The antisense strand is sufficiently complementary to an endogenous transcription product such that translation of the endogenous transcription product is often inhibited.
  • “Encoding” or “encoded”, with respect to a specified nucleic acid, means 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 are present in some plant, animal, and fungal mitochondria, the bacterium Mycoplasma capricolum, or the ciliate Macronucleus, may be used when the nucleic acid is expressed therein.
  • 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 invention may be expressed in both monocotyledonous and dicotyledonous plant species, sequences can be modified to account for the specific codon preferences and GC content preferences of monocotyledons or dicotyledons as these preferences have been shown to differ (Murray et al. Nucl. Acids Res. 17: 477-498 (1989)). Thus, the maize preferred codon for a particular amino acid may be derived from known gene sequences from maize. Maize codon usage for 28 genes from maize plants is listed in Table 4 of Murray et al., supra.
  • As used herein “full-length sequence” in reference to a specified polynucleotide or its encoded protein means having the entire amino acid sequence of, a native (non-synthetic), endogenous, biologically active form of the specified protein. Methods to determine whether a sequence is full-length are well known in the art including such exemplary techniques as northern or western blots, primer extension, S1 protection, and ribonuclease protection. See, e.g., Plant Molecular Biology: A Laboratory Manual, Clark, Ed., Springer-Verlag, Berlin (1997). Comparison to known full-length homologous (orthologous and/or paralogous) sequences can also be used to identify full-length sequences of the present invention. Additionally, consensus sequences typically present at the 5′ and 3′ untranslated regions of mRNA aid in the identification of a polynucleotide as full-length. For example, the consensus sequence ANNNNAUGG, where the underlined codon represents the N-terminal methionine, aids in determining whether the polynucleotide has a complete 5′ end. Consensus sequences at the 3′ end, such as polyadenylation sequences, aid in determining whether the polynucleotide has a complete 3′ end.
  • 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 nucleotide sequence can be from a species different from that from which the nucleotide sequence was derived, or, if from the same species, the promoter is not naturally found operably linked to the nucleotide sequence. 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” means a cell which contains a vector and supports the replication and/or expression of the vector. Host cells may be prokaryotic cells such as E. coli, or eukaryotic cells such as yeast, insect, amphibian, or mammalian cells, excluding human cells. Preferably, host cells are monocotyledonous or dicotyledonous plant cells. A particularly preferred monocotyledonous host cell is a maize host cell.
  • 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 term “isolated” refers to material, such as a nucleic acid or a protein, which is: (1) substantially or essentially free from components that normally accompany or interact with it as it is found in its naturally occurring environment. The isolated material optionally comprises material not found with it in its natural environment; or (2) if the material is in its natural environment, the material has been synthetically (non-naturally) altered by deliberate human intervention to a composition and/or placed at a location in the cell (e.g., genome or subcellular organelle) not native to a material found in that environment. The alteration to yield the synthetic material can be performed on the material within or removed from its natural state. For example, a naturally occurring nucleic acid becomes an isolated nucleic acid if it is altered, or if it is transcribed from DNA which has been altered, by means of human intervention performed within the cell from which it originates. See, e.g., Compounds and Methods for Site Directed Mutagenesis in Eukaryotic Cells, Kmiec, U.S. Pat. No. 5,565,350; In Vivo Homologous Sequence Targeting in Eukaryotic Cells; Zarling et al., PCT/US93/03868. Likewise, a naturally occurring nucleic acid (e.g., a promoter) becomes isolated if it is introduced by non-naturally occurring means to a locus of the genome not native to that nucleic acid. Nucleic acids which are “isolated” as defined herein, are also referred to as “heterologous” nucleic acids.
  • As used herein, “nucleic acid” and “polynucleotide” are used interchangeably and include reference to a deoxyribonucleotide or ribonucleotide polymer, or chimeras thereof, 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. A polynucleotide can be full-length or a subsequence of a native or heterologous structural or regulatory gene. Unless otherwise indicated, the term includes reference to the specified sequence as well as the complementary sequence thereof. Thus, DNAs or RNAs with backbones modified for stability or for other reasons are “polynucleotides” as that term is intended herein. Moreover, DNAs or RNAs comprising unusual bases, such as inosine, or modified bases, such as tritylated bases, to name just two examples, are polynucleotides as the term is used herein. It will be appreciated that a great variety of modifications have been made to DNA and RNA that serve many useful purposes known to those of skill in the art. The term polynucleotide as it is employed herein embraces such chemically, enzymatically or metabolically modified forms of polynucleotides, as well as the chemical forms of DNA and RNA characteristic of viruses and cells, including among other things, simple and complex cells.
  • “Nucleic acid library” means a collection of isolated DNA or RNA molecules which comprise and substantially represent the entire transcribed fraction of a genome of a specified organism or of a tissue from that 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, Guide to Molecular Cloning Techniques, Methods in Enzymology, Vol. 152, Academic Press, Inc., San Diego, Calif. (Berger); Sambrook et al., Molecular Cloning—A Laboratory Manual, 2nd ed., Vol. 1-3 (1989) (hereinafter Sambrook); and Current Protocols in Molecular Biology, F. M. Ausubel et al., Eds., Current Protocols, a joint venture between Greene Publishing Associates, Inc. and John Wiley & Sons, Inc. (1994) (hereinafter Ausubel).
  • As used herein, “operably linked” includes reference to a functional linkage between a promoter and a second sequence, wherein the promoter sequence initiates and mediates transcription of the DNA sequence 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, seeds, suspension cultures, embryos, meristematic regions, callus tissue, leaves, roots, shoots, gametophytes, sporophytes, pollen, and microspores. The classes of plants which can be used in the methods of the invention include both monocotyledonous and dicotyledonous plants. A particularly preferred plant is Zea mays.
  • 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. The essential nature of such analogues of naturally occurring amino acids is that, when incorporated into a protein, that protein is specifically reactive to antibodies elicited to the same protein but consisting entirely of naturally occurring amino acids. The terms “polypeptide”, “peptide” and “protein” are also inclusive of modifications including, but not limited to, glycosylation, lipid attachment, sulfation, gamma-carboxylation of glutamic acid residues, hydroxylation and ADP-ribosylation. Further, this invention contemplates the use of both the methionine-containing and the methionine-less amino terminal variants of the protein of the invention.
  • 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 whether or not its origin is from a plant cell. 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 of promoters under developmental control include promoters that preferentially initiate transcription in certain tissues, such as leaves, roots, or seeds. Such promoters are referred to as “tissue preferred”. Promoters which initiate transcription only in certain tissues are referred to as “tissue specific”. 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 “repressible” 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. Tissue specific, tissue preferred, cell type specific, and inducible promoters constitute the class of “non-constitutive” promoters. A “constitutive” promoter is a promoter which is active under most environmental conditions.
  • 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. 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 host 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 terms “residue,” “amino acid residue,” and “amino acid” are used interchangeably herein to refer to an amino acid that is incorporated into a protein, polypeptide, or peptide (collectively “protein”). The amino acid may be a naturally occurring amino acid and, unless otherwise limited, may encompass non-natural analogs of natural amino acids that can function in a similar manner as naturally occurring amino acids.
  • 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 80% sequence identity, 90% sequence identity, 95% or 100% sequence identity (i.e., complementary) with each other.
  • The term “stringent conditions” or “stringent hybridization conditions” includes reference to conditions under which a probe will selectively hybridize to its target sequence, to a detectably greater degree than to 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 are 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). Generally, a probe is less than about 1000 nucleotides in length, and optionally less than 500 nucleotides in length.
  • 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 a destabilizing agent such as formamide. Exemplary low stringency conditions include hybridization with a buffer solution of 30 to 35% formamide, 1 M NaCl, 1% SDS (sodium dodecyl sulphate) at 37° C., and a wash in 1× to 2×SSC (20×SSC=3.0 M NaCl/0.3 M trisodium citrate) at 50 to 55° C. Exemplary moderate stringency conditions include hybridization in 40 to 45% formamide, 1 M NaCl, 1% SDS at 37° C., and a wash in 0.5× to 1×SSC at 55 to 60° C. Exemplary high stringency conditions include hybridization in 50% formamide, 1 M NaCl, 1% SDS at 37° C. for at least 4 hours, more preferably up to 12 hours or longer, and a final wash in 0.1×SSC at 60 to 65° C. for 30 minutes.
  • Specificity is typically a 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 (thermal melting point) can be approximated from the equation of Meinkoth and Wahl, Anal. Biochem., 138:267-284 (1984): 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 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 Tm; moderately stringent conditions can utilize a hybridization and/or wash at 6, 7, 8, 9, or 10° C. lower than the Tm; low stringency conditions can utilize a hybridization and/or wash at 11, 12, 13, 14, 15, or 20° C. lower than the 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 Ausubel.
  • 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 a 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 the introduction of a polynucleotide of the present invention into a host cell. 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 a polynucleotide/polypeptide of the present invention with a reference polynucleotide/polypeptide: (a) “reference sequence”, (b) “comparison window”, (c) “sequence identity”, and (d) “percentage of sequence identity”.
  • (a) As used herein, a “reference sequence” is a defined sequence used as a basis for sequence comparison with a polynucleotide/polypeptide of the present invention. A reference sequence may be a subset of, 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.
  • (b) As used herein, a “comparison window” includes reference to a contiguous and specified segment of a polynucleotide/polypeptide sequence, wherein the polynucleotide/polypeptide sequence may be compared to a reference sequence and wherein the portion of the polynucleotide/polypeptide 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 nucleotide or amino acid residues 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/polypeptide sequence, a gap penalty is typically introduced and is subtracted from the number of matches.
  • Methods of alignment of sequences for comparison are well-known in the art. Thus, the determination of percent identity between any two sequences can be accomplished using a mathematical algorithm. Optimal alignment of sequences for comparison may be conducted by the local alignment algorithm of Smith and Waterman, Adv. Appl. Math. 2: 482 (1981); by the global alignment algorithm of Needleman and Wunsch, J. Mol. Biol. 48: 443 (1970); by the local alignment method of Pearson and Lipman, Proc. Natl. Acad. Sci. 85: 2444 (1988), by the algorithm of Karlin and Altschul (1990) Proc Natl Acad Sci USA 87: 2264, modified as in Karlin and Altschul (1993) Proc Natl Acad Sci USA 90: 5873-5877.; by computerized implementations of these algorithms, including, but not limited to: CLUSTAL in the PC/Gene program by Intelligenetics, Mountain View, Calif.; GAP, BESTFIT, BLAST, FASTA, and TFASTA in the Wisconsin Genetics Software Package®, Genetics Computer Group (GCG®), (Accelrys, Inc., San Diego, Calif.). The CLUSTAL program is well described by Higgins and Sharp, Gene 73: 237-244 (1988); Higgins and Sharp, CABIOS 5: 151-153 (1989); Corpet, et al., Nucleic Acids Research 16: 10881-90 (1988); Huang, et al., Computer Applications in the Biosciences 8: 155-65 (1992), and Pearson, et al., Methods in Molecular Biology 24: 307-331 (1994).
  • 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-Interscience, New York (1995).
  • Software for performing BLAST analyses is publicly available, e.g., through the National Center for Biotechnology Information website, located on the world wide web at the address ncbi.nlm.nih.gov, preceded by the www prefix. This algorithm involves first identifying high scoring sequence pairs (HSPs) by identifying short words of length W in the query sequence, which either match or satisfy some positive-valued threshold score T when aligned with a word of the same length in a database sequence. T is referred to as the neighborhood word score threshold. These initial neighborhood word hits act as seeds for initiating searches to find longer HSPs containing them. The word hits are then extended in both directions along each sequence for as far as the cumulative alignment score can be increased. Cumulative scores are calculated using, for nucleotide sequences, the parameters M (reward score for a pair of matching residues; always >0) and N (penalty score for mismatching residues; always <0). For amino acid sequences, a scoring matrix is used to calculate the cumulative score. Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached. The BLAST algorithm parameters W, T, and X determine the sensitivity and speed of the alignment. The BLASTN program (for nucleotide sequences) uses as defaults a wordlength (W) of 11, an expectation (E) of 10, a cutoff of 100, M=5, N=−4, and a comparison of both strands. For amino acid sequences, the BLASTP program uses as defaults a wordlength (W) of 3, an expectation (E) of 10, and the BLOSUM62 scoring matrix (see Henikoff & Henikoff (1989) Proc. Natl. Acad. Sci. USA 89:10915).
  • In addition to calculating percent sequence identity, the BLAST algorithm also performs a statistical analysis of the similarity between two sequences (see, e.g., Karlin & Altschul, Proc. Nat'l. Acad. Sci. USA 90:5873-5877, 1993). One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P(N)), which provides an indication of the probability by which a match between two nucleotide or amino acid sequences would occur by chance.
  • 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, Comput. Chem., 17:149-163, 1993) and XNU (Clayerie and States, Comput. Chem., 17:191-201, 1993) low-complexity filters can be employed alone or in combination.
  • GAP can also be used to compare a polynucleotide or polypeptide of the present invention with a reference sequence. GAP uses the algorithm of Needleman and Wunsch (J. Mol. Biol. 48:443-453, 1970) 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 for protein sequences are 8 and 2, respectively. For nucleotide sequences the default gap creation penalty is 50 while the default gap extension penalty is 3. The gap creation and gap extension penalties can be expressed as an integer selected from the group of integers consisting of from 0 to 200. Thus, for example, the gap creation and gap extension penalties can each independently be: 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, 50, 60, 65 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 & 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 al., Nucleic Acids Res. 25:3389-3402, 1997; Altschul et al., J. Mol. Bio. 215: 403-410, 1990) or to the value obtained using the GAP program using default parameters (see the Wisconsin Genetics Software Package, (Accelrys, Inc., San Diego, Calif.)).
  • (c) 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 the 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, Computer Applic. Biol. Sci., 4: 11-17 (1988) e.g., as implemented in the program PC/GENE (Intelligenetics, Mountain View, Calif., USA).
  • (d) 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.
  • (e) As used herein, “substantial identity” of polynucleotide sequences means that a polynucleotide comprises a sequence that has at least 70% sequence identity, preferably at least 80%, more preferably at least 90%, and most preferably at least 95%, compared to a reference sequence using one of the alignment programs described using standard parameters. One of skill in the art 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 at least 60%, more preferably at least 70%, 80%, 90%, and most preferably at least 95%.
  • The present invention provides, inter alia, compositions and methods for modulating the total level of proteins of the present invention and/or altering their ratios in a plant. “Modulation” is intended to mean an increase or decrease in a particular character, quality, substance, or response.
  • The compositions comprise the nucleotide and amino acid sequence for 38 homologs of cysteine proteinases from maize, wheat, rice, and soybean, as presented in Table 1. These plant cystatin genes are characterized by their cysteine proteinase inhibitory activity. “Plant cystatin genes” is intended to mean genes that are structurally related to plant cystatins, also known as plant cysteine proteinases. As is well known in the art, “proteinases” are also called “proteases” and “peptidases” interchangeably. Thus, “cystatin-like” activity is intended to include the activity of peptides that inhibit the activity of cysteine proteinases in plants. In addition, at least some of these peptides also retain antifungal and/or antibacterial activity. The genes of the present invention are called cystatins after a structural classification of proteins (SCOP) classification system.
    TABLE 1
    The Cystatin Genes of the Present Invention
    Full Length Full Length
    Nucleotide Peptide
    Nucleotide Corresponding Peptide Sequence Sequence
    Plant Gene Name SEQ ID NO: SEQ ID NO: Length (nt) Length (aa)
    Maize Zm-Cys1 1 2 786 135
    Maize Zm-Cys3 3 4 915 134
    Maize Zm-Cys4 5 6 915 134
    Maize Zm-Cys5 7 8 1102 245
    Maize Zm-Cys6 9 10 944 176
    Maize Zm-Cys7 11 12 688 116
    Maize Zm-Cys8 13 14 622 110
    Maize Zm-Cys9 15 16 802 157
    Maize Zm-Cys10 17 18 871 174
    Maize Zm-Cys11 19 20 716 174
    Maize Zm-Cys12 21 22 1102 245
    Maize Zm-Cys13 23 24 761 127
    Maize Zm-Cys14 25 26 749 150
    Soybean Gm-Cys1 27 28 1140 245
    Soybean Gm-Cys2 29 30 552 97
    Soybean Gm-Cys3 31 32 484 103
    Soybean Gm-Cys4 33 34 814 92
    Soybean Gm-Cys5 35 36 504 112
    Soybean Gm-Cys6 37 38 708 104
    Soybean Gm-Cys7 39 40 505 97
    Soybean Gm-Cys8 41 42 573 142
    Soybean Gm-Cys9 43 44 473 114
    Rice Os-Cys1 45 46 797 102
    Rice Os-Cys2 47 48 1091 250
    Rice Os-Cys3 49 50 744 108
    Rice Os-Cys4 51 52 919 184
    Rice Os-Cys5 53 54 798 151
    Rice Os-Cys6 55 56 780 123
    Wheat Ta-Cys1 57 58 626 142
    Wheat Ta-Cys2 59 60 609 125
    Wheat Ta-Cys3 61 62 557 128
    Wheat Ta-Cys4 63 64 608 107
    Wheat Ta-Cys6 65 66 622 107
    Wheat Ta-Cys8 67 68 750 152
    Wheat Ta-Cys9 69 70 801 152
    Wheat Ta-Cys10 71 72 1149 243
    Wheat Ta-Cys11 73 74 959 180
    Wheat Ta-Cys13 75 76 518 127
  • Cystatins are a group of proteins which inhibit the activity of cysteine proteinases. The cystatins identified in vertebrates, insects, and plants have been classified into four groups, all belonging to a cystatin superfamily. Groups 1 through 3 are primarily vertebrate cystatin molecules, while group 4 comprises all the known plant cystatins. Group 1 cystatins are referred to as the stefins, single chain proteins with molecular weights of about 11 kDa, which contain no disulfide bonds or carbohydrates. The second group is referred to as the cystatins, and comprise single chain proteins of about 13 kDa, with two disulfide bonds located toward the carboxyl terminus. The kininogens, group 3, are the largest of the cystatins. They are characterized by having three Type 2-like domains, bound carbohydrates, and an additional polypeptide (kinin) unrelated to the cystatin segments.
  • The cysteine proteinase inhibitors of plant origin have been grouped into a fourth cystatin family, the “phytocystatins,” or “plant cystatins” based on their sequence similarity and absence of disulfide bonds or cysteine residues. The phytocystatins are single polypeptide chains with molecular weights ranging from 10 to 16 kDa. Many share several reported conserved sequence motifs, including glycine residue(s) in the vicinity of the N-terminal region, a Gln-Xaa-Val-Xaa-Gly (SEQ ID NO: 77) motif in the first hairpin loop, and a Pro-Trp in the second hairpin loop. In addition, many also share a longer conserved sequence at a part of the N-terminal α-1 helix identified as Leu-Ala-Arg-[Phe or Tyr]-Ala-[Val or Ile]-Xaa-Xaa-Xaa-Asn (SEQ ID NO: 78) (Margis et al. (1998) Arch Biochem Biophys 359(1): 24-30). After an examination of 32 members of the plant cystatin family, Margis et al. (supra) indicate this conserved region of the N-terminal α-1 helix can be rewritten as [Leu or Val or Ile]-[Ala or Gly or Thr]-[Arg or Lys or Glu]-[Phe or Tyr]-[Ala or Ser]-[Val or Ile]-Xaa-[Glu or Asp or Gln or Val]-[His or Tyr or Phe or Gln]-Asn (SEQ ID NO: 79).
  • Analysis of the 38 plant cystatins (see Table 62—multiple sequence alignment) of the present invention when compared with those analyzed by Margis et al. (supra) shows this N-terminal domain analysis to be generally consistent across the plant cystatin group. The sequences examined by Margis et al. were primarily dicot sequences, only 5 of 32 sequences were monocot species. The present analysis (Table 62) shows some trends that are more particular to monocot species. In particular, Table 62 shows that the fourth residue of the N-terminal α-1 helix, shown by Margis et al. to be [Phe or Tyr], should be expanded to be [Phe or Tyr or Trp]. This is supported by the fact that all eight of the sequences of the instant invention not having a phenylalanine or tyrosine at position 4 had a tryptophan residue instead, and is further supported by the chemical similarity of tryptophan to both phenylalanine and tyrosine, which are often referred to as the aromatics group of amino acids. Similarly, in the first hairpin loop, while the Margis et al. analysis showed the consensus sequence of the final six amino acids to be Thr-Met-Tyr-Tyr-Ile-Thr (SEQ ID NO: 80), the present analysis showed this consensus to be Thr-Leu-Tyr-Tyr-Leu-Thr (SEQ ID NO: 81), in which quite often the Tyr-Tyr was replaced with His-His. The replacement of the methionine residue with a leucine and the isoleucine residue with a leucine is not surprising in view of the fact that methionine, leucine, isoleucine and valine are all in the same family and are considered to be conservative substitutes for each other.
  • Table 2 shows the sequences of the first and second hairpin loops and their locations in the cystatin sequences of the present invention. In particular, the highly conserved nature of the QXVXG (SEQ ID NO: 77) motif within the first hairpin loop is evident. Furthermore, the second hairpin loop, while less highly conserved than the first, generally presents a tryptophan residue at the end of the motif.
    TABLE 2
    First Hairpin Loop (FHL) and Second Hairpin Loop (SHL)
    Motifs of Plant Cystatin Genes
    FHL FHL SHL SHL
    Full Motif Start Motif Start
    Protein SEQ Amino SEQ Amino
    SEQ Gene ID Acid ID Acid
    ID NO: Name FHL Motif NO: Position SHL Motif NO: Position
    28 gm-cys1 QVVAGTLHHLT 82 93 EAKVWVKPW 120 116
    30 gm-cys2 QVVSGTLYTIT 83 49 EAKVWEKSW 121 72
    32 gm-cys3 QVVSGTLYYIT 84 49 ETKVLEKPW 122 72
    34 gm-cys4 QVVEGFIYYIT 85 40 ETKVWVRSW 123 63
    36 gm-cys5 QVVSGTNYRLV 86 68 EAIVWEKPW 124 91
    38 gm-cys6 QVVSGMKYYLK 87 54 TSVVVVKPW 125 77
    40 gm-cys7 QVVSGTLYTIT 88 49 EAKVWEKAW 126 72
    42 gm-cys8 QVVSGMKYYLK 89 80 NSVVVVKPW 127 103
    44 gm-cys9 QVVAGLNYRLS 90 70 QAIVYEKAW 128 90
    46 os-cys1 QVVAGTLYYFT 91 53 EAKVWEKPW 129 76
    48 os-cys2 QVVAGTLHHLT 92 93 EAKVWVKPW 130 116
    50 os-cys3 QVVGGFMHYLT 93 59 EAKVWERAW 131 83
    52 os-cys4 QVVTGTLHDLM 94 90 SAKVWVKPW 132 113
    54 os-cys5 QVVSDVAYYLK 95 96 DAVVVVKAW 133 127
    56 os-cys6 QVVSGMNYRLV 96 76 VAVVYEQSW 134 100
    58 ta-cys1 QTVAGTMHYIT 97 93 EAKVWEKPW 135 116
    72 ta-cys10 QTVAGTVHHLT 98 86 EAKVWVKPW 136 109
    74 ta-cys11 QVVAGTLHDLM 99 85 KAKVWVKPW 137 108
    76 ta-cys13 QVVAGTMYYLT 100 78 EAKVWEKPW 138 101
    60 ta-cys2 QTVAGTMHYIT 101 76 EAKVWEKPW 139 99
    62 ta-cys3 QLVSGMNYELI 102 83 KAEVYEQTW 140 107
    64 ta-cys4 QVVAGCMHYFT 103 63 EAKVWEKAW 141 86
    66 ta-cys6 QVVAGCMHYFT 104 63 EAKVWEKAW 142 86
    68 ta-cys8 QVVSGIKYYLR 105 98 DAVVVVKPW 143 129
    70 ta-cys9 QVVSGIKYYLR 106 98 DAVVVVKPW 144 129
     2 ZmCys1 QVVAGTMYYLT 107 86 EAKVWEKPW 145 109
    18 ZmCys10 QVVTGTLHDLI 108 77 RAKVWVKSW 146 100
    20 ZmCys11 QVVAGTNYKLN 109 131 QAVVFDPLP 147 152
    22 ZmCys12 QVVAGTLHHLT 110 87 EAKVWVKPW 148 110
    24 ZmCys13 QIVAGKNYRLR 111 83 RAVVYEQLT 149 107
    26 ZmCys14 QVVSGLKYYLR 112 99 DAVVVVKPW 150 127
     4 ZmCys3 QVVAGTMYYLT 113 85 EAKVWEKPW 151 108
     6 ZmCys4 QVVAGTMYYLT 114 85 EAKVWEKPW 152 108
     8 ZmCys5 QVVAGTLHHLT 115 87 EAKVWVKPW 153 110
    10 ZmCys6 QVVTGTLHDLI 116 80 RAKVWVKPW 154 103
    12 ZmCys7 QVVSGMNYKLV 117 71 GAFVYEQSW 155 95
    14 ZmCys8 QVVAGTLHHFT 118 59 EAKVWEKAW 156 84
    16 ZmCys9 QVVSGMNYRLY 119 81 VAVVYEQVW 157 105
  • Table 3 shows the conserved region of the N-terminal alpha-1-helix in each of the sequences of the present invention.
    TABLE 3
    N-terminal Alpha-1 Helix Motif
    Full Motif
    Protein Motif Start
    SEQ SEQ Amino
    ID Gene Motif ID Acid
    NO: Name Sequence NO: Position
    28 gm-cys1 LARFAVDEHN 158 66
    30 gm-cys2 LARFAVEEHN 159 22
    32 gm-cys3 LARFAVDEHN 160 22
    34 gm-cys4 LARFAVEEQN 161 13
    36 gm-cys5 IANYALSEYD 162 41
    38 gm-cys6 LGRFAVEEYN 163 20
    40 gm-cys7 LARFAVEEHN 164 22
    42 gm-cys8 LGRFAVEEYN 165 42
    44 gm-cys9 IANFAVTEYD 166 43
    46 os-cys1 LARFAVTEHN 167 26
    48 os-cys2 LARFAVDEHN 168 66
    50 os-cys3 LARFAVAEHN 169 32
    52 os-cys4 AARFAVAEYN 170 63
    54 os-cys5 LGRFAVAEHN 171 57
    56 os-cys6 LGGWAVERHA 172 49
    58 ta-cys1 LARFAVSEHN 173 66
    72 ta-cys10 LARFAVDEHN 174 59
    74 ta-cys11 AARFAVAEHN 175 58
    76 ta-cys13 LARFAVDEHN 176 51
    60 ta-cys2 LARFAVSEHN 177 49
    62 ta-cys3 LGRWAVLEFG 178 56
    64 ta-cys4 LARFAVAEHN 179 36
    66 ta-cys6 LARFAVAEHN 180 36
    68 ta-cys8 LGRYSVEEHN 181 61
    70 ta-cys9 LGRYSVEEHN 182 61
     2 ZmCys1 LARFAVNEHN 183 59
    18 ZmCys10 AARFAVAHYN 184 50
    20 ZmCys11 VGEWAVKEHN 185 104
    22 ZmCys12 LGRFAVDEHN 186 60
    24 ZmCys13 IGRWAVSEHI 187 56
    26 ZmCys14 LGRFSVAEYN 188 66
     4 ZmCys3 LARFAVDEHN 189 58
     6 ZmCys4 LARFAVDEHN 190 58
     8 ZmCys5 LGRFAVDEHN 191 60
    10 ZmCys6 AARFAVAYHN 192 53
    12 ZmCys7 LGGWAVTEHV 193 44
    14 ZmCys8 LARFAVAEHN 194 32
    16 ZmCys9 LGGWALGQAK 195 35
  • The protein sequences of the present invention were analyzed for percent identities and similarities using the GAP algorithm. These analyses were performed by species, such that the maize sequences were compared to the other maize sequences, and so on for each of soybean, rice, and wheat. It is evident from the tables that follow that the sequences of the present invention, although they are all cystatins, can vary markedly at the protein level while still retaining cystatin activity. In Tables 4 through 11, sequence similarities and identities among crop plant sequence groups are presented. Those SEQ ID NOs: for which activity data are provided in the instant application are shown in bold faced type (see Examples).
    TABLE 4
    GAP Analysis: Maize Amino Acid Sequence Percent Identities
    SEQ ID NO: 3 5 7 9 11 13 15 18 20 22 24 26
    1 90.3 90.3 44.2 38.7 36.2 56.0 33.0 37.3 34.1 44.3 35.2 33.3
    3 100 45.0 38.2 39.1 56.0 33.9 37.3 33.3 45.0 37.1 33.3
    5 45.0 38.2 39.1 56.0 33.9 37.3 33.3 45.0 37.1 33.3
    7 38.9 33.9 50.5 28.7 36.9 32.3 100 29.5 37.6
    9 36.8 44.9 25.5 88.4 32.5 39.0 24.6 38.1
    11 29.0 57.4 33.9 37.5 33.9 53.9 38.8
    13 25.2 43.5 32.3 50.5 34.7 36.9
    15 28.0 35.2 28.7 43.0 36.7
    18 29.9 36.9 22.5 34.9
    20 32.3 44.3 29.6
    22 29.5 37.6
    24 37.1
  • TABLE 5
    GAP Analysis: Maize Amino Acid Sequence Percent Similarities
    SEQ ID NO: 3 5 7 9 11 13 15 18 20 22 24 26
    1 92.5 92.5 53.4 48.1 44.2 69.2 40.3 46.8 40.0 53.4 44.0 43.2
    3 100 54.2 46.9 47.8 70.1 42.7 45.2 41.7 54.2 48.4 43.4
    5 54.2 46.9 47.8 70.1 42.7 45.2 41.7 54.2 48.4 43.4
    7 46.5 40.0 57.0 35.3 44.6 35.4 100 40.2 44.8
    9 44.7 49.5 34.6 91.3 37.5 46.5 38.5 45.2
    11 38.0 61.1 42.6 45.5 40.0 64.3 46.6
    13 32.7 47.2 38.4 57.0 46.5 45.6
    15 36.7 43.4 35.3 49.6 42.2
    18 36.8 44.6 33.3 41.5
    20 35.4 54.9 35.2
    22 40.2 44.8
    24 48.4
  • TABLE 6
    GAP Analysis: Glycine max Amino Acid Sequence Percent Identities
    SEQ ID NO: 30 32 34 36 38 40 42 44
    28 67.0 55.3 45.7 33.9 41.2 65.0 37.8 27.7
    30 69.1 61.4 41.3 42.2 94.9 38.0 30.8
    32 67.4 39.1 34.0 69.1 33.0 32.2
    34 34.5 37.8 60.2 35.9 32.9
    36 34.4 41.3 32.1 59.6
    38 42.2 82.7 31.8
    40 38.0 31.9
    42 28.4
  • TABLE 7
    GAP Analysis: Glycine max Amino Acid Sequence Percent Similarities
    SEQ ID NO: 30 32 34 36 38 40 42 44
    28 77.3 63.1 59.8 44.6 55.7 75.3 48.8 38.4
    30 78.4 68.2 48.9 51.1 94.9 47.8 42.9
    32 72.8 48.9 47.4 78.4 42.7 41.1
    34 44.0 47.8 67.0 44.6 40.2
    36 42.2 48.9 39.3 67.0
    38 51.1 85.6 42.0
    40 47.8 44.0
    42 36.7
  • TABLE 8
    GAP Analysis: Oryza sativa Amino Acid Sequence Percent Identities
    SEQ ID NO: 48 50 52 54 56
    46 54.9 60.0 40.2 34.7 31.3
    48 48.1 37.0 40.2 34.7
    50 38.3 34.9 36.9
    52 31.3 25.0
    54 42.3
  • TABLE 9
    GAP Analysis: Oryza sativa Amino Acid
    Sequence Percent Similarities
    SEQ ID NO: 48 50 52 54 56
    46 62.7 71.0 49.0 44.9 40.4
    48 57.5 42.0 46.5 41.3
    50 46.7 44.3 40.8
    52 36.7 31.7
    54 48.0
  • TABLE 10
    GAP Analysis: Wheat Amino Acid Sequence Percent Identities
    SEQ ID NO: 60 62 64 66 68 70 72 74 76
    58 92.8 28.6 61.7 61.7 29.9 31.3 46.7 38.8 63.8
    60 33.3 63.6 63.6 31.4 33.1 48.7 38.0 62.6
    62 33.6 33.6 29.7 29.7 30.3 24.4 35.0
    64 98.1 34.0 34.9 47.6 43.0 63.2
    66 33.0 34.0 47.6 42.1 63.2
    68 91.5 38.8 43.4 37.2
    70 35.7 40.0 33.1
    72 38.3 47.9
    74 39.5
  • TABLE 11
    GAP Analysis: Wheat Amino Acid Sequence Percent Similarities
    SEQ ID NO: 60 62 64 66 68 70 72 74 76
    58 93.6 34.1 66.4 65.4 41.8 41.8 54.1 47.8 70.9
    60 39.3 67.3 66.4 43.0 43.8 57.1 47.1 68.3
    62 40.2 40.2 37.5 39.1 41.0 34.4 44.2
    64 98.1 42.5 43.4 57.1 51.4 70.8
    66 41.5 42.5 57.1 50.5 70.8
    68 94.1 46.5 50.0 47.1
    70 44.2 47.7 46.0
    72 43.4 57.0
    74 51.6
  • The phytocystatins play a role in a wide range of plant physiological processes, including plant defense mechanisms. Plant cystatins have been found in various tissues in numerous plant species, including, but not limited to, crop plants such as rice (Abe et al. (1987) J Biol Chem 262: 16793-16797; Kondo et al. (1990) J Biol Chem 265: 15832-15837), tomato (Wu et al. (2000) Comp Biochem Phys C 127: 209-220), maize (Yamada et al. (2000) Plant Cell Physiol 42(7): 710-716), sunflower (Doi-Kawano et al. (1998) J Biochem 124: 11-916), soybean (Misaka et al. (1996) Eur J Biochem 240: 609-614) and potato (Rowan et al. (1990) FEBS Lett 269: 328-330; Waldron et al. (1993) Plant Mol Biol 23: 801-812). Information on their amino acid sequences has been obtained through either protein or cDNA sequencing.
  • Cystatins play a role in many plant physiological functions, including defense, more specifically plant defense against pathogens. A range of functions performed by plant cystatins are responsible for enhancing plant defense against different pathogens. While not wishing to be bound by any one mechanism of action, the sequences and related genes of the present invention encode proteins with antimicrobial and antifungal activity. These proteins may inhibit the proteinases of the pathogen, so as to thwart their utilization of the plant tissue. In addition, cystatins which are expressed around disease-induced lesions may control symptom development, as in a hypersensitive response (HR), by controlling the proteinase-mediated cell death mechanism.
  • Compositions of the present invention include the sequences for maize, soybean, rice and wheat nucleotide sequences which have been identified as cystatins that are involved in plant defense response and development. In particular, the present invention provides for isolated nucleic acid molecules comprising nucleotide sequences encoding the amino acid sequences shown in SEQ ID NOs: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, and 76. Further provided are polypeptides having an amino acid sequence encoded by a nucleic acid molecule described herein, for example those nucleotide sequences set forth in SEQ ID NOs: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 43, 45, 47, 49, 51, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, and 75.
  • The nucleotide sequences of the invention are maize, soybean, rice and wheat sequences comprising plant cysteine proteinases. The claimed sequences are members of the plant cystatin class of genes and polypeptides. These plant cystatins are identified herein as “Zm-Cys”, “Ta-Cys”, “Gm-Cys” and “Os-Cys” for cystatins originating from Zea mays, Triticum aestivum, Glycine max and Oryza sativa, respectively, and are numbered for easy reference (e.g. Zm-Cys10 or Gm-Cys8). These sequences represent a diverse and conserved supergene family in plants.
  • The compositions of the invention can be used in a variety of methods whereby the protein products can be expressed in crop plants to function as antimicrobial proteins. Such expression results in the alteration or modulation of the level, tissue, or timing of expression to achieve enhanced disease or stress resistance. The compositions of the invention may be expressed in the same species from which the particular cystatin originates, or alternatively, can be expressed in any plant of interest. In this manner, the coding sequence for the cystatin can be used in combination with a promoter that is introduced into a crop plant. In one embodiment, a high-level expressing constitutive promoter may be utilized and would result in high levels of expression of the cystatin. In other embodiments, the coding sequence may be operably linked to a tissue-specific promoter to direct the expression to a plant tissue known to be susceptible to a pathogen. Likewise, manipulation of the timing of expression may be utilized. For example, by judicious choice of promoter, expression can be enhanced early in plant growth to prime the plant to be responsive to pathogen attack. Likewise, pathogen inducible promoters can be used wherein expression of the cystatin is turned on in the presence of the pathogen.
  • The cystatin genes of the present invention additionally find use in enhancing the plant pathogen defense system. The compositions and methods of the invention can be used for enhancing resistance to plant pathogens including fungal pathogens, plant viruses, and the like. The method involves stably transforming a plant with a nucleotide sequence capable of modulating the plant pathogen defense system operably linked with a promoter capable of driving expression of a gene in a plant cell. “Enhancing resistance” means that the plant's tolerance to pathogens is increased. That is, the cystatin may slow or prevent pathogen infection and spread.
  • In specific embodiments, methods for increasing pathogen resistance in a plant comprise stably transforming a plant with a DNA construct comprising an anti-pathogenic nucleotide sequence of the invention operably linked to a promoter that drives expression in a plant. Such methods find use in agriculture, particularly in limiting the impact of plant pathogens on crop plants. While the choice of promoter will depend on the desired timing and location of expression of the anti-pathogenic nucleotide sequences, preferred promoters include constitutive and pathogen-inducible promoters.
  • Additionally, the compositions can be used in formulations used for their disease resistance activities. The proteins of the invention can be formulated with an acceptable carrier into a pesticidal composition(s) that is for example, a suspension, a solution, an emulsion, a dusting powder, a dispersible granule, a wettable powder, an emulsifiable concentrate, an aerosol, an impregnated granule, an adjuvant, a coatable paste, or an encapsulation in, for example, polymer substances.
  • Transformed plants, plant cells, plant tissues and seeds thereof are additionally provided.
  • It is recognized that the present invention is not dependent upon a particular mechanism of defense. Rather, the genes and methods of the invention work to increase resistance of the plant to pathogens independent of how that resistance is increased or achieved.
  • It is understood in the art that plant DNA viruses and fungal pathogens remodel the control of the host replication and gene expression machinery to accomplish their own replication and effective infection. The present invention may be useful in preventing such corruption of the cell.
  • The cystatin sequences find use in disrupting cellular function of plant pathogens or insect pests as well as altering the defense mechanisms of a host plant to enhance resistance to disease or insect pests. While the invention is not bound by any particular mechanism of action to enhance disease resistance, the gene products, probably proteins or polypeptides, function to inhibit or prevent diseases in a plant.
  • The methods of the invention can be used with other methods available in the art for enhancing disease resistance in plants. For example, any one of a variety of second nucleotide sequences may be utilized, embodiments of the invention encompass those second nucleotide sequences that, when expressed in a plant, help to increase the resistance of a plant to pathogens. It is recognized that such second nucleotide sequences may be used in either the sense or antisense orientation depending on the desired outcome. Other plant defense proteins include those described in PCT patent publications WO 99/43823 and WO 99/43821, both of which are herein incorporated by reference.
  • Plant senescence is an important trait affecting life cycle duration or maturity, seed dry down, the disease resistance profile, and ‘stay green’, which in turn affect yield, stalk strength, appearance, and nutritional value (silage quality). All of these factors, which relate to cell death processes, are considered in maize breeding efforts. Plant proteinases have been implicated in these processes and thus comprise an area of active research.
  • In particular, cysteine proteinases are induced upon plant organ senescence, such as in tomato leaves (Drake et al. (1996) Plant Mol Biol 30(4): 755-767), sweet potato (Chen et al. (2002) Plant and Cell Phys 43(9): 984-991), and day-lily flowers (Valpuesta et al. (1995) Plant Mol Biol 28(3): 575-582). Furthermore, the maize cysteine proteinase Seel (“Senescence enhanced”) has been linked to the stay green phenotype in maize (Griffiths et al. (1997) Plant Mol Biol 34: 815-821). Cysteine proteinase inhibitors have also been shown to delay flower senescence (Eason et al. (2002) Functional Plant Biol 29(9): 1055-1064). While the biology is undoubtedly complex in senescence, to the extent that the cysteine proteinases are involved in senescence related processes, then their cognate proteinase inhibitors, here cystatins, are involved in the control of the timing and onset of senescence as well. Modulation of cystatins can provide agronomic advantages by promoting or delaying senescence and other developmental signals.
  • In a plant breeding effort, the position of the starting breeding material relative to the desired outcome, will dictate what direction one will want to push a trait. For example, one may want increase or decrease maturity time; generally decrease. To increase senescence one may want to suppress cystatin expression, and to decrease senescence one may want to increase cystatin expression. Methods for increasing or decreasing cystatin expression are outlined elsewhere in this specification. However, tissue-targeted or developmental-targeted expression may be desirable to reach these ends. The proteinase promoters, such as those from Seel, can be useful in conjunction with forward or antisense constructs of the proteinase inhibitor gene in question, to coordinately augment or cancel, respectively, the death-promoting capacity of the cysteine proteinase.
  • The proteinase inhibitor genes herein are useful for controlling the senescence of special crop plant tissues. For certain crops particular tissue or organs are desired to senesce. This includes controlled dropping of cotton leaves to facilitate cotton boll harvesting. Sometimes organs are desired not to senesce, as in the petioles of fruit; premature fruit drop can cause loss of yield. For maize, delay of senescence of the pedicel/hilum region of kernels may be desirable to allow for prolonged kernel fill or delayed maturation of seed, with higher yield and/or higher digestibility as possible outcomes.
  • Over-expression or transgenic expression of proteinase inhibitors provides effective control of both cyst and root knot nematodes. The primary mechanism by which cystatins confer nematode resistance is most likely associated with disruption of nematode development. Currently over-expression of proteinase inhibitors (PIs) offers the most advanced approach for nematode control. Furthermore, transgenic expression of PIs provides effective control of both cyst and root knot nematodes. Recent research has shown the value of using proteinase inhibitors in controlling certain species of nematodes.
  • Oryzacystatin-I (Oc-I) is a cysteine proteinase inhibitor from rice seeds (Abe et al. (1987) Supra), while Oc-1D86 is a modified form of Oc-1 which has shown stronger inhibitory activity (Urwin et al. (1995) Plant J 8: 121-131). When expressed in tomato hairy roots both Oc-1 and Oc-ID86 had a detrimental effect on the growth and development of potato cyst nematode G. pallida (Id.). Similarly, when expressed in transgenic Arabidopsis thaliana Oc-ID86 had a profound effect on the size and fecundity of females of both the beet-cyst nematode Heterodera schachtii and the root-knot nematode Meloidogyne incognita as well as reniform nematode Rotylenchulus reniformis (Urwin et al. (1997) Plant J 12: 455-461; Urwin et al. (1998) Planta 204: 472-479; Urwin et al. (2000) Mol Breeding 6: 257-264). Compositions of the instant invention indicate that cystatins can also confer resistance to soybean cyst nematode (SCN) in soybean and other crops, by inhibiting nematode growth and development.
  • The recovery of viable transgenic plants from crop plants, in particular for monocot cereal crop plants such as maize, rice and wheat, is still a laborious and expensive process. This can be a particular problem when transformation-recalcitrant varieties, often those with desirable breeding characteristics, perform poorly in the transgenic production transformation process. Methods are consequently sought to identify new methods that will improve the transformation and recovery of viable plants.
  • One of the chief problems is cell death in tissue culture. This is caused not only by the general poor viability of some lines in culture, but also by the fact that in order to select for the transformants, often antibiotics are added that kill the non-transformed cells. Amidst this cell death the positively transformed lines are also killed. Recalcitrance to death, or the signals of death, and as well positive cell growth, are thus desirable features.
  • To the extent that these proteinase inhibitors can retard cell death by suppressing proteinase inhibitor activity, they can be used to help transformed cells survive. Cysteine proteinases are known to be induced in the plant HR response, and transgenic ectopically expressed cystatins can counteract this response (Pechan et al. (2000) Plant Cell 12(7): 1031-1040; Solomon et al. (1999) Plant Cell 11 (3): 431-443). The transformed cells receive a copy of one or more of these proteinase inhibitor coding region(s) driven by an appropriate promoter. Promoter choices are discussed elsewhere in this application, however, this embodiment can benefit from the use of a constitutive promoter, or by a transiently expressed promoter targeted to the cell culture phase or induced by plant hormones used in culture. Constitutive expression may help disease resistance generally, and as such, constitutive promoters, for example the ubiquitin promoter, can be useful beyond cell culture. Of course a variety of promoters would be effective. The resulting transformed cells would be more viable. This would effect a cleaner separation of the dying non-transformed cells and allow for cleaner and more rapid growth of the transformed line. Plant transformation techniques would be improved as a result.
  • It should also be recognized that plants can be wounded abiotically, as by drought stress, wind stress (which includes damage by wind-blown soil particles), and chemical and nutrient stress. Such stresses can precipitate cell death that can reduce plant yield. To the extent that these proteinase inhibitors may retard cell death by thwarting proteinase inhibitor activity, they can retard the symptom development of necrosis resulting from these stresses when driven by a death-induced promoter.
  • Second, the proteinase inhibitor genes can have application in the development and implementation of herbicide resistance mechanisms in crop plants. Ectopic expression of the proteinase inhibitors, as in leaves, can result in a retardation of cell death following the application of herbicides. This would be subject to the kind of herbicide used and its mode of action, but it is an area of utility for these genes. Herbicides and herbicide resistance systems are often used as selectable markers in plant transformation experiments. Thus, in a way similar to the herbicide resistance application, these proteinase inhibitor genes can be used as selectable markers—only cells expressing the proteinase inhibitor genes (ectopically) would grow or stay alive in the face of an antibiotic/herbicide medium. This application of course bears direct overlap with the examples given above for improving plant transformation.
  • Cell death can also be a mechanism of male infertility. Consequently similar methods, probably with anther- or tapetum- or pollen-preferred expression, could be a means of enhancing or controlling male fertility. For example, expressing cystatins can suppress cell death and thus suppress sterility, rendering the plants male fertile. This could be used in a conditional situation, where the plants would be sterile until induced to be fertile.
  • Many proteinase inhibitors, including some of the present invention, are expressed in seeds. The chief biological role of seed expression of proteinase inhibitors is to inhibit, or otherwise control, proteinase activities in the seeds. This is especially important during seed development/maturation, in order to regulate protein processing by proteinases. Cereal cysteine proteinases play a chief role in the digestion of seed storage proteins, especially during germination (Gruis et al. (2002) Plant Cell 14(11):2863-2882; Debarros & Larkins (1994) Plant Sci 99(2) 189-197; Koehler & Ho (1990) Plant Physiol 94(1):251-258; Poulle & Jones (1988) Plant Physiol 88(4): 1454-1460). Regulating the activity of cysteine proteinases in seeds prevents undesirable loss of seed proteins, including storage proteins, and also prevents premature germination (Corre et al (2002) Plant Mol Biol 50(4-5):687-698). Furthermore, regulating the processing of proteins can serve as an anti-nutritional/protective agent against microbes, insects, and herbivores. However, crop plant seeds, such as maize caryopses, are mostly intended for animal consumption as feed grain, and some also for human food consumption. As such, the proteinase inhibitors from the seed can inhibit digestive proteinases in the gastrointestinal tract of livestock and humans. This can change the site and extent of digestion of protein and other grain components within, as well as elicit hyper-secretion of pancreatic enzymes. The impact on overall nutritional status may either be positive or negative.
  • Lowering the digestibility of grain proteins lowers the effectiveness of the grain for weight gain for monogastric animals and humans. The reduced protein digestibility will also reduce access of starch-degrading enzymes to starch granules (which are encompassed by a protein matrix), thereby reducing digestible energy content in addition to digestible protein. Moreover, various proteinase inhibitors induce the release of pancreatic cholecystokinin, a known satiety factor resulting in lower feed or food intake (Elsaesser et al. (1990) Cell Tissue Res 262(1): 143-148; Garlicki et al. (1990) Am J Physiol 258: E40-E45; Schwartz et al. (1994) Diabetes Care 17(4): 255-262; Choi et al. (2000) Domest Anim Endocrin 19(3): 159-175). For livestock, this is clearly a negative factor, as well as for undernourished humans in developing countries; reduced caloric value and reduced food intake may be very positive, however, for overweight people.
  • Lowering fermentative proteolysis to reduce the formation of non-protein-nitrogen (NPN) is beneficial, however, for ruminant livestock (dairy and beef cattle, sheep and goats), especially if the protein is of high Biological Value (i.e., of balanced amino acid composition, containing especially lysine, tryptophan, threonine, and methionine). First, silage made of various forages, such as ryegrass and alfalfa, is subject to excessive proteolysis during the ensiling process. Total protein losses can amount to 50% and the dairy cow poorly utilizes the resulting NPN. Proteinase inhibitors can be employed to reduce these proteolytic losses. Second, lowering the proteolysis in the rumen is beneficial to allow otherwise easily digestible high-protein concentrates (such as fat-extracted soybean & canola meals) and high-protein forages (such as alfalfa) to bypass rumen fermentation. Rapid and extensive ruminal breakdown of protein leads to decreased protein efficiency because 1) the rumen microbes do not use the degraded protein as fast as it is broken down, leading to excessive formation of ammonia, much of which will be excreted in the urine as urea, and 2) the microbial protein that is re-synthesized from ammonia is generally of lower biological value than soybean or canola protein.
  • Consequently, to the extent that these cystatin proteinase inhibitors can alter digestion characteristics of the grain, it would be desirable to reduce the level of their expression to increase protein digestibility and energy availability for monogastric livestock and humans. On the other hand, given the reasoning above, it would be desirable to increase the level of cystatin expression to reduce proteolysis in silage and generate rumen by-pass protein for ruminant livestock, and to produce diet foods by reducing the caloric value of cereals and/or inducing satiety.
  • It is this over-expression that would be the best mode of using these cystatin genes, which would be achieved by over-expression of one or more of the cystatin genes. The various advantages and disadvantages of using different promoters to drive such over-expression is well known by those skilled in the art. However, by way of example, a constitutive promoter could drive the expression, but a more ideal promoter would target tissues, such as the grain. For silage production, a high-level vegetative promoter would be desirable. Over-expression of one or several of the cystatins would be technically more easy to achieve than suppression of most of these cystatins, especially given their sequence diversity.
  • The different proteinase inhibitor genes have somewhat different expression profiles. Based upon the maize EST library distributions, Zm-Cys5, Zm-Cys6, Zm-Cys9, Zm-Cys10, Zm-Cys13, and Zm-Cys14 are abundant in, or specific to, maize endosperm or other kernel tissues. Generally, the best mode for this invention (in maize or cereals) is to express the sense version of the proteinase inhibitor genes under the control of a seed-preferred, especially endosperm-preferred, especially R3-R5-preferred promoter, or, in the case of alfalfa, a constitutive promoter. Thus when the sense transcript is produced, it will result in either over-expression or silencing of the targeted proteinase inhibitor gene. There are other methods for suppression of gene expression that may be applied. This strategy could be applied to one or several of the proteinase inhibitor genes in the same crop plant.
  • Sequences of the invention, as discussed in more detail below, encompass coding sequences, antisense sequences, and fragments and variants thereof. Expression of the sequences of the invention can be used to modulate or regulate the expression of corresponding cystatin proteins. The invention encompasses isolated or substantially purified nucleic acid or protein compositions. An “isolated” or “purified” nucleic acid molecule or protein, or biologically active portion thereof, is substantially free of other cellular material, or culture medium when produced by recombinant techniques, or substantially free of chemical precursors or other chemicals when chemically synthesized. Preferably, an “isolated” nucleic acid is free of sequences (preferably protein encoding sequences) that naturally flank the nucleic acid (i.e., sequences located at the 5′ and 3′ ends of the nucleic acid) in the genomic DNA of the organism from which the nucleic acid is derived. For example, in various embodiments, the isolated nucleic acid molecule can contain less than about 5 kb, 4 kb, 3 kb, 2 kb, 1 kb, 0.5 kb, or 0.1 kb of nucleotide sequences that naturally flank the nucleic acid molecule in genomic DNA of the cell from which the nucleic acid is derived. A protein that is substantially free of cellular material includes preparations of protein having less than about 30%, 20%, 10%, 5%, (by dry weight) of contaminating protein. When the protein of the invention or biologically active portion thereof is recombinantly produced, preferably culture medium represents less than about 30%, 20%, 10%, or 5% (by dry weight) of chemical precursors or non-protein-of-interest chemicals.
  • Fragments and variants of the disclosed nucleotide sequences and proteins encoded thereby are also encompassed by the present invention. “Fragment” means a portion of the nucleotide sequence or a portion of the amino acid sequence and hence protein encoded thereby. Fragments of a nucleotide sequence may encode protein fragments that retain the biological activity of the native protein and hence have cystatin-like activity and thereby affect development, developmental pathways, and defense responses. Alternatively, fragments of a nucleotide sequence that are useful as hybridization probes generally do not encode fragment proteins retaining biological activity. Thus, fragments of a nucleotide sequence may range from at least about 20 nucleotides, about 50 nucleotides, about 100 nucleotides, and up to the full-length nucleotide sequence encoding the proteins of the invention.
  • A fragment of a cystatin nucleotide sequence that encodes a biologically active portion of a cystatin protein of the invention will encode at least 15, 25, 30, 50, 100, 150, 200, or 250 contiguous amino acids, or up to the total number of amino acids present in a full-length protein of the invention (for example, 135, 134, 134, 245, 176, 116, 110 or 157 amino acids for SEQ ID NO:2, 4, 6, 8, 10, 12, 14, or 16, respectively). Fragments of a cystatin nucleotide sequence that are useful as hybridization probes for PCR primers generally need not encode a biologically active portion of a cystatin protein.
  • Thus, a fragment of a cystatin nucleotide sequence may encode a biologically active portion of a cystatin protein, or it may be a fragment that can be used as a hybridization probe or PCR primer using methods disclosed below. A biologically active portion of a cystatin protein can be prepared by isolating a portion of one of the cystatin nucleotide sequences of the invention, expressing the encoded portion of the cystatin protein (e.g., by recombinant expression in vitro), and assessing the activity of the encoded portion of the cystatin protein. Nucleic acid molecules that are fragments of a cystatin nucleotide sequence comprise at least 16, 20, 50, 75, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, or 800 nucleotides, or up to the number of nucleotides present in a full-length cystatin nucleotide sequence disclosed herein (for example, 408, 405, 405, 738, 531, 351, 333, or 474 nucleotides for SEQ ID NO:1, 3, 5, 7, 9, 11, 13, or 15, respectively).
  • “Variants” is intended to mean substantially similar sequences. For nucleotide sequences, conservative variants include those sequences that, because of the degeneracy of the genetic code, encode the amino acid sequence of one of the cystatin polypeptides of the invention. Naturally occurring allelic variants such as these can be identified with the use of well-known molecular biology techniques, as, for example, with polymerase chain reaction (PCR) and hybridization techniques as outlined below. Variant nucleotide sequences also include synthetically derived nucleotide sequences, such as those generated, for example, by using site-directed mutagenesis but which still encode a cystatin protein of the invention. Generally, variants of a particular nucleotide sequence of the invention will have at least about 50%, 60%, 65%, 70%, generally at least about 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to that particular nucleotide sequence as determined by sequence alignment programs described elsewhere herein using default parameters.
  • These variant nucleotide sequences can also be evaluated by comparison of the percent sequence identity shared by the polypeptides they encode. For example, isolated nucleic acids which encode a polypeptide with a given percent sequence identity to the polypeptide of SEQ ID NO: 2, 4, 6, 8 and 10 are disclosed. Identity can be calculated using, for example, the BLAST, CLUSTALW, or GAP algorithms under default conditions. The percentage of identity to a reference sequence is at least 50% and, rounded upwards to the nearest integer, can be expressed as an integer selected from the group of integers consisting of from 50 to 99. Thus, for example, the percentage of identity to a reference sequence can be at least 60%, 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%.
  • A “variant” protein is intended to mean a protein derived from the native protein by deletion (so-called truncation) or addition of one or more amino acids to the N-terminal and/or C-terminal end of the native protein; deletion or addition of one or more amino acids at one or more sites in the native protein; or substitution of one or more amino acids at one or more sites in the native protein. Variant proteins encompassed by the present invention are biologically active, that is they continue to possess the desired biological activity of the native protein, that is, cystatin-like activity as described herein. Such variants may result from, for example, genetic polymorphism or from human manipulation. Biologically active variants of a native cystatin protein of the invention will have at least about 40%, 50%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to the amino acid sequence of the native protein as determined by sequence alignment programs described elsewhere herein using default parameters. A biologically active variant of a protein of the invention may differ from that protein by as few as 1-15 amino acid residues, as few as 1-10, such as 6-10, as few as 5, as few as 4, 3, 2, or even 1 amino acid residue.
  • The polypeptides of the invention may be altered in various ways including amino acid substitutions, deletions, truncations, and insertions. Novel proteins having properties of interest may be created by combining elements and fragments of proteins of the present invention as well as other proteins. Methods for such manipulations are generally known in the art. For example, amino acid sequence variants of the cystatin proteins can be prepared by mutations in the DNA. Methods for mutagenesis and nucleotide sequence alterations are well known in the art. See, for example, Kunkel (1985) Proc Nat Acad Sci USA 82:488-492; Kunkel et al. (1987) Method Enzymol 154:367-382; U.S. Pat. No. 4,873,192; Walker and Gaastra, eds. (1983) Techniques in Molecular Biology (MacMillan Publishing Company, New York) and the references cited therein. Guidance as to appropriate amino acid substitutions that do not affect biological activity of the protein of interest are well known in the art and may be found in the model of Dayhoff et al. (1978) Atlas of Protein Sequence and Structure (Natl. Biomed. Res. Found., Washington, D.C.), herein incorporated by reference. Conservative substitutions, such as exchanging one amino acid with another having similar properties, may be preferred. Table 12, below, shows potential amino acid substitution groups which are considered to be highly conserved.
    TABLE 12
    Conservative Substitution Groups
    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)
    6 Phenylalanine (F) Tyrosine (Y) Tryptophan (W)
  • Thus, the genes and nucleotide sequences of the invention include both the naturally occurring sequences as well as mutant forms. Likewise, the proteins of the invention encompass both naturally occurring proteins as well as variations and modified forms thereof. Such variants will continue to possess the desired developmental activity, or defense response activity. Obviously, the mutations that will be made in the DNA encoding the variant must not place the sequence out of reading frame and preferably will not create complementary regions that could produce secondary mRNA structures. See, EP Patent Application Publication No. 0075444.
  • In nature, some polypeptides are produced as complex precursors which, in addition to targeting labels such as the signal peptides discussed elsewhere in this application, also contain other fragments of peptides which are removed (processed) at some point during protein maturation, resulting in a mature form of the polypeptide that is different from the primary translation product (aside from the removal of the signal peptide). “Mature protein” refers to a post-translationally processed polypeptide; i.e., one from which any pre- or propeptides present in the primary translation product have been removed. “Precursor protein” or “prepropeptide” or “preproprotein” all refer to the primary product of translation of mRNA; i.e., with pre- and propeptides still present. Pre- and propeptides may include, but are not limited to, intracellular localization signals. “Pre” in this nomenclature generally refers to the signal peptide. The form of the translation product with only the signal peptide removed but not further processing yet is called a “propeptide” or “proprotein”. The fragments or segments to be removed may themselves also be referred to as “propeptides.” A proprotein or propeptide thus has had the signal peptide removed, but contains propeptides (here referring to propeptide segments) and the portions that will make up the mature protein. The skilled artisan is able to determine, depending on the species in which the proteins are being expressed and the desired intracellular location, if higher expression levels might be obtained by using a gene construct encoding just the mature form of the protein, the mature form with a signal peptide, or the proprotein (i.e., a form including propeptides) with a signal peptide. For optimal expression in plants or fungi, the pre- and propeptide sequences may be needed. The propeptide segments may play a role in aiding correct peptide folding.
  • The deletions, insertions, and substitutions of the protein sequences encompassed herein are not expected to produce radical changes in the characteristics of the protein. However, when it is difficult to predict the exact effect of the substitution, deletion, or insertion in advance of doing so, one skilled in the art will appreciate that the effect can be evaluated by routine screening assays. That is, the activity can be evaluated by cystatin activity assays. Additionally, differences in the expression of specific genes between uninfected and infected plants can be determined using gene expression profiling.
  • Variant nucleotide sequences and proteins also encompass sequences and proteins derived from a mutagenic and recombinogenic procedure such as DNA shuffling. With such a procedure, one or more different cystatin coding sequences can be manipulated to create a new cystatin protein possessing the desired properties. In this manner, libraries of recombinant polynucleotides are generated from a population of related sequence polynucleotides comprising sequence regions that have substantial sequence identity and can be homologously recombined in vitro or in vivo. For example, using this approach, sequence motifs encoding a domain of interest may be shuffled between the cystatin genes and partial sequences of the invention and other known cystatin genes to obtain a new gene coding for a protein with an improved property of interest, such as an increased Km in the case of an enzyme. Such shuffling of domains may also be used to assemble novel proteins having novel properties. Strategies for such DNA shuffling are known in the art. See, for example, Stemmer (1994) Proc Natl Acad Sci USA 91: 10747-10751; Stemmer (1994) Nature 370: 389-391; Crameri et al. (1997) Nature Biotech 15: 436-438; Moore et al. (1997) J Mol Biol 272: 336-347; Zhang et al. (1997) Proc Natl Acad Sci USA 94: 4504-4509; Crameri et al. (1998) Nature 391: 288-291; and U.S. Pat. Nos. 5,605,793 and 5,837,458.
  • The nucleotide sequences of the invention can be used to isolate corresponding sequences from other organisms, particularly other plants, more particularly other monocots. In this manner, methods such as PCR, hybridization, and the like can be used to identify such sequences based on their sequence homology to the sequences set forth herein. Sequences isolated based on their sequence identity to the entire cystatin sequences set forth herein or to fragments thereof are encompassed by the present invention. Such sequences include sequences that are orthologs of the disclosed sequences. “Orthologs” means genes derived from a common ancestral gene and which are found in different species as a result of speciation. Genes found in different species are considered orthologs when their nucleotide sequences and/or their encoded protein sequences share substantial identity as defined elsewhere herein. Functions of orthologs are often highly conserved among species.
  • In a PCR approach, oligonucleotide primers can be designed for use in PCR reactions to amplify corresponding DNA sequences from cDNA or genomic DNA extracted from any plant of interest. Methods for designing PCR primers and PCR cloning are generally known in the art and are disclosed in, for example, Sambrook. See also Innis et al., eds. (1990) PCR Protocols: A Guide to Methods and Applications (Academic Press, New York); Innis and Gelfand, eds. (1995) PCR Strategies (Academic Press, New York); and Innis and Gelfand, eds. (1999) PCR Methods Manual (Academic Press, New York). Known methods of PCR include, but are not limited to, methods using paired primers, nested primers, single specific primers, degenerate primers, gene-specific primers, vector-specific primers, partially-mismatched primers, and the like.
  • In hybridization techniques, all or part of a known nucleotide sequence is used as a probe that selectively hybridizes to other corresponding nucleotide sequences present in a population of cloned genomic DNA fragments or cDNA fragments (i.e., genomic or cDNA libraries) from a chosen organism. The hybridization probes may be genomic DNA fragments, cDNA fragments, RNA fragments, or other oligonucleotides, and may be labeled with a detectable group such as 32P, or any other detectable marker. Thus, for example, probes for hybridization can be made by labeling synthetic oligonucleotides based on the cystatin sequences of the invention. Methods for preparation of probes for hybridization and for construction of cDNA and genomic libraries are generally known in the art and are disclosed in Sambrook.
  • For example, an entire cystatin sequence disclosed herein, or one or more portions thereof, may be used as a probe capable of specifically hybridizing to corresponding cystatin sequences and messenger RNAs. To achieve specific hybridization under a variety of conditions, such probes include sequences that are unique among cystatin sequences and are preferably at least about 10 nucleotides in length, and most preferably at least about 20 nucleotides in length. Such probes may be used to amplify corresponding sequences from a chosen organism by PCR. This technique may be used to isolate additional coding sequences from a desired organism or as a diagnostic assay to determine the presence of coding sequences in an organism. Hybridization techniques include hybridization screening of plated DNA libraries (either plaques or colonies; see, for example, Sambrook.
  • Thus, isolated sequences that encode for a cystatin polypeptide and which hybridize under stringent conditions to the cystatin sequences disclosed herein, or to fragments thereof, are encompassed by the present invention.
  • Biological activity of the cystatin polypeptides (i.e., influencing the plant defense response and various developmental pathways, including, for example, influencing cell division) can be assayed by any method known in the art. Biological activity of the polypeptides of the present invention can be assayed by any method known in the art. For example, most published cystatin activity assays are based on inhibition of papain-mediated substrate hydrolysis. A variety of synthetic papain substrates are known in the art and can be used for this purpose, such as N-benzoyl-asparaginyl-p-nitroanilide (Schlereth et al. (2001) Planta 212: 718-727), α-N-benzoyl-L-arginine-p-nitroanilide (Masoud et al. (1993) Plant Mol Biol 21: 655-663), N-Cbz-Phe-Arg-7-amido-4-methylcoumarin (Urwin et al. (1998) Supra), and Z-Phe-Arg-7-(4-methylcoumarylamide) (Barrett & Kirschke (1981) Method Enzymol 80: 535-561), all of which are herein incorporated by reference. Furthermore, papain could be substituted by a cysteine proteinase that is more relevant to the biological system studied (e.g., a Fusarium cysteine proteinase). Assays to detect cystatin-like activity include, for example, assessing antifungal and/or antimicrobial activity (Soares-Costa et al. (2002) Biochem Biophys Res Comm 296: 1194-1199; Duvick et al. (1992) J Biol Chem 267(26): 18814-18820; Pernas-Monica et al. (1999) Mol Plant Microbe In 12 (7): 624-627; Blankenvoorde-Michiel et al. (1998) Biol Chem 379(11): 1371-1375, all of which are herein incorporated by reference).
  • Assays that measure antipathogenic activity are commonly known in the art, as are methods to quantitate disease resistance in plants following pathogen infection. See, for example, U.S. Pat. No. 5,614,395, herein incorporated by reference. Such techniques include, measuring over time, the average lesion diameter, the pathogen biomass, and the overall percentage of decayed plant tissues. For example, a plant either expressing an antipathogenic polypeptide or having an antipathogenic composition applied to its surface shows a decrease in tissue necrosis (i.e., lesion diameter) or a decrease in plant death following pathogen challenge when compared to a control plant that was not exposed to the antipathogenic composition. Alternatively, antipathogenic activity can be measured by a decrease in pathogen biomass. For example, a plant expressing an antipathogenic polypeptide or exposed to an antipathogenic composition is challenged with a pathogen of interest. Over time, tissue samples from the pathogen-inoculated tissues are obtained and RNA is extracted. The percent of a specific pathogen RNA transcript relative to the level of a plant specific transcript allows the level of pathogen biomass to be determined. See, for example, Thomma et al. (1998) Plant Biology 95:15107-15111, herein incorporated by reference.
  • Furthermore, in vitro antipathogenic assays include, for example, the addition of varying concentrations of the antipathogenic composition to paper disks and placing the disks on agar containing a suspension of the pathogen of interest. Following incubation, clear inhibition zones develop around the discs that contain an effective concentration of the antipathogenic polypeptide (Liu et al. (1994) Plant Biology 91:1888-1892, herein incorporated by reference). Additionally, microspectrophotometrical analysis can be used to measure the in vitro antipathogenic properties of a composition (Hu et al. (1997) Plant Mol. Biol. 34:949-959 and Cammue et al. (1992) J. Biol. Chem. 267: 2228-2233, both of which are herein incorporated by reference).
  • Compositions and methods for controlling pathogenic agents are provided. The anti-pathogenic compositions comprise maize, soybean, rice and wheat cystatin nucleotide and amino acid sequences. Particularly, the nucleic acid and amino acid sequences and fragments and variants thereof set forth herein. Accordingly, the compositions and methods are also useful in protecting plants against fungal pathogens, viruses, nematodes, insects and the like.
  • “Plant pathogen” or “plant pest” is intended to mean any microorganism that can cause harm to a plant, such as by inhibiting or slowing the growth of a plant, by damaging the tissues of a plant, by weakening the immune system of a plant or the resistance of a plant to abiotic stresses, and/or by causing the premature death of the plant, etc. Plant pathogens and plant pests include microbes such as fungi, viruses, bacteria, and nematodes.
  • “Disease resistance” or “pathogen resistance” is intended to mean that the organisms avoid the disease symptoms which are the outcome of organism-pathogen interactions. That is, pathogens are prevented from causing diseases and the associated disease symptoms, or alternatively, the disease symptoms caused by the pathogen is minimized or lessened. The methods of the invention can be utilized to protect plants from disease, particularly those diseases that are caused by plant pathogens. “Anti-pathogenic compositions” is intended to mean that the compositions of the invention are capable of suppressing, controlling, and/or killing the invading pathogenic organism. An antipathogenic composition of the invention will reduce the disease symptoms resulting from pathogen challenge by at least about 5% to about 50%, at least about 10% to about 60%, at least about 30% to about 70%, at least about 40% to about 80%, or at least about 50% to about 90% or greater. Hence, the methods of the invention can be utilized to protect plants from disease, particularly those diseases that are caused by plant pathogens.
  • An “antimicrobial agent,” a “pesticidal agent,” a “cystatin,” and/or a “fungicidal agent” will act similarly to suppress, control, and/or kill the invading pathogen. A defensive agent will possess defensive activity. “Defensive activity” means an antipathogenic, antimicrobial, or antifungal activity.
  • “Antipathogenic compositions” is intended to mean that the compositions of the invention have activity against pathogens; including fungi, microorganisms, viruses, and nematodes, and thus are capable of suppressing, controlling, and/or killing the invading pathogenic organism. An antipathogenic composition of the invention will reduce the disease symptoms resulting from plant pathogen challenge by at least about 5% to about 50%, at least about 10% to about 60%, at least about 30% to about 70%, at least about 40% to about 80%, or at least about 50% to about 90% or greater. Hence, the methods of the invention can be utilized to protect organisms, particularly plants, from disease, particularly those diseases that are caused by invading pathogens.
  • Pathogens of the invention include, but are not limited to, viruses or viroids, bacteria, insects, nematodes, fungi, and the like. Viruses include any plant virus, for example, tobacco or cucumber mosaic virus, ringspot virus, necrosis virus, maize dwarf mosaic virus, etc. Specific fungal and viral pathogens for the major crops include, but are not limited to: Soybeans: Phytophthora megasperma fsp. glycinea, Macrophomina phaseolina, Rhizoctonia solani, Sclerotinia sclerotiorum, Fusarium oxysporum, Diaporthe phaseolorum var. sojae (Phomopsis sojae), Diaporthe phaseolorum var. caulivora, Sclerotium rolfsii, Cercospora kikuchii, Cercospora sojina, Peronospora manshurica, Colletotrichum dematium (Colletotichum truncatum), Corynespora cassiicola, Septoria glycines, Phyllosticta sojicola, Alternaria alternata, Pseudomonas syringae p.v. glycinea, Xanthomonas campestris p.v. phaseoli, Microsphaera diffusa, Fusarium semitectum, Phialophora gregata, Soybean mosaic virus, Glomerella glycines, Tobacco Ring spot virus, Tobacco Streak virus, Phakopsora pachyrhizi, Pythium aphanidermatum, Pythium ultimum, Pythium debaryanum, Tomato spotted wilt virus, Heterodera glycines, Fusarium solani; Canola: Albugo candida, Alternaria brassicae, Leptosphaeria maculans, Rhizoctonia solani, Sclerotinia sclerotiorum, Mycosphaerella brassiccola, Pythium ultimum, Peronospora parasitica, Fusarium roseum, Alternaria alternata; Alfalfa: Clavibater michiganese subsp. insidiosum, Pythium ultimum, Pythium irregulare, Pythium splendens, Pythium debaryanum, Pythium aphanidermatum, Phytophthora megasperma, Peronospora trifoliorum, Phoma medicaginis var. medicaginis, Cercospora medicaginis, Pseudopeziza medicaginis, Leptotrochila medicaginis, Fusarium, Xanthomonas campestris p.v. alfalfae, Aphanomyces euteiches, Stemphylium herbarum, Stemphylium alfalfae; Wheat: Pseudomonas syringae p.v. atrofaciens, Urocystis agropyri, Xanthomonas campestris p.v. translucens, Pseudomonas syringae p.v. syringae, Alternaria alternata, Cladosporium herbarum, Fusarium graminearum, Fusarium avenaceum, Fusarium culmorum, Ustilago tritici, Ascochyta tritici, Cephalosporium gramineum, Collotetrichum graminicola, Erysiphe graminis f.sp. tritici, Puccinia graminis f.sp. tritici, Puccinia recondita f.sp. tritici, Puccinia striiformis, Pyrenophora tritici-repentis, Septoria nodorum, Septoria tritici, Septoria avenae, Pseudocercosporella herpotrichoides, Rhizoctonia solani, Rhizoctonia cerealis, Gaeumannomyces graminis var. tritici, Pythium aphanidermatum, Pythium arrhenomanes, Pythium ultimum, Bipolaris sorokiniana, Barley Yellow Dwarf Virus, Brome Mosaic Virus, Soil Borne Wheat Mosaic Virus, Wheat Streak Mosaic Virus, Wheat Spindle Streak Virus, American Wheat Striate Virus, Claviceps purpurea, Tilletia tritici, Tilletia laevis, Tilletia indica, Pythium gramicola, High Plains Virus, European wheat striate virus; Sunflower: Broomrape, Plasmophora halstedii, Sclerotinia sclerotiorum, Aster Yellows, Septoria helianthi, Phomopsis helianthi, Alternaria helianthi, Alternaria zinniae, Botrytis cinerea, Phoma macdonaldii, Macrophomina phaseolina, Erysiphe cichoracearum, Rhizopus oryzae, Rhizopus arrhizus, Rhizopus stolonifer, Puccinia helianthi, Verticillium dahliae, Erwinia carotovorum pv. carotovora, Cephalosporium acremonium, Phytophthora cryptogea, Albugo tragopogonis; Corn: Fusarium moniliforme var. subglutinans, Erwinia stewartii, Fusarium moniliforme, Gibberella zeae (Fusarium graminearum), Stenocarpella maydi (Diplodia maydis), Pythium irregulare, Pythium debaryanum, Pythium graminicola, Pythium splendens, Pythium ultimum, Pythium aphanidermatum, Aspergillus flavus, Bipolaris maydis O, T (Cochliobolus heterostrophus), Helminthosporium carbonum I, II & III (Cochliobolus carbonum), Exserohilum turcicum I, II & III, Helminthosporium pedicellatum, Physoderma maydis, Phyllosticta maydis, Kabatiella-maydis, Cercospora sorghi, Ustilago maydis, Puccinia sorghi, Puccinia polysora, Macrophomina phaseolina, Penicillium oxalicum, Nigrospora oryzae, Cladosporium herbarum, Curvularia lunata, Curvularia inaequalis, Curvularia pallescens, Clavibacter michiganense subsp. nebraskense, Trichoderma viride, Maize Dwarf Mosaic Virus A & B, Wheat Streak Mosaic Virus, Maize Chlorotic Dwarf Virus, Claviceps sorghi, Pseudonomas avenae, Erwinia chrysanthemi pv. zea, Erwinia carotovora, Corn stunt spiroplasma, Diplodia macrospora, Sclerophthora macrospora, Peronosclerospora sorghi, Peronosclerospora philippinensis, Peronosclerospora maydis, Peronosclerospora sacchari, Sphacelotheca reiliana, Physopella zeae, Cephalosporium maydis, Cephalosporium acremonium, Maize Chlorotic Mottle Virus, High Plains Virus, Maize Mosaic Virus, Maize Rayado Fino Virus, Maize Streak Virus, Maize Stripe Virus, Maize Rough Dwarf Virus; Sorghum: Exserohilum turcicum, Colletotrichum graminicola (Glomerella graminicola), Cercospora sorghi, Gloeocercospora sorghi, Ascochyta sorghina, Pseudomonas syringae p.v. syringae, Xanthomonas campestris p.v. holcicola, Pseudomonas andropogonis, Puccinia purpurea, Macrophomina phaseolina, Perconia circinata, Fusarium moniliforme, Alternaria alternata, Bipolaris sorghicola, Helminthosporium sorghicola, Curvularia lunata, Phoma insidiosa, Pseudomonas avenae (Pseudomonas alboprecipitans), Ramulispora sorghi, Ramulispora sorghicola, Phyllachara sacchari, Sporisorium reilianum (Sphacelotheca reiliana), Sphacelotheca cruenta, Sporisorium sorghi, Sugarcane mosaic H, Maize Dwarf Mosaic Virus A & B, Claviceps sorghi, Rhizoctonia solani, Acremonium strictum, Sclerophthona macrospora, Peronosclerospora sorghi, Peronosclerospora philippinensis, Sclerospora graminicola, Fusarium graminearum, Fusarium oxysporum, Pythium arrhenomanes, Pythium graminicola; Rice: rice brownspot fungus (Cochliobolus miyabeanus), rice blast fungus—Magnaporthe grisea (Pyricularia grisea), Magnaporthe salvinii (Sclerotium oryzae), Xanthomomas oryzae pv. oryzae, Xanthomomas oryzae pv. oryzicola, Rhizoctonia spp. (including but not limited to Rhizoctonia solani, Rhizoctonia oryzae and Rhizoctonia oryzae-sativae), Pseudomonas spp. (including but not limited to Pseudomonas plantarii, Pseudomonas avenae, Pseudomonas glumae, Pseudomonas fuscovaginae, Pseudomonas alboprecipitans, Pseudomonas syringae pv. panici, Pseudomonas syringae pv. syringae, Pseudomonas syringae pv. oryzae and Pseudomonas syringae pv. aptata), Erwinia spp. (including but not limited to Erwinia herbicola, Erwinia amylovaora, Erwinia chrysanthemi and Erwinia carotovora), Achyla spp. (including but not limited to Achyla conspicua and Achyla klebsiana), Pythium spp. (including but not limited to Pythium dissotocum, Pythium irregulare, Pythium arrhenomanes, Pythium myriotylum, Pythium catenulatum, Pythium graminicola and Pythium spinosum), Saprolegnia spp., Dictyuchus spp., Pythiogeton spp., Phytophthora spp., Alternaria padwickii, Cochliobolus miyabeanus, Curvularia spp. (including but not limited to Curvularia lunata, Curvularia affinis, Curvularia clavata, Curvularia eragrostidis, Curvularia fallax, Curvularia geniculata, Curvularia inaequalis, Curvularia intermedia, Curvularia oryzae, Curvularia oryzae-sativae, Curvularia pallescens, Curvularia senegalensis, Curvularia tuberculata, Curvularia uncinata and Curvularia verruculosa), Sarocladium oryzae, Gerlachia oryzae, Fusarium spp. (including but not limited Fusarium graminearum, Fusarium nivale and to different pathovars of Fusarium monoliforme, including pvs. fujikuroi and zeae), Sclerotium rolfsii, Phoma exigua, Mucor fragilis, Trichoderma viride, Rhizopus spp., Cercospora oryzae, Entyloma oryzae, Dreschlera gigantean, Sclerophthora macrospora, Mycovellosiella oryzae, Phomopsis oryzae-sativae, Puccinia graminis, Uromyces coronatus, Cylindrocladium scoparium, Gaeumannomyces graminis pv. graminis, Myrothecium verrucaria, Pyrenochaeta oryzae, Ceratobasidium oryzae-sativae, Microdochium oryzae (Rhynchosporium oryzae), Cercospora janseana, Thanatephorus cucumeris, Ustilaginoidea virens, Neovossia spp. (including but not limited to Neovossia horrida), Tilletia spp., Balansia oryzae-sativae, Phoma spp. (including but not limited to Phoma sorghina, Phoma insidiosa, Phoma glumarum, Phoma glumicola and Phoma oryzina), Nigrospora spp. (including but not limited to Nigrospora oryzae, Nigrospora sphaerica, Nigrospora panici and Nigrospora padwickii), Epiococcum nigrum, Phyllostica spp., Wolkia decolorans, Monascus purpureus, Aspergillus spp., Penicillium spp., Absidia spp., Mucor spp., Chaetomium spp., Dematium spp., Monilia spp., Streptomyces spp., Syncephalastrum spp., Verticillium spp., Nematospora coryli, Nakataea sigmoidea, Cladosporium spp., Bipolaris spp., Coniothyrium spp., Diplodia oryzae, Exserophilum rostratum, Helococera oryzae, Melanomma glumarum, Metashaeria spp., Mycosphaerella spp., Oidium spp., Pestalotia spp., Phaeoseptoria spp., Sphaeropsis spp., Trematosphaerella spp., rice black-streaked dwarf virus, rice dwarf virus, rice gall dwarf virus, barley yellow dwarf virus, rice grassy stunt virus, rice hoja blanca virus, rice necrosis mosaic virus, rice ragged stunt virus, rice stripe virus, rice stripe necrosis virus, rice transitory yellowing virus, rice tungro bacilliform virus, rice tungro spherical virus, rice yellow mottle virus, rice tarsonemid mite virus, Echinochloa hoja blanca virus, Echinochloa ragged stunt virus, rice bunchy stunt virus, rice giallume virus, orange leaf mycoplasma-like organism, yellow dwarf mycoplasma-like organism, Aphelenchoides besseyi, Ditylenchus angustus, Hirschmanniella spp., Criconemella spp., Meloidogyne spp., Heterodera spp., Pratylenchus spp., Hoplolaimus indicus.
  • Nematodes include plant-parasitic nematodes such as root-knot, cyst, and lesion nematodes, including Heterodera and Globodera spp. such as Globodera rostochiensis and Globodera pailida (potato cyst nematodes); Heterodera glycines (soybean cyst nematode); Heterodera schachtii (beet cyst nematode); and Heterodera avenae (cereal cyst nematode).
  • Insect pests include insects selected from the orders Coleoptera, Diptera, Hymenoptera, Lepidoptera, Mallophaga, Homoptera, Hemiptera, Orthoptera, Thysanoptera, Dermaptera, Isoptera, Anoplura, Siphonaptera, Trichoptera, etc., particularly Coleoptera and Lepidoptera. Insect pests of the invention for the major crops include, but are not limited to: Maize: Ostrinia nubilalis, European corn borer; Agrotis ipsilon, black cutworm; Helicoverpa zea, corn earworm; Spodoptera frugiperda, fall armyworm; Diatraea grandiosella, southwestern corn borer; Elasmopalpus lignosellus, lesser cornstalk borer; Diatraea saccharalis, surgarcane borer; Diabrotica virgifera, western corn rootworm; Diabrotica longicornis barberi, northern corn rootworm; Diabrotica undecimpunctata howardi, southern corn rootworm; Melanotus spp., wireworms; Cyclocephala borealis, northern masked chafer (white grub); Cyclocephala immaculata, southern masked chafer (white grub); Popillia japonica, Japanese beetle; Chaetocnema pulicaria, corn flea beetle; Sphenophorus maidis, maize billbug; Rhopalosiphum maidis, corn leaf aphid; Anuraphis maidiradicis, corn root aphid; Blissus leucopterus leucopterus, chinch bug; Melanoplus femurrubrum, redlegged grasshopper; Melanoplus sanguinipes, migratory grasshopper; Hylemya platura, seedcorn maggot; Agromyza parvicornis, corn blot leafminer; Anaphothrips obscrurus, grass thrips; Solenopsis milesta, thief ant; Tetranychus urticae, two-spotted spider mite; Sorghum: Chilo partellus, sorghum borer; Spodoptera frugiperda, fall armyworm; Helicoverpa zea, corn earworm; Elasmopalpus lignosellus, lesser cornstalk borer; Feltia subterranea, granulate cutworm; Phyllophaga crinita, white grub; Eleodes, Conoderus, and Aeolus spp., wireworms; Oulema melanopus, cereal leaf beetle; Chaetocnema pulicaria, corn flea beetle; Sphenophorus maidis, maize billbug; Rhopalosiphum maidis; corn leaf aphid; Sipha flava, yellow sugarcane aphid; Blissus leucopterus leucopterus, chinch bug; Contarinia sorghicola, sorghum midge; Tetranychus cinnabarinus, carmine spider mite; Tetranychus urticae, twospotted spider mite; Wheat: Pseudaletia unipunctata, army worm; Spodoptera frugiperda, fall armyworm; Elasmopalpus lignosellus, lesser cornstalk borer; Agrotis orthogonia, western cutworm; Elasmopalpus lignosellus, lesser cornstalk borer; Oulema melanopus, cereal leaf beetle; Hypera punctata, clover leaf weevil; Diabrotica undecimpunctata howardi, southern corn rootworm; Russian wheat aphid; Schizaphis graminum, greenbug; Macrosiphum avenae, English grain aphid; Melanoplus femurrubrum, redlegged grasshopper; Melanoplus differentialis, differential grasshopper; Melanoplus sanguinipes, migratory grasshopper; Mayetiola destructor, Hessian fly; Sitodiplosis mosellana, wheat midge; Meromyza americana, wheat stem maggot; Hylemya coarctata, wheat bulb fly; Frankliniella fusca, tobacco thrips; Cephus cinctus, wheat stem sawfly; Aceria tulipae, wheat curl mite; Sunflower: Suleima helianthana, sunflower bud moth; Homoeosoma electellum, sunflower moth; zygogramma exclamationis, sunflower beetle; Bothyrus gibbosus, carrot beetle; Neolasioptera murtfeldtiana, sunflower seed midge; Cotton: Heliothis virescens, cotton budworm; Helicoverpa zea, cotton bollworm; Spodoptera exigua, beet armyworm; Pectinophora gossypiella, pink bollworm; Anthonomus grandis grandis, boll weevil; Aphis gossypii, cotton aphid; Pseudatomoscelis seriatus, cotton fleahopper; Trialeurodes abutilonea, banded-winged whitefly; Lygus lineolaris, tarnished plant bug; Melanoplus femurrubrum, redlegged grasshopper; Melanoplus differentialis, differential grasshopper; Thrips tabaci, onion thrips; Franklinkiella fusca, tobacco thrips; Tetranychus cinnabarinus, carmine spider mite; Tetranychus urticae, two-spotted spider mite; Rice: Diatraea saccharalis, sugarcane borer; Spodoptera frugiperda, fall armyworm; Helicoverpa zea, corn earworm; Colaspis brunnea, grape colaspis; Lissorhoptrus oryzophilus, rice water weevil; Sitophilus oryzae, rice weevil; Nephotettix nigropictus, rice leafhopper; Blissus leucopterus leucopterus, chinch bug; Acrosternum hilare, green stink bug; Soybean: Pseudoplusia includens, soybean looper; Anticarsia gemmatalis, velvetbean caterpillar; Plathypena scabra, green cloverworm; Ostrinia nubilalis, European corn borer; Agrotis ipsilon, black cutworm; Spodoptera exigua, beet armyworm; Heliothis virescens, cotton budworm; Helicoverpa zea, cotton bollworm; Epilachna varivestis, Mexican bean beetle; Myzus persicae, green peach aphid; Empoasca fabae, potato leafhopper; Acrosternum hilare, green stink bug; Melanoplus femurrubrum, redlegged grasshopper; Melanoplus differentialis, differential grasshopper; Hylemya platura, seedcorn maggot; Sericothrips variabilis, soybean thrips; Thrips tabaci, onion thrips; Tetranychus turkestani, strawberry spider mite; Tetranychus urticae, two-spotted spider mite; Barley: Ostrinia nubilalis, European corn borer; Agrotis ipsilon, black cutworm; Schizaphis graminum, greenbug; Blissus leucopterus leucopterus, chinch bug; Acrosternum hilare, green stink bug; Euschistus servus, brown stink bug; Delia platura, seedcorn maggot; Mayetiola destructor, Hessian fly; Petrobia latens, brown wheat mite; Oil Seed Rape: Brevicoryne brassicae, cabbage aphid; Phyllotreta cruciferae, Flea beetle; Mamestra configurata, Bertha armyworm; Plutella xylostella, Diamond-back moth; Delia ssp., Root maggots.
  • The nucleic acid sequences of the present invention can be expressed in a host cell such as bacteria, yeast, insect, mammalian, or preferably plant cells. 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 invention. No attempt to describe in detail the various methods known for the expression of proteins in prokaryotes or eukaryotes will be made.
  • The cystatin sequences of the invention are provided in expression cassettes or DNA constructs for expression in the plant of interest. The cassette will include 5′ and 3′ regulatory sequences operably linked to a cystatin sequence of the invention. The cassette may additionally contain at least one additional gene to be cotransformed into the organism. Alternatively, the additional gene(s) can be provided on multiple expression cassettes.
  • Such an expression cassette is provided with a plurality of restriction sites for insertion of the cystatin sequence to be under the transcriptional regulation of the regulatory regions. The expression cassette may additionally contain selectable marker genes.
  • The expression cassette will include in the 5′-3′ direction of transcription, a transcriptional initiation region (i.e., a promoter), translational initiation region, a polynucleotide of the invention, a translational termination region and, optionally, a transcriptional termination region functional in the host organism. The regulatory regions (i.e., promoters, transcriptional regulatory regions, and translational termination regions) and/or the polynucleotide of the invention may be native/analogous to the host cell or to each other. Alternatively, the regulatory regions and/or the polynucleotide of the invention may be heterologous to the host cell or to each other. As used herein, “heterologous” in reference to a sequence is a sequence 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 polynucleotide is from a species different from the species from which the polynucleotide was derived, or, if from the same/analogous species, one or both are substantially modified from their original form and/or genomic locus, or the promoter is not the native promoter for the operably linked polynucleotide.
  • While it may be preferable to express the sequences using heterologous promoters, the native promoter sequences may be used. Such constructs would change expression levels of cystatin in the host cell (i.e., plant or plant cell). Thus, the phenotype of the host cell (i.e., plant or plant cell) is altered.
  • The termination region may be native with the transcriptional initiation region, may be native with the operably linked DNA sequence of interest, or may be derived from another source. Convenient termination regions are available from the Ti-plasmid of A. tumefaciens, such as the octopine synthase and nopaline synthase termination regions. See also Guerineau et al. (1991) Mol. Gen. Genet. 262:141-144; Proudfoot (1991) Cell 64:671-674; Sanfacon et al. (1991) Genes Dev. 5:141-149; Mogen et al. (1990) Plant Cell 2:1261-1272; Munroe et al. (1990) Gene 91:151-158; Ballas et al. (1989) Nucleic Acids Res. 17:7891-7903; and Joshi et al. (1987) Nucleic Acid Res. 15:9627-9639.
  • Where appropriate, the gene(s) may be optimized for increased expression in the transformed plant. That is, the genes can be synthesized using plant-preferred codons for improved expression. Methods are available in the art for synthesizing plant-preferred genes. See, for example, U.S. Pat. Nos. 5,380,831, and 5,436,391, and Murray et al. (1989) Nucleic Acids Res. 17:477-498, herein incorporated by reference.
  • Additional sequence modifications are known to enhance gene expression in a cellular host. These include elimination of sequences encoding spurious polyadenylation signals, exon-intron splice site signals, transposon-like repeats, and other such well-characterized sequences that may be deleterious to gene expression. The G-C content of the sequence may be adjusted to levels average for a given cellular host, as calculated by reference to known genes expressed in the host cell. When possible, the sequence is modified to avoid predicted hairpin secondary mRNA structures.
  • The expression cassettes may additionally contain 5′ leader sequences in the expression cassette construct. Such leader sequences can act to enhance translation. Translation leaders are known in the art and include: picornavirus leaders, for example, EMCV leader (Encephalomyocarditis 5′ noncoding region) (Elroy-Stein et al. (1989) PNAS USA 86:6126-6130); potyvirus leaders, for example, TEV leader (Tobacco Etch Virus) (Allison et al. (1986) Virology 154:9-20); and human immunoglobulin heavy-chain binding protein (BiP), (Macejak et al. (1991) Nature 353:90-94); untranslated leader from the coat protein mRNA of alfalfa mosaic virus (AMV RNA 4) (Jobling et al. (1987) Nature 325:622-625); tobacco mosaic virus leader (TMV) (Gallie et al. (1989) in Molecular Biology of RNA, ed. Cech (Liss, New York), pp. 237-256); and maize chlorotic mottle virus leader (MCMV) (Lommel et al. (1991) Virology 81:382-385). See also, Della-Cioppa et al. (1987) Plant Physiol. 84:965-968. Other methods known to enhance transcription can also be utilized.
  • In preparing the expression cassette, the various DNA fragments may be manipulated, so as to provide for the DNA sequences in the proper orientation and, as appropriate, in the proper reading frame. Toward this end, adapters or linkers may be employed to join the DNA fragments or other manipulations may be involved to provide for convenient restriction sites, removal of superfluous DNA, removal of restriction sites, or the like. For this purpose, in vitro mutagenesis, primer repair, restriction, annealing, resubstitutions, e.g., transitions and transversions, may be involved.
  • Generally, the expression cassette will 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, glyphosate, ammonium, bromoxynil, imidazolinones, and 2,4-dichlorophenoxyacetate (2,4-D). 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; Baim 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 et al. (1991) Antimicrob. Agents Chemother. 35:1591-1595; Kleinschnidt et al. (1988) Biochemistry 27:1094-1104; 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; and WO Publication No. 02/36782. 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 present invention.
  • A number of promoters can be used in the practice of the invention. The promoters can be selected based on the desired outcome. That is, the nucleic acids can be combined with constitutive, tissue-preferred, or other promoters for expression in the host cell of interest. Such constitutive promoters include, for example, the core promoter of the Rsyn7 (WO 99/48338 and U.S. Pat. No. 6,072,050); the core CaMV 35S promoter (Odell et al. (1985) Nature 313:810-812); rice actin (McElroy et al. (1990) Plant Cell 2:163-171); ubiquitin (Christensen et al. (1989) Plant Mol. Biol. 12:619-632 and Christensen et al. (1992) Plant Mol. Biol. 18:675-689); PEMU (Last et al. (1991) Theor. Appl. Genet. 81:581-588); MAS (Velten et al. (1984) EMBO J. 3:2723-2730); ALS promoter (U.S. Pat. No. 5,659,026), and the like. Other constitutive promoters include, for example, those disclosed in U.S. Pat. Nos. 5,608,149; 5,608,144; 5,604,121; 5,569,597; 5,466,785; 5,399,680; 5,268,463; and 5,608,142.
  • Generally, it will be beneficial to express the gene from an inducible promoter, particularly from a pathogen-inducible promoter. Such promoters include those from pathogenesis-related proteins (PR proteins), which are induced following infection by a pathogen; e.g., PR proteins, SAR proteins, beta-1,3-glucanase, chitinase, etc. See, for example, Redolfi et al. (1983) Neth. J. Plant Pathol. 89:245-254; Uknes et al. (1992) Plant Cell 4:645-656; and Van Loon (1985) Plant Mol. Virol. 4:111-116. See also, U.S. application Ser. No. 09/257,583 and WO 99/43819, herein incorporated by reference.
  • Of interest are promoters that are expressed locally at or near the site of pathogen infection. See, for example, Marineau et al. (1987) Plant Mol. Biol. 9:335-342; Matton et al. (1989) Molecular Plant-Microbe Interactions 2:325-331; Somsisch et al. (1986) Proc. Natl. Acad. Sci. USA 83:2427-2430; Somsisch et al. (1988) Mol. Gen. Genet. 2:93-98; and Yang (1996) Proc. Natl. Acad. Sci. USA 93:14972-14977. See also, Chen et al. (1996) Plant J. 10:955-966; Zhang et al. (1994) Proc. Natl. Acad. Sci. USA 91:2507-2511; Warner et al. (1993) Plant J. 3:191-201; Siebertz et al. (1989) Plant Cell 1:961-968; U.S. Pat. No. 5,750,386 (nematode-inducible); and the references cited therein. Of particular interest is the inducible promoter for the maize PRms gene, whose expression is induced by the pathogen Fusarium moniliforme (see, for example, Cordero et al. (1992) Physiol. Mol. Plant. Path. 41:189-200).
  • Additionally, as pathogens find entry into plants through wounds or insect damage, a wound-inducible promoter may be used in the constructions of the invention. Such wound-inducible promoters include potato proteinase inhibitor (pin 11) gene (Ryan (1990) Ann. Rev. Phytopath. 28:425-449; Duan et al. (1996) Nature Biotechnology 14:494-498); wun1 and wun2, U.S. Pat. No. 5,428,148; win1 and win2 (Stanford et al. (1989) Mol. Gen. Genet. 215:200-208); systemin (McGurl et al. (1992) Science 225:1570-1573); WIP1 (Rohmeier et al. (1993) Plant Mol. Biol. 22:783-792; Eckelkamp et al. (1993) FEBS Letters 323:73-76); MPI gene (Corderok et al. (1994) Plant J. 6(2):141-150); and the like, herein incorporated by reference.
  • Chemical-regulated promoters can be used to modulate the expression of a gene in a plant through the application of an exogenous chemical regulator. Depending upon the objective, the promoter may be a chemical-inducible promoter, where application of the chemical induces gene expression, or a chemical-repressible promoter, where application of the chemical represses gene expression. Chemical-inducible promoters are known in the art and include, but are not limited to, the maize ln2-2 promoter, which is activated by benzenesulfonamide herbicide safeners, the maize GST promoter, which is activated by hydrophobic electrophilic compounds that are used as pre-emergent herbicides, and the tobacco PR-1a promoter, which is activated by salicylic acid. Other chemical-regulated promoters of interest include steroid-responsive promoters (see, for example, the glucocorticoid-inducible promoter in Schena et al. (1991) Proc. Natl. Acad. Sci. USA 88:10421-10425 and McNellis et al. (1998) Plant J. 14(2):247-257) and tetracycline-inducible and tetracycline-repressible promoters (see, for example, Gatz et al. (1991) Mol. Gen. Genet. 227:229-237, and U.S. Pat. Nos. 5,814,618 and 5,789,156), herein incorporated by reference.
  • Tissue-preferred promoters can be utilized to target enhanced cystatin expression within a particular plant tissue. Tissue-preferred promoters include those disclosed in Yamamoto et al. (1997) Plant J. 12(2):255-265; Kawamata et al. (1997) Plant Cell Physiol. 38(7):792-803; Hansen et al. (1997) Mol. Gen. Genet. 254(3):337-343; Russell et al. (1997) Transgenic Res. 6(2):157-168; Rinehart et al. (1996) Plant Physiol. 112(3):1331-1341; Van Camp et al. (1996) Plant Physiol. 112(2):525-535; Canevascini et al. (1996) Plant Physiol. 112(2):513-524; Yamamoto et al. (1994) Plant Cell Physiol. 35(5):773-778; Lam (1994) Results Probl. Cell Differ. 20:181-196; Orozco et al. (1993) Plant Mol. Biol. 23(6):1129-1138; Matsuoka et al. (1993) Proc Natl. Acad. Sci. USA 90(20):9586-9590; and Guevara-Garcia et al. (1993) Plant J. 4(3):495-505. Such promoters can be modified, if necessary, for weak expression.
  • Leaf-specific promoters are known in the art. See, for example, Yamamoto et al. (1997) Plant J. 12(2):255-265; Kwon et al. (1994) Plant Physiol. 105:357-67; Yamamoto et al. (1994) Plant Cell Physiol. 35(5):773-778; Gotor et al. (1993) Plant J. 3:509-18; Orozco et al. (1993) Plant Mol. Biol. 23(6):1129-1138; and Matsuoka et al. (1993) Proc. Natl. Acad. Sci. USA 90(20):9586-9590.
  • Just as expression of an antipathogenic polypeptide of the invention may be targeted to specific plant tissues or cell types through the use of appropriate promoters, it may also be targeted to different locations within the cell through the use of targeting information or “targeting labels”. Unlike the promoter, which acts at the transcriptional level, such targeting information is part of the initial translation product. Depending on the mode of infection of the pathogen or the metabolic function of the tissue or cell type, the location of the protein in different compartments of the cell may make it more efficacious against a given pathogen or make it interfere less with the functions of the cell. For example, one may produce a protein preceded by a signal peptide, which directs the translation product into the endoplasmic reticulum, by including in the construct (i.e. expression cassette) sequences encoding a signal peptide (such sequences may also be called the “signal sequence”). The signal sequence used could be, for example, one associated with the gene encoding the polypeptide, or it may be taken from another gene.
  • There are many signal peptides described in the literature, and they are largely interchangeable (Raikhel N, Chrispeels M J (2000) Protein sorting and vesicle traffic. In B Buchanan, W Gruissem, R Jones, eds, Biochemistry and Molecular Biology of Plants. American Society of Plant Physiologists, Rockville, Md., pp 160-201, herein incorporated by reference). The addition of a signal peptide will result in the translation product entering the endoplasmic reticulum (in the process of which the signal peptide itself is removed from the polypeptide), but the final intracellular location of the protein depends on other factors, which may be manipulated to result in localization most appropriate for the pathogen and cell type. The default pathway, that is, the pathway taken by the polypeptide if no other targeting labels are included, results in secretion of the polypeptide across the cell membrane (Raikhel and Chrispeels, supra) into the apoplast. The apoplast is the region outside the plasma membrane system and includes cell walls, intercellular spaces, and the xylem vessels that form a continuous, permeable system through which water and solutes may move.
  • The method of transformation/transfection is not critical to the instant invention; various methods of transformation or transfection are currently available. As newer methods are available to transform crops or other host cells they may be directly applied. Accordingly, a wide variety of methods have been developed to insert a DNA sequence into the genome of a host cell to obtain the transcription and/or translation of the sequence to effect phenotypic changes in the organism. Thus, any method, which provides for effective transformation/transfection may be employed.
  • Transformation protocols as well as protocols for introducing nucleotide sequences into plants may vary depending on the type of plant or plant cell, i.e., monocot or dicot, targeted for transformation. Suitable methods of introducing nucleotide sequences into plant cells and subsequent insertion into the plant genome include microinjection (Crossway et al. (1986) Biotechniques 4:320-334), electroporation (Riggs et al. (1986) Proc. Natl. Acad. Sci. USA 83:5602-5606), Agrobacterium-mediated transformation (Townsend et al., U.S. Pat. No. 5,563,055 and Zhao et al., U.S. Pat. No. 5,981,840), direct gene transfer (Paszkowski et al. (1984) EMBO J. 3:2717-2722), and ballistic particle acceleration (see, for example, Sanford et al., U.S. Pat. No. 4,945,050; Tomes et al. (1995) “Direct DNA Transfer into Intact Plant Cells via Microprojectile Bombardment,” in Plant Cell, Tissue, and Organ Culture: Fundamental Methods, ed. Gamborg and Phillips (Springer-Verlag, Berlin); and McCabe et al. (1988) Biotechnology 6:923-926). Also see 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); McCabe et al. (1988) Bio/Technology 6:923-926 (soybean); Finer and McMullen (1991) In vitro Cell Dev. Biol. 27P:175-182 (soybean); Singh et al. (1998) Theor. Appl. Genet. 96:319-324 (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); Tomes, U.S. Pat. No. 5,240,855; Buising et al., U.S. Pat. Nos. 5,322,783 and 5,324,646; Klein et al. (1988) Plant Physiol. 91:440-444 (maize); Fromm et al. (1990) Biotechnology 8:833-839 (maize); Hooykaas-Van Slogteren et al. (1984) Nature (London) 311:763-764; Bytebier 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. Chapman et al. (Longman, New York), pp. 197-209 (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); 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 Biotechnology 14:745-750 (maize via Agrobacterium tumefaciens); all of which are herein incorporated by reference.
  • The cells that have been transformed may be grown into plants in accordance with conventional ways. See, for example, McCormick et al. (1986) Plant Cell Reports 5:81-84. These plants may then be grown, and either pollinated with the same transformed strain or different strains, and the resulting hybrid having constitutive expression of the desired phenotypic characteristic identified. Two or more generations may be grown to ensure that constitutive expression of the desired phenotypic characteristic is stably maintained and inherited and then seeds harvested to ensure constitutive expression of the desired phenotypic characteristic. One of skill will recognize that after the recombinant expression cassette is stably incorporated in transgenic plants and confirmed to be operable, it can be introduced into other plants by sexual crossing. Any of number of standard breeding techniques can be used, depending upon the species to be crossed.
  • In vegetatively propagated crops, mature transgenic plants can be propagated by the taking of cuttings or by tissue culture techniques to produce multiple identical plants. Selection of desirable transgenics is made and new varieties are obtained and propagated vegetatively for commercial use. In seed propagated crops, mature transgenic plants can be self-crossed to produce a homozygous inbred plant. The inbred plant produces seed containing the newly introduced heterologous nucleic acid. These seeds can be grown to produce plans that would produce the selected phenotype.
  • Parts obtained from the regenerated plant, such as flowers, seeds, leaves, branches, fruit, and the like are included in the invention, provided that these parts comprise cells comprising the isolated nucleic acid of the present invention. Progeny and variants, and mutants of the regenerated plants are also included within the scope of the invention, provided that these parts comprise the introduced nucleic acid sequences.
  • A preferred embodiment is a transgenic plant that is homozygous for the added heterologous nucleic acid; i.e., a transgenic plant that contains two added nucleic acid sequences, one gene at the same locus on each chromosome of a chromosome pair. A homozygous transgenic plant can be obtained by sexually mating (selfing) a heterozygous transgenic plant that contains a single added heterologous nucleic acid, germinating some of the seed produced and analyzing the resulting plants produced for altered expression of a polynucleotide of the present invention relative to a control plant (i.e., native, non-transgenic). Backcrossing to a parental plant and out-crossing with a non-transgenic plant are also contemplated.
  • The present invention may be used for transformation of any plant species, including, but not limited to, monocots and dicots. Examples of plants of interest include, but are not limited to, corn (Zea mays), Brassica sp. (e.g., B. napus, B. rapa, B. juncea), particularly those Brassica species useful as sources of seed oil, alfalfa (Medicago sativa), rice (Oryza sativa), rye (Secale cereale), sorghum (Sorghum bicolor, Sorghum vulgare), millet (e.g., pearl millet (Pennisetum glaucum), proso millet (Panicum miliaceum), foxtail millet (Setaria italica), finger millet (Eleusine coracana)), sunflower (Helianthus annuus), safflower (Carthamus tinctorius), wheat (Triticum aestivum), soybean (Glycine max), tobacco (Nicotiana tabacum), potato (Solanum tuberosum), peanuts (Arachis hypogaea), cotton (Gossypium barbadense, Gossypium hirsutum), sweet potato (Ipomoea batatus), cassaya (Manihot esculenta), coffee (Coffea 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), papaya (Carica papaya), cashew (Anacardium occidentale), macadamia (Macadamia integrifolia), almond (Prunus amygdalus), sugar beets (Beta vulgaris), sugarcane (Saccharum spp.), oats, barley, vegetables, ornamentals, and conifers.
  • Vegetables include tomatoes (Lycopersicon esculentum), lettuce (e.g., Lactuca sativa), green beans (Phaseolus vulgaris), lima beans (Phaseolus limensis), peas (Lathyrus spp.), and members of the genus Cucumis such as cucumber (C. sativus), cantaloupe (C. cantalupensis), and musk melon (C. melo). Ornamentals include azalea (Rhododendron spp.), hydrangea (Macrophylla hydrangea), hibiscus (Hibiscus rosasanensis), roses (Rosa spp.), tulips (Tulipa spp.), daffodils (Narcissus spp.), petunias (Petunia hybrida), carnation (Dianthus caryophyllus), poinsettia (Euphorbia pulcherrima), and chrysanthemum. Conifers that may be employed in practicing the present invention include, for example, pines such as loblolly pine (Pinus taeda), slash pine (Pinus elliotii), ponderosa pine (Pinus ponderosa), lodgepole pine (Pinus contorta), and Monterey pine (Pinus radiata); Douglas-fir (Pseudotsuga menziesii); Western hemlock (Tsuga canadensis); Sitka spruce (Picea glauca); redwood (Sequoia sempervirens); true firs such as silver fir (Abies amabilis) and balsam fir (Abies balsamea); and cedars such as Western red cedar (Thuja plicata) and Alaska yellow-cedar (Chamaecyparis nootkatensis). Preferably, plants of the present invention are crop plants (for example, corn, alfalfa, sunflower, Brassica, soybean, cotton, safflower, peanut, sorghum, wheat, millet, tobacco, etc.), more preferably corn and soybean plants, yet more preferably corn plants.
  • Prokaryotic cells may be used as hosts for expression. Prokaryotes most frequently are represented by various strains of E. coli, 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 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). Examples of selection markers for E. coli include, for example, genes specifying resistance to ampicillin, tetracycline, or chloramphenicol.
  • The vector is selected to allow introduction 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 invention are available using Bacillus sp. and Salmonella (Palva et al. (1983) Gene 22:229-235 and Mosbach et al. (1983) Nature 302:543-545).
  • 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, a polynucleotide of the present invention 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 invention. Such antimicrobial proteins can be used for any application including coating surfaces to target microbes. In this manner, target microbes include human pathogens or microorganisms.
  • Synthesis of heterologous nucleotide sequences in yeast is well known. Sherman, F., et al. (1982) Methods in Yeast Genetics, Cold Spring Harbor Laboratory is a well recognized work describing the various methods available to produce a 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 invention, once expressed, can be isolated from yeast by lysing the cells and applying standard protein isolation techniques to the lysates. The monitoring of the purification process can be accomplished by using Western blot techniques, radioimmunoassay, or other standard immunoassay techniques.
  • The sequences of the present invention can also be ligated to various expression vectors for use in transfecting cell cultures of, for instance, mammalian, insect, or plant origin. Illustrative cell cultures useful for the production of the peptides are mammalian cells. 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 invention are available, for instance, from the American Type Culture Collection.
  • Appropriate vectors for expressing proteins of the present invention 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, Schneider, J. Embryol. Exp. Morphol. 27:353-365 (1987)).
  • As with yeast, when higher animal or plant host cells are employed, polyadenylation 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. 45:773-781). 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. See, Saveria-Campo, M., (1985) Bovine Papilloma Virus DNA a Eukaryotic Cloning Vector in DNA Cloning Vol. II a Practical Approach, D. M. Glover, Ed., IRL Press, Arlington, Va. pp. 213-238.
  • Animal and lower eukaryotic (e.g., yeast) host cells are competent or rendered competent for transfection by various means. There are several well-known methods of introducing DNA into animal cells. These include: calcium phosphate precipitation, fusion of the recipient cells with bacterial protoplasts containing the DNA, treatment of the recipient cells with liposomes containing the DNA, DEAE dextrin, electroporation, biolistics, and micro-injection of the DNA directly into the cells. The transfected cells are cultured by means well known in the art. See, Kuchler, R. J. (1997) Biochemical Methods in Cell Culture and Virology, Dowden, Hutchinson and Ross, Inc.
  • It is recognized that with these nucleotide sequences, antisense constructions, complementary to at least a portion of the messenger RNA (mRNA) for the cystatin sequences can be constructed. Antisense nucleotides are constructed to hybridize with the corresponding mRNA. Modifications of the antisense sequences may be made as long as the sequences hybridize to and interfere with expression of the corresponding mRNA. In this manner, antisense constructions having 70%, preferably 80%, more preferably 85% sequence identity to the corresponding antisensed sequences may be used. 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, or greater may be used.
  • The nucleotide sequences of the present invention may also be used in the sense orientation to suppress the expression of endogenous genes in plants. Methods for suppressing gene expression in plants using nucleotide sequences in the sense orientation are known in the art. The methods generally involve transforming plants with a DNA construct comprising a promoter that drives expression in a plant operably linked to at least a portion of a nucleotide sequence that corresponds to the transcript of the endogenous gene. Typically, such a nucleotide sequence has substantial sequence identity to the sequence of the transcript of the endogenous gene, preferably greater than about 65% sequence identity, more preferably greater than about 85% sequence identity, most preferably greater than about 95% sequence identity. See, U.S. Pat. Nos. 5,283,184 and 5,034,323; herein incorporated by reference.
  • The present invention further provides a method for modulating (i.e., increasing or decreasing) the concentration or composition of the polypeptides of the present invention in a plant or part thereof. Increasing or decreasing the concentration and/or the composition of polypeptides in a plant can affect modulation. For example, increasing the ratio of polypeptides of the invention to native polypeptides can affect modulation. The method comprises: introducing a polynucleotide of the present invention into a plant cell with a recombinant expression cassette as described above to obtain a transformed plant cell, culturing the transformed plant cell under appropriate growing conditions, and inducing or repressing expression of a polynucleotide of the present invention in the plant for a time sufficient to modulate the concentration and/or the composition of polypeptides in the plant or plant part.
  • In some embodiments, the content and/or composition of polypeptides of the present invention in a plant may be modulated by altering, in vivo or in vitro, the promoter of the nucleotide sequence to up- or down-regulate expression. For instance, an isolated nucleic acid comprising a promoter sequence is transfected into a plant cell. Subsequently, a plant cell comprising the promoter operably linked to a polynucleotide of the present invention is selected for by means known to those of skill in the art such as, but not limited to, Southern blot, DNA sequencing, or PCR analysis using primers specific to the promoter and to the gene and detecting amplicons produced therefrom. A plant or plant part altered or modified by the foregoing embodiments is grown under plant forming conditions for a time sufficient to modulate the concentration and/or composition of polypeptides of the present invention in the plant. Plant forming conditions are well known in the art and discussed briefly, supra.
  • In general, concentration or composition is increased or decreased by at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% relative to a native control plant, plant part, or cell lacking the aforementioned recombinant expression cassette. Modulation in the present invention may occur during and/or subsequent to growth of the plant to the desired stage of development. Modulating nucleic acid expression temporally and/or in particular tissues can be controlled by employing the appropriate promoter operably linked to a polynucleotide of the present invention in, for example, sense or antisense orientation as discussed in greater detail, supra. Induction of expression of a polynucleotide of the present invention can also be controlled by exogenous administration of an effective amount of inducing compound. Inducible promoters and inducing compounds, which activate expression from these promoters, are well known in the art. In preferred embodiments, the polypeptides of the present invention are modulated in monocots, particularly maize.
  • In certain embodiments the nucleic acid sequences of the present invention can be stacked with any combination of polynucleotide sequences of interest in order to create plants with a desired phenotype. For example, the polynucleotides of the present invention may be stacked with any other polynucleotides of the present invention, such as any combination of the maize, soybean, rice and wheat cystatin sequences presented (SEQ ID NOS: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 43, 45, 47, 49, 51, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, and 75), or with other genes implicated in disease resistance pathways, especially those that have antimicrobial activity like cystatins, such as: (a) other proteinase inhibitors, such as trypsin proteinase inhibitors (Chen et al. (1999) Appl Environ Microb 65(3): 1320-1324.), subtilisin/chymotrypsin inhibitors (Cordero et al. (1994) Plant J 6(2): 141-150), trypsin/alpha-amylase inhibitors (Wen et al. (1992) Plant Mol. Biol. 18(4): 813-814), and Bowman-Birk proteinase inhibitors (Prakash et al. (1996) J Mol Evol 1996; 42 (5): 560-569); (b) small cysteine-rich antimicrobial proteins, such as defensins (Thomma et al. (2002) Planta 261(2): 193-202) or gamma-thionins (Nitti et al. (1995) Eur J Biochem 228(2): 250-6), kistrin-like cysteine-rich proteins (Segura, et al. (1999) Mol Plant Microbe Interact 12: 16-23), cyclotides (Craik et al. (1999) J Mol Biol 294(5):1327-36), and basal layer antifungal peptides (Hueros et al. (1995) Plant Cell 7: 747-757); (c) antimicrobial enzymes, such as endo-1,3-beta-glucanases, chitinases (Simmons (1994) CRC Cr Rev Plant Sci 13: 325-387), RNAses (Hugot et al. (2002) Mol Plant Microbe Interact 15(3): 243-250); (d) other pathogenesis-related proteins, such as PR-1 homologs (Tornero et al. (1997) Mol Plant Microbe Interact 10(5): 624-34), PR-10/ocatin/major latex protein homologs (McGee et al. (2001) Mol Plant Microbe Interact 14(7): 877-86), PR-5/thaumatin/osmotin homologs (Barre et al. (2000) Planta. 211 (6):791-9); (e) and other antimicrobial proteins such as lipid transfer proteins (Park et al. (2002) Plant Mol. Biol. 48(3): 243-54), puroindolines (Krishnamurthy et al. (2001). Mol Plant Microbe Interact 14: 1255-1260), alpha- and beta-thionins (Rodriguez-Palenzuela et al. (1988) Gene 70: 271-281; Van Campenhout et al. (1998) Theor Appl Genet 96: 80-86), maize basic proteins (Duvick et al. (1992) J Biol Chem 267(26): 18814-18820), and small histidine-glycine rich proteins (Park et al. (2000) Plant Mol Biol 44: 187-197) and the like, the disclosures of which are herein incorporated by reference. It is understood that other genes and their products that are not themselves antimicrobial, but which contribute to the disease response by a number mechanisms, could also be employed in conjunction, among them LRR-proteins (Mondragon-Palomino et al. (2002) Genome Res (9): 1305-15) and other R gene analogues, transcription factors such as WRKY (Eulgem et al. (2000) Trends Plant Sci (5): 199-206), multidrug transporters (Diener et al. (2001) Plant Cell 13(7): 1625-38), phenylpropanoid pathway enzymes (Dixon et al. (1996) Gene 179(1):61-71), and polygalacturonase inhibitor proteins (Berger et al. (2000). Phys Mol Plant Pathol. 57 (1): 5-14). Such genes could come from maize or non-maize sources, including non-plant sources.
  • The combinations generated can also include multiple copies of any one of the polynucleotides of interest. The polynucleotides of the present invention can also be stacked with any other 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., U.S. Pat. No. 6,232,529); balanced amino acids (e.g. hordothionins (U.S. Pat. Nos. 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 98/20122); 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 (U.S. application Ser. No. 10/053,410, filed Nov. 7, 2001)); and thioredoxins (U.S. application Ser. No. 10/005,429, filed Dec. 3, 2001), the disclosures of which are herein incorporated by reference.
  • The polynucleotides of the present invention can also be stacked with traits desirable for insect, disease or herbicide resistance (e.g., Bacillus thuringiensis toxic proteins (U.S. Pat. Nos. 5,366,892; 5,747,450; 5,737,514; 5,723,756; 5,593,881; Geiser et al (1986) Gene 48:109); lectins (Van Damme et al. (1994) Plant Mol. Biol. 24:825); fumonisin detoxification genes (U.S. Pat. No. 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 GAT gene)); and traits desirable for processing or process products such as high oil (U.S. Pat. No. 6,232,529); modified oils (e.g., fatty acid desaturase genes (U.S. Pat. No. 5,952,544; WO 94/11516)); modified starches (e.g., ADPG pyrophosphorylases (AGPase), starch synthases (SS), starch branching enzymes (SBE) and starch debranching enzymes (SDBE)); and polymers or bioplastics (U.S. Pat. No. 5,602,321); beta-ketothiolase, polyhydroxybutyrate synthase, and acetoacetyl-CoA reductase (Schubert et al. (1988) J. Bacteriol. 170:5837-5847), which facilitate expression of polyhydroxyalkanoates (PHAs)), the disclosures of which are herein incorporated by reference. One could also combine the polynucleotides of the present invention with polynucleotides providing agronomic traits such as male sterility (see U.S. Pat. No. 5,583,210), stalk strength, flowering time, or transformation technology traits such as cell cycle regulation or gene targeting (see, WO 99/61619; WO 00/17364; WO 99/25821), the disclosures of which are herein incorporated by reference.
  • These stacked combinations can be created by any method including, but not limited to, polynucleotide sequences of interest can be combined at any time and in any order. For example, a transgenic plant comprising one or more desired traits can be used as the target to introduce further traits by subsequent transformation. The traits can be introduced simultaneously in a co-transformation protocol with the polynucleotides of interest provided by any combination of transformation cassettes. For example, if two sequences will be introduced, the two sequences can be contained in separate transformation cassettes (trans) or contained on the same transformation cassette (cis). Expression of the sequences can be driven by the same promoter or by different promoters. In certain cases, it may be desirable to introduce a transformation cassette that will suppress the expression of the polynucleotide of interest. This may be combined with any combination of other suppression cassettes or overexpression cassettes to generate the desired combination of traits in the plant.
  • The present invention provides a method of genotyping a plant comprising a polynucleotide of the present invention. Optionally, the plant is a monocot, such as maize or sorghum. Genotyping provides a means of distinguishing homologs of a chromosome pair and can be used to differentiate segregants in a plant population. Molecular marker methods can be used for phylogenetic studies, characterizing genetic relationships among crop varieties, identifying crosses or somatic hybrids, localizing chromosomal segments affecting monogenic traits, map based cloning, and the study of quantitative inheritance. See, e.g., Plant Molecular Biology: A Laboratory Manual, Chapter 7, Clark, Ed., Springer-Verlag, Berlin (1997). For molecular marker methods, see generally, The DNA Revolution by Andrew H. Paterson 1996 (Chapter 2) in: Genome Mapping in plants (Ed., Andrew H. Paterson) by Academic Press/R.G. Lands Company, Austin, Tex., pp. 7-21.
  • The particular method of genotyping in the present invention may employ any number of molecular marker analytic techniques such as, but not limited to, restriction fragment length polymorphisms (RFLPs). RFLPs are the product of allelic differences between DNA restriction fragments resulting from nucleotide sequence variability. As is well known to those of skill in the art, RFLPs are typically detected by extraction of genomic DNA and digestion with a restriction enzyme. Generally, the resulting fragments are separated according to size and hybridized with a probe; single copy probes are preferred. Restriction fragments from homologous chromosomes are revealed. Differences in fragment size among alleles represent an RFLP. Thus, the present invention further provides a means to follow segregation of a gene or nucleic acid of the present invention as well as chromosomal sequences genetically linked to these genes or nucleic acids using such techniques as RFLP analysis. Linked chromosomal sequences are within 50 centiMorgans (cM), often within 40 or 30 cM, preferably within 20 or 10 cM, more preferably within 5, 3, 2, or 1 cM of a gene of the present invention.
  • In the present invention, the nucleic acid probes employed for molecular marker mapping of plant nuclear genomes hybridize, under selective hybridization conditions, to a gene encoding a polynucleotide of the present invention. In preferred embodiments, the probes are selected from polynucleotides of the present invention. Typically, these probes are cDNA probes or restriction enzyme treated (e.g., PST I) genomic clones. The length of the probes is typically at least 15 bases in length, more preferably at least 20, 25, 30, 35, 40, or 50 bases in length. Generally, however, the probes are less than about 1 kilobase in length. Preferably, the probes are single copy probes that hybridize to a unique locus in a haploid chromosome compliment. Some exemplary restriction enzymes employed in RFLP mapping are EcoRI, EcoRV, and SstI. As used herein the term “restriction enzyme” includes reference to a composition that recognizes and, alone or in conjunction with another composition, cleaves at a specific nucleotide sequence.
  • The method of detecting an RFLP comprises the steps of (a) digesting genomic DNA of a plant with a restriction enzyme; (b) hybridizing a nucleic acid probe, under selective hybridization conditions, to a sequence of a polynucleotide of the present invention of the genomic DNA; (c) detecting therefrom a RFLP. Other methods of differentiating polymorphic (allelic) variants of polynucleotides of the present invention can be had by utilizing molecular marker techniques well known to those of skill in the art including such techniques as: 1) single stranded conformation analysis (SSCA); 2) denaturing gradient gel electrophoresis (DGGE); 3) RNase protection assays; 4) allele-specific oligonucleotides (ASOs); 5) the use of proteins which recognize nucleotide mismatches, such as the E. coli mutS protein; and 6) allele-specific PCR. Other approaches based on the detection of mismatches between the two complementary DNA strands include clamped denaturing gel electrophoresis (CDGE); heteroduplex analysis (HA); and chemical mismatch cleavage (CMC). Thus, the present invention further provides a method of genotyping comprising the steps of contacting, under stringent hybridization conditions, a sample suspected of comprising a polynucleotide of the present invention with a nucleic acid probe. Generally, the sample is a plant sample, preferably, a sample suspected of comprising a maize polynucleotide of the present invention (e.g., gene, mRNA). The nucleic acid probe selectively hybridizes, under stringent conditions, to a subsequence of a polynucleotide of the present invention comprising a polymorphic marker. Selective hybridization of the nucleic acid probe to the polymorphic marker nucleic acid sequence yields a hybridization complex. Detection of the hybridization complex indicates the presence of that polymorphic marker in the sample. In preferred embodiments, the nucleic acid probe comprises a polynucleotide of the present invention.
  • The use of the term “nucleotide constructs” herein is not intended to limit the present invention to nucleotide constructs comprising DNA. Those of ordinary skill in the art will recognize that nucleotide constructs, particularly polynucleotides and oligonucleotides, comprised of ribonucleotides and combinations of ribonucleotides and deoxyribonucleotides may also be employed in the methods disclosed herein. Thus, the nucleotide constructs of the present invention encompass all nucleotide constructs that can be employed in the methods of the present invention for transforming plants including, but not limited to, those comprised of deoxyribonucleotides, ribonucleotides, and combinations thereof. Such deoxyribonucleotides and ribonucleotides include both naturally occurring molecules and synthetic analogues. The nucleotide constructs of the invention also encompass all forms of nucleotide constructs including, but not limited to, single-stranded forms, double-stranded forms, hairpins, stem-and-loop structures, and the like.
  • Furthermore, it is recognized that the methods of the invention may employ a nucleotide construct that is capable of directing, in a transformed plant, the expression of at least one protein, or at least one RNA, such as, for example, an antisense RNA that is complementary to at least a portion of an mRNA. Typically such a nucleotide construct is comprised of a coding sequence for a protein or an RNA operably linked to 5′ and 3′ transcriptional regulatory regions. Alternatively, it is also recognized that the methods of the invention may employ a nucleotide construct that is not capable of directing, in a transformed plant, the expression of a protein or an RNA.
  • In addition, it is recognized that methods of the present invention do not depend on the incorporation of the entire nucleotide construct into the genome, only that the plant or cell thereof is altered as a result of the introduction of the nucleotide construct into a cell. In one embodiment of the invention, the genome may be altered following the introduction of the nucleotide construct into a cell. For example, the nucleotide construct, or any part thereof, may incorporate into the genome of the plant. Alterations to the genome of the present invention include, but are not limited to, additions, deletions, and substitutions of nucleotides in the genome. While the methods of the present invention do not depend on additions, deletions, or substitutions of any particular number of nucleotides, it is recognized that such additions, deletions, or substitutions comprise at least one nucleotide.
  • The nucleotide constructs of the invention also encompass nucleotide constructs that may be employed in methods for altering or mutating a genomic nucleotide sequence in an organism, including, but not limited to, chimeric vectors, chimeric mutational vectors, chimeric repair vectors, mixed-duplex oligonucleotides, self-complementary chimeric oligonucleotides, and recombinogenic oligonucleobases. Such nucleotide constructs and methods of use, such as, for example, chimeraplasty, are known in the art. Chimeraplasty involves the use of such nucleotide constructs to introduce site-specific changes into the sequence of genomic DNA within an organism. See, U.S. Pat. Nos. 5,565,350; 5,731,181; 5,756,325; 5,760,012; 5,795,972; and 5,871,984; all of which are herein incorporated by reference. See also, WO 98/49350, WO 99/07865, WO 99/25821, and Beetham et al. (1999) Proc. Natl. Acad. Sci. USA 96:8774-8778; herein incorporated by reference.
  • The methods of the invention can be used with other methods available in the art for enhancing disease resistance in plants. Similarly, the antimicrobial compositions described herein may be used alone or in combination with other nucleotide sequences, polypeptides, or agents to protect against plant diseases and pathogens. Although any one of a variety of second nucleotide sequences may be utilized, specific embodiments of the invention encompass those second nucleotide sequences that, when expressed in a plant, help to increase the resistance of a plant to pathogens.
  • Proteins, peptides, and lysozymes that naturally occur in insects (Jaynes et al. (1987) Bioassays 6:263-270), plants (Broekaert et al. (1997) Critical Reviews in Plant Sciences 16:297-323), animals (Vunnam et al. (1997) J. Peptide Res. 49:59-66), and humans (Mitra and Zang (1994) Plant Physiol. 106:977-981; Nakajima et al. (1997) Plant Cell Reports 16:674-679) are also a potential source of plant disease resistance (Ko, K. (2000) www.scisoc.org/feature/BioTechnology/antimicrobial.html). Examples of such plant resistance-conferring sequences include those encoding sunflower rhoGTPase-Activating Protein (rhoGAP), lipoxygenase (LOX), Alcohol Dehydrogenase (ADH), and Sclerotinia-Inducible Protein-1 (SCIP-1) described in U.S. application Ser. No. 09/714,767, herein incorporated by reference. These nucleotide sequences enhance plant disease resistance through the modulation of development, developmental pathways, and the plant pathogen defense system. Other plant defense proteins include those described in WO 99/43823 and WO 99/43821, all of which are herein incorporated by reference. It is recognized that such second nucleotide sequences may be used in either the sense or antisense orientation depending on the desired outcome.
  • In another embodiment, the cystatins comprise isolated polypeptides of the invention. The cystatins of the invention find use in the decontamination of plant pathogens during the processing of grain for animal or human food consumption; during the processing of feedstuffs, and during the processing of plant material for silage. In this embodiment, the cystatins of the invention, are presented to grain, plant material for silage, or a contaminated food crop, or during an appropriate stage of the processing procedure, in amounts effective for anti-microbial activity. The compositions can be applied to the environment of a plant pathogen by, for example, spraying, atomizing, dusting, scattering, coating or pouring, introducing into or on the soil, introducing into irrigation water, by seed treatment, or dusting at the time when the plant pathogen has begun to appear or before the appearance of pests as a protective measure. It is recognized that any means to bring the defensive agent polypeptides in contact with the plant pathogen can be used in the practice of the invention.
  • Additionally, the compositions can be used in formulations used for their antimicrobial activities. Methods are provided for controlling plant pathogens comprising applying a decontaminating amount of a polypeptide or composition of the invention to the environment of the plant pathogen. The polypeptides of the invention can be formulated with an acceptable carrier into a composition(s) that is, for example, a suspension, a solution, an emulsion, a dusting powder, a dispersible granule, a wettable powder, an emulsifiable concentrate, an aerosol, an impregnated granule, an adjuvant, a coatable paste, and also encapsulations in, for example, polymer substances.
  • Such compositions disclosed above may be obtained by the addition of a surface-active agent, an inert carrier, a preservative, a humectant, a feeding stimulant, an attractant, an encapsulating agent, a binder, an emulsifier, a dye, a UV protectant, a buffer, a flow agent or fertilizers, micronutrient donors or other preparations that influence plant growth. One or more agrochemicals including, but not limited to, herbicides, insecticides, fungicides, bacteriocides, nematocides, molluscicides, acaracides, plant growth regulators, harvest aids, and fertilizers, can be combined with carriers, surfactants, or adjuvants customarily employed in the art of formulation or other components to facilitate product handling and application for particular target pathogens. Suitable carriers and adjuvants can be solid or liquid and correspond to the substances ordinarily employed in formulation technology, e.g., natural or regenerated mineral substances, solvents, dispersants, wetting agents, tackifiers, binders, or fertilizers. The active ingredients of the present invention are normally applied in the form of compositions and can be applied to the crop area or plant to be treated, simultaneously or in succession, with other compounds. In some embodiments, methods of applying an active ingredient of the present invention or an agrochemical composition of the present invention (which contains at least one of the proteins of the present invention) are foliar application, seed coating, and soil application.
  • Suitable surface-active agents include, but are not limited to, anionic compounds such as a carboxylate of, for example, a metal; a carboxylate of a long chain fatty acid; an N-acylsarcosinate; mono or di-esters of phosphoric acid with fatty alcohol ethoxylates or salts of such esters; fatty alcohol sulfates such as sodium dodecyl sulfate, sodium octadecyl sulfate, or sodium cetyl sulfate; ethoxylated fatty alcohol sulfates; ethoxylated alkylphenol sulfates; lignin sulfonates; petroleum sulfonates; alkyl aryl sulfonates such as alkyl-benzene sulfonates or lower alkylnaphtalene sulfonates, e.g., butyl-naphthalene sulfonate; salts of sulfonated naphthalene-formaldehyde condensates; salts of sulfonated phenol-formaldehyde condensates; more complex sulfonates such as the amide sulfonates, e.g., the sulfonated condensation product of oleic acid and N-methyl taurine; or the dialkyl sulfosuccinates, e.g., the sodium sulfonate or dioctyl succinate. Non-ionic agents include condensation products of fatty acid esters, fatty alcohols, fatty acid amides or fatty-alkyl- or alkenyl-substituted phenols with ethylene oxide, fatty esters of polyhydric alcohol ethers, e.g., sorbitan fatty acid esters, condensation products of such esters with ethylene oxide, e.g. polyoxyethylene sorbitar fatty acid esters, block copolymers of ethylene oxide and propylene oxide, acetylenic glycols such as 2,4,7,9-tetraethyl-5-decyn-4,7-diol, or ethoxylated acetylenic glycols. Examples of a cationic surface-active agent include, for instance, an aliphatic mono-, di-, or polyamine such as an acetate, naphthenate, or oleate; or oxygen-containing amine such as an amine oxide of polyoxyethylene alkylamine; an amide-linked amine prepared by the condensation of a carboxylic acid with a di- or polyamine; or a quaternary ammonium salt.
  • Examples of inert materials include, but are not limited to, inorganic minerals such as kaolin, phyllosilicates, carbonates, sulfates, phosphates, or botanical materials such as cork, powdered corncobs, peanut hulls, rice hulls, and walnut shells.
  • The compositions of the present invention can be in a suitable form for direct application or as concentrate of primary composition, which requires dilution with a suitable quantity of water or other diluent before application. The decontaminating concentration will vary depending upon the nature of the particular formulation, specifically, whether it is a concentrate or to be used directly.
  • In a further embodiment, the compositions, as well as the polypeptides of the present invention can be treated prior to formulation to prolong their activity when applied to the environment of a plant pathogen as long as the pretreatment is not deleterious to the activity. Such treatment can be by chemical and/or physical means as long as the treatment does not deleteriously affect the properties of the composition(s). Examples of chemical reagents include, but are not limited to, halogenating agents; aldehydes such as formaldehyde and glutaraldehyde; anti-infectives, such as zephiran chloride; alcohols, such as isopropanol and ethanol; and histological fixatives, such as Bouin's fixative and Helly's fixative (see, for example, Humason (1967) Animal Tissue Techniques (W.H. Freeman and Co.)).
  • In an embodiment of the invention, the compositions of the invention comprise a microbe having stably integrated the nucleotide sequence of a defensive agent. The resulting microbes can be processed and used as a microbial spray. Any suitable microorganism can be used for this purpose. See, for example, Gaertner et al. (1993) in Advanced Engineered Pesticides, Kim (Ed.). In one embodiment, the nucleotide sequences of the invention are introduced into microorganisms that multiply on plants (epiphytes) to deliver the cystatins to potential target crops. Epiphytes can be, for example, gram-positive or gram-negative bacteria.
  • It is further recognized that whole, i.e., unlysed, cells of the transformed microorganism can be treated with reagents that prolong the activity of the polypeptide produced in the microorganism when the microorganism is applied to the environment of a target plant. A secretion signal sequence may be used in combination with the gene of interest such that the resulting enzyme is secreted outside the microorganism for presentation to the target plant.
  • In this manner, a gene encoding a defensive agent of the invention may be introduced via a suitable vector into a microbial host, and said transformed host applied to the environment, plants, or animals. Microorganism hosts that are known to occupy the “phytosphere” (phylloplane, phyllosphere, rhizosphere, and/or rhizoplane) of one or more crops of interest may be selected for transformation. These microorganisms are selected so as to be capable of successfully competing in the particular environment with the wild-type microorganisms, to provide for stable maintenance and expression of the gene expressing the detoxifying polypeptide, and for improved protection of the enzymes of the invention from environmental degradation and inactivation.
  • Such microorganisms include bacteria, algae, and fungi. Of particular interest are microorganisms, such as bacteria, e.g., Pseudomonas, Erwinia, Serratia, Klebsiella, Xanthomonas, Streptomyces, Rhizobium, Rhodopseudomonas, Methylius, Agrobacterium, Acetobacter, Lactobacillus, Arthrobacter, Azotobacter, Leuconostoc, and Alcaligenes; fungi, particularly yeast, e.g., Saccharomyces, Pichia, Cryptococcus, Kluyveromyces, Sporobolomyces, Rhodotorula, Aureobasidium, and Gliocladium. Of particular interest are such phytosphere bacterial species as Pseudomonas syringae, Pseudomonas fluorescens, Serratia marcescens, Acetobacter xylinum, Agrobacteria, Rhodopseudomonas spheroides, Xanthomonas campestris, Rhizobium melioti, Alcaligenes entrophus, Clavibacter xyli, and Azotobacter vinlandii; and phytosphere yeast species such as Rhodotorula rubra, R. glutinis, R. marina, R. aurantiaca, Cryptococcus albidus, C. diffluens, C. laurentii, Saccharomyces rosei, S. pretoriensis, S. cerevisiae, Sporobolomyces rosues, S. odorus, Kluyveromyces veronae, and Aureobasidium pullulans.
  • Illustrative prokaryotes, both Gram-negative and -positive, include Enterobacteriaceae, such as Escherichia, Erwinia, Shigella, Salmonella, and Proteus; Bacillaceae; Rhizobiaceae, such as Rhizobium; Spirillaceae, such as photobacterium, Zymomonas, Serratia, Aeromonas, Vibrio, Desulfovibrio, Spirillum; Lactobacillaceae; Pseudomonadaceae, such as Pseudomonas and Acetobacter; Azotobacteraceae; and Nitrobacteraceae. Among eukaryotes are fungi, such as Phycomycetes and Ascomycetes, which includes yeast, such as Saccharomyces and Schizosaccharomyces; and Basidiomycetes yeast, such as Rhodotorula, Aureobasidium, Sporobolomyces, and the like.
  • In an embodiment of the invention, the cystatins of the invention may be used as a pharmaceutical compound for treatment of fungal and microbial pathogens in humans and other animals. Diseases and disorders caused by fungal and microbial pathogens include but are not limited to fungal meningoencephalitis, superficial fungal infections, ringworm, Athlete's foot, histoplasmosis, candidiasis, thrush, coccidioidoma, pulmonary cryptococcus, trichosporonosis, piedra, tinea nigra, fungal keratitis, onychomycosis, tinea capitis, chromomycosis, aspergillosis, endobronchial pulmonary aspergillosis, mucormycosis, chromoblastomycosis, dermatophytosis, tinea, fusariosis, pityriasis, mycetoma, pseudallescheriasis, and sporotrichosis.
  • The compositions of the invention may be used as pharmaceutical compounds to provide treatment for diseases and disorders associated with, but not limited to, the following fungal pathogens: Histoplasma capsulatum, Candida spp. (C. albicans, C. tropicalis, C. parapsilosis, C. guilliermondii, C. glabratal Torulopsis glabrata, C. krusei, C. lusitaniae), Aspergillus fumigatus, A. flavus, A. niger, Rhizopus spp., Rhizomucor spp., Cunninghamella spp., Apophysomyces spp., Saksenaee spp., Mucor spp., and Absidia spp. Efficacy of the compositions of the invention as anti-fungal treatments may be determined through anti-fungal assays known to one of skill in the art.
  • The cystatins may be administered to a patient through numerous means. Systemic administration can also be by transmucosal or transdermal means. For transmucosal or transdermal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives. Transmucosal administration can be accomplished through the use of nasal sprays or suppositories. For transdermal administration, the active compounds are formulated into ointments, salves, gels, or creams as generally known in the art. The compounds can also be prepared in the form of suppositories (e.g., with conventional suppository bases such as cocoa butter and other glycerides) or retention enemas for rectal delivery.
  • In one embodiment, the active compounds are prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art. The materials can also be obtained commercially from Alza Corporation and Nova Pharmaceuticals, Inc. Liposomal suspensions (including liposomes targeted to infected cells with monoclonal antibodies to viral antigens) can also be used as pharmaceutically acceptable carriers. These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Pat. No. 4,522,811.
  • It is especially advantageous to formulate oral or parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subject to be treated with each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. Depending on the type and severity of the disease, about 1 μg/kg to about 15 mg/kg (e.g., 0.1 to 20 mg/kg) of antibody is an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion. A typical daily dosage might range from about 1 μg/kg to about 100 mg/kg or more, depending on the factors mentioned above. For repeated administrations over several days or longer, depending on the condition, the treatment is sustained until a desired suppression of disease symptoms occurs. However, other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques and assays. An exemplary dosing regimen is disclosed in WO 94/04188. The specification for the dosage unit forms of the invention are dictated by and directly dependent on the unique characteristics of the active compound and the particular therapeutic effect to be achieved, and the limitations inherent in the art of compounding such an active compound for the treatment of individuals.
  • “Treatment” is herein defined as the application or administration of a therapeutic agent to a patient, or application or administration of a therapeutic agent to an isolated tissue or cell line from a patient, who has a disease, a symptom of disease or a predisposition toward a disease, with the purpose to cure, heal, alleviate, relieve, alter, remedy, ameliorate, improve or affect the disease, the symptoms of disease or the predisposition toward disease. A “therapeutic agent” includes, but is not limited to, small molecules, peptides, antibodies, ribozymes and antisense oligonucleotides.
  • The cystatins of the invention can be used for any application including coating surfaces to target microbes. In this manner, target microbes include human pathogens or microorganisms. Surfaces that might be coated with the cystatins of the invention include carpets and sterile medical facilities. Polymer bound polypeptides of the invention may be used to coat surfaces. Methods for incorporating compositions with anti-microbial properties into polymers are known in the art. See U.S. Pat. No. 5,847,047 herein incorporated by reference.
  • An isolated polypeptide of the invention can be used as an immunogen to generate antibodies that bind cystatins using standard techniques for polyclonal and monoclonal antibody preparation. The full-length cystatins can be used or, alternatively, the invention provides antigenic peptide fragments of cystatins for use as immunogens. The antigenic peptide of a defensive agent comprises at least 8, preferably 10, 15, 20, or 30 amino acid residues of the amino acid sequence shown in any of SEQ ID NOs: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, and 76, and encompasses an epitope of a cystatin such that an antibody raised against the peptide forms a specific immune complex with the anti-microbial polypeptides. Preferred epitopes encompassed by the antigenic peptide are regions of cystatins that are located on the surface of the protein, e.g., hydrophilic regions.
  • Accordingly, another aspect of the invention pertains to anti-cystatin polyclonal and monoclonal antibodies that bind a cystatin. Polyclonal cystatin-like antibodies can be prepared by immunizing a suitable subject (e.g., rabbit, goat, mouse, or other mammal) with an defensive agent-like immunogen. The anti-cystatin antibody titer in the immunized subject can be monitored over time by standard techniques, such as with an enzyme linked immunosorbent assay (ELISA) using immobilized anti-microbial polypeptides. At an appropriate time after immunization, e.g., when the anti-defensive agent antibody titers are highest, antibody-producing cells can be obtained from the subject and used to prepare monoclonal antibodies by standard techniques, such as the hybridoma technique originally described by Kohler and Milstein (1975) Nature 256:495-497, the human B cell hybridoma technique (Kozbor et al. (1983) Immunol. Today 4:72), the EBV-hybridoma technique (Cole et al. (1985) in Monoclonal Antibodies and Cancer Therapy, ed. Reisfeld and Sell (Alan R. Liss, Inc., New York, N.Y.), pp. 77-96) or trioma techniques. The technology for producing hybridomas is well known (see generally Coligan et al., eds. (1994) Current Protocols in Immunology (John Wiley & Sons, Inc., New York, N.Y.); Galfre et al. (1977) Nature 266:55052; Kenneth (1980) in Monoclonal Antibodies: A New Dimension In Biological Analyses (Plenum Publishing Corp., NY; and Lerner (1981) Yale J. Biol. Med., 54:387-402).
  • Alternatively to preparing monoclonal antibody-secreting hybridomas, a monoclonal anti-cystatin-like antibody can be identified and isolated by screening a recombinant combinatorial immunoglobulin library (e.g., an antibody phage display library) with a cystatin to thereby isolate immunoglobulin library members that bind the defensive agent. Kits for generating and screening phage display libraries are commercially available (e.g., the Pharmacia Recombinant Phage Antibody System, Catalog No. 27-9400-01; and the Stratagene SurfZAP™ Phage Display Kit, Catalog No. 240612). Additionally, examples of methods and reagents particularly amenable for use in generating and screening an antibody display library can be found in, for example, U.S. Pat. No. 5,223,409; PCT Publication Nos. WO 92/18619; WO 91/17271; WO 92/20791; WO 92/15679; 93/01288; WO 92/01047; 92/09690; and 90/02809; Fuchs et al. (1991) Bio/Technology 9:1370-1372; Hay et al. (1992) Hum. Antibod. Hybridomas 3:81-85; Huse et al. (1989) Science 246:1275-1281; Griffiths et al. (1993) EMBO J. 12:725-734. The antibodies can be used to identify homologs of the cystatins of the invention.
  • The following examples are offered by way of illustration and not by way of limitation.
  • The following stock solutions and media were used for transformation and regeneration of soybean roots:
  • Stock Solutions (Per Liter):
    • 10×B-5 Majors: 25.00 g KNO3, 1.34 g (NH4)2 SO4, 2.50 g MgSO4.7H2O, 1.50 g CaCl2.2H2O, 1.31 g NaH2PO4 (anhydrous).
    • 100×B-5 Minors: 1.00 g MnSO4.H2O, 0.30 g H3BO3, 0.20 g ZnSO4.7H2O, 0.075 g KI.
    • 100×B-5 Vitamins with Thiamine: 10.00 g myo-Inositol, 1.00 g Thiamine*HCl, 0.10 g Nicotinic acid, 0.10 g Pyridoxine HCl.
    • 100× Iron Mix: 3.73 g. Na2EDTA, 2.78 g FeSO4.7H2O. Media (per Liter):
    • Minimal A medium: 10.5 g K2HPO4, 4.5 g KH2PO4, 1.0 g (NH4)2SO4, 0.5 g (Na)2C6H5O7.2H2O, 246.5 mg MgSO4.7H2O, 2 g sucrose, 15 g agar.
    • 0 B-5 medium: 0.6 g MES [2-(N-Morpholino) ethane-sulfonic acid (Sigma, M5287), 20 g sucrose, 10 mL 100×B-5 minors, 100 mL 10×B-5 majors, 10 mL 100×B-5 vitamins with Thiamine, 10 mL 100×Iron mix.
    • MXB medium: Murashige and Skoog Basal nutrient salts (M5524, Sigma), 10 mL 100×B-5 Vitamins with thiamine, 30 g sucrose, 3 g gelrite, pH 5.7.
    • SOC medium: 20 g bactotryptone, 5 g yeast extract (Difco), 2 mL 5 M NaCl, 2.5 mL 1 M KCl, 10 mL 1 M MgCl2, 10 mL 2 M glucose, 10 mL 1 M MgSO4.
    EXAMPLE 1 Transformation and Regeneration of Transgenic Plants
  • Immature maize embryos from greenhouse donor plants are bombarded with a plasmid containing the cystatin sequences of the present invention operably linked to a ubiquitin promoter and the selectable marker gene PAT (Wohlleben et al. (1988) Gene 70:25-37), which confers resistance to the herbicide Bialaphos. Alternatively, the selectable marker gene is provided on a separate plasmid. Transformation is performed as follows. Media recipes follow below.
  • Preparation of Target Tissue
  • The ears are husked and surface sterilized in 30% Clorox bleach plus 0.5% Micro detergent for 20 minutes, and rinsed two times with sterile water. The immature embryos are excised and placed embryo axis side down (scutellum side up), 25 embryos per plate, on 560Y medium for 4 hours and then aligned within the 2.5 cm target zone in preparation for bombardment.
  • Preparation of DNA
  • This plasmid DNA plus plasmid DNA containing a PAT selectable marker is precipitated onto 1.1 μm (average diameter) tungsten pellets using a CaCl2 precipitation procedure as follows:
  • 100 μL prepared tungsten particles in water
  • 10 μL (1 μg) DNA in Tris EDTA buffer (1 μg total DNA)
  • 100 μL 2.5 M CaCl2
  • 10 μL 0.1 M spermidine
  • Each reagent is added sequentially to the tungsten particle suspension, while maintained on the multitube vortexer. The final mixture is sonicated briefly and allowed to incubate under constant vortexing for 10 minutes. After the precipitation period, the tubes are centrifuged briefly, liquid removed, washed with 500 μL 100% ethanol, and centrifuged for 30 seconds. Again the liquid is removed, and 105 μl 100% ethanol is added to the final tungsten particle pellet. For particle gun bombardment, the tungsten/DNA particles are briefly sonicated and 10 μL spotted onto the center of each macrocarrier and allowed to dry about 2 minutes before bombardment.
  • Particle Gun Treatment
  • The sample plates are bombarded at level #4 in particle gun #HE34-1 or #HE34-2. All samples receive a single shot at 650 PSI, with a total of ten aliquots taken from each tube of prepared particles/DNA.
  • Subsequent Treatment
  • Following bombardment, the embryos are kept on 560Y medium for 2 days, then transferred to 560R selection medium containing 3 mg/L Bialaphos, and subcultured every 2 weeks. After approximately 10 weeks of selection, selection-resistant callus clones are transferred to 288J medium to initiate plant regeneration. Following somatic embryo maturation (2-4 weeks), well-developed somatic embryos are transferred to medium for germination and transferred to the lighted culture room. Approximately 7-10 days later, developing plantlets are transferred to 272V hormone-free medium in tubes for 7-10 days until plantlets are well established. Plants are then transferred to inserts in flats (equivalent to 2.5″ pot) containing potting soil and grown for 1 week in a growth chamber, subsequently grown an additional 1-2 weeks in the greenhouse, then transferred to classic 600 pots (1.6 gallon) and grown to maturity. Plants are monitored and scored for and altered level of expression of the cystatin sequence of the invention. Alternatively, the cysteine proteinase activity can be assayed.
  • Bombardment and Culture Media
  • Bombardment medium (560Y) comprises 4.0 g/L N6 basal salts (SIGMA C-1416), 1.0 mL/L Eriksson's Vitamin Mix (1000X SIGMA-1511), 0.5 mg/L thiamine HCl, 120.0 g/L sucrose, 1.0 mg/L 2,4-D, and 2.88 g/L L-proline (brought to volume with D-1 H20 following adjustment to pH 5.8 with KOH); 2.0 g/L Gelrite (added after bringing to volume with D-I H20); and 8.5 mg/L silver nitrate (added after sterilizing the medium and cooling to room temperature). Selection medium (560R) comprises 4.0 g/L N6 basal salts (SIGMA C-1416), 1.0 mL/L Eriksson's Vitamin Mix (1000X SIGMA-1511), 0.5 mg/L thiamine HCl, 30.0 g/L sucrose, and 2.0 mg/L 2,4-D (brought to volume with D-I H20 following adjustment to pH 5.8 with KOH); 3.0 g/L Gelrite (added after bringing to volume with D-I H20); and 0.85 mg/L silver nitrate and 3.0 mg/L bialaphos (both added after sterilizing the medium and cooling to room temperature).
  • Plant regeneration medium (288J) comprises 4.3 g/L MS salts (GIBCO 11117-074), 5.0 mL/L MS vitamins stock solution (0.100 g nicotinic acid, 0.02 g/L thiamine HCL, 0.10 g/L pyridoxine HCL, and 0.40 g/L glycine brought to volume with polished D-I H20) (Murashige and Skoog (1962) Physiol. Plant. 15:473), 100 mg/L myo-inositol, 0.5 mg/L zeatin, 60 g/L sucrose, and 1.0 mL/L of 0.1 mM abscisic acid (brought to volume with polished D-I H20 after adjusting to pH 5.6); 3.0 g/L Gelrite (added after bringing to volume with D-I H20); and 1.0 mg/L indoleacetic acid and 3.0 mg/L bialaphos (added after sterilizing the medium and cooling to 60° C.). Hormone-free medium (272V) comprises 4.3 g/L MS salts (GIBCO 11117-074), 5.0 mL/L MS vitamins stock solution (0.100 g/L nicotinic acid, 0.02 g/L thiamine HCL, 0.10 g/L pyridoxine HCL, and 0.40 g/L glycine brought to volume with polished D-I H20), 0.1 g/L myo-inositol, and 40.0 g/L sucrose (brought to volume with polished D-I H20 after adjusting pH to 5.6); and 6 g/L bacto-agar (added after bringing to volume with polished D-I H20), sterilized and cooled to 60° C.
  • EXAMPLE 2 Agrobacterium-Mediated Transformation
  • For Agrobacterium-mediated transformation of maize with a cystatin nucleotide sequence of the invention, operably linked to a ubiquitin promoter, preferably the method of Zhao is employed (U.S. Pat. No. 5,981,840, and PCT patent publication WO98/32326; the contents of which are hereby incorporated by reference). Briefly, immature embryos are isolated from maize and the embryos contacted with a suspension of Agrobacterium, where the bacteria are capable of transferring the DNA construct containing the cystatin nucleotide sequence to at least one cell of at least one of the immature embryos (step 1: the infection step). In this step the immature embryos are preferably immersed in an Agrobacterium suspension for the initiation of inoculation. The embryos are co-cultured for a time with the Agrobacterium (step 2: the co-cultivation step). Preferably the immature embryos are cultured on solid medium following the infection step. Following this co-cultivation period an optional “resting” step is contemplated. In this resting step, the embryos are incubated in the presence of at least one antibiotic known to inhibit the growth of Agrobacterium without the addition of a selective agent for plant transformants (step 3: resting step). Preferably the immature embryos are cultured on solid medium with antibiotic, but without a selecting agent, for elimination of Agrobacterium and for a resting phase for the infected cells. Next, inoculated embryos are cultured on medium containing a selective agent and growing transformed callus is recovered (step 4: the selection step). Preferably, the immature embryos are cultured on solid medium with a selective agent resulting in the selective growth of transformed cells. The callus is then regenerated into plants (step 5: the regeneration step), and preferably calli grown on selective medium are cultured on solid medium to regenerate the plants.
  • EXAMPLE 3 Identification of the Gene from a Computer Homology Search
  • Gene identities were determined by conducting BLAST (Basic Local Alignment Search Tool; Altschul, S. F., et al. (1993) J. Mol. Biol. 215:403-410) searches under default parameters for similarity to sequences contained in the BLAST “nr” database (comprising all non-redundant GenBank CDS translations, sequences derived from the 3-dimensional structure Brookhaven Protein Data Bank, the last major release of the SWISS-PROT protein sequence database, EMBL, and DDBJ databases).
  • The cDNA sequences of the present invention were analyzed for similarity to all publicly available DNA sequences contained in the “nr” database using the BLASTN algorithm. The DNA sequences were translated in all reading frames and compared for similarity to all publicly available protein sequences contained in the “nr” database using the BLASTX algorithm (Gish, W. and States, D. J. Nature Genetics 3:266-272 (1993)) provided by the NCBI. In some cases, the sequencing data from two or more clones containing overlapping segments of DNA were used to construct contiguous DNA sequences.
  • Sequence alignments and percent identity calculations were performed using GAP, as well as the Megalign program of the LASERGENE bioinformatics computing suite (DNASTAR Inc., Madison, Wis.). Multiple alignments of the sequences were performed using the Clustal method of alignment (Higgins and Sharp (1989) CABIOS. 5:151-153) with the default parameters (GAP PENALTY=10, GAP LENGTH PENALTY=10). Default parameters for pairwise alignments using the Clustal method are KTUPLE 1, GAP PENALTY=3, WINDOW=5 and DIAGONALS SAVED=5.
  • EXAMPLE 4 Soybean Embryo Transformation
  • Soybean embryos are bombarded with a plasmid containing the cystatin sequence operably linked to a ubiquitin promoter as follows. To induce somatic embryos, cotyledons, 3-5 mm in length dissected from surface-sterilized, immature seeds of the soybean cultivar A2872, are 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 multiplied as early, globular-staged embryos, the suspensions are maintained as described below.
  • Soybean embryogenic suspension cultures are maintained in 35 mL liquid media on a rotary shaker, 150 rpm, at 26° C. with florescent lights on a 16:8 hour day/night schedule. Cultures are subcultured 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, U.S. Pat. No. 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 comprising the cystatin sequence operably linked to the ubiquitin promoter 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 μL of a 60 mg/mL 1 μm gold particle suspension is added (in order): 5 μL DNA (1 μg/μL), 20 μL spermidine (0.1 M), and 50 μL CaCl2 (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 μL 70% ethanol and resuspended in 40 μL 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 60×15 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 1100 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 5 Sunflower Meristem Tissue Transformation
  • Sunflower meristem tissues are transformed with an expression cassette containing the cystatin sequence operably linked to a ubiquitin promoter as follows (see also European Patent Number EP 0 486233, herein incorporated by reference, and Malone-Schoneberg et al. (1994) Plant Science 103:199-207). Mature sunflower seed (Helianthus annuus L.) are dehulled using a single wheat-head thresher. Seeds are surface sterilized for 30 minutes in a 20% Clorox bleach solution with the addition of two drops of Tween 20 per 50 mL of solution. The seeds are rinsed twice with sterile distilled water.
  • Split embryonic axis explants are prepared by a modification of procedures described by Schrammeijer et al. (Schrammeijer et al. (1990) Plant Cell Rep. 9: 55-60). Seeds are imbibed in distilled water for 60 minutes following the surface sterilization procedure. The cotyledons of each seed are then broken off, producing a clean fracture at the plane of the embryonic axis. Following excision of the root tip, the explants are bisected longitudinally between the primordial leaves. The two halves are placed, cut surface up, on GBA medium consisting of Murashige and Skoog mineral elements (Murashige et al. (1962) Physiol. Plant., 15: 473-497), Shepard's vitamin additions (Shepard (1980) in Emergent Techniques for the Genetic Improvement of Crops (University of Minnesota Press, St. Paul, Minn.), 40 mg/L adenine sulfate, 30 g/L sucrose, 0.5 mg/L 6-benzyl-aminopurine (BAP), 0.25 mg/L indole-3-acetic acid (IAA), 0.1 mg/L gibberellic acid (GA3), pH 5.6, and 8 g/L Phytagar.
  • The explants are subjected to microprojectile bombardment prior to Agrobacterium treatment (Bidney et al. (1992) Plant Mol. Biol. 18: 301-313). Thirty to forty explants are placed in a circle at the center of a 60×20 mm plate for this treatment. Approximately 4.7 mg of 1.8 mm tungsten microprojectiles are resuspended in 25 mL of sterile TE buffer (10 mM Tris HCl, 1 mM EDTA, pH 8.0) and 1.5 mL aliquots are used per bombardment. Each plate is bombarded twice through a 150 mm nytex screen placed 2 cm above the samples in a PDS 1000® particle acceleration device.
  • Disarmed Agrobacterium tumefaciens strain EHA105 is used in all transformation experiments. A binary plasmid vector comprising the expression cassette described above is introduced into Agrobacterium strain EHA105 via freeze-thawing as described by Holsters et al. (1978) Mol. Gen. Genet. 163:181-187. This plasmid further comprises a kanamycin selectable marker gene (i.e, nptII). Bacteria for plant transformation experiments are grown overnight (28° C. and 100 RPM continuous agitation) in liquid YEP medium (10 g/L yeast extract, 10 g/L Bactopeptone, and 5 g/L NaCl, pH 7.0) with the appropriate antibiotics required for bacterial strain and binary plasmid maintenance. The suspension is used when it reaches an OD600 of about 0.4 to 0.8. The Agrobacterium cells are pelleted and resuspended at a final OD600 of 0.5 in an inoculation medium comprised of 12.5 mM MES pH 5.7, 1 g/L NH4Cl, and 0.3 g/L MgSO4.
  • Freshly bombarded explants are placed in an Agrobacterium suspension, mixed, and left undisturbed for 30 minutes. The explants are then transferred to GBA medium and co-cultivated, cut surface down, at 26° C. and 18-hour days. After three days of co-cultivation, the explants are transferred to 374B (GBA medium lacking growth regulators and a reduced sucrose level of 1%) supplemented with 250 mg/L cefotaxime and 50 mg/L kanamycin sulfate. The explants are cultured for two to five weeks on selection and then transferred to fresh 374B medium lacking kanamycin for one to two weeks of continued development. Explants with differentiating, antibiotic-resistant areas of growth that have not produced shoots suitable for excision are transferred to GBA medium containing 250 mg/L cefotaxime for a second 3-day phytohormone treatment. Leaf samples from green, kanamycin-resistant shoots are assayed for the presence of NPTII by ELISA and for the presence of transgene expression by assaying for the activity of the cystatin sequences.
  • NPTII-positive shoots are grafted to Pioneer® hybrid 6440 in vitro-grown sunflower seedling rootstock. Surface sterilized seeds are germinated in 48-0 medium (half-strength Murashige and Skoog salts, 0.5% sucrose, 0.3% gelrite, pH 5.6) and grown under conditions described for explant culture. The upper portion of the seedling is removed, a 1 cm vertical slice is made in the hypocotyl, and the transformed shoot inserted into the cut. The entire area is wrapped with parafilm to secure the shoot. Grafted plants can be transferred to soil following one week of in vitro culture. Grafts in soil are maintained under high humidity conditions followed by a slow acclimatization to the greenhouse environment. Transformed sectors of T0 plants (parental generation) maturing in the greenhouse are identified by NPTII ELISA and/or by the analysis of the activity of the cystatin sequences in the leaf extracts while transgenic seeds harvested from NPTII-positive T0 plants are identified by the analysis of the activity the cystatin sequences in small portions of dry seed cotyledon.
  • An alternative sunflower transformation protocol allows the recovery of transgenic progeny without the use of chemical selection pressure. Seeds are dehulled and surface-sterilized for 20 minutes in a 20% Clorox bleach solution with the addition of two to three drops of Tween 20 per 100 mL of solution, then rinsed three times with distilled water. Sterilized seeds are imbibed in the dark at 26° C. for 20 hours on filter paper moistened with water. The cotyledons and root radical are removed, and the meristem explants are cultured on 374E (GBA medium consisting of MS salts, Shepard vitamins, 40 mg/L adenine sulfate, 3% sucrose, 0.5 mg/L 6-BAP, 0.25 mg/L IAA, 0.1 mg/L GA, and 0.8% Phytagar at pH 5.6) for 24 hours under the dark. The primary leaves are removed to expose the apical meristem, around 40 explants are placed with the apical dome facing upward in a 2 cm circle in the center of 374M (GBA medium with 1.2% Phytagar), and then cultured on the medium for 24 hours in the dark.
  • Approximately 18.8 mg of 1.8 μm tungsten particles are resuspended in 150 μL absolute ethanol. After sonication, 8 μl of it is dropped on the center of the surface of macrocarrier. Each plate is bombarded twice with 650 psi rupture discs in the first shelf at 26 mm of Hg helium gun vacuum.
  • The plasmid of interest is introduced into Agrobacterium tumefaciens strain EHA105 via freeze thawing as described previously. The pellet of overnight-grown bacteria at 28° C. in a liquid YEP medium (10 g/L yeast extract, 10 g/L Bactopeptone, and 5 g/L NaCl, pH 7.0) in the presence of 50 μg/L kanamycin is resuspended in an inoculation medium (12.5 mM 2-mM 2-(N-morpholino) ethanesulfonic acid, MES, 1 g/L NH4Cl and 0.3 g/L MgSO4 at pH 5.7) to reach a final concentration of 4.0 at OD 600. Particle-bombarded explants are transferred to GBA medium (374E), and a droplet of bacteria suspension is placed directly onto the top of the meristem. The explants are co-cultivated on the medium for 4 days, after which the explants are transferred to 374C medium (GBA with 1% sucrose and no BAP, IAA, GA3 and supplemented with 250 μg/mL cefotaxime). The plantlets are cultured on the medium for about two weeks under 16-hour day and 26° C. incubation conditions.
  • Explants (around 2 cm long) from two weeks of culture in 374C medium are screened for cystatin activity using assays known in the art. After positive (i.e., for cystatin expression) explants are identified, those shoots that fail to exhibit cystatin activity are discarded, and every positive explant is subdivided into nodal explants. One nodal explant contains at least one potential node. The nodal segments are cultured on GBA medium for three to four days to promote the formation of auxiliary buds from each node. Then they are transferred to 374C medium and allowed to develop for an additional four weeks. Developing buds are separated and cultured for an additional four weeks on 374C medium. Pooled leaf samples from each newly recovered shoot are screened again by the appropriate protein activity assay. At this time, the positive shoots recovered from a single node will generally have been enriched in the transgenic sector detected in the initial assay prior to nodal culture.
  • Recovered shoots positive for defense-inducible expression are grafted to Pioneer hybrid 6440 in vitro-grown sunflower seedling rootstock. The rootstocks are prepared in the following manner. Seeds are dehulled and surface-sterilized for 20 minutes in a 20% Clorox bleach solution with the addition of two to three drops of Tween 20 per 100 mL of solution, and are rinsed three times with distilled water. The sterilized seeds are germinated on the filter moistened with water for three days, then they are transferred into 48 medium (half-strength MS salt, 0.5% sucrose, 0.3% gelrite pH 5.0) and grown at 26° C. under the dark for three days, then incubated at 16-hour-day culture conditions. The upper portion of selected seedling is removed, a vertical slice is made in each hypocotyl, and a transformed shoot is inserted into a V-cut. The cut area is wrapped with parafilm. After one week of culture on the medium, grafted plants are transferred to soil. In the first two weeks, they are maintained under high humidity conditions to acclimatize to a greenhouse environment.
  • EXAMPLE 6 Transformation of BL21 Star Cells and Cystatin Expression and Purification
  • Strain Transformation
  • One μL samples of mini-prep DNA were incubated on ice with 20 μL of BL21 Star cells (Invitrogen) for 30 minutes. The DNA/cell mixtures were then heated at 42° C. for 45 sec, then iced for 2 min. 200 μL of SOC were added to each tube, and these mixtures were then incubated at 37° C. for 1 hour. 50 μL samples were spread out on LB broth plates containing 100 mg/L carbenicillin and 0.1 M MgSO4 to incubate overnight at 37° C.
  • Protein Expression
  • Bacteria samples in duplicate were selected from the plates, and these were incubated overnight in 2.0 mL of 2xYT media containing 100 μg/mL carbenicillin. The incubations took place in a 48 well, pyramid bottom plate (Innovative Microplate) at 37° C. on a shaker (225 rpm).
  • The next day, the OD readings for the wells were obtained after diluting 10 μL bacterial samples with 90 μL of water in a standard 96-well, flat bottomed microtiter plate, with the absorbance read at 600 nm. Based on these values, the cultures were diluted with fresh 2xYT (+carb.) to the two target induction OD600 values of 0.1 and 0.6, to a final volume of 2 mL. Each target OD value was diluted in quadruplicate to account for two isopropyl-beta-D-thiogalactopyranoside (IPTG) induction values each and two incubation temperatures each. When the plates were ready, the wells were induced with IPTG at concentrations of either 0.05 mM or 1.0 mM. The plates were then incubated overnight on shakers (225 rpm) at temperatures of either 16° C. or 30° C. After approximately 20 hours of incubation, the OD values for all the wells were obtained as described supra. The cells were then harvested by centrifuging the plates at 1700 rpm for 10 minutes. The supernatants were discarded and the pelleted cells frozen at −20° C. until purification could take place.
  • Protein Purification
  • The cells were allowed to thaw for several minutes, then 200 μL of B-Peril protein extraction reagent (Pierce) containing 0.2 units of benzonase (Novagen) were added to each well, and the solution was pipetted repeatedly to resuspend the pellet. The cells were incubated for 10 min in the B-Peril solution.
  • 25-30 μm MBPP 800 μL purification filter plates (Whatman) were prepared by adding 200 μL of 50% slurry of glutathione Sepharose 4B resin (Amersham) for GST-tagged proteins, or TALON resin (Clontech) for HIS-tagged proteins into each well. The plates were centrifuged briefly to remove excess buffer. The lysates were transferred directly from the growth plate to the plates with the resins, and the solutions were pipetted up and down several times to mix them well. The plates were then centrifuged at 760 rpm for 5 min, and the flow-through was discarded. The resins with the bound proteins were washed twice with 200 μL of buffer A (50 mM Tris, pH 8.0, 200 mM NaCl, 10% glycerol), and each time the plates were centrifuged at 760 rpm for 5 min and the wash was discarded. Standard 96 well plates were placed underneath the Whatman filter plates, and the proteins were eluted by adding and mixing 100 μL of buffer A containing either 20 mM reduced glutathione (Sigma) for GST tags, or 500 mM imidazole (Sigma) for HIS tags. The plates were centrifuged at 1000 rpm for 5 min, and the purified proteins were collected in the standard 96 well plates placed underneath.
  • Bradford reactions were performed to determine the protein concentrations for all the samples. These values were used to establish normalization plates for the cystatin assays where equal volumes contained equal amounts of protein. However, when the protein concentration was too low for a given sample, 20 μL were used directly in the cystatin assay to come as close as possible to the target amount of 6 μg of protein.
  • EXAMPLE 7 Anti-Fungal and Anti-Bacterial Assays
  • The anti-fungal and anti-bacterial activity of the cystatin polypeptides of the invention can be tested using a variety of assays.
  • BL21 cells were transformed with cystatin genes using the Lambda CE6 Induction Kit (Stratagene) according to the manufacturer's protocol. The induced BL21 cells were grown and harvested, and then resuspended in the binding buffer from a His-Bind Kit (Novagen). The suspended cells were sonicated to release the expressed protein from the cells. The crude protein extract was denatured by dissolving it into a 6 M urea solution for 1 hour on ice in order to allow the His-tag to efficiently bind to the resin. The denatured cystatins were then purified by His-Bind Resin Chromatography according to the manufacturer's protocol, except that 6M Urea was added to the binding, washing and elution buffers.
  • The purified proteins were renatured by dialysis using 1×PBS buffer with gradually decreased concentration of urea (5M, 4M, 3M, 1.5M, and 0.75M). The dialyses were performed with each concentration of urea for one hour and then with 1×PBS buffer without urea overnight at 4° C. Bradford reactions were performed to determine the protein concentrations for all the samples. The isolated polypeptide was then tested for activity.
  • Anti-Fungal Assays:
  • Fusarium verticillioides (strain M033) was grown on ½× potato dextrose agar plates: (For each liter, 12 grams Difco Potato Dextrose Broth (#0549-17-9) and 15 grams agar were suspended in dH2O, final volume was raised to 1 liter and autoclaved at 121° C. for 20 minutes. Plates were poured in sterile hood.) Spores were collected from 10 day old to 3 week old culture plates of Fusarium verticillioides by rinsing a portion of the plate with potato dextrose broth (24 grams Difco Potato Dextrose Broth (#0549-17-9) per liter+0.08% tween-20). The collected spore solution was then vortexed and placed on ice. The spores were counted with a hemocytometer and used within 2 hours. 4× assay medium with spores was prepared by diluting the collected spores with potato dextrose broth+0.08% tween-20 to 16,000 spores per milliliter.
  • The two purified cystatins used in this assay were supplied in PBS buffer, which was prepared by diluting a 10× commercial stock buffer (10× phosphate buffered saline [137 mM NaCl, 2.7 mM KCl, 10 mM phosphate pH 7.3-7.5] EM Science Catalog #6505) with water. The protein concentration of each cystatin was measured using the Pierce BCA protein assay (BCA Protein Assay Reagent, Pierce catalog #23225; Smith, P. K., et al. (1985). Measurement of protein using bicinchoninic acid (Anal. Biochem. 150, 76-85.) calibrated against BSA. The purified GmCys-2 (SEQ ID NO: 30) protein was supplied at 5.7 mg/mL and the ZmCys-4 (SEQ ID NO: 6) protein was supplied at 3.2 mg/mL.
  • For an assay, 25 μL of the 4× assay medium with spores was diluted to 1× with 75 μL test sample/water. Assays were conducted in 96 well microtiter plates.
  • Plates were covered and incubated at 280 in the dark for 24-48 h. Growth was evaluated visually using an inverted microscope, and a scale of 0-4 was used to rate the effect of added peptide
  • Antifungal activity was rated as follows:
  • 0=no observable inhibition relative to water control
  • 1=slight inhibition
  • 2=strong inhibition, contained growth (fuzzy balls)
  • 3=strong inhibition, very little branching
  • 4=complete inhibition of germination
  • PBS buffer was tested at an equivalent dilution and no effect was observed (score=0) for all control samples.
    TABLE 13
    Anti-Fusarium activity of maize cystatin Zm-Cys4
    and soybean cystatin Gm-Cys2.
    ZmCys-4 GmCys-2 (SEQ ID NO: 30)
    (SEQ ID NO: 6) Score
    Concentration Score Concentration 48
    (μg/mL) 26 hours (μg/mL) 24 hours hours
    400 3.5 570 4 3.5
    200 3.5 280 4 0
    100 3.5 140 3 0
    50 3 71 2 0
    25 2 35 1 0
    12.5 1 18 0 0
    6 1 9 0 0
    3 1 4 0 0

    The results shown in Table 13, above, are a clear indication that both ZmCys-4 (SEQ ID NO: 6) and GmCys-2 (SEQ ID NO: 30) have a significant anti-fungal activity towards F. verticillioides.
  • Anti-bacterial Assays. Cultures are grown to midlog phase (E. coli in LB broth and C. nebraskense in NBY) and are then harvested by centrifugation (2000×g for 10 min). Cells are washed with 10 mM sodium phosphate buffer, pH 5.8 (C. nebraskense) or pH 6.5 (E. coli) by centrifugation and then colony forming units are estimated spectrophotometrically at 600 nm with previously established colony forming unit-optical density relationships used as a reference.
  • Assays for bactericidal activity are performed by incubating 105 bacterial colony forming units in 90 μL with 10 mL of peptide (or water for control). After 60 min at 37° C. (E. coli) or 25° C. (C. nebraskense), four serial, 10-fold dilutions are made in sterile phosphate buffer. Aliquots of 100 μL are plated on LB or NBY plates, using 1 or 2 plates/dilution. Resulting colonies are counted, and the effect of the peptide is expressed as percent of initial colony count (Selsted et al. (1984) Infect. Immun. 45:150-154).
  • Assays for bacteriostatic activity are performed by incubating 105 bacteria with MBP-1 in 200 μL of dilute medium (1 part NBY broth to 4 parts 10 mM sodium phosphate, pH 5.8) in microtiter plate wells. Plates are covered, sealed, and incubated at 28° C. Growth is monitored spectrophotometrically at 600 nm. After 41 h controls will have grown sufficiently (optical density >0.20) to measure effect of peptide as percent of control.
  • EXAMPLE 8 Proteinase Inhibition Assays
  • Cystatin Assay
  • The measurement of inhibition activity of cystatins toward papain was based on the method by Kouzuma et al. (1996) (J. Biochem. 119: 1106-1113) (see FIG. 1). The assay was performed in a 96 well plate. Twenty μL of a cystatin solution containing either 6 μg or 0.06 μg of cystatin protein were incubated with 20 μL of papain solution (0.1 mg/mL papain (Calbiochem) stock in 100 mM phosphate buffer, pH 6.5, 0.3 M KCl, 0.1 mM EDTA, and 1 mM freshly added DTT) at 37° C. for 15 min. 200 μL of filtered substrate solution (504 μM L-pyroglutamyl-L-phenylalanyl-L-leucine p-nitroanilide (Pyr-Phe-Leu-pNA; Sigma, P3169) with 10% DMSO (Sigma) in the same buffer as above) were then added to the reaction mix and the plate incubated at 37° C. for one hour. Formation of p-nitroaniline was measured by measuring absorbance at 420 nm. Further 37° C. incubation took place with 420 nm readings being taken at desired time intervals. The reaction was stopped by the addition of 30 μL of 30% acetic acid without significantly affecting the absorbance values. Cysteine proteinase inhibitory activity was indicated by smaller absorbance values for the 6 μg protein wells versus the 0.06 μg wells for each cystatin. Negative controls consisted of buffer without cystatin protein. Positive controls consisted of previously tested cystatins.
    TABLE 14
    Impact of N-terminal tag and induction conditions on cystatin activity.
    Cystatin activity
    Gene SEQ ID NO: GST-tagged His-tagged Conditions for highest activity
    GmCys2 29 low/medium Low GST: 16 C.
    His: 25 C., OD 0.8
    GmCys4 33 not not detected
    detected
    GmCys5 35 low/medium not detected GST: 30 C., 0.05 mM IPTG, OD
    0.1
    GmCys7 39 low not detected GST: 30 C., 0.05 mM IPTG, OD
    0.1
    GmCys9 43 high High GST: 30 C., 0.05 mM IPTG, OD
    0.1
    His: 16 C., 1 mM IPTG, OD 0.6
    OsCys4 51 not not detected
    detected
    OsCys6 55 high High GST: 30 C., 1 mM IPTG, OD 0.6
    His: 16 C., 0.05 mM IPTG, OD
    0.6
    TaCys8 67 low/medium low/medium GST: 30 C.
    (transient) (transient) His: 30 C.
    ZmCys4 5 high High GST: 25 C.
    HIS: 30 C.
    ZmCys6 9 not not detected
    detected
    ZmCys7 11 high Low GST: 30 C.
    His: 30 C., OD 0.6
    ZmCys8 13 not not detected
    detected
    ZmCys10 17 not not detected
    detected
    ZmCys11 19 not very low His: 16 C., 0.05 mM IPTG, OD
    detected 0.1
    ZmCys12 21 low Low GST: 30 C., 0.05 mM IPTG, OD
    0.1
    His: 30 C., 1 mM IPTG, OD 0.6
    ZmCys13 23 high High GST: 30 C.
    His: 16 C., OD 0.6
    ZmCys14 25 low/medium low/medium GST: 30 C.
    His: 30 C.
  • Table 14, above, is a summary of the cystatin activity detected in the tested genes of the present invention. Detailed data for the assay results on each of the above cystatins is presented in tables 15 through 60.
  • Some of the identified cystatins did not show any proteinase inhibitor activity in the conditions of the assay as conducted above. There can be numerous reasons for these results. As is well known in the art, certain microorganisms perform better than others for expression of a given protein. Often it is difficult to predict which one will work best. The cystatins of the present invention have been expressed in BL21 Star cells (Invitrogen). Other bacterial lines or expression systems may be more effective for a selection of the genes. Furthermore, the cystatin proteins were expressed with a N-terminal His- or GST-tag to facilitate cystatin purification from the bacterial culture. It is possible that the BL21 bacteria were unable to express sufficient amounts of correctly folded protein or, that for unknown reasons, recovery of certain cystatin proteins from the bacterial culture was low. It is also possible that correct folding and cystatin activity are impacted by the presence of the GST or His tag. See results given in Table 14. A C-terminal tag may be more effective for certain cystatins. A preferred method for expression of these proteins in plants would be to express the cystatins without a tag. Untagged cystatins can be expected to have improved activity compared to tagged cystatins. It is also possible that certain cystatins have a pH optimum that is distinct from pH 6.5 at which papain inhibition was measured. Another possibility is that while certain cystatins appear to be inactive as inhibitors of papain the expressed proteins inhibit other proteinases.
    TABLE 15
    Cystatin Activity Results
    GST fusion tagged protein, first replicate
    GmCys4 (SEQ ID NO: 34)
    Cystatin Assay 9/26 First Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (mM) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 16 0.05 0.1 2.404 0.082 0.281 0.206 −0.08 0.556 0.537 −0.02
    16 1.0 0.1 2.312 0.048 0.258 0.207 −0.05 0.552 0.553 0.00
    16 0.05 0.6 3.654 0.079 0.276 0.205 −0.07 0.550 0.541 −0.01
    16 1.0 0.6 3.747 0.039 0.260 0.209 −0.05 0.557 0.552 −0.01
    30 0.05 0.1 5.419 0.194 0.303 0.247 −0.06 0.545 0.552 0.01
    30 1.0 0.1 6.305 0.161 0.293 0.240 −0.05 0.548 0.556 0.01
    30 0.05 0.6 6.259 0.179 0.305 0.241 −0.06 0.551 0.554 0.00
    30 1.0 0.6 6.259 0.150 0.302 0.245 −0.06 0.552 0.555 0.00
    BL21 Star 16 0.05 0.1 0.882 0.021 0.229 0.204 −0.03 0.562 0.552 −0.01
    16 1.0 0.1 0.882 0.026 0.237 0.206 −0.03 0.551 0.550 0.00
    16 0.05 0.6 1.712 0.064 0.262 0.214 −0.05 0.547 0.552 0.01
    16 1.0 0.6 1.389 0.046 0.255 0.219 −0.04 0.549 0.555 0.01
    30 0.05 0.1 4.025 0.096 0.242 0.226 −0.02 0.544 0.554 0.01
    30 1.0 0.1 4.303 0.154 0.269 0.221 −0.05 0.542 0.556 0.01
    30 0.05 0.6 4.675 0.154 0.261 0.224 −0.04 0.551 0.547 0.00
    30 1.0 0.6 5.047 0.110 0.202 0.224 0.02 0.547 0.550 0.00
  • TABLE 16
    Cystatin Activity Results
    GST fusion tagged protein, second replicate
    GmCys4 (SEQ ID NO: 34)
    Cystatin Assay 9/26 Second Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (Mm) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 16 0.05 0.1 2.543 0.095 0.292 0.246 −0.05 0.549 0.564 0.01
    16 1.0 0.1 2.404 0.077 0.302 0.250 −0.05 0.554 0.564 0.01
    16 0.05 0.6 3.191 0.064 0.288 0.230 −0.06 0.551 0.556 0.01
    16 1.0 0.6 3.283 0.066 0.313 0.224 −0.09 0.549 0.549 0.00
    30 0.05 0.1 5.233 0.119 0.296 0.247 −0.05 0.549 0.552 0.00
    30 1.0 0.1 5.746 0.165 0.295 0.251 −0.04 0.543 0.552 0.01
    30 0.05 0.6 6.119 0.182 0.311 0.259 −0.05 0.557 0.553 0.00
    30 1.0 0.6 6.445 0.152 0.303 0.251 −0.05 0.557 0.552 −0.01
    BL21 Star 16 0.05 0.1 0.928 0.023 0.254 0.222 −0.03 0.568 0.551 −0.02
    16 1.0 0.1 0.882 0.019 0.245 0.228 −0.02 0.561 0.554 −0.01
    16 0.05 0.6 1.758 0.064 0.285 0.221 −0.06 0.546 0.545 0.00
    16 1.0 0.6 1.620 0.05 0.283 0.226 −0.06 0.544 0.549 0.01
    30 0.05 0.1 4.350 0.125 0.259 0.234 −0.03 0.545 0.550 0.01
    30 1.0 0.1 4.396 0.152 0.272 0.235 −0.04 0.543 0.556 0.01
    30 0.05 0.6 5.419 0.1 0.278 0.242 −0.04 0.546 0.544 0.00
    30 1.0 0.6 5.419 0.137 0.273 0.240 −0.03 0.525 0.538 0.01
  • TABLE 17
    Cystatin Activity Results
    His fusion tagged protein, first replicate
    GmCys4 (SEQ ID NO: 34)
    Cystatin Assay 9/26 First Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (Mm) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 16 0.05 0.1 2.404 0.233 0.254 0.205 −0.05 0.543 0.554 0.01
    16 1.0 0.1 1.896 0.228 0.247 0.202 −0.05 0.536 0.549 0.01
    16 0.05 0.6 3.422 0.347 0.273 0.197 −0.08 0.535 0.544 0.01
    16 1.0 0.6 2.543 0.302 0.273 0.202 −0.07 0.542 0.543 0.00
    30 0.05 0.1 6.212 0.457 0.270 0.229 −0.04 0.541 0.552 0.01
    30 1.0 0.1 5.187 0.410 0.267 0.223 −0.04 0.542 0.548 0.01
    30 0.05 0.6 5.839 0.570 0.275 0.225 −0.05 0.536 0.550 0.01
    30 1.0 0.6 5.559 0.463 0.266 0.215 −0.05 0.533 0.547 0.01
    BL21 Star 16 0.05 0.1 1.389 0.084 0.271 0.206 −0.07 0.543 0.547 0.00
    16 1.0 0.1 1.389 0.079 0.271 0.218 −0.05 0.542 0.544 0.00
    16 0.05 0.6 2.867 0.302 0.273 0.206 −0.07 0.538 0.540 0.00
    16 1.0 0.6 2.219 0.246 0.280 0.200 −0.08 0.534 0.540 0.01
    30 0.05 0.1 5.001 0.435 0.269 0.226 −0.04 0.527 0.552 0.03
    30 1.0 0.1 5.187 0.456 0.267 0.229 −0.04 0.521 0.549 0.03
    30 0.05 0.6 4.629 0.412 0.282 0.219 −0.06 0.545 0.546 0.00
    30 1.0 0.6 4.861 0.456 0.275 0.216 −0.06 0.535 0.544 0.01
  • TABLE 18
    Cystatin Activity Results
    His fusion tagged protein, second replicate
    GmCys4 (SEQ ID NO: 34)
    Cystatin Assay 9/26 Second Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (Mm) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 16 0.05 0.1 2.219 0.255 0.289 0.223 −0.07 0.539 0.547 0.01
    16 1.0 0.1 1.989 0.261 0.291 0.225 −0.07 0.543 0.553 0.01
    16 0.05 0.6 2.867 0.3 0.298 0.221 −0.08 0.538 0.547 0.01
    16 1.0 0.6 2.635 0.282 0.284 0.213 −0.07 0.536 0.540 0.00
    30 0.05 0.1 5.746 0.528 0.282 0.232 −0.05 0.539 0.544 0.01
    30 1.0 0.1 4.954 0.312 0.274 0.231 −0.04 0.543 0.551 0.01
    30 0.05 0.6 5.932 0.534 0.282 0.243 −0.04 0.531 0.548 0.02
    30 1.0 0.6 5.606 0.44 0.278 0.239 −0.04 0.537 0.542 0.01
    BL21 Star 16 0.05 0.1 1.297 0.086 0.294 0.226 −0.07 0.546 0.544 0.00
    16 1.0 0.1 1.389 0.084 0.291 0.223 −0.07 0.544 0.549 0.01
    16 0.05 0.6 2.728 0.016 0.221 0.215 −0.01 0.550 0.543 −0.01
    16 1.0 0.6 2.497 0.212 0.289 0.214 −0.08 0.538 0.537 0.00
    30 0.05 0.1 5.094 0.471 0.277 0.209 −0.07 0.540 0.541 0.00
    30 1.0 0.1 5.140 0.515 0.281 0.235 −0.05 0.533 0.546 0.01
    30 0.05 0.6 4.303 0.463 0.280 0.239 −0.04 0.528 0.546 0.02
    30 1.0 0.6 4.629 0.377 0.286 0.231 −0.06 0.530 0.529 0.00
  • TABLE 19
    Cystatin Activity Results
    GST fusion tagged protein, first replicate
    GmCys2 (SEQ ID NO: 30)
    Cystatin Assay 6/12
    Cell Temp IPTG Bradford Overnight
    Type ° C. (Mm) OD600 Final OD (ug/uL) 600 ng 60 ng 6 ng Δ
    BL21 16 0.5 0.1 4.559 0.089 0.270 0.232 0.233 −0.038
    16 0.5 0.8 6.894 0.129 0.258 0.230 0.224 −0.031
    16 0.05 0.1 7.694 0.150 0.275 0.229 0.240 −0.040
    16 0.05 0.8 11.171 0.143 0.263 0.229 0.220 −0.038
    25 0.5 0.1 3.140 0.000 0.237 0.206 0.201 −0.034
    25 0.5 0.8 3.186 0.004 0.233 0.211 0.197 −0.029
    25 0.05 0.1 4.285 0.062 0.245 0.216 0.214 −0.030
    25 0.05 0.8 4.461 0.067 0.244 0.211 0.206 −0.036
    BL21 Star 16 0.5 0.1 1.906 0.134 0.258 0.222 0.223 −0.036
    16 0.5 0.8 5.131 0.171 0.216 0.224 0.219 0.006
    16 0.05 0.1 1.726 0.024 0.241 0.223 0.221 −0.019
    16 0.05 0.8 4.415 0.159 0.201 0.219 0.218 0.017
    25 0.5 0.1 3.547 0.212 0.210 0.195 0.193 −0.016
    25 0.5 0.8 4.248 0.276 0.209 0.188 0.194 −0.018
    25 0.05 0.1 3.241 0.259 0.215 0.199 0.195 −0.019
    25 0.05 0.8 3.714 0.524 0.204 0.194 0.189 −0.012
  • TABLE 20
    Cystatin Activity Results
    GST fusion tagged protein, second replicate
    GmCys2 (SEQ ID NO: 30)
    Cystatin Assay 7/24
    Cell Temp IPTG Bradford Overnight
    Type ° C. (Mm) OD600 Final OD (ug/uL) 600 ng 60 ng 6 ng Δ
    BL21 16 0.5 0.1 5.331 0.058 0.243 0.200 0.186 −0.049
    16 0.5 0.8 7.142 0.081 0.240 0.200 0.198 −0.041
    16 0.05 0.1 7.769 0.082 0.239 0.200 0.193 −0.043
    16 0.05 0.8 10.354 2.948 0.254 0.201 0.203 −0.052
    25 0.5 0.1 2.894 0.000 0.253 0.204 0.191 −0.055
    25 0.5 0.8 3.010 0.009 0.243 0.196 0.185 −0.052
    25 0.05 0.1 3.686 0.048 0.250 0.208 0.211 −0.040
    25 0.05 0.8 4.294 0.052 0.244 0.197 0.198 −0.046
    BL21 Star 16 0.5 0.1 3.269 0.135 0.226 0.181 0.186 −0.042
    16 0.5 0.8 6.576 0.119 0.211 0.190 0.191 −0.020
    16 0.05 0.1 2.899 0.155 0.215 0.188 0.185 −0.028
    16 0.05 0.8 5.401 0.097 0.178 0.186 0.187 0.008
    25 0.5 0.1 3.862 0.542 0.232 0.185 0.183 −0.048
    25 0.5 0.8 3.802 0.246 0.221 0.186 0.180 −0.038
    25 0.05 0.1 3.492 0.302 0.231 0.186 0.190 −0.043
    25 0.05 0.8 3.329 0.669 0.215 0.192 0.195 −0.022
  • TABLE 21
    Cystatin Activity Results
    His fusion tagged protein, first replicate
    GmCys2 (SEQ ID NO: 30)
    Cystatin Assay 6/12
    Cell Temp IPTG Bradford Overnight
    Type ° C. (Mm) OD600 Final OD (ug/uL) 600 ng 60 ng 6 ng Δ
    BL21 16 0.5 0.1 5.312 0.351 0.256 0.227 0.225 −0.030
    16 0.5 0.8 7.076 0.456 0.297 0.295 0.285 −0.007
    16 0.05 0.1 7.324 0.410 0.260 0.224 0.228 −0.033
    16 0.05 0.8 10.935 0.578 0.294 0.290 0.296 −0.001
    25 0.5 0.1 3.241 0.280 0.235 0.198 0.210 −0.031
    25 0.5 0.8 3.468 0.203 0.258 0.208 0.216 −0.046
    25 0.05 0.1 4.498 0.300 0.238 0.244 0.215 −0.008
    25 0.05 0.8 4.215 0.345 0.259 0.227 0.222 −0.035
    BL21 Star 16 0.5 0.1 5.760 0.342 0.263 0.220 0.225 −0.041
    16 0.5 0.8 7.058 0.456 0.283 0.294 0.290 0.009
    16 0.05 0.1 4.698 0.295 0.259 0.224 0.219 −0.037
    16 0.05 0.8 5.690 0.453 0.285 0.288 0.294 0.005
    25 0.5 0.1 4.016 0.337 0.215 0.192 0.194 −0.022
    25 0.5 0.8 3.714 0.353 0.243 0.216 0.210 −0.030
    25 0.05 0.1 3.492 0.383 0.224 0.199 0.197 −0.026
    25 0.05 0.8 2.941 0.410 0.249 0.216 0.212 −0.035
  • TABLE 22
    Cystatin Activity Results
    His fusion tagged protein, second replicate
    GmCys2 (SEQ ID NO: 30)
    Cystatin Assay 7/24
    Cell Temp IPTG Bradford Overnight
    Type ° C. (Mm) OD600 Final OD (ug/uL) 600 ng 60 ng 6 ng Δ
    BL21 16 0.5 0.1 5.131 0.245 0.234 0.188 0.206 −0.037
    16 0.5 0.8 6.049 0.421 0.254 0.205 0.201 −0.051
    16 0.05 0.1 7.268 0.306 0.244 0.200 0.203 −0.042
    16 0.05 0.8 8.599 0.559 0.245 0.213 0.538 0.130
    25 0.5 0.1 2.728 0.204 0.234 0.197 0.241 −0.015
    25 0.5 0.8 2.880 0.290 0.266 0.219 0.206 −0.053
    25 0.05 0.1 3.473 0.300 0.223 0.240 0.217 0.006
    25 0.05 0.8 3.825 0.352 0.251 0.207 0.202 −0.046
    BL21 Star 16 0.5 0.1 4.991 0.302 0.258 0.184 0.222 −0.055
    16 0.5 0.8 6.553 0.434 0.245 0.208 0.194 −0.044
    16 0.05 0.1 3.927 0.144 0.220 0.191 0.188 −0.030
    16 0.05 0.8 5.014 0.458 0.245 0.201 0.203 −0.043
    25 0.5 0.1 3.556 0.259 0.229 0.203 0.208 −0.024
    25 0.5 0.8 3.427 0.219 0.247 0.213 0.207 −0.038
    25 0.05 0.1 3.093 0.298 0.225 0.210 0.220 −0.010
    25 0.05 0.8 3.019 0.259 0.271 0.189 0.214 −0.069
  • TABLE 23
    Cystatin Activity Results
    GST fusion tagged protein, first replicate
    OsCys4 (SEQ ID NO: 52)
    Cystatin Assay 9/26 First Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (Mm) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 16 0.05 0.1 2.312 0.073 0.275 0.203 −0.07 0.554 0.536 −0.02
    16 1.0 0.1 2.035 0.046 0.264 0.211 −0.05 0.553 0.550 0.00
    16 0.05 0.6 3.839 0.073 0.282 0.204 −0.08 0.559 0.539 −0.02
    16 1.0 0.6 3.515 0.059 0.271 0.187 −0.08 0.549 0.540 −0.01
    30 0.05 0.1 6.352 0.171 0.304 0.242 −0.06 0.543 0.546 0.00
    30 1.0 0.1 5.746 0.156 0.296 0.235 −0.06 0.540 0.548 0.01
    30 0.05 0.6 5.932 0.100 0.292 0.235 −0.06 0.550 0.552 0.00
    30 1.0 0.6 6.305 0.110 0.285 0.233 −0.05 0.542 0.554 0.01
    BL21 Star 16 0.05 0.1 1.067 0.019 0.232 0.194 −0.04 0.566 0.552 −0.01
    16 1.0 0.1 1.021 0.017 0.239 0.195 −0.04 0.558 0.547 −0.01
    16 0.05 0.6 1.896 0.070 0.263 0.198 −0.07 0.541 0.546 0.01
    16 1.0 0.6 1.989 0.075 0.269 0.191 −0.08 0.542 0.545 0.00
    30 0.05 0.1 4.489 0.182 0.275 0.224 −0.05 0.539 0.545 0.01
    30 1.0 0.1 4.954 0.123 0.205 0.220 0.02 0.539 0.551 0.01
    30 0.05 0.6 4.582 0.158 0.265 0.217 −0.05 0.536 0.549 0.01
    30 1.0 0.6 5.094 0.150 0.232 0.219 −0.01 0.552 0.545 −0.01
  • TABLE 24
    Cystatin Activity Results
    GST fusion tagged protein, second replicate
    OsCys4 (SEQ ID NO: 52)
    Cystatin Assay 9/26 Second Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (Mm) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 16 0.05 0.1 2.035 0.075 0.291 0.214 −0.08 0.552 0.540 −0.01
    16 1.0 0.1 2.173 0.062 0.297 0.210 −0.09 0.554 0.543 −0.01
    16 0.05 0.6 3.654 0.098 0.297 0.222 −0.08 0.551 0.545 −0.01
    16 1.0 0.6 3.144 0.068 0.303 0.228 −0.08 0.548 0.557 0.01
    30 0.05 0.1 6.072 0.228 0.296 0.244 −0.05 0.542 0.545 0.00
    30 1.0 0.1 6.025 0.154 0.285 0.240 −0.05 0.535 0.551 0.02
    30 0.05 0.6 6.679 0.085 0.280 0.241 −0.04 0.549 0.547 0.00
    30 1.0 0.6 6.119 0.182 0.294 0.245 −0.05 0.542 0.554 0.01
    BL21 Star 16 0.05 0.1 0.975 0.023 0.252 0.220 −0.03 0.562 0.549 −0.01
    16 1.0 0.1 0.975 0.025 0.262 0.230 −0.03 0.559 0.554 −0.01
    16 0.05 0.6 1.804 0.062 0.294 0.225 −0.07 0.548 0.558 0.01
    16 1.0 0.6 1.850 0.077 0.295 0.230 −0.07 0.544 0.562 0.02
    30 0.05 0.1 4.350 0.217 0.256 0.238 −0.02 0.530 0.550 0.02
    30 1.0 0.1 4.954 0.282 0.266 0.233 −0.03 0.535 0.546 0.01
    30 0.05 0.6 5.513 0.266 0.243 0.238 −0.01 0.537 0.553 0.02
    30 1.0 0.6 5.419 0.163 0.275 0.236 −0.04 0.541 0.548 0.01
  • TABLE 25
    Cystatin Activity Results
    His fusion tagged protein, first replicate
    OsCys4 (SEQ ID NO: 52)
    Cystatin Assay 9/26 First Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (Mm) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 16 0.05 0.1 2.219 0.194 0.257 0.197 −0.06 0.542 0.540 0.00
    16 1.0 0.1 1.942 0.212 0.263 0.193 −0.07 0.538 0.544 0.01
    16 0.05 0.6 3.283 0.304 0.275 0.186 −0.09 0.533 0.542 0.01
    16 1.0 0.6 2.728 0.313 0.262 0.186 −0.08 0.537 0.542 0.01
    30 0.05 0.1 6.072 0.459 0.279 0.218 −0.06 0.535 0.543 0.01
    30 1.0 0.1 5.466 0.391 0.278 0.210 −0.07 0.532 0.542 0.01
    30 0.05 0.6 5.979 0.448 0.273 0.219 −0.05 0.530 0.542 0.01
    30 1.0 0.6 5.559 0.438 0.266 0.228 −0.04 0.522 0.558 0.04
    BL21 Star 16 0.05 0.1 1.389 0.095 0.272 0.198 −0.07 0.543 0.547 0.00
    16 1.0 0.1 1.343 0.100 0.281 0.215 −0.07 0.541 0.543 0.00
    16 0.05 0.6 3.329 0.295 0.260 0.190 −0.07 0.534 0.536 0.00
    16 1.0 0.6 2.820 0.270 0.265 0.196 −0.07 0.534 0.541 0.01
    30 0.05 0.1 5.326 0.433 0.279 0.200 −0.08 0.534 0.534 0.00
    30 1.0 0.1 6.072 0.459 0.278 0.203 −0.08 0.528 0.537 0.01
    30 0.05 0.6 5.280 0.415 0.260 0.216 −0.04 0.533 0.542 0.01
    30 1.0 0.6 4.954 0.423 0.271 0.218 −0.05 0.539 0.538 0.00
  • TABLE 26
    Cystatin Activity Results
    His fusion tagged protein, second replicate
    OsCys4 (SEQ ID NO: 52)
    Cystatin Assay 9/26 Second Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (Mm) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 16 0.05 0.1 1.989 0.217 0.291 0.224 −0.07 0.544 0.552 0.01
    16 1.0 0.1 2.035 0.237 0.284 0.219 −0.07 0.543 0.548 0.01
    16 0.05 0.6 2.820 0.304 0.297 0.222 −0.08 0.545 0.549 0.00
    16 1.0 0.6 2.867 0.288 0.279 0.225 −0.05 0.543 0.547 0.00
    30 0.05 0.1 5.606 0.463 0.277 0.232 −0.05 0.531 0.542 0.01
    30 1.0 0.1 5.280 0.412 0.269 0.231 −0.04 0.529 0.545 0.02
    30 0.05 0.6 5.885 0.505 0.276 0.231 −0.05 0.537 0.547 0.01
    30 1.0 0.6 5.466 0.49 0.275 0.231 −0.04 0.538 0.549 0.01
    BL21 Star 16 0.05 0.1 1.435 0.1 0.291 0.221 −0.07 0.541 0.548 0.01
    16 1.0 0.1 1.389 0.088 0.289 0.207 −0.08 0.539 0.536 0.00
    16 0.05 0.6 2.682 0.205 0.279 0.228 −0.05 0.543 0.561 0.02
    16 1.0 0.6 2.312 0.156 0.297 0.229 −0.07 0.539 0.550 0.01
    30 0.05 0.1 5.140 0.517 0.267 0.237 −0.03 0.521 0.544 0.02
    30 1.0 0.1 5.326 0.509 0.274 0.236 −0.04 0.523 0.540 0.02
    30 0.05 0.6 5.792 0.469 0.223 0.235 0.01 0.546 0.547 0.00
    30 1.0 0.6 5.140 0.45 0.276 0.223 −0.05 0.529 0.534 0.01
  • TABLE 27
    Cystatin Activity Results
    GST fusion tagged protein, first replicate
    ZmCys4 (SEQ ID NO: 6)
    Cystatin Assay 6/12
    Cell Temp IPTG Bradford Overnight
    Type ° C. (mM) OD600 Final OD (ug/uL) 600 ng 60 ng 6 ng Δ
    BL21 16 0.5 0.1 5.298 0.061 0.272 0.238 0.236 −0.035
    16 0.5 0.8 6.749 0.097 0.229 0.231 0.223 −0.002
    16 0.05 0.1 8.848 0.093 0.277 0.246 0.244 −0.032
    16 0.05 0.8 12.435 0.137 0.270 0.247 0.233 −0.030
    25 0.5 0.1 3.038 0.830 0.234 0.206 0.207 −0.027
    25 0.5 0.8 3.237 0.145 0.222 0.206 0.189 −0.025
    25 0.05 0.1 5.452 0.188 0.240 0.226 0.221 −0.017
    25 0.05 0.8 4.577 0.241 0.226 0.208 0.195 −0.025
    BL21 Star 16 0.5 0.1 1.550 0.124 0.233 0.231 0.229 −0.003
    16 0.5 0.8 3.552 0.364 0.188 0.237 0.222 0.042
    16 0.05 0.1 1.906 0.088 0.215 0.218 0.212 0.000
    16 0.05 0.8 2.991 0.588 0.223 0.227 0.222 0.002
    25 0.5 0.1 3.575 0.112 0.184 0.198 0.196 0.013
    25 0.5 0.8 3.830 0.354 0.169 0.200 0.197 0.029
    25 0.05 0.1 3.459 0.150 0.141 0.235 0.202 0.077
    25 0.05 0.8 3.839 0.353 0.116 0.209 0.190 0.084
  • TABLE 28
    Cystatin Activity Results
    GST fusion tagged protein, second replicate
    ZmCys4 (SEQ ID NO: 6)
    Cystatin Assay 7/24
    Cell Temp IPTG Bradford Overnight
    Type ° C. (mM) OD600 Final OD (ug/uL) 600 ng 60 ng 6 ng Δ
    BL21 16 0.5 0.1 5.243 0.195 0.198 0.202 0.200 0.003
    16 0.5 0.8 7.029 0.112 0.194 0.187 0.321 0.060
    16 0.05 0.1 8.322 0.153 0.247 0.217 0.204 −0.037
    16 0.05 0.8 11.639 0.170 0.244 0.252 0.224 −0.006
    25 0.5 0.1 2.982 0.094 0.239 0.195 0.222 −0.031
    25 0.5 0.8 2.968 0.082 0.234 0.195 0.199 −0.036
    25 0.05 0.1 4.601 0.272 0.240 0.211 0.203 −0.033
    25 0.05 0.8 4.294 0.167 0.236 0.210 0.201 −0.030
    BL21 Star 16 0.5 0.1 1.744 0.082 0.162 0.198 0.200 0.037
    16 0.5 0.8 4.275 0.319 0.127 0.205 0.206 0.078
    16 0.05 0.1 2.002 0.154 0.165 0.214 0.191 0.038
    16 0.05 0.8 4.164 0.414 0.124 0.200 0.218 0.085
    25 0.5 0.1 3.167 0.812 0.216 0.192 0.190 −0.025
    25 0.5 0.8 3.965 0.237 0.092 0.208 0.191 0.107
    25 0.05 0.1 3.140 0.725 0.116 0.203 0.194 0.082
    25 0.05 0.8 3.478 0.273 0.097 0.191 0.194 0.095
  • TABLE 29
    Cystatin Activity Results
    His fusion tagged protein, first replicate
    ZmCys4 (SEQ ID NO: 6)
    Cystatin Assay 9/26 First Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (mM) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 16 0.05 0.1 1.850 0.226 0.274 0.193 −0.08 0.540 0.537 0.00
    16 1.0 0.1 1.804 0.211 0.268 0.192 −0.08 0.539 0.534 −0.01
    16 0.05 0.6 2.913 0.338 0.270 0.185 −0.09 0.540 0.538 0.00
    16 1.0 0.6 2.358 0.325 0.260 0.198 −0.06 0.536 0.532 0.00
    30 0.05 0.1 5.326 0.490 0.295 0.248 −0.05 0.524 0.526 0.00
    30 1.0 0.1 5.419 0.478 0.291 0.258 −0.03 0.518 0.538 0.02
    30 0.05 0.6 5.513 0.455 0.303 0.268 −0.04 0.529 0.537 0.01
    30 1.0 0.6 5.140 0.490 0.303 0.273 −0.03 0.536 0.538 0.00
    BL21 Star 16 0.05 0.1 1.297 0.045 0.082 0.194 0.11 0.402 0.535 0.13
    16 1.0 0.1 1.343 0.051 0.047 0.203 0.16 0.144 0.529 0.39
    16 0.05 0.6 2.358 0.213 0.045 0.205 0.16 0.123 0.539 0.42
    16 1.0 0.6 2.450 0.215 0.046 0.199 0.15 0.115 0.535 0.42
    30 0.05 0.1 5.094 0.467 0.053 0.265 0.21 0.120 0.538 0.42
    30 1.0 0.1 5.140 0.533 0.048 0.261 0.21 0.123 0.531 0.41
    30 0.05 0.6 4.768 0.400 0.265 0.278 0.01 0.545 0.535 −0.01
    30 1.0 0.6 4.768 0.488 0.052 0.274 0.22 0.119 0.531 0.41
  • TABLE 30
    Cystatin Activity Results
    His fusion tagged protein, second replicate
    ZmCys4 (SEQ ID NO: 6)
    Cystatin Assay 9/26 Second Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (mM) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 16 0.05 0.1 1.758 0.425 0.318 0.247 −0.07 0.536 0.545 0.01
    16 1.0 0.1 1.712 0.453 0.313 0.233 −0.08 0.537 0.539 0.00
    16 0.05 0.6 2.312 0.414 0.328 0.260 −0.07 0.539 0.540 0.00
    16 1.0 0.6 2.219 0.348 0.325 0.258 −0.07 0.543 0.546 0.00
    30 0.05 0.1 5.699 0.216 0.290 0.267 −0.02 0.561 0.569 0.01
    30 1.0 0.1 5.699 0.222 0.292 0.263 −0.03 0.559 0.566 0.01
    30 0.05 0.6 5.466 0.293 0.297 0.521
    30 1.0 0.6 5.979 0.316 0.294 0.530
    BL21 Star 16 0.05 0.1 1.297 0.427 0.265 0.234 −0.03 0.549 0.547 0.00
    16 1.0 0.1 1.251 0.446 0.235 0.232 0.00 0.536 0.536 0.00
    16 0.05 0.6 2.404 0.427 0.079 0.233 0.15 0.305 0.541 0.24
    16 1.0 0.6 2.589 0.453 0.063 0.222 0.16 0.243 0.542 0.30
    30 0.05 0.1 4.629 0.083 0.095 0.260 0.17 0.371 0.557 0.19
    30 1.0 0.1 4.118 0.109 0.102 0.269 0.17 0.426 0.558 0.13
    30 0.05 0.6 4.582 0.175 0.067 0.150
    30 1.0 0.6 5.280 0.354 0.068 0.272
  • TABLE 31
    Cystatin Activity Results
    GST fusion tagged protein, first replicate
    ZmCys6 (SEQ ID NO: 10)
    Cystatin Assay 9/26 First Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (Mm) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 16 0.05 0.1 2.358 0.093 0.293 0.211 −0.08 0.553 0.544 −0.01
    16 1.0 0.1 2.219 0.060 0.286 0.221 −0.07 0.552 0.561 0.01
    16 0.05 0.6 3.237 0.103 0.292 0.220 −0.07 0.549 0.554 0.01
    16 1.0 0.6 2.589 0.072 0.291 0.223 −0.07 0.558 0.557 0.00
    30 0.05 0.1 5.606 0.142 0.298 0.247 −0.05 0.552 0.549 0.00
    30 1.0 0.1 6.165 0.197 0.289 0.248 −0.04 0.539 0.554 0.02
    30 0.05 0.6 5.746 0.166 0.296 0.256 −0.04 0.547 0.552 0.01
    30 1.0 0.6 6.445 0.170 0.305 0.262 −0.04 0.549 0.558 0.01
    BL21 Star 16 0.05 0.1 1.113 0.027 0.252 0.200 −0.05 0.559 0.545 −0.01
    16 1.0 0.1 0.975 0.037 0.268 0.210 −0.06 0.549 0.549 0.00
    16 0.05 0.6 1.758 0.128 0.266 0.213 −0.05 0.543 0.543 0.00
    16 1.0 0.6 1.758 0.170 0.279 0.214 −0.07 0.540 0.544 0.00
    30 0.05 0.1 4.396 0.632 0.266 0.226 −0.04 0.537 0.552 0.02
    30 1.0 0.1 4.257 0.516 0.269 0.231 −0.04 0.536 0.550 0.01
    30 0.05 0.6 4.954 0.676 0.274 0.241 −0.03 0.544 0.551 0.01
    30 1.0 0.6 4.536 0.710 0.268 0.237 −0.03 0.542 0.553 0.01
  • TABLE 32
    Cystatin Activity Results
    GST fusion tagged protein, second replicate
    ZmCys6 (SEQ ID NO: 10)
    Cystatin Assay 9/26 Second Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (Mm) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 16 0.05 0.1 1.896 0.068 0.252 0.213 −0.04 0.548 0.548 0.00
    16 1.0 0.1 1.896 0.072 0.271 0.222 −0.05 0.555 0.550 −0.01
    16 0.05 0.6 2.635 0.116 0.291 0.212 −0.08 0.549 0.545 0.00
    16 1.0 0.6 2.266 0.076 0.291 0.232 −0.06 0.550 0.549 0.00
    30 0.05 0.1 5.932 0.174 0.335 0.274 −0.06 0.540 0.542 0.00
    30 1.0 0.1 6.212 0.178 0.350 0.307 −0.04 0.544 0.546 0.00
    30 0.05 0.6 5.746 0.145 0.337 0.283 −0.05 0.553 0.540 −0.01
    30 1.0 0.6 6.259 0.178 0.366 0.302 −0.06 0.547 0.542 −0.01
    BL21 Star 16 0.05 0.1 0.836 0.024 0.233 0.207 −0.03 0.566 0.550 −0.02
    16 1.0 0.1 0.882 0.025 0.232 0.210 −0.02 0.565 0.551 −0.01
    16 0.05 0.6 1.389 0.153 0.265 0.201 −0.06 0.543 0.535 −0.01
    16 1.0 0.6 1.389 0.159 0.282 0.205 −0.08 0.528 0.538 0.01
    30 0.05 0.1 5.001 0.657 0.355 0.312 −0.04 0.545 0.543 0.00
    30 1.0 0.1 4.768 0.594 0.357 0.311 −0.05 0.539 0.542 0.00
    30 0.05 0.6 4.489 0.63 0.375 0.297 −0.08 0.539 0.532 −0.01
    30 1.0 0.6 4.582 0.68 0.384 0.231 −0.15 0.547 0.511 −0.04
  • TABLE 33
    Cystatin Activity Results
    His fusion tagged protein, first replicate
    ZmCys6 (SEQ ID NO: 10)
    Cystatin Assay 9/26 First Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (Mm) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 16 0.05 0.1 1.942 0.292 0.282 0.202 −0.08 0.546 0.549 0.00
    16 1.0 0.1 2.035 0.238 0.266 0.199 −0.07 0.537 0.542 0.01
    16 0.05 0.6 2.959 0.338 0.274 0.213 −0.06 0.542 0.552 0.01
    16 1.0 0.6 2.913 0.300 0.284 0.197 −0.09 0.544 0.538 −0.01
    30 0.05 0.1 5.373 0.416 0.219 0.233 0.01 0.540 0.546 0.01
    30 1.0 0.1 4.954 0.452 0.263 0.231 −0.03 0.533 0.548 0.02
    30 0.05 0.6 5.419 0.448 0.275 0.245 −0.03 0.544 0.553 0.01
    30 1.0 0.6 5.373 0.459 0.275 0.236 −0.04 0.542 0.553 0.01
    BL21 Star 16 0.05 0.1 1.297 0.093 0.290 0.210 −0.08 0.542 0.543 0.00
    16 1.0 0.1 1.343 0.091 0.287 0.218 −0.07 0.542 0.552 0.01
    16 0.05 0.6 2.266 0.284 0.264 0.206 −0.06 0.536 0.542 0.01
    16 1.0 0.6 2.266 0.253 0.274 0.220 −0.05 0.532 0.545 0.01
    30 0.05 0.1 4.675 0.463 0.214 0.228 0.01 0.548 0.550 0.00
    30 1.0 0.1 5.094 0.490 0.252 0.226 −0.03 0.526 0.539 0.01
    30 0.05 0.6 4.814 0.402 0.263 0.230 −0.03 0.538 0.546 0.01
    30 1.0 0.6 4.861 0.452 0.266 0.226 −0.04 0.540 0.545 0.01
  • TABLE 34
    Cystatin Activity Results
    His fusion tagged protein, second replicate
    ZmCys6 (SEQ ID NO: 10)
    Cystatin Assay 9/26 Second Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (Mm) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 16 0.05 0.1 1.758 0.267 0.270 0.199 −0.07 0.539 0.541 0.00
    16 1.0 0.1 1.666 0.25 0.271 0.196 −0.08 0.544 0.541 0.00
    16 0.05 0.6 2.635 0.306 0.277 0.187 −0.09 0.539 0.532 −0.01
    16 1.0 0.6 2.497 0.286 0.276 0.190 −0.09 0.532 0.528 0.00
    30 0.05 0.1 5.979 0.448 0.340 0.299 −0.04 0.541 0.546 0.01
    30 1.0 0.1 5.979 0.414 0.349 0.303 −0.05 0.536 0.542 0.01
    30 0.05 0.6 6.072 0.436 0.356 0.296 −0.06 0.540 0.538 0.00
    30 1.0 0.6 6.072 0.431 0.366 0.303 −0.06 0.533 0.536 0.00
    BL21 Star 16 0.05 0.1 1.113 0.207 0.258 0.206 −0.05 0.552 0.540 −0.01
    16 1.0 0.1 1.159 0.132 0.267 0.220 −0.05 0.543 0.546 0.00
    16 0.05 0.6 2.219 0.257 0.253 0.205 −0.05 0.526 0.533 0.01
    16 1.0 0.6 2.081 0.186 0.284 0.219 −0.07 0.531 0.534 0.00
    30 0.05 0.1 5.885 0.452 0.362 0.324 −0.04 0.535 0.538 0.00
    30 1.0 0.1 6.259 0.471 0.341 0.284 −0.06 0.530 0.533 0.00
    30 0.05 0.6 5.140 0.509 0.379 0.305 −0.07 0.529 0.534 0.01
    30 1.0 0.6 5.652 0.476 0.345 0.269 −0.08 0.528 0.524 0.00
  • TABLE 35
    Cystatin Activity Results
    GST fusion tagged protein, first replicate
    ZmCys7 (SEQ ID NO: 12)
    Cystatin Assay 9/26 First Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (Mm) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 16 0.05 0.1 2.127 0.078 0.295 0.245 −0.05 0.550 0.565 0.01
    16 1.0 0.1 2.127 0.072 0.291 0.224 −0.07 0.557 0.555 0.00
    16 0.05 0.6 3.191 0.224 0.291 0.221 −0.07 0.544 0.543 0.00
    16 1.0 0.6 2.959 0.261 0.281 0.225 −0.06 0.544 0.547 0.00
    30 0.05 0.1 5.606 0.353 0.314 0.261 −0.05 0.559 0.551 −0.01
    30 1.0 0.1 6.025 0.370 0.320 0.266 −0.05 0.554 0.553 0.00
    30 0.05 0.6 5.979 0.313 0.335 0.262 −0.07 0.533 0.532 0.00
    30 1.0 0.6 6.445 0.277 0.328 0.268 −0.06 0.529 0.537 0.01
    BL21 Star 16 0.05 0.1 1.067 0.093 0.218 0.202 −0.02 0.552 0.538 −0.01
    16 1.0 0.1 0.975 0.097 0.173 0.209 0.04 0.525 0.540 0.02
    16 0.05 0.6 1.896 0.375 0.042 0.207 0.17 0.068 0.537 0.47
    16 1.0 0.6 1.758 0.373 0.040 0.208 0.17 0.065 0.542 0.48
    30 0.05 0.1 4.025 0.600 0.047 0.224 0.18 0.069 0.539 0.47
    30 1.0 0.1 4.536 0.621 0.045 0.219 0.17 0.064 0.532 0.47
    30 0.05 0.6 4.164 0.594 0.042 0.252 0.21 0.058 0.535 0.48
    30 1.0 0.6 4.350 0.602 0.042 0.259 0.22 0.057 0.545 0.49
  • TABLE 36
    Cystatin Activity Results
    GST fusion tagged protein, second replicate
    ZmCys7 (SEQ ID NO: 12)
    Cystatin Assay 9/26 Second Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (Mm) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 16 0.05 0.1 1.942 0.19 0.308 0.251 −0.06 0.548 0.536 −0.01
    16 1.0 0.1 1.804 0.196 0.292 0.240 −0.05 0.538 0.539 0.00
    16 0.05 0.6 2.774 0.167 0.321 0.248 −0.07 0.544 0.536 −0.01
    16 1.0 0.6 2.404 0.152 0.310 0.245 −0.07 0.531 0.544 0.01
    30 0.05 0.1 6.072 0.054 0.327 0.262 −0.07 0.534 0.556 0.02
    30 1.0 0.1 6.539 0.051 0.319 0.266 −0.05 0.529 0.562 0.03
    30 0.05 0.6 5.932 0.075 0.329 0.273 −0.06 0.567 0.561 −0.01
    30 1.0 0.6 6.585 0.086 0.320 0.276 −0.04 0.556 0.572 0.02
    BL21 Star 16 0.05 0.1 0.928 0.713 0.235 0.210 −0.03 0.551 0.532 −0.02
    16 1.0 0.1 0.928 0.425 0.228 0.225 0.00 0.553 0.547 −0.01
    16 0.05 0.6 1.666 0.785 0.149 0.227 0.08 0.489 0.543 0.05
    16 1.0 0.6 1.527 0.421 0.056 0.204 0.15 0.140 0.529 0.39
    30 0.05 0.1 4.257 0.019 0.043 0.244 0.20 0.061 0.545 0.48
    30 1.0 0.1 4.443 0.021 0.041 0.251 0.21 0.065 0.556 0.49
    30 0.05 0.6 4.489 0.077 0.044 0.246 0.20 0.060 0.562 0.50
    30 1.0 0.6 4.350 0.105 0.045 0.250 0.21 0.062 0.550 0.49
  • TABLE 37
    Cystatin Activity Results
    His fusion tagged protein, first replicate
    ZmCys7 (SEQ ID NO: 12)
    Cystatin Assay 9/26 First Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (Mm) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 16 0.05 0.1 1.989 0.275 0.287 0.227 −0.06 0.542 0.555 0.01
    16 1.0 0.1 1.850 0.244 0.282 0.194 −0.09 0.545 0.529 −0.02
    16 0.05 0.6 2.404 0.311 0.323 0.206 −0.12 0.536 0.541 0.01
    16 1.0 0.6 2.266 0.280 0.271 0.187 −0.08 0.531 0.531 0.00
    30 0.05 0.1 5.652 0.396 0.279 0.248 −0.03 0.544 0.549 0.01
    30 1.0 0.1 5.326 0.461 0.265 0.236 −0.03 0.539 0.539 0.00
    30 0.05 0.6 5.280 0.486 0.301 0.268 −0.03 0.519 0.537 0.02
    30 1.0 0.6 5.187 0.448 0.304 0.262 −0.04 0.523 0.534 0.01
    BL21 Star 16 0.05 0.1 1.159 0.122 0.147 0.211 0.06 0.503 0.539 0.04
    16 1.0 0.1 1.251 0.112 0.220 0.211 −0.01 0.540 0.535 −0.01
    16 0.05 0.6 2.219 0.263 0.043 0.215 0.17 0.076 0.541 0.47
    16 1.0 0.6 2.266 0.358 0.058 0.203 0.15 0.148 0.520 0.37
    30 0.05 0.1 5.187 0.554 0.175 0.236 0.06 0.517 0.543 0.03
    30 1.0 0.1 5.094 0.537 0.212 0.230 0.02 0.521 0.535 0.01
    30 0.05 0.6 5.187 0.020 0.196 0.271 0.08 0.504 0.534 0.03
    30 1.0 0.6 4.768 0.520 0.234 0.265 0.03 0.506 0.535 0.03
  • TABLE 38
    Cystatin Activity Results
    His fusion tagged protein, second replicate
    ZmCys7 (SEQ ID NO: 12)
    Cystatin Assay 9/26 Second Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (Mm) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 16 0.05 0.1 1.804 0.482 0.286 0.222 −0.06 0.529 0.529 0.00
    16 1.0 0.1 1.804 0.453 0.295 0.212 −0.08 0.532 0.527 −0.01
    16 0.05 0.6 2.266 0.463 0.291 0.220 −0.07 0.532 0.533 0.00
    16 1.0 0.6 2.358 0.491 0.288 0.222 −0.07 0.527 0.537 0.01
    30 0.05 0.1 5.606 0.237 0.298 0.263 −0.04 0.530 0.562 0.03
    30 1.0 0.1 5.094 0.224 0.290 0.255 −0.04 0.530 0.540 0.01
    30 0.05 0.6 5.513 0.32 0.301 0.264 −0.04 0.558 0.565 0.01
    30 1.0 0.6 5.513 0.337 0.295 0.266 −0.03 0.554 0.534 −0.02
    BL21 Star 16 0.05 0.1 1.021 0.47 0.214 0.227 0.01 0.549 0.540 −0.01
    16 1.0 0.1 1.113 0.485 0.238 0.224 −0.01 0.544 0.540 0.00
    16 0.05 0.6 2.127 0.431 0.051 0.220 0.17 0.110 0.539 0.43
    16 1.0 0.6 1.989 0.395 0.088 0.219 0.13 0.325 0.535 0.21
    30 0.05 0.1 5.280 0.036 0.173 0.258 0.09 0.504 0.548 0.04
    30 1.0 0.1 5.140 0.049 0.230 0.262 0.03 0.515 0.553 0.04
    30 0.05 0.6 5.373 0.164 0.193 0.265 0.07 0.535 0.552 0.02
    30 1.0 0.6 5.187 0.203 0.208 0.268 0.06 0.533 0.549 0.02
  • TABLE 39
    Cystatin Activity Results
    GST fusion tagged protein, first and second replicates
    OsCys6 (SEQ ID NO: 56)
    Cystatin Assay 10/22 First Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (Mm) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 Star 16 0.05 0.1 1.758 −0.018 0.651 0.482 −0.169 1.235 1.4 0.165
    16 1.0 0.1 1.067 −0.025 0.638 0.49 −0.148 1.286 1.382 0.096
    16 0.05 0.6 2.45 0.058 0.701 0.477 −0.224 1.222 1.333 0.111
    16 1.0 0.6 1.758 0.015 0.657 0.427 −0.230 1.239 1.103 −0.136
    30 0.05 0.1 5.094 0.313 0.057 0.494 0.437 0.14 1.037 0.897
    30 1.0 0.1 3.052 0.196 0.051 0.489 0.438 0.126 1.204 1.078
    30 0.05 0.6 2.682 0.276 0.06 0.457 0.397 0.117 1.002 0.885
    30 1.0 0.6 2.589 0.191 0.053 0.486 0.433 0.12 1.402 1.282
    Cystatin Assay 10/22 Second Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (mM) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 Star 16 0.05 0.1 1.573 −0.016 0.501 0.38 −0.121 1.231 1.36 0.129
    16 1.0 0.1 1.021 −0.025 0.447 0.361 −0.086 1.269 1.176 −0.093
    16 0.05 0.6 2.035 0.076 0.525 0.406 −0.119 1.386 1.689 0.303
    16 1.0 0.6 1.85 −0.034 0.477 0.367 −0.110 1.456 1.325 −0.131
    30 0.05 0.1 3.237 0.287 0.061 0.453 0.392 0.129 1.198 1.069
    30 1.0 0.1 3.283 0.142 0.077 0.426 0.349 0.288 1.39 1.102
    30 0.05 0.6 3.607 0.268 0.06 0.462 0.402 0.116 1.242 1.126
    30 1.0 0.6 3.283 −0.004 0.415 0.385 −0.030 1.114 1.188 0.074
  • TABLE 40
    Cystatin Activity Results
    His fusion tagged protein, first and second replicates
    OsCys6 (SEQ ID NO: 56)
    Cystatin Assay 10/22 First Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (Mm) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 Star 16 0.05 0.1 1.113 0.004 0.429 0.389 −0.040 1.107 1.284 0.177
    16 1.0 0.1 1.251 0.024 0.297 0.38 0.083 1.291 1.32 0.029
    16 0.05 0.6 2.173 0.265 0.056 0.389 0.333 0.111 1.148 1.037
    16 1.0 0.6 2.358 −0.034 0.359 0.378 0.019 1.18 1.293 0.113
    30 0.05 0.1 4.675 0.462 0.567 0.453 −0.114 1.123 1.294 0.171
    30 1.0 0.1 4.629 0.429 0.599 0.459 −0.140 1.346 1.45 0.104
    30 0.05 0.6 6.305 0.374 0.585 0.455 −0.130 1.321 1.282 −0.039
    30 1.0 0.6 3.932 0.368 0.559 0.447 −0.112 1.484 1.359 −0.125
    Cystatin Assay 10/22 Second Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (mM) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 Star 16 0.05 0.1 1.021 −0.01 0.479 0.427 −0.052 1.252 1.597 0.345
    16 1.0 0.1 1.159 −0.013 0.405 0.368 −0.037 1.187 1.311 0.124
    16 0.05 0.6 1.896 0.21 0.056 0.424 0.368 0.116 1.326 1.210
    16 1.0 0.6 2.035 0.216 0.07 0.384 0.314 0.126 1.392 1.266
    30 0.05 0.1 3.654 0.394 0.609 0.499 −0.110 1.22 1.291 0.071
    30 1.0 0.1 3.607 0.286 0.599 0.503 −0.096 1.314 1.409 0.095
    30 0.05 0.6 3.468 0.292 0.599 0.51 −0.089 1.693 1.392 −0.301
    30 1.0 0.6 3.329 0.347 0.522 0.495 −0.027 1.321 1.721 0.400
  • TABLE 41
    Cystatin Activity Results
    GST fusion tagged protein, first and second replicates
    GmCys5 (SEQ ID NO: 36)
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (mM) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    Cystatin Assay 10/22 First Replicate
    BL21 Star 16 0.05 0.1 1.758 −0.013 0.654 0.423 −0.231 1.163 1.368 0.205
    16 1.0 0.1 1.343 −0.016 0.608 0.468 −0.140 1.241 1.252 0.011
    16 0.05 0.6 1.896 0.04 0.662 0.454 −0.208 1.244 1.304 0.060
    16 1.0 0.6 1.804 0.085 0.656 0.438 −0.218 1.176 1.309 0.133
    30 0.05 0.1 2.959 0.174 0.288 0.494 0.206 1.195 1.382 0.187
    30 1.0 0.1 2.867 0.134 0.359 0.491 0.132 0.985 1.17 0.185
    30 0.05 0.6 3.422 0.147 0.299 0.492 0.193 1.309 1.434 0.125
    30 1.0 0.6 3.747 0.119 0.362 0.446 0.084 1.126 0.993 −0.133
    Cystatin Assay 10/22 Second Replicate
    BL21 Star 16 0.05 0.1 1.021 0.004 0.463 0.347 −0.116 1.185 1.256 0.071
    16 1.0 0.1 1.389 −0.002 0.496 0.342 −0.154 1.48 1.099 −0.381
    16 0.05 0.6 2.127 0.036 0.479 0.351 −0.128 1.17 1.417 0.247
    16 1.0 0.6 2.081 0.044 0.486 0.35 −0.136 1.067 1.248 0.181
    30 0.05 0.1 3.237 0.187 0.114 0.425 0.311 0.731 1.534 0.803
    30 1.0 0.1 3.052 0.162 0.126 0.38 0.254 0.717 1.194 0.477
    30 0.05 0.6 3.515 0.113 0.206 0.428 0.222 1.214 1.437 0.223
    30 1.0 0.6 2.635 0.091 0.182 0.392 0.210 1.063 1.279 0.216
  • TABLE 42
    Cystatin Activity Results
    His fusion tagged protein, first and second replicates
    GmCys5 (SEQ ID NO: 36)
    Cystatin Assay 10/22 First Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (Mm) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 Star 16 0.05 0.1 1.251 −0.006 0.451 0.362 −0.089 1.166 1.283 0.117
    16 1.0 0.1 2.219 −0.015 0.442 0.356 −0.086 1.19 1.143 −0.047
    16 0.05 0.6 2.404 0.218 0.266 0.374 0.108 1.118 1.124 0.006
    16 1.0 0.6 2.543 0.193 0.238 0.357 0.119 1.336 1.344 0.008
    30 0.05 0.1 3.561 0.345 0.586 0.431 −0.155 1.355 1.374 0.019
    30 1.0 0.1 5.932 0.38 0.569 0.435 −0.134 1.339 1.308 −0.031
    30 0.05 0.6 4.303 0.386 0.576 0.435 −0.141 1.29 1.521 0.231
    30 1.0 0.6 4.303 0.372 0.555 0.388 −0.167 1.06 1.048 −0.012
    Cystatin Assay 10/22 Second Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (mM) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 Star 16 0.05 0.1 1.113 −0.021 0.465 0.364 −0.101 1.273 1.347 0.074
    16 1.0 0.1 1.021 −0.023 0.467 0.359 −0.108 1.232 1.176 −0.056
    16 0.05 0.6 2.312 0.165 0.306 0.38 0.074 1.323 1.369 0.046
    16 1.0 0.6 2.173 0.206 0.202 0.388 0.186 0.973 1.686 0.713
    30 0.05 0.1 3.468 0.284 0.581 0.478 −0.103 1.223 1.173 −0.050
    30 1.0 0.1 3.376 0.286 0.593 0.422 −0.171 1.541 1.766 0.225
    30 0.05 0.6 3.098 0.276 0.61 0.49 −0.120 1.265 1.205 −0.060
    30 1.0 0.6 2.867 0.255 0.594 0.445 −0.149 1.784 1.802 0.018
  • TABLE 43
    Cystatin Activity Results
    GST fusion tagged protein, first and second replicates
    GmCys7 (SEQ ID NO: 40)
    Cystatin Assay 10/22 First Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (Mm) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 Star 16 0.05 0.1 1.435 −0.013 0.609 0.452 −0.157 1.179 1.263 0.084
    16 1.0 0.1 1.113 −0.02 0.562 0.433 −0.129 1.155 1.184 0.029
    16 0.05 0.6 1.573 0.067 0.556 0.361 −0.195 1.187 1.191 0.004
    16 1.0 0.6 1.573 0.053 0.52 0.376 −0.144 1.171 1.231 0.060
    30 0.05 0.1 3.515 0.274 0.246 0.501 0.255 1.164 1.217 0.053
    30 1.0 0.1 2.867 0.17 0.336 0.45 0.114 1.295 1.061 −0.234
    30 0.05 0.6 3.422 0.225 0.216 0.461 0.245 1.05 1.126 0.076
    30 1.0 0.6 2.82 0.179 0.258 0.446 0.188 1.156 1.069 −0.087
    Cystatin Assay 10/22 Second Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (mM) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 Star 16 0.05 0.1 0.975 −0.019 0.525 0.404 −0.121 1.204 1.393 0.189
    16 1.0 0.1 1.389 −0.034 0.492 0.366 −0.126 1.343 1.333 −0.010
    16 0.05 0.6 1.666 0.054 0.512 0.402 −0.110 1.298 1.29 −0.008
    16 1.0 0.6 1.573 0.024 0.486 0.374 −0.112 1.333 1.285 −0.048
    30 0.05 0.1 3.654 0.216 0.212 0.482 0.270 1.115 1.348 0.233
    30 1.0 0.1 3.098 0.104 0.3 0.475 0.175 1.222 1.297 0.075
    30 0.05 0.6 3.191 0.245 0.236 0.445 0.209 1.244 1.262 0.018
    30 1.0 0.6 2.82 0.145 0.322 0.462 0.140 1.185 1.326 0.141
  • TABLE 44
    Cystatin Activity Results
    His fusion tagged protein, first and second replicates
    GmCys7 (SEQ ID NO: 40)
    Cystatin Assay 10/22 First Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (Mm) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 Star 16 0.05 0.1 1.113 0 0.486 0.403 −0.083 1.134 1.185 0.051
    16 1.0 0.1 1.159 −0.017 0.423 0.374 −0.049 1.051 1.178 0.127
    16 0.05 0.6 2.266 0.12 0.387 0.397 0.010 1.075 1.144 0.069
    16 1.0 0.6 2.404 0.094 0.289 0.367 0.078 1.858 1.2 −0.658
    30 0.05 0.1 3.886 0.429 0.475 0.515 0.040 1.277 1.771 0.494
    30 1.0 0.1 3.515 0.411 0.471 0.479 0.008 1.318 1.317 −0.001
    30 0.05 0.6 3.747 0.405 0.478 0.456 −0.022 1.499 1.364 −0.135
    30 1.0 0.6 4.21 0.386 0.443 0.479 0.036 1.475 1.615 0.140
    Cystatin Assay 10/22 Second Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (mM) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 Star 16 0.05 0.1 0.975 0 0.571 0.473 −0.098 1.236 1.18 −0.056
    16 1.0 0.1 1.159 −0.011 0.54 0.438 −0.102 1.259 1.11 −0.149
    16 0.05 0.6 1.942 0.087 0.453 0.429 −0.024 1.035 1.078 0.043
    16 1.0 0.6 1.942 0.067 0.345 0.446 0.101 1.033 1.178 0.145
    30 0.05 0.1 3.283 0.322 0.641 0.539 −0.102 1.249 1.331 0.082
    30 1.0 0.1 3.376 0.308 0.622 0.54 −0.082 1.345 1.453 0.108
    30 0.05 0.6 3.329 0.285 0.579 0.552 −0.027 1.263 1.36 0.097
    30 1.0 0.6 3.329 0.302 0.605 0.503 −0.102 1.551 1.294 −0.257
  • TABLE 45
    Cystatin Activity Results
    GST fusion tagged protein, first and second replicates
    GmCys9 (SEQ ID NO: 44)
    Cystatin Assay 10/22 First Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (Mm) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 Star 16 0.05 0.1 1.021 −0.016 0.591 0.434 −0.157 1.131 1.066 −0.065
    16 1.0 0.1 1.067 −0.018 0.586 0.45 −0.136 1.069 1.173 0.104
    16 0.05 0.6 3.237 0.056 0.541 0.454 −0.087 1.178 1.196 0.018
    16 1.0 0.6 2.127 0.053 0.543 0.452 −0.091 1.111 1.256 0.145
    30 0.05 0.1 3.144 0.187 0.066 0.501 0.435 0.194 1.236 1.042
    30 1.0 0.1 3.191 −0.002 0.44 0.471 0.031 1.267 1.162 −0.105
    30 0.05 0.6 3.607 0.153 0.09 0.554 0.464 0.371 1.364 0.993
    30 1.0 0.6 2.728 0.094 0.217 0.481 0.264 1.007 1.203 0.196
    Cystatin Assay 10/22 Second Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (mM) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 Star 16 0.05 0.1 1.343 −0.007 0.477 0.371 −0.106 1.416 1.062 −0.354
    16 1.0 0.1 1.113 −0.013 0.444 0.362 −0.082 1.286 1.167 −0.119
    16 0.05 0.6 1.989 0.033 0.501 0.405 −0.096 1.036 1.251 0.215
    16 1.0 0.6 2.035 0.011 0.471 0.403 −0.068 1.222 1.511 0.289
    30 0.05 0.1 3.747 0.238 0.063 0.455 0.392 0.145 1.119 0.974
    30 1.0 0.1 5.094 0.134 0.123 0.458 0.335 0.725 1.233 0.508
    30 0.05 0.6 3.376 0.151 0.131 0.496 0.365 0.73 1.314 0.584
    30 1.0 0.6 3.005 0.085 0.236 0.456 0.220 1.048 1.32 0.272
  • TABLE 46
    Cystatin Activity Results
    His fusion tagged protein, first and second replicates
    GmCys9 (SEQ ID NO: 44)
    Cystatin Assay 10/22 First Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (Mm) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 Star 16 0.05 0.1 1.113 0.007 0.476 0.402 −0.074 1.148 1.388 0.240
    16 1.0 0.1 1.113 0 0.295 0.393 0.098 1.246 1.421 0.175
    16 0.05 0.6 2.45 0.206 0.056 0.405 0.349 0.137 1.084 0.947
    16 1.0 0.6 2.312 0.201 0.059 0.404 0.345 0.114 1.286 1.172
    30 0.05 0.1 5.047 0.417 0.497 0.469 −0.028 1.369 1.199 −0.170
    30 1.0 0.1 3.839 0.392 0.558 0.474 −0.084 1.263 1.429 0.166
    30 0.05 0.6 4.443 0.382 0.423 0.489 0.066 1.115 1.221 0.106
    30 1.0 0.6 3.839 0.321 0.432 0.496 0.064 0.937 1.26 0.323
    Cystatin Assay 10/22 Second Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (mM) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 Star 16 0.05 0.1 1.297 0.014 0.613 0.495 −0.118 1.303 1.166 −0.137
    16 1.0 0.1 1.067 0.012 0.381 0.513 0.132 1.081 1.422 0.341
    16 0.05 0.6 1.942 0.236 0.08 0.477 0.397 0.33 1.115 0.785
    16 1.0 0.6 2.081 0.259 0.054 0.474 0.420 0.114 1.228 1.114
    30 0.05 0.1 3.561 0.397 0.292 0.53 0.238 1.077 1.269 0.192
    30 1.0 0.1 3.376 0.304 0.48 0.501 0.021 1.195 1.305 0.110
    30 0.05 0.6 3.144 0.257 0.364 0.511 0.147 1.372 1.222 −0.150
    30 1.0 0.6 3.191 0.332 0.383 0.491 0.108 1.291 1.293 0.002
  • TABLE 47
    Cystatin Activity Results
    GST fusion tagged protein, first and second replicates
    TaCys8 (SEQ ID NO: 68)
    Cystatin Assay 10/22 First Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (Mm) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 Star 16 0.05 0.1 1.481 −0.007 0.608 0.392 −0.216 1.323 1.516 0.193
    16 1.0 0.1 1.021 −0.007 0.587 0.428 −0.159 1.272 1.198 −0.074
    16 0.05 0.6 2.173 0.076 0.546 0.417 −0.129 0.95 1.197 0.247
    16 1.0 0.6 1.896 0.04 0.537 0.433 −0.104 1.338 1.188 −0.150
    30 0.05 0.1 3.005 0.208 0.121 0.471 0.350 0.879 1.15 0.271
    30 1.0 0.1 2.774 0.166 0.191 0.444 0.253 1.025 1.156 0.131
    30 0.05 0.6 4.536 0.183 0.134 0.46 0.326 0.985 1.058 0.073
    30 1.0 0.6 2.913 0.159 0.142 0.448 0.306 0.838 1.037 0.199
    Cystatin Assay 10/22 Second Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (mM) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 Star 16 0.05 0.1 1.343 −0.002 0.45 0.34 −0.110 1.227 1.191 −0.036
    16 1.0 0.1 1.62 0.002 0.398 0.357 −0.041 1.092 1.304 0.212
    16 0.05 0.6 2.035 0.031 0.4 0.352 −0.048 1.118 1.364 0.246
    16 1.0 0.6 2.081 0.024 0.336 0.358 0.022 1.091 1.19 0.099
    30 0.05 0.1 3.515 0.272 0.095 0.406 0.311 0.753 1.103 0.350
    30 1.0 0.1 2.774 0.164 0.116 0.392 0.276 0.916 1.159 0.243
    30 0.05 0.6 3.468 0.191 0.117 0.433 0.316 0.818 1.184 0.366
    30 1.0 0.6 2.728 0.189 0.111 0.416 0.305 0.76 1.099 0.339
  • TABLE 48
    Cystatin Activity Results
    His fusion tagged protein, first and second replicates
    TaCys8 (SEQ ID NO: 68)
    Cystatin Assay 10/22 First Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (Mm) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 Star 16 0.05 0.1 1.389 −0.006 0.484 0.387 −0.097 1.28 1.614 0.334
    16 1.0 0.1 1.113 −0.008 0.417 0.354 −0.063 1.163 1.451 0.288
    16 0.05 0.6 2.266 0.158 0.38 0.414 0.034 1.21 1.428 0.218
    16 1.0 0.6 2.404 0.152 0.357 0.406 0.049 1.307 1.227 −0.080
    30 0.05 0.1 3.839 0.517 0.093 0.438 0.345 0.796 1.514 0.718
    30 1.0 0.1 3.839 0.53 0.097 0.416 0.319 0.78 1.245 0.465
    30 0.05 0.6 4.814 0.462 0.099 0.455 0.356 0.678 1.101 0.423
    30 1.0 0.6 3.839 0.415 0.092 0.422 0.330 0.676 0.99 0.314
    Cystatin Assay 10/22 Second Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (mM) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 Star 16 0.05 0.1 1.205 0 0.636 0.498 −0.138 1.461 1.677 0.216
    16 1.0 0.1 0.975 0 0.63 0.494 −0.136 1.463 1.354 −0.109
    16 0.05 0.6 2.127 0.197 0.339 0.455 0.116 0.98 1.102 0.122
    16 1.0 0.6 2.035 0.22 0.261 0.447 0.186 1.035 1.056 0.021
    30 0.05 0.1 3.839 0.277 0.386 0.503 0.117 1.165 1.225 0.060
    30 1.0 0.1 3.191 0.306 0.425 0.498 0.073 1.169 1.406 0.237
    30 0.05 0.6 3.7 0.297 0.223 0.479 0.256 1.097 1.226 0.129
    30 1.0 0.6 3.191 0.304 0.216 0.488 0.272 1.062 1.071 0.009
  • TABLE 49
    Cystatin Activity Results
    GST fusion tagged protein, first and second replicates
    ZmCys8 (SEQ ID NO: 14)
    Cystatin Assay 10/22 First Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (Mm) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 Star 16 0.05 0.1 1.666 −0.009 0.606 0.425 −0.181 1.435 1.288 −0.147
    16 1.0 0.1 0.882 −0.011 0.581 0.437 −0.144 1.283 1.369 0.086
    16 0.05 0.6 1.758 0.047 0.526 0.351 −0.175 1.027 1.126 0.099
    16 1.0 0.6 1.758 0.067 0.541 0.368 −0.173 1.168 1.156 −0.012
    30 0.05 0.1 2.589 0.213 0.484 0.464 −0.020 0.914 1.219 0.305
    30 1.0 0.1 2.635 0.174 0.508 0.444 −0.064 0.951 1.116 0.165
    30 0.05 0.6 2.173 0.189 0.463 0.416 −0.047 1.069 0.968 −0.101
    30 1.0 0.6 2.173 0.138 0.523 0.434 −0.089 1.349 1.404 0.055
    Cystatin Assay 10/22 Second Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (mM) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 Star 16 0.05 0.1 1.159 −0.017 0.49 0.348 −0.142 1.326 1.308 −0.018
    16 1.0 0.1 1.251 −0.013 0.495 0.37 −0.125 1.411 1.256 −0.155
    16 0.05 0.6 1.297 0.052 0.512 0.364 −0.148 1.306 1.447 0.141
    16 1.0 0.6 1.297 0.062 0.51 0.369 −0.141 1.365 1.387 0.022
    30 0.05 0.1 2.682 0.173 0.469 0.426 −0.043 1.212 1.153 −0.059
    30 1.0 0.1 2.543 0.149 0.544 0.447 −0.097 1.349 1.282 −0.067
    30 0.05 0.6 2.543 0.165 0.481 0.426 −0.055 1.146 1.359 0.213
    30 1.0 0.6 2.173 0.118 0.541 0.422 −0.119 1.57 1.603 0.033
  • TABLE 50
    Cystatin Activity Results
    His fusion tagged protein, first and second replicates
    ZmCys8 (SEQ ID NO: 14)
    Cystatin Assay 10/22 First Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (Mm) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 Star 16 0.05 0.1 1.527 −0.013 0.464 0.382 −0.082 1.168 1.229 0.061
    16 1.0 0.1 1.343 −0.021 0.413 0.368 −0.045 1.028 1.08 0.052
    16 0.05 0.6 2.543 0.099 0.487 0.368 −0.119 1.117 1.147 0.030
    16 1.0 0.6 2.358 0.064 0.486 0.366 −0.120 1.311 1.355 0.044
    30 0.05 0.1 4.025 0.403 0.557 0.464 −0.093 1.346 1.576 0.230
    30 1.0 0.1 5.28 0.446 0.548 0.445 −0.103 1.337 1.448 0.111
    30 0.05 0.6 4.071 0.353 0.517 0.466 −0.051 1.199 1.389 0.190
    30 1.0 0.6 3.376 0.308 0.491 0.438 −0.053 1.062 1.665 0.603
    Cystatin Assay 10/22 Second Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (mM) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 Star 16 0.05 0.1 0.975 −0.013 0.522 0.436 −0.086 1.283 1.163 −0.120
    16 1.0 0.1 0.975 −0.013 0.53 0.421 −0.109 1.314 1.256 −0.058
    16 0.05 0.6 1.942 0.161 0.527 0.427 −0.100 1.148 1.055 −0.093
    16 1.0 0.6 1.804 0.095 0.505 0.417 −0.088 1.071 1.075 0.004
    30 0.05 0.1 3.144 0.257 0.646 0.554 −0.092 1.176 1.318 0.142
    30 1.0 0.1 3.515 0.332 0.675 0.509 −0.166 1.541 1.5 −0.041
    30 0.05 0.6 2.774 0.214 0.661 0.505 −0.156 1.396 1.296 −0.100
    30 1.0 0.6 2.543 0.234 0.678 0.505 −0.173 1.532 1.418 −0.114
  • TABLE 51
    Cystatin Activity Results
    GST fusion tagged protein, first and second replicates
    ZmCys10 (SEQ ID NO: 18)
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (mM) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    Cystatin Assay 10/22 First Replicate
    BL21 Star 16 0.05 0.1 2.127 −0.027 0.581 0.463 −0.118 1.184 1.334 0.150
    16 1.0 0.1 1.481 −0.011 0.632 0.478 −0.154 1.324 1.385 0.061
    16 0.05 0.6 2.127 0.056 0.612 0.457 −0.155 1.252 1.243 −0.009
    16 1.0 0.6 2.219 0.047 0.668 0.464 −0.204 1.208 1.353 0.145
    30 0.05 0.1 4.35 0.1 0.622 0.482 −0.140 1.158 1.187 0.029
    30 1.0 0.1 4.443 0.106 0.629 0.483 −0.146 1.122 1.151 0.029
    30 0.05 0.6 3.7 0.072 0.592 0.466 −0.126 1.23 1.146 −0.084
    30 1.0 0.6 2.635 0.068 0.617 0.477 −0.140 1.29 1.268 −0.022
    Cystatin Assay 10/22 Second Replicate
    BL21 Star 16 0.05 0.1 1.021 −0.018 0.446 0.341 −0.105 1.12 1.218 0.098
    16 1.0 0.1 1.113 −0.009 0.473 0.352 −0.121 1.217 1.172 −0.045
    16 0.05 0.6 2.266 0.009 0.459 0.334 −0.125 1.494 1.18 −0.314
    16 1.0 0.6 2.127 0.038 0.486 0.358 −0.128 1.229 1.161 −0.068
    30 0.05 0.1 2.728 0.074 0.547 0.412 −0.135 1.193 1.136 −0.057
    30 1.0 0.1 2.959 0.083 0.512 0.403 −0.109 1.094 1.178 0.084
    30 0.05 0.6 2.173 0.047 0.528 0.424 −0.104 1.122 1.315 0.193
    30 1.0 0.6 2.358 0.045 0.564 0.425 −0.139 1.594 1.58 −0.014
  • TABLE 52
    Cystatin Activity Results
    His fusion tagged protein, first and second replicates
    ZmCys10 (SEQ ID NO: 18)
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (mM) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    Cystatin Assay 10/22 First Replicate
    BL21 Star 16 0.05 0.1 1.62 0.002 0.481 0.357 −0.124 1.291 1.21 −0.081
    16 1.0 0.1 1.113 0.011 0.468 0.336 −0.132 1.161 1.278 0.117
    16 0.05 0.6 2.497 0.214 0.499 0.365 −0.134 1.192 1.136 −0.056
    16 1.0 0.6 2.266 0.214 0.495 0.36 −0.135 1.175 1.211 0.036
    30 0.05 0.1 4.722 0.446 0.56 0.415 −0.145 1.233 1.295 0.062
    30 1.0 0.1 3.654 0.339 0.58 0.387 −0.193 1.2 1.276 0.076
    30 0.05 0.6 5.001 0.339 0.543 0.418 −0.125 1.14 1.254 0.114
    30 1.0 0.6 4.118 0.38 0.576 0.39 −0.186 1.291 1.29 −0.001
    Cystatin Assay 10/22 Second Replicate
    BL21 Star 16 0.05 0.1 1.021 −0.008 0.491 0.356 −0.135 1.137 1.392 0.255
    16 1.0 0.1 1.021 −0.004 0.434 0.353 −0.081 1.163 1.474 0.311
    16 0.05 0.6 1.896 0.287 0.54 0.386 −0.154 1.48 1.423 −0.057
    16 1.0 0.6 2.173 0.227 0.51 0.362 −0.148 1.242 1.362 0.120
    30 0.05 0.1 3.468 0.31 0.597 0.472 −0.125 1.308 1.232 −0.076
    30 1.0 0.1 3.329 0.325 0.559 0.469 −0.090 1.042 1.169 0.127
    30 0.05 0.6 2.959 0.272 0.632 0.485 −0.147 1.597 1.161 −0.436
    30 1.0 0.6 3.098 0.308 0.577 0.484 −0.093 1.119 1.107 −0.012
  • TABLE 53
    Cystatin Activity Results
    GST fusion tagged protein, first and second replicates
    ZmCys11 (SEQ ID NO: 20)
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (mM) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    Cystatin Assay 10/22 First Replicate
    BL21 Star 16 0.05 0.1 1.942 −0.016 0.598 0.443 −0.155 1.414 1.123 −0.291
    16 1.0 0.1 2.081 −0.042 0.581 0.456 −0.125 1.247 1.244 −0.003
    16 0.05 0.6 3.283 0.053 0.603 0.427 −0.176 1.136 1.193 0.057
    16 1.0 0.6 2.589 0.091 0.621 0.45 −0.171 1.125 1.282 0.157
    30 0.05 0.1 3.7 0.104 0.554 0.434 −0.120 1.192 1.171 −0.021
    30 1.0 0.1 4.025 0.085 0.557 0.454 −0.103 1.001 1.202 0.201
    30 0.05 0.6 3.098 0.074 0.485 0.446 −0.039 0.964 1.082 0.118
    30 1.0 0.6 2.913 0.089 0.529 0.46 −0.069 1.013 1.1 0.087
    Cystatin Assay 10/22 Second Replicate
    BL21 Star 16 0.05 0.1 1.067 −0.013 0.438 0.335 −0.103 1.093 1.296 0.203
    16 1.0 0.1 1.067 0.011 0.455 0.338 −0.117 1.167 1.015 −0.152
    16 0.05 0.6 2.404 0.082 0.461 0.331 −0.130 0.946 1.235 0.289
    16 1.0 0.6 2.404 0.013 0.46 0.358 −0.102 1.106 1.1 −0.006
    30 0.05 0.1 3.237 0.079 0.509 0.401 −0.108 1.149 1.099 −0.050
    30 1.0 0.1 3.793 0.07 0.506 0.404 −0.102 1.082 1.01 −0.072
    30 0.05 0.6 2.867 0.068 0.5 0.44 −0.060 1.264 1.201 −0.063
    30 1.0 0.6 3.747 0.072 0.433 0.442 0.009 1.293 1.236 −0.057
  • TABLE 54
    Cystatin Activity Results
    His fusion tagged protein, first and second replicates
    ZmCys11 (SEQ ID NO: 20)
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (mM) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    Cystatin Assay 10/22 First Replicate
    BL21 Star 16 0.05 0.1 1.712 0.024 0.467 0.372 −0.095 1.176 1.526 0.350
    16 1.0 0.1 1.159 0.404 0.456 0.364 −0.092 1.088 1.235 0.147
    16 0.05 0.6 2.682 0.265 0.492 0.431 −0.061 1.122 1.418 0.296
    16 1.0 0.6 2.312 0.319 0.479 0.454 −0.025 1.04 1.543 0.503
    30 0.05 0.1 4.768 0.433 0.514 0.439 −0.075 1.308 1.335 0.027
    30 1.0 0.1 3.468 0.356 0.527 0.435 −0.092 1.453 1.511 0.058
    30 0.05 0.6 3.839 0.339 0.525 0.461 −0.064 1.228 1.229 0.001
    30 1.0 0.6 3.283 0.272 0.523 0.454 −0.069 1.337 1.172 −0.165
    Cystatin Assay 10/22 Second Replicate
    BL21 Star 16 0.05 0.1 1.021 0.032 0.518 0.48 −0.038 1.326 1.467 0.141
    16 1.0 0.1 1.067 0.026 0.64 0.491 −0.149 1.298 1.255 −0.043
    16 0.05 0.6 1.989 0.344 0.519 0.431 −0.088 1.041 1.007 −0.034
    16 1.0 0.6 1.989 0.336 0.656 0.507 −0.149 1.222 1.288 0.066
    30 0.05 0.1 3.237 0.314 0.599 0.493 −0.106 1.147 1.117 −0.030
    30 1.0 0.1 2.913 0.318 0.646 0.501 −0.145 1.453 1.682 0.229
    30 0.05 0.6 3.237 0.275 0.599 0.49 −0.109 1.262 1.523 0.261
    30 1.0 0.6 3.561 0.269 0.621 0.483 −0.138 1.33 1.337 0.007
  • TABLE 55
    Cystatin Activity Results
    GST fusion tagged protein, first and second replicates
    ZmCys12 (SEQ ID NO: 22)
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (mM) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    Cystatin Assay 10/22 First Replicate
    BL21 Star 16 0.05 0.1 0.882 −0.02 0.552 0.428 −0.124 1.307 1.093 −0.214
    16 1.0 0.1 1.021 −0.007 0.561 0.434 −0.127 1.609 1.369 −0.240
    16 0.05 0.6 1.573 0.04 0.505 0.359 −0.146 1.18 1.41 0.230
    16 1.0 0.6 1.942 0.071 0.491 0.355 −0.136 1.171 1.108 −0.063
    30 0.05 0.1 4.814 0.198 0.165 0.455 0.290 0.881 1.121 0.240
    30 1.0 0.1 3.793 0.136 0.435 0.458 0.023 1.069 1.252 0.183
    30 0.05 0.6 2.173 0.234 0.257 0.415 0.158 0.994 1.083 0.089
    30 1.0 0.6 3.283 0.213 0.211 0.405 0.194 0.934 1.406 0.472
    Cystatin Assay 10/22 Second Replicate
    BL21 Star 16 0.05 0.1 1.067 −0.013 0.511 0.375 −0.136 1.372 1.221 −0.151
    16 1.0 0.1 0.882 −0.015 0.491 0.347 −0.144 1.234 1.209 −0.025
    16 0.05 0.6 1.527 0.041 0.477 0.367 −0.110 1.309 1.418 0.109
    16 1.0 0.6 2.404 0.037 0.468 0.348 −0.120 1.246 1.229 −0.017
    30 0.05 0.1 2.635 0.165 0.219 0.436 0.217 1.26 1.424 0.164
    30 1.0 0.1 3.144 0.132 0.417 0.462 0.045 1.773 1.297 −0.476
    30 0.05 0.6 3.654 0.12 0.322 0.414 0.092 1.559 1.443 −0.116
    30 1.0 0.6 3.607 0.106 0.389 0.437 0.048 1.27 1.249 −0.021
  • TABLE 56
    Cystatin Activity Results
    His fusion tagged protein, first and second replicates
    ZmCys12 (SEQ ID NO: 22)
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (mM) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    Cystatin Assay 10/22 First Replicate
    BL21 Star 16 0.05 0.1 1.297 −0.025 0.508 0.364 −0.144 1.289 1.036 −0.253
    16 1.0 0.1 1.067 −0.015 0.538 0.375 −0.163 1.589 1.006 −0.583
    16 0.05 0.6 2.035 0.186 0.49 0.365 −0.125 1.376 1.348 −0.028
    16 1.0 0.6 2.219 0.154 0.47 0.377 −0.093 1.276 1.171 −0.105
    30 0.05 0.1 3.932 0.425 0.423 0.47 0.047 1.345 1.489 0.144
    30 1.0 0.1 3.561 0.411 0.491 0.432 −0.059 1.141 1.016 −0.125
    30 0.05 0.6 3.839 0.513 0.321 0.458 0.137 1.676 1.602 −0.074
    30 1.0 0.6 3.747 0.38 0.396 0.456 0.060 1.118 1.771 0.653
    Cystatin Assay 10/22 Second Replicate
    BL21 Star 16 0.05 0.1 0.882 0.01 0.533 0.418 −0.115 1.266 1.19 −0.076
    16 1.0 0.1 1.021 0.01 0.553 0.435 −0.118 1.328 1.209 −0.119
    16 0.05 0.6 1.666 0.324 0.33 0.442 0.112 1.202 1.172 −0.030
    16 1.0 0.6 1.85 0.273 0.344 0.438 0.094 1.1 1.188 0.088
    30 0.05 0.1 2.497 0.299 0.136 0.493 0.357 1.092 1.511 0.419
    30 1.0 0.1 2.728 0.359 0.263 0.482 0.219 1.251 1.224 −0.027
    30 0.05 0.6 3.376 0.365 0.2 0.518 0.318 1.117 1.177 0.060
    30 1.0 0.6 2.589 0.336 0.15 0.453 0.303 0.962 1.2 0.238
  • TABLE 57
    Cystatin Activity Results
    GST fusion tagged protein, first and second replicates
    ZmCys13 (SEQ ID NO: 24)
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (mM) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    Cystatin Assay 10/22 First Replicate
    BL21 Star 16 0.05 0.1 2.82 0 0.701 0.5 −0.201 1.364 1.272 −0.092
    16 1.0 0.1 1.62 −0.011 0.66 0.512 −0.148 1.244 1.55 0.306
    16 0.05 0.6 3.191 0.071 0.672 0.478 −0.194 1.303 1.18 −0.123
    16 1.0 0.6 3.005 0.047 0.654 0.489 −0.165 1.286 1.261 −0.025
    30 0.05 0.1 4.025 0.387 0.056 0.473 0.417 0.135 1.148 1.013
    30 1.0 0.1 2.82 0.257 0.055 0.486 0.431 0.158 1.344 1.186
    30 0.05 0.6 4.35 0.347 0.053 0.472 0.419 0.126 1.727 1.601
    30 1.0 0.6 2.45 0.291 0.051 0.432 0.381 0.133 0.99 0.857
    Cystatin Assay 10/22 Second Replicate
    BL21 Star 16 0.05 0.1 1.527 0.011 0.473 0.36 −0.113 1.096 1.082 −0.014
    16 1.0 0.1 1.343 0.015 0.622 0.47 −0.152 1.004 1.124 0.120
    16 0.05 0.6 2.219 0.089 0.478 0.365 −0.113 1.07 1.225 0.155
    16 1.0 0.6 2.312 0.091 0.641 0.492 −0.149 1.158 1.236 0.078
    30 0.05 0.1 3.283 0.357 0.053 0.371 0.318 0.132 0.989 0.857
    30 1.0 0.1 3.932 0.249 0.056 0.406 0.350 0.142 1.265 1.123
    30 0.05 0.6 2.82 0.349 0.05 0.397 0.347 0.113 1.207 1.094
    30 1.0 0.6 4.443 0.323 0.054 0.42 0.366 0.121 1.324 1.203
  • TABLE 58
    Cystatin Activity Results
    His fusion tagged protein, first and second replicates
    ZmCys13 (SEQ ID NO: 24)
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (mM) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    Cystatin Assay 10/22 First Replicate
    BL21 Star 16 0.05 0.1 1.343 0.019 0.417 0.386 −0.031 1.141 1.314 0.173
    16 1.0 0.1 1.804 0.022 0.429 0.396 −0.033 1.205 1.318 0.113
    16 0.05 0.6 2.82 0.276 0.066 0.409 0.343 0.232 1.148 0.916
    16 1.0 0.6 3.005 0.231 0.068 0.4 0.332 0.239 1.155 0.916
    30 0.05 0.1 5.14 0.411 0.459 0.412 −0.047 1.052 1.256 0.204
    30 1.0 0.1 3.561 0.376 0.463 0.44 −0.023 1.205 1.383 0.178
    30 0.05 0.6 5.233 0.362 0.129 0.448 0.319 0.917 1.397 0.480
    30 1.0 0.6 3.422 0.358 0.132 0.433 0.301 0.895 1.308 0.413
    Cystatin Assay 10/22 Second Replicate
    BL21 Star 16 0.05 0.1 1.666 0.004 0.504 0.448 −0.056 1.094 1.401 0.307
    16 1.0 0.1 1.205 −0.013 0.424 0.385 −0.039 1.182 1.215 0.033
    16 0.05 0.6 2.543 0.291 0.064 0.475 0.411 0.241 1.301 1.060
    16 1.0 0.6 2.404 0.178 0.059 0.409 0.350 0.26 1.592 1.332
    30 0.05 0.1 3.376 0.368 0.446 0.45 0.004 1.052 1.036 −0.016
    30 1.0 0.1 4.396 0.394 0.457 0.415 −0.042 0.915 0.932 0.017
    30 0.05 0.6 4.164 0.364 0.174 0.474 0.300 0.8 1.355 0.555
    30 1.0 0.6 4.396 0.315 0.181 0.464 0.283 1.06 1.272 0.212
  • TABLE 59
    Cystatin Activity Results
    GST fusion tagged protein, first and second replicates
    ZmCys14 (SEQ ID NO: 26)
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (mM) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    Cystatin Assay 10/22 First Replicate
    BL21 Star 16 0.05 0.1 1.113 −0.018 0.604 0.477 −0.127 1.298 1.255 −0.043
    16 1.0 0.1 1.343 −0.016 0.643 0.493 −0.150 1.39 1.257 −0.133
    16 0.05 0.6 2.45 0.056 0.476 0.456 −0.020 1.185 1.314 0.129
    16 1.0 0.6 3.052 0.051 0.488 0.436 −0.052 1.025 1.012 −0.013
    30 0.05 0.1 3.654 0.245 0.082 0.459 0.377 0.836 1.257 0.421
    30 1.0 0.1 3.283 0.204 0.08 0.484 0.404 0.813 1.27 0.457
    30 0.05 0.6 5.326 0.264 0.062 0.436 0.374 0.592 1.122 0.530
    30 1.0 0.6 3.747 0.251 0.064 0.416 0.352 0.538 0.923 0.385
    Cystatin Assay 10/22 Second Replicate
    BL21 Star 16 0.05 0.1 1.989 0.002 0.426 0.348 −0.078 1.043 1.017 −0.026
    16 1.0 0.1 1.113 −0.002 0.587 0.46 −0.127 1.215 1.019 −0.196
    16 0.05 0.6 1.942 0.053 0.377 0.354 −0.023 1.092 0.95 −0.142
    16 1.0 0.6 2.913 0.022 0.526 0.488 −0.038 0.943 1.159 0.216
    30 0.05 0.1 3.144 0.289 0.061 0.374 0.313 0.532 0.949 0.417
    30 1.0 0.1 3.329 0.221 0.07 0.414 0.344 0.647 1.23 0.583
    30 0.05 0.6 4.768 0.274 0.059 0.43 0.371 0.512 1.246 0.734
    30 1.0 0.6 2.959 0.221 0.085 0.455 0.370 0.716 1.224 0.508
  • TABLE 60
    Cystatin Activity Results
    His fusion tagged protein, first and second replicates
    ZmCys14 (SEQ ID NO: 26)
    Cystatin Assay 10/22 First Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (Mm) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 Star 16 0.05 0.1 1.251 0.032 0.372 0.408 0.036 1.206 1.292 0.086
    16 1.0 0.1 1.251 0.017 0.41 0.396 −0.014 1.249 1.293 0.044
    16 0.05 0.6 2.497 0.265 0.13 0.539 0.409 0.764 1.31 0.546
    16 1.0 0.6 3.191 0.235 0.093 0.477 0.384 0.705 1.498 0.793
    30 0.05 0.1 3.515 0.481 0.065 0.453 0.388 0.602 1.365 0.763
    30 1.0 0.1 3.932 0.481 0.078 0.465 0.387 0.672 1.29 0.618
    30 0.05 0.6 5.047 0.394 0.069 0.454 0.385 0.504 1.465 0.961
    30 1.0 0.6 5.513 0.472 0.062 0.449 0.387 0.517 1.373 0.856
    Cystatin Assay 10/22 Second Replicate
    Cell Temp IPTG Bradford 1 hour Overnight
    Type ° C. (mM) OD600 Final OD (ug/uL) 6000 ng 60 ng Δ 6000 ng 60 ng Δ
    BL21 Star 16 0.05 0.1 1.021 0.022 0.518 0.541 0.023 1.22 1.407 0.187
    16 1.0 0.1 1.021 0.02 0.489 0.589 0.100 1.147 1.308 0.161
    16 0.05 0.6 2.035 0.306 0.092 0.499 0.407 0.785 1.188 0.403
    16 1.0 0.6 2.081 0.273 0.081 0.475 0.394 0.636 1.037 0.401
    30 0.05 0.1 3.607 0.324 0.17 0.489 0.319 0.881 1.484 0.603
    30 1.0 0.1 3.561 0.359 0.181 0.496 0.315 0.941 1.592 0.651
    30 0.05 0.6 3.515 0.365 0.065 0.462 0.397 0.566 1.169 0.603
    30 1.0 0.6 3.237 0.355 0.076 0.485 0.409 0.64 1.223 0.583
  • EXAMPLE 9 Microinjection Assay for Anti-Nematodal Activity of Cystatins
  • Two of the cystatin genes of the present invention, ZmCys4 (SEQ ID NO: 5) and GmCys2 (SEQ ID NO: 29), were expressed in, and their encoded proteins purified from E. coli as set forth in Example 7. A control expression vector was also prepared which contained no cystatin gene. The purified cystatins and control were injected into sugar beet nematode-induced syncytia in Arabidopsis roots one week after inoculation, as per the methods outlined in Bockenhoff and Grundler ((1994), Parasitology. 109: 249-255), hereby incorporated by reference. Fluorescent dye was used to monitor the growth of the nematodes following injection, as per Bockenhoff and Grundler (1994). The anti-nematodal activity of the cystatins was measured by comparing the nematode growth and development 10 days following injection, and comparing it with the control. Results of the experiment are presented in Table 61, below.
    TABLE 61
    Effect of Two Cystatins on Nematode Development
    Protein Injected Lethality (%)
    Control 30
    Zm-Cys4 (SEQ ID NO: 6) 50
    Gm-Cys2 (SEQ ID NO: 30) 58
  • These results show that both Zm-Cys4 and Gm-Cys2 had significant inhibitory effects on the growth and development of sugar beet nematode juveniles in Arabidopsis. Sugar beet nematode is a genetically close relative of soybean cyst nematode. It has been reported that there is a high cysteine proteinase activity in SCN intestines (Lilley, C. J. et al. (1996) 113: 415-424). These data indicate that both Zm-Cys4 and Gm-Cys2 confer resistance to nematodes by inhibiting SCN growth and development in roots.
  • EXAMPLE 10 Use of C. elegans as a Model to Analyze Cystatin Anti-Nematodal Activity
  • C. elegans populations are cultured on NGM agar carrying a lawn of E. coli OP50 cells as described by Wood ((1988) The nematode C. elegans, Cold Spring Harbor, N.Y. Cold Spring Harbor Laboratory Press). After populations are maintained for five days, agar plugs are removed to fresh plates. Cystatins are added to the medium to a final concentration of 2.5 mg/L just prior to pouring. In order to study the effect of the cystatins on egg laying, hermaphrodite nematodes are taken from their normal growth media and transferred individually to fresh plates containing the cystatin(s) to be used for testing. Egg laying is carefully monitored.
  • Half of the eggs laid on each plate are removed to fresh plates containing media not supplemented with the cystatin(s) being tested. Development of the hatched larvae is monitored.
  • Alternately, groups of larvae hatched on normal media can be transferred to plates containing the cystatin(s) to be tested at time points corresponding to larval stages L1, L2, L3 and L4. The larvae for each stage should be removed, respectively, 6, 12, 24, and 30 hours post hatching. Development of the various larval stages on the supplemented media is monitored.
  • All publications and patent applications mentioned in the specification are indicative of the level of those skilled in the art to which this invention pertains. All publications, patents and patent applications are herein incorporated by reference to the same extent as if each individual publication, patent or patent application was specifically and individually indicated to be incorporated by reference.
  • Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it will be obvious that certain changes and modifications may be practiced within the scope of the appended claims.
    TABLE 62
    Multiple Alignment of All Cystatin Sequences
    Plurality: 2.00 Threshold: 4 AveWeight 1.00 AveMatch 2.78 AvMisMatch −2.25
    Symbol comparison table: blosum62.cmp CompCheck: 1102
    GapWeight:       8
    GapLengthWeight: 2
    Pileup MSF: 314 Type: P May 13, 2003 15:11 Check: 6646 . . .
    //
        1  50
    gm-cys6 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ (SEQ ID NO:38)
    gm-cys8 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ (SEQ ID NO:42)
    ta-cys8 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜MAR VIGASGACAL (SEQ ID NO:68)
    ta-cys9 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜MAR LVGAAGACAL (SEQ ID NO:70)
    ZmCys14 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜MAR ...ALGACVL (SEQ ID NO:26)
    os-cys5 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜M (SEQ ID NO:54)
    ZmCys3 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜MRK (SEQ ID NO:4)
    ZmCys4 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜MRK (SEQ ID NO:6)
    ta-cys13 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ (SEQ ID NO:76)
    ZmCys1 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜MRK (SEQ ID NO:2)
    os-cys1 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ (SEQ ID NO:46)
    ta-cys1 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜MEMWKYRVV (SEQ ID NO:58)
    ta-cys2 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ (SEQ ID NO:60)
    ta-cys4 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ (SEQ ID NO:64)
    ta-cys6 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ (SEQ ID NO:66)
    os-cys3 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ (SEQ ID NO:50)
    ZmCys8 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ (SEQ ID NO:14)
    ZmCys12 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ MRVAAT...R (SEQ ID NO:22)
    ZmCys5 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ MRVAAT...R (SEQ ID NO:8)
    os-cys2 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ MRVAATTRPA (SEQ ID NO:48)
    ta-cys10 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ MRVAATRPAS (SEQ ID NO:72)
    gm-cys2 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ (SEQ ID NO:30)
    gm-cys7 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ (SEQ ID NO:40)
    gm-cys1 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ MRALTSSSST (SEQ ID NO:28)
    gm-cys3 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ (SEQ ID NO:32)
    gm-cys4 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ (SEQ ID NO:34)
    ZmCys10 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ (SEQ ID NO:18)
    ZmCys6 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ (SEQ ID NO:10)
    os-cys4 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜M LRRRGFCCCS (SEQ ID NO:52)
    ta-cys11 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜MVRRCGCS (SEQ ID NO:74)
    gm-cys5 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ (SEQ ID NO:36)
    gm-cys9 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ (SEQ ID NO:44)
    ZmCys7 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ (SEQ ID NO:12)
    os-cys6 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ (SEQ ID NO:56)
    ZmCys9 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ (SEQ ID NO:16)
    ZmCys13 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜M (SEQ ID NO:24)
    ta-cys3 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜M (SEQ ID NO:62)
    ZmCys11 MAFLSTNALM SVPITAAAAP RHRRSLVVVR AAAVKSNEHL QEEQASVADG (SEQ ID NO:20)
    51                                                 100
    gm-cys6 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜MVGG KTEVP.DVRT
    gm-cys8 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜MAVALT ILVTLLSVLS SASCARMVGG KTEIP.EVRK
    ta-cys8 LVVLLVACA. ASAARTE... .PGAA.RQLW ED..GRKVGG RTEVR.DVES
    ta-cys9 LVILLMACA. ASAARSE... .PGAA.RQLW DD..GRKVGG RTEVT.DVEG
    ZmCys14 LAVLLGALAP AAAARAHDDQ GSGAGIRQPS GEYRGRKVGA RTEVR.DVEG
    os-cys5 ATSPMLFLVS LLLVLVAAAT GDEASPSNAA APAAPVLVGG RTEIR.DVGS
    ZmCys3 HRIVSLVAAL LILLAL.AVS STRNAQEDSM ADNTGTLAGG IKDVP.GNEN
    ZmCys4 HRIVSLVAAL LILLAL.AVS STRNAQEDSM ADNTGTLAGG IKDVP.GNEN
    ta-cys13 ˜˜˜˜SLVAAL LILLAL.AVS STRNAQEDSM ADNTGTLAGG IKDVP.GNEN
    ZmCys1 HRIVSLVAAL LVLLALAAVS STRSAQKESV ADNAGMLAGG IKDVP.ANEN
    os-cys1 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜M SSDGGPVLGG VEPV..GNEN
    ta-cys1 GSVAALLLLL AIVVPFTQTQ TQSARDKAAM AEDAGPLVGG ISDSPMGQEN
    ta-cys2 ˜˜˜˜˜˜˜˜LL AIVVPFTQTR TQSARDKAAM AEDAGPLVGG IKDSPMGQEN
    ta-cys4 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜MAEAAQGGG LRGRGALLGG VQDAPAGREN
    ta-cys6 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜MAEAAQGGG LRGRGVLLGG VQDAPAGREN
    os-cys3 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜MAEEAQ... .QPRGVKVGG IHDAPAGREN
    ZmCys8 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜MAEVHN ERPVG.MVGD VRDAPVGREN
    ZmCys12 AAAAAHPPSA FLLLLLLLGC ASL.AIGGA. .AMAGHVLGG VKENP.AAAN
    ZmCys5 AAAAAHPPSA FLLLLLLLGC ASL.AIGGA. .AMAGHVLGG VKENP.AAAN
    os-cys2 SSSAAAPLPL FLLLAVAAAA AALFLVGSAS LAMAGHVLGG AHDAP.SAAN
    ta-cys10 SAPVA..LLA ALALLFLVGS ASL.AIG... .AMASHVLGG KSENP.DAAN
    gm-cys2 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜MAALGG NRDVT.GSQN
    gm-cys7 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜MAALGG NRDVA.GSQN
    gm-cys1 FIPKRYSFFF FLSILFALRS SSGGCSEYHH HHAPMATIGG LRDSQ.GSQN
    gm-cys3 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜MAALGG FTDIT.GAQN
    gm-cys4 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜MP.TLGA
    ZmCys10 ˜˜˜MMPRRAL LFAAVLLAAS A.AAVSGFHL GGDESGLVRG VL.AALRER.
    ZmCys6 ˜˜MTMPRRAL LFAAVLLAAS A.AAVSGFHL AGDESGLVRG VL.TAVRERA
    os-cys4 GAPAAAAAAL LLLAV..AAA A.PRAAGFHL GGDESVLVRG ML.AAIR.RE
    ta-cys11 GAMLLA.... LSLAVLLAAS AVPGAAGFHL GGDESGLVRG ML.AAVRER.
    gm-cys5 ˜˜˜˜˜˜˜˜˜˜ ˜˜MRHHCL.L LVSLVLVSYA AR.SESALGG WS..PIKDVN
    gm-cys9 ˜˜˜˜˜˜˜˜˜˜ ˜˜MKQKCLVV LVFVVLLACA VGWDEGTPGG WN..PIKNIN
    ZmCys7 ˜˜˜˜˜˜˜˜MS ARALLLTTAT LLLLVAAAR. ..AGQPLAGG WS..PIRNVS
    os-cys6 ˜˜˜MARIPLL LALLLAVSAA AAAQVGGNR. ..GHGPLVGG WS..PITDVG
    ZmCys9 ˜˜˜MATHRHC LPLLLLVAAA LAAVPARAAL GGGRGPLLGG WN..PIPDVS
    ZmCys13 AMTMTLGSML IAAAAVVGLC SVAPAASARE EPLQPQIVGG WK..PIKNVN
    ta-cys3 RTSSFLLIIV VAFLYAIGSP AIGCGERMGN QLWNTAIENG WE..PIGNIN
    ZmCys11 ARGRRRAMVL LAATAAVTGS SVAICRSARA AGV.TTLSGQ YV..KIENVK
    101                                                150
    gm-cys6 NREVQELGRF AVEEYNRGLK Q.WKN....N GSEQLNFSEV VEAQQQVVSG
    gm-cys8 NRQVQELGRF AVEEYNLGLK L.LKNNNVDN GREQLNFSAV VEAQQQVVSG
    ta-cys8 DREVQELGRY SVEEHNRRRE EGCEGGGGVC GR..LEFARV VSAQRQVVSG
    ta-cys9 DREVQELGRY SVEEHNRRRE EGCEGGGGVC GR..LEFARV VSAQRQVVSG
    ZmCys14 DGEVQELGRF SVAEYNRQLR EG..GGGG.. GR..LEFGRV VAAQRQVVSG
    os-cys5 NKAVQSLGRF AVAEHNRRLR HGGSGGPADP VPVKLAFARV VEAQKQVVSD
    ZmCys3 DLHLQELARF AVDEHN...K KA........ .NALLGFEKL VKAKTQVVAG
    ZmCys4 DLHLQELARF AVDEHN...K KA........ .NALLGFEKL VKAKTQVVAG
    ta-cys13 DLHLQELARF AVDEHN...K KA........ .NALLGFEKL VKAKTQVVAG
    ZmCys1 DLQLQELARF AVNEHN...Q KA........ .NALLGFEKL VKAKTQVVAG
    os-cys1 DLHLVDLARF AVTEHN...K KA........ .NSLLEFEKL VSVKQQVVAG
    ta-cys1 DLDVIALARF AVSEHN.. N KA........ .NALLEFENV VKVKKQTVAG
    ta-cys2 DLDVIALARF AVSEHN.. N KA........ .NALLEFENV VKLKKQTVAG
    ta-cys4 DLETIELARF AVAEHN...I KA........ .NALLEFERL VKVRQQVVAG
    ta-cys6 DLATIELARF AVAEHN...I KA........ .NALLEFERL VKVRQQVVAG
    os-cys3 DLTTVELARF AVAEHN...S KA........ .NAMLELERV VKVRQQVVGG
    ZmCys8 DLEAIELARF AVAEHN...S KT........ .NAMLEFERL VKVRHQVVAG
    ZmCys12 SAESDGLGRF AVDEHN...R RE........ .NALLEFVRV VEAKEQVVAG
    ZmCys5 SAESDGLGRF AVDEHN...R RE........ .NALLEFVRV VEAKEQVVAG
    os-cys2 SVETDALARF AVDEHN...K RE........ .NALLEFVRV VEAKEQVVAG
    ta-cys10 SLETDGLARF AVDEHN...K RE........ .NALLEFVRV VEAKEQTVAG
    gm-cys2 SVEIDALARF AVEEHN...K KQ........ .NALLEFEKV VIAKQQVVSG
    gm-cys7 SLEIDGLARF AVEEHN...K KQ........ .NALLEFEKV VSAKQQVVSG
    gm-cys1 SVQTEALARF AVDEHN...K KQ........ .NSLLEFSRV VRTQEQVVAG
    gm-cys3 SIDIENLARF AVDEHN...K KE........ .NAVLEFVRV ISAKKQVVSG
    gm-cys4 GGEIDHLARF AVEEQN...K RE........ .NANLEFVGV IRAKQQVVEG
    ZmCys10 .AEAEDAARF AVAHYN...K NQ........ .GAALEFTRV LKSKRQVVTG
    ZmCys6 EAEAEDAARF AVAYHN...R NQ........ .GAALEFTRV LKSKRQVVTG
    os-cys4 QAEAEDAARF AVAEYN...K NQ........ .GAELEFARI VKAKRQVVTG
    ta-cys11 .AXAXDAARF XVAEHN...R XQ........ .GSALEFTRV VNAKXQVVAG
    gm-cys5 DSHVAEIANY ALSEYD...K RS........ .GAKLTLVKV VKGETQVVSG
    gm-cys9 DPHVTEIANF AVTEYD...K QS........ .GEKLKLVKV TKGDLQVVAG
    ZmCys7 DPHIQELGGW AVTEHV...R RA........ .NDGLRFGEV TGGEEQVVSG
    os-cys6 DPHIQELGGW AVERHA...S LS........ .SDGLRFRRV TSGEQQVVSG
    ZmCys9 DSHIQELGGW ALGQA.KHQK LA........ .ADGLRFRRV VRGEQQVVSG
    ZmCys13 DPHVQEIGRW AVSEHI...K TA........ .NDGLGFGRV VSGEEQIVAG
    ta-cys3 DQHIQELGRW AVLEFGKHVN CV........ ....LKFNKV VSGRQQLVSG
    ZmCys11 DPYVQGVGEW AVKEHN...R QT........ .GESLQFAEV VSGMEQVVAG
    151                                                200
    gm-cys6 MKYYLKISAT HKG....... .VHKMFTSVV VVKPWLHS.. KQLLHFAPAA
    gm-cys8 MKYYLKISAT HNG....... .VHEMFNSVV VVKPWLHS.. KQLLHFAPAS
    ta-cys8 IKYYLRVAAA EENGAGSNVV SDGRVFDAVV VVKPWLQS.. RALVRFAPAD
    ta-cys9 IKYYLRVAAA EEGGAGSNGV TDGRVFDAVV VVKPWLQS.. RALIRFAPAD
    ZmCys14 LKYYLRVVAV EEGGAGNGG. ..ERVFDAVV VVKPWLDS.. RTLLTFAPAA
    os-cys5 VAYYLKVAAS ARDPRGGAAA GGDRVFDAVV VVKAWLKS.. KELVSFTPAS
    ZmCys3 TMYYLTIEVK D..GEVK... ...KLYEAKV WEKPWEN..F KELQEFKPVE
    ZmCys4 TMYYLTIEVK D..GEVK... ...KLYEAKV WEKPWEN..F KELQEFKPVE
    ta-cys13 TMYYLTIEVK D..GEVK... ...KLYEAKV WEKPWEN..F KELQEFKPVE
    ZmCys1 TMYYLTIEVK D..GEVN... ...KLYEAKV WEKPWEN..F KQLQEFKPVE
    os-cys1 TLYYFTIEVK E..GDAK... ...KLYEAKV WEKPWMD..F KELQEFKPVD
    ta-cys1 TMHYITIRVT E..GGAK... ...KLYEAKV WEKPWEN..F KKLEEFKLVE
    ta-cys2 TMHYITIRVT E..GGAK... ...KLYEAKV WEKPWEN..F KQLQEFKPVE
    ta-cys4 CMHYFTIEVK E.GGA.K... ...KLYEAKV WEKAWEN..F KQLQDFKPAA
    ta-cys6 CMHYFTIEVK E.GGA.K... ...KLYEAKV WEKAWEN..F KQLQDFKPAA
    os-cys3 FMHYLTVEVK EPGGA.N... ...KLYEAKV WERAWEN..F KQLQDFKPLD
    ZmCys8 TLHHFTVEVK EAGGGEK... ...KLYEAKV WEKAWEN..F KQLQSFELVG
    ZmCys12 TLHHLTLEAV F..AGRK... ...KLYEAKV WVKPWLD..F KELQEFSHKG
    ZmCys5 TLHHLTLEAV F..AGRK... ...KLYEAKV WVKPWLD..F KELQEFSHKG
    os-cys2 TLHHLTLEAL F..AGRK... ...KVYEAKV WVKPWLD..F KELQEFRNTG
    ta-cys10 TVHHLTLEAL F..AGRK... ...KLYEAKV WVKPWLD..F KELQEFRHTG
    gm-cys2 TLYTITLEAK D..GGQK... ...KVYEAKV WEKSWLN..F KEVQEFKLVG
    gm-cys7 TLYTITLEAK D..GGQK... ...KVYEAKV WEKAWLN..F KEVQEFKLVG
    gm-cys1 TLHHLTLEAI F..AGEK... ...KLYEAKV WVKPWLN..F KELQEFKPAG
    gm-cys3 TLYYITLEAN D..GVTK... ...KVYETKV LEKPWLN..I KEVQEFKPIT
    gm-cys4 FIYYITLEAK D..GETK... ...NVYETKV WVRSWLN..S KEVLEFKPIS
    ZmCys10 TLHDLILEAA D..AGKK... ...SVYRAKV WVKSWED..F KSVVEFRLVG
    ZmCys6 TLHDLILEAA D..AGKK... ...SLYRAKV WVKPWED..F KSVVEFRLAG
    os-cys4 TLHDLMLEVV D..SGKK... ...SLYSAKV WVKPWLD..F KAVVEFRHVG
    ta-cys11 TLHDLMVEVV D..SGXK... ....ICTTQS LGEAWQN..F XAVVEFRHAG
    gm-cys5 TNYRLVLKAK D.GSATA... ...S.YEAIV WEKPWL..HF MNLTSFKPLH
    gm-cys9 LNYRLSLTAS D.SN...... ...N.YQAIV YEKAWAREHY RNLTSFTPLH
    ZmCys7 MNYKLVLDAT DADGKVA... ...A.YGAFV YEQSWTNT.. RELVSFAPAS
    os-cys6 MNYRLVVSAS DPAGATA... ...S.YVAVV YEQSWTNT.. RQLTSFKPAA
    ZmCys9 MNYRLYVDAA DPAGRTV... ...P.YVAVV YEQVWTAP.. ...ASSPPST
    ZmCys13 KNYRLRIQAT KVGGQKA... ...M.YRAVV YEQL.TNT.. RQLLSFDPAN
    ta-cys3 MNYELIIEAS DIGGKED... ...K.YKAEV YEQTWTHK.. RQLLSFAKVK
    ZmCys11 TNYKLNLATK DP...TS... ...S.YQAVV FDPLPNSSKN RQLMSFKSI˜
    201                                                250
    gm-cys6 PSSKDF˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    gm-cys8 SSTTTTNNNM HPIVRKDN˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    ta-cys8 AK˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    ta-cys9 AK˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    ZmCys14 AK˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    os-cys5 STK˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    ZmCys3 EGASA˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    ZmCys4 EGASA˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    ta-cys13 EGASA˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    ZmCys1 EGASA˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    os-cys1 ASANA˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    ta-cys1 DVPSA˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    ta-cys2 DAAIA˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    ta-cys4 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    ta-cys6 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    os-cys3 DATA˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    ZmCys8 DAAVA˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    ZmCys12 DATAFTNADL GAKQGGHEPG WREVPVEDPV VKDAAHHAVK SIQERSNSLF
    ZmCys5 DATAFTNADL GAKQGGHEPG WREVPVEDPV VKDAAHHAVK SIQERSNSLF
    os-cys2 DATTFTNADL GAKKGGHEPG WRDVPVHDPV VKDAADHAVK SIQQRSNSLF
    ta-cys10 D˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    gm-cys2 DAPA˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    gm-cys7 DAPA˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    gm-cys1 DVPSFTSADL GVKKDGHQPG WQSVPTHDPQ VQDAANHAIK TIQQRSNSLV
    gm-cys3 VAVNPLSVTV ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    gm-cys4 INPLSVSV˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    ZmCys10 DSESEPEPSV ASDVSSGQAI AKLSLEADIV QEEARLHTIE NDGLSGDFTS
    ZmCys6 DSESEPEPSV ASDEGSGQGV AKLSLEADII HEEAHLHTIE NDGLSSDFAS
    os-cys4 DSQS..QSAT AADDNAGQDT AD.....PTV ASRNDLHNTE NNKVSVVLST
    ta-cys11 TXSXLPLLYG RXGKLPQASL KXHAXRXQHX NTXSVTHLSR KHXVWCXYIX
    gm-cys5 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    gm-cys9 A˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    ZmCys7 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    os-cys6 AH˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    ZmCys9 RCPAPTETIR TRVGRSDVLR QLRVELILLL FLLL˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    ZmCys13 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    ta-cys3 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    ZmCys11 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    251                                                300
    gm-cys6 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    gm-cys8 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    ta-cys8 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    ta-cys9 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    ZmCys14 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    os-cys5 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    ZmCys3 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    ZmCys4 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    ta-cys13 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    ZmCys1 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    os-cys1 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    ta-cys1 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    ta-cys2 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    ta-cys4 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    ta-cys6 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    os-cys3 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    ZmCys8 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    ZmCys12 PYELLEILRA HAQVVEDFAK FDILMKLKRG SKEEKIKAEV HKSLEGAFVL
    ZmCys5 PYELLEILRA HAQVVEDFAK FDILMKLKRG SKEEKIKAEV HKSLEGAFVL
    os-cys2 PYELLEIVRA KAEVVEDFAK FDILMKLKRG NKEEKFKAEV HKNLEGAFVL
    ta-cys10 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    gm-cys2 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    gm-cys7 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    gm-cys1 PYELHEVADA KAEVIDDFAK FNLLLKVKRG QKEEKFKVEV HKNNQGGFHL
    gm-cys3 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    gm-cys4 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    ZmCys10 SSS˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    ZmCys6 SA˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    os-cys4 FSQTYSV˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    ta-cys11 LQLXXE˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    gm-cys5 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    gm-cys9 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    ZmCys7 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    os-cys6 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    ZmCys9 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    ZmCys13 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    ta-cys3 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    ZmCys11 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜˜˜˜˜˜˜
    301            314
    gm-cys6 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜
    gm-cys8 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜
    ta-cys8 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜
    ta-cys9 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜
    ZmCys14 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜
    os-cys5 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜
    ZmCys3 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜
    ZmCys4 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜
    ta-cys13 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜
    ZmCys1 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜
    os-cys1 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜
    ta-cys1 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜
    ta-cys2 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜
    ta-cys4 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜
    ta-cys6 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜
    os-cys3 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜
    ZmCys8 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜
    ZmCys12 NQHQPAEHDE SSSQ
    ZmCys5 NQHQPAEHDE SSSQ
    os-cys2 NQMQ.QEHDE SSSQ
    ta-cys10 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜
    gm-cys2 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜
    gm-cys7 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜
    gm-cys1 NQMEQDHS˜˜ ˜˜˜˜
    gm-cys3 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜
    gm-cys4 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜
    ZmCys10 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜
    ZmCys6 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜
    os-cys4 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜
    ta-cys11 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜
    gm-cys5 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜
    gm-cys9 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜
    ZmCys7 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜
    os-cys6 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜
    ZmCys9 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜
    ZmCys13 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜
    ta-cys3 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜
    ZmCys11 ˜˜˜˜˜˜˜˜˜˜ ˜˜˜˜

Claims (19)

1. An isolated polynucleotide comprising a nucleic acid sequence selected from the group consisting of:
(a) a nucleic acid sequence set forth in SEQ ID NO: 5;
(b) a nucleotide sequence that encodes a polypeptide having the amino acid sequence set forth in SEQ ID NO: 6;
(c) a nucleic acid sequence having at least 95% sequence identity over the entire length of SEQ ID NO: 5 as determined by the GAP algorithm under default parameters, wherein said nucleic acid sequence encodes a polypeptide with cysteine proteinase inhibitor activity;
(d) a nucleic acid sequence that encodes a polypeptide with cysteine proteinase inhibitor activity, wherein said polypeptide has at least 95% sequence identity to SEQ ID NO: 6; and
(e) a nucleic acid sequence that comprises the full length complement of any one of (a) to (d).
2. The isolated polynucleotide of claim 1, wherein said polynucleotide is optimized for expression in a plant.
3. A DNA construct comprising the isolated polynucleotide of claim 1, wherein said polynucleotide is operably linked to a promoter that drives expression in a host cell.
4. The DNA construct of claim 3, wherein said polynucleotide is operably linked in an antisense orientation to said promoter.
5. An expression cassette comprising the DNA construct of claim 3.
6. A host cell having stably incorporated into its genome at least one DNA construct of claim 3.
7. The host cell of claim 6, wherein said host cell is a plant cell.
8. A transgenic plant having stably incorporated into its genome the DNA construct of claim 3.
9. The transgenic plant according to claim 8, wherein said plant is a monocot.
10. The transgenic plant according to claim 8, wherein said plant is a dicot.
11. The transgenic plant according to claim 8, wherein said plant is selected from the group consisting of: corn, soybean, wheat, rice, alfalfa, barley, millet, sunflower, sorghum, canola and cotton.
12. Transformed seed of the transgenic plant of claim 8, wherein said transformed seed comprises the DNA construct of claim 3.
13. A method for enhancing the disease resistance of a plant comprising:
(a) introducing into a plant cell at least one DNA construct comprising a polynucleotide operably linked to a promoter that drives expression of a cysteine proteinase inhibitor polypeptide in plant cells, wherein said polynucleotide comprises a nucleotide sequence selected from the group consisting of:
(i) a nucleic acid sequence set forth in SEQ ID NO: 5;
(ii) a nucleic acid sequence that encodes a polypeptide having the amino acid sequence set forth in SEQ ID NO: 6;
(iii) a nucleic acid sequence having at least 95% sequence identity over the entire length of SEQ ID NO: 5 as determined by the GAP algorithm under default parameters, wherein said nucleic acid sequence encodes a polypeptide with cysteine proteinase inhibitor activity;
(iv) a nucleic acid sequence that encodes a polypeptide with cysteine proteinase inhibitor activity, wherein said polypeptide has at least 95% sequence identity to SEQ ID NO: 6; and
(v) a nucleotide sequence that comprises the full length complement of any one of (i) through (iv).
(b) growing the plant cell under plant growing conditions to produce a regenerated plant; and
(c) inducing expression of said polynucleotide for a time sufficient to enhance the disease resistance of said plant.
14. The method of claim 13, wherein said promoter is selected from the group consisting of:
(a) a strong constitutive promoter;
(b) a tissue-specific promoter;
(c) a temporally-defined promoter; and
(d) an inducible promoter.
15. The method of claim 13, wherein said plant expresses a polypeptide having pesticidal activity against fungal pathogens.
16. The method of claim 15, wherein said fungus is Fusarium ssp.
17. The method of claim 13, wherein said plant expresses a polypeptide having pesticidal activity against insects.
18. The method of claim 13, wherein said plant expresses a polypeptide having pesticidal activity against nematodes.
19. The method of claim 18, wherein said nematode is a Soybean Cyst Nematode.
US11/674,765 2003-09-25 2007-02-14 Crop Plant Cystatin Proteinase Inhibitors and Methods of Use Abandoned US20070162999A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/674,765 US20070162999A1 (en) 2003-09-25 2007-02-14 Crop Plant Cystatin Proteinase Inhibitors and Methods of Use

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US50594803P 2003-09-25 2003-09-25
US10/947,979 US7205453B2 (en) 2003-09-25 2004-09-23 Crop plant cystatin proteinase inhibitors encoding nucleic acids and methods of use
US11/674,765 US20070162999A1 (en) 2003-09-25 2007-02-14 Crop Plant Cystatin Proteinase Inhibitors and Methods of Use

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/947,979 Division US7205453B2 (en) 2003-09-25 2004-09-23 Crop plant cystatin proteinase inhibitors encoding nucleic acids and methods of use

Publications (1)

Publication Number Publication Date
US20070162999A1 true US20070162999A1 (en) 2007-07-12

Family

ID=34393092

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/947,979 Expired - Fee Related US7205453B2 (en) 2003-09-25 2004-09-23 Crop plant cystatin proteinase inhibitors encoding nucleic acids and methods of use
US11/674,765 Abandoned US20070162999A1 (en) 2003-09-25 2007-02-14 Crop Plant Cystatin Proteinase Inhibitors and Methods of Use

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US10/947,979 Expired - Fee Related US7205453B2 (en) 2003-09-25 2004-09-23 Crop plant cystatin proteinase inhibitors encoding nucleic acids and methods of use

Country Status (2)

Country Link
US (2) US7205453B2 (en)
WO (1) WO2005030967A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110120958A1 (en) * 2009-11-22 2011-05-26 United Laboratories International, Llc Wastewater Treatment

Families Citing this family (56)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP4817751B2 (en) * 2005-08-17 2011-11-16 独立行政法人農業・食品産業技術総合研究機構 Novel antibacterial proteins and their uses
AR059448A1 (en) * 2006-02-13 2008-04-09 Divergence Inc UNION VEGETABLE CHEMICAL POLIPEPTIDES FOR UNIVERSAL MOLECULAR RECOGNITION
DE102006018583B4 (en) * 2006-04-21 2017-05-11 Alyoniq Ag Defense against the formation of haustoria as a plant protection product against plant parasites
KR100897763B1 (en) * 2007-04-20 2009-05-15 경상대학교산학협력단 Transgenic plant for retardation of seed germination and inhibition of cysteine peptidase activity by over-expression of phytocystatin gene
US8829282B2 (en) 2008-05-14 2014-09-09 Monsanto Technology, Llc Plants and seeds of spring canola variety SCV425044
US7947877B2 (en) 2008-05-14 2011-05-24 Monosanto Technology LLC Plants and seeds of spring canola variety SCV328921
US7964774B2 (en) 2008-05-14 2011-06-21 Monsanto Technology Llc Plants and seeds of spring canola variety SCV384196
US7935870B2 (en) 2008-05-14 2011-05-03 Monsanto Technology Llc Plants and seeds of spring canola variety SCV354718
EP2315518B1 (en) 2008-07-23 2014-05-21 Temasek Life Sciences Laboratory Limited Somatic embryogenesis of jatropha curcas from ovules
US20110230396A1 (en) * 2008-08-21 2011-09-22 Ith Immune Therapy Holdings Ab Anti-infective compounds and uses thereof
AU2010259295B2 (en) 2009-06-10 2015-05-07 Temasek Life Sciences Laboratory Limited Virus induced gene silencing (VIGS) for functional analysis of genes in cotton.
US8071848B2 (en) 2009-06-17 2011-12-06 Monsanto Technology Llc Plants and seeds of spring canola variety SCV218328
US8440891B2 (en) 2009-09-22 2013-05-14 Board of Trustees of the University of Akransas, N.A. Rice cultivar CL 142-AR
US8440892B2 (en) 2009-10-15 2013-05-14 Board Of Trustees Of The University Of Arkansas, N.A. Rice cultivar CL 181-AR
US8143488B2 (en) 2010-02-26 2012-03-27 Monsanto Technoloy LLC Plants and seeds of spring canola variety SCV470336
US8148611B2 (en) 2010-02-26 2012-04-03 Monsanto Technology Llc Plants and seeds of spring canola variety SCV453784
US8138394B2 (en) 2010-02-26 2012-03-20 Monsanto Technology Llc Plants and seeds of spring canola variety SCV431158
US8581048B2 (en) 2010-03-09 2013-11-12 Monsanto Technology, Llc Plants and seeds of spring canola variety SCV119103
US8153865B2 (en) 2010-03-11 2012-04-10 Monsanto Technology Llc Plants and seeds of spring canola variety SCV152154
US8513487B2 (en) 2011-04-07 2013-08-20 Zenon LISIECZKO Plants and seeds of spring canola variety ND-662c
US8513494B2 (en) 2011-04-08 2013-08-20 Chunren Wu Plants and seeds of spring canola variety SCV695971
US8507761B2 (en) 2011-05-05 2013-08-13 Teresa Huskowska Plants and seeds of spring canola variety SCV372145
US8513495B2 (en) 2011-05-10 2013-08-20 Dale Burns Plants and seeds of spring canola variety SCV291489
US9631196B2 (en) 2011-06-10 2017-04-25 Temasek Life Sciences Laboratory Limited Genetic manipulation and expression systems for Pucciniomycotina and Ustilaginomycotina subphyla
US8785729B2 (en) 2011-08-09 2014-07-22 Nunhems, B.V. Lettuce variety redglace
US8754293B2 (en) 2011-09-09 2014-06-17 Nunhems B.V. Lettuce variety intred
US9204603B2 (en) 2011-12-21 2015-12-08 The Curators Of The University Of Missouri Soybean variety S05-11482
US20130167262A1 (en) 2011-12-21 2013-06-27 The Curators Of The University Of Missouri Soybean variety s05-11268
US9380756B2 (en) 2012-01-04 2016-07-05 Nunhems B.V. Lettuce variety multigreen 50
US8802935B2 (en) 2012-04-26 2014-08-12 Monsanto Technology Llc Plants and seeds of spring canola variety SCV942568
US8835720B2 (en) 2012-04-26 2014-09-16 Monsanto Technology Llc Plants and seeds of spring canola variety SCV967592
US8878009B2 (en) 2012-04-26 2014-11-04 Monsanto Technology, LLP Plants and seeds of spring canola variety SCV318181
US8859857B2 (en) 2012-04-26 2014-10-14 Monsanto Technology Llc Plants and seeds of spring canola variety SCV259778
CA2905743C (en) 2013-03-13 2021-09-28 Pioneer Hi-Bred International, Inc. Glyphosate application for weed control in brassica
CN115960896A (en) 2013-03-14 2023-04-14 先锋国际良种公司 Compositions and methods for controlling insect pests
RU2015143825A (en) 2013-03-15 2017-04-26 Пайонир Хай-Бред Интернэшнл, Инк. PHI-4 POLYPEPTIDES AND WAYS OF THEIR APPLICATION
EA030896B1 (en) 2013-08-16 2018-10-31 Пайонир Хай-Бред Интернэшнл, Инк. Insecticidal proteins and methods for their use
BR112016005543B1 (en) 2013-09-13 2022-03-08 Pioneer Hi-Bred International, Inc RECOMBINANT PIP-72 POLYPEPTIDE, DNA CONSTRUCTION, METHOD FOR OBTAINING A TRANSGENIC PLANT, HOST CELL, COMPOSITION, FUSION PROTEIN, METHOD FOR CONTROLLING AN INSECT PEST POPULATION, METHOD FOR INHIBITING THE GROWTH OR KILLING AN INSECT PEST, METHOD FOR TO CONTROL AN INSECT INFESTATION IN A TRANSGENIC PLANT, METHOD FOR IDENTIFYING A NUCLEOTIDE SEQUENCE IN A BIOLOGICAL SAMPLE, METHOD FOR IDENTIFYING A PIP-72 POLYPEPTIDE IN A SAMPLE
BR112016018287A2 (en) 2014-02-07 2017-10-10 Du Pont insecticide proteins and methods for their use
BR112016018103B1 (en) 2014-02-07 2024-01-16 E.I. Du Pont De Nemours And Company POLYPEPTIDE AND ITS USE, POLYNUCLEOTIDE, COMPOSITION, FUSION PROTEIN, METHOD FOR CONTROLING A POPULATION, METHOD FOR INHIBITING GROWTH, METHOD FOR CONTROLING INFESTATION, METHOD FOR OBTAINING A PLANT OR PLANT CELL, CONSTRUCTION
WO2016044092A1 (en) 2014-09-17 2016-03-24 Pioneer Hi Bred International Inc Compositions and methods to control insect pests
CA2963558C (en) 2014-10-16 2023-04-04 Pioneer Hi-Bred International, Inc. Insecticidal proteins and methods for their use
EP3209775A4 (en) 2014-10-22 2018-09-12 Temasek Life Sciences Laboratory Limited Terpene synthases from ylang ylang (cananga odorata var. fruticosa)
WO2016099916A1 (en) 2014-12-19 2016-06-23 E. I. Du Pont De Nemours And Company Polylactic acid compositions with accelerated degradation rate and increased heat stability
CN108064233B (en) 2015-05-19 2022-07-15 先锋国际良种公司 Insecticidal proteins and methods of use thereof
BR112017027382A2 (en) 2015-06-16 2018-08-28 Pioneer Hi-Bred International, Inc. silencing element, dna construct, expression construct, expression cassette, host cell, composition, plant cell, plant or plant part, transgenic seed, method for controlling a plant insect pest, kit for controlling insect pests
EP3331352B1 (en) 2015-08-06 2022-07-06 Pioneer Hi-Bred International, Inc. Plant derived insecticidal proteins and methods for their use
BR102015028125A2 (en) * 2015-11-06 2017-05-09 Spinomics Ltda - Epp gene construction, use, pharmaceutical composition for cancer treatment and process for its preparation
WO2017105987A1 (en) 2015-12-18 2017-06-22 Pioneer Hi-Bred International, Inc. Insecticidal proteins and methods for their use
US11008585B2 (en) 2016-05-04 2021-05-18 Pioneer Hi-Bred International, Inc. Insecticidal proteins and methods for their use
US20190185867A1 (en) 2016-06-16 2019-06-20 Pioneer Hi-Bred International, Inc. Compositions and methods to control insect pests
WO2018005411A1 (en) 2016-07-01 2018-01-04 Pioneer Hi-Bred International, Inc. Insecticidal proteins from plants and methods for their use
WO2018013333A1 (en) 2016-07-12 2018-01-18 Pioneer Hi-Bred International, Inc. Compositions and methods to control insect pests
US11021716B2 (en) 2016-11-01 2021-06-01 Pioneer Hi-Bred International, Inc. Insecticidal proteins and methods for their use
BR112022027035A2 (en) 2020-07-14 2023-04-11 Pioneer Hi Bred Int INSECTICIDAL PROTEINS AND METHODS FOR THE USE OF THEM
CN112391394B (en) * 2020-11-26 2022-10-04 南京农业大学 Rice blast resistance related gene OsCYS and application thereof in genetic engineering

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5494813A (en) * 1991-03-05 1996-02-27 Nickerson Biocem Limited Nematode control with proteinase inhibitors
US6703224B2 (en) * 2000-06-05 2004-03-09 Zymogenetics, Inc. Zcys6: a member of the cystatin superfamily
US20040214272A1 (en) * 1999-05-06 2004-10-28 La Rosa Thomas J Nucleic acid molecules and other molecules associated with plants

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997032007A1 (en) * 1996-02-28 1997-09-04 Purdue Research Foundation Soybean cysteine proteinase inhibitors, nucleotides encoding the same, and methods of use thereof

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5494813A (en) * 1991-03-05 1996-02-27 Nickerson Biocem Limited Nematode control with proteinase inhibitors
US20040214272A1 (en) * 1999-05-06 2004-10-28 La Rosa Thomas J Nucleic acid molecules and other molecules associated with plants
US6703224B2 (en) * 2000-06-05 2004-03-09 Zymogenetics, Inc. Zcys6: a member of the cystatin superfamily

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110120958A1 (en) * 2009-11-22 2011-05-26 United Laboratories International, Llc Wastewater Treatment
US8419948B2 (en) 2009-11-22 2013-04-16 United Laboratories International, Llc Wastewater treatment

Also Published As

Publication number Publication date
WO2005030967A2 (en) 2005-04-07
US20050102717A1 (en) 2005-05-12
US7205453B2 (en) 2007-04-17
WO2005030967A3 (en) 2005-09-01

Similar Documents

Publication Publication Date Title
US7205453B2 (en) Crop plant cystatin proteinase inhibitors encoding nucleic acids and methods of use
US6911577B2 (en) Defensin polynucleotides and methods of use
US7238781B2 (en) Plant defensin polynucleotides and methods of use thereof
US20030017566A1 (en) Maize peroxidase genes and their use for improving plant disease resistance and stalk strength
US6720480B2 (en) Maize proteinase inhibitor-like polynucleotides and defense-activated promoter, transformed plants, and methods of use
CA2724158C (en) Defensin polynucleotides and methods of use
US6660907B2 (en) Genes encoding SCIP-1 orthologs and methods of use
US20030028921A1 (en) Maize basal layer antimicrobial protein polynucleotides and method of use

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION