POLYNUCLEOTIDES AND POLYPEPTIDES OF PLANT AND BACTERIAL ORIGIN FOR PROTECTING PLANTS FROM PATHOGENIC FUNGI

Information

  • Patent Application
  • 20240102043
  • Publication Number
    20240102043
  • Date Filed
    July 27, 2023
    9 months ago
  • Date Published
    March 28, 2024
    a month ago
Abstract
The present invention relates to polynucleotides and polypeptides derived from plants and bacteria which are associated with increased resistance of plants to pathogenic fungi and/or Oomycetes. The fungicidal polypeptides of the invention are particularly effective in combating fungi inducing root rot and stalk rot in plants. The present invention further provides methods of using the polynucleotides and polypeptides of the invention for controlling plant fungal/Oomycetes pathogens and for producing transgenic plants having increased resistance to the pathogens.
Description
SEQUENCE LISTING

The Sequence Listing, submitted herewith through Patent Center, as an eXtensible Markup Language file (2023-12-05_Sequence_listing_169.0121-US02.xml, created on Dec. 8, 2023; 8,134,164 bytes) is incorporated by reference.


FIELD OF THE INVENTION

The present invention relates to polynucleotides and polypeptides of plant and bacterial origin which are involved in enhancing resistance of plants towards pathogenic fungi and/or Oomycetes, particularly to fungi inducing root rot and stalk rot in plants, and use thereof for controlling plant diseases associated with the fungal pathogens and for producing genetically engineered plants having increased resistance to the pathogenic fungi.


BACKGROUND OF THE INVENTION

During all developmental stages, plants are exposed to an extremely wide range of biotic and abiotic stress conditions leading to plant diseases. In the production of crop plants, damages caused by biotic stresses, particularly by pathogenic agents, which may be further enhanced under conditions of abiotic stress, pose a major problem and significantly affect the crop yield and profitability.


Many plant diseases are caused by plant pathogenic fungi, and damages to both monocotyledonous and dicotyledonous crop plants are of billions of US$ loss in yield in the U.S. only. For example, stalk rot, caused by a complex of Fusarium spp. and other fungi is one of the most serious challenges in maize production. At present, the majority of both inbreed and hybrid maize lines are susceptible. Fusarium graminearum (Fg) and Fusarium verticillioides (Fv) are the two main causal agents of stalk rot caused by Fusarium spp. in maize, but more than 10 additional Fusarium spp. can cause stalk rot. Natural infection is initiated by a mixture of the local Fusarium spp., but typically during the progress of the disease one species predominates. Gibberella stalk rot (caused by Fusarium graminearum Schwabe) is more prevalent in maize grown in cool regions; while Fusarium stalk rot (caused by Fusarium verticillioides) is most common in dry, warm regions. Fusarium graminearum and other Fusarium species are also responsible for Fusarium head blight (FHB) of wheat, which is a major disease problem for wheat and barley production worldwide, and for various root rots in wide range of different hosts. Colletotrichum spp. infects many grain crops such as barley, wheat, sorghum and corn. C. graminicola is one of the other major pathogens causing mainly maize stalk rots and being capable of infecting many other parts of the corn plant. It has been suggested that C. graminicola behaves as a wilt fungus which efficiently colonizes and moves through the fiber cells that surround the vascular bundles and underlie the epidermal cells in the stalk rind. Movement through the mostly non-living fibers may allow the fungus to avoid host defenses, providing a base from which it can invade adjacent parenchyma cells.


Powdery mildews are fungal diseases caused by different species of fungi in the order Erysiphales that affect a wide range of plants including cereals, grasses, vegetables, ornamentals, weeds, shrubs, fruit trees, broad-leaved shade and forest trees.


Traditionally, plant diseases have been controlled by agronomic practices that include crop rotation, the use of agrochemicals, and conventional breeding techniques. The use of chemicals to control plant pathogens, while being effective, increases the production costs, and moreover, is opposed to by the public and government regulators due to the increased awareness to the harmful effects of such chemicals on the ecosystem and animal health.


Upon the plant recognition of an agent as a pathogen, an array of biochemical responses is activated by the plant. As of today, it is acknowledged that the initial plant response involves induction of several local responses in the cells immediately surrounding the infection site. In the hypersensitive response, cells contacted by the pathogen, and often neighboring cells, rapidly collapse and dry in a necrotic fleck. Other responses include the deposition of callose, the physical thickening of cell walls by lignification, and the synthesis of various antibiotic small molecules and proteins. Genetic factors in both the host and the pathogen determine the specificity of these local responses, which can be very effective in limiting spread of the infection.


Resistance to Fusarium, as an example, is a polygenic trait and can be seen as consisting of two major components: (1) resistance to initial penetration, and (2) resistance to the spreading of the pathogen in host tissue. Though there is no evidence of complete resistance to Fusarium stalk rot in maize, genetic variation for resistance exists within maize germplasm. Resistance to C. graminicola is also primarily quantitative, although a few sources of major gene resistance have been described.


The constantly growing volume of research regarding the mechanisms involved in plant resistance to pathogens and the genetic basis of such mechanism, together with advances in biotechnology have presented new opportunities for protecting plants against pathogen infection through genetic engineering.


Many genes have been identified to participate in the plant defense mechanisms. For example, Sanghyun S et al. (2008. J Exp Bot. 2008:59(9):2371-8) showed that transgenic wheat expressing a barley class II chitinase exhibited enhanced resistance against F. graminearum in greenhouse and field conditions. Zhu X et al. (2012. Funct Integr Genomics. 12(3):481-488) described that overexpression of wheat lipid transfer protein gene TaLTP5 increases resistances to Cochiobolus sativus and Fusarium graminearum in transgenic wheat. Perochon A et al. (2015. Plant Physiol 169(4):2895-2906) reported the functional characterization of an orphan gene (Triticum aestivum Fusarium Resistance Orphan Gene [TaFROG]) as a component of resistance to Fusarium head blight (FHB). Zuo D Y et al. (2016. Phytopatol. 106(6):614-623) showed that a deoxynivalenol-activated methionyl-tRNA synthetase gene from wheat encodes a nuclear localized protein and protects plants against Fusarium pathogens infection and mycotoxins. Dowd P F and Johnson E T (2016. J Plant Res. 129(1):13-20) showed that the maize peroxidase Px5, the pericarp expression of which has been shown to be associated with resistance to Aspergillus flavus growth and to insects in a set of inbred plant lines has a highly conserved sequence which enhances fungal and insect resistance.


Many defense response genes are induced in wheat and other plants during F. graminearum infection and may play a role in reducing FHB. These response genes were thus investigated in an attempt to produce resistant wheat lines. Mackintosh C A et al. (2007. Plant Cell Rep 26(4):479-488) examined overexpression of the defense response genes alpha-1-purothionin, thaumatin-like protein 1 (tlp-1), and beta-1,3-glucanase in wheat, and reported that all the genes reduced at least part of the disease symptoms. A beta-1,3-glucanase transgenic line had enhanced resistance, showing lower FHB severity, deoxynivalenol (DON) mycotoxin concentration, and percent of visually scabby kernels (VSK) compared to a control plant. Sasaki K et al. (2016. J Biotechnol 228:3-7) also showed that overexpression of TAD1 (Triticum aestivum defensin 1), a protein induced during cold acclimation in winter wheat and encoding a plant defensin with antimicrobial activity, increased resistance against Fusarium graminearum in the transformed wheat plants.


Various additional genetic manipulation of gene expression for improving resistance to pathogenic fungi has also been reported. For example, Brewer H C et al. (2014. BMC Plant Biol 14(1):317) showed that mutations in the Arabidopsis homoserine kinase gene DMR1 confer enhanced resistance to F. culmorum and F. graminearum. Tundo S et al. (2016. Mol Plant Microbe Interact 29(8):629-639) produced transgenic plants with a combination of genes encoding proteins involved in inhibiting the activity of cell wall-degrading enzymes (CWDEs) secreted by pathogens to breach the plant cell-wall barrier. They showed that pyramiding polygalacturonase (PG) inhibiting protein (PGIP2) and TAXI-III, a xylanase inhibitor that controls the activity of xylanases, enhanced resistance against Fusarium graminearum, while pyramiding PGIP2 and pectin methyl esterase inhibitor (PMEI) did not reach the same effect. Li X et al. (2015. Mol Plant Microbe Interact 28(11):1237-1246) demonstrated that transgenic wheat expressing a barley UDP-glucosyltransferase detoxifies deoxynivalenol and provides high levels of resistance to Fusarium graminearum.


Among others, International Application Publication Nos. WO/2006/085965 and WO 2006/091219 disclose methods for protecting plants from plant pathogenic fungi by enhancing fungal pathogen resistance in a plant using the nucleotide sequences disclosed therein. Further disclosed therein are methods comprising introducing into a plant an expression cassette comprising a promoter operably linked to a nucleotide sequence that encodes an antifungal polypeptide as well as transformed plants, plant cells, seeds, and microorganisms comprising a nucleotide sequence that encodes an antifungal polypeptide or variant or fragment thereof.


U.S. Pat. No. 9,359,615 discloses plants which overexpress a p33kD or BURP protein, or an ortholog thereof, and exhibit an increased pre-formed resistance to pathogens, particularly fungal pathogens.


U.S. Pat. No. 9,732,354 discloses new gene that is able to provide plants with resistance against pathogens, particularly Verticillium, Ralstonia or Fusarium. The gene is typical for Brassicaceae, but may confer resistance to other plants. Further provided are host cells with a nucleotide construct encoding the protein and methods for enhancing the pathogen resistance of plants.


International (PCT) Patent Application Publication No. WO 2018/131037 to the Applicant of the present invention discloses polynucleotides and polypeptides associated with increased resistance of plant to pathogenic fungi, particularly to fungi inducing root rot and stalk rot in plants, and use thereof for controlling plant fungal pathogens and for producing transgenic plants having increased resistance to pathogenic fungi.


Intact microorganisms, particularly bacteria, which can survive on or be toxic to fungi, have been used as biocontrol agents. For example, U.S. Pat. No. 9,485,994 discloses methods and compositions for control of pathogenic fungal or Oomycetous infection. Particularly, the patent discloses an antifungal or an anti-Oomycetous composition comprising bacteria of the genus Collimonas and bacteria of the genus Bacillus, together exhibiting a synergistic antifungal or a synergistic anti-Oomycetous effect, and methods of use thereof. However, it is difficult both to apply biological organisms to large areas and to cause such living organisms to remain in the treated area or on the treated plant species for an extended time, and therefore use of intact bacteria as biocontrol agents is still limited.


Therefore, attempts were made to identify bacterial proteins and genes encoding same responsible for the biocontrol activity. Targets, mechanism of action and prospective applications of anti-fungal proteins of various organisms, including bacteria are reviewed, for example, by Theis and Stahl (Theis T and Stahl U. 2004 CMLS, Cell. Mol. Life Sci. 61:437-455). Proteomic approach taken to investigate Bacillus strains having anti-fungal activity revealed the presence of lytic enzymes, cellulases, proteases, 1,4-β-glucanases and hydrolases, all of which contribute to degradation of the pathogen cell wall. Further proteomic investigations showed that proteins involved in metabolism, protein folding, protein degradation, translation, recognition and signal transduction cascade play an important role in the control of Fusarium oxysporum (Baysal O. et al., 2013. Plose One 8(1):e53182). Li et al., (Li Jing et al., J Zhejiang Univ Sci B 10(4):264-272) describe the purification of a particular antifungal protein from Bacillus subtilis, designated B29I, which exhibited inhibitory activity on mycelial growth of Fusarium oxysporum, Rhizoctonia solani, Fusarium moniliforme, and Sclerotinia sclerotiorum. Yadav et al. (Yadav V et al., 2010. Intl J Microbiol, Article ID 196363, doi:10.1155/2010/196363) described the isolation of an antifungal protein from Escherichia coli BL21 (PPEBL21), predicted to be alcohol dehydrogenase (ADH). Additional example of enzymes having a significant role in the anti-fungal activity of bacteria includes chitinases (see, e.g. Hjort K et al., 2013. Appl Microbiol Biotechnol DOI 10.1007/s00253-013-5287-x).


Song et al. (Song C et al., 2015. BMC Genomics 16:1103) explored the genomic traits of the fungus-feeding bacterial genus Collimonas, and Haack et al. (Haack F S et al., 2016. Frontiers in Microbiology 7 Article 1668) investigated the molecular keys of the interactions of Janthinobacterium and Duganella species with the plant pathogenic fungi Fusarium graminearum.


However, as of today, there is a need for further developed methods and compositions for protecting plants from fungal pathogen.


SUMMARY OF THE INVENTION

The present invention provides isolated polynucleotides, constructs comprising same and isolated polypeptides useful in conferring and/or enhancing resistance of a plant towards pathogenic fungi and/or Oomycetes. The present invention further provides genetically altered plants and plant cells with enhanced resistance to pathogenic fungi and/or Oomycetes and methods for producing and selecting same as well as compositions comprising the polypeptides of the invention useful in protecting plants from the pathogenic fungi and/or Oomycetes.


According to one aspect, the present invention provides a method for enhancing the resistance of a plant or a part thereof to at least one pathogenic fungus and/or Oomycete, comprising modulating the expression and/or activity of at least one polypeptide at least 80% identical to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:326, 360, 308-325, 327-359, 361-506 within at least one cell of the plant or part thereof, thereby enhancing the resistance of said plant or part thereof to the at least one pathogenic fungus and/or Oomycete compared to the resistance of a control plant. Each possibility represents a separate embodiment of the present invention.


According to certain embodiments, the method comprises modulating the expression and/or activity of at least one polypeptide having the amino acid sequence selected from the group consisting of SEQ ID NOs:326, 360, 308-325, 327-359, 361-508. Each possibility represents a separate embodiment of the present invention.


According to certain embodiments, the at least one polypeptide is encoded by a polynucleotide comprising a nucleic acid sequence at least 80% identical to a nucleic acid sequence selected from the group consisting of SEQ ID NOs:40, 80, 22-39, 41-79, 81-301. Each possibility represents a separate embodiment of the present invention.


According to other embodiments, the at least one polypeptide is encoded by a polynucleotide comprising the nucleic acid sequence set forth in any one of SEQ ID NOs:40, 80, 22-39, 41-79, 81-305. Each possibility represents a separate embodiment of the present invention.


According to certain embodiments, enhancing the resistance of the plant or part thereof to the pathogenic fungus and/or Oomycete comprises enhancing the expression and/or activity of at least one polypeptide compared to its expression and/or activity in the control plant.


According to certain exemplary embodiments, the polypeptide the expression and/or activity of which is to be enhanced comprises an amino acid sequence at least 80% identical to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:326, 360, 308-325, 327, 329-347, 349-359, 362-506. Each possibility represents a separate embodiment of the present invention. According to some embodiments, the polypeptide the expression and/or activity of which is to be enhanced comprises an amino acid sequence selected from the group consisting of SEQ ID NOs:326, 360, 308-325, 327, 329-347, 349-359, 362-508. Each possibility represents a separate embodiment of the present invention.


According to certain embodiments, the polypeptide the expression and/or activity of which is to be enhanced is an endogenous polypeptide of the at least one cell. According to other embodiments, the polypeptide the expression and/or activity of which is to be enhanced is a polypeptide heterologous to the at least one plant cell. According to certain embodiments, enhancing the expression and/or activity of the polypeptide comprises modulating the expression of an endogenous polynucleotide encoding said polypeptide within the at least one cell of the plant or part thereof.


Modulating, according to certain embodiments enhancing, the expression of the endogenous polynucleotide can be affected at the genomic and/or the transcript level using a variety of methods that induce the transcription and/or translation of the polypeptide.


According to certain embodiments, enhancing the expression and/or activity of the endogenous polypeptide comprises subjecting the at least one cell of the plant or part thereof to genome editing using artificially engineered nucleases as is known in the art.


According to certain embodiments, isolated nucleic acids which serve as promoter or enhancer elements may be introduced in an appropriate position as to enhance transcription of the endogenous polynucleotide. According to some embodiments, the regulatory element is selected from the group consisting of, but not limited to, a promoter and an enhancer.


According to certain embodiments, the polypeptide the expression of which is to be enhanced is encoded by a polynucleotide at least 80% identical to a nucleic acid sequence selected from the group consisting of SEQ ID NOs:40, 80, 22-39, 41, 43-61, 63-79, 81, 83-301. According to some embodiments, the polypeptide the expression of which is to be enhanced is encoded by a polynucleotide comprising a nucleic acid sequence selected from the group consisting of SEQ ID NOs:40, 80, 22-39, 41, 43-61, 63-79, 81, 83-305.


According to other embodiments, at least one mutation may be inserted within the endogenous polynucleotide as long as the mutation results in enhanced expression of the encoded polypeptide. Any method for mutagenesis as is known in the art can be used according to the teachings of the present invention including chemical mutagenesis, radio-mutagenesis and site directed mutagenesis, for example using genome editing techniques.


According to certain embodiments, enhancing the resistance of the plant to the pathogenic fungus and/or Oomycete comprises reducing the expression of at least one polypeptide compared to its expression and/or activity in the control plant. According to certain exemplary embodiments, the polypeptide the expression and/or activity of which is to be reduced comprises an amino acid sequence at least 80% identical to the amino acid sequence set forth in any one of SEQ ID NO:328, SEQ ID NO:361, and SEQ ID NO:348. According to further exemplary embodiments, the polypeptide the expression and/or activity of which is to be reduced comprises the amino acid sequence set forth in any one of SEQ ID NO:328, SEQ ID NO:361 and SEQ ID NO:348.


According to certain embodiments, enhancing the resistance of the plant to the pathogenic fungus and/or Oomycete comprises reducing the expression and/or activity of at least one polynucleotide compared to its expression in the control plant. According to certain exemplary embodiments, the polynucleotide having reduced expression comprises a nucleic acid sequence at least 80% identical to the nucleic acid sequence set forth in any one of SEQ ID NO:42, SEQ ID NO:82, and SEQ ID NO:62. According to certain exemplary embodiments, the polynucleotide having reduced expression comprises the nucleic acid sequence set forth in any one of SEQ ID NO:42, SEQ ID NO:82 and SEQ ID NO:62.


Any method as is known in the art for reducing the expression and/or activity of a plant endogenous protein and the polynucleotide encoding same can be used according to the teachings of the resent invention.


According to certain embodiments, reducing the expression and/or activity of the polypeptide comprises down-regulating the expression of the endogenous polynucleotide encoding said polypeptide within the at least one cell of the plant or part thereof.


According to certain embodiments, reducing the expression and/or activity of the polypeptide comprises modulating the endogenous polynucleotide as to encode a non-functional polypeptide.


According to certain embodiments, expression of the polynucleotide is affected at the genomic and/or the transcript level using a variety of molecules that interfere with transcription and/or translation (e.g., antisense, siRNA, Ribozyme, or DNAzyme) of the polynucleotide. Inserting a mutation into the polynucleotide, including deletions, insertions, site specific mutations, mutations mediated by artificially engineered nucleases (including zinc-finger nucleases (ZFNs), transcription-activator like effector nucleases (TALENs) and CRISPR/Cas system) can be also used, as long as the mutations result in down-regulation of the gene expression or in the production of non-functional protein. Alternatively, expression can be inhibited at the protein level using, e.g., antagonists, enzymes that cleave the polypeptide, and the like.


According to additional aspect, the present invention provides a method for enhancing the resistance of a plant or a part thereof to at least one pathogenic fungus and/or Oomycete, comprising enhancing the expression and/or activity of at least one polypeptide at least 70% identical to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:2635, 2444-2634, 2636-4252, an active fragment and/or a variant thereof within at least one cell of the plant or part thereof, thereby enhancing the resistance of said plant or part thereof to the at least one pathogenic fungus and/or Oomycete compared to the resistance of a control plant. Each possibility represents a separate embodiment of the present invention.


According to certain embodiments, the method comprises enhancing the expression and/or activity of at least one polypeptide having the amino acid sequence selected from the group consisting of SEQ ID NOs:2635, 2444-2634, 2636-4252, an active fragment and/or a variant thereof. Each possibility represents a separate embodiment of the present invention.


According to the certain exemplary embodiments, the fragment comprises an amino acid sequence selected from the group consisting of SEQ ID NOs:4661, 4647-4660, and 4662-4799.


According to certain embodiments, the at least one polypeptide is encoded by a polynucleotide comprising a nucleic acid sequence at least 70% identical to a nucleic acid sequence selected from the group consisting of SEQ ID NOs:783, 592-782, 784-2443, a fragment and/or a variant thereof. Each possibility represents a separate embodiment of the present invention.


According to other embodiments, the at least one polypeptide is encoded by a polynucleotide comprising the nucleic acid sequence set forth in any one of SEQ ID NOs:783, 592-782, 784-2443, a fragment and/or a variant thereof. Each possibility represents a separate embodiment of the present invention.


Enhancing the polypeptide expression can be affected at the genomic and/or the transcript and/or translation level using a variety of methods that induce the transcription and/or translation of the polypeptide.


According to certain embodiments, enhancing the expression and/or activity of the polypeptide comprises expressing an exogenous polynucleotide encoding said at least one polypeptide within the at least one cell of the plant or the part thereof.


According to certain embodiments, enhancing the expression and/or activity of the polypeptide comprises transforming at least one cell of the plant or part thereof with an exogenous polynucleotide encoding the polypeptide, thereby producing a transgenic plant over-expressing said polypeptide.


Any method as is known in the art for introducing an exogenous polynucleotide into a plant cell can be used according to the teachings of the present invention. According to some embodiments, the exogenous polynucleotide is transformed into the plant cell using a suitable vector.


According to certain embodiments, genome editing is employed to edit the genome of the at least one cell as to express a heterologous polypeptide of the invention.


According to certain exemplary embodiments, the polynucleotide the expression of which is to be enhanced comprises a nucleic acid sequence at least 80% identical to a nucleic acid sequence selected from the group consisting of SEQ ID NOs:40, 80, 22-39, 41, 43-61, 63-79, 81, and 83-301. Each possibility represents a separate embodiment of the present invention. According to some embodiments, the polynucleotide the expression of which is to be enhanced comprises a nucleic acid sequence selected from the group consisting of SEQ ID NOs:40, 80, 22-39, 41, 43-61, 63-79, 81, and 83-305.


According to further exemplary embodiments, the polynucleotide the expression of which is to be enhanced comprises a nucleic acid sequence at least 70% identical to a nucleic acid sequence selected from the group consisting of SEQ ID NOs:783, 592-782, 784-2443 and a fragment thereof. According to some embodiments, the polynucleotide the expression of which is to be enhanced comprises a nucleic acid sequence selected from the group consisting of SEQ ID NOs:783, 592-782, 784-2443 and a fragment thereof.


Gene stacking is often desirable in the field of plant genetic engineering, leading to a desired phenotype and/or improvement of a desired phenotype. According to certain aspects, the present invention provides a method for enhancing the resistance of a plant or a part thereof to at least one pathogenic fungus and/or Oomycete, comprising transforming at least one cell of the plant or part thereof with at least one DNA construct comprising nucleic acid sequence encoding at least two polypeptides, wherein each polypeptide is capable of enhancing resistance to the at least one pathogenic fungus and/or Oomycete, thereby enhancing the resistance of said plant or part thereof to said at least one pathogenic fungus and/or Oomycete compared to the resistance of a control plant. According to certain embodiments, the present invention provides a method for enhancing the resistance of a plant or a part thereof to at least one pathogenic fungus and/or Oomycete, comprising transforming at least one cell of a plant or a part thereof with a DNA construct comprising a first polynucleotide encoding a polypeptide having the amino acid sequence set forth in SEQ ID NO:311 and a second polynucleotide encoding a polypeptide designated LFS57 having the amino acid sequence set forth in SEQ ID NO:617 of International Application Publication No. WO 2018/131037, thereby enhancing the resistance of said plant or part thereof to the at least one pathogenic fungus and/or Oomycete compared to the resistance of a control plant.


According to certain embodiments, the first polynucleotide comprises the nucleic acid sequence set forth in SEQ ID NO:66 and the second polynucleotide comprises the nucleic acid sequence set forth in any one of SEQ ID NO:101 and SEQ ID NO:165 of WO 2018/131037.


According to certain embodiments, the present invention provides a method for enhancing the resistance of a plant or a part thereof to at least one pathogenic fungus and/or Oomycete, comprising transforming at least one cell of a plant or a part thereof with a DNA construct comprising a first polynucleotide encoding a polypeptide having the amino acid sequence set forth in SEQ ID NO:310 and a second polynucleotide encoding a polypeptide designated LFS46 having the amino acid sequence set forth in SEQ ID NO:608 of International Application publication No. WO 2018/131037, thereby enhancing the resistance of said plant or part thereof to the at least one pathogenic fungus and/or Oomycete compared to the resistance of a control plant.


According to certain embodiments, the first polynucleotide comprises the nucleic acid sequence set forth in SEQ ID NO:65 and the second polynucleotide comprises the nucleic acid sequence set forth in any one of SEQ ID NO:92 and SEQ ID NO:157 of WO 2018/131037.


According to some embodiments, the control plant is a plant not manipulated to have modulated expression and/or activity of the polypeptide. According to some embodiments, the control plant is of the same species. According to some embodiments, the control plant comprises the same genetic background.


According to another aspect, the present invention provides a method for producing a population of plants each having an enhanced resistance to at least one pathogenic fungus and/or Oomycete, comprising the steps of:

    • (a) modulating the expression and/or activity of at least one polypeptide at least 80% identical to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:326, 360, 308-325, 327-359, and 361-506 within at least one cell of each plant of a plant population as to produce a genetically engineered plant population;
    • (b) inoculating each plant of the genetically engineered plant population with the at least one pathogenic fungus and/or Oomycete; and
    • (c) selecting plants showing an enhanced resistance to said at least one pathogenic fungus and/or Oomycete compared to a control plant or to a pre-determined resistance score value;


thereby producing a population of genetically engineered plants having enhanced resistance to said at least one pathogenic fungus and/or Oomycete.


According to certain embodiments, the method comprises modulating the expression and/or activity of at least one polypeptide having the amino acid sequence set forth in any one of SEQ ID NOs:326, 360, 308-325, 327-359, 361-508. According to certain embodiments, the method comprises modulating the expression and/or activity of at least one polypeptide having the amino acid sequence set forth in any one of SEQ ID NOs:507-508. According to certain embodiments, the method comprises modulating the expression and/or activity of at least one polypeptide having the amino acid sequence set forth in any one of SEQ ID NOs:507-509. According to other embodiments, the method comprises modulating the expression and/or activity of at least one polypeptide having the amino acid sequence set forth in SEQ ID NO:509. Each possibility represents a separate embodiment of the present invention.


According to certain embodiments, the method comprises enhancing the expression and/or activity of the at least one polypeptide. According to these embodiments, the method comprises enhancing the expression and/or activity of at least one polypeptide at least 80% identical to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:326, 360, 308-325, 327, 329-347, 349-359, 362-506. According to some embodiments, the method comprises enhancing the expression and/or activity of at least one polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:326, 360, 308-325, 327, 329-347, 349-359, 362-509. Each possibility represents a separate embodiment of the present invention.


According to certain embodiments, the method comprises reducing the expression and/or activity of the at least one polypeptide. According to these embodiments, the method comprises reducing the expression and/or activity of at least one polypeptide at least 80% identical to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:328, SEQ ID NO:361 and SEQ ID NO:348. According to some embodiments, the method comprises reducing the expression and/or activity of at least one polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:328, SEQ ID NO:361 and SEQ ID NO:348.


According to yet another aspect, the present invention provides a method for producing a population of plants each having an enhanced resistance to at least one pathogenic fungus and/or Oomycete, comprising the steps of:

    • (a) enhancing the expression and/or activity of at least one polypeptide at least 70% identical to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:2635, 2444-2634, 2636-4252, an active fragment and/or variant thereof within at least one cell of each plant of a plant population as to produce a genetically engineered plant population;
    • (b) inoculating each plant of the genetically engineered plant population with the at least one pathogenic fungus and/or Oomycete; and
    • (c) selecting plants showing an enhanced resistance to said at least one pathogenic fungus and/or Oomycete compared to a control plant or to a pre-determined resistance score value;


thereby producing a population of genetically engineered plants having enhanced resistance to said at least one pathogenic fungus and/or Oomycete.


According to certain embodiments, the method comprises modulating the expression and/or activity of at least one polypeptide having the amino acid sequence set forth in any one of SEQ ID NOs:2635, 2444-2634, 2636-4252, an active fragment and/or a variant thereof.


According to the certain exemplary embodiments, the polypeptide fragment comprises an amino acid sequence selected from the group consisting of SEQ ID NOs:4661, 4647-4660, and 4662-4799. The methods by which the expression and/or activity of the at least polypeptide can be enhanced or reduced are as is known in the art and as described hereinabove.


According to certain embodiments, the method comprises modulating the expression of at least one polynucleotide comprising a nucleic acid sequence at least 80% identical to a nucleic acid sequence selected from the group consisting of SEQ ID NOs:40, 80, 22-39, 41-79, 81-301. According to some embodiments, the method comprises modulating the expression of at least one polynucleotide comprising a nucleic acid sequence selected from the group consisting of SEQ ID NOs:40, 80, 22-39, 41-79, and 81-305. According to some embodiments, the method comprises modulating the expression of at least one polynucleotide comprising a nucleic acid sequence selected from the group consisting of SEQ ID NOs:304-305. According to some embodiments, the method comprises modulating the expression of at least one polynucleotide comprising a nucleic acid sequence selected from the group consisting of SEQ ID NOs:304-306. According to some embodiments, the method comprises modulating the expression of at least one polynucleotide comprising a nucleic acid sequence set forth in SEQ ID NO:306. Each possibility represents a separate embodiment of the present invention. According to some embodiments, modulating the expression of the at least one polynucleotide comprises enhancing its expression. According to these embodiments, the polynucleotide is at least 80% identical to a polynucleotide having the nucleic acid sequence selected from the group consisting of 40, 80, 22-39, 41, 43-61, 63-79, 81, and 83-301. According to some embodiments, the the polynucleotide comprises the nucleic acid sequence selected from the group consisting of SEQ ID NOs:40, 80, 22-39, 41, 43-61, 63-79, 81, and 83-306. Each possibility represents a separate embodiment of the present invention.


According to certain embodiments, enhancing the expression comprises transforming the at least one cell of the plant or part thereof with a polynucleotide at least 80% identical to a polynucleotide having an amino acid sequence selected from the group consisting of SEQ ID NOs:40, 80, 22-39, 41-79, 81, and 83-301. According to certain embodiments, enhancing the expression comprises transforming the at least one cell of the plant or part thereof with a polynucleotide comprising a nucleic acid sequence selected from the group consisting of SEQ ID NOs:40, 80, 22-39, 41-79, 81, and 83-306.


According to some embodiments, modulating the expression of the at least one polynucleotide comprises reducing its expression. According to these embodiments, the polynucleotide is at least 80% identical to a polynucleotide having the nucleic acid sequence selected from the group consisting of SEQ ID NO:42, SEQ ID NO:82 and SEQ ID NO:62. According to some embodiments, the the polynucleotide comprises the nucleic acid sequence selected from the group consisting of SEQ ID NO:42, SEQ ID NO:82 and SEQ ID NO:62.


According to certain embodiments, the method comprises enhancing the expression of at least one polynucleotide comprising a nucleic acid sequence at least 70% identical to a nucleic acid sequence selected from the group consisting of SEQ ID NOs:783, 592-782, 784-2443, a variant and/or a fragment thereof. According to some embodiments, the method comprises enhancing the expression of at least one polynucleotide comprising a nucleic acid sequence selected from the group consisting of SEQ ID NOs:783, 592-782, 784-2443, a variant and/or a fragment thereof.


The methods by which the expression of the at least one polynucleotide can be enhanced or reduced are as is known in the art and as described hereinabove.


According to additional aspect, the present invention provides a method for selecting a plant having an enhanced resistance to at least one pathogenic fungus and/or Oomycete, comprising the steps of

    • (a) providing a plurality of plants each comprising at least one cell with modulated expression and/or activity of a polypeptide at least 80% identical to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:326, 360, 308-325, 327-359, 361-506.
    • (b) inoculating the plurality of plants with the at least one pathogenic fungus and/or Oomycete; and
    • (c) selecting plants showing an enhanced resistance to said at least one pathogenic fungus and/or Oomycete compared to a control plant or to a pre-determined resistance score value.


According to certain embodiments, the method comprises providing a plurality of plants each having a modulated expression and/or activity of at least one polypeptide having the amino acid sequence set forth in any one of SEQ ID NOs:326, 360, 308-325, 327-359, 361-508. Each possibility represents a separate embodiment of the present invention. According to other embodiments, the method comprises providing a plurality of plants each having a modulated expression and/or activity of a polypeptide having the amino acid sequence set forth in any one of SEQ ID NOs:507-509. Each possibility represents a separate embodiment of the present invention. According to some embodiments, the method comprises providing a plurality of plants each having a modulated expression and/or activity of a polypeptide having the amino acid sequence set forth in SEQ ID NO:509.


According to certain embodiments, the method comprises providing a plurality of plants each having modulated expression of at least one polynucleotide comprising a nucleic acid sequence at least 80% identical to a polynucleotide having the nucleic acid sequence set forth in any one of SEQ ID NOs:40, 80, 22-39, 41-79, 81-301. According to some embodiments, the at least one polynucleotide comprises a nucleic acid sequence selected from the group consisting of SEQ ID NOs:40, 80, 22-39, 41-79, 81-305. Each possibility represents a separate embodiment of the present invention. According to some embodiments, the at least one polynucleotide comprises a nucleic acid sequence set forth in SEQ ID NO:306.


According to certain embodiments, the modulated expression and/or activity is selected from enhanced expression and/or activity and reduced expression and/or activity. Modulating the expression can be performed by any method as is known in the art and as described hereinabove. The polypeptides and polynucleotides the expression of which is to be enhanced or reduced are as described hereinabove. According to yet further aspect, the present invention provides a method for selecting a plant having an enhanced resistance to at least one pathogenic fungus and/or Oomycete, comprising the steps of:

    • (a) providing a plurality of plants each comprising at least one cell with enhanced expression and/or activity of a polypeptide at least 70% identical to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:2635, 2444-2634, 2636-4252, an active fragment and/or a variant thereof;
    • (b) inoculating the plurality of plants with the at least one pathogenic fungus and/or Oomycete; and
    • (c) selecting plants showing an enhanced resistance to said at least one pathogenic fungus and/or Oomycete compared to a control plant or to a pre-determined resistance score value.


According to certain embodiments, the method comprises providing a plurality of plants each having an enhanced expression and/or activity of at least one polypeptide having the amino acid sequence set forth in any one of SEQ ID NOs:2635, 2444-2634, 2636-4252, an active fragment and/or a variant thereof. Each possibility represents a separate embodiment of the present invention.


According to the certain exemplary embodiments, the fragment comprises an amino acid sequence selected from the group consisting of SEQ ID NOs:4661, 4647-4660, and 4662-4799. Each possibility represents a separate embodiment of the present invention.


According to certain embodiments, the method comprises providing a plurality of plants each having enhanced expression of at least one polynucleotide comprising a nucleic acid sequence at least 70% identical to a polynucleotide having the nucleic acid sequence set forth in any one of SEQ ID NOs:783, 592-782, 784-2443. According to some embodiments, the at least one polynucleotide comprises a nucleic acid sequence selected from the group consisting of SEQ ID NOs:4783, 592-782, 784-2443.


According to certain embodiments, the control plant is a plant not manipulated to have modulated expression and/or activity of the polypeptide. According to some embodiments, the control plant is of the same species. According to some embodiments, the control plant comprises the same genetic background.


According to certain embodiments, the pre-determined resistance score value is obtained by a method comprising the steps of inoculating a plurality of corresponding plants susceptible to the at least one pathogenic fungus or Oomycete; scoring the infection degree; and setting an average resistance score value.


Modulating (enhancing or reducing) the expression and/or activity of the polypeptide can be achieved as described hereinabove and by any other method as is known in the art.


According to certain embodiments, the plant part is selected from the group consisting of seeds, roots, shoots, leaves, flowers and the like. Each possibility represents separate embodiment of the present invention. According to certain exemplary embodiments, the plant part is a root. Tissue cultures comprising cells derived from the plant having a modulated expression and/or activity of a polypeptide of the invention are also encompassed within the scope of the present invention.


According to additional aspect, the present invention provides a genetically engineered plant having enhanced resistance to at least one fungus and/or Oomycete compared to a non-engineered control plant, the genetically engineered plant comprising at least one cell having modified expression and/or activity of at least one polypeptide at least 80% identical to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:326, 360, 308-325, 327-359, 361-506 compared to the polypeptide expression and/or activity in the non-engineered control plant. Each possibility represents a separate embodiment of the present invention.


According to certain embodiments, the genetically engineered plant comprises at least one cell having enhanced expression and/or activity of at least one polypeptide at least 80% identical to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:326, 360, 308-325, 327, 329-347, 349-359, 362-506.


According to certain embodiments, the genetically engineered plant comprises at least one cell having reduced expression and/or activity of at least one polypeptide at least 80% identical to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:328, 361 and 348.


According to additional aspect, the present invention provides a genetically engineered plant having enhanced resistance to at least one fungus and/or Oomycete compared to a non-engineered control plant, the genetically engineered plant comprising at least one cell having enhanced expression and/or activity of at least one polypeptide at least 70% identical to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:2635, 2444-2634, 2636-4252, an active fragment or a variant thereof compared to the polypeptide expression and/or activity in the non-engineered control plant. Each possibility represents a separate embodiment of the present invention.


According to certain embodiments, the genetically engineered plant comprises at least one cell having modified expression and/or activity of at least one polypeptide having the amino acid sequence selected from the group consisting of SEQ ID NOs:326, 360, 308-325, 327-359, 361-508. Each possibility represents a separate embodiment of the present invention. According to certain embodiments, the genetically engineered plant comprises at least one cell having modified expression and/or activity of at least one polypeptide having the amino acid sequence selected from the group consisting of SEQ ID NOs:507-508. Each possibility represents a separate embodiment of the present invention.


According to certain embodiments, the genetically engineered plant comprises at least one cell having enhanced expression and/or activity of at least one polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:326, 360, 308-325, 327, 329-347, 349-359, 362-508. Each possibility represents a separate embodiment of the present invention.


According to certain embodiments, the genetically engineered plant comprises at least one cell having reduced expression and/or activity of at least one polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:328, 361, and 348. Each possibility represents a separate embodiment of the present invention.


According to certain embodiments, the genetically engineered plant comprises at least one cell having enhanced expression and/or activity of at least one polypeptide having the amino acid sequence selected from the group consisting of SEQ ID NOs:2635, 2444-2634, 2636-4252, an active fragment and/or a variant thereof. Each possibility represents a separate embodiment of the present invention.


According to certain embodiments, the genetically engineered plant comprises at least one cell having modified expression of at least one polynucleotide comprising a nucleic acid sequence at least 80% identical to a polynucleotide having the nucleic acid sequence set forth in any one of SEQ ID NOs:40, 80, 22-39, 41-79, 81-301. According to some embodiments, the at least one polynucleotide comprises a nucleic acid sequence selected from the group consisting of SEQ ID NOs:40, 80, 22-39, 41-79, 81-305. According to some embodiments, the at least one polynucleotide comprises a nucleic acid sequence selected from the group consisting of SEQ ID NOs:302-305. Each possibility represents a separate embodiment of the present invention.


According to certain embodiments, the genetically engineered plant comprises at least one cell having enhanced expression of at least one polynucleotide comprising a nucleic acid sequence at least 80% identical to a polynucleotide having the nucleic acid sequence set forth in any one of SEQ ID NOs: 40, 80, 22-39, 41, 43-61, 63-79, 81, and 83-301. Each possibility represents a separate embodiment of the present invention.


According to certain embodiments, the genetically engineered plant comprises at least one cell having reduced expression of at least one polynucleotide comprising a nucleic acid sequence at least 80% identical to a polynucleotide having the nucleic acid sequence set forth in any one of SEQ ID NOs:42, 82 and 62. Each possibility represents a separate embodiment of the present invention.


According to certain embodiments, the genetically engineered plant comprises at least one cell having enhanced expression of at least one polynucleotide comprising a nucleic acid sequence at least 70% identical to a polynucleotide having the nucleic acid sequence set forth in any one of SEQ ID NOs:783, 592-782, 784-2443. According to some embodiments, the at least one polynucleotide comprises a nucleic acid sequence selected from the group consisting of SEQ ID NOs:783, 592-782, 784-2443. Each possibility represents a separate embodiment of the present invention.


According to certain embodiments, the genetically engineered plant comprises a polynucleotide encoding a modified form of the at least one polypeptide, wherein the modified form has reduced or no activity compared to the unmodified form, thereby having an enhanced resistance to the at least one fungus.


According to additional aspect, the present invention provides an isolated or recombinant polynucleotide encoding a polypeptide comprising an amino acid sequence at least 80% identical to an amino acid sequence selected from the group consisting of SEQ ID NOs:326, 360, 308-325, 327-359, and 362-506, wherein the polypeptide, when expressed in a plant, is capable of modulating the resistance of the plant to at least one pathogenic fungus and/or Oomycete. According to certain embodiments, the polynucleotide encodes a polypeptide comprising the amino acid sequence set forth in any one of SEQ ID NOs:326, 360, 308-325, 327-359, 362-506. According to certain embodiments, the polynucleotide encodes a polypeptide comprising the amino acid sequence set forth in any one of SEQ ID NOs:507-508. Each possibility represents a separate embodiment of the present invention.


According to certain embodiments, the polypeptide, when expressed in a plant, is capable of enhancing the resistance of the plant to at least one pathogenic fungus and/or Oomycete, said polypeptide is at least 80% identical to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:326, 360, 308-325, 327, 329-347, 349-359, and 362-506. According to certain embodiments, said polypeptide comprises an amino acid sequence selected from the group consisting of SEQ ID NOs:326, 360, 308-325, 327, 329-347, 349-359, and 362-508. Each possibility represents a separate embodiment of the present invention.


According to certain embodiments, the polynucleotide encoding a polypeptide capable of modulating the resistance of the plant to at least one pathogenic fungus and/or Oomycete when expressed in said plant comprises a nucleic acid sequence at least 80% identical to a nucleic acids sequence set forth in any one of SEQ ID NOs:40, 80, 22-39, 41-79, 81-301. According to other embodiments, the polynucleotide comprises the nucleic acid sequence set forth in any one of SEQ ID NOs:40, 80, 22-39, 41-79, 81-301. According to yet further embodiments, the polynucleotide comprises the nucleic acid sequence set forth in any one of SEQ ID NOs:302-305.


According to certain embodiments, the polynucleotide encoding a polypeptide capable of enhancing the resistance of the plant to at least one pathogenic fungus and/or Oomycete when expressed in said plant comprises a nucleic acid sequence at least 80% identical to a nucleic acids sequence set forth in any one of SEQ ID NOs: 40, 80, 22-39, 41, 43-61, 63-79, 81, and 83-301. According to other embodiments, the polynucleotide comprises the nucleic acid sequence set forth in any one of SEQ ID NOs: 40, 80, 22-39, 41, 43-61, 63-79, 81, and 83-301. According to yet further embodiments, the polynucleotide comprises the nucleic acid sequence set forth in any one of SEQ ID NOs:302-305.


According to yet additional aspect, the present invention provides an isolated or recombinant polynucleotide encoding a polypeptide comprising an amino acid sequence at least 70% identical to an amino acid sequence selected from the group consisting of SEQ ID NOs:2635, 2444-2634, 2636-4252, wherein the polypeptide, when expressed in a plant, is capable of enhancing the resistance of the plant to at least one pathogenic fungus and/or Oomycete. According to certain embodiments, the polynucleotide encodes a polypeptide comprising the amino acid sequence set forth in any one of SEQ ID NOs:2635, 2444-2634, 2636-4252.


According to yet another aspect, the present invention provides an isolated or recombinant polynucleotide, a fragment or a variant thereof, the polynucleotide comprising a nucleic acid sequence at least 80% identical to a nucleic acids sequence selected from the group consisting of SEQ ID NOs:40, 80, 22-39, 41, 43-61, 63-79, 81, and 83-301, wherein said polynucleotide, when expressed in a plant, is capable of enhancing the resistance of the plant to at least one pathogenic fungus and/or Oomycete. According to certain embodiments, the polynucleotide comprises the nucleic acid sequence set forth in any one if SEQ ID NOs:40, 80, 22-39, 41, 43-61, 63-79, 81, and 83-305.


According to yet further aspect, the present invention provides an isolated or recombinant polynucleotide, a fragment or a variant thereof, the polynucleotide comprising a nucleic acid sequence at least 70% identical to a nucleic acids sequence selected from the group consisting of SEQ ID NOs:783, 592-782, 784-2443, wherein said polynucleotide, when expressed in a plant, is capable of enhancing the resistance of the plant to at least one pathogenic fungus and/or Oomycete. According to certain embodiments, the polynucleotide comprises the nucleic acid sequence set forth in any one if SEQ ID NOs:783, 592-782, 784-2443.


According to additional aspect, the present invention provides a recombinant polynucleotide optimized for polypeptide expression in a bacterial expression system, wherein the polypeptide is capable of inhibiting the growth of at least one pathogenic fungus and/or Oomycete.


According to certain exemplary embodiment the bacterium is Escherichia coli. According to these embodiments, the polypeptide comprises an amino acid sequence selected from the group consisting of SEQ ID NOs: 4468, 4450-4467, and 4469-4646. According to certain embodiments, the recombinant polynucleotide comprises a nucleic acid sequence selected from the group consisting of SEQ ID NOs:4271, 4253-4270, and 4272-4449.


According to certain embodiments, the recombinant polynucleotide optimized for polypeptide expression in the bacterial expression system further comprises said bacterium signal peptide.


According to a further aspect, the present invention provides a fungicidal composition comprising an effective amount of at least one polypeptide at least 70% identical to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:2635, 2444-2634, 2636-4252, a fragment and/or a variant thereof, wherein the fungicidal composition is effective in inhibiting the growth and/or development of at least one plant pathogenic fungi and/or Oomycetes. According to certain embodiments, the fungicidal composition comprises at least one polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:2635, 4468, 4661, 2444-2634, 2636-4252, 4450-4467, 4469-4660, and 4662-4799. According to some embodiments, the fungicidal composition comprises at least one polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:4468, 4661, 4450-4467, 4469-4660, and 4662-4799. According to certain embodiments, the fungicidal composition further comprises at least one agriculturally compatible agent selected from the group consisting of a carrier, a stabilizer, a diluent, a surfactant, a mineral and an adjuvant.


According to yet additional aspect, the present invention provides a fungicidal composition comprising bacteria comprising at least one polypeptide at least 70% identical to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:2635, 2444-2634, 2636-4252, a fragment and/or a variant thereof, wherein the fungicidal composition is effective in inhibiting the growth and/or development of at least one plant pathogenic fungi and/or Oomycetes. According to certain embodiments, the bacteria comprise at least one polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:2635, 4468, 4661, 2444-2634, 2636-4252, 4450-4467, 4469-4660, and 4662-4799. According to some embodiments, the bacteria comprise at least one polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:4468, 4661, 4450-4467, 4469-4660, and 4662-4799. Each possibility represents a separate embodiment of the present invention. The bacteria can be of the same species or of different species, with the proviso that the bacteria have no adverse effect on a plant. The bacteria can be alive or dead. According to certain exemplary embodiments, the bacteria are selected from the group consisting of Bacillus subtilis Lactococcus lacti, Corynebacterium glutamicum; and Bacillus brevis.


According to certain embodiments, the fungicidal composition further comprises at least one agriculturally compatible agent selected from the group consisting of a carrier, a stabilizer, a diluent, a surfactant, a mineral and an adjuvant.


According to additional aspect, the present invention provides a nucleic acid construct comprising a polynucleotide according to some embodiments of the present invention, further comprising at least one regulatory element for directing the expression of the polynucleotide within a plant cell. According to certain embodiment, the regulatory element is a promoter. The promoter can be endogenous or heterologous to the plant comprising the nucleic acid construct.


The polypeptides and polynucleotides disclosed herein may be used to confer resistance to a wide variety of fungal and Oomycetous pathogens that cause commercial damage to crop and ornamental plants.


According to certain embodiments, the fungal or Oomycetous pathogens can be one or more fungi or Oomycetes from a class selected from the group consisting of Ascomycetes, Plasmodiophoromycetes, Chytridiomycetes, Zygomycetes, Basidiomycetes, Deuteromycetes, and Sordariomycetes. Each possibility represents a separate embodiment of the present invention.


According to some embodiments, the fungal pathogens can be one or more fungi from a genus selected from the group consisting of Fusarium, Colletotrichum, Geotrichum, Aspergillus, Alternaria, Athelia, Botryosphaeria, Botrytis, Cryphonectria, Choanephora, Cercospora, Magnaporthe Monilinia, Mycosphaerella, Melampsora, Puccinia, Phakopsora, Rhizoctonia, Septoria, Uromyces, Ustilago and Verticillium.


According to some embodiments, the Oomycetous pathogen can be from the class Oomycetes (synonym Peronosporomycetes). In some embodiments, said Oomycetous infection comprises infection by an Oomycete from a genus selected from the group consisting of Blumeria, Macrophomina, Oidium, Pythium, and Phytophthora. Each possibility represents a separate embodiment of the present invention. According to some embodiments, the fungal or Oomycetous pathogen is selected from the group consisting of Botrytis cinerea, Mycosphaerella graminicola, Mycosphaerella fijiensis, Septoria lycopersici, Magnaporthe oryza, Rhizoctonia solani, Ustilago maydis, Sclerotium rolfsii, and Blumeria graminis.


According to certain exemplary embodiments, the fungus of the genus Fusarium is selected from the group consisting of Fusarium verticilloides and Fusarium graminearum. According to additional exemplary embodiments, the F. verticilloides is F. verticillioides strain A-00149-FGSC 7600. According to further exemplary embodiments, the F. graminearum is F. graminearum strain CBS 110260.


According to other exemplary embodiments, the fungus of the genus Colletotrichum is Colletotrichum graminicola.


The polynucleotides and polypeptides of the present invention can be used to confer resistance to any plant type. According to certain embodiments, the plant is a cereal plant. According to some embodiments, the cereal plant is selected from the group consisting of maize, wheat, barley, sorghum, rice, oat, and rye. Each possibility represents a separate embodiment of the present invention. According to certain exemplary embodiments, the cereal plant is maize (Zea mays). According to other embodiments, the plant is a field-crop plant. According to some embodiments, the field crop plant is selected from the group consisting of tomato, potato, sweet potato, cassava, beets, ginger, horseradish, radish, ginseng, turnip, any root or tuber crop, pepper, eggplant, ground cherry, tomatillo, okra, other fruiting vegetables, cucumber, cantaloupe, melon, muskmelon, squash, watermelon and other cucurbit plants.


Further embodiments and the full scope of applicability of the present invention will become apparent from the detailed description given hereinafter. However, it should be understood that the detailed description and specific examples, while indicating preferred embodiments of the invention, are given by way of illustration only, since various changes and modifications within the spirit and scope of the invention will become apparent to those skilled in the art from this detailed description.





BRIEF DESCRIPTION OF THE FIGURES


FIG. 1 is a schematic illustration of the pQ6sVN plasmid. pQ6sVN used for expression of the isolated polynucleotide sequences of some embodiments of the invention in Brachypodium. “35S(V)”=35S promoter (SEQ ID NO:37); “NOS ter”=nopaline synthase terminator; “Bar_GA”=BAR open reading frame optimized for expression in Brachypodium (SEQ ID NO:39); “Hyg”=Hygromycin resistance gene. “Ubi1 promoter”=SEQ ID NO: 11; the isolated polynucleotide sequences of some embodiments of the invention were cloned into the Multiple cloning site of the vector (downstream of the “35S(V)” promoter) using one or more of the indicated restriction enzyme sites.



FIGS. 2A-H depict an exemplary design of Homology Directed Repair according to some embodiments of the invention. FIG. 2A depicts the sequence of the endogenous 5′-upstream flanking region of the genomic sequence GRMZM2G069095 (SEQ ID NO:45). FIG. 2B depicts the sequence of the endogenous 3′-downstream flanking region of the genomic sequence GRMZM2G069095 having the nucleic acid sequence set forth in SEQ ID NO:46. FIG. 2C depicts the sequence of the 5′-UTR gRNA (SEQ ID NO:43). FIG. 2D depicts the sequence of the 5′-UTR gRNA without NGG nucleotides (SEQ ID NO:47). FIG. 2E depicts the sequence of the 3′-UTR gRNA (SEQ ID NO:44). FIG. 2F depicts the sequence of the 3′-UTR gRNA after cut (SEQ ID NO:48). FIG. 2G depicts the coding sequence (from the “ATG” start codon to the “TGA” termination codon, marked by bold and underlined) of the desired LFS127_H4 sequence (SEQ ID NO:50) encoding the polypeptide set forth by SEQ ID NO:487. FIG. 2H depicts the exemplary repair template (SEQ ID NO:49) which includes (1) the upstream flanking region (1 kbp) sequence including part of the gRNA after cutting (SEQ ID NO:47; shown in bold and italics); (2) 5′ UTR of genomic DNA from Cas9 cutting site to ATG; (3) the coding sequence (CDS) of the desired LFS127_H4 sequence (SEQ ID NO:50) marked in lower case with the start (ATG) and the stop (TGA) codons marked in bold and underlined; (4) 3′ UTR of genomic DNA from the stop codon to Cas9 cutting site including the predicted part of the gRNA after cutting (SEQ ID NO:48, shown in bold and underlined and (5) the downstream flanking region (1kbp) sequence.



FIG. 3A-E depicts an exemplary design of polynucleotide knockout (KO) using CRISPR/CAS system. FIG. 3A depicts the sequence of the KO gRNA (SEQ ID NO:51); FIG. 3B depicts the sequence of the KO gRNA after cut (SEQ ID NO:52); FIG. 3C depicts the coding sequence (from the “ATG” start codon to the “TAG” termination codon, marked by bold and underlined) of the desired LFS132 sequence (SEQ ID NO:53); FIG. 3D (targeted region in bold) and FIG. 3E depict the anticipated change in the coding sequence of the exemplified KO gene (SEQ ID NO:54).





DETAILED DESCRIPTION OF THE INVENTION

The present invention discloses means and methods for conferring and/or enhancing the resistance of a plant to pathogenic fungi and/or Oomycetes. Particularly, the present invention provides isolated polypeptides conferring or enhancing plant resistance to pathogenic fungi and/or Oomycetes, isolated polynucleotides encoding same, nucleic acid constructs comprising the polynucleotides and plant cells transformed with same. The present invention further provides methods for producing and selecting plants having increased resistance to at least one pathogenic fungus and/or Oomycete and plant with enhanced resistance to the at least one pathogenic fungus and/or Oomycete, in which the expression of the polynucleotides and/or polypeptides of the invention is modulated. In additional aspects, the present invention provides fungicidal compositions comprising isolated fungicidal polypeptides of the invention and/or bacteria comprising same.


The present invention is based in part on bioinformatics tools that have been used to identify polynucleotides associated with resistance or reduced sensitivity of plants to at least one pathogenic fungus or pathogenic Oomycete, as well as to identify polynucleotides of bacterial origin that encode for proteins with potential antifungal activity. Cereal plants, including maize (Zea), wheat (Triticum), millet (Sitaria italica) and the closely related Aegilops and Brachypodium species were used as representative genera to identify genes overexpressed or downregulated in plants showing increased resistance to fungal/Oomycetes infection. Genes comprising the nucleic acids sequence set forth in any one of SEQ ID NOs:22-62 and 302-303, encoding polypeptides having the amino acid sequence set forth in any one of SEQ ID NOs:308-348, 507, and 510 were identified. Homologous genes and encoded proteins were also identified in wider genera of plant, as described in details and presented in Table 7 hereinbelow. Bacteria isolated from soils known to contain Fusaria species were used for screening and identification of potential anti-fungal bacterial proteins. Genes comprising the nucleic acids sequence set forth in any one of SEQ ID NOs:592-789, encoding polypeptides having the amino acid sequence set forth in any one of SEQ ID NOs:2444-2641 were identified. Homologous genes and encoded proteins were also identified in wider genera of bacteria, as described in details and presented in Table 16 hereinbelow.


Polynucleotides of plant origin according to some embodiments of the present invention were cloned into binary vectors, and transformed into plants of the species Brachypodium distachyon (Example 6 hereinbelow) to validate the effect of the genes on the resistance of the transformed plants towards the fungi/Oomycetes (Example 7).


Polynucleotides of bacterial origin according to some embodiments of the present invention were cloned, transformed into E. coli, and the expressed proteins were isolated (Examples 14-15). The anti-fungal effect of the isolated proteins was validated by their ability to inhibit the growth of Fusarium verticillioides mycelia (Example 16, Table 21).


Definitions

The terms “comprise”, “comprising”, “includes”, “including”, “having” and their conjugates mean “including but not limited to”.


The term “consisting of” means “including and limited to”.


The term “consisting essentially of” means that the composition, method or structure may include additional ingredients, steps and/or parts, but only if the additional ingredients, steps and/or parts do not materially alter the basic and novel characteristics of the claimed composition, method or structure.


As used herein, the singular form “a”, “an” and “the” include plural references unless the context clearly dictates otherwise. For example, the term “a compound” or “at least one compound” may include a plurality of compounds, including mixtures thereof.


Throughout this application, various embodiments of this invention may be presented in a range format. It should be understood that the description in range format is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of the invention. Accordingly, the description of a range should be considered to have specifically disclosed all the possible sub ranges as well as individual numerical values within that range. For example, description of a range such as from 1 to 6 should be considered to have specifically disclosed sub ranges such as from 1 to 3, from 1 to 4, from 1 to 5, from 2 to 4, from 2 to 6, from 3 to 6 etc., as well as individual numbers within that range, for example, 1, 2, 3, 4, 5, and 6. This applies regardless of the breadth of the range.


Whenever a numerical range is indicated herein, it is meant to include any cited numeral (fractional or integral) within the indicated range. The phrases “ranging/ranges between” a first indicate number and a second indicate number and “ranging/ranges from” a first indicate number “to” a second indicate number are used herein interchangeably and are meant to include the first and second indicated numbers and all the fractional and integral numerals therebetween.


As used herein the term “method” refers to manners, means, techniques and procedures for accomplishing a given task including, but not limited to, those manners, means, techniques and procedures either known to, or readily developed from known manners, means, techniques and procedures by practitioners of the agricultural, chemical, pharmacological, biological, biochemical and medical arts.


When reference is made to particular sequence listings, such reference is to be understood to also encompass sequences that substantially correspond to its complementary sequence as including minor sequence variations, resulting from, e.g., sequencing errors, cloning errors, or other alterations resulting in base substitution, base deletion or base addition, provided that the frequency of such variations is less than 1 in 50 nucleotides, alternatively, less than 1 in 100 nucleotides, alternatively, less than 1 in 200 nucleotides, alternatively, less than 1 in 500 nucleotides, alternatively, less than 1 in 1000 nucleotides, alternatively, less than 1 in 5,000 nucleotides, alternatively, less than 1 in 10,000 nucleotides.


As used herein, the phrase “exogenous polynucleotide” refers to a heterologous nucleic acid sequence which is not naturally expressed within the plant (e.g., a nucleic acid sequence from a different species) or to an endogenous nucleic acid of which overexpression in the plant is desired. The exogenous polynucleotide may be introduced into the plant in a stable or transient manner, so as to produce a ribonucleic acid (RNA) molecule and/or a polypeptide molecule. The term “endogenous” as used herein refers to a polynucleotide or polypeptide which is naturally present and/or naturally expressed within a plant or a cell thereof.


The terms “modulating”, “modifying” and “altering” with reference to the expression or activity of a polynucleotide, gene, polypeptide or a protein within a cell or a plurality of cells, particularly plant cell(s), are used herein interchangeably and refer to changing their level of expression within the cell, particularly plant cell. The change can be an increase or a decrease; and it can be measured as compared to any one of the polynucleotide, gene, polypeptide and/or protein level within the same cell(s) before modulation and as compared to the level in a control plant or an average level from a plurality of control plants in which the expression was not modified by man.


According to certain embodiments, the control plant is a wild type plant not manipulated to have modulated expression and/or activity of the polynucleotide and/or polypeptide. According to some embodiments, the control plant is of the same species. According to some embodiments, the control plant comprises the same genetic background.


According to certain embodiments, the examined plant and the control plant are grown under the same growing conditions.


As used herein, the term “resistance” with regard to plants pathogenic fungus and/or Oomycete refers to a plant that is resistant to infection by a fungal or Oomycetous pathogen or resistant to the symptoms of fungal or Oomycetous pathogen infection. For example, a plant resistant to a fungal or Oomycetous pathogen can exhibit a lack of infection, or reduced symptoms of infection, when challenged with a pathogen. As another example, a plant resistant to a fungal or Oomycetous pathogen can be infected by the fungal or Oomycetous pathogen and yet exhibit a reduced number or degree of symptoms of said infection. As yet another example, a plant resistant to a fungal or Oomycetous pathogen can be infected by the pathogen and exhibit one or more symptoms of infection by the pathogen and yet exhibit a reduction in an effect of the infection or symptom thereof. For instance, a plant resistant to a fungal or Oomycetous pathogen can be infected by the pathogen, and exhibit one or more symptoms selected from the group consisting of leaf wilt, leaf or vascular discoloration (e.g., yellowing), spike bleaching etc., and yet not exhibit a reduction in yield loss in comparison to a plant that is not resistant (susceptible) to the fungal or Oomycetous pathogen.


Accordingly, “confer resistance to a pathogenic fungus and/or Oomycete” or “enhanced resistance to a pathogenic fungus and/or Oomycete” refer to a phenotype in which a plant has greater health, growth, multiplication, fertility, vigor, strength (e.g., stem strength and resistance), yield, or less severe symptoms associated with infection of the pathogenic fungus or Oomycete during or after a fungal or Oomycete infection than an organism that does not have enhanced resistance to the pathogen. Where a plant is tested for resistance, a control plant is used to assess the degree of the plant resistance. According to certain embodiments of the present invention, the control plant is a plant not manipulated to have modified expression of at least one polynucleotide and/or polypeptide of the present invention. The control plant is typically, but not necessarily of the same species as the examined plant. According to some embodiments the control plant is of the same specifies and has the same genetic background as the examined plant. The enhancement can be manifested as an increase of 0.1%, 0.2%, 0.3%, 0.5%, 0.75%, 1%, 1.5%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 12%, 15%, 17%, 20%, 25%, 30%, 35%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or more in health, growth, multiplication, fertility, vigor, strength, or yield, as compared to a control plant. The enhancement can be a decrease of 0.1%, 0.2%, 0.3%, 0.5%, 0.75%, 1%, 1.5%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 12%, 15%, 17%, 20%, 25%, 30%, 35%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% in the symptoms associated with the pathogenic fungus and/or Oomycete as compared to the control plant. According to certain exemplary embodiments, the examined plant and the control plant are grown under the same conditions.


According to certain embodiments of the invention, enhancing the resistance of a plant to a pathogenic fungus comprises enhancing the expression and/or activity of a polypeptide of the invention within at least one cell of the plant. As used herein, the expression of a polynucleotide or polypeptide of the invention is “enhanced” or “up-regulated” if the level of the polynucleotide or polypeptide is enhanced by at least 50%, i.e. the polynucleotide or polypeptide level is at least 1.5-fold higher compared to its level in a control plant or compared to a predetermined threshold level. According to some embodiments, the level of the polynucleotide or polypeptide expression is enhanced by at least 60%, 70%, 80%, 90%, 100%, 200%, 300% and more.


According to certain embodiments, the pre-determined resistance score value is obtained by inoculating a population of corresponding plants susceptible to the at least one pathogenic fungus, scoring the infection degree and setting an average resistance score value.


According to certain embodiments of the invention, enhancing the resistance of a plant to a pathogenic fungus comprises reducing the expression and/or activity of a polynucleotide and/or polypeptide of the invention within at least one cell of the plant. As used herein, the expression of a polynucleotide or polypeptide of the invention is “reduced”, “inhibited”, “down regulated” or “knocked out” or “knocked down” if the level of the polynucleotide or polypeptide is reduced by at least 30% compared to its level in a control plant or compared to a predetermined threshold level. According to certain embodiments, the level of the polynucleotide or polypeptide is reduced by at least 40%, 50%, 60%, 70%, 80%, 90% and more. According to some embodiments, the term “reduced expression” refers to 100% inhibition or “knockout” of a polynucleotide function and/or expression.


As used herein the term “polynucleotide” refers to a single or double stranded nucleic acid sequence which is isolated and provided in the form of an RNA sequence, a complementary polynucleotide sequence (cDNA), a genomic polynucleotide sequence and/or a composite polynucleotide sequences (e.g., a combination of the above).


As used herein the phrase “complementary polynucleotide sequence” refers to a sequence, which results from reverse transcription of messenger RNA using a reverse transcriptase or any other RNA dependent DNA polymerase. Such a sequence can be subsequently amplified in vivo or in vitro using a DNA dependent DNA polymerase.


The term “isolated” refers to at least partially separated from the natural environment e.g., from a plant cell.


As used herein the phrase “genomic polynucleotide sequence” refers to a sequence derived (isolated) from a chromosome and thus it represents a contiguous portion of a chromosome.


As used herein the phrase “composite polynucleotide sequence” refers to a sequence, which is at least partially complementary and at least partially genomic. A composite sequence can include some exonal sequences required to encode the polypeptide of the present invention, as well as some intronic sequences interposing therebetween. The intronic sequences can be of any source, including of other genes, and typically will include conserved splicing signal sequences. Such intronic sequences may further include cis acting expression regulatory elements.


It should be noted that the nucleic acid sequence of a polynucleotide encoding a polypeptide which is provided in the sequence listing as a single strand refers to the sense direction which is equivalent to the mRNA transcribed from the polynucleotide.


When reference is made to particular sequence listings, such reference is to be understood to also encompass sequences that substantially correspond to its complementary sequence as including minor sequence variations, resulting from, e.g., sequencing errors, cloning errors, or other alterations resulting in base substitution, base deletion or base addition, provided that the frequency of such variations is less than 1 in 50 nucleotides, alternatively, less than 1 in 100 nucleotides, alternatively, less than 1 in 200 nucleotides, alternatively, less than 1 in 500 nucleotides, alternatively, less than 1 in 1000 nucleotides, alternatively, less than 1 in 5,000 nucleotides, alternatively, less than 1 in 10,000 nucleotides.


As used herein, the phrase “exogenous polynucleotide” refers to a heterologous nucleic acid sequence which is not naturally expressed within the plant (e.g., a nucleic acid sequence from a different species) or to an endogenous nucleic acid of which overexpression in the plant is desired. The exogenous polynucleotide may be introduced into the plant in a stable or transient manner, so as to produce a ribonucleic acid (RNA) molecule and/or a polypeptide molecule.


The term “endogenous” as used herein refers to a polynucleotide or polypeptide which is naturally present and/or naturally expressed within a plant or a cell thereof.


The term “heterologous” as used herein refers to polynucleotide or polypeptide which is not naturally present and/or naturally expressed within a plant or a cell thereof.


According to one aspect, the present invention provides a method for enhancing the resistance of a plant or a part thereof to at least one pathogenic fungus and/or Oomycete, comprising modulating the expression and/or activity of at least one polypeptide at least about 70%, at least about 71%, at least about 72%, at least about 73%, at least about 74%, at least about 75%, at least about 76%, at least about 77%, at least about 78%, at least about 79% or more homologous to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:2635, 2444-2634, 2636-4252; or at least about 80%, at least about 81%, at least about 82%, at least about 83%, at least about 84%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99% or more homologous to, or identical to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:326, 360, 308-325, 327-359, 361-506 and 2635, 2444-2634, 2636-4252, a fragment and/or a variant thereof within at least one cell of the plant or part thereof, thereby enhancing the resistance of said plant or part thereof to the at least one pathogenic fungus and/or Oomycetes compared to the resistance of a control plant. Each possibility represents a separate embodiment of the present invention.


According to another aspect, the present invention provides a method for enhancing the resistance of a plant or a part thereof to at least one pathogenic fungus and/or Oomycete, comprising modulating the expression of at least one polynucleotide encoding a polypeptide at least about 70%, at least about 71%, at least about 72%, at least about 73%, at least about 74%, at least about 75%, at least about 76%, at least about 77%, at least about 78%, at least about 79% or more homologous to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:2380-4175; or at least about 80%, at least about 81%, at least about 82%, at least about 83%, at least about 84%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99% or more homologous to, or identical to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:339-537 and 2380-4175, a fragment and/or a variant thereof within at least one cell of the plant or part thereof, thereby enhancing the resistance of said plant or part thereof to the at least one pathogenic fungus and/or Oomycete compared to the resistance of a corresponding control plant. Each possibility represents a separate embodiment of the present invention.


According to certain embodiments, the polypeptide is 70%-79% homologous to any one of the polypeptides set forth in SEQ ID NOs:2635, 2444-2634, 2636-4252. According to other embodiments, the polypeptide is 75%-79% homologous to any one of the polypeptides set forth in SEQ ID NOs:2635, 2444-2634, 2636-4252.


According to certain embodiments, the polypeptide is 80%-99% homologous to any one of the polypeptides set forth in SEQ ID NOs:326, 360, 308-325, 327-359, 361-506, 2635, 2444-2634, and 2636-4252. According to other embodiments, the polypeptide is 85%-95% homologous to any one of the polypeptides set forth in SEQ ID NOs: 326, 360, 308-325, 327-359, 361-506, 2635, 2444-2634, and 2636-4252. According to other embodiments, the polypeptide is 90%-99% homologous to any one of the polypeptides set forth in SEQ ID NOs:326, 360, 308-325, 327-359, 361-506, 2635, 2444-2634, and 2636-4252. According to certain embodiments, the polypeptide comprises an amino acid sequence selected from the group consisting of SEQ ID NOs: 326, 360, 308-325, 327-359, 361-508, 2635, 2444-2634, and 2636-4252. Each possibility represents a separate embodiment of the present invention. According to certain embodiments, the polypeptide consists of the amino acid sequence selected from the group consisting of SEQ ID NOs:326, 360, 308-325, 327-359, 361-508 and 2635, 2444-2634, 2636-4252. Each possibility represents a separate embodiment of the present invention.


According to yet additional aspect, the present invention provides a method for enhancing the resistance of a plant or a part thereof to at least one pathogenic fungus and/or Oomycete, comprising modulating the expression of at least one polynucleotide at least about 70%, at least about 71%, at least about 72%, at least about 73%, at least about 74%, at least about 75%, at least about 76%, at least about 77%, at least about 78%, at least about 79% or more homologous to a polynucleotide having a nucleic acid sequence selected from the group consisting of SEQ ID NOs:783, 592-782, 784-2443; or at least about 80%, at least about 81%, at least about 82%, at least about 83%, at least about 84%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99% or more homologous to, or identical to a polynucleotide having an nucleic acid sequence selected from the group consisting of SEQ ID NOs:40, 80, 22-39, 41-79, 81-301, 783, 592-782, 784-2443 within at least one cell of the plant or part thereof, thereby enhancing the resistance of said plant or part thereof to the at least one pathogenic fungus and/or Oomycetes compared to the resistance of a control plant. Each possibility represents a separate embodiment of the present invention.


According to certain embodiments, the polynucleotide is 70%-79% homologous to any one of the polynucleotides set forth in SEQ ID NOs:783, 592-782, 784-2443. According to other embodiments, the polynucleotide is 75%-79% homologous to any one of the polynucleotides set forth in SEQ ID NOs:783, 592-782, 784-2443. Each possibility represents a separate embodiment of the present invention.


According to certain embodiments, the polynucleotide is 80%-99% homologous to any one of the polynucleotides set forth in SEQ ID NOs:40, 80, 22-39, 41-79, 81-301, 783, 592-782, and 784-2443. According to other embodiments, the polynucleotide is 85%-95% homologous to any one of the polynucleotides set forth in SEQ ID NOs:40, 80, 22-39, 41-79, 81-301, 783, 592-782, and 784-2443. According to other embodiments, the polynucleotide is 90%-99% homologous to any one of the polynucleotides set forth in SEQ ID NOs:40, 80, 22-39, 41-79, 81-301, 783, 592-782, and 784-2443. Each possibility represents a separate embodiment of the present invention.


According to certain embodiments, the polynucleotide comprises a nucleic acids sequence set forth in any one of SEQ ID NOs:40, 80, 22-39, 41-79, 81-305, 783, 592-782, 784-2443. According to other embodiments, the exogenous polynucleotide consists of a nucleic acids sequence set forth in any one of SEQ ID NOs:40, 80, 22-39, 41-79, 81-305, 783, 592-782, and 784-2443. Each possibility represents a separate embodiment of the present invention.


According to another aspect, the present invention provides a method for producing a population of plants each having an enhanced resistance to at least one pathogenic fungus or Oomycete, comprising the steps of:

    • (a) modulating the expression and/or activity of at least one polypeptide at least about 70%, at least about 71%, at least about 72%, at least about 73%, at least about 74%, at least about 75%, at least about 76%, at least about 77%, at least about 78%, at least about 79% or more homologous to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:2635, 2444-2634, 2636-4252; or at least about 80%, at least about 81%, at least about 82%, at least about 83%, at least about 84%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99% or more homologous to, or identical to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:326, 360, 308-325, 327-359, 361-506, 2635, 2444-2634, and 2636-4252, a fragment and/or a variant thereof within at least one cell of each plant of the plant population as to produce a genetically engineered plant population;
    • (b) inoculating each plant of the genetically engineered plant population with the at least one pathogenic fungus; and
    • (c) selecting plants showing an enhanced resistance to said at least one pathogenic fungus compared to a control or to a pre-determined resistance score value;


thereby producing a population of genetically engineered plants having enhanced resistance to said at least one pathogenic fungus.


According to certain embodiments, the method comprises modulating the expression and/or activity of a polypeptide having the amino acid sequence set forth in any one of SEQ ID NOs:326, 360, 308-325, 327-359, 361-508, 2635, 2444-2634, and 2636-4252. According to other embodiments, the method comprises enhancing the expression and/or activity of a polypeptide having the amino acid sequence set forth in SEQ ID NO:509. According to yet additional embodiments, the method comprises enhancing the expression and/or activity of a polypeptide consisting of the amino acid sequence set forth in any one of SEQ ID NOs:326, 360, 308-325, 327-359, 361-509, 2635, 2444-2634, and 2636-4252.


According to additional aspect, the present invention provides a method for selecting a plant having an enhanced resistance to at least one pathogenic fungus and/or Oomycete, comprising the steps of:

    • (a) providing a plurality of plants each comprising at least one cell with modulated expression and/or activity of a polypeptide at least about 70%, at least about 71%, at least about 72%, at least about 73%, at least about 74%, at least about 75%, at least about 76%, at least about 77%, at least about 78%, at least about 79% or more homologous to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:2635, 2444-2634, 2636-4252; or at least about 80%, at least about 81%, at least about 82%, at least about 83%, at least about 84%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99% or more homologous to, or identical to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:326, 360, 308-325, 327-359, 361-506, 2635, 2444-2634, and 2636-4252, a fragment and/or a variant thereof;
    • (b) inoculating the plurality of plants with the at least one pathogenic fungus or Oomycete; and
    • (c) selecting plants showing an enhanced resistance to said at least one pathogenic fungus or Oomycete compared to a control plant or to a pre-determined resistance score value.


According to certain embodiments, the method comprises providing a plurality of plants each comprising at least one cell with modulated expression and/or activity of at least one polypeptide having the amino acid sequence set forth in any one of SEQ ID NOs:326, 360, 308-325, 327-359, 361-508, 2635, 2444-2634, and 2636-4252. Each possibility represents a separate embodiment of the present invention. According to other embodiments, the method comprises providing a plurality of plants each having modulated expression and/or activity of a polypeptide having the amino acid sequence set forth SEQ ID NO:509. According to other embodiments, the method comprises providing a plurality of plants each having modulated expression and/or activity of at least one polypeptide consisting of the amino acid sequence set forth in any one of SEQ ID NOs: 326, 360, 308-325, 327-359, 361-509, 2635, 2444-2634, and 2636-4252.


The plurality of plants having modulated expression and/or activity of the polypeptide may include plants having at least one cell with enhanced expression and/or activity of a polypeptide, plants having at least one cell with reduced expression and/or activity of a polypeptide or a combination thereof. Enhancing or reducing the expression and/or activity of the polypeptide can be performed as is known in the Art and as described hereinbelow.


According to certain embodiments, the polypeptide the expression and/or activity of which is to be enhanced comprises an amino acid sequence at least 70% identical to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:2635, 2444-2634, 2636-4252 or at least 80% identical to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:326, 360, 308-325, 327, 329-347, 349-359, 362-506. According to some embodiments, the polypeptide the expression and/or activity of which is to be enhanced comprises an amino acid sequence selected from the group consisting of SEQ ID NOs:326, 360, 308-325, 327, 329-347, 349-359, 362-508, 2635, 2444-2634, and 2636-4252. Each possibility represents a separate embodiment of the present invention.


According to certain embodiments, enhancing the resistance of the plant to the pathogenic fungus and/or Oomycete comprises reducing the expression and/or activity of at least one polypeptide compared to its expression and/or activity in the control plant. According to certain exemplary embodiments, the polypeptide the expression and/or activity of which is to be reduced comprises an amino acid sequence at least 80% identical to the amino acid sequence set forth in any one of SEQ ID NO:328, SEQ ID NO:361, and SEQ ID NO:348. According to further exemplary embodiments, the polypeptide the expression and/or activity of which is to be reduced comprises the amino acid sequence set forth in any one of SEQ ID NO:328, SEQ ID NO:361 and SEQ ID NO:348.


According to another aspect, the present invention provides a method for conferring and/or enhancing the resistance of a grafted plant to at least one pathogenic fungus and/or Oomycete, the method comprising providing a scion and a rootstock, wherein the rootstock exhibits enhanced resistance to the at least one pathogenic fungus and/or Oomycete, said rootstock comprises at least one cell with a modulated expression of a polynucleotide encoding a polypeptide at least about 70%, at least about 71%, at least about 72%, at least about 73%, at least about 74%, at least about 75%, at least about 76%, at least about 77%, at least about 78%, at least about 79% or more homologous to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:2635, 2444-2634, 2636-4252; or at least about 80%, at least about 81%, at least about 82%, at least about 83%, at least about 84%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99% or more homologous, or identical to the amino acid sequence selected from the group consisting of SEQ ID NOs:326, 360, 308-325, 327-359, 361-506, 2635, 2444-2634, 2636-4252, a fragment and/or a variant thereof compared to the scion and grafting said scion onto said rootstock, thereby producing a grafted plant having an enhanced resistance to said at least one pathogenic fungus and/or Oomycete.


According to another aspect, the present invention provides a method for conferring and/or enhancing the resistance of a grafted plant to at least one pathogenic fungus and/or Oomycete, the method comprising providing a scion and a rootstock, the scion having an enhances resistance to the at least one pathogenic fungus and/or Oomycete, said scion comprises at least one cell with modulated expression of at least one polynucleotide encoding a polypeptide at least about 70%, at least about 71%, at least about 72%, at least about 73%, at least about 74%, at least about 75%, at least about 76%, at least about 77%, at least about 78%, at least about 79% or more homologous to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:2635, 2444-2634, 2636-4252; or at least about 80%, at least about 81%, at least about 82%, at least about 83%, at least about 84%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99% or more homologous, or identical to the amino acid sequence selected from the group consisting of SEQ ID NOs:326, 360, 308-325, 327-359, 361-506, 2635, 2444-2634, 2636-4252, a fragment and/or a variant thereof compared to the rootstock, and grafting said scion onto said rootstock, thereby producing a grafted plant having an enhanced resistance to the at least one pathogenic fungus and/or Oomycete.


According to certain embodiments, the polypeptide expressed in the scion or in the rootstock comprises an amino acid sequence selected from the group consisting of SEQ ID NOs:326, 360, 308-325, 327-359, 361-508, 2635, 2444-2634, and 2636-4252.


According to certain embodiments, the scion or rootstock having enhanced resistance to the at least one fungus and/or Oomycete comprises at least one cell with enhanced expression and/or activity of at least one polypeptide or the nucleotide encoding same. According to other embodiments, the scion or rootstock having enhanced resistance to the at least one fungus and/or Oomycete comprises at least one cell with reduced expression and/or activity of at least one polypeptide or the nucleotide encoding same. The polypeptides the expression of which is enhanced or reduced are as described hereinabove.


According to some embodiments, the at least one polynucleotide is constitutively expressed in the transgenic rootstock. According to some embodiments, the at least one polynucleotide is expressed in the transgenic rootstock in a tissue specific or inducible manner. According to some embodiments, the expression of the at least one polynucleotide is induced by biotic stress, particularly by fungi infection.


According to additional aspect, the present invention provides a method of growing a crop plant having enhanced resistance to at least one pathogenic fungus and/or Oomycete comprising the steps of:

    • (a) selecting a parent plant having a modulated expression of at least one polynucleotide comprising a nucleic acid sequence encoding a polypeptide at least about 70%, at least about 71%, at least about 72%, at least about 73%, at least about 74%, at least about 75%, at least about 76%, at least about 77%, at least about 78%, at least about 79% or more homologous to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:2635, 2444-2634, 2636-4252; or at least about 80%, at least about 81%, at least about 82%, at least about 83%, at least about 84%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99% or more homologous, or identical to the amino acid sequence selected from the group consisting of SEQ ID NOs:326, 360, 308-325, 327-359, 361-506, 2635, 2444-2634, 2636-4252, a fragment and/or a variant thereof for enhanced resistance to at least one pathogenic fungus and/or Oomycete; and
    • (b) growing a progeny crop plant of the parent plant, wherein the progeny crop plant having modulated expression of the polynucleotide has an enhanced resistance to the at least one pathogenic fungus and/or Oomycete.


According to certain embodiments, the encoded polypeptide comprises the amino acid sequence set forth in any one of SEQ ID NOs:326, 360, 308-325, 327-359, 361-508, 2635, 2444-2634, and 2636-4252. According to certain embodiments, the encoded polypeptide comprises the amino acid sequence set forth in SEQ ID NOs:5509. According to some embodiments, the encoded polypeptide consists of the amino acid sequence set forth in any one of SEQ ID NOs: 326, 360, 308-325, 327-359, 361-509, 2635, 2444-2634, 2636-4252. Each possibility represents a separate embodiment of the present invention.


According to certain embodiments, the polynucleotide comprises a nucleic acid sequence at least about 70%, at least about 71%, at least about 72%, at least about 73%, at least about 74%, at least about 75%, at least about 76%, at least about 77%, at least about 78%, at least about 79% or more homologous to a polynucleotide having a nucleic acid sequence selected from the group consisting of SEQ ID NOs:783, 592-782, 784-2443; or at least about 80%, at least about 81%, at least about 82%, at least about 83%, at least about 84%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99% or more homologous to, or identical to a nucleic acid sequence set forth in any one of SEQ ID NOs:40, 80, 22-39, 41-79, 81-301, 783, 592-782, and 784-2443. Each possibility represents a separate embodiment of the present invention.


According to certain embodiments, the polynucleotide comprises the nucleic acid sequence set forth in any one of SEQ ID NOs:40, 80, 22-39, 41-79, 81-305, 783, 592-782, and 784-2443. According to certain embodiments, the polynucleotide comprises the nucleic acid sequence set forth in SEQ ID NO:306. According to some embodiments, the polynucleotide consists of the nucleic acid sequence set forth in any one of SEQ ID NOs: 40, 80, 22-39, 41-79, 81-306, 783, 592-782, 784-2443. Each possibility represents a separate embodiment of the present invention.


According to certain embodiments, the modulated expression comprises up-regulation of at least one polynucleotide expression. According to certain embodiments, the modulated expression comprises down-regulation of at least one polynucleotide expression.


According to certain exemplary embodiments, the gene to be upregulated comprises a nucleic acid sequence at least about 80%, at least about 81%, at least about 82%, at least about 83%, at least about 84%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99% or more homologous to, or identical to a nucleic acid sequence set forth in any one of SEQ ID NOs:40, 80, 22-39, 41, 43-61, 63-79, 81, 83-301. According to some embodiments, the gene to be upregulated comprises a polynucleotide selected from the group consisting of SEQ ID NOs:40, 80, 22-39, 41, 43-61, 63-79, 81, 83-306.


According to certain exemplary embodiments, the gene the expression of which is to be downregulated comprises a nucleic acid sequence at least about 80%, at least about 81%, at least about 82%, at least about 83%, at least about 84%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99% or more homologous to, or identical to a nucleic acid sequence set forth in any one of SEQ ID NOs:42, 62 and 82. According to certain embodiments, the gene the expression of which is to be downregulated comprises a nucleic acid sequence selected from the group consisting of SEQ ID NOs:42, 62 and 82.


According to certain embodiments, the encoded polypeptide the expression and/or activity of which is to be downregulated is at least about 80%, at least about 81%, at least about 82%, at least about 83%, at least about 84%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99% or more homologous to, or identical to an amino acid sequence set forth in any one of SEQ ID NOs:328, 248 and 361. According to certain embodiments, the polypeptide the expression and/or activity of which is to be downregulated comprises an amino acid sequence selected from the group consisting of SEQ ID NOs:328, 248 and 361. Each possibility represents a separate embodiment of the present invention.


According to yet additional aspect, the present invention provides a method of producing seeds of a crop comprising the steps of:

    • (a) selecting a parent plant having a modulated expression of at least one polynucleotide comprising a nucleic acid sequence encoding a polypeptide at least about 70%, at least about 71%, at least about 72%, at least about 73%, at least about 74%, at least about 75%, at least about 76%, at least about 77%, at least about 78%, at least about 79% or more homologous to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:2635, 2444-2634, 2636-4252; or at least about 80%, at least about 81%, at least about 82%, at least about 83%, at least about 84%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99% or more homologous, or identical to the amino acid sequence selected from the group consisting of SEQ ID NOs:326, 360, 308-325, 327-359, 361-506, 2635, 2444-2634, 2636-4252, a fragment and/or a variant thereof for enhanced resistance to at least one pathogenic fungus and/or Oomycete;
    • (b) growing the selected parent plant of step (a) to produce seeds;
    • (c) harvesting the produced seeds.


According to certain embodiments, the encoded polypeptide comprises the amino acid sequence set forth in any one of SEQ ID NOs:326, 360, 308-325, 327-359, 361-508 and 2635, 2444-2634, 2636-4252. According to certain embodiments, the encoded polypeptide comprises the amino acid sequence set forth in SEQ ID NO:509. According to some embodiments, the encoded polypeptide consists of the amino acid sequence set forth in any one of SEQ ID NOs:326, 360, 308-325, 327-359, 361-508 and 2635, 2444-2634, 2636-4252. Each possibility represents a separate embodiment of the present invention.


According to certain embodiments, the modulated expression of the least one polynucleotide comprises up-regulation of a polynucleotide expression as described hereinabove. According to certain embodiments, the modulated expression of the least one polynucleotide comprises down-regulation of a polynucleotide expression as described hereinabove.


According to certain embodiments, the seeds produced by the methods of the invention comprise at least one cell having modulated expression of the polynucleotide. According to some embodiments, plants grown from the produced seeds have enhanced resistance to at least one pathogenic fungus and/or Oomycete.


The present invention encompasses polynucleotides identified to be associated with resistance to at least one pathogenic fungus and/or Oomycete polypeptides encoded by same and homologs thereto.


According to certain embodiments, the exogenous isolated or recombinant polynucleotides employed in the methods of the present invention encode a polypeptide comprising an amino acid sequence at least about 80%, at least about 81%, at least about 82%, at least about 83%, at least about 84%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99% or more homologous to, or identical to an amino acid sequence set forth in any one of SEQ ID NOs:326, 360, 308-325, 327-359, 361-509 and 2635, 2444-2634, 2636-4252.


According to certain embodiments, the exogenous isolated or recombinant polynucleotides employed in the methods of the present invention comprise a nucleic acid sequence at least about 80%, at least about 81%, at least about 82%, at least about 83%, at least about 84%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99% or more homologous to, or identical to a nucleic acid sequence set forth in any one of SEQ ID NOs:40, 80, 22-39, 41-79, 81-306 and 783, 592-782, 784-2443.


Homologous sequences include both orthologous and paralogous sequences. The term “paralogous” relates to gene-duplications within the genome of a species leading to paralogous genes. The term “orthologous” relates to homologous genes in different organisms due to ancestral relationship. Thus, orthologs are evolutionary counterparts derived from a single ancestral gene in the last common ancestor of given two species (Koonin E V and Galperin M Y 2003. Sequence—Evolution-Function: Computational Approaches in Comparative Genomics. Boston: Kluwer Academic; Chapter 2, Evolutionary Concept in Genetics and Genomics) and therefore have great likelihood of having the same function.


One option to identify orthologues in monocot or in dicot plant species is by performing a reciprocal blast search. This may be done by a first blast involving blasting the sequence-of-interest against any sequence database, such as the publicly available NCBI database which may be found at: ncbi.nlm.nih.gov. If orthologues in rice were sought, the sequence-of-interest would be blasted against, for example, the 28,469 full-length cDNA clones from Oryza sativa Nipponbare available at NCBI. The blast results may be filtered. The full-length sequences of either the filtered results or the non-filtered results are then blasted back (second blast) against the sequences of the organism from which the sequence-of-interest is derived. The results of the first and second blasts are then compared. An ortholog is identified when the sequence resulting in the highest score (best hit) in the first blast identifies in the second blast the query sequence (the original sequence-of-interest) as the best hit. Using the same rational a paralog (homolog to a gene in the same organism) is found. In case of large sequence families, the ClustalW program may be used (ebi.ac.uk/Tools/clustalw2/index.html), followed by a neighbor-joining tree (Wikipedia.org/wiki/Neighbor-joining) which helps visualizing the clustering.


Homology (e.g., percent homology, sequence identity+sequence similarity) can be determined using any homology comparison software computing a pairwise sequence alignment.


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. When percentage of sequence identity is used in reference to proteins it is recognized that residue positions which are not identical often differ by conservative amino acid substitutions, where amino acid residues are substituted for other amino acid residues with similar chemical properties (e.g. charge or hydrophobicity) and therefore do not change the functional properties of the molecule. Where sequences differ in conservative substitutions, the percent sequence identity may be adjusted upwards to correct for the conservative nature of the substitution. Sequences which differ by such conservative substitutions are considered 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 Henikoff S and Henikoff J G. (Amino acid substitution matrices from protein blocks. Proc. Natl. Acad. Sci. U.S.A. 1992, 89(22): 10915-9).


Identity (e.g., percent homology) can be determined using any homology comparison software, including for example, the BlastN software of the National Center of Biotechnology Information (NCBI) such as by using default parameters.


According to some embodiments of the invention, the identity is a global identity, i.e., an identity over the entire amino acid or nucleic acid sequences of the invention and not over portions thereof.


According to some embodiments of the invention, the term “homology” or “homologous” refers to identity of two or more nucleic acid sequences; or identity of two or more amino acid sequences; or the identity of an amino acid sequence to one or more nucleic acid sequence.


According to some embodiments of the invention, the homology is a global homology, i.e., a homology over the entire amino acid or nucleic acid sequences of the invention and not over portions thereof.


According to some embodiments of the invention, the homology is a partial homology, i.e. a homology over part of the amino acid or nucleic acid sequences of the invention. According to certain exemplary embodiments, the partial sequence is of polynucleotides and polypeptides of embodiments of the present invention lacking a signal or transit peptide.


The degree of homology or identity between two or more sequences can be determined using various known sequence comparison tools. Following is a non-limiting description of such tools which can be used along with some embodiments of the invention.


Pairwise global alignment was defined by S. B. Needleman and C. D. Wunsch, “A general method applicable to the search of similarities in the amino acid sequence of two proteins” Journal of Molecular Biology, 1970, pages 443-53, volume 48).


For example, when starting from a polypeptide sequence and comparing to other polypeptide sequences, the EMBOSS-6.0.1 Needleman-Wunsch algorithm (available from emboss(dot)sourceforge(dot)net/apps/cvs/emboss/apps/needle(dot)html) can be used to find the optimum alignment (including gaps) of two sequences along their entire length—a “Global alignment”. Default parameters for Needleman-Wunsch algorithm (EMBOSS-6.0.1) include: gapopen=10; gapextend=0.5; datafile=EBLOSUM62; brief=YES.


According to some embodiments of the invention, the parameters used with the EMBOSS-6.0.1 tool (for protein-protein comparison) include: gapopen=8; gapextend=2; datafile=EBLOSUM62; brief=YES.


According to some embodiments of the invention, the threshold used to determine homology using the EMBOSS-6.0.1 Needleman-Wunsch algorithm is 80%, 81%, 82%, 83%, 84%, 8%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 9%, 96%, 97%, 98%, 99%, or 100%.


When starting from a polypeptide sequence and comparing to polynucleotide sequences, the OneModel FramePlus algorithm [Halperin, E., Faigler, S. and Gill-More, R. (1999)—FramePlus: aligning DNA to protein sequences. Bioinformatics, 15, 867-873) (available from biocceleration(dot)com/Products(dot)html] can be used with following default parameters: model=frame+_p2n.model mode=local.


According to some embodiments of the invention, the parameters used with the OneModel FramePlus algorithm are model=frame+_p2n.model, mode=qglobal.


According to some embodiments of the invention, the threshold used to determine homology using the OneModel FramePlus algorithm is 80%, 81%, 82%, 83%, 84%, 85%, 86%, 8%, 88%, 89%, 90%, 91%, 9%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%.


When starting with a polynucleotide sequence and comparing to other polynucleotide sequences the EMBOSS-6.0.1 Needleman-Wunsch algorithm (available from emboss(dot)sourceforge(dot)net/apps/cvs/emboss/apps/needle(dot)html) can be used with the following default parameters: (EMBOSS-6.0.1) gapopen=10; gapextend=0.5; datafile=EDNAFULL; brief=YES.


According to some embodiments of the invention, the parameters used with the EMBOSS-6.0.1 Needleman-Wunsch algorithm are gapopen=10; gapextend=0.2; datafile=EDNAFULL; brief=YES.


According to some embodiments of the invention, the threshold used to determine homology using the EMBOSS-6.0.1 Needleman-Wunsch algorithm for comparison of polynucleotides with polynucleotides is 80%, 81%, 82%, 83%, 84%, 85%, 86%, 8%, 88%, 89%, 90%, 91%, 9%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%.


According to some embodiment, determination of the degree of homology further requires employing the Smith-Waterman algorithm (for protein-protein comparison or nucleotide-nucleotide comparison).


Default parameters for GenCore 6.0 Smith-Waterman algorithm include: model=sw.model.


According to some embodiments of the invention, the threshold used to determine homology using the Smith-Waterman algorithm is 80%, 81%, 82%, 83%, 84%, 85%, 86%, 8%, 88%, 89%, 90%, 91%, 9%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%.


According to some embodiments of the invention, the global homology is performed on sequences which are pre-selected by local homology to the polypeptide or polynucleotide of interest (e.g., 60% identity over 60% of the sequence length), prior to performing the global homology to the polypeptide or polynucleotide of interest (e.g., 80% global homology on the entire sequence). For example, homologous sequences are selected using the BLAST software with the Blastp and tBlastn algorithms as filters for the first stage, and the needle (EMBOSS package) or Frame+algorithm alignment for the second stage. Local identity (Blast alignments) is defined with a very permissive cutoff—60% Identity on a span of 60% of the sequences lengths because it is used only as a filter for the global alignment stage. In this specific embodiment (when the local identity is used), the default filtering of the Blast package is not utilized (by setting the parameter “−F F”).


In the second stage, homologs are defined based on a global identity of at least 80% to the core gene polypeptide sequence.


According to some embodiments of the invention, two distinct forms for finding the optimal global alignment for protein or nucleotide sequences are used:


1. Between Two Proteins (Following the Blastp Filter):

EMBOSS-6.0.1 Needleman-Wunsch algorithm with the following modified parameters: gapopen=8 gapextend=2. The rest of the parameters are unchanged from the default options listed here:


Standard (Mandatory) Qualifiers:
















[-asequence]
sequence
filename and optional format, or reference




(input USA)


[-bsequence]
seqall
Sequence(s) filename and optional format, or




reference (input USA)











    • -gapopen float [10.0 for any sequence]. The gap open penalty is the score taken away when a gap is created. The best value depends on the choice of comparison matrix. The default value assumes you are using the EBLOSUM62 matrix for protein sequences, and the EDNAFULL matrix for nucleotide sequences. (Floating point number from 1.0 to 100.0)

    • -gapextend float [0.5 for any sequence]. The gap extension, penalty is added to the standard gap penalty for each base or residue in the gap. This is how long gaps are penalized. Usually you will expect a few long gaps rather than many short gaps, so the gap extension penalty should be lower than the gap penalty. An exception is where one or both sequences are single reads with possible sequencing errors in which case you would expect many single base gaps. You can get this result by setting the gap open penalty to zero (or very low) and using the gap extension penalty to control gap scoring. (Floating point number from 0.0 to 10.0)





















[-outfile]
align [*needle] Output alignment file name










Additional (Optional) Qualifiers:

    • datafile matrixf [EBLOSUM62 for protein, EDNAFULL for DNA]. This is the scoring matrix file used when comparing sequences. By default, it is the file ‘EBLOSUM62’ (for proteins) or the file ‘EDNAFULL’ (for nucleic sequences). These files are found in the ‘data’ directory of the EMBOSS installation.


Advanced (Unprompted) Qualifiers:

















-[no]brief
boolean [Y] Brief identity and similarity









Associated Qualifiers:














“-asequence” associated qualifiers











-sbegin1
integer
Start of the sequence to be used



-send1
integer
End of the sequence to be used



-sreverse1
boolean
Reverse (if DNA)



-sask1
boolean
Ask for begin/end/reverse



-snucleotide1
boolean
Sequence is nucleotide



-sprotein1
boolean
Sequence is protein



-slower1
boolean
Make lower case



-supper1
boolean
Make upper case



-sformat1
string
Input sequence format



-sdbname1
string
Database name



-sid1
string
Entryname



-ufo1
string
UFO features



-fformat1
string
Features format



-fopenfile1
string
Features file name







“-bsequence” associated qualifiers











-sbegin2
integer
Start of each sequence to be used



-send2
integer
End of each sequence to be used



-sreverse2
boolean
Reverse (if DNA)



-sask2
boolean
Ask for begin/end/reverse



-snucleotide2
boolean
Sequence is nucleotide



-sprotein2
boolean
Sequence is protein



-slower2
boolean
Make lower case



-supper2
boolean
Make upper case



-sformat2
string
Input sequence format



-sdbname2
string
Database name



-sid2
string
Entryname



-ufo2
string
UFO features



-fformat2
string
Features format



-fopenfile2
string
Features file name







“-outfile” associated qualifiers











-aformat3
string
Alignment format



-aextension3
string
File name extension



-adirectory3
string
Output directory



-aname3
string
Base file name



-awidth3
integer
Alignment width



-aaccshow3
boolean
Show accession number in the header



-adesshow3
boolean
Show description in the header



-ausashow3
boolean
Show the full USA in the alignment



-aglobal3
boolean
Show the full sequence in alignment










General Qualifiers:
















-auto
boolean
Turn off prompts


-stdout
boolean
Write first file to standard output


-filter
boolean
Read first file from standard input, write




first file to standard output


-options
boolean
Prompt for standard and additional values


-debug
boolean
Write debug output to program.dbg


-verbose
boolean
Report some/full command line options


-help
boolean
Report command line options. More information




on associated and general qualifiers can be




found with -help -verbose


-warning
boolean
Report warnings


-error
boolean
Report errors


-fatal
boolean
Report fatal errors


-die
boolean
Report dying program messages










2. Between a protein sequence and a nucleotide sequence (following the tblastn filter): GenCore 6.0 OneModel application utilizing the Frame+ algorithm with the following parameters: model=frame+_p2n.model mode=qglobal -q=protein.sequence -db=nucleotide. sequence. The rest of the parameters are unchanged from the default options:














  Usage:


  om -model =< model_fname> [-q=]query [-db=]database [options]


  -model =<model_fname> Specifies the model that you want to run. All models supplied by


Compugen are located in the directory $CGNROOT/models/.


  Valid command line parameters:


  -dev =<dev_name>


  Selects the device to be used by the application.


  Valid devices are:


  bic - Bioccelerator (valid for SW, XSW, FRAME_N2P, and FRAME_P2N models).


  xlg - BioXL/G (valid for all models except XSW).


  xlp - BioXL/P (valid for SW, FRAME+_N2P, and


  FRAME_P2N models).


  xlh - BioXL/H (valid for SW, FRAME+ _N2P, and


  FRAME_P2N models).


  soft - Software device (for all models).











    • q=<query> Defines the query set. The query can be a sequence file or a database reference. You can specify a query by its name or by accession number. The format is detected automatically. However, you may specify a format using the -qfmt parameter. If you do not specify a query, the program prompts for one. If the query set is a database reference, an output file is produced for each sequence in the query.

    • -db=<database name> Chooses the database set. The database set can be a sequence file or a database reference. The database format is detected automatically. However, you may specify a format using -dfmt parameter.

    • -qacc Add this parameter to the command line if you specify query using accession numbers.

    • dacc Add this parameter to the command line if you specify a database using accession numbers.

















-dfmt/-qfmt -<format_type> Chooses the database/query format type. Possible formats are:


fasta - fasta with seq type auto-detected.


fastap - fasta protein seq.


fastan - fasta nucleic seq.


gcg - gcg format, type is auto-detected.


gcg9seq - gcg9 format, type is auto-detected.


gcg9seqp - gcg9 format protein seq.


gcg9seqn - gcg9 format nucleic seq.


nbrf - nbrf seq, type is auto-detected.


nbrfp - nbrf protein seq.


nbrfn - nbrf nucleic seq.


embl - embl and swissprot format.


genbank - genbank format (nucleic).


blast - blast format.


nbrf_gcg - nbrf-gcg seq, type is auto-detected.


nbrf_gcgp - nbrf-gcg protein seq.


nbrf_gcgn - nbrf-gcg nucleic seq.


raw - raw ascii sequence, type is auto-detected.


rawp - raw ascii protein sequence.


rawn - raw ascii nucleic sequence.


pir - pir codata format, type is auto-detected.


profile - gcg profile (valid only for -qfmt


in SW, XSW, FRAME_P2N, and FRAME+_P2N).


-out =<out_fname>  The name of the output file.


-suffix =<name>  The output file name suffix.











    • -gapop=<n> Gap open penalty. This parameter is not valid for FRAME+. For FrameSearch the default is 12.0. For other searches the default is 10.0.

    • -gapext=<n> Gap extend penalty. This parameter is not valid for FRAME+. For FrameSearch the default is 4.0. For other models: the default for protein searches is 0.05, and the default for nucleic searches is 1.0.

    • -qgapop=<n> The penalty for opening a gap in the query sequence. The default is 10.0. Valid for XSW.

    • -qgapext=<n> The penalty for extending a gap in the query sequence. The default is 0.05. Valid for XSW.

    • -start=<n> The position in the query sequence to begin the search.

    • -end=<n> The position in the query sequence to stop the search.

    • -qtrans Performs a translated search, relevant for a nucleic query against a protein database.


      The nucleic query is translated to six reading frames and a result is given for each frame.





Valid for SW and XSW.

    • -dtrans Performs a translated search, relevant for a protein query against a DNA database. Each database entry is translated to six reading frames and a result is given for each frame.


Valid for SW and XSW.


Note: “-qtrans” and “-dtrans” options are mutually exclusive.


-matrix=<matrix_file> Specifies the comparison matrix to be used in the search. The matrix must be in the BLAST format. If the matrix file is not located in $CGNROOT/tables/matrix, specify the full path as the value of the -matrix parameter.

    • -trans=<transtab_name> Translation table. The default location for the table is $CGNROOT/tables/trans.
    • -onestrand Restricts the search to just the top strand of the query/database nucleic sequence.
    • -list=<n> The maximum size of the output hit list. The default is 50.
    • -docalign=<n> The number of documentation lines preceding each alignment. The default is 10.
    • -thr_score=<score_name> The score that places limits on the display of results. Scores that are smaller than -thr_min value or larger than -thr_max value are not shown. Valid options are: quality.
    • zscore.
    • escore.
    • -thr_max=<n> The score upper threshold. Results that are larger than -thr_max value are not shown.
    • -thr_min=<n> The score lower threshold. Results that are lower than -thr_min value are not shown.
    • -align=<n> The number of alignments reported in the output file.
    • -noalign Do not display alignment.


Note: “-align” and “-noalign” parameters are mutually exclusive.

    • -outfmt=<format_name> Specifies the output format type. The default format is PFS.


      Possible values are:
    • PFS—PFS text format
    • FASTA—FASTA text format
    • BLAST—BLAST text format
    • -nonorm Do not perform score normalization.
    • -norm=<norm name> Specifies the normalization method. Valid options are:
    • log—logarithm normalization.
    • std—standard normalization.
    • stat—Pearson statistical method.


Note: “-nonorm” and “-norm” parameters cannot be used together.


Note: Parameters -xgapop, -xgapext, -fgapop, -fgapext, -ygapop, -ygapext, -delop, and -delext apply only to FRAME+.

    • -xgapop=<n> The penalty for opening a gap when inserting a codon (triplet). The default is 12.0.
    • -xgapext=<n> The penalty for extending a gap when inserting a codon (triplet). The default is 4.0.
    • -ygapop=<n> The penalty for opening a gap when deleting an amino acid. The default is 12.0.
    • -ygapext=<n> The penalty for extending a gap when deleting an amino acid. The default is 4.0.
    • -fgapop=<n> The penalty for opening a gap when inserting a DNA base. The default is 6.0.
    • -fgapext=<n> The penalty for extending a gap when inserting a DNA base. The default is 7.0.
    • -delop=<n> The penalty for opening a gap when deleting a DNA base. The default is 6.0.
    • -delext=<n> The penalty for extending a gap when deleting a DNA base. The default is 7.0.
    • -silent No screen output is produced.
    • -host=<host_name> The name of the host on which the server runs. By default, the application uses the host specified in the file $CGNROOT/cgnhosts.
    • -wait Do not go to the background when the device is busy. This option is not relevant for the Parseq or Soft pseudo device.
    • -batch Run the job in the background. When this option is specified, the file “$CGNROOT/defaults/batch.defaults” is used for choosing the batch command. If this file does not exist, the command “at now” is used to run the job.


Note: “-batch” and “-wait” parameters are mutually exclusive.

    • -version Prints the software version number.
    • -help Displays this help message. To get more specific help type:
      • “om -model=<model_fname>-help”.


According to some embodiments the homology is a local homology or a local identity.


Local alignments tools include, but are not limited to the BlastP, BlastN, BlastX or TBLASTN software of the National Center of Biotechnology Information (NCBI), FASTA, and the Smith-Waterman algorithm.


A tblastn search allows the comparison between a protein sequence to the six-frame translations of a nucleotide database. It can be a very productive way of finding homologous protein coding regions in unannotated nucleotide sequences such as expressed sequence tags (ESTs) and draft genome records (HTG), located in the BLAST databases est and htgs, respectively.


Default parameters for blastp include: Max target sequences: 100; Expected threshold: e-5; Word size: 3; Max matches in a query range: 0; Scoring parameters: Matrix—BLOSUM62; filters and masking: Filter—low complexity regions.


Local alignments tools, which can be used include, but are not limited to, the tBLASTX algorithm, which compares the six-frame conceptual translation products of a nucleotide query sequence (both strands) against a protein sequence database. Default parameters include: Max target sequences: 100; Expected threshold: 10; Word size: 3; Max matches in a query range: 0; Scoring parameters: Matrix—BLOSUM62; filters and masking: Filter—low complexity regions.


Modulating the expression and/or activity of the polypeptides of the present invention within a plant cell as to enhance the resistance of the plant to the pathogenic fungi and/or Oomycetes may include enhancing the expression and/or activity of polypeptides identified to positively contribute to the plant defense mechanism against the pathogenic fungi, or reducing the expression and/or activity of those polypeptides found to be associated with susceptibility to the fungus or Oomycete infection.


According to certain embodiments, enhancing the resistance of the plant or part thereof to the pathogenic fungus and/or Oomycete comprises enhancing the expression and/or activity of the at least one polypeptide compared to its expression and/or activity in the control plant.


According to certain embodiments, enhancing the expression and/or activity of the polypeptide comprises introducing into at least one cell of the plant or part thereof an exogenous polynucleotide encoding said polypeptide, thereby producing a transgenic plant over-expressing said polypeptide compared to the control plant.


According to certain embodiments, the exogenous polynucleotide encodes a polypeptide endogenous to the at least one cell. According to other embodiments, the exogenous polynucleotide encodes a polypeptide heterologous to the at least one plant cell.


Nucleic acid sequences encoding the polypeptides of the present invention may be optimized for expression. Examples of such sequence modifications include, but are not limited to, an altered G/C content to more closely approach that typically found in the plant species of interest, and the removal of codons atypically found in the plant species commonly referred to as codon optimization.


The phrase “codon optimization” refers to the selection of appropriate DNA nucleotides for use within a structural gene or fragment thereof that approaches codon usage within the plant of interest, and/or to a process of modifying a nucleic acid sequence for enhanced expression in the host cells of interest by replacing at least one codon (e.g., about or more than about 1, 2, 3, 4, 5, 10, 15, 20, 25, 50, or more codons) of the native sequence with codons that are more frequently or most frequently used in the genes of that host cell while maintaining the native amino acid sequence. Various species exhibit particular bias for certain codons of a particular amino acid. Codon bias (differences in codon usage between organisms) often correlates with the efficiency of translation of messenger RNA (mRNA), which is in turn believed to be dependent on, among other things, the properties of the codons being translated and the availability of particular transfer RNA (tRNA) molecules. The predominance of selected tRNAs in a cell is generally a reflection of the codons used most frequently in peptide synthesis. Accordingly, genes can be tailored for optimal gene expression in a given organism based on codon optimization. Therefore, an optimized gene or nucleic acid sequence refers to a gene in which the nucleotide sequence of a native or naturally occurring gene has been modified in order to utilize statistically-preferred or statistically-favored codons within the plant. The nucleotide sequence typically is examined at the DNA level and the coding region optimized for expression in the plant species determined using any suitable procedure. For example (see U.S. Pat. No. 7,214,862), the standard deviation of codon usage (SDCU), a measure of codon usage bias, may be calculated by first finding the squared proportional deviation of usage of each codon of the native gene relative to that of highly expressed plant genes, followed by a calculation of the average squared deviation. The formula used is:









n
=
1

N



[


(


X
n

-

Y
n


)

/

Y
n


]


2
/
N





wherein Xn refers to the frequency of usage of codon n in highly expressed plant genes, where Yn to the frequency of usage of codon n in the gene of interest and N refers to the total number of codons in the gene of interest. A Table of codon usage from highly expressed genes of dicotyledonous plants is compiled using the data of Murray et al. (1989, Nuc Acids Res. 17:477-498).


Alternative method of optimizing the nucleic acid sequence in accordance with the preferred codon usage for a particular plant cell type is based on the direct use, without performing any extra statistical calculations, of codon optimization Tables such as those provided on-line at the Codon Usage Database through the NIAS (National Institute of Agrobiological Sciences) DNA bank in Japan (www.kazusa.or.jp/codon/). The Codon Usage Database contains codon usage tables for a number of different species, with each codon usage Table having been statistically determined based on the data present in Genbank.


By using the tables described above to determine the most preferred or most favored codons for each amino acid in a particular species (for example, rice), a naturally-occurring nucleotide sequence encoding a protein of interest can be codon optimized for that particular plant species. This is affected by replacing codons that may have a low statistical incidence in the particular species genome with corresponding codons, in regard to an amino acid, that are statistically more favored. However, one or more less-favored codons may be selected to delete existing restriction sites, to create new ones at potentially useful junctions (5′ and 3′ ends to add signal peptide or termination cassettes, internal sites that might be used to cut and splice segments together to produce a correct full-length sequence), or to eliminate nucleotide sequences that may negatively affect mRNA stability or expression.


The naturally-occurring encoding nucleotide sequence may already, in advance of any modification, contain a number of codons that correspond to a statistically-favored codon in a particular plant species. Therefore, codon optimization of the native nucleotide sequence may comprise determining which codons, within the native nucleotide sequence, are not statistically-favored with regards to a particular plant, and modifying these codons in accordance with a codon usage table of the particular plant to produce a codon optimized derivative. A modified nucleotide sequence may be fully or partially optimized for plant codon usage provided that the protein encoded by the modified nucleotide sequence is produced at a level higher than the protein encoded by the corresponding naturally occurring or native gene. Construction of synthetic genes by altering the codon usage is described in for example PCT Patent Application Publication No. WO 93/07278.


Bacterial genes quite often possess multiple methionine initiation codons in proximity to the start of the open reading frame. Translation initiation at one or more of these start codons often leads to generation of a functional protein, and it is not always predetermined which of these codons are used naturally in the bacterium. These start codons can include ATG codons, but additional codons, such GTG, may be used, for example by Bacillus sp. as a start codon, and proteins that initiate translation at GTG codons contain a methionine at the first amino acid. Thus, it is understood that use of one of the alternate methionine codons may also lead to generation of proteins capable of conferring resistance to plants against pathogenic filamentous fungi and/or Oomycetes. These proteins are encompassed within the scope of the present invention. It will be understood that, when expressed in plants, it will be necessary to alter the alternate start codon to ATG for proper translation. In addition, the translation initiator methionine of a polypeptide of the disclosure may be cleaved off post translationally. One skilled in the art understands that the N-terminal translation initiator methionine can be removed by methionine aminopeptidase in many cellular expression systems.


As is known to the skilled Artisan, the polynucleotide coding sequence can be modified to add a codon at the position following the methionine start codon to create a restriction enzyme site for recombinant cloning purposes and/or for expression purposes.


A “signal peptide” is an amino acid sequence which is translated in conjunction with a protein and directs the protein to the secretory system (Chrispeels, J. J., (1991) Ann. Rev. Plant Phys. Plant Mol. Biol. 42:21-53). A signal peptide may form part of the polypeptides of the invention or may be added as described hereinabove. In plants, the signal peptide may preferably direct the protein to the apoplast, the primary site of fungal colonization. When the antifungal proteins of the invention are of bacterial origin, the native fungal signal peptide should be replaced by a plant signal peptide, for example by the maize β-expansin 1 signal peptide (Fursova O et al., Ann Bot. 2012 110(1):47-56; Wu Y et al., Plant Physiol. 2001 126(1) 222-232). Bacterial-originated anti-fungal protein may also include lipid-anchor sequences, which should be modified and/or replaced for effective expression in plant cells.


According to certain embodiments of the present invention, a signal peptide required for expression in specific bacterium or plant species needs to be added or replace the native signal peptide. It is to be explicitly understood that polynucleotides and polypeptides optimized for expression in plant or bacterial cells by modification of their native N-terminus are encompassed within the scope of the present invention, although the global identity of the modified polypeptide to its parent peptide may be less than 70%. A polypeptide that was modified by removal of a native signal peptide thereof is considered herein as a “fragment polypeptide” or a “derived polypeptide”, which includes the amino acid sequence of the mature polypeptide, without the native signal peptide of either a curated or an isolated natural polypeptide. As used herein, the term “optimized polypeptide” refers to a polypeptide encoded by a polynucleotide modified for optimized expression in a desired organism.


According to additional aspect, the present invention provides an isolated or recombinant polynucleotide encoding a polypeptide at least about 70%, at least about 71%, at least about 72%, at least about 73%, at least about 74%, at least about 75%, at least about 76%, at least about 77%, at least about 78%, at least about 79% or more homologous to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:2635, 2444-2634, 2636-4252; or at least about 80%, at least about 81%, at least about 82%, at least about 83%, at least about 84%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99% or more homologous, or identical to an amino acid sequence selected from the group consisting of SEQ ID NOs:326, 360, 308-325, 327-359, 361-506 and 2635, 2444-2634, 2636-4252, a fragment and/or a variant thereof. According to certain embodiments, the present invention provides an isolated polynucleotide encoding a polypeptide comprising the amino acid sequence set forth in any one of SEQ ID NOs:326, 360, 308-325, 327-359, 361-508 and 2635, 2444-2634, 2636-4252. According to additional embodiments, the present invention provides an isolated polypeptide consisting of the amino acid sequence set forth in any one of SEQ ID NOs:326, 360, 308-325, 327-359, 361-508 and 2635, 2444-2634, 2636-4252, a fragment and/or a variant thereof. According to certain embodiments, the present invention provides an isolated or recombinant polynucleotide encoding a polypeptide comprising the amino acid sequence set forth in any one of SEQ ID NOs:507-598. According to certain embodiments, the present invention provides an isolated or recombinant polynucleotide encoding a polypeptide consisting of the amino acid sequence set forth in any one of SEQ ID NOs:507-508. Each possibility represents a separate embodiment of the present invention.


The invention also encompasses fragments of the above described polynucleotides and polypeptides having mutations, such as deletions, insertions or substitutions of one or more amino acids, either naturally occurring or man induced, either randomly or in a targeted fashion.


Expression in plants of polypeptides isolated from bacteria according to some embodiments of the present invention may require modifying the polypeptide sequences for optimized expression. For example, signal peptides forming part of bacterial polypeptides should be typically removed, and optionally replaced with adequate signal peptides which will direct the localization of the polypeptide to a desired plant compartment.


According to certain embodiments, the bacterial polypeptide fragment comprises an amino acid sequence selected from the group consisting of SEQ ID NOs:4647-4799.


According to additional aspect, the present invention provides an isolated polynucleotide, a fragment or a mutant thereof, the polynucleotide comprising a nucleic acids sequence at least about 70%, at least about 71%, at least about 72%, at least about 73%, at least about 74%, at least about 75%, at least about 76%, at least about 77%, at least about 78%, at least about 79% or more homologous to a polynucleotide having a nucleic acid sequence selected from the group consisting of SEQ ID NOs:783, 592-782, 784-2443; or at least about 80%, at least about 81%, at least about 82%, at least about 83%, at least about 84%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99% or more homologous, or identical to a nucleic acid sequence selected from the group consisting of SEQ ID NOs:40, 80, 22-39, 41-79, 81-301 and 783, 592-782, 784-2443. According to certain embodiments, the present invention provides an isolated polynucleotide, a fragment or a mutant thereof, the polynucleotide comprising the nucleic acid sequence set forth in any one of SEQ ID NOs:40, 80, 22-39, 41-79, 81-305 and 783, 592-782, 784-2443. According to additional embodiments, the present invention provides an isolated polynucleotide consisting of the nucleic acid sequence set forth in any one of SEQ ID NOs:40, 80, 22-39, 41-79, 81-305 and 783, 592-782, 784-2443.


According to certain aspects, the present invention provides a recombinant polynucleotide optimized for polypeptide expression in a bacterial expression system, wherein the polypeptide is capable of inhibiting the growth of at least one pathogenic fungus and/or Oomycete.


According to certain exemplary embodiment the bacterium is Escherichia coli. According to these embodiments, the polypeptide comprises an amino acid sequence selected from the group consisting of SEQ ID NOs:4468, 4450-4467, and 4469-4646. According to certain embodiments, the recombinant polynucleotide comprises a nucleic acid sequence selected from the group consisting of SEQ ID NOs:4271, 4253-4270, and 4272-4449.


According to certain exemplary embodiments, the recombinant polynucleotide optimized for polypeptide expression in the bacterial expression system further comprises said bacterium signal peptide.


The isolated polynucleotides and polypeptides of the present invention and the fragment thereof are associated with conferring and/or increasing the resistance of a plant to at least one pathogenic fungus and/or Oomycete.


According to some embodiments of the invention, the exogenous polynucleotide is a non-coding RNA.


As used herein the phrase “non-coding RNA” refers to an RNA molecule which does not encode an amino acid sequence (a polypeptide). Examples of such non-coding RNA molecules include, but are not limited to, an antisense RNA, a pre-miRNA (precursor of a microRNA), or a precursor of a Piwi-interacting RNA (piRNA).


Thus, the invention encompasses nucleic acid sequences described hereinabove; fragments thereof, sequences hybridizable therewith, sequences homologous thereto, sequences encoding similar polypeptides with different codon usage, altered sequences characterized by mutations, such as deletion, insertion or substitution of one or more nucleotides, either naturally occurring or man induced, either randomly or in a targeted fashion.


According to additional aspect, the present invention provides a nucleic acid construct comprising the isolated polynucleotide of the invention, further comprising at least one regulatory element for directing transcription of the nucleic acid sequence in a host plant cell.


According to some embodiments of the invention, expressing the exogenous polynucleotide of the invention within a plant is affected by transforming one or more cells of the plant with the exogenous polynucleotide, followed by generating a mature plant from the transformed cells and cultivating the mature plant under conditions suitable for expressing the exogenous polynucleotide within the mature plant.


According to some embodiments of the invention, the transformation is affected by introducing to the plant cell a nucleic acid construct which includes the exogenous polynucleotide of some embodiments of the invention and at least one promoter for directing transcription of the exogenous polynucleotide in a host cell (a plant cell). Further details of suitable transformation approaches are provided hereinbelow.


As mentioned, the nucleic acid construct according to some embodiments of the invention comprises a promoter sequence and the isolated polynucleotide of some embodiments of the invention.


According to some embodiments of the invention, the isolated polynucleotide is operably linked to the promoter sequence.


A coding nucleic acid sequence is “operably linked” to a regulatory sequence (e.g., promoter) if the regulatory sequence is capable of exerting a regulatory effect on the coding sequence linked thereto.


As used herein, the term “promoter” refers to a region of DNA which lies upstream of the transcriptional initiation site of a gene to which RNA polymerase binds to initiate transcription of RNA. The promoter controls where (e.g., which portion of a plant) and/or when (e.g., at which stage or condition in the lifetime of an organism) the gene is expressed.


According to some embodiments of the invention, the promoter is heterologous to the isolated polynucleotide and/or to the host cell.


As used herein the phrase “heterologous promoter” refers to a promoter from a different species or from the same species but from a different gene locus as of the isolated polynucleotide sequence.


According to some embodiments of the invention, the isolated polynucleotide is heterologous to the plant cell (e.g., the polynucleotide is derived from a different plant species when compared to the plant cell, thus the isolated polynucleotide and the plant cell are not from the same plant species).


Any suitable promoter sequence can be used by the nucleic acid construct of the present invention. Preferably the promoter is selected from the group consisting of a constitutive promoter, a tissue-specific, or biotic-stress specific promoter, particularly promoters inducible by fungi infection.


According to some embodiments of the invention, the promoter is a plant promoter, which is suitable for expression of the exogenous polynucleotide in a plant cell.


Suitable promoters for expression in planta include, but are not limited to, Wheat SPA promoter (SEQ ID NO:1; Albani et al, 1997. Plant Cell, 9:171-184); wheat LMW [SEQ ID NO:2 (longer LMW promoter) and SEQ ID NO:3 (LMW promoter)]; HMW glutenin-1 [SEQ ID NO:4; (Wheat HMW glutenin-1 longer promoter); and SEQ ID NO:5 (Wheat HMW glutenin-1 Promoter); Thomas and Flavell, 1990. The Plant Cell 2:1171-1180; Furtado et al., 2009. Plant Biotechnology Journal 7:240-253]; wheat alpha, beta and gamma gliadins [e.g., SEQ ID NO:6 (wheat alpha gliadin, B genome, promoter); SEQ ID NO:7 (wheat gamma gliadin promoter); Rafalski J A et al. 1984. EMBO 3:1409-1415], wheat TdPR60 [SEQ ID NO:8 (wheat TdPR60 longer promoter) or SEQ ID NO:9 (wheat TdPR60 promoter); Kovalchuk et al., 2009. Plant Mol Biol 71:81-98], maize Ubl Promoter [cultivar Nongda 105 (SEQ ID NO:10); GenBank: DQ141598.1; Taylor et al., 1993. Plant Cell Rep 12: 491-495; and cultivar B73 (SEQ ID NO: 11; Christensen, A H et al. 1992. Plant Mol. Biol. 18(4):675-689); rice actin 1 (SEQ ID NO:12; Mc Elroy et al. 1990, The Plant Cell (2):163-171 rice GOS2 [SEQ ID NO:13 (rice GOS2 longer promoter) and SEQ ID NO:14 (rice GOS2 Promoter); De Pater et al. 1992. Plant J. 2: 837-44], Arabidopsis Pho1 [SEQ ID NO:15 (Arabidopsis Pho1 Promoter); Hamburger et al., Plant Cell. 2002; 14: 889-902,], ExpansinB promoters, e.g., rice ExpB5 [SEQ ID NO:16 (rice ExpB5 longer promoter) and SEQ ID NO:17 (rice ExpB5 promoter)] and Barley ExpB1 [SEQ ID NO: 18 (barley ExpB1 Promoter); Won et al. Mol Cells. 2010. 30:369-76], barley SS2 (sucrose synthase 2; SEQ ID NO: 19; Guerin and Carbonero, 1997. Plant Physiology 114(1):55-62), and rice PG5a (SEQ ID NO:20; U.S. Pat. No. 7,700,835; Nakase et al., 1996. Plant Mol Biol. 32:621-30).


Suitable constitutive promoters include, for example, CaMV 35S promoter [SEQ ID NO:21 (CaMV 35S (pQXNc) Promoter); SEQ ID NO:22 (PJJ 35S from Brachypodium); SEQ ID NO:23 (CaMV 35S (OLD) Promoter; Odell et al., Nature 313:810-812, 1985)], Arabidopsis At6669 promoter [SEQ ID NO:24 (Arabidopsis At6669 (OLD) Promoter; see PCT Publication No. WO04081173 or the new At6669 promoter (SEQ ID NO:25 (Arabidopsis At6669 (NEW) Promoter)]; maize Ubl Promoter [cultivar Nongda 105 (SEQ ID NO: 10); and cultivar B73 (SEQ ID NO:11)]; rice actin 1 (SEQ ID NO:12); pEMU (Last et al., 1991. Theor. Appl. Genet. 81:581-588); CaMV 19S (Nilsson et al., 1997. Physiol. Plant 100:456-462); rice GOS2 [SEQ ID NO:13 (rice GOS2 longer Promoter) and SEQ ID NO: 14 (rice GOS2 Promoter); RBCS promoter (SEQ ID NO:26); Rice cyclophilin (Bucholz et al., 1994 Plant Mol Biol. 25(5):837-43); Maize H3 histone (Lepetit et al., 1992 Mol. Gen. Genet. 231: 276-285); Actin 2 (An et al., 1996. Plant J. 10(1); 107-121) and Synthetic Super MAS (Ni et al., 1995. The Plant Journal 7: 661-676). Other constitutive promoters include those in U.S. Pat. Nos. 5,659,026; 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.


Suitable tissue-specific promoters include, but are not limited to, leaf-specific promoters [e.g., AT5G06690 (Thioredoxin), high expression, SEQ ID NO:27); AT5G61520 (AtSTP3, low expression, SEQ ID NO:28, described in Buttner et al., 2000. Plant, Cell and Environment 23:175-184); or the promoters described in Yamamoto et al., 1997. Plant J. 12:255-265; Kwon et al., 1994. Plant Physiol. 105:357-67; Yamamoto et al., 1994. Plant Cell Physiol. 35:773-778; Gotor et al., 1993. Plant J. 3:509-18; Orozco et al., Plant Mol. Biol. 1993. 23:1129-1138; and Matsuoka et al., 1993. Proc. Natl. Acad. Sci. USA 90:9586-9590; as well as Arabidopsis STP3 (AT5G61520) promoter (Buttner et al., 2000. Plant, Cell and Environment 23:175-184]; seed-preferred promoters [e.g., Napin (originated from Brassica napus which is characterized by a seed specific promoter activity; Stuitje A. R. et. al. 2003. Plant Biotechnology Journal 1(4):301-309; SEQ ID NO:29 (Brassica napus NAPIN Promoter) from seed specific genes (Simon, et al., 1985. Plant Mol. Biol. 5:191; Scofield, et al., 1987. J. Biol. Chem. 262:12202; Baszczynski, et al., 1990. Plant Mol. Biol. 14:633), rice PG5a (SEQ ID NO:20; U.S. Pat. No. 7,700,835), early seed development Arabidopsis BAN (AT1G61720) (SEQ ID NO:30, US 2009/0031450), late seed development Arabidopsis ABI3 (AT3G24650) (SEQ ID NO:31 (Arabidopsis ABI3 (AT3G24650) longer Promoter) or SEQ ID NO:32 (Arabidopsis ABI3 (AT3G24650) Promoter)) (Ng et al., 2004. Plant Molecular Biology 54: 25-38), Brazil Nut albumin (Pearson' et al., 1992. Plant Mol. Biol. 18: 235-245), legumin (Ellis, et al. 1988. Plant Mol. Biol. 10: 203-214), Glutelin (rice) (Takaiwa et al., 1986. Mol. Gen. Genet. 208:15-22; Takaiwa et al., 1987. FEBS Letts. 221: 43-47), Zein (Matzke et al., 1990. Plant Mol Biol, (143):323-332), napA (Stalberg et al., 1996. Planta 199:515-519); Wheat SPA (SEQ ID NO:1); sunflower oleosin (Cummins et al., 1992. Plant Mol. Biol. 19: 873-876); endosperm specific promoters [e.g., wheat LMW (SEQ ID NO:2; Wheat LMW Longer Promoter), and SEQ ID NO:3 (Wheat LMW Promoter)] and HMW glutenin-1 [(SEQ ID NO:4 (Wheat HMW glutenin-1 longer Promoter); and SEQ ID NO:5 (Wheat HMW glutenin-1 Promoter); Colot et al., Mol Gen Genet 216:81-90, 1989; Olin et al., NAR 17:461-2, 1989), wheat alpha, beta and gamma gliadins (SEQ ID NO:6 (wheat alpha gliadin (B genome) promoter); SEQ ID NO:7 (wheat gamma gliadin promoter); Barley ltrl promoter, barley B1, C, D hordein (Cho et al., Theor Appl Gen 98:1253-62, 1999; Muller et al., Plant J 4:343-55, 1993; Sorenson et al., Mol Gen Genet 250:750-60, 1996), Barley DOF (Mena et al., 1998. The Plant Journal 116(1):53-62), Biz2 (EP99106056.7), Barley SS2 (SEQ ID NO:19), wheat Tarp60 (Kovalchuk et al., 2009. Plant Mol Biol 71:81-98), barley D-hordein (D-Hor) and B-hordein (B-Hor) (Agnelo F et al., 2009. Plant Biotech J 793):240-253)], Synthetic promoter (Vicente-Carbajosa et al., 1998. Plant J. 13: 629-640), rice prolamin NRP33, rice-globulin Glb-1 (Wu et al., 1998. Plant Cell Physiology 39(8) 885-889), rice alpha-globulin REB/OHP-1 (Nakase et al. 1997. Plant Mol. Biol. 33: 513-S22), rice ADP-glucose PP (Russell et al., Trans Res 6:157-68, 1997), maize ESR gene family (Opsahl-Ferstad et al., Plant J 12:235-46, 1997), sorgum gamma-kafirin (DeRose et al., PMB 32:1029-35, 1996)], embryo specific promoters [e.g., rice OSHI (Sato et al., Proc. Natl. Acad. Sci. USA, 93: 8117-8122, 1996), KNOX (Postma-Haarsma et al., 1999. Plant Mol. Biol. 39:257-71), rice oleosin (Wu et al., 1998. J. Biochem., 123:386], and flower-specific promoters [e.g., AtPRP4, chalene synthase (chsA) (Van der Meer et al., 1990. Plant Mol. Biol. 15, 95-109), LAT52 (Twell et al., 1989. Mol. Gen Genet 217:240-245), Arabidopsis apetala-3 (Tilly et al., 1998. Development 125:1647-57), Arabidopsis APETALA 1 (AT1G69120, API) (SEQ ID NO:33 (Arabidopsis (AT1G69120) APETALA 1)) (Hempel et al., 1997. Development 124:3845-3853)], and root promoters [e.g., the ROOTP promoter [SEQ ID NO:34]; rice ExpB5 [SEQ ID NO:17 (rice ExpB5 Promoter); or SEQ ID NO:16 (rice ExpB5 longer Promoter)] and barley ExpB1 promoters (SEQ ID NO:18) (Won et al. 2010. Mol. Cells 30: 369-376); Arabidopsis ATTPS-CIN (AT3G25820) promoter (SEQ ID NO:35; Chen et al., 2004. Plant Phys 135:1956-66); Arabidopsis Pho1 promoter (SEQ ID NO: 15), which is also slightly induced by stress].


The nucleic acid construct of some embodiments of the invention can further include an appropriate selectable marker and/or an origin of replication. According to some embodiments of the invention, the nucleic acid construct utilized is a shuttle vector, which can propagate both in E. coli (wherein the construct comprises an appropriate selectable marker and origin of replication) and be compatible with propagation in cells. The construct according to the present invention can be, for example, a plasmid, a bacmid, a phagemid, a cosmid, a phage, a virus or an artificial chromosome.


The nucleic acid construct of some embodiments of the invention can be utilized to stably or transiently transform plant cells. In stable transformation, the exogenous polynucleotide is integrated into the plant genome and as such it represents a stable and inherited trait. In transient transformation, the exogenous polynucleotide is expressed by the cell transformed but it is not integrated into the genome and as such it represents a transient trait.


There are various methods of introducing foreign genes into both monocotyledonous and dicotyledonous plants (Potrykus, I., Annu. Rev. Plant. Physiol. & Plant. Mol. Biol. 1991. 42:205-225; Shimamoto et al., 1989. Nature 338:274-276).


The principle methods of causing stable integration of exogenous DNA into plant genomic DNA include two main approaches:

    • (i) Agrobacterium-mediated gene transfer: Klee et al., (1987) Annu. Rev. Plant Physiol. 38:467-486; Klee and Rogers in Cell Culture and Somatic Cell Genetics of Plants, Vol. 6, Molecular Biology of Plant Nuclear Genes, eds. Schell, J., and Vasil, L. K., Academic Publishers, San Diego, Calif (1989) p. 2-25; Gatenby, in Plant Biotechnology, eds. Kung, S. and Arntzen, C. J., Butterworth Publishers, Boston, Mass. (1989) p. 93-112.
    • (ii) Direct DNA uptake: Paszkowski et al., in Cell Culture and Somatic Cell Genetics of Plants, Vol. 6, Molecular Biology of Plant Nuclear Genes eds. Schell, J., and Vasil, L. K., Academic Publishers, San Diego, Calif. (1989) p. 52-68; including methods for direct uptake of DNA into protoplasts, Toriyama, K. et al. (1988) Bio/Technology 6:1072-1074. DNA uptake induced by brief electric shock of plant cells: Zhang et al. Plant Cell Rep. (1988) 7:379-384. Fromm et al. Nature (1986) 319:791-793. DNA injection into plant cells or tissues by particle bombardment, Klein et al. Bio/Technology (1988) 6:559-563; McCabe et al. Bio/Technology (1988) 6:923-926; Sanford, Physiol. Plant. (1990) 79:206-209; by the use of micropipette systems: Neuhaus et al., Theor. Appl. Genet. (1987) 75:30-36; Neuhaus and Spangenberg, Physiol. Plant. (1990) 79:213-217; glass fibers or silicon carbide whisker transformation of cell cultures, embryos or callus tissue, U.S. Pat. No. 5,464,765 or by the direct incubation of DNA with germinating pollen, DeWet et al. in Experimental Manipulation of Ovule Tissue, eds. Chapman, G. P. and Mantell, S. H. and Daniels, W. Longman, London, (1985) p. 197-209; and Ohta, Proc. Natl. Acad. Sci. USA (1986) 83:715-719.


The Agrobacterium system includes the use of plasmid vectors that contain defined DNA segments that integrate into the plant genomic DNA. Methods of inoculation of the plant tissue vary depending upon the plant species and the Agrobacterium delivery system. A widely used approach is the leaf disc procedure which can be performed with any tissue explant that provides a good source for initiation of whole plant differentiation. See, e.g., Horsch et al. in Plant Molecular Biology Manual A5, Kluwer Academic Publishers, Dordrecht (1988) p. 1-9. A supplementary approach employs the Agrobacterium delivery system in combination with vacuum infiltration. The Agrobacterium system is especially viable in the creation of transgenic dicotyledonous plants.


There are various methods of direct DNA transfer into plant cells. In electroporation, the protoplasts are briefly exposed to a strong electric field. In microinjection, the DNA is mechanically injected directly into the cells using very small micropipettes. In microparticle bombardment, the DNA is adsorbed on microprojectiles such as magnesium sulfate crystals or tungsten particles, and the microprojectiles are physically accelerated into cells or plant tissues.


Following stable transformation plant propagation is exercised. The most common method of plant propagation is by seed. Regeneration by seed propagation, however, has the deficiency that due to heterozygosity there is a lack of uniformity in the crop, since seeds are produced by plants according to the genetic variances governed by Mendelian rules. Basically, each seed is genetically different and each will grow with its own specific traits. Therefore, it is preferred that the transformed plant be produced such that the regenerated plant has the identical traits and characteristics of the parent transgenic plant. Therefore, it is preferred that the transformed plant be regenerated by micropropagation which provides a rapid, consistent reproduction of the transformed plants.


Micropropagation is a process of growing new generation plants from a single piece of tissue that has been excised from a selected parent plant or cultivar. This process permits the mass reproduction of plants having the preferred tissue expressing the fusion protein. The new generation plants which are produced are genetically identical to, and have all of the characteristics of, the original plant. Micropropagation allows mass production of quality plant material in a short period of time and offers a rapid multiplication of selected cultivars in the preservation of the characteristics of the original transgenic or transformed plant. The advantages of cloning plants are the speed of plant multiplication and the quality and uniformity of plants produced.


Micropropagation is a multi-stage procedure that requires alteration of culture medium or growth conditions between stages. Thus, the micropropagation process involves four basic stages: Stage one, initial tissue culturing; stage two, tissue culture multiplication; stage three, differentiation and plant formation; and stage four, greenhouse culturing and hardening. During stage one, initial tissue culturing, the tissue culture is established and certified contaminant-free. During stage two, the initial tissue culture is multiplied until a sufficient number of tissue samples are produced from the seedlings to meet production goals. During stage three, the tissue samples grown in stage two are divided and grown into individual plantlets. At stage four, the transformed plantlets are transferred to a greenhouse for hardening where the plants' tolerance to light is gradually increased so that it can be grown in the natural environment.


According to some embodiments of the invention, the transgenic plant is generated by transient transformation of leaf cells, meristematic cells or the whole plant.


Transient transformation can be performed by any of the direct DNA transfer methods described above or by viral infection using modified plant viruses.


Viruses that have been shown to be useful for the transformation of plant hosts include, for example, Cauliflower mosaic virus (CaMV), Tobacco mosaic virus (TMV), brome mosaic virus (BMV) and Bean Common Mosaic Virus (BV or BCMV). Methods for transformation of plants using plant viruses are well known in the art; see, e.g. U.S. Pat. No. 4,855,237; Gluzman, Y. et al., Communications in Molecular Biology: Viral Vectors, Cold Spring Harbor Laboratory, New York, pp. 172-189 (1988); and Mortimer C et al., 2015. Current Opinion in Biotechnology 32:85-92). Pseudovirus particles for use in expressing foreign DNA in many hosts, including plants are described in WO 87/06261.


According to some embodiments of the invention, the virus used for transient transformations is avirulent and thus is incapable of causing severe symptoms such as reduced growth rate, mosaic, ring spots, leaf roll, yellowing, streaking, pox formation, tumor formation and pitting. A suitable avirulent virus may be a naturally occurring avirulent virus or an artificially attenuated virus. Virus attenuation may be performed by using methods well known in the art including, but not limited to, sub-lethal heating, chemical treatment or by directed mutagenesis techniques such as described, for example, by Kurihara and Watanabe (Molecular Plant Pathology 4:259-269, 2003), Gal-On et al., J Gener Viriol 73: 2183-87 (1992), Atreya et al. Viriology 191:106-11 (1992) and Huet et al. Viriology 75: 1407-14 (1994).


Suitable virus strains can be obtained from available sources such as, for example, the American Type culture Collection (ATCC) or by isolation from infected plants. Isolation of viruses from infected plant tissues can be effected by techniques well known in the art such as described, for example by Foster and Taylor, Eds. “Plant Virology Protocols: From Virus Isolation to Transgenic Resistance (Methods in Molecular Biology Vol 81 Humana Press, 1998). Briefly, tissues of an infected plant believed to contain a high concentration of a suitable virus, preferably young leaves and flower petals, are ground in a buffer solution (e.g., phosphate buffer solution) to produce a virus infected sap which can be used in subsequent inoculations.


Construction of plant RNA viruses for the introduction and expression of non-viral exogenous polynucleotide sequences in plants is demonstrated by the above references as well as by Dawson, W. O. et al., 1989. Virology 172:285-292; Takamatsu et al. 1987. EMBO J 6:307-311; French et al. 1986. Science 231:1294-1297; Takamatsu et al. 1990. FEBS Letters 269:73-76; and U.S. Pat. No. 5,316,931.


When the virus is a DNA virus, suitable modifications can be made to the virus itself. Alternatively, the virus can first be cloned into a bacterial plasmid for ease of constructing the desired viral vector with the foreign DNA. The virus can then be excised from the plasmid. If the virus is a DNA virus, a bacterial origin of replication can be attached to the viral DNA, which is then replicated by the bacteria. Transcription and translation of this DNA will produce the coat protein which will encapsidate the viral DNA. If the virus is an RNA virus, the virus is generally cloned as a cDNA and inserted into a plasmid. The plasmid is then used to make all of the constructions. The RNA virus is then produced by transcribing the viral sequence of the plasmid and translation of the viral genes to produce the coat protein(s) which encapsidate the viral RNA.


In one embodiment, a plant viral polynucleotide is provided in which the native coat protein coding sequence has been deleted from a viral polynucleotide, a non-native plant viral coat protein coding sequence and a non-native promoter, preferably the subgenomic promoter of the non-native coat protein coding sequence, capable of expression in the plant host, packaging of the recombinant plant viral polynucleotide, and ensuring a systemic infection of the host by the recombinant plant viral polynucleotide, has been inserted. Alternatively, the coat protein gene may be inactivated by insertion of the non-native polynucleotide sequence within it, such that a protein is produced. The recombinant plant viral polynucleotide may contain one or more additional non-native subgenomic promoters. Each non-native subgenomic promoter is capable of transcribing or expressing adjacent genes or polynucleotide sequences in the plant host and incapable of recombination with each other and with native subgenomic promoters. Non-native (foreign) polynucleotide sequences may be inserted adjacent the native plant viral subgenomic promoter or the native and a non-native plant viral subgenomic promoters if more than one polynucleotide sequence is included. The non-native polynucleotide sequences are transcribed or expressed in the host plant under control of the subgenomic promoter to produce the desired products.


In a second embodiment, a recombinant plant viral polynucleotide is provided as in the first embodiment except that the native coat protein coding sequence is placed adjacent one of the non-native coat protein subgenomic promoters instead of a non-native coat protein coding sequence.


In a third embodiment, a recombinant plant viral polynucleotide is provided in which the native coat protein gene is adjacent its subgenomic promoter and one or more non-native subgenomic promoters have been inserted into the viral polynucleotide. The inserted non-native subgenomic promoters are capable of transcribing or expressing adjacent genes in a plant host and are incapable of recombination with each other and with native subgenomic promoters. Non-native polynucleotide sequences may be inserted adjacent the non-native subgenomic plant viral promoters such that the sequences are transcribed or expressed in the host plant under control of the subgenomic promoters to produce the desired product.


In a fourth embodiment, a recombinant plant viral polynucleotide is provided as in the third embodiment except that the native coat protein coding sequence is replaced by a non-native coat protein coding sequence.


The viral vectors are encapsidated by the coat proteins encoded by the recombinant plant viral polynucleotide to produce a recombinant plant virus. The recombinant plant viral polynucleotide or recombinant plant virus is used to infect appropriate host plants. The recombinant plant viral polynucleotide is capable of replication in the host, systemic spread in the host, and transcription or expression of foreign gene(s) (exogenous polynucleotide) in the host to produce the desired protein.


Techniques for inoculation of viruses to plants may be found, for example, in Foster and Taylor, Eds. “Plant Virology Protocols: From Virus Isolation to Transgenic Resistance (Methods in Molecular Biology (Humana Pr), Vol 81)”, Humana Press, 1998; Maramorosh and Koprowski, Eds. “Methods in Virology” 7 vols, Academic Press, New York 1967-1984; Hill, S. A. “Methods in Plant Virology”, Blackwell, Oxford, 1984; Walkey, D. G. A. “Applied Plant Virology”, Wiley, New York, 1985; and Kado and Agrawa, eds. “Principles and Techniques in Plant Virology”, Van Nostrand-Reinhold, New York.


In addition to the above, the polynucleotide of the present invention can also be introduced into a chloroplast genome thereby enabling chloroplast expression.


A technique for introducing exogenous polynucleotide sequences to the genome of the chloroplasts is known. This technique involves the following procedures. First, plant cells are chemically treated so as to reduce the number of chloroplasts per cell to about one. Then, the exogenous polynucleotide is introduced via particle bombardment into the cells with the aim of introducing at least one exogenous polynucleotide molecule into the chloroplasts. The exogenous polynucleotides selected such that it is integratable into the chloroplast's genome via homologous recombination which is readily performed by enzymes inherent to the chloroplast. To this end, the exogenous polynucleotide includes, in addition to a gene of interest, at least one polynucleotide stretch which is derived from the chloroplast's genome. In addition, the exogenous polynucleotide includes a selectable marker, which serves by sequential selection procedures to ascertain that all or substantially all of the copies of the chloroplast genomes following such selection will include the exogenous polynucleotide. Further details relating to this technique are found in U.S. Pat. Nos. 4,945,050; and 5,693,507 which are incorporated herein by reference. A polypeptide can thus be produced by the protein expression system of the chloroplast and become integrated into the chloroplast's inner membrane.


According to some embodiments of the invention, the transformed plant is homozygote to the transgene (i.e., the exogenous polynucleotide of some embodiments of the invention), and accordingly all seeds generated thereby include the transgene.


Expressing a plurality of exogenous polynucleotides in a single host plant can be effected by co-introducing multiple nucleic acid constructs, each including a different exogenous polynucleotide, into a single plant cell. The transformed cell can then be regenerated into a mature plant using the methods described hereinabove.


Alternatively, expressing a plurality of exogenous polynucleotides in a single host plant can be effected by co-introducing into a single plant-cell a single nucleic-acid construct including a plurality of different exogenous polynucleotides. Such a construct can be designed with a single promoter sequence which can transcribe a polycistronic messenger RNA including all the different exogenous polynucleotide sequences. To enable co-translation of the different polypeptides encoded by the polycistronic messenger RNA, the polynucleotide sequences can be inter-linked via an internal ribosome entry site (IRES) sequence which facilitates translation of polynucleotide sequences positioned downstream of the IRES sequence. In this case, a transcribed polycistronic RNA molecule encoding the different polypeptides described above will be translated from both the capped 5′ end and the two internal IRES sequences of the polycistronic RNA molecule to thereby produce in the cell all different polypeptides. Alternatively, the construct can include several promoter sequences each linked to a different exogenous polynucleotide sequence.


The plant cell transformed with the construct including a plurality of different exogenous polynucleotides can be regenerated into a mature plant, using the methods described hereinabove.


Alternatively, expressing a plurality of exogenous polynucleotides in a single host plant can be effected by introducing different nucleic acid constructs, including different exogenous polynucleotides, into a plurality of plants. The regenerated transformed plants can then be cross-bred and resultant progeny selected for superior abiotic stress tolerance, water use efficiency, fertilizer use efficiency, early flowering, grain filling period, harvest index, plant height, growth, biomass, yield and/or vigor traits, using conventional plant breeding techniques.


According to certain embodiments, enhancing the resistance of the plant or part thereof to the pathogenic fungus comprises reducing the expression and/or activity of the at least one polypeptide compared to its expression and/or activity in the control plant.


Any method as is known in the art for reducing the expression and/or activity of a plant endogenous protein and the polynucleotide encoding same can be used according to the teachings of the resent invention.


According to certain embodiment of the invention, reducing the expression and/or activity of a polypeptide of the invention within a plant cell comprising transforming the plant cell with a polynucleotide that inhibits the expression of said polypeptide. The polynucleotide may inhibit the transcription or translation of a polynucleotide encoding said polypeptide or can encode for an inhibitory polypeptide interfering with the translation or activity of said polypeptide.


Polynucleotide-Based Methods

According to some embodiments of the present disclosure, a plant is transformed with a polynucleotide that inhibits the expression of a polypeptide of the invention. According to certain exemplary embodiments, the plant cell is transformed within a construct capable of expressing the inhibitory polynucleotide. The term “expression” as used herein refers to the biosynthesis of a gene product, including the transcription and/or translation of said gene product. For example, for the purposes of the present invention, a construct capable of expressing the inhibitory polynucleotide is capable of producing an RNA molecule that inhibits the transcription and/or translation of a polypeptide of the invention. The “expression” or “production” of a protein or polypeptide from a DNA molecule refers to the transcription and translation of the coding sequence to produce the protein or polypeptide, while the “expression” or “production” of a protein or polypeptide from an RNA molecule refers to the translation of the RNA coding sequence to produce the protein or polypeptide. Examples of polynucleotides that inhibit the expression of a CCT polypeptide are given below.


Sense Suppression/Co-Suppression

According to certain embodiments, inhibition of the expression of a polypeptide of the invention may be obtained by sense suppression or co-suppression. For co-suppression, a construct is designed to express an RNA molecule corresponding to all or part of a messenger RNA encoding the polypeptide in the “sense” orientation. Over-expression of the RNA molecule can result in reduced expression of the native gene. Accordingly, multiple plant lines transformed with the co-suppression constructs are screened to identify those that show the greatest inhibition of the polypeptide expression.


The polynucleotide used for co-suppression may correspond to all or part of the sequence encoding the polypeptide of the invention, all or part of the 5′ and/or 3′ untranslated region of said polypeptide transcript or all or part of both the coding sequence and the untranslated regions of a transcript encoding said polypeptide. In some embodiments where the polynucleotide comprises all or part of the coding region for said polypeptide, the construct is designed to eliminate the start codon of the polynucleotide so that no protein product will be translated.


Co-suppression may be used to inhibit the expression of plant genes to produce plants having undetectable protein levels for the proteins encoded by these genes. See, for example, Broin, et al., (2002) Plant Cell 14:1417-1432. Co-suppression may also be used to inhibit the expression of multiple proteins in the same plant. See, for example, U.S. Pat. No. 5,942,657. Methods for using co-suppression to inhibit the expression of endogenous genes in plants are described, for example, in Yu, et al., Phytochemistry (2003) 63:753-763 and U.S. Pat. Nos. 5,034,323, 5,283,184 and 5,942,657. The efficiency of co-suppression may be increased by including a poly-dT region in the expression cassette at a position 3′ to the sense sequence and 5′ of the polyadenylation signal. See for example, US Patent Application Publication Number 2002/0048814. Typically, such a nucleotide sequence has substantial sequence identity to the sequence of the transcript of the endogenous gene, optimally greater than about 65% sequence identity, more optimally greater than about 85% sequence identity, most optimally greater than about 95% sequence identity (U.S. Pat. Nos. 5,283,184 and 5,034,323).


Antisense Suppression

According to some embodiments, inhibition of the expression of a polypeptide of the invention may be obtained by antisense suppression. For antisense suppression, the construct is designed to express an RNA molecule complementary to all or part of a messenger RNA encoding the polypeptide. Over-expression of the antisense RNA molecule can result in reduced expression of the native gene. Accordingly, multiple plant lines transformed with the antisense RNA are screened to identify those that show the greatest inhibition of said polypeptide expression.


The polynucleotide for use in antisense suppression may correspond to all or part of the complement of the sequence encoding the polypeptide of the invention, all or part of the complement of the 5′ and/or 3′ untranslated region of its transcript or all or part of the complement of both the coding sequence and the untranslated regions of a transcript encoding said polypeptide. In addition, the antisense polynucleotide may be fully complementary (i.e., 100% identical to the complement of the target sequence) or partially complementary (i.e., less than 100% identical to the complement of the target sequence) to the target sequence. Antisense suppression may be used to inhibit the expression of multiple proteins in the same plant. See, for example, U.S. Pat. No. 5,942,657. Furthermore, portions of the antisense nucleotides may be used to disrupt the expression of the target gene. Generally, sequences of at least 50 nucleotides, 100 nucleotides, 200 nucleotides, 300, 400, 450, 500, 550 or greater may be used. Methods for using antisense suppression to inhibit the expression of endogenous genes in plants are described, for example, in Liu, et al., (2002) Plant Physiol. 129:1732-1743 and U.S. Pat. Nos. 5,759,829 and 5,942,657. Efficiency of antisense suppression may be increased by including a poly-dT region in the expression cassette at a position 3′ to the antisense sequence and 5′ of the polyadenylation signal (see, e.g. US Patent Application Publication Number 2002/0048814).


Double-Stranded RNA Interference

According to some embodiments, inhibition of the expression of a polypeptide of the invention may be obtained by double-stranded RNA (dsRNA) interference. For dsRNA interference, a sense RNA molecule like that described above for co-suppression and an antisense RNA molecule that is fully or partially complementary to the sense RNA molecule are expressed in the same cell, resulting in inhibition of the expression of the corresponding endogenous messenger RNA.


Expression of the sense and antisense molecules can be accomplished by designing the DNA construct to comprise both a sense sequence and an antisense sequence. Alternatively, separate constructs may be used for the sense and antisense sequences. Multiple plant lines transformed with the dsRNA interference construct(s) are then screened to identify plant lines that show the greatest inhibition of the expression of the polypeptide. Methods for using dsRNA interference to inhibit the expression of endogenous plant genes are described in Waterhouse, et al., (1998) Proc. Natl. Acad. Sci. USA 95:13959-13964, Liu, et al., (2002) Plant Physiol. 129:1732-1743 and WO 1999/49029, WO 1999/53050, WO 1999/61631 and WO 2000/49035.


Hairpin RNA Interference and Intron-Containing Hairpin RNA Interference

According to some embodiments, inhibition of the expression of a polypeptide of the invention may be obtained by hairpin RNA (hpRNA) interference or intron-containing hairpin RNA (ihpRNA) interference. These methods are highly efficient at inhibiting the expression of endogenous genes. See, Waterhouse and Helliwell, (2003) Nat. Rev. Genet. 4:29-38 and the references cited therein.


For hpRNA interference, the construct is designed to express an RNA molecule that hybridizes with itself to form a hairpin structure that comprises a single-stranded loop region and a base-paired stem. The base-paired stem region comprises a sense sequence corresponding to all or part of the endogenous messenger RNA encoding the gene whose expression is to be inhibited and an antisense sequence that is fully or partially complementary to the sense sequence. Thus, the base-paired stem region of the molecule generally determines the specificity of the RNA interference. hpRNA molecules are highly efficient at inhibiting the expression of endogenous genes, and the RNA interference they induce is inherited by subsequent generations of plants. See, for example, Chuang and Meyerowitz, (2000) Proc. Natl. Acad. Sci. USA 97:4985-4990; Stoutjesdijk, et al., (2002) Plant Physiol. 129:1723-1731 and Waterhouse and Helliwell, (2003) Nat. Rev. Genet. 4:29-38. Methods for using hpRNA interference to inhibit or silence the expression of genes are described, for example, in US Patent Application Publication Number 2003/0175965. A transient assay for the efficiency of hpRNA constructs to silence gene expression in vivo has been described by Panstruga, et al., (2003) Mol. Biol. Rep. 30:135-140.


For Intron-Containing Hairpin RNA (ihpRNA) interference, the interfering molecules have the same general structure as for hpRNA, but the RNA molecule additionally comprises an intron that is capable of being spliced in the cell in which the ihpRNA is expressed. The use of an intron minimizes the size of the loop in the hairpin RNA molecule following splicing, and this increases the efficiency of interference. See, for example, Smith, et al., (2000) Nature 407:319-320. In fact, Smith, et al., shows 100% suppression of endogenous gene expression using ihpRNA-mediated interference. Methods for using ihpRNA interference to inhibit the expression of endogenous plant genes are described, for example, in US Patent Application Publication Number 2003/0180945.


The expression cassette for hpRNA interference may also be designed such that the sense sequence and the antisense sequence do not correspond to an endogenous RNA. In this embodiment, the sense and antisense sequence flank a loop sequence that comprises a nucleotide sequence corresponding to all or part of the endogenous messenger RNA of the target gene. Thus, it is the loop region that determines the specificity of the RNA interference. See, for example, WO 2002/00904.


Amplicon-Mediated Interference

Amplicon expression cassettes comprise a plant virus-derived sequence that contains all or part of the target gene but generally not all of the genes of the native virus. The viral sequences present in the transcription product of the construct allow the transcription product to direct its own replication. The transcripts produced by the amplicon may be either sense or antisense relative to the target sequence (i.e., the messenger RNA for the polypeptide of the invention). Methods of using amplicons to inhibit the expression of endogenous plant genes are described, for example, in Angell and Baulcombe, (1997) EMBO J. 16:3675-3684, Angell and Baulcombe, (1999) Plant J. 20:357-362.


Ribozymes

According to some embodiments, the polynucleotide expressed by the construct of the invention is catalytic RNA or has ribozyme activity specific for the messenger RNA of a polypeptide of the invention. Thus, the polynucleotide causes the degradation of the endogenous messenger RNA, resulting in reduced expression of said polypeptide. This method is described, for example, in U.S. Pat. No. 4,987,071.


Small Interfering RNA or Micro RNA

According to certain embodiments of the invention, inhibition of the expression of a polypeptide of the invention may be obtained by RNA interference by expression of a gene encoding a micro RNA (miRNA). miRNAs are regulatory agents consisting of about 22 ribonucleotides. miRNAs are highly efficient at inhibiting the expression of endogenous genes. See, for example, Palatnik1 J F et al., (2003) Nature 425:257-263.


For miRNA interference, the construct is designed to express an RNA molecule that is modeled on an endogenous miRNA gene. The miRNA gene encodes an RNA that forms a hairpin structure containing a 22-nucleotide sequence that is complementary to another endogenous gene (target sequence). For suppressing the expression of a polypeptide of the invention, the 22-nucleotide sequence is selected from the polypeptide transcript sequence and contains 22 nucleotides of said transcript sequence in sense orientation and 21 nucleotides of a corresponding antisense sequence that is complementary to the sense sequence. miRNA molecules are highly efficient at inhibiting the expression of endogenous genes and the RNA interference they induce is inherited by subsequent generations of plants.


Polypeptide-Based Inhibition of Gene Expression

According to certain additional or alternative embodiments, the inhibitory polynucleotide encodes a zinc finger protein that binds to a gene encoding a polypeptide of the invention, resulting in reduced expression of the gene. In particular embodiments, the zinc finger protein binds to a regulatory region of a polypeptide encoding gene. In other embodiments, the zinc finger protein binds to a messenger RNA encoding said polypeptide and prevents its translation. Methods of selecting sites for targeting by zinc finger proteins have been described, for example, in U.S. Pat. No. 6,453,242 and methods for using zinc finger proteins to inhibit the expression of genes in plants are described, for example, in U.S. Pat. No. 7,151,201.


Polypeptide-Based Inhibition of Protein Activity

According to certain additional or alternative embodiments, the polynucleotide encodes an antibody that binds to a polypeptide of the invention and reduces the activity of the polypeptide. In another embodiment, the binding of the antibody results in increased turnover of the antibody-polypeptide complex by cellular quality control mechanisms. The expression of antibodies in plant cells and the inhibition of molecular pathways by expression and binding of antibodies to proteins in plant cells are well known in the art. See, for example, Conrad and Sonnewald, (2003) Nature Biotech. 21:35-36.


According to some embodiments of the invention, up-regulation or down regulation of the expression and/or activity of the polypeptide of the invention is achieved by means of genome editing.


Genome editing is a reverse genetics method which uses artificially engineered nucleases to cut and create specific double-stranded breaks at a desired location(s) in the genome, which are then repaired by cellular endogenous processes such as, homology directed repair (HDR) and non-homologous end-joining (NHEJ). NHEJ directly joins the DNA ends in a double-stranded break, while HDR utilizes a homologous sequence as a template for regenerating the missing DNA sequence at the break point. In order to introduce specific nucleotide modifications to the genomic DNA, a DNA repair template containing the desired sequence must be present during HDR. Genome editing cannot be performed using traditional restriction endonucleases since most restriction enzymes recognize a few base pairs on the DNA as their target and the probability is very high that the recognized base pair combination will be found in many locations across the genome resulting in multiple cuts not limited to a desired location. To overcome this challenge and create site-specific single- or double-stranded breaks, several distinct classes of nucleases have been discovered and bioengineered to date. These include the meganucleases, Zinc finger nucleases (ZFNs), transcription-activator like effector nucleases (TALENs) and CRISPR/Cas system.


Genome editing is a powerful tool to impact target traits by modifications of the target plant genome sequence. Such modifications can result in new or modified alleles or regulatory elements.


In addition, the traces of genome-edited techniques can be used for marker assisted selection (MAS) as is further described hereinunder. Target plants for the mutagenesis/genome editing methods according to the invention are any plants of interest including monocot or dicot plants.


Over-expression of a polypeptide by genome editing can be achieved by: (i) replacing an endogenous sequence encoding the polypeptide of interest or a regulatory sequence under which it is placed, and/or (ii) inserting a new gene encoding the polypeptide of interest in a targeted region of the genome, and/or (iii) introducing point mutations which result in up-regulation of the gene encoding the polypeptide of interest (e.g., by altering the regulatory sequences such as promoter, enhancers, 5′-UTR and/or 3′-UTR, or mutations in the coding sequence).


Down regulation of the expression of a polypeptide by gnome editing can be achieved by (i) replacing an endogenous sequence encoding a polypeptide negatively affecting a desired plant trait, according to some embodiments of the invention enhancing susceptibility of the plant to pathogenic fungi and/or Oomycete or replacing a regulatory sequence under which the endogenous sequence encoding the polypeptide is placed, and/or (ii) introducing point mutations which result in down-regulation of the gene encoding the polypeptide of interest (e.g., by altering the regulatory sequences such as promoter, enhancers, 5′-UTR and/or 3′-UTR, or mutations in the coding sequence).


Genome Editing Systems Overview

Several systems have been reported to enable genome editing implementation. Examples detailed herein below:


Meganucleases—Meganucleases are commonly grouped into four families: the LAGLIDADG family, the GIY-YIG family, the His-Cys box family and the HNH family. These families are characterized by structural motifs, which affect catalytic activity and recognition sequence. For instance, members of the LAGLIDADG family are characterized by having either one or two copies of the conserved LAGLIDADG motif. The four families of meganucleases are widely separated from one another with respect to conserved structural elements and, consequently, DNA recognition sequence specificity and catalytic activity. Meganucleases are found commonly in microbial species and have the unique property of having very long recognition sequences (>14 bp) thus making them naturally very specific for cutting at a desired location. This can be exploited to make site-specific double-stranded breaks directing modifications in regulatory elements or coding regions upon introduction of the desired sequence. One of skill in the art can use these naturally occurring meganucleases, however the number of such naturally occurring meganucleases is limited. To overcome this challenge, mutagenesis and high throughput screening methods have been used to create meganuclease variants that recognize unique sequences. For example, various meganucleases have been fused to create hybrid enzymes that recognize a new sequence. Alternatively, DNA interacting amino acids of the meganuclease can be altered to design sequence specific meganucleases (see e.g., U.S. Pat. No. 8,021,867). Meganucleases can be designed using the methods described in e.g., Certo, M T et al., 2012. Nature Methods 9:073-975; U.S. Pat. Nos. 8,304,222; 8,021,867; 8,119,381; 8,124,369; 8,129,134; 8,133,697; 8,143,015; 8,143,016; 8,148,098; or 8,163,514. Alternatively, meganucleases with site specific cutting characteristics can be obtained using commercially available technologies e.g., Precision Biosciences' Directed Nuclease Editor™ genome editing technology.


ZFNs and TALENs—Two distinct classes of engineered nucleases, zinc-finger nucleases (ZFNs) and transcription activator-like effector nucleases (TALENs), have both proven to be effective at producing targeted double-stranded breaks (Christian et al., 2010. Genetics 186:757-761; Kim et al., 1996. Proc. Natl. Acad. Sci. 93:1156-1160; Li et al., 2011. Nucleic Acids Res 39:359-372; Mahfouz et al., 2011. Proc. Natl. Acad. Sci; 108:2623-2628; Miller et al., 2010. Nat Biotechnol. 29:143-148).


Basically, ZFNs and TALENs restriction endonuclease technology utilizes a non-specific DNA cutting enzyme which is linked to a specific DNA binding domain (either a series of zinc finger domains or TALE repeats, respectively). Typically, a restriction enzyme whose DNA recognition site and cleaving site are separate from each other is selected. The cleaving portion is separated and then linked to a DNA binding domain, thereby yielding an endonuclease with very high specificity for a desired sequence. An exemplary restriction enzyme with such properties is Fokl. Additionally, Fokl has the advantage of requiring dimerization to have nuclease activity and this means the specificity increases dramatically as each nuclease partner recognizes a unique DNA sequence. To enhance this effect, Fokl nucleases have been engineered that can only function as heterodimers and have increased catalytic activity. The heterodimer functioning nucleases avoid the possibility of unwanted homodimer activity and thus increase specificity of the double-stranded break.


Thus, for example to target a specific site, ZFNs and TALENs are constructed as nuclease pairs, with each member of the pair designed to bind adjacent sequences at the targeted site. Upon transient expression in cells, the nucleases bind to their target sites and the FokI domains heterodimerize to create a double-stranded break. Repair of these double-stranded breaks through the nonhomologous end-joining (NHEJ) pathway most often results in small deletions or small sequence insertions. Since each repair made by NHEJ is unique, the use of a single nuclease pair can produce an allelic series with a range of different deletions at the target site. The deletions typically range anywhere from a few base pairs to a few hundred base pairs in length, but larger deletions have successfully been generated in cell culture by using two pairs of nucleases simultaneously (Carlson et al., 2012. Proc. Natl. Acad. Sci 109:17382-17387; Lee et al., 2010. Genome Res 20:81-89). In addition, when a fragment of DNA with homology to the targeted region is introduced in conjunction with the nuclease pair, the double-stranded break can be repaired via homology directed repair to generate specific modifications (Li et al., 2011, ibid; Miller et al., 2010, ibid; Umov et al., 2005. Nature, 435:646-651).


Although the nuclease portions of both ZFNs and TALENs have similar properties, the difference between these engineered nucleases is in their DNA recognition peptide. ZFNs rely on Cys2-His2 zinc fingers and TALENs on TALEs. Both of these DNA recognizing peptide domains have the characteristic that they are naturally found in combinations in their proteins. Cys2-His2 Zinc fingers typically found in repeats that are 3 bp apart and are found in diverse combinations in a variety of nucleic acid interacting proteins. TALEs on the other hand are found in repeats with a one-to-one recognition ratio between the amino acids and the recognized nucleotide pairs. Because both zinc fingers and TALEs happen in repeated patterns, different combinations can be tried to create a wide variety of sequence specificities. Approaches for making site-specific zinc finger endonucleases include, e.g., modular assembly (where Zinc fingers correlated with a triplet sequence are attached in a row to cover the required sequence), OPEN (low-stringency selection of peptide domains vs. triplet nucleotides followed by high-stringency selections of peptide combination vs. the final target in bacterial systems), and bacterial one-hybrid screening of zinc finger libraries, among others. ZFNs can also be designed and obtained commercially from e.g., Sangamo Biosciences™ (Richmond, CA).


Method for designing and obtaining TALENs are described in e.g. Reyon et al., 2012. Nature Biotechnology 30(5):460-5; Miller et al., 2011. Nat Biotechnol. 29:143-148; Cermak et al., 2011. Nucleic Acids Research 39 (12):e82 and Zhang et al., 2011 Nature Biotechnology 29 (2):149-53. A recently developed web-based program named Mojo Hand was introduced by Mayo Clinic for designing TAL and TALEN constructs for genome editing applications (can be accessed through www(dot)talendesign(dot)org). TALEN can also be designed and obtained commercially from e.g., Sangamo Biosciences™ (Richmond, CA).


The ZFN/TALEN system capability for precise targeting can be utilized for directing modifications in regulatory elements and/or coding regions upon introduction of the sequence of interest for trait improvement.


CRISPR/Cas9—The CRISPR/Cas system for genome editing contains two distinct components: a gRNA (guide RNA) and an endonuclease e.g. Cas9.


The gRNA is typically a 20-nucleotide sequence encoding a combination of the target homologous sequence (crRNA) and the endogenous bacterial RNA that links the crRNA to the Cas9 nuclease (tracrRNA) in a single chimeric transcript. The gRNA/Cas9 complex is recruited to the target sequence by the base-pairing between the gRNA sequence and the complement genomic DNA. For successful binding of Cas9, the genomic target sequence must also contain the correct Protospacer Adjacent Motif (PAM) sequence immediately following the target sequence. The binding of the gRNA/Cas9 complex localizes the Cas9 to the genomic target sequence so that the Cas9 can cut both strands of the DNA causing a double-strand break. Just as with ZFNs and TALENs, the double-stranded brakes produced by CRISPR/Cas can undergo homologous recombination or NHEJ.


The Cas9 nuclease has two functional domains: RuvC and HNH, each cutting a different DNA strand. When both of these domains are active, the Cas9 causes double strand breaks in the genomic DNA.


A significant advantage of CRISPR/Cas is that the high efficiency of this system coupled with the ability to easily create synthetic gRNAs enables multiple genes to be targeted simultaneously. In addition, the majority of cells carrying the mutation present biallelic mutations in the targeted genes.


However, apparent flexibility in the base-pairing interactions between the gRNA sequence and the genomic DNA target sequence allows imperfect matches to the target sequence to be cut by Cas9.


Modified versions of the Cas9 enzyme containing a single inactive catalytic domain, either RuvC- or HNH-, are called ‘nickases’. With only one active nuclease domain, the Cas9 nickase cuts only one strand of the target DNA, creating a single-strand break or ‘nick’. A single-strand break, or nick, is normally quickly repaired through the HDR pathway, using the intact complementary DNA strand as the template. However, two proximal, opposite strand nicks introduced by a Cas9 nickase are treated as a double-strand break, in what is often referred to as a ‘double nick’ CRISPR system. A double-nick can be repaired by either NHEJ or HDR depending on the desired effect on the gene target. Thus, if specificity and reduced off-target effects are crucial, using the Cas9 nickase to create a double-nick by designing two gRNAs with target sequences in close proximity and on opposite strands of the genomic DNA would decrease off-target effect as either gRNA alone will result in nicks that will not change the genomic DNA.


Modified versions of the Cas9 enzyme containing two inactive catalytic domains (dead Cas9, or dCas9) have no nuclease activity while still able to bind to DNA based on gRNA specificity. The dCas9 can be utilized as a platform for DNA transcriptional regulators to activate or repress gene expression by fusing the inactive enzyme to known regulatory domains. For example, the binding of dCas9 alone to a target sequence in genomic DNA can interfere with gene transcription.


There is a number of publically available tools available to help choose and/or design target sequences as well as lists of bioinformatically determined unique gRNAs for different genes in different species such as the Feng Zhang lab's Target Finder, the Michael Boutros lab's Target Finder (E-CRISP), the RGEN Tools: Cas-OFFinder, the CasFinder: Flexible algorithm for identifying specific Cas9 targets in genomes and the CRISPR Optimal Target Finder.


In order to use the CRISPR system, both gRNA and Cas9 should be expressed in a target cell. The insertion vector can contain both cassettes on a single plasmid or the cassettes are expressed from two separate plasmids. CRISPR plasmids are commercially available such as the px330 plasmid from Addgene.


Recombinant adeno-associated virus (rAAV) platform—this genome-editing platform is based on rAAV vectors which enable insertion, deletion or substitution of DNA sequences in the genomes of live mammalian cells. The rAAV genome is a single-stranded deoxyribonucleic acid (ssDNA) molecule, either positive- or negative-sensed, which is about 4.7 kb long. These single-stranded DNA viral vectors have high transduction rates and have a unique property of stimulating endogenous homologous recombination in the absence of double-strand DNA breaks in the genome. One of skill in the art can design a rAAV vector to target a desired genomic locus and perform both gross and/or subtle endogenous gene alterations in a cell. rAAV genome editing has the advantage in that it targets a single allele and does not result in any off-target genomic alterations. rAAV genome editing technology is commercially available, for example, the rAAV GENESIS™ system from Horizon™ (Cambridge, UK).


Methods for qualifying efficacy and detecting sequence alteration are well known in the art and include, but not limited to, DNA sequencing, electrophoresis, an enzyme-based mismatch detection assay and a hybridization assay such as PCR, RT-PCR, RNase protection, in-situ hybridization, primer extension, Southern blot, Northern Blot and dot blot analysis. Sequence alterations in a specific gene can also be determined at the protein level using e.g. chromatography, electrophoretic methods, immunodetection assays such as ELISA and Western blot analysis and immunohistochemistry.


In addition, one ordinarily skilled in the art can readily design a knock-in/knock-out construct including positive and/or negative selection markers for efficiently selecting transformed cells that underwent a homologous recombination event with the construct. Positive selection provides a means to enrich the population of clones that have taken up foreign DNA. Non-limiting examples of such positive markers include glutamine synthetase, dihydrofolate reductase (DHFR), markers that confer antibiotic resistance, such as neomycin, hygromycin, puromycin, and blasticidin S resistance cassettes. Negative selection markers are necessary to select against random integrations and/or elimination of a marker sequence (e.g. positive marker). Non-limiting examples of such negative markers include the herpes simplex-thymidine kinase (HSV-TK) which converts ganciclovir (GCV) into a cytotoxic nucleoside analog, hypoxanthine phosphoribosyltransferase (HPRT) and adenine phosphoribosytransferase (ARPT).


Recombination Procedures—Common to Different Genome Editing Systems

“Hit and run” or “in-out”—involves a two-step recombination procedure. In the first step, an insertion-type vector containing a dual positive/negative selectable marker cassette is used to introduce the desired sequence alteration. The insertion vector contains a single continuous region of homology to the targeted locus and is modified to carry the mutation of interest. This targeting construct is linearized with a restriction enzyme at a one site within the region of homology, electroporated into the cells, and positive selection is performed to isolate homologous recombinants. These homologous recombinants contain a local duplication that is separated by intervening vector sequence, including the selection cassette. In the second step, targeted clones are subjected to negative selection to identify cells that have lost the selection cassette via intrachromosomal recombination between the duplicated sequences. The local recombination event removes the duplication and, depending on the site of recombination, the allele either retains the introduced mutation or reverts to wild type. The end result is the introduction of the desired modification without the retention of any exogenous sequences.


The “double-replacement” or “tag and exchange” strategy—involves a two-step selection procedure similar to the hit and run approach, but requires the use of two different targeting constructs. In the first step, a standard targeting vector with 3′ and 5′ homology arms is used to insert a dual positive/negative selectable cassette near the location where the mutation is to be introduced. After electroporation and positive selection, homologous targeted clones are identified. Next, a second targeting vector that contains a region of homology with the desired mutation is electroporated into targeted clones, and negative selection is applied to remove the selection cassette and introduce the mutation. The final allele contains the desired mutation while eliminating unwanted exogenous sequences.


Site-Specific Recombinases—The Cre recombinase derived from the P1 bacteriophage and Flp recombinase derived from the yeast Saccharomyces cerevisiae are site-specific DNA recombinases each recognizing a unique 34 base pair DNA sequence (termed “Lox” and “FRT”, respectively) and sequences that are flanked with either Lox sites or FRT sites can be readily removed via site-specific recombination upon expression of Cre or Flp recombinase, respectively. For example, the Lox sequence is composed of an asymmetric eight base pair spacer region flanked by 13 base pair inverted repeats. Cre recombines the 34 base pair lox DNA sequence by binding to the 13 base pair inverted repeats and catalyzing strand cleavage and religation within the spacer region. The staggered DNA cuts made by Cre in the spacer region are separated by 6 base pairs to give an overlap region that acts as a homology sensor to ensure that only recombination sites having the same overlap region recombine. Basically, the site-specific recombinase system offers means for the removal of selection cassettes after homologous recombination. This system also allows for the generation of conditional altered alleles that can be inactivated or activated in a temporal or tissue-specific manner. Of note, the Cre and Flp recombinases leave behind a Lox or FRT “scar” of 34 base pairs. The Lox or FRT sites that remain are typically left behind in an intron or 3′ UTR of the modified locus, and current evidence suggests that these sites usually do not interfere significantly with gene function. Thus, Cre/Lox and Flp/FRT recombination involves introduction of a targeting vector with 3′ and 5′ homology arms containing the mutation of interest, two Lox or FRT sequences and typically a selectable cassette placed between the two Lox or FRT sequences. Positive selection is applied and homologous recombinants that contain targeted mutation are identified. Transient expression of Cre or Flp in conjunction with negative selection results in the excision of the selection cassette and selects for cells where the cassette has been lost. The final targeted allele contains the Lox or FRT scar of exogenous sequences.


Transposases—As used herein, the term “transposase” refers to an enzyme that binds to the ends of a transposon and catalyzes the movement of the transposon to another part of the genome.


As used herein the term “transposon” refers to a mobile genetic element comprising a nucleotide sequence which can move around to different positions within the genome of a single cell. In the process the transposon can cause mutations and/or change the amount of a DNA in the genome of the cell. A number of transposon systems that are able to also transpose in cells e.g. vertebrates have been isolated or designed, such as Sleeping Beauty [Izsvák and Ivics Molecular Therapy (2004) 9: 147-156], piggyBac [Wilson et al. Molecular Therapy (2007) 15: 139-145], Tol2 [Kawakami et al. PNAS (2000) 97 (21): 11403-11408] or Frog Prince [Miskey et al. Nucleic Acids Res. December 1, (2003) 31(23): 6873-6881]. Generally, DNA transposons translocate from one DNA site to another in a simple, cut-and-paste manner. Each of these elements has their own advantages, for example, Sleeping Beauty is particularly useful in region-specific mutagenesis, whereas Tol2 has the highest tendency to integrate into expressed genes. Hyperactive systems are available for Sleeping Beauty and piggyBac. Most importantly, these transposons have distinct target site preferences, and can therefore introduce sequence alterations in overlapping, but distinct sets of genes. Therefore, to achieve the best possible coverage of genes, the use of more than one element is particularly preferred. The basic mechanism is shared between the different transposases, therefore the piggyBac (PB) is described as an example. PB is a 2.5 kb insect transposon originally isolated from the cabbage looper moth, Trichoplusia ni. The PB transposon consists of asymmetric terminal repeat sequences that flank a transposase, PBase. PBase recognizes the terminal repeats and induces transposition via a “cut-and-paste” based mechanism, and preferentially transposes into the host genome at the tetranucleotide sequence TTAA. Upon insertion, the TTAA target site is duplicated such that the PB transposon is flanked by this tetranucleotide sequence. When mobilized, PB typically excises itself precisely to reestablish a single TTAA site, thereby restoring the host sequence to its pretransposon state. After excision, PB can transpose into a new location or be permanently lost from the genome. Typically, the transposase system offers an alternative means for the removal of selection cassettes after homologous recombination quite similar to the use Cre/Lox or Flp/FRT. Thus, for example, the PB transposase system involves introduction of a targeting vector with 3′ and 5′ homology arms containing the mutation of interest, two PB terminal repeat sequences at the site of an endogenous TTAA sequence and a selection cassette placed between PB terminal repeat sequences. Positive selection is applied and homologous recombinants that contain targeted mutation are identified. Transient expression of PBase removes in conjunction with negative selection results in the excision of the selection cassette and selects for cells where the cassette has been lost. The final targeted allele contains the introduced mutation with no exogenous sequences.


For PB to be useful for the introduction of sequence alterations, there must be a native TTAA site in relatively close proximity to the location where a particular mutation is to be inserted.


Homology Directed Repair (HDR) Homology Directed Repair (HDR) can be used to generate specific nucleotide changes (also known as gene “edits”) ranging from a single nucleotide change to large insertions. In order to utilize HDR for gene editing, a DNA “repair template” containing the desired sequence must be delivered into the cell type of interest with e.g. the guide RNA [gRNA(s)] and Cas9 or Cas9 nickase or other genome editing method (examples herein below). The repair template must contain the desired edit as well as additional homologous sequence immediately upstream and downstream of the target (termed left and right homology arms). The length and binding position of each homology arm is dependent on the size of the change being introduced. The repair template can be a single stranded oligonucleotide, double-stranded oligonucleotide, or double-stranded DNA plasmid depending on the specific application.


The HDR method was successfully used for targeting a specific modification in a coding sequence of a gene in plants [Budhagatapalli Nagaveni et al. (2015) “Targeted Modification of Gene Function Exploiting Homology-Directed Repair of TALEN-Mediated Double-Strand Breaks in Barley”. G3 (Bethesda). 5(9): 1857-1863). Thus, the gfp-specific transcription activator-like effector nucleases were used along with a repair template that, via HDR, facilitates conversion of gfp into yfp, which is associated with a single amino acid exchange in the gene product. The resulting yellow-fluorescent protein accumulation along with sequencing confirmed the success of the genomic editing.


Similarly, Zhao Yongping et al. 2016 (An alternative strategy for targeted gene replacement in plants using a dual-sgRNA/Cas9 design. Scientific Reports 6, Article number: 23890 (2016)) describe co-transformation of Arabidopsis plants with a combinatory dual-sgRNA/Cas9 vector that successfully deleted miRNA gene regions (MIR169a and MIR827a) and second construct that contains sites homologous to Arabidopsis TERMINAL FLOWER 1 (TFL1) for homology-directed repair (HDR) with regions corresponding to the two sgRNAs on the modified construct to provide both targeted deletion and donor repair for targeted gene replacement by HDR.


Specific considerations for Homology Directed Repair (HDR) utilizing CRISPR/Cas9 system are described herein: It should be noted that the repair template should not include a sequence that exhibits more than 90% identity to the gRNA designed to the genomic DNA or to the reverse complement sequence of the gRNA which is designed to the genomic sequence, otherwise the repair template becomes a suitable target for Cas9 cleavage. Additionally or alternatively, when using a short repair template (e.g., about 40-200 base pairs) the repair template should preferably lack the Protospacer Adjacent Motif (PAM) sequence. For example, the PAM could be mutated such that it is no longer present, but the coding region of the gene is not affected (i.e. a silent mutation).


Introduction of large double stranded DNA as repair template can be performed using plasmids, yet, the plasmid should be linearized before transfection.


Activation of Target Genes Using CRISPR/Cas9 System

Many bacteria and archaea contain endogenous RNA-based adaptive immune systems that can degrade nucleic acids of invading phages and plasmids. These systems consist of clustered regularly interspaced short palindromic repeat (CRISPR) genes that produce RNA components and CRISPR associated (Cas) genes that encode protein components. The CRISPR RNAs (crRNAs) contain short stretches of homology to specific viruses and plasmids and act as guides to direct Cas nucleases to degrade the complementary nucleic acids of the corresponding pathogen. Studies of the type II CRISPR/Cas system of Streptococcus pyogenes have shown that three components form an RNA/protein complex and together are sufficient for sequence-specific nuclease activity: the Cas9 nuclease, a crRNA containing 20 base pairs homologous to the target sequence, and a trans-activating crRNA (tracrRNA) (Jinek et al. Science (2012) 337: 816-821). It was further demonstrated that a synthetic chimeric guide RNA (gRNA) composed of a fusion between crRNA and tracrRNA could direct Cas9 to cleave DNA targets that are complementary to the crRNA in vitro. It was also demonstrated that transient expression of CRISPR-associated endonuclease (Cas9) in conjunction with synthetic gRNAs can be used to produce targeted double-stranded brakes in a variety of different species.


The CRISPR/Cas9 system is a remarkably flexible tool for genome manipulation. A unique feature of Cas9 is its ability to bind target DNA independently of its ability to cleave target DNA. Specifically, both RuvC- and HNH-nuclease domains can be rendered inactive by point mutations (D10A and H840A in SpCas9), resulting in a nuclease dead Cas9 (dCas9) molecule that cannot cleave target DNA. The dCas9 molecule retains the ability to bind to target DNA based on the gRNA targeting sequence. The dCas9 can be tagged with transcriptional activators, and targeting these dCas9 fusion proteins to the promoter region results in robust transcription activation of downstream target genes. The simplest dCas9-based activators consist of dCas9 fused directly to a single transcriptional activator. Importantly, unlike the genome modifications induced by Cas9 or Cas9 nickase, dCas9-mediated gene activation is reversible, since it does not permanently modify the genomic DNA.


Indeed, genome editing was successfully used to over-express a protein of interest in a plant by, for example, mutating a regulatory sequence, such as a promoter to overexpress the endogenous polynucleotide operably linked to the regulatory sequence. For example, U.S. Patent Application Publication No. 20160102316 to Rubio Munoz, Vicente et al., describes plants with increased expression of an endogenous DDA1 plant nucleic acid sequence wherein the endogenous DDA1 promoter carries a mutation introduced by mutagenesis or genome editing which results in increased expression of the DDA1 gene, using for example, CRISPR. The method involves targeting of Cas9 to the specific genomic locus, in this case DDA1, via a 20-nucleotide guide sequence of the single-guide RNA. An online CRISPR Design Tool can identify suitable target sites (tools.genome-engineering.org; Ran et al. (2013) Nature Protocols, 8911:2281-2308).


The CRISPR-Cas system was used for altering (increasing or decreasing) gene expression in plants as described in U.S. Patent Application publication No. 20150067922 to Yang; Yinong et al. The engineered, non-naturally occurring gene editing system comprises two regulatory elements, wherein the first regulatory element (a) operable in a plant cell operably linked to at least one nucleotide sequence encoding a CRISPR-Cas system guide RNA (gRNA) that hybridizes with the target sequence in the plant, and a second regulatory element (b) operable in a plant cell operably linked to a nucleotide sequence encoding a Type-II CRISPR-associated nuclease, wherein components (a) and (b) are located on same or different vectors of the system, whereby the guide RNA targets the target sequence and the CRISPR-associated nuclease cleaves the DNA molecule, thus altering the expression of a gene product in a plant. It should be noted that the CRISPR-associated nuclease and the guide RNA do not naturally occur together.


In addition, as described above, point mutations which activate a gene-of-interest and/or which result in over-expression of a polypeptide-of-interest can be also introduced into plants by means of genome editing. Such mutation can be for example, deletions of repressor sequences which result in activation of the gene-of-interest; and/or mutations which insert nucleotides and result in activation of regulatory sequences such as promoters and/or enhancers.


According to additional aspect, the present invention provides a genetically engineered plant having enhanced resistance to at least one fungus and/or Oomycete compared to a non-engineered control plant, the genetically engineered plant comprises at least one cell having modified expression and/or activity of at least one polypeptide at least 70% identical to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:2635, 2444-2634, 2636-4252 or at least 80% identical to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:326, 360, 308-325, 327-359, 361-506 and 2635, 2444-2634, 2636-4252, a fragment and/or a variant thereof compared to the polypeptide expression and/or activity in the non-engineered control plant. Each possibility represents a separate embodiment of the present invention.


According to certain embodiments, the genetically engineered plant comprises at least one cell having modified expression and/or activity of at least one polypeptide having the amino acid sequences selected from the group consisting of SEQ ID NOs:326, 360, 308-325, 327-359, 361-508 and 2635, 2444-2634, 2636-4252. Each possibility represents a separate embodiment of the present invention.


According to certain embodiments, the genetically engineered plant having enhanced resistance to the at least one fungus and/or Oomycete comprises at least one cell with enhanced expression and/or activity of at least one polypeptide at least 70% identical to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs: 2635, 2444-2634, 2636-4252 or at least 80% identical to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:326, 360, 308-325, 327, 329-347, 349-359, 362-506.


According to certain embodiments, the genetically engineered plant comprises at least one cell transformed with an exogenous polynucleotide encoding the at least one polypeptide, the polynucleotide is at least 70% identical to a nucleic acid sequence selected from the group consisting of SEQ ID NOs:783, 592-782, 784-2443 or at least 80% identical to a polynucleotide sequence selected from the group consisting of SEQ ID NOs:40, 80, 22-39, 41, 43-61, 63-79, 81, and 83-301, thereby having an enhanced resistance to the at least one fungus. The exogenous polynucleotide can be endogenous to the plant cell or heterologous to the plant cell.


According to certain embodiments, the genetically engineered plant comprises at least one cell edited to express an exogenous polynucleotide encoding the at least one polypeptide, thereby having an enhanced resistance to the at least one fungus.


According to certain embodiments, the genetically engineered plant comprises at least one cell edited to over-express an endogenous polynucleotide encoding the at least one polypeptide, thereby having an enhanced resistance to the at least one fungus.


According to certain embodiments, the genetically engineered plant having enhanced resistance to the at least one fungus comprises at least one cell with enhanced expression of at least one polynucleotide encoding a polypeptide at least 70% identical to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:2635, 2444-2634, 2636-4252 or at least 80% identical to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:326, 360, 308-325, 327, 329-347, 349-359, 362-506. According to certain exemplary embodiments, the polynucleotide expression in the genetically engineered plant is enhanced in comparison to the polynucleotide expression in a control plant.


According to certain embodiments, the genetically engineered plant having enhanced resistance to the at least one fungus comprises at least one cell with reduced expression and/or activity of the at least one polypeptide at least 80% identical to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:328, 348 and 361. According to certain exemplary embodiments, the polypeptide expression and/or activity in the genetically engineered plant is reduced in comparison to the polypeptide expression and/or activity in a control plant.


According to certain embodiments, the genetically engineered plant having reduced expression and/or activity of the at least one polypeptide comprises at least one cell having reduced expression of a polynucleotide encoding said at least one polypeptide, thereby having an enhanced resistance to the at least one fungus.


According to certain embodiments, the genetically engineered plant comprises a polynucleotide encoding a modified form of the at least one polypeptide, wherein the modified form has reduced or no activity compared to the unmodified form, thereby having an enhanced resistance to the at least one fungus.


According to additional aspect, the present invention provides a genetically engineered plant having enhanced resistance to at least one fungus and/or Oomycete compared to a control plant, the genetically engineered plant comprises at least one cell having modified expression and/or activity of at least one polypeptide at least about 70%, at least about 71%, at least about 72%, at least about 73%, at least about 74%, at least about 75%, at least about 76%, at least about 77%, at least about 78%, at least about 79% or more homologous to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:2635, 2444-2634, 2636-4252; or at least about 80%, at least about 81%, at least about 82%, at least about 83%, at least about 84%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99% or more homologous, or identical to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:326, 360, 308-325, 327-359, 361-506 and 2635, 2444-2634, 2636-4252, a fragment and/or a variant thereof compared to the polypeptide expression and/or activity in a control plant. Each possibility represents a separate embodiment of the present invention.


According to certain embodiments, the genetically engineered plant comprises at least one cell having modified expression and/or activity of at least one polypeptide having the amino acid sequence selected from the group consisting of SEQ ID NOs:326, 360, 308-325, 327-359, 361-508 and 2635, 2444-2634, 2636-4252. Each possibility represents a separate embodiment of the present invention.


According to certain embodiments, the genetically engineered plant comprises at least one cell having modified expression of a polynucleotide encoding the at least one polypeptide.


According to additional aspect, the present invention provides a genetically engineered plant having enhanced resistance to at least one fungus and/or Oomycete compared to a control plant, the genetically engineered plant comprises at least one cell having modified expression of at least one polynucleotide at least about 70%, at least about 71%, at least about 72%, at least about 73%, at least about 74%, at least about 75%, at least about 76%, at least about 77%, at least about 78%, at least about 79% or more homologous to a polynucleotide having a nucleic acid sequence selected from the group consisting of SEQ ID NOs:783, 592-782, 784-2443; or at least about 80%, at least about 81%, at least about 82%, at least about 83%, at least about 84%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99% or more homologous, or identical to a polynucleotide having an nucleic acid sequence selected from the group consisting of SEQ ID NOs:40, 80, 22-39, 41-79, 81-301 and 783, 592-782, 784-2443 compared to the polynucleotide expression and/or activity in a control plant. Each possibility represents a separate embodiment of the present invention.


According to certain embodiments, the genetically engineered plant comprises at least one cell having modified expression of at least one polynucleotide having the nucleic acid sequence selected from the group consisting of SEQ ID NOs:40, 80, 22-39, 41-79, 81-305 and 783, 592-782, 784-2443. Each possibility represents a separate embodiment of the present invention.


According to certain embodiments, modified expression/and or activity of a polypeptide or polynucleotide encoding same comprises enhanced expression and/or activity. According to certain embodiments, modified expression/and or activity of a polypeptide or polynucleotide encoding same comprises reduced expression and/or activity.


According to certain embodiments, the genetically engineered plant having enhanced resistance to the at least one fungus and/or Oomycete comprises at least one cell with enhanced expression and/or activity of at least one polypeptide at least 80% identical to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:326, 360, 308-325, 327, 329-347, 349-359, 362-506 and 2635, 2444-2634, 2636-4252. According to certain exemplary embodiments, the polypeptide expression and/or activity in the genetically engineered plant is enhanced in comparison to the polypeptide expression and/or activity in a control plant.


According to certain embodiments, the genetically engineered plant having enhanced resistance to the at least one fungus and/or Oomycete comprises at least one cell with reduced expression and/or activity of at least one polypeptide at least 80% identical to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:328, 361 and 348. According to certain exemplary embodiments, the polypeptide expression and/or activity in the genetically engineered plant is reduced in comparison to the polypeptide expression and/or activity in a control plant.


According to certain embodiments, the genetically engineered plant having reduced expression and/or activity of the at least one polypeptide comprises at least one cell having reduced expression of a polynucleotide encoding said at least one polypeptide, thereby having an enhanced resistance to the at least one fungus.


According to certain embodiments, the genetically engineered plant comprises a polynucleotide encoding a modified form of the at least one polypeptide, wherein the modified form has reduced or no activity compared to the unmodified form, thereby having an enhanced resistance to the at least one fungus.


Once expressed within the plant cell or the entire plant, the level of the polypeptide encoded by the exogenous polynucleotide can be determined by methods well known in the art such as, activity assays, Western blots using antibodies capable of specifically binding the polypeptide, Enzyme-Linked Immuno Sorbent Assay (ELISA), radio-immuno-assays (RIA), immunohistochemistry, immunocytochemistry, immunofluorescence and the like.


Methods of determining the level in the plant of the RNA transcribed from the exogenous polynucleotide are well known in the art and include, for example, Northern blot analysis, reverse transcription polymerase chain reaction (RT-PCR) analysis (including quantitative, semi-quantitative or real-time RT-PCR) and RNA-in situ hybridization.


The sequence information and annotations uncovered by the present teachings can be harnessed in favor of classical breeding. Thus, sub-sequence data of those polynucleotides described above, can be used as markers for marker assisted selection (MAS), in which a marker is used for indirect selection of a genetic determinant or determinants of a trait of interest (e.g., biomass, growth rate, oil content, yield, abiotic stress tolerance, water use efficiency, nitrogen use efficiency and/or fertilizer use efficiency). Nucleic acid data of the present teachings (DNA or RNA sequence) may contain or be linked to polymorphic sites or genetic markers on the genome such as restriction fragment length polymorphism (RFLP), microsatellites and single nucleotide polymorphism (SNP), DNA fingerprinting (DFP), amplified fragment length polymorphism (AFLP), expression level polymorphism, polymorphism of the encoded polypeptide and any other polymorphism at the DNA or RNA sequence.


Examples of marker assisted selections include, but are not limited to, selection for a morphological trait (e.g., a gene that affects form, coloration, male sterility or resistance such as the presence or absence of awn, leaf sheath coloration, height, grain color, aroma of rice); selection for a biochemical trait (e.g., a gene that encodes a protein that can be extracted and observed; for example, isozymes and storage proteins); selection for a biological trait (e.g., pathogen races or insect biotypes based on host pathogen or host parasite interaction can be used as a marker since the genetic constitution of an organism can affect its susceptibility to pathogens or parasites).


The polynucleotides and polypeptides described hereinabove can be used in a safe and cost-effective manner in a wide range of economical plants, exemplary species of which are described hereinabove.


It will be appreciated that some genes involved in a plant defense mechanism conferring resistance to a particular fungus species may also be involved in resistance to other species, regulated by the same or homologous genes. Of course, the overall defense mechanism is related, not identical, and therefore not all genes involved in resistance to one pathogen will confer resistance to other pathogens. Nonetheless, if a gene confers or enhances resistance to one of the pathogen species, it would be apparent to one skilled in the art to test for resistance to other pathogens, specifically to pathogen of the same genus or that cause similar symptoms.


According to certain embodiments, the fungus and/or Oomycete is selected from, but not limited to, Fusarium verticillioides; Fusarium graminearum; Collotetrichum graminicola; Fusarium avenaceum; Fusarium culmorum; Fusarium oxysporum; Fusarium roseum; Fusarium semitectum; Fusarium solani; Fusarium verticillioides; Fusarium verticillioides var. subglutinans; Acremonium strictum; Albugo candida; Albugo tragopogonis; Alternaria alternate; Alternaria brassicae; Alternaria helianthi; Alternaria zinnia; Aphanomyces euteiches; Ascochyta sorghina; Ascochyta tritici; Aspergillus flavus; Bipolaris maydis O; Bipolaris sorghicola; Bipolaris sorokiniana; Botrytis cinerea; Cephalosporium acremonium; Cephalosporium gramineum; Cephalosporium maydis; Cercospora kikuchii; Cercospora medicaginis; Cercospora sojina; Cercospora sorghi; Cladosporium herbarum; Clavibacter michiganense subsp. Nebraskense; Clavibacter michiganese subsp. Insidiosum; Claviceps purpurea; Claviceps sorghi; Cochliobolus heterostrophus; Colletotrichum dematium (Colletotichum truncatum); Colletotrichum trifolii; Colletotrichum sublineolum; Corn stunt Spiroplasma; Corynespora cassiicola; Curvularia inaequalis; Curvularia lunata; Curvularia pallescens; Diaporthe phaseolorum var. caulivora; Diaporthe phaseolorum var. sojae (Phomopsis sojae); Diplodia macrospora; Erwinia carotovora; Erwinia carotovorum pv. Carotovora; Erwinia chrysanthemi pv. Zea; Erwinia stewartii; Erysiphe cichoracearum; Erysiphe graminis fsp. tritici; Exserohilum turcicum I, II & III; Gaeumannomyces graminis var. tritici; Gibberella zeae (Fusarium graminearum); Gloeocercospora sorghi; Glomerella glycines; Helminthosporium carbonum I, II & III (Cochliobolus carbonum); Helminthosporium pedicellatum; Helminthosporium sorghicola; Kabatiella maydis; Leptosphaeria maculans; Leptosphaerulina briosiana; Leptotrichila medicaginis; Macrophomina phaseolina; Microsphaera diffusa; Mycosphaerella brassicicola; Nigrospora oryzae; Penicillium oxalicum; Perconia circinata; Peronosclerospora maydis; Peronosclerospora philippinensis; Peronosclerospora sacchari; Peronosclerospora sorghi; Peronospora manshurica; Peronospora parasitica; Peronospora trifoliorum; Phakopsora pachyrhizi; Phialophora gregata; Phoma insidiosa; Phoma macdonaldii; Phoma medicaginis var. medicaginis; Phomopsis helianthi; Phyllachara sacchari; Phyllosticta maydis; Phyllosticta sojicola; Physoderma maydis; Physopella zeae; Phytophthora cryptogea; Phytophthora megasperma; Phytophthora megasperma fsp. Glycinea; Plasmopora halstedii; Pseudocercosporella herpotrichoides; Pseudomonas andropogonis; Pseudomonas avenae; Pseudomonas avenae (Pseudomonas alboprecipitans); Pseudomonas syringae p.v. atrofaciens; Pseudomonas syringae p.v. glycinea; Pseudomonas syringae p.v. syringae; Pseudopeziza medicaginis; Puccinia graminis fsp. tritici; Puccinia helianthi; Puccinia polysora; Puccinia purpurea; Puccinia recondita fsp. tritici; Puccinia sorghi; Puccinia striiformis; Pyrenophora tritici-repentis; Pythium aphanidermatum; Pythium arrhenomanes; Pythium debaryanum; Pythium gramicola; Pythium graminicola; Pythium irregular; Pythium splendens; Pythium ultimum; Ramulispora sorghi; Ramulispora sorghicola; Rhizoctonia cerealis; Rhizoctonia solani; Rhizopus arrhizus; Rhizopus oryzae; Rhizopus stolonifera; Sclerophthona macrospora; Sclerospora graminicola; Sclerotinia sclerotiorum; Sclerotinia trifoliorum; Sclerotium rolfsii; Septoria avenae; Septoria glycines; Septoria helianthi; Septoria nodorum; Septoria tritici; Exserohilum turcicum; Sphacelotheca cruenta; Sporisorium reilianum (Sphacelotheca reiliana); Sporisorium sorghi; Stagonospora meliloti; Stemphylium alfalfa; Stemphylium botryosum; Stemphylium herbarum; Stenocarpella maydi (Diplodia maydis); Tilletia indica; Tilletia laevis; Tilletia tritici; Trichoderma viride; Urocystis agropyri; Uromyces striatus; Ustilago maydis; Ustilago tritici; Verticillium albo-atrum; Verticillium dahlia; Xanthomonas campestris p.v. alfalfa; Xanthomonas campestris p.v. holcicola; Xanthomonas campestris p.v. phaseoli; and Xanthomonas campestris p.v. translucens. Each possibility represents a separate embodiment of the present invention.


Specific pathogenic fungi or Oomycetes are known to cause dramatic crop lose due to disease symptoms which negatively affect the quality of the crop. For example, Fusarium verticilloides and Fusarium graminearum cause rot in maize (specifically stalk rot), wheat, sweet paper, eggplants and head blight is wheat. Fusarium oxysporum causes sudden death syndrome (SDS) in soybeans, yellow spots in sugar beet, Panama disease in Banana, and wilt in tomato, sweet pepper, eggplants, potatoes and various plant of the Cucurbitaceae family. Colletotrichum spp. cause stalk rot in maize, anthracnose in sugar beet, tomato and sweet pepper. Botrytis cinerea causes gray mold in tomato, sweet pepper, eggplants and potato. Rust is caused by Puccinia spp. in maize, wheat and sunflower, by Uromyces spp. in sunflower and by Phakopsora in soybean. Phytophthora causes root rot in soybean, late blight in tomato and potato, blight in eggplant and blight fruit rot in sweet pepper. Mycosphaerella graminicola causes leaf blotch in wheat. Mycosphaerella fijiensis causes black leaf streak disease (BLSD; aka black Sigatoka leaf spot) in banana. Septoria lycopersici causes leaf spots in tomato. Verticillium spp. cause wilt disease in canola, sugar beet, tomato, sweet pepper, eggplant and potato. Magnaporthe oryza causes rice blast. Pythium spp. cause damping off disease in maize, soybean, tomato, sweet pepper, eggplant and potato and black vessels in sugar beet. Sclerotinia causes stem rot in soybean and white mold in tomato, sweet pepper, eggplant and potato. Rhizoctonia solani causes root crown rot in sugar beet, sheath blight in rice, and damping off disease in tomato, sweet pepper, eggplant and potato. Maize smut is caused by Ustilago maydis. Alternaria spp. cause leaf spots in sugar beet and sweet pepper, early blight in tomato and potato, and fruit rot in sweet pepper and eggplants. Cercospora causes leaf blight in soybean and leaf spots in sugar beet, sweet pepper, eggplants and potato. Macrophomina causes charcoal rot in maize, wheat, soybean, tomato and potato. Sclerotium rolfsii causes Southern blight in sweet pepper and eggplants. Oidium spp. cause powdery mildew in tomato, sweet pepper, eggplants and potato. Powdery mildew is also caused by Blumeria graminis.


Methods for identifying symptoms caused by various fungi and Oomycetes upon infection of specific plant species, and for measuring the degree of the plant susceptibility/resistance to the infection are well known to those skilled in the art.


The term “plant” as used herein encompasses a whole plant, a grafted plant, ancestor(s) and progeny of the plants and plant parts, including seeds, shoots, stems, roots (including tubers), rootstock, scion, and plant cells, tissues and organs. The plant or part thereof may be in any form including suspension cultures, embryos, meristematic regions, callus tissue, leaves, gametophytes, sporophytes, pollen, and microspores. Plants that are particularly useful in the methods of the invention include all plants which belong to the superfamily Viridiplantae, in particular monocotyledonous and dicotyledonous plants including a fodder or forage legume, ornamental plant, food crop, tree, or shrub selected from the list comprising Acacia spp., Acer spp., Actinidia spp., Aesculus spp., Agathis australis, Albizia amara, Alsophila tricolor, Andropogon spp., Arachis spp, Areca catechu, Astelia fragrans, Astragalus cicer, Baikiaea plurijuga, Betula spp., Brassica spp., Bruguiera gymnorrhiza, Burkea africana, Butea frondosa, Cadaba farinosa, Calliandra spp, Camellia sinensis, Canna indica, Capsicum spp., Cassia spp., Centroema pubescens, Chacoomeles spp., Cinnamomum cassia, Coffea arabica, Colophospermum mopane, Coronillia varia, Cotoneaster serotina, Crataegus spp., Cucumis spp., Cupressus spp., Cyathea dealbata, Cydonia oblonga, Cryptomeria japonica, Cymbopogon spp., Cynthea dealbata, Cydonia oblonga, Dalbergia monetaria, Davallia divaricata, Desmodium spp., Dicksonia squarosa, Dibeteropogon amplectens, Dioclea spp, Dolichos spp., Dorycnium rectum, Echinochloa pyramidalis, Ehraffia spp., Eleusine coracana, Eragrestis spp., Erythrina spp., Eucalypfus spp., Euclea schimperi, Eulalia vilosa, Pagopyrum spp., Feijoa sellowlana, Fragaria spp., Flemingia spp, Freycinetia banks/i, Geranium thunbergii, GinAgo biloba, Glycine javanica, Gliricidia spp, Gossypium hirsutum, Grevillea spp., Guibourtia coleosperma, Hedysarum spp., Hemaffhia altissima, Heteropogon contoffus, Hordeum vulgare, Hyparrhenia rufa, Hypericum erectum, Hypefjhelia dissolute, Indigo incamata, Iris spp., Leptarrhena pyrolifolia, Lespediza spp., Lettuca spp., Leucaena leucocephala, Loudetia simplex, Lotonus bainesli, Lotus spp., Macrotyloma axillare, Malus spp., Manihot esculenta, Medicago saliva, Metasequoia glyptostroboides, Musa sapientum, Nicotianum spp., Onobrychis spp., Ornithopus spp., Oryza spp., Peltophorum africanum, Pennisetum spp., Persea gratissima, Petunia spp., Phaseolus spp., Phoenix canariensis, Phormium cookianum, Photinia spp., Picea glauca, Pinus spp., Pisum sativam, Podocarpus totara, Pogonarthria fleckii, Pogonaffhria squarrosa, Populus spp., Prosopis cineraria, Pseudotsuga menziesii, Pterolobium stellatum, Pyrus communis, Quercus spp., Rhaphiolepsis umbellata, Rhopalostylis sapida, Rhus natalensis, Ribes grossularia, Ribes spp., Robinia pseudoacacia, Rosa spp., Rubus spp., Salix spp., Schyzachyrium sanguineum, Sciadopitys vefficillata, Sequoia sempervirens, Sequoiadendron giganteum, Sorghum bicolor, Spinacia spp., Sporobolus fimbriatus, Stiburus alopecuroides, Stylosanthos humilis, Tadehagi spp, Taxodium distichum, Themeda triandra, Trifolium spp., Triticum spp., Tsuga heterophylla, Vaccinium spp., Vicia spp., Vitis vinifera, Watsonia pyramidata, Zantedeschia aethiopica, Zea mays, amaranth, artichoke, asparagus, broccoli, Brussels sprouts, cabbage, canola, carrot, cauliflower, celery, collard greens, flax, kale, lentil, oilseed rape, okra, onion, potato, rice, soybean, straw, sugar beet, sugar cane, sunflower, tomato, squash tea, maize, wheat, barley, rye, oat, peanut, pea, lentil and alfalfa, cotton, rapeseed, canola, pepper, sunflower, tobacco, eggplant, eucalyptus, a tree, an ornamental plant, a perennial grass and a forage crop. Alternatively, algae and other non-Viridiplantae can be used for the methods of the present invention.


According to some embodiments, the plant used according to the teachings of the present invention is a crop plant such as maize, rice, wheat, barley, peanut, potato, sesame, olive tree, palm oil, banana, soybean, sunflower, canola, sugarcane, alfalfa, millet, Leguminosae (bean, pea), flax, lupinus, rapeseed, tobacco, poplar and cotton. According to certain exemplary embodiments, the crop plant is maize (Zea mays).


According to some embodiment, the plant used according to the teachings of the present invention is a field crop plant selected from the group consisting of tomato, potato, sweet potato, cassava, beets, ginger, horseradish, radish, ginseng, turnip, any root or tuber crop, pepper, eggplant, ground cherry, tomatillo, okra, other fruiting vegetables, cucumber cantaloupe, melon, muskmelon, squash, watermelon and other cucurbit plants.


According to some embodiments of the invention the plant is a dicotyledonous plant.


According to some embodiments of the invention the plant is a monocotyledonous plant.


According to some embodiments the present invention provides a plant cell expressing the exogenous polynucleotide of some embodiments of the invention, the nucleic acid construct comprising the exogenous polynucleotide of some embodiments of the invention and/or the polypeptide of some embodiments of the invention.


According to a further aspect, the present invention provides a fungicidal composition comprising an effective amount of at least one polypeptide at least 70% identical to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:2635, 2444-2634, 2636-4252, a fragment and/or a variant thereof, wherein the fungicidal composition is effective in inhibiting the growth and/or development of at least one plant pathogenic fungi and/or oomycetes. According to certain embodiments, the fungicidal composition comprises at least one polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:2635, 4468, 4661, 2444-2634, 2636-4252, 4450-4467, 4469-4660, and 4662-4799. According to some embodiments, the fungicidal composition comprises at least one polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:4468, 4661, 4450-4467, 4469-4660, and 4662-4799.


According to yet additional aspect, the present invention provides a fungicidal composition comprising bacteria comprising at least one polypeptide at least 70% identical to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:2635, 2444-2634, 2636-4252, a fragment and/or a variant thereof, wherein the fungicidal composition is effective in inhibiting the growth and/or development of at least one plant pathogenic fungi and/or oomycetes. According to certain embodiments, the bacteria comprise at least one polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:2635, 4468, 4661, 2444-2634, 2636-4252, 4450-4467, 4469-4660, and 4662-4799. According to some embodiments, the bacteria comprise at least one polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs:4468, 4661, 4450-4467, 4469-4660, and 4662-4799. Each possibility represents a separate embodiment of the present invention. The bacteria can be of the same species or of different species, with the proviso that the bacteria have no adverse effect on the plant. The bacteria can be alive or dead. Any bacteria suitable for expression of exogenous genes and not having negative effects on plant can be used (see, e.g. Billman-Jacobe H. “Expression in bacteria other than E. coli 1996. Curr. Opinion in Biotech 7:500). According to certain exemplary embodiments, the bacteria are selected from the group consisting of Bacillus subtilis, Lactococcus lacti, Corynebacterium glutamicum; and Bacillus brevis. Particular example is Bacillus Expression systems Lonza XS™ ToolBox.


According to certain embodiments, the fungicidal composition further comprises at least one agriculturally compatible carrier or diluent.


It is appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention, which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable subcombination or as suitable in any other described embodiment of the invention. Certain features described in the context of various embodiments are not to be considered essential features of those embodiments, unless the embodiment is inoperative without those elements.


The following examples are presented in order to more fully illustrate some embodiments of the invention. They should, in no way be construed, however, as limiting the broad scope of the invention. One skilled in the art can readily devise many variations and modifications of the principles disclosed herein without departing from the scope of the invention.


EXAMPLES

Reference is now made to the following examples, which together with the above descriptions illustrate some embodiments of the invention in a non-limiting fashion.


Generally, the nomenclature used herein and the laboratory procedures utilized in the present invention include molecular, biochemical, microbiological and recombinant DNA techniques. Such techniques are thoroughly explained in the literature. See, for example, “Molecular Cloning: A laboratory Manual” Sambrook et al., (1989); “Current Protocols in Molecular Biology” Volumes I-III Ausubel, R. M., ed. (1994); Ausubel et al., “Current Protocols in Molecular Biology”, John Wiley and Sons, Baltimore, Maryland (1989); Perbal, “A Practical Guide to Molecular Cloning”, John Wiley & Sons, New York (1988); Watson et al., “Recombinant DNA”, Scientific American Books, New York; Birren et al. (eds) “Genome Analysis: A Laboratory Manual Series”, Vols. 1-4, Cold Spring Harbor Laboratory Press, New York (1998); methodologies as set forth in U.S. Pat. Nos. 4,666,828; 4,683,202; 4,801,531; 5,192,659 and 5,272,057; “Cell Biology: A Laboratory Handbook”, Volumes I-III Cellis, J. E., ed. (1994); “Current Protocols in Immunology” Volumes I-III Coligan J. E., ed. (1994); Stites et al. (eds), “Basic and Clinical Immunology” (8th Edition), Appleton & Lange, Norwalk, C T (1994); Mishell and Shiigi (eds), “Selected Methods in Cellular Immunology”, W. H. Freeman and Co., New York (1980); “Oligonucleotide Synthesis” Gait, M. J., ed. (1984); “Nucleic Acid Hybridization” Hames, B. D., and Higgins S. J., eds. (1985); “Transcription and Translation” Hames, B. D., and Higgins S. J., Eds. (1984); “A Practical Guide to Molecular Cloning” Perbal, B., (1984) and “Methods in Enzymology” Vol. 1-317, Academic Press; “PCR Protocols: A Guide To Methods and Applications”, Academic Press, San Diego, C A (1990); Marshak et al., “Strategies for Protein Purification and Characterization—A Laboratory Course Manual” CSHL Press (1996); all of which are incorporated by reference as if fully set forth herein. Other general references are provided throughout this document. The procedures therein are believed to be well known in the art and are provided for the convenience of the reader. All the information contained therein is incorporated herein by reference.


Section I: Plant Genes Associated with Fungal Resistance
Example 1: Identification of Plant Genes Associated with Fungal Resistance

The inventors of the present invention have identified polynucleotides related to resistant to fungal infection, particularly to infection by Fusarium verticillioides, Fusarium graminearum or Colletotrichum graminicola. Modulating the expression of the polynucleotide in plants can increase the plant resistance to fungal infection.


The nucleotide sequence datasets used for the polynucleotide identification were originated from publicly available databases as well as from Applicant proprietary sequencing data obtained using the Solexa technology (form e.g. wheat, maize and sorghum). Sequence data from 200 different plant species was introduced into a single, comprehensive database. The information used to build the datasets included gene expression levels, protein annotation, enzymatic activity and involvement in biosynthetic pathways.


Major databases used included:


Genomic Databases






    • Arabidopsis genome [TAIR genome version 6 (Arabidopsis.org/)];

    • Rice genome [IRGSP build 4.0 (rgp.dna.affrc.go.jp/IRGSP/)];

    • Poplar [Populus trichocarpa release 1.1 from JGI (assembly release v1.0) (genome.jgi-psf.org/)];


    • Brachypodium [JGI 4×assembly, brachpodium.org)];

    • Soybean [DOE-JGI SCP, version Glyma0 (phytozome.net/)];

    • Grape [French-Italian Public Consortium for Grapevine Genome Characterization grapevine genome (genoscope.cns.fr/)];

    • Castorbean [TIGR/J Craig Venter Institute 4×assembly [msc.jcvi.org/r communis];

    • Sorghum [DOE-JGI SCP, version Sbi1 [phytozome.net/)]; and

    • Maize “B73” [DOE-JGI SCP, version AGPv2 [phytozome.net/)];


      Databases of Expressed Sequence Tag (EST) and mRNA Sequences:

    • GenBank ncbi.nlm.nih.gov/dbEST;

    • RefSeq (ncbi.nlm.nih.gov/RefSeq/);

    • TAIR (Arabidopsis.org/);





Protein and Pathway Databases:





    • Uniprot [uniprot.org/];

    • AraCyc [Arabidopsis.org/biocyc/index.jsp];

    • ENZYME [expasy.org/enzyme/];





Microarray datasets were downloaded from: GEO (ncbi.nlm.nih.gov/geo/); TAIR (Arabidopsis.org/); and Applicant proprietary microarray data as described in PCT Patent Application Publication No. WO 2008/122980.


QTL (quantitative trait loci) and SNPs information was retrieved from Gramene [gramene.org/qtl/]; and Panzea [panzea.org/index.html].


Database Assembly

Database assembly was performed to build a wide, rich, reliable annotated and easy to analyze database. The assembly comprised data retrieved from publicly available genomic sequences, mRNA sequences, ESTs DNA sequences and QTL data, as well as information regarding gene expression, protein annotation, and involvement in biosynthesis pathway, all in various plant types. The assembly further comprised data retrieved from Applicant proprietary databases produced from various plant types including genomic sequences, mRNA sequences, expression, proteomic and metabolomic data, QTL and GWAS (genome-wide association studies) data.


Database assembly is comprised of a toolbox of gene refining, structuring and annotation as well as analysis tools enabling to construct a tailored database for each gene-discovery project. Gene refining and structuring tools enable to reliably detect splice variants and antisense transcripts, and understand various potential phenotypic outcomes of a single gene. The capabilities of the “LEADS” platform of Compugen LTD. for analyzing human genome have been confirmed and accepted by the scientific community [see e.g., “Widespread Antisense Transcription”, Yelin, et al. (2003) Nature Biotechnology 21, 379-85; “Splicing of Alu Sequences”, Lev-Maor, et al. (2003) Science 300 (5623), 1288-91; “Computational analysis of alternative splicing using EST tissue information”, Xie H et al. Genomics 2002], and have been proven most efficient in plant genomics as well.


EST Clustering and Gene Assembly

For gene clustering and assembly of organisms with available genome sequence data (Arabidopsis, rice, castorbean, grape, Brachypodium, poplar, soybean, sorghum, maize) the genomic LEADS version (GANG) was employed. This tool allows most accurate clustering of ESTs and mRNA sequences on genome, and predicts gene structure as well as alternative splicing events and antisense transcription.


For organisms with no available full genome sequence data, “expressed LEADS” clustering software was applied.


Gene Annotation

Predicted genes and proteins were annotated as follows: BLAST™ search [blast.ncbi.nlm.nih.gov/Blast.cgi] against all plant UniProt [uniprot.org/] sequences was performed. Open reading frames (ORFs) of each putative transcript were analyzed and longest ORF with highest number of homologues was selected as a predicted protein of the transcript. The predicted proteins were analyzed by InterPro [ebi.ac.uk/interpro/].


BLAST™ against proteins from AraCyc and ENZYME databases was used to map the predicted transcripts to AraCyc pathways.


Predicted proteins from different species were compared using BLAST™ algorithm (ncbi.nlm.nih.gov/Blast.cgi) to validate the accuracy of the predicted protein sequence, and for efficient detection of orthologs.


Gene Expression Profiling

Several data sources were exploited for gene expression profiling, namely microarray data and digital expression profile (see below). Genes were analyzed for expression patterns in different plant species and varieties. The analysis was based on differential expression under uninfected and infected conditions, wherein the infection was induced by different pathogens and the gene expression was measured in different plant organs and at different time points along the disease development. Publicly available microarray datasets were downloaded from TAIR and NCBI GEO sites, renormalized, and integrated into the database. Expression profiling is one of the most important resource data for identifying genes related to disease resistance.


A digital expression profile summary was compiled for each gene cluster according to all keywords included in the sequence records comprising the cluster. Digital expression, also known as electronic Northern Blot, is a tool that displays virtual expression profile based on the expressed sequence tag (EST) sequences forming the gene cluster. The tool provides the expression profile of a cluster in terms of plant anatomy (e.g., the tissue/organ in which the gene is expressed), developmental stage (the developmental stages at which a gene can be found) and profile of treatment (provides the physiological conditions under which a gene is expressed such as drought, cold, pathogen infection, etc.). Given a random distribution of ESTs in the different clusters, the digital expression provides a probability value that describes the probability of a cluster having a total of N ESTs to contain X ESTs from a certain collection of libraries. For the probability calculations, the following is taken into consideration: a) the number of ESTs in the cluster, b) the number of ESTs of the implicated and related libraries, c) the overall number of ESTs available representing the species. Thereby clusters with low probability values are highly enriched with ESTs from the group of libraries of interest indicating a specialized expression.


Recently, the accuracy of this system was demonstrated by Portnoy et al., 2009 (Analysis of The Melon Fruit Transcriptome Based on 454 Pyrosequencing, in: Plant & Animal Genomes XVII Conference, San Diego, CA). Transcriptomic analysis, based on relative EST abundance in data was performed by 454 pyrosequencing of cDNA representing mRNA of the melon fruit. Fourteen double strand cDNA samples obtained from two genotypes, two fruit tissues (flesh and rind) and four developmental stages were sequenced. GS FLX pyrosequencing (Roche/454 Life Sciences) of non-normalized and purified cDNA samples yielded 1,150,657 expressed sequence tags that assembled into 67,477 unigenes (32,357 singletons and 35,120 contigs). Analysis of the data obtained against the Cucurbit Genomics Database [icugi.org/] confirmed the accuracy of the sequencing and assembly. Expression patterns of selected genes fitted well their qRT-PCR data.


The produced datasets were used for further comparative analyses of gene expression in various plant species and lines in response to fungal infection.


Example 2: Production of Plant Transcriptomes for Discovery of Genes Correlating with Resistance to the Fungal Infection
Experimental Procedures

The association of gene expression with fungal infection was examined in Brachypodium distachyon, Aegilops spp., Setaria italica, Triticum aestivum and Zea mays.


The following pathogenic fungi were used: Fusarium verticillioides (hereinafter F. verticillioides or Fv). A GFP transformant of F. verticillioides strain A-00149-FGSC 7600 (Oren et al., 2003, Appl. Environ. Microbiol. 69:1695) was used throughout the data generation and validation experiments; and Fusarium graminearum (hereinafter F. graminearum or Fg).


Fungi Infection

Fungal infection of plant roots: this method was used for inoculating plant root with F. verticillioides (Fv) and Fusarium graminearum (Fg). Sterilized seeds from tolerant and sensitive lines of the examined plant were germinated on water agar supplemented with Ampicillin (100 μg/ml) and inoculated after 6 days with an agar plug (1 cm diameter) covered with one-week old fungal mycelium applied to the root site (1 cm below the seed) for 2 hours four days after germination. Control plants were mock inoculated with an empty plug.


RNA extraction was performed using TRIzol Reagent from Invitrogen (invitrogen.com/content.cfm?pageid=469). Approximately 30-50 mg of sample tissue was taken for the analysis. The weighed tissues were ground using pestle and mortar in liquid nitrogen and resuspended in 500 μl of TRIzol Reagent. To the homogenized lysate, 100 μl of chloroform were added followed by precipitation using isopropanol and two washes with 75% ethanol. The RNA was eluted in 30 μl of RNase-free water. RNA samples were cleaned up using Qiagen's RNeasy minikit clean-up protocol as per the manufacturer's protocol (QIAGEN Inc, CA USA).


RNA was extracted from tissues of the infected and mock plants obtained from each treatment as follows:


Direct root infection—Root and basal stem tissues from plants growing under normal or fungal infection conditions were sampled at 6, 24 and 72 hours post infection (hpi) and RNA was extracted as described hereinabove.


Identification and Validation of Gene Associated with Fungal Infection


In order to discover gene expressed in the examined plant species and plant lines with fungal infection, the present inventors utilized available micro-arrays as described in details hereinbelow for each plant species examined. To define correlations between the levels of RNA expression and fungal resistance, parameters related to plant response to fungal infection were analyzed under normal and infected conditions. From plant identified as encompassing variance in the resistance or susceptibility spectrum, hybrids were selected for further association analysis between fungal infection and gene expression after the plants were challenged with F. verticillioides or F. graminearum as described hereinabove.


Fungal infection was phenotyped as follows:


Plants infected via the direct root infection were phenotyped (24 and 72 hpi) for fungal recovery from root and stem tissues by culturing sterilized explants on PDA for 4-5 days and validating the presence of the Fv-GFP strain. For plants infected with Fg, development of necrosis on the roots was monitored along 7 days.


Example 2.1: Production of B. distachyon Plant Transcriptomes

The association of gene expression in Brachypodium lines with fungal infection was investigated utilizing a Brachypodium oligonucleotide micro-array, produced by Agilent Technologies [chem.agilent.com/Scripts/PDS.asp?lPage=50879]. The array oligonucleotide represents about 27,500 Brachypodium genes and transcripts. To define correlations between the levels of RNA expression and fungal resistance, parameters related to responses to fungal infection were analyzed in 30 different Brachypodium lines under normal and infected conditions as described hereinabove. Among them, 5 lines encompassing susceptibility to F. graminearum (designated as ““sensitive”, Table 1) were selected for RNA expression analysis after challenge with F. verticillioides or F. graminearum as described hereinabove.









TABLE 1








Brachypodium varieties used for production of transcriptomic



data and their phenotypic response to F. graminearum infection









Response to


Variety

F. graminearum






Bd20-1
Sensitive


Bd25-1
Sensitive


PI254868
Sensitive


Bd9-1
Sensitive


Bd26-1
Sensitive









Example 2.2: Production of Setaria italica Transcriptome for Discovery of Gene Correlating with Resistance to the Fungal Infection

The association of gene expression in Setaria italica lines with fungal infection was investigated utilizing a Setaria italica oligonucleotide micro-array, produced by Agilent Technologies [chem.agilent.com/Scripts/PDS.asp?lPage=50879]. The array oligonucleotide represents about 27,500 Setaria italica genes and transcripts designed based on data from Public databases (Example 1). To define correlations between the levels of RNA expression and fungal resistance related parameters in responses to fungal infection various plant characteristics of 30 different Setaria italica hybrids were analyzed under normal and infected conditions as described hereinabove. Among them, 5 hybrids encompassing medium resistance to F. graminearum (designated as “medium resistant”, Table 2) were selected for RNA expression analysis after challenge with F. verticillioides or F. graminearum









TABLE 2








Setaria italica varieties used for production of transcriptomic



data and their phenotypic response to F. graminearum infection









Response to


Variety

F. graminearum






PI231736
Medium resistant


PI473596
Medium resistant


PI253494
Medium resistant


PI269971
Medium resistant


Ames27267
Medium resistant









Example 2.3: Production of Aegilops Spp. Transcriptome

In order to define correlations between the levels of RNA expression with fungal resistance related parameters, responses to fungal infection of 30 different Aegilops varieties were analyzed under normal and infected conditions as described hereinabove. Among them, 5 hybrids encompassing resistance to F. graminearum (designated resistant”, Table 3) were selected for RNAseq expression analysis (Illumina) after challenge with F. verticillioides or F. graminearum.









TABLE 3








Aegilops varieties used for production of transcriptomic data



and their phenotypic response to F. garminearum infection













Response to



Species
Variety

F. graminearum









Aegilops geniculata

Coll3-III
Resistant




Aegilops geniculata

Coll3-VI
Resistant




Aegilops tauschii

L84 TQ20
Resistant




Aegilops tauschii

TQ106
Resistant




Aegilops sharonensis

6118VI
Resistant










Example 2.4: Production of Maize Transcriptome

The association of gene expression in Maize lines with fungal infection was investigated utilizing a Maize oligonucleotide micro-array, produced by Agilent Technologies [chem.agilent.com/Scripts/PDS.asp?lPage=50879]. The array oligonucleotide represents about 60K Maize genes and transcripts designed based on data from Public databases (Example 1). To define correlations between the levels of RNA expression and fungal resistance related parameters responses to fungal infection of various plant characteristics of 30 different Maize hybrids were analyzed under normal and infected conditions as described hereinabove. Among them, 6 hybrids encompassing variance in the resistance spectrum to F. verticillioides or C. graminicola (designated as “tolerant” and “sensitive”, Table 4) were selected for RNA expression analysis after challenge with F. verticillioides or F. graminearum









TABLE 4







Maize varieties used for production of transcriptomic data and


their phenotypic response to F. verticillioides infection











Response to



Variety

F. verticillioides








32W86
Tolerant



Klips
Sensitive



W182E
Tolerant



B84
Sensitive



NC350
Tolerant



Ky WS4
Sensitive










Example 2.5: Production of Wheat Transcriptome

The association of gene expression in wheat lines to fungal infection was investigated utilizing a wheat oligonucleotide micro-array, produced by Agilent Technologies [chem.agilent.com/Scripts/PDS.asp?lPage=50879]. The array oligonucleotide represents about 50,000 wheat genes and transcripts.


In order to define correlations between the levels of RNA expression with fungal resistance related parameters, responses to fungal infection of 30 different wheat varieties were analyzed under normal and infected conditions as described hereinabove. Among them, 6 hybrids encompassing variance in the resistance spectrum to F. verticillioides (designated as “tolerant” and “sensitive”, Table 5) were selected for RNA expression analysis after challenge with F. verticillioides or F. graminearum.









TABLE 5







Wheat varieties used for production of transcriptomic data and


their phenotypic response to F. verticillioides infection









Response to


Variety

F. verticillioides






Aurora
Sensitive


Precoce
Tolerant


Barani
Sensitive


N46
Tolerant


Bobwhite
Sensitive


Thacher
Tolerant









Differential Expression Analysis

The analysis was preformed via proprietary differential expression algorithm.


The default query parameters used were: >2 fold change, p value<0.01, FDR<0.5 (FDR=false discovery rate). Stringency varied due to specific experimental context. The following queries were performed across species (aggregated through the use of proprietary ortholog determination), germplasm, organs, types of pathogens treated, and time post infection:

    • 1. Up regulation upon infection: the gene's expression level is higher in infected samples than in mock controls (both resistant and susceptible lines are queried).
    • 2. Stronger expression induction in resistant lines: the gene's expression induction is higher in resistant than in susceptible lines upon infection.
    • 3. Higher basal expression in resistant lines: the gene's expression is higher in resistant than in susceptible lines in uninfected samples.


No type of query is necessary nor sufficient but overall enrichment of positive indications is considered to identify genes significantly qualifying the above criteria


Results

The genes identified using the above differential expression analyses and the indications found per gene are described hereinbelow:


LFS90


Aegilops—The gene was upregulated in the roots in response to Fusarium verticillioides (Fv) at early infection stage (6 and 24 hours post infection—hpi).


Wheat—The wheat gene ortholog was upregulated in the roots and stem in response to Fv at early infection stages (6 and 72 hpi, respectively), and in the inflorescence in response to Fusarium graminearum (Fg) at early infection stages (30 and 50 hpi).


LFS91


Aegilops—The gene was upregulated in the roots in response to Fusarium verticillioides (Fv) at early infection stage (6 and 24 hpi).


Barley—The barley gene ortholog was upregulated at both early and late stages following inflorescence infection with Fusarium graminearum (Fg) or challenging assays with the mycotoxin Deoxynivalenol (DON) 1, 2, 3, 4- and 6-days post infection (dpi).



Brachypodium—The Brachypodium ortholog was upregulated in the roots of several genotypes in response to Fg at early infection stages (48 hpi).


Maize—The maize gene ortholog was upregulated in response to Colletotrichum graminicola (Cg) at early infection stage (120 hpi).



Sorghum—The sorghum ortholog was upregulated in the roots in response to Fv at early infection stages (6, 24 and 72 hpi).


Wheat—The wheat ortholog was upregulated in the inflorescence in response to Fg at early infection stages (50 hpi). QTL—The rice ortholog mapped to a QTL for blast disease resistance.


LFS92


Aegilops—The gene was upregulated in the roots in response to Fusarium verticillioides (Fv) at early infection stage (6 and 24 hpi).


Wheat—The wheat ortholog was upregulated in the inflorescence in response to Fusarium graminearum (Fg) at early infection stages (50 hpi).


Maize—The maize ortholog was upregulated in the inflorescence in response to Fv at early infection stages (72 and 96 hpi). In addition, the ortholog was upregulated in the leaves in response to Colletotrichum graminicola (Cg) at early infection stages (96 and 120 hpi).


QTL—The soybean ortholog mapped to a QTL for fungal disease resistance.


LFS93


Aegilops—The gene was upregulated in the roots in response to Fusarium verticillioides (Fv) at early infection stage (6 and 24 hpi).


QTL—The maize ortholog mapped to QTLs for Fusarium ear rot resistance and for resistance to contamination with the mycotoxin fumonisin.


LFS94


Aegilops—The gene was upregulated in the roots in response to Fusarium verticillioides (Fv) at early infection stage (6 and 24 hpi).


Wheat—The wheat ortholog was upregulated in the roots of resistant genotypes in response to Fv at early infection stages (1.5 and 2.5 hpi) and in the inflorescence in response to Fusarium graminearum (Fg) at early infection stages (30 and 50 hpi) LFS95



Brachypodium—The Brachypodium ortholog gene was upregulated following spikes inoculation with Fusarium graminearum (Fg) (96 hpi).


Wheat—The wheat ortholog was upregulated in the roots of resistant genotypes in response to Fv at early infection stages (1.5 and 2.5 hpi) and in the inflorescence in response to Fusarium graminearum (Fg) at early infection stages (30 and 50 hpi).


Maize—The maize ortholog was upregulated in the inflorescence in response to Fv at early infection stages (96 hpi). In addition, the ortholog was upregulated in the leaves in response to Colletotrichum graminicola (Cg) at early infection stages (96 and 120 hpi).


LFS96


Aegilops—The gene was upregulated in the roots in response to Fusarium verticillioides (Fv), and in several genotypes also to Fusarium graminearum (Fg) at early infection stage (6 and 24 hpi).


Maize—The maize ortholog was upregulated in the inflorescence in response to Fv at early infection stages (72 and 96 hpi). In addition, the ortholog was upregulated in the leaves in response to Colletotrichum graminicola (Cg) at early infection stages (72, 96 and 120 hpi).


QTL—The rice ortholog mapped to a QTL for blast disease resistance.


LFS97


Aegilops—The gene was upregulated in the roots in response to Fusarium verticillioides (Fv) at early infection stage (6 and 24 hpi). In addition, an upregulation in several genotypes was observed in the roots in response to Fusarium graminearum (Fg) at early infection stages (6 and 24 hpi).


Barley—The barley ortholog was upregulated at early and late stages following inflorescence infection with Fg (72, 96 and 144 hpi) or challenging assays with the mycotoxin Deoxynivalenol (DON) (12 and 24 hpi).


Wheat—The wheat ortholog was upregulated in the inflorescence in response to Fg at early infection stages (30 and 50 hpi).


Maize—The maize ortholog was upregulated in the inflorescence in response to Fv at early infection stage (72 hpi). In addition, the gene was upregulated in the leaves in response to Colletotrichum graminicola (Cg) at early infection stage (120 hpi) and was upregulated in the inflorescence following Cg infection (72 hpi).



Sorghum—The sorghum ortholog was upregulated in the roots in response to Fv at early infection stages (2.5 and 6 hpi).


QTL—The maize ortholog mapped to a QTL for resistance to the mycotoxin fumonisin contamination. The Sorghum ortholog mapped to a QTL for rust resistance.


LFS98


Aegilops—The gene was upregulated in the roots in response to Fusarium verticillioides (Fv) at early infection stage (6 and 24 hours post infection—hpi). In addition, an upregulation in several genotypes was observed in the roots, in response to Fusarium graminearum (Fg) at early infection stages (24 hpi).


Maize—The maize ortholog was upregulated in the roots in response to Fv and Fg at early infection stages (6 hpi and 72 hpi, respectively). In addition, the ortholog was upregulated in the leaves in response to Colletotrichum graminicola (Cg) at early infection stage (96 and 120 hpi).



Sorghum—The Sorghum ortholog was upregulated in the roots in response to Fv at early infection stages (24 hpi).


Wheat—The wheat ortholog was upregulated in the roots in response to Fv at late infection stages (10 days post infection—dpi), and in the inflorescence in response to Fg at early infection stages (50 hpi).


QTL—The maize ortholog mapped to a QTL for Fusarium ear rot resistance. The rice ortholog mapped to a QTL for sheath blight disease resistance.


LFS99

Barley—The barley ortholog was upregulated at both early and late stages following inflorescence infection with Fusarium graminearum (Fg) or challenging assays with the mycotoxin Deoxynivalenol (DON) (1, 2, 3, 4 and 6 dpi).


Wheat—The wheat ortholog was upregulated in the roots in response to Fv at early infection stages (6 and 24 hpi). In addition, the ortholog was upregulated in the inflorescence in response to Fg at early infection stages (30 and 50 hpi).


LFS100


Aegilops—The gene was upregulated in the roots in response to Fusarium verticillioides (Fv) at early infection stage (6 and 24 hours post infection—hpi). In addition, an upregulation in several genotypes was observed in the roots, in response to Fusarium graminearum (Fg) at early infection stages (6 and 24 hpi).


Wheat—The wheat ortholog was upregulated in the roots of resistant genotypes in response to Fv at early infection stages (6 and 24 hpi) and in the inflorescence in response to Fg at early infection stages (30 and 50 hpi).



Brachypodium—The Brachypodium ortholog was upregulated in the roots at early stages of both Fv and Fg infections (6 and 24 hpi).


Barley—The barley ortholog was upregulated at both early and late stages following inflorescence infection with Fg or challenging assays with the mycotoxin Deoxynivalenol (DON) (1, 2, 3, 4 and 6 dpi).


QTL—The rice ortholog mapped to a QTL for blast disease resistance


LFS102


Aegilops—The gene was upregulated in the roots in response to Fusarium verticillioides (Fv) at early infection stage (6 and 24 hours post infection—hpi).


Wheat—The wheat ortholog was upregulated in the inflorescence in response to Fg at early infection stages (30 and 50 hpi).


Barley—The barley ortholog was upregulated at late stages following inflorescence infection with Fg (3, 4- and 6-days post infection—dpi) or at early stage following challenging assays with the mycotoxin Deoxynivalenol (DON) (2 and 4 dpi).


Maize—The maize ortholog was upregulated in the leaves in response to Colletotrichum graminicola (Cg) at early and late infection stages (120 hpi) and was upregulated in the inflorescence following Cg infection (4 and 6 dpi). QTL—The maize ortholog mapped to a QTL for fumonisin contamination resistance.


LFS104


Aegilops—The gene was upregulated in the roots in response to Fusarium verticillioides (Fv) at early infection stage (6 and 24 hpi). In addition, an upregulation in several genotypes was observed in the roots, in response to Fusarium graminearum (Fg) at early infection stages (24 hpi).


Wheat—The wheat ortholog was upregulated in the roots in response to Fv at early infection stages (6 and 24 hpi) and in the inflorescence in response to Fg at early infection stages (30 and 50 hpi).



Brachypodium—The Brachypodium ortholog was moderately upregulated following spikes inoculation with Fg (96 hpi).


Barley—The barley ortholog was upregulated at both early and late stages following inflorescence infection with Fg or challenging assays with the mycotoxin Deoxynivalenol (DON) (1, 2, 3, 4- and 6-days post infection—dpi).


Maize—The maize ortholog was upregulated in the leaves in response to Colletotrichum graminicola (Cg) at early and late infection stages (120 hpi).



Sorghum—The Sorghum ortholog was upregulated in the roots in response to Fv at early infection stages (6 hpi).


QTL—The maize ortholog mapped to QTLs for fumonisin contamination resistance. The rice ortholog mapped to a QTL for blast disease resistance.


LFS105


Aegilops—The gene was upregulated in the roots in response to Fusarium verticillioides (Fv) at early infection stage (6 and 24 hpi).


Maize—The maize ortholog was upregulated in the inflorescence in response to Fv at early infection stages (72 hpi). In addition, the ortholog was upregulated in the leaves in response to Colletotrichum graminicola (Cg) at early infection stage (120 hpi).


Wheat—The wheat ortholog was upregulated in the roots in response to Fv at early infection stages (6 and 24 hpi) and in the inflorescence in response to Fusarium graminearum (Fg) at early infection stages (30 and 50 hpi).


QTL—The rice ortholog mapped to a QTL for blast disease resistance


LFS106


Aegilops—The gene was upregulated in the roots in response to Fusarium verticillioides (Fv) at early infection stage (6 and 24 hpi).



Brachypodium—The Brachypodium ortholog was upregulated in the roots of several genotypes in response to Fv at early infection stage (48 hpi). In addition, the gene was upregulated following spikes inoculation with Fusarium graminearum (Fg) (96 hpi).


Wheat—The wheat ortholog was upregulated in the inflorescence in response to Fg at early infection stages (50 hpi).


Maize—The maize ortholog was upregulated in the inflorescence in response to Fv at early infection stage (72 hpi). In addition, the gene was upregulated in the leaves in response to Colletotrichum graminicola (Cg) at early infection stage (120 hpi).



Sorghum—The Sorghum ortholog was upregulated in the roots in response to Fv at early infection stages (6 and 24 hpi)


LFS107


Aegilops—The gene was upregulated in the roots in response to Fusarium verticillioides (Fv) at early infection stage (6 and 24 hpi). In addition, in several genotypes upregulation was observed in the roots in response to Fusarium graminearum (Fg) at early infection stages (24 hpi).


Foxtail millet (Setaria italica)—The foxtail millet ortholog was moderately upregulated in the roots in response to Fg at early infection stages (24 and 48 hpi).


Wheat—The wheat ortholog was upregulated in the roots in response to Fv at early infection stages (6 hpi) and in the inflorescence in response to Fg at early infection stages (30 and 50 hpi).


QTL—The rice ortholog mapped to a QTL for blast disease resistance.


LFS109


Aegilops—The gene was upregulated in the roots in response to Fusarium verticillioides (Fv) at early infection stage (6 and 24 hpi) and upregulated moderately in the roots in response to Fusarium graminearum (Fg) at early infection stage (6 and 24 hpi).



Brachypodium—The Brachypodium ortholog was upregulated in the roots of several genotypes in response to Fg at early infection stages (48 hpi).


Maize—The maize ortholog was upregulated in the inflorescence in response to Fv at early infection stages (72 hpi). In addition, the ortholog was upregulated in the leaves in response to Colletotrichum graminicola (Cg) at early infection stage (48, 96 and 120 hpi).



Sorghum—The Sorghum ortholog was upregulated in the roots in response to Fv at early infection stages (6 and 24 hpi).


LFS110


Aegilops—The gene was upregulated in the roots in response to Fusarium verticillioides (Fv) at early infection stage (6 and 24 hpi). In addition, an upregulation in several genotypes was observed in the roots, in response to Fusarium graminearum (Fg) at early infection stages (24 hpi).


Barley—The barley ortholog was upregulated at early infection stages following inflorescence infection with Fg (96 hpi) or challenging assays with the mycotoxin Deoxynivalenol (DON) (1 and 12 hpi).


Maize—The maize ortholog was upregulated in the leaves in response to Colletotrichum graminicola (Cg) at early infection stages (120 hpi).


QTL—The maize ortholog mapped to QTLs for Fusarium ear rot resistance. The rice ortholog mapped to a QTL for blast disease resistance. The soybean ortholog mapped to a QTL for fungal disease resistance.


LFS111


Aegilops—The gene was upregulated in the roots in response to Fusarium verticillioides (Fv) at early infection stage (6 and 24 hpi).


Foxtail millet (Setaria italica)—The foxtail millet ortholog was moderately upregulated in the roots of several genotypes in response to Fusarium graminearum (Fg) at early infection stages (48 hpi).



Brachypodium—The Brachypodium ortholog was moderately upregulated in the roots of several genotypes in response to Fg at early infection stages (48 hpi).


QTL—The maize ortholog mapped to QTLs for resistance to fumonisin contamination. The rice ortholog mapped to a QTL for blast disease resistance.


LFS112


Aegilops—The gene was upregulated in the roots in response to Fusarium verticillioides (Fv) at early infection stage (6 and hpi).


Foxtail millet (Setaria italica)—The foxtail millet ortholog was upregulated in the roots of several genotypes at early infection stages of Fv infection (6 and 24 hpi).


Maize—The maize ortholog was upregulated in the inflorescence in response to Fv at early infection stage (72 hpi). In addition, the gene was upregulated in the leaves in response to Colletotrichum graminicola (Cg) at early and late infection stages (120 hpi) and was upregulated in the inflorescence following Cg infection (4 and 6 dpi).


QTL—The rice ortholog mapped to a QTL for sheath blast disease resistance. The soybean ortholog is mapped to a QTL for a fungal disease resistance


LFS113


Aegilops—The gene was upregulated in the roots in response to Fusarium verticillioides (Fv) at early infection stage (6 and 24 hpi).


Barley—The barley ortholog was upregulated at both early and late stages following inflorescence infection with Fusarium graminearum (Fg) or challenging assays with the mycotoxin Deoxynivalenol (DON) (1, 2, 3, 4 and 6 dpi).


Foxtail millet (Setaria italica)—The foxtail millet ortholog was upregulated in the roots of several genotypes in response to Fg at early infection stages (24 and 48 hpi).



Brachypodium—The Brachypodium ortholog was upregulated in the roots of several genotypes in response to Fg at early infection stages (48 hpi).


Wheat—The wheat ortholog was upregulated in the inflorescence in response to Fg at early infection stages (30 and 50 hpi).


QTL—The Sorghum ortholog mapped to a QTL for anthracnose resistance. The rice ortholog mapped to a QTL for blast disease resistance.


LFS114


Aegilops—The gene was upregulated in the roots in response to Fusarium verticillioides (Fv) and Fusarium graminearum (Fg) at early infection stage (6 and 24 hpi).



Brachypodium—The gene was upregulated in the roots in response to Fg at early infection stage (24 and 48 hpi).


Barley—The barley ortholog was upregulated at early stages following inflorescence challenging with the mycotoxin Deoxynivalenol (DON) (1 and 12 hpi).


Wheat—The wheat ortholog was upregulated in inflorescence in response to Fg at early infection stages (30 and 50 hpi).


Maize—The maize ortholog was upregulated in the roots in response to Fv at early (6 hpi) and late (6 and 14 dpi) infection stages. In addition, the ortholog was upregulated in the inflorescence in response to Fv at early infection stage (72 hpi). The ortholog was also upregulated in the roots in response to Fg at early infection stages (24 and 72 hpi). An upregulation was also observed in the leaves in response to Colletotrichum graminicola (Cg) infection (96 and 120 hpi).



Sorghum—The Sorghum ortholog was upregulated in the roots in response to Fv at early infection stages (6 and 24 hpi)


LFS115


Aegilops—The gene was upregulated in the roots in response to Fusarium graminearum (Fg) at early infection stage (6 and 24 hpi).


LFS116


Aegilops—The gene was upregulated in the roots in response to Fusarium verticillioides (Fv) at early infection stage (6 and 24 hpi).



Brachypodium—The Brachypodium ortholog was upregulated following spikes inoculation with Fusarium graminearum (Fg) (96 hpi).


Wheat—The wheat ortholog was upregulated in the inflorescence in response to Fg at early infection stages (30 and 50 hpi).


QTL—The rice ortholog mapped to a QTL for blast disease resistance.


LFS117


Aegilops—The gene was upregulated in the roots in response to Fusarium verticillioides (Fv) at early infection stage (6 and 24 hours post infection—hpi).



Brachypodium—The Brachypodium ortholog was upregulated following spikes inoculation with Fusarium graminearum (Fg) (96 hpi).


Maize—The maize ortholog was upregulated in the roots in response to Fg at early infection stages (72 hpi).


QTL—The maize ortholog mapped to a QTL for Fusarium ear rot resistance


LFS121

Foxtail millet (Setaria italica)—The gene was upregulated in the roots at early stages of Fusarium verticillioides (Fv) and Fusarium graminearum (Fg) infection (6 and 24 hpi).



Aegilops—The Aegilops ortholog was upregulated in the roots at early infection stages of Fg and Fv infection (6 and 24 hpi).



Sorghum—The Sorghum ortholog was upregulated in the roots in response to Fv at early infection stages (6 and 24 hpi).


Wheat—The wheat ortholog was upregulated in the roots of resistant genotypes in response to Fv at early infection stages (6 hpi) and in the inflorescence in response to Fg at early infection stages (30 and 50 hpi).


Maize—The maize ortholog was upregulated in the leaves in response to Colletotrichum graminicola (Cg) at early infection stages (120 hpi).


QTL—The maize ortholog mapped to a QTL for Fusarium ear rot resistance.


LFS122

Foxtail millet (Setaria italica)—The gene was upregulated in the roots of several genotypes in response to Fusarium verticillioides (Fv) and Fusarium graminearum (Fg) at early infection stages (6 and 24 hpi).



Aegilops—The Aegilops ortholog was upregulated in the roots in response to Fv at early infection stage (6 and 24 hpi)


LFS123

Foxtail millet (Setaria italica)—The gene was upregulated in the roots at early stages of Fusarium graminearum (Fg) infection (24 and 48 hpi).


LFS124

Foxtail millet (Setaria italica)—The gene was upregulated in the roots at early infection stages of both Fusarium verticillioides (Fv) and Fusarium graminearum (Fg) infections (6 and 24 hpi).



Aegilops—The Aegilops ortholog was upregulated in the roots in response to Fv at early infection stage (6 and 24 hpi). In addition, an upregulation in several genotypes was observed in the roots, in response to Fg at early infection stages (24 hpi).


Wheat—The wheat ortholog was upregulated in the roots in response to Fv at early infection stages (24 and 72 hpi) and in the inflorescence in response to Fg at early infection stages (50 hpi).


Barley—The barley ortholog was upregulated at both early and late stages following inflorescence infection with Fg or challenging assays with the mycotoxin Deoxynivalenol (DON) (1, 2, 3, 4 and 6 dpi).


Maize—The maize ortholog was upregulated in the inflorescence in response to Fv at early infection stage (72 hpi).



Sorghum—The Sorghum ortholog was upregulated in the roots in response to Fv at early infection stages (6 and 24 hpi).


QTL—The rice ortholog mapped to a QTL for blast disease resistance. The Sorghum ortholog mapped to a QTL for rust resistance.


LFS125

Foxtail millet (Setaria italica)—The foxtail millet gene was upregulated in the roots in response to Fusarium verticillioides (Fv) and Fusarium graminearum (Fg) at early infection stages (24 hpi).



Aegilops—The Aegilops ortholog was upregulated in the roots in response to Fv at early infection stage (6 and 24 hpi).


Maize—The maize ortholog was upregulated in the inflorescence of resistant genotype in response to Fv at early infection stages (96 hpi). In addition, the ortholog was upregulated in the leaves of resistant genotype in response to Colletotrichum graminicola (Cg) at early infection stage (48 and 120 hpi).



Sorghum—The Sorghum ortholog was upregulated in the roots in response to Fv at early infection stages (6 hpi).


QTL—The maize ortholog mapped to a QTL for fumonisin contamination resistance.


LFS126

Foxtail millet (Setaria italica)—The gene was upregulated in the roots at early stages of Fusarium verticillioides (Fv) and Fusarium graminearum (Fg) infection (24 and 48 hpi).



Sorghum—The Sorghum ortholog was upregulated in the roots in response to Fusarium verticillioides (Fv) at early infection stages (72 hpi).


QTL—The Sorghum ortholog mapped to a QTL for anthracnose resistance.


LFS127

Foxtail millet (Setaria italica)—The gene was upregulated in the roots at early stages of Fusarium graminearum (Fg) infection (24 and 48—hpi).



Brachypodium—The Brachypodium ortholog was upregulated in the roots at early stages of Fg and Fusarium verticillioides (Fv) infection (48 hpi).



Aegilops—The Aegilops ortholog was upregulated in the roots of several genotypes at early stages of Fg and Fv infection (24 hpi).



Sorghum—The Sorghum ortholog was upregulated in the roots in response to Fv at late infection stages (5 dpi).


Maize—The maize ortholog was upregulated in the leaves of a resistant genotype in response to Colletotrichum graminicola (Cg) at early infection stage (120 hpi).


Wheat—The wheat ortholog was upregulated in the roots in response to Fv at early and late infection stages (6, 24 and hpi, 5 and 10 dpi). QTL—The rice ortholog mapped to a QTL for blast disease resistance. The sorghum ortholog mapped to QTLs for anthracnose resistance and for rust resistance.


LFS128

Foxtail millet (Setaria italica)—The gene was upregulated in the roots at early infection stages of Fusarium graminearum (Fg) infection (24 and 48 hpi).



Aegilops—The Aegilops ortholog was moderately upregulated in the roots in response to Fusarium verticillioides (Fv) at early infection stage (6 hpi).



Sorghum—The Sorghum ortholog was upregulated in the roots in response to Fv at early infection stages (2.5 hpi)


LFS129


Aegilops—The Aegilops ortholog was upregulated in the roots in response to Fusarium verticillioides (Fv) at early infection stage (6 and 24 hpi).


Wheat—The wheat ortholog was upregulated in the inflorescence in response to Fg at early infection stages (30 and 50 hpi).


Barley—The barley ortholog was upregulated at both early and late stages following inflorescence infection with Fg (72, 96 and 144 hpi).



Brachypodium—The Brachypodium ortholog was upregulated in the roots of several genotypes in response to Fg at early infection stage (48 hpi).


QTL—The rice ortholog mapped to a QTL for sheath blight disease resistance.


LFS130


Aegilops—Computational evidences indicate up-regulation of the gene mainly at early infection stages of Fusarium verticillioides (Fv) and Fusarium graminearum (Fg) in roots.


LFS131

Foxtail mille (Setaria italica)—The gene was upregulated in the roots and stem in response to Fusarium verticillioides (Fv) at early infection stages.


LFS132

Maize—Computational evidences indicate upregulation of the gene in roots of sensitive lines at early infection stages of Fusarium verticillioides (Fv).


LFS82

Maize—Computational evidences indicate upregulation of the gene mainly at early infection stages of Fusarium verticillioides (Fv) and Fusarium graminearum (Fg) in different organs


Wheat—The wheat ortholog was upregulated in the roots and stem in response to Fv at early infection stages.


LFS83

Wheat—Computational evidences indicate upregulation of the gene mainly at early infection stages of Fusarium verticillioides (Fv) and Fusarium graminearum (Fg) in different organs. Aegilops and Maize—The gene orthologs were upregulated in the roots and stem in response to Fusarium verticillioides (Fv) at early infection stages.


LFS84

Wheat—Computational evidences indicate upregulation of the gene and its Aegilops ortholog mainly at early infection stages of Fusarium verticillioides (Fv) and Fusarium graminearum (Fg) in different organs


Maize—The gene was up regulated as a result of Colletotrichum graminicola (Cg) infection in stem tissue.


LFS86

Maize—Computational evidences indicate upregulation of the gene and its Sorghum ortholog mainly at early infection stages of Fusarium verticillioides (Fv) and Fusarium graminearum (Fg) in different organs.


LFS87

Maize—Computational evidences indicate upregulation of the gene and its wheat ortholog mainly at early infection stages of Fusarium verticillioides (Fv) and Fusarium graminearum (Fg) in different organs


LFS89

Wheat—Computational evidences indicate upregulation of the gene at late infection stages of Fusarium graminearum (Fg) in heads and its Aegilops ortholog mainly at early infection stages of Fusarium verticilloides (Fv) and Fusarium graminearum (Fg) roots.


Maize—Computational evidences indicate a positive correlation of the gene expression to the base diameter, stalk width.


The plant genes listed in Table 6 below were identified as candidates to have a major impact on plant resistance to at least one of Fusarium verticilloides, Fusarium graminearum and Colletotrichum graminicola when expression thereof is modulated in plants. The identified gene name, the plant from which it is derived, and the amino acid and nucleic acid sequences of each gene are summarized in Table 6, hereinbelow.









TABLE 6







Genes associated with plant resistance to fungal infection














Polyn.
Polyp.





SEQ ID
SEQ ID



Gene Name
Organism
NO:
NO:
















LFS82

Zea mays

22
308



LFS83

Triticum aestivum

23
309



LFS84

Triticum aestivum

24
310



LFS86

Zea mays

25
311



LFS87

Zea mays

26
312



LFS89

Triticum aestivum

27
313



LFS90

Aegilops tauschii

28
314



LFS91

Aegilops tauschii

29
315



LFS92

Aegilops tauschii

30
316



LFS93

Aegilops tauschii

31
317



LFS94

Aegilops tauschii

32
318



LFS95

Aegilops tauschii

33
319



LFS96

Aegilops tauschii

34
320



LFS97

Aegilops tauschii

35
321



LFS98

Aegilops tauschii

36
322



LFS99

Aegilops tauschii

37
323



LFS100

Aegilops tauschii

38
324



LFS102

Aegilops tauschii

39
325



LFS104

Aegilops tauschii

40
326



LFS105

Aegilops tauschii

302
510



LFS106

Aegilops tauschii

41
327



LFS107

Aegilops tauschii

42
328



LFS109

Aegilops tauschii

43
329



LFS110

Aegilops tauschii

44
330



LFS111

Aegilops tauschii

45
331



LFS112

Aegilops tauschii

303
507



LFS113

Aegilops tauschii

46
332



LFS114

Aegilops tauschii

47
333



LFS115

Aegilops tauschii

48
334



LFS116

Aegilops tauschii

49
335



LFS117

Aegilops tauschii

50
336



LFS121

Setaria italica

51
337



LFS122

Setaria italica

52
338



LFS123

Setaria italica

53
339



LFS124

Setaria italica

54
340



LFS125

Setaria italica

55
341



LFS126

Setaria italica

56
342



LFS127

Setaria italica

57
343



LFS128

Setaria italica

58
344



LFS129

Setaria italica

59
345



LFS130

Aegilops tauschii

60
346



LFS131

Setaria italica

61
347



LFS132

Zea mays

62
348







“polyn.” = polynucleotide;



“polyp.” = polypeptide.






Example 3: Identification of Homologous Sequences Associated with Fungal Resistance

The concepts of orthology and paralogy have recently been applied to functional characterizations and classifications on the scale of whole-genome comparisons. Orthologs and paralogs constitute two major types of homologs: The first evolved from a common ancestor by specialization, and the latter are related by duplication events. It is assumed that paralogs arising from ancient duplication events are likely to have diverged in function while true orthologs are more likely to retain identical function over evolutionary time.


To further investigate and identify putative orthologs of the genes identified to affect plant resistance to Fusarium verticilloides, Fusarium graminearum or Colletotrichum graminicola, all sequences were aligned using the BLAST™ (Basic Local Alignment Search Tool). Sequences sufficiently similar were tentatively grouped. These putative orthologs were further organized under a Phylogram—a branching diagram (tree) assumed to be a representation of the evolutionary relationships among the biological taxa. Putative ortholog groups were analyzed as to their agreement with the phylogram and in cases of disagreements these ortholog groups were broken accordingly.


Expression data were analyzed and the EST libraries were classified using a fixed vocabulary of custom terms such as developmental stages (e.g., genes showing similar expression profile through development with up-regulation at specific stage, such as at seed germination, time points during seedling growth and at maturity) and/or plant organ (e.g., genes showing similar expression profile across their organs with up-regulation at specific organs such as roots and stems). The annotations from all the ESTs clustered to a gene were analyzed statistically by comparing their frequency in the cluster versus their abundance in the database, allowing the construction of a numeric and graphic expression profile of that gene, which is termed “digital expression”. The rationale of using these two complementary methods with methods of phenotypic association studies of QTLs, SNPs and phenotype expression correlation is based on the assumption that true orthologs are likely to retain identical function over evolutionary time. These methods provide different sets of indications on function similarities between two homologous genes, including similarities in the sequence level—identical amino acids in the protein domains and similarity in expression profiles.


The search and identification of homologous genes involves the screening of sequence information available, for example, in public databases such as the DNA Database of Japan (DDBJ), GenBank, and the European Molecular Biology Laboratory Nucleic Acid Sequence Database (EMBL) or versions thereof or the MIPS database. A number of different search algorithms have been developed, including but not limited to the suite of programs referred to as BLAST™ programs. There are five implementations of BLAST™, three designed for nucleotide sequence queries (BLASTN, BLASTX, and TBLASTX) and two designed for protein sequence queries (BLASTP and TBLASTN) (Altschul, S. F et al., (1990) “Basic local alignment search tool.” J. Mol. Biol. 215:403-410). Such methods involve alignment and comparison of sequences. The BLAST™ algorithm calculates percent sequence identity and performs a statistical analysis of the similarity between the two sequences. The software for performing BLAST™ analysis is publicly available through the National Centre for Biotechnology Information. Other such software or algorithms are GAP, BESTFIT, FASTA and TFASTA. 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.


The homologous genes may belong to the same gene family. The analysis of a gene family may be carried out using sequence similarity analysis. To perform this analysis, one may use standard programs for multiple alignments e.g. Clustal W. A neighbor-joining tree of the proteins homologous to the proteins encoded by the genes identified in this invention may be used to provide an overview of structural and ancestral relationships. Sequence identity may be calculated using an alignment program as described above. It is expected that plants other that those examined in the present invention will carry a similar functional gene (ortholog) or a family of similar genes and those genes will provide the same preferred phenotype as the genes presented here. Advantageously, these family members may be useful in the methods of the invention. Example of other plants are included here but not limited to, barley (Hordeum vulgare), Arabidopsis (Arabidopsis thaliana), maize (Zea mays), cotton (Gossypium), Oilseed rape (Brassica napus), Rice (Oryza sativa), Sugar cane (Saccharum officinarum), Sorghum (Sorghum bicolor), Soybean (Glycine max), Sunflower (Helianthus annuus), Tomato (Lycopersicon esculentum), and Wheat (Triticum aestivum).


The above-mentioned analyses for sequence homology can be carried out on a full-length sequence, but may also be based on a comparison of certain regions such as conserved domains. The identification of such domains would also be well within the realm of the person skilled in the art and would involve, for example, a computer readable format of the nucleic acids of the present invention, the use of alignment software programs and the use of publicly available information on protein domains, conserved motifs and boxes. This information is available in the PRODOM (biochem.ucl.ac.uk/bsm/dbbrowser/protocol/prodomqry.html), PIR (pir.Georgetown.edu/) or Pfam (sanger.ac.uk/Software/Pfam/) databases. Sequence analysis programs designed for motif searching may be used for identification of fragments, regions and conserved domains as mentioned above. Preferred computer programs include, but are not limited to, MEME, SIGNALSCAN, and GENESCAN.


A person skilled in the art may use the homologous sequences provided herein to find similar sequences in other species and other organisms. Homologues of a protein encompass peptides, oligopeptides, polypeptides, proteins and enzymes having amino acid substitutions, deletions and/or insertions relative to the unmodified protein in question and having similar biological and functional activity as the unmodified protein from which they are derived. To produce such homologues, amino acids of the protein may be replaced by other amino acids having similar properties (conservative changes, such as similar hydrophobicity, hydrophilicity, antigenicity, propensity to form or break α-helical structures or β-sheet structures). Conservative substitution Tables are well known in the art (see for example Creighton T E (1984) Proteins. W.H. Freeman and Company). Homologues of a nucleic acid encompass nucleic acids having nucleotide substitutions, deletions and/or insertions relative to the unmodified nucleic acid in question and having similar biological and functional activity as the unmodified nucleic acid from which they are derived.


Polynucleotides and polypeptides with significant homology to the identified genes described in Table 6 (Example 2) were identified from the databases using BLAST™ Software with the Blastp and tBlastn algorithms as filters for the first stage, and the needle (EMBOSS package) or Frame+algorithm alignment for the second stage. Local identity (BLAST™ alignments) was defined with a very permissive cutoff—60% Identity on a span of 60% of the sequences lengths because it is used only as a filter for the global alignment stage. The default filtering of the BLAST™ package was not utilized (by setting the parameter “−F F”).


In the second stage, homologs were defined based on a global identity of at least 80% to the core gene polypeptide sequence. Two distinct forms for finding the optimal global alignment for protein or nucleotide sequences were used in this application:


1.Between Two Proteins (Following the BLASTP Filter):


EMBOSS-6.0.1 Needleman-Wunsch algorithm with the following modified parameters: gapopen=8 gapextend=2. The rest of the parameters were unchanged from the default options described hereinabove.


2.Between a Protein Sequence and a Nucleotide Sequence (Following the TBLASTN Filter):


GenCore 6.0 OneModel application utilizing the Frame+algorithm with the following parameters: model=frame+p2n.model mode=qglobal -q=protein.sequence -db=nucleotide. sequence. The rest of the parameters are unchanged from the default options described hereinabove.


The query polypeptide sequences were the sequences listed in Table 6 (Example 2). The subject sequences are protein sequences identified in the database based on greater than 80% global identity to the predicted translated sequences of the query nucleotide sequences or to the polypeptide sequences. Homology was calculated as 00 of identity over the aligned sequences. The identified orthologous and homologous sequences having at least 80% global sequence identity to said sequences are provided in Table 7, below. These homologous genes are expected to increase plant resistance to fungal infection caused by the mentioned pathogens









TABLE 7







Homologues (e.g., orthologues) of genes associated


with plant resistance to fungal infection













Polyn.
Hom. to

Polyp.
Hom. to
%



SEQ ID
Gene

SEQ ID
SEQ ID
glob.



NO:
Name
Organism
NO:
NO:
Iden.
Algor.
















100
LFS122

Panicum virgatum


338
90.76
glotblastn


101
LFS122

Panicum virgatum

369
338
84.3
globlastp


102
LFS109

Hordeum vulgare

370
329
81.2
globlastp


103
LFS86

Sorghum bicolor

371
311
80.3
globlastp


104
LFS132

Sorghum bicolor


348
80.33
glotblastn


105
LFS107

Triticum aestivum

372
328
90.6
globlastp


106
LFS116

Triticum aestivum

373
335
99.5
globlastp


107
LFS116

Hordeum vulgare

374
335
90.9
globlastp


108
LFS121

Panicum virgatum

375
337
85
globlastp


109
LFS121

Panicum virgatum

376
337
84.3
globlastp


110
LFS121

Zea mays

377
337
80.9
globlastp


111
LFS92

Triticum aestivum

378
316
94.6
globlastp


112
LFS92

Secale cereale

379
316
87.5
globlastp


113
LFS95

Aegilops tauschii

380
354
88.7
globlastp


114
LFS95

Triticum aestivum

381
354
88.3
globlastp


115
LFS95

Secale cereale

382
354
86
globlastp


116
LFS95

Secale cereale

383
354
84.8
globlastp


117
LFS95

Secale cereale

384
354
84.3
globlastp


118
LFS95

Secale cereale


354
83.88
glotblastn


119
LFS95

Secale cereale


354
83.47
glotblastn


120
LFS95

Triticum aestivum

385
354
83.1
globlastp


121
LFS95

Triticum aestivum

386
354
80.2
globlastp


122
LFS106

Secale cereale

387
327
89.2
globlastp


123
LFS106

Triticum aestivum

388
327
85.5
globlastp


124
LFS106

Hordeum vulgare

389
327
83.1
globlastp


125
LFS111

Triticum aestivum


331
96.71
glotblastn


126
LFS111

Hordeum vulgare

390
331
88.6
globlastp


127
LFS97

Triticum aestivum

391
321
99.8
globlastp


128
LFS97

Triticum aestivum

392
321
99
globlastp


129
LFS97

Triticum aestivum

393
321
99
globlastp


130
LFS97

Hordeum vulgare

394
321
98.1
globlastp


131
LFS97

Secale cereale

395
321
97.7
globlastp


132
LFS97

Brachypodium distachyon

396
321
94.7
globlastp


133
LFS97

Avena sativa

397
321
94.7
globlastp


134
LFS97

Brachypodium distachyon

398
321
93
globlastp


135
LFS97

Oryza sativa

399
321
89.1
globlastp


136
LFS97

Panicum virgatum

400
321
84.4
globlastp


137
LFS97

Setaria italica

401
321
83.6
globlastp


138
LFS97

Aegilops tauschii


321
82.56
glotblastn


139
LFS97

Sorghum bicolor

402
321
81.2
globlastp


140
LFS98

Triticum aestivum

403
357
97.2
globlastp


141
LFS113

Triticum aestivum

404
332
98.8
globlastp


142
LFS113

Hordeum vulgare

405
332
97.5
globlastp


143
LFS113

Brachypodium distachyon

406
332
84.6
globlastp


144
LFS113

Oryza sativa

407
332
82.7
globlastp


145
LFS113

Oryza sativa


332
81.53
glotblastn


146
LFS113

Secale cereale


363
98.25
glotblastn


147
LFS113

Secale cereale

408
363
97.9
globlastp


148
LFS113

Setaria italica

409
363
84
globlastp


149
LFS113

Sorghum bicolor

410
363
83.1
globlastp


150
LFS113

Zea mays

411
363
82.2
globlastp


151
LFS117

Triticum aestivum


365
96.87
glotblastn


152
LFS100

Triticum aestivum


359
92.49
glotblastn


153
LFS100

Aegilops tauschii


359
89.67
glotblastn


154
LFS100

Secale cereale


359
80.47
glotblastn


155
LFS94

Secale cereale


318
94.7
glotblastn


156
LFS94

Secale cereale


318
93.32
glotblastn


157
LFS94

Triticum aestivum


318
93.09
glotblastn


158
LFS94

Hordeum vulgare

412
318
92.2
globlastp


159
LFS94

Secale cereale

413
318
91.2
globlastp


160
LFS94

Aegilops tauschii

414
318
90.1
globlastp


161
LFS94

Brachypodium distachyon

415
318
86.3
globlastp


162
LFS94

Brachypodium distachyon

416
318
85.8
globlastp


163
LFS94

Oryza sativa

417
318
83.3
globlastp


164
LFS94

Secale cereale

418
318
83.1
globlastp


165
LFS94

Zea mays

419
318
80
globlastp


166
LFS99

Triticum aestivum


358
80
glotblastn


167
LFS95

Hordeum vulgare

420
319
89
globlastp


168
LFS95

Aegilops tauschii

421
319
87.9
globlastp


169
LFS96

Triticum aestivum

422
320
98.1
globlastp


170
LFS96

Aegilops tauschii


320
86.79
glotblastn


171
LFS130

Triticum aestivum

423
346
98.5
globlastp


172
LFS130

Hordeum vulgare

424
346
94.8
globlastp


173
LFS130

Brachypodium distachyon

425
346
83.1
globlastp


174
LFS82

Panicum virgatum

426
349
94.7
globlastp


175
LFS82

Panicum virgatum

427
349
93.9
globlastp


176
LFS82

Oryza sativa

428
349
88.6
globlastp


177
LFS82

Brachypodium distachyon

429
349
87.1
globlastp


178
LFS82

Triticum aestivum

430
349
86.4
globlastp


179
LFS82

Secale cereale

431
349
86.2
globlastp


180
LFS82

Hordeum vulgare

432
349
86.1
globlastp


306
LFS112

Triticum aestivum

509
507
99.2
globlastp


305
LFS112

Hordeum vulgare

508
507
95.6
globlastp


181
LFS129

Panicum virgatum

433
345
92
globlastp


182
LFS129

Panicum virgatum

434
345
80.5
globlastp


183
LFS94

Triticum aestivum

435
353
99.8
globlastp


184
LFS94

Secale cereale

436
353
96.8
globlastp


185
LFS94

Secale cereale

437
353
90.8
globlastp


186
LFS94

Setaria italica

438
353
80.7
globlastp


187
LFS89

Aegilops tauschii

313
313
100
globlastp


188
LFS84

Hordeum vulgare

439
310
94.3
globlastp


189
LFS84

Secale cereale

440
310
90
globlastp


190
LFS84

Aegilops tauschii

441
310
89.4
globlastp


191
LFS84

Brachypodium distachyon

442
310
87.4
globlastp


192
LFS84

Panicum virgatum

443
310
85.8
globlastp


193
LFS84

Sorghum bicolor

444
310
85.3
globlastp


194
LFS84

Setaria italica

445
310
84.9
globlastp


195
LFS84

Zea mays


310
83.65
glotblastn


196
LFS84

Zea mays

446
310
83.2
globlastp


197
LFS84

Oryza sativa


310
80.27
glotblastn


198
LFS125

Zea mays

447
341
83.8
globlastp


199
LFS115

Secale cereale

448
334
99.8
globlastp


200
LFS115

Triticum aestivum

449
334
92.7
globlastp


201
LFS114

Hordeum vulgare

450
364
92.3
globlastp


202
LFS114

Secale cereale

451
364
91.4
globlastp


203
LFS114

Triticum aestivum

452
364
80.8
globlastp


204
LFS93

Hordeum vulgare

453
317
92.8
globlastp


205
LFS93

Aegilops tauschii


317
89.24
glotblastn


206
LFS93

Triticum aestivum


317
84.39
glotblastn


207
LFS93

Brachypodium distachyon


317
80.38
glotblastn


208
LFS126

Panicum virgaium

454
342
91.2
globlastp


209
LFS126

Panicum virgatum

455
342
91.2
globlastp


210
LFS126

Zea mays

456
342
88
globlastp


211
LFS126

Sorghum bicolor

457
342
88
globlastp


212
LFS126

Oryza sativa

458
342
80.5
globlastp


213
LFS100

Triticum aestivum

459
324
81.9
globlastp


214
LFS83

Triticum aestivum

460
350
99.7
globlastp


215
LFS83

Secale cereale

461
350
95.4
globlastp


216
LFS83

Lolium multiflorum

462
350
87.5
globlastp


217
LFS83

Lolium multiflorum

463
350
87.5
globlastp


218
LFS83

Brachypodium distachyon

464
350
83.7
globlastp


219
LFS83

Setaria italica

465
350
80.5
globlastp


220
LFS131

Setaria italica

466
347
83.2
globlastp


221
LFS131

Setaria italica

467
347
81.2
globlastp


222
LFS131

Sorghum bicolor

468
347
81.2
globlastp


223
LFS91

Triticum aestivum

315
315
100
globlastp


224
LFS91

Triticum aestivum

469
315
93.1
globlastp


225
LFS91

Secale cereale


315
91.16
glotblastn


226
LFS91

Secale cereale


315
89.23
glotblastn


227
LFS91

Secale cereale

470
315
81.4
globlastp


228
LFS91

Brachypodium distachyon

471
315
80.7
globlastp


229
LFS83

Secale cereale

472
309
95.6
globlastp


230
LFS83

Secale cereale


309
82.07
glotblastn


231
LFS83

Aegilops tauschii

473
309
82
globlastp


232
LFS83

Sorghum bicolor

474
309
80.8
globlastp


233
LFS83

Triticum aestivum

475
309
80.5
globlastp


234
LFS102

Triticum aestivum

476
325
96.9
globlastp


235
LFS102

Hordeum vulgare

477
325
90.2
globlastp


236
LFS102

Aegilops tauschii


325
87.5
glotblastn


237
LFS104

Hordeum vulgare

478
326
97.8
globlastp


238
LFS104

Secale cereale

479
326
83.1
globlastp


239
LFS104

Zea mays


326
81.37
glotblastn


240
LFS99

Avena sativa


323
93
glotblastn


241
LFS99

Triticum aestivum


323
87
glotblastn


242
LFS99

Triticum aestivum

480
323
86
globlastp


243
LFS99

Triticum aestivum


323
82
glotblastn


244
LFS90

Triticum aestivum

481
314
99.8
globlastp


245
LFS90

Triticum aestivum

482
314
97.2
globlastp


246
LFS90

Triticum aestivum

483
314
95.5
globlastp


247
LFS90

Triticum aestivum

484
314
93.9
globlastp


248
LFS90

Triticum aestivum

485
314
93.7
globlastp


249
LFS82

Sorghum bicolor


308
97.24
glotblastn


250
LFS87

Zea mays

486
312
96.9
globlastp


251
LFS87

Zea mays

486
312
96.9
globlastp


252
LFS87

Zea mays


312
95.92
glotblastn


253
LFS87

Sorghum bicolor


312
95.92
glotblastn


254
LFS87

Saccharum officinarum


312
95.92
glotblastn


255
LFS87

Cenchrus ciliaris


312
94.9
glotblastn


256
LFS87

Setaria italica


312
94.9
glotblastn


257
LFS87

Zea mays


312
94.9
glotblastn


258
LFS87

Panicum virgatum


312
94.9
glotblastn


259
LFS87

Panicum virgatum


312
94.9
glotblastn


260
LFS87

Zea mays


312
93.88
glotblastn


261
LFS87

Pennisetum glaucum


312
93.88
glotblastn


262
LFS87

Echinochloa colona


312
91.84
glotblastn


263
LFS87

Zea mays


312
89.8
glotblastn


264
LFS87

Oryza sativa


312
88.78
glotblastn


265
LFS87

Oryza sativa


312
87.76
glotblastn


266
LFS87

Hordeum vulgare


312
86.73
glotblastn


267
LFS87

Aegilops tauschii


312
85.71
glotblastn


268
LFS87

Secale cereale


312
85.71
glotblastn


269
LFS87

Secale cereale


312
85.71
glotblastn


270
LFS87

Triticum aestivum


312
85.71
glotblastn


271
LFS87

Triticum aestivum


312
85.71
glotblastn


272
LFS87

Brachypodium distachyon


312
84.69
glotblastn


273
LFS87

Cocos nucifera


312
82.65
glotblastn


274
LFS87

Phalaenopsis aphrodite


312
82.65
glotblastn


275
LFS87

Cocos nucifera


312
81.63
glotblastn


276
LFS87

Cynodon dactylon


312
81.63
glotblastn


277
LFS87

Elaeis guineensis


312
81.63
glotblastn


278
LFS87

Lolium multiflorum


312
80.61
glotblastn


279
LFS87

Avena sativa


312
80.61
glotblastn


280
LFS87

Elaeis guineensis


312
80.61
glotblastn


281
LFS104

Brachypodium distachyon

487
360
92.6
globlastp


282
LFS104

Setaria italica

488
360
86.7
globlastp


283
LFS104

Sorghum bicolor

489
360
85.6
globlastp


284
LFS104

Zea mays

490
360
83.8
globlastp


285
LFS104

Oryza sativa

491
360
80.9
globlastp


286
LFS127

Panicum virgatum

492
343
86.2
globlastp


287
LFS127

Sorghum bicolor

493
343
85.2
globlastp


288
LFS127

Panicum virgatum

494
343
82.1
globlastp


289
LFS127

Zea mays

495
343
81.5
globlastp


290
LFS125

Panicum virgatum

496
367
92.3
globlastp


291
LFS125

Panicum virgatum

497
367
91.6
globlastp


292
LFS125

Zea mays

498
367
85.5
globlastp


293
LFS125

Sorghum bicolor

499
367
84.7
globlastp


294
LFS125

Oryza sativa

500
367
82.1
globlastp


295
LFS117

Hordeum vulgare

501
336
94.3
globlastp


296
LFS117

Secale cereale


336
80.97
glotblastn


297
LFS124

Setaria italica

502
340
90.1
globlastp


298
LFS124

Sorghum bicolor

503
340
85.7
globlastp


299
LFS124

Zea mays

504
340
83.3
globlastp


300
LFS124

Panicum virgatum

505
340
82.3
globlastp


301
LFS124

Panicum virgatum

506
340
81.8
globlastp





“Polyn.” = polynucleotide; “Polyp.” = polypeptide; “Algor.” = algorithm (used for sequence alignment and determination of percent homology); “Hom.”—homology; “iden.”—identity; “glob.”—global.







The output of the functional genomics approach described herein is a set of genes highly predicted to improve resistance of a plant to fungal infection by Fusarium verticilloides, Fusarium graminearum or Colletotrichum graminicola when their expression is modulated (enhanced or down-regulated) in said plant. Although each gene is predicted to have its own impact, modifying the mode of expression of more than one gene is expected to provide an additive or synergistic effect on the plant resistance Altering the expression of each gene described herein alone or of a set of genes together increases the overall plant resistance, hence expects to increase agricultural productivity.


Example 4: Identification of Domains Comprised within Identified Genes

A polypeptide domain refers to a set of conserved amino acids located at specific positions along an alignment of sequences of evolutionarily related proteins. While amino acids at other positions can vary between homologues, amino acids that are highly conserved, and particularly amino acids that are highly conserved at specific positions indicate amino acids that are likely essential in the structure, stability or function of a protein. Identified by their high degree of conservation in aligned sequences of a family of protein homologues, they can be used as identifiers to determine if any polypeptide in question belongs to a previously identified polypeptide family.


The Integrated Resource of Protein Families, Domains and Sites (InterPro) database is an integrated interface for the commonly used signature databases for text- and sequence-based searches. The InterPro database combines these databases, which use different methodologies and varying degrees of biological information about well-characterized proteins to derive protein signatures. Collaborating databases include SWISS-PROT, PROSITE, TrEMBL, PRINTS, ProDom and Pfam, Smart and TIGRFAMs. Pfam is a large collection of multiple sequence alignments and hidden Markov models covering many common protein domains and families. Pfam is hosted at the Sanger Institute server in the United Kingdom.


Interpro is hosted at the European Bioinformatics Institute in the United Kingdom. InterProScan is the software package that allows sequences (protein and nucleic) to be scanned against InterPro's signatures. Signatures are predictive models, provided by several different databases that make up the InterPro consortium.


InterProScan 5.32-71.0 was used to analyze the polypeptides of the present invention (core and homologues/orthologs) for common domains (Jones P et al., 2014. Bioinformatics, January 2014 (doi:10.1093/bioinformatics/btu031). Briefly, InterProScan is based on scanning methods native to the InterPro member databases. It is distributed with pre-configured method cut-offs recommended by the member database experts and which are believed to report relevant matches. All cut-offs are defined in configuration files of the InterProScan programs. Matches obtained with the fixed cut-off are subject to the following filtering:


Pfam filtering: Each Pfam family is represented by two hidden Markov models (HMMs)—ls and fs (full-length and fragment). An HMM model has bit score cut-offs (for each domain match and the total model match) and these are defined in the Gathering threshold (GA) lines of the Pfam database. Initial results are obtained with quite a high common cut-off and then the matches of the signature with a lower score than the family specific cut-offs are dropped.


If both the fs and ls model for a particular Pfam hits the same region of a sequence, the AM field in the Pfam database is used to determine which model should be chosen—globalfirst(LS); localfirst(FS) or byscore (whichever has the highest e-value).


Another type of filtering has been implemented since release of version 4.1. It is based on Clan filtering and nested domains. Further information on Clan filtering can be found in the Pfam website (www.sanger.ac.uk/Pfam) for more information on Clan filtering.


TIGRFAMs filtering: Each TIGRFAM HMM model has its own cut-off scores for each domain match and the total model match. These bit score cut-offs are defined in the “trusted cut-offs” (TC) lines of the database. Initial results are obtained with quite a high common cut-off and then the matches (of the signature or some of its domains) with a lower score compared to the family specific cut-offs are dropped.


PRINTS filtering: All matches with p-value more than a pre-set minimum value for the signature, are dropped.


SMART filtering: The publicly distributed version of InterProScan has a common e-value cut-off corresponding to the reference database size. A more sophisticated scoring model is used on the SMART web server and in the production of pre-calculated InterPro Match data.


Exact scoring thresholds for domain assignments are proprietary data. The InterProMatches data production procedure uses these additional smart.thresholds data. It is to be noted that the given cut-offs are e-values (i.e. the number of expected random hits) and therefore are valid only in the context of reference database size and smart.desc data files to filter out results obtained with higher cut-off.


It implements the following logic: If the whole sequence E-value of a found match is worse than the ‘cut_low’ the match is dropped. If the domain E-value of a found match is worse than the ‘repeat’ cut-off (where defined) the match is dropped. If a signature is a repeat, the number of significant matches of that signature to a sequence must be greater than the value of ‘repeats’ in order for all matches to be accepted as true (T).


If the signature is part of a family (‘family_cut’ is defined), if the domain E-value is worse than the domain cut off (‘cutoff’), the match is dropped. If the signature has “siblings” (because it has a family_cut defined), and they overlap, the preferred sibling is chosen as the true match according to information in the overlaps file.


PROSITE patterns CONFIRMation: ScanRegExp is able to verify PROSITE matches using corresponding statistically-significant CONFIRM patterns. The default status of the PROSITE matches is unknown (?) and the true positive (T) status is assigned if the corresponding CONFIRM patterns match as well. The CONFIRM patterns were generated based on the true positive SWISS-PROT PROSITE matches using eMOTIF software with a stringency of 10e-9 P-value.


PANTHER filtering: Panther has pre- and post-processing steps. The pre-processing step is intended to speed up the HMM-based searching of the sequence and involves blasting the HMM sequences with the query protein sequence in order to find the most similar models above a given e-value. The resulting HMM hits are then used in the HMM-based search.


Panther consists of families and sub-families. When a sequence is found to match a family in the blast run, the sub-families are also scored using HMMER tool (that is, unless there is only 1 sub-family, in which case, the family alone is scored against).


Any matches that score below the e-value cut-off are discarded. Any remaining matches are searched to find the HMM with the best score and e-value and the best hit is then reported (including any sub-family hit).


GENE3D filtering: Gene3D also employs post-processing of results by using a program called DomainFinder. This program takes the output from searching the Gene3D HMMs against the query sequence and extracts all hits that are more than 10 residues long and have an e-value better than 0.001. If hits overlap at all, the match with the better e-value is chosen.


The polypeptides of the invention associated with conferring and/or enhancing the resistance of a plant to at least one fungal pathogen can be characterized by specific amino acid domains. According to certain embodiments, particular domains are conserved within a family of polypeptides as described in Table 8 hereinbelow. Without wishing to be bound by any specific theory or mechanism of action, the conserved domain(s) may indicate common functionally of the polypeptides comprising same. The domains are presented by an identifier (number). Table 9 provides the details of each domain.









TABLE 8







Core and homologous polypeptides comprising the same domains










Core
Gene

Homologous Polypeptides


Polypeptide
Name
Characteristic Domains
Comprising the Domains


(SEQ ID NO)
(Core)
(Domain identifier)
(SEQ ID NOs)





308
LFS82
1; 2; 3; 4; 5; 6; 7; 8; 9; 10 in core,
426; 427; 428; 429; 430; 431; 432




cloned and homologs



309
LFS83
11; 12; 13; 14; 15; 16 in core, cloned
460; 461; 462; 463; 464; 465; 472; 473;




and homologs
474; 475


310
LFS84
2; 17; 18; 5 in core, cloned and
439; 440; 441; 442; 443; 444; 445; 446




homologs



311
LFS86
19 in core, cloned and homologs
371


312
LFS87
20; 21 in core and homologs
486


313
LFS89
22; 23; 24; 25; 26; 27 in core, cloned
313




and homologs



314
LFS90
28; 29; 30 in core, cloned and
481; 482; 483; 484; 485




homologs



315
LFS91
31; 32 in core, cloned and
315; 469; 470; 471




homologs



316
LFS92
23; 24; 33 in core, cloned and
378; 379




homologs



317
LFS93
34 in core, cloned and homologs
453


318
LFS94
35; 36 in core, cloned and
412; 413; 414; 415; 416; 417; 418; 419;




homologs
435; 436; 437; 438


319
LFS95
37 in core, cloned and homologs
380; 381; 382; 383; 384; 385; 386; 420;





421


320
LFS96
38; 39 in core, cloned and
422




homologs



321
LFS97
40; 41; 42; 43 in core, cloned and
391; 392; 393; 394; 395; 396; 397; 398;




homologs
399; 400; 401; 402


322
LFS98
44; 45; 32; 46 in core, cloned and
403




homologs



323
LFS99
no domains in core



324
LFS100
47; 48 in core, cloned and
459




homologs



325
LFS102
49; 50 in core, cloned and
476; 477




homologs



326
LFS104
51; 52; 53; 54; 55; 56; 57 in core,
478; 479; 487; 488; 489; 490; 491




cloned and homologs



327
LFS106
no domains in core



328
LFS107
35; 36 in core, cloned and
372




homologs



329
LFS109
58; 59; 60; 21; 61; 20; 62 in core,
370




cloned and homologs



330
LFS110
63 in core and cloned



331
LFS111
34 in core, cloned and homologs
390


332
LFS113
65; 66; 67 in core, cloned and
404; 405; 406; 407; 408; 409; 410; 411




homologs



333
LFS114
68; 69 in core, cloned and
450; 451; 452




homologs



334
LFS115
1; 2; 3; 4; 5; 70; 9; 71; 10 in core,
448; 449




cloned and homologs



335
LFS116
20; 72; 21 in core, cloned and
373; 374




homologs



336
LFS117
73; 74; 75 in core, cloned and
501




homologs



337
LFS121
76; 77; 78 in core, cloned and
375; 376; 377




homologs



338
LFS122
79 in core, cloned and homologs
369


339
LFS123
80; 81; 82; 83; 84 in core and cloned



340
LFS124
85; 86; 87; 88 in core, cloned and
502; 503; 504; 505; 506




homologs



341
LFS125
89 in core, cloned and homologs
447; 496; 497; 498; 499; 500


342
LFS126
62; 59; 60 in core, cloned and
454; 455; 456; 457; 458




homologs



343
LFS127
90; 91; 92; 93; 94 in core, cloned and
492; 493; 494; 495




homologs



344
LFS128
95; 96; 97; 98 in core and cloned



345
LFS129
99; 100; 101; 48; 102; 103; 104; 105;
433; 434




106; 107 in core, cloned and





homologs



346
LFS130
108 in core and homologs
423; 424; 425


347
LFS131
109; 110; 111; 112 in core and
466; 467; 468




homologs



348
LFS132
35; 36 in core



507
LFS112
64 in core, cloned and homologs
508; 509


510
LFS105
no domains in core
















TABLE 9







Details of identified domains










Domain
InterPro




Identifier
No.
Accession No.
Description













1
IPR023298
SSF81665
P-type ATPase, transmembrane domain





superfamily


2
IPR036412
SSF56784
HAD-like superfamily


3
IPR023299
G3DSA:3.40.1110.10
P-type ATPase, cytoplasmic domain N


4
IPR008250
SSF81653
P-type ATPase, A domain superfamily


5
IPR023214
G3DSA:3.40.50.1000
HAD superfamily


6
IPR032631
PF16209
Phospholipid-translocating ATPase N-terminal P-





type ATPase, N-terminal


7
IPR032630
PF16212
Phospholipid-translocating P-type ATPase C-





terminal P-type ATPase, C-terminal


8
IPR006539
TIGR01652
ATPase-Plipid: phospholipid-translocating P-type





ATPase, flippase P-type ATPase, subfamily IV


9
IPR001757
TIGR01494
ATPase_P-type: HAD ATPase, P-type, family IC





P-type ATPase


10
IPR018303
PS00154
E1-E2 ATPases phosphorylation site. P-type





ATPase, phosphorylation site


11
IPR000909
PF00388
Phosphatidylinositol-specific phospholipase C, X





domain Phosphatidylinositol-specific





phospholipase C, X domain


12
IPR017946
G3DSA:3.20.20.190
PLC-like phosphodiesterase, TIM beta/alpha-





barrel domain superfamily


13
IPR001192
PR00390
Phospholipase C signature Phosphoinositide





phospholipase C family


14
IPR035892
G3DSA:2.60.40.150
C2 domain superfamily


15
IPR000008
SM00239
C2 domain


16
IPR001711
PF00387
Phosphatidylinositol-specific phospholipase C, Y





domain Phospholipase C, phosphatidylinositol-





specific, Y domain


17
IPR006379
TIGR01484
HAD-SF-IIB: HAD hydrolase, family IIB HAD-





superfamily hydrolase, subfamily IIB


18
IPR003337
PF02358
Trehalose-phosphatase


19
IPR008889
PF05678
VQ motif


20
IPR000719
PS50011
Protein kinase domain profile. Protein kinase





domain


21
IPR011009
SSF56112
Protein kinase-like domain superfamily


22
IPR012967
PF08100
Dimerisation domain Plant methyltransferase


23
IPR036390
SSF46785
Winged helix DNA-binding domain superfamily


24
IPR036388
G3DSA:1.10.10.10
Winged helix-like DNA-binding domain





superfamily


25
IPR029063
SSF53335
S-adenosyl-L-methionine-dependent





methyltransferase


26
IPR001077
PF00891
O-methyltransferase domain


27
IPR016461
PIRSF005739
O-methyltransferase COMT-type


28
IPR008998
PF07468
Agglutinin domain


29
IPR036242
SSF50382
Agglutinin domain superfamily


30
IPR005830
PF01117
Aerolysin toxin Aerolysin


31
IPR001509
PF01370
NAD dependent epimerase/dehydratase family


32
IPR036291
SSF51735
NAD(P)-binding domain superfamily


33
IPR000232
PS00434
HSF-type DNA-binding domain signature. Heat





shock factor (HSF)-type, DNA-binding


34
IPR005202
PS50985
GRAS family profile. Transcription factor GRAS


35
IPR003480
PF02458
Transferase family Transferase


36
IPR023213
G3DSA:3.30.559.10
Chloramphenicol acetyltransferase-like domain





superfamily


37
IPR007770
PF05078
Protein of unknown function (DUF679) Protein





DMP


38
IPR035995
SSF57247
Bowman-Birk type proteinase inhibitor


39
IPR000877
cd00023
BBI Proteinase inhibitor I12, Bowman-Birk


40
IPR003593
SM00382
AAA+ ATPase domain


41
IPR003959
PF00004
ATPase family associated with various cellular





activities (AAA) ATPase, AAA-type, core


42
IPR025753
PF14363
Domain associated at C-terminal with AAA





AAA-type ATPase, N-terminal domain


43
IPR027417
SSF52540
P-loop containing nucleoside triphosphate





hydrolase


44
IPR011032
SSF50129
GroES-like superfamily


45
IPR020843
SM00829
Polyketide synthase, enoylreductase domain


46
IPR013149
PF00107
Zinc-binding dehydrogenase Alcohol





dehydrogenase, C-terminal


47
IPR003245
PS51485
Phytocyanin domain profile. Phytocyanin domain


48
IPR008972
SSF49503
Cupredoxin


49
IPR035897
SSF52200
Toll/interleukin-1 receptor homology (TIR)





domain superfamily


50
IPR000157
PF01582
TIR domain Toll/interleukin-1 receptor homology





(TIR) domain


51
IPR020833
PS00711
Lipoxygenases iron-binding region signature 1.





Lipoxygenase, iron binding site


52
IPR036392
SSF49723
PLAT/LH2 domain superfamily


53
IPR036226
SSF48484
Lipoxigenase, C-terminal domain superfamily


54
IPR013819
PR00087
Lipoxygenase signature Lipoxygenase, C-terminal


55
IPR027433
G3DSA:4.10.372.10
Lipoxygenase, domain 3


56
IPR020834
PS00081
Lipoxygenases iron-binding region signature 2.





Lipoxygenase, conserved site


57
IPR001246
PR00468
Plant lipoxygenase signature Lipoxygenase, plant


58
IPR017441
PS00107
Protein kinases ATP-binding region signature.





Protein kinase, ATP binding site


59
IPR001611
PS51450
Leucine-rich repeat profile. Leucine-rich repeat


60
IPR032675
G3DSA:3.80.10.10
Leucine-rich repeat domain superfamily


61
IPR008271
PS00108
Serine/Threonine protein kinases active-site





signature.


62
IPR003591
SM00369
Leucine-rich repeat, typical subtype


63
IPR015425
PS51444
Formin homology-2 (FH2) domain profile.





Formin, FH2 domain


64
IPR020864
SM00457
Membrane attack complex component/perforin





(MACPF) domain


65
IPR036259
SSF103473
MFS transporter superfamily


66
IPR020846
cd06174
MFS Major facilitator superfamily domain


67
IPR000109
PF00854
POT family Proton-dependent oligopeptide





transporter family


68
IPR029058
SSF53474
Alpha/Beta hydrolase fold


69
IPR013094
PF07859
alpha/beta hydrolase fold Alpha/beta hydrolase





fold-3


70
IPR004014
PF00690
Cation transporter/ATPase, N-terminus Cation-





transporting P-type ATPase, N-terminal


71
IPR006534
TIGR01647
ATPase-IIIA_H: plasma-membrane proton-efflux





P-type ATPase P-type ATPase, subfamily IIIA


72
IPR001245
PF07714
Protein tyrosine kinase Serine-threonine/tyrosine-





protein kinase, catalytic domain


73
IPR036955
G3DSA:3.30.730.10
AP2/ERF domain superfamily


74
IPR016177
SSF54171
DNA-binding domain superfamily


75
IPR001471
PR00367
Ethylene responsive element binding protein





signature AP2/ERF domain


76
IPR001005
cd00167
SANT SANT/Myb domain


77
IPR009057
SSF46689
Homeobox-like domain superfamily


78
IPR017930
PS51294
Myb-type HTH DNA-binding domain profile.





Myb domain


79
IPR025886
PF14299
Phloem protein 2 Phloem protein 2-like


80
IPR005630
PF03936
Terpene synthase family, metal binding domain





Terpene synthase, metal-binding domain


81
IPR036965
G3DSA:1.50.10.130
Terpene synthase, N-terminal domain





superfamily


82
IPR008949
G3DSA:1.10.600.10
Isoprenoid synthase domain superfamily


83
IPR008930
SSF48239
Terpenoid cyclases/protein prenyltransferase





alpha-alpha toroid


84
IPR001906
PF01397
Terpene synthase, N-terminal domain


85
IPR016039
SSF53901
Thiolase-like


86
IPR001099
PF00195
Chalcone and stilbene synthases, N-terminal





domain Chalcone/stilbene synthase, N-terminal


87
IPR011141
PIRSF000451
Polyketide synthase, type III


88
IPR012328
PF02797
Chalcone and stilbene synthases, C-terminal





domain Chalcone/stilbene synthase, C-terminal


89
IPR002123
SM00563
Phospholipid/glycerol acyltransferase


90
IPR011051
SSF51182
RmlC-like cupin domain superfamily


91
IPR006045
SM00835
Cupin 1


92
IPR014710
G3DSA:2.60.120.10
RmlC-like jelly roll fold


93
IPR001929
PR00325
Germin signature Germin


94
IPR019780
PS00725
Germin family signature. Germin, manganese





binding site


95
IPR026961
PF13962
Domain of unknown function PGG domain


96
IPR036770
G3DSA:1.25.40.20
Ankyrin repeat-containing domain superfamily


97
IPR020683
cd00204
ANK Ankyrin repeat-containing domain


98
IPR002110
PS50088
Ankyrin repeat profile. Ankyrin repeat


99
IPR034289
cd13897
CuRO_3_LCC_plant Laccase, third cupredoxin





domain


100
IPR011706
PF07731
Multicopper oxidase Multicopper oxidase, type 2


101
IPR011707
PF07732
Multicopper oxidase Multicopper oxidase, type 3


102
IPR033138
PS00079
Multicopper oxidases signature 1. Multicopper





oxidases, conserved site


103
IPR002355
PS00080
Multicopper oxidases signature 2. Multicopper





oxidase, copper-binding site


104
IPR001117
PF00394
Multicopper oxidase Multicopper oxidase, type 1


105
IPR017761
TIGR03389
laccase: laccase


106
IPR034285
cd13875
CuRO_2_LCC_plant Laccase, second cupredoxin





domain


107
IPR034288
cd13849
CuRO_1_LCC_plant Laccase, first cupredoxin





domain


108
IPR004326
PF03094
Mlo family Mlo-related protein


109
IPR036396
G3DSA:1.10.630.10
Cytochrome P450 superfamily


110
IPR001128
PF00067
Cytochrome P450 Cytochrome P450


111
IPR017972
PS00086
Cytochrome P450 cysteine heme-iron ligand





signature. Cytochrome P450, conserved site


112
IPR002401
PR00463
E-class P450 group I signature Cytochrome P450,





E-class, group I









Example 5: Gene Cloning and Generation of Binary Vectors for Expression in Plants

To validate the role of genes identified hereinabove in increasing resistance to fungal infection, selected genes were over-expressed in plants, as follows.


Cloning Strategy

Selected genes from those presented in Examples 1-4 hereinabove were cloned into binary vectors for the generation of transgenic plants. In addition, to examine the effect of gene stacking, two pairs of genes were also cloned. For cloning, the full-length open reading frames (ORFs) were identified. EST clusters and in some cases mRNA sequences were analyzed to identify the entire open reading frame by comparing the results of several translation algorithms to known proteins from other plant species.


In order to clone the full-length cDNAs, reverse transcription (RT) followed by polymerase chain reaction (PCR; RT-PCR) was performed on total RNA extracted from roots or shoots challenged by the relevant pathogen. Total RNA extraction, production of cDNA and PCR amplification was performed using standard protocols described elsewhere (Sambrook J., E. F. Fritsch, and T. Maniatis. 1989. Molecular Cloning. A Laboratory Manual, 2nd Ed. Cold Spring Harbor Laboratory Press, New York) which are well known to those skilled in the art. PCR products were purified using PCR purification kit (Qiagen).


Typically, 2 sets of primers were prepared for the amplification of each gene, via nested PCR (if required). Both sets of primers were used for amplification on a cDNA. In case no product was obtained, a nested PCR reaction was performed. Nested PCR was performed by amplification of the gene using external primers and then using the produced PCR product as a template for a second PCR reaction, where the internal set of primers was used. Alternatively, one or two of the internal primers were used for gene amplification, both in the first and the second PCR reactions (meaning only 2-3 primers are designed for a gene). To facilitate further cloning of the cDNAs, an 8-12 base pairs (bp) extension was added to the 5′ of each internal primer. The primer extension includes an endonuclease restriction site. The restriction sites were selected using two parameters: (a) the restriction site does not exist in the cDNA sequence; and (b) the restriction sites in the forward and reverse primers were designed such that the digested cDNA was inserted in the sense direction into the binary vector utilized for transformation.


PCR products were digested with the restriction endonucleases (New England BioLabs Inc.) according to the sites designed in the primers. Each digested/undigested PCR product was inserted into a high copy vector pUC19 (New England BioLabs Inc.), or into plasmids originating from this vector. In some cases, the undigested PCR product was inserted into pCR-Blunt II-TOPO (Invitrogen) or into pJET1.2 (CloneJET PCR Cloning Kit, Thermo Scientific) or directly into the binary vector. The digested/undigested products and the linearized plasmid vector were ligated using T4 DNA ligase enzyme (Roche, Switzerland or other manufacturers). In cases where pCR-Blunt II-TOPO is used no T4 ligase was needed.


Sequencing of the inserted genes was performed using the ABI 377 sequencer (Applied Biosystems). In some cases, after confirming the sequences of the cloned genes, the cloned cDNA was introduced into pQ6sVN plasmid.


Several DNA sequences of the selected genes were synthesized by GeneArt™ (Life Technologies, Grand Island, NY, USA). Synthetic DNA was designed in silico. Suitable restriction enzyme sites were added to the cloned sequences at the 5′ end and at the 3′ end to enable later cloning into the desired binary vector.


Binary vectors—The pPI plasmid vector was constructed by inserting a synthetic poly-(A) signal sequence, originating from pGL3 basic plasmid vector (Promega, GenBank Accession No. U47295; nucleotides 4658-4811) into the HindIII restriction site of the binary vector pBI101.3 (Clontech, GenBank Accession No. U12640). pGI is similar to pPI, but the original gene in the backbone is GUS-Intron and not GUS.


The modified pGI vector (e.g., pQFN, pQFNc, pQFNd, pQYN_6669, pQNa_RP, pQFYN, pQXNc or pQ6sVN (FIG. 1) is a modified version of the pGI vector in which the cassette is inverted between the left and right borders so the gene and its corresponding promoter are close to the right border and the hygromycin-resistance conferring gene is close to the left border.


In case of Brachypodium transformation, after confirming the sequences of the cloned genes, the cloned cDNAs were introduced into pQ6sVN (FIG. 1) containing 35S promoter (SEQ ID NO:37) and the NOS terminator (SEQ ID NO:36) via digestion with appropriate restriction endonucleases. The genes were cloned downstream to the 35S promoter and upstream to the NOS terminator. In the pQ6sVN vector the Hygromycin resistance gene cassette and the Bar_GA resistance gene cassette replaced the NPTII resistance gene cassette. pQ6sVN contains the 35S promoter (SEQ ID NO:37). Bar_GA resistance gene (SEQ ID NO: 39) is an optimized sequence of the BAR gene for expression in Brachypodium plants (ordered from GeneArt™).


BAR gene coding sequence is provided in GenBank Accession No. JQ293091.1 (SEQ ID NO:38); further description is provided in Akama K, et al. “Efficient Agrobacterium-mediated transformation of Arabidopsis thaliana using the bar gene as selectable marker”, Plant Cell Rep. 1995, 14(7):450-4; Christiansen P, et al. “A rapid and efficient transformation protocol for the grass Brachypodium distachyon”, Plant Cell Rep. 2005 March; 23(10-11):751-8. Epub 2004 Oct. 19; and Pacurar D I, et al. “A high-throughput Agrobacterium-mediated transformation system for the grass model species Brachypodium distachyon L”, Transgenic Res. 2008 17(5):965-75; each of which is fully incorporated herein by reference in its entirety.


The pQ6sVN vector, in addition to the multiple cloning sites (MCS) between the promoter and terminator, contains additional restriction sites (PmeI, I-SceI, and BsaI) downstream of the expression cassette to allow cloning of a second expression cassette into the vector.


Stacking two genes (two expression cassettes) into pQ6sVN was performed as follows: the first gene was cloned into pQ6sVN via the MCS. The second gene was cloned into the vector pUCsVN. Genes cloned into this high copy vector via its MCS are flanked by a 35S promoter and a NOS terminator—the same elements found in the pQ6sVN vector. The expression cassette of pUCsVN is flanked by restriction sites for PmeI, I-SceI and BsaI. The cassette containing the second gene was excised from pUCsVN by digestion with one of the aforementioned enzymes, and ligated into the pQ6sVN plasmid already carrying the first gene, linearized with the same restriction enzyme. This resulted in a single vector harboring two “stacked” cassettes. The sequences and orientations of both cassettes were verified by Sanger sequencing and restriction digests.


In case genomic DNA was cloned, the genes were amplified by direct PCR on genomic DNA extracted from leaf tissue using the DNAeasy kit (Qiagen Cat. No. 69104).


Table 10 hereinbelow provides a list of the gene cloned, including gene name, the organism from which the gene is derived, the SEQ ID NO. of the primer used and the SEQ ID NO. of the gene polynucleotide and encoded polypeptide.









TABLE 10







Cloned genes














Polyn.
Polyp.


Gene

Primers used
SEQ ID
SEQ ID


Name
Organism
SEQ ID NOs
NO
NO














LFS82

Zea mays

535, 561, 574, 588
63
349


LFS83

Triticum aestivum

536, 562, 575, 589
64
350


LFS84

Triticum aestivum


65
310


LFS86

Zea mays


66
311


LFS89

Triticum aestivum


67
313


LFS90

Aegilops tauschii

511, 537, 563, 576
68
351


LFS91

Aegilops tauschii

512, 538
69
315


LFS92

Aegilops tauschii

513, 539
70
352


LFS93

Aegilops tauschii


71
317


LFS94

Aegilops tauschii

514, 540, 564
72
353


LFS95

Aegilops tauschii

515, 541, 565, 577
73
354


LFS96

Aegilops tauschii

516, 542
74
355


LFS97

Aegilops tauschii

517, 543, 566, 578
75
356


LFS98

Aegilops tauschii

518, 544, 567, 579
76
357


LFS99

Aegilops tauschii

519, 545, 568, 580
77
358


LFS100

Aegilops tauschii

520, 546, 569, 581
78
359


LFS102

Aegilops tauschii

521, 547, 582
79
325


LFS104

Aegilops tauschii

547, 582, 522, 548
80
360


LFS105

Aegilops tauschii

590, 591
307
510


LFS106

Aegilops tauschii

523, 549
81
327


LFS107

Aegilops tauschii

524, 550
82
361


LFS109

Aegilops tauschii

525, 551
83
362


LFS110

Aegilops tauschii


84
330


LFS111

Aegilops tauschii


85
331


LFS112

Aegilops tauschii


304
507


LFS113

Aegilops tauschii

526, 552, 570, 583
86
363


LFS114

Aegilops tauschii

527, 553, 571, 584
87
364


LFS115

Aegilops tauschii


88
334


LFS116

Aegilops tauschii

528, 554, 572, 585
89
335


LFS117

Aegilops tauschii

529, 555
90
365


LFS121

Setaria italica


91
337


LFS122

Setaria italica

530, 556, 573, 586
92
338


LFS123

Setaria italica

531, 557, 587
93
366


LFS124

Setaria italica


94
340


LFS125

Setaria italica

532, 558
95
367


LFS126

Setaria italica


96
342


LFS127

Setaria italica


97
343


LFS128

Setaria italica

533, 559
98
368


LFS129

Setaria italica

534, 560
99
345





“polyn.” = polynucleotide;


“polyp.” = polypeptide.






Example 6: Transformation of Brachypodium distachyon Plants with the Polynucleotides of the Invention

Similar to the Arabidopsis model plant, Brachypodium distachyon has several features that recommend it as a model plant for functional genomics studies, especially in the grasses. Traits that make it an ideal model include its small genome (˜160 Mbp for a diploid genome and 355 Mbp for a polyploidy genome), small physical stature, a short lifecycle, and few growth requirements. Brachypodium is related to the major cereal grain species but is understood to be more closely related to the Triticeae (wheat, barley) than to the other cereals. Brachypodium, with its polyploidy accessions, can serve as an ideal model for these grains (whose genomic size and complexity is a major barrier to biotechnological improvement).



Brachypodium distachyon embryogenic calli were transformed using the procedure described by Vogel and Hill (2008. High-efficiency Agrobacterium-mediated transformation of Brachypodium distachyon inbred line Bd21-3. Plant Cell Rep 27:471-478); Vain et al (2008. Agrobacterium-mediated transformation of the temperate grass Brachypodium distachyon (genotype Bd21) for T-DNA insertional mutagenesis. Plant Biotechnology J 6: 236-245), and Vogel J, et al. (2006. Agrobacterium mediated transformation and inbred line development in the model grass Brachypodium distachyon. Plant Cell Tiss Org. Cult. 85:199-211), each of which is fully incorporated herein by reference, with some minor modifications, which are briefly summarized hereinbelow.


Callus initiation—Immature spikes (about 2 months after seeding) were harvested at the very beginning of seeds filling. Spikes were then husked and surface sterilized with 3% NaClO containing 0.1% Tween 20, shaken on a gyratory shaker at low speed for 20 minutes. Following three rinses with sterile distilled water, embryos were excised under a dissecting microscope in a laminar flow hood using fine forceps.


Excised embryos (size ˜0.3 mm, bell shaped) were placed on callus induction medium (CIM) [LS salts (Linsmaier, E. M. & Skoog, F. 1965. Physiol. Plantarum 18, 100) and vitamins plus 3% sucrose, 6 mg/L CuSO4, 2.5 mg/l 2,4-Dichlorophenoxyacetic Acid, pH 5.8 and 0.25% phytagel (Sigma)] with the scutellum side down, 50 or 100 embryos on a plate, and incubated at 28° C. in the dark. One week later, the embryonic calli were cleaned from emerging shoots and somatic calli, and subcultured onto fresh CIM medium. During culture, yellowish embryogenic calli (EC) appear and were further selected (e.g., picked and transferred) for further incubation in the same conditions for additional 2 weeks. Twenty-five pieces of sub-cultured calli were then separately placed on 90×15 mm petri plates, and incubated as before for three additional weeks.


Transformation—As described in Vogel and Hill (2008, Supra), Agrobacterium is scraped off 2-day-old MGL plates (plates with the MGL medium which contains: Tryptone 5 gr/L, Yeast Extract 2.5 gr/L, NaCl 5 gr/L, D-Mannitol 5 g/l, MgSO4*7H2O 0.204 gr/L, K2HPO4 0.25 gr/L, Glutamic Acid 1.2 gr/L, Plant Agar 7.5 gr/L) and resuspended in liquid MS medium supplemented with 200 μM acetosyringone to an optic density (OD) at 600 nm (OD600) of 0.6 to 1.0. Once the desired OD was attained, 1 ml of 10% Synperonic PE/F68 (Sigma) per 100 ml of inoculation medium is added.


To begin inoculation, 300 callus pieces were placed in approximately 12 plates (25 callus pieces in each plate) and covered with the Agrobacterium suspension (8-10 ml). The callus was incubated in the Agrobacterium suspension for 5 to 20 minutes. After incubation, the Agrobacterium suspension was aspirated off and the calli were then transferred into co-cultivation plates, prepared by placing a sterile 7-cm diameter filter paper in an empty 90×15 mm petri plate. The calli pieces are then gently distributed on the filter paper. One co-cultivation plate is used for two starting callus plates (50 initial calli pieces). The co-cultivation plates were then sealed with Parafilm M® or a plastic wrap [e.g., Saran™ wrap (Dow Chemical Company)] and incubated at 24° C. in the dark for 3 days.


The callus pieces were then individually transferred into CIM medium as described above, which was further supplemented with 200 mg/L Ticarcillin (to kill the Agrobacterium) and Bialaphos (5 mg/L) or Hygromycin B (40 mg/L) (for selection of the transformed resistant embryogenic calli sections), and incubated at 28° C. in the dark for 14 days.


The calli pieces were then transferred to shoot induction media (SIM; LS salts and vitamins plus 3% Maltose monohydrate) supplemented with 400 mg/L Ticarcillin, Bialaphos (5 mg/L) or Hygromycin B (40 mg/L), Indol-3-acetic acid (IAA) (0.25 mg/L), and 6-Benzylaminopurine (BAP) (1 mg/L), and were cultivated in conditions as described below. After 10-15 days calli were sub-cultured on the same fresh media for additional 10-15 days (total of 20-30 days). At each sub-culture all the pieces from a single callus were kept together to maintain their independence and were incubated under the following conditions: light to a level of 60 lE m−2 s−1, a 16-hours light, 8-hours dark photoperiod and a constant 24° C. temperature. During the period of 20 to 30 days from the beginning of cultivation of calli on shoot induction media (SIM) plantlets start to emerge from the transformed calli.


When plantlets were large enough to handle without damage, they were transferred to plates containing the above-mentioned shoot induction media (SIM) with Bialaphos or Hygromycin B. Each plantlet is considered as a different event. After two weeks of growth, the plantlets were transferred to 2-cm height Petri plates (De Groot, Catalog No. 60-664160) containing MSnoH media (MS salts 4.4 gr/L, sucrose 30 gr/L, supplemented with Hygromycin B (40 mg/L) and Ticarcillin (400 mg/L). Roots usually appear within 2 weeks. Rooted and non-rooted plants were transferred to a fresh MSnoH media supplemented with Hygromycin B and Ticarcillin as described above. In case roots do not appear in the non-rooted plants after two weeks on the MSnoH media (which is supplemented with Hygromycin B and Ticarcillin), then the non-rooted plants are further transferred to the rooting induction medium [RIM; MS salts and vitamins 4.4 gr/L, sucrose 30 gr/L with Ticarcillin 400 mg/L, Indol-3-acetic acid (IAA) (1 mg/L), and α-Naphthalene acetic acid (NAA) (2 mg/L)]. After additional two weeks of incubation at 24° C., the plantlets are transferred to 0.5 modified RIM medium [MS modified salts 4.4 gr/L, MS vitamins 103 mg/L, sucrose 30 gr/L with α-Tocopherol (2 mg/L), Indol-3-acetic acid (IAA) (1 mg/L), and α-Naphthalene acetic acid (NAA) (2 mg/L)] and are incubated at 28° C. for additional 15-20 days, till the roots appear.


If needed, in the tillering stage the plantlets can grow axillary tillers and eventually become bushy on the above-mentioned media (SIM) without Bialaphos or Hygromycin B. Each bush from the same plant (event ID) is then divided to tissue culture boxes (“Humus”) containing “rooting medium” [MS basal salts, 3% sucrose, 3 gr/L phytagel, 2 mg/L α-Naphthalene Acetic Acid (NAA) and 1 mg/L IAA and Ticarcillin 400 mg/L, PH 5.8]. All plants in a “Humus box” are individual plants of the same transformation event.


When plantlets establish roots, they are transplanted to the soil and grown in the greenhouse. Before transfer to greenhouse, 20 randomly selected events are tested every month for expression of the BAR_GA gene (SEQ ID NO:39, BAR gene) which is responsible for resistance to Bialaphos, using AgraStrip® LL strip test seed check (Romer labs). Briefly, the expression of the BAR gene is determined as follows: Leaves (about 0.5 cm long leave) are grounded using a pellet pestle in an Eppendorf tube containing 150 μl of water until the water turns green in color. A strip test is then added to the Eppendorf tube and the results are read within 30-60 seconds. Appearance of two pink bands means that the plant is transgenic. On the other hand, appearance of one pink band means that the plant is not transgenic or not expressing BAR gene.


To verify the transgenic status of plants containing the gene of interest, T1 plants are subjected to PCR as previously described by Vogel et al. 2006 [Agrobacterium mediated transformation and inbred line development in the model grass Brachypodium distachyon. Plant Cell Tiss Org. Cult. 85:199-211].


Example 7: Validation Assays

The transgenic Brachypodium plants obtained as described hereinabove were used to validate the effect of the transformed gene(s) on fungal penetration and spreading within the plant by evaluation of fungal penetration and spreading within inoculated seedlings grown under controlled conditions.


Each validation assay evaluates the gene performance by quantitative and/or qualitative measure of specific traits as described in Table 11 below.









TABLE 11







Allocation of fungal parameters to specific traits











#
Parameters
Traits







1
Fungal biomass in root
Fungal presence in tissue










The validation assay was performed with inoculated transgenic plants grown under controlled conditions till seedling stage (1-2 tillers).


Transgenic Brachypodium seeds were sown in trays contained sterilized vermiculite soaked with 0.1% BASTA solution for selection of transgenic plants. The trays were placed in the refrigerator at 4° C. for cold treatment for 3 days (stratification step) following by trays placement in controlled growth chamber for germination up to 5 days (26° C.±1, humidity of 50%).


The viable transgenic seedlings were inoculated with Fusarium verticillioides (Fv 149 strain, GFP-tagged and bar-resistant) spore solution (106/ml) and then transplanted to 180 cm3 pots filled with sterilized vermiculite. The plants were grown for 7 days in the growth chamber.


For fungal biomass recovery, roots externally sterilized and then grinded. Serial dilutions of the extracts from each one of the biological repeats (1 biological repeat is a pool of 6 roots) were plated in Petri dishes and incubated for 5 days at 25° C. Developing colonies were counted to provide Colony Forming Units (CFU) using a florescent binocular and data were transformed to logarithmic scale for further analysis.


Fungal Biomass was computed as Log CFU of fungal root extraction 7 days after inoculation


Statistical analyses—To identify genes conferring significantly improved tolerance to fungal penetration and fungal spreading, the results obtained from the transgenic plants were compared to those obtained from control plants. To identify outperforming genes and constructs, results from the independent transformation events tested were analyzed separately. Analysis was conducted on the log CFU data using Student's t-test. The JMP statistics software package was used (Version 5.2.1, SAS Institute Inc., Cary, NC, USA). Table 12 hereinbelow shows the reduction in CFU in the tested infected plants and its significance compared to control plants.









TABLE 12







Validation results (single genes): reduction of fungi burden in



Brachypodium plant infected with Fusarium verticilloides











Gene

% change of logCFU relative to control
% change of CFU


Name
Origin
(p value)
relative to control





LFS90

Aegilops

first assay:
first assay:




Event 13869.1: 11.45% (significant, p_value = 0.00)
Event 13869.1: 179.54%




Event 13870.1: 6.93% (significant, p_value = 0.02)
Event 13870.1: 86.38%




Event 13865.1: 9.40% (significant, p_value = 0.00)
Event 13865.1: 132.57%




Event 13866.1: 18.38% (significant, p_value = 0.00)
Event 13866.1: 420.96%




Event 13868.1: 10.57% (significant, p_value = 0.00)
Event 13868.1: 158.22%




Event 13874.1: 9.78% (significant, p_value = 0.00)
Event 13874.1: 140.59%




second assay:
second assay:




Event 13869.1: −1.72% (not significant, p_value = 0.25)
Event 13869.1: -14.02%




Event 13865.1: −2.28% (not significant, p_value = 0.18)
Event 13865.1: −18.22%




Event 13866.1: 3.23% (not significant, p_value = 0.11)
Event 13866.1: 32.90%




Event 13868.1: −3.61% (significant, p_value = 0.08)
Event 13868.1: −27.22%




Event 13874.1: 0.44% (not significant, p_value = 0.44)
Event 13874.1: 3.95%


LFS91

Aegilops

first assay:
first assay:




Event 13884.1: 8.87% (significant, p_value = 0.00)
Event 13884.1: 134.98%




Event 13883.1: −6.99% (significant, p_value = 0.01)
Event 13883.1: −49.02%




Event 13885.1: −3.76% (not significant, p_value = 0.10)
Event 13885.1: −30.38%




Event 13891.1: 0.26% (not significant, p_value = 0.46)
Event 13891.1: 2.55%




Event 13890.1: 9.90% (significant, p_value = 0.00)
Event 13890.1: 159.62%




Event 13894.1: 1.62% (not significant, p_value = 0.29)
Event 13894.1: 16.93%




second assay:
second assay:




Event 13883.1: −9.31% (significant, p_value = 0.00)
Event 13883.1: −61.15%


LFS92

Aegilops

first assay:
first assay:




Event 14188.1: −0.38% (not significant, p_value = 0.44)
Event 14188.1: −3.18%




Event 14202.1: −4.87% (significant, p_value = 0.08)
Event 14202.1: −33.67%




Event 14152.1: −5.65% (significant, p_value = 0.04)
Event 14152.1: −37.92%




Event 14155.1: −1.66% (not significant, p_value = 0.31)
Event 14155.1: −13.09%




Event 14187.1: −1.17% (not significant, p_value = 0.37)
Event 14187.1: −9.42%




Event 14201.1: −0.06% (not significant, p_value = 0.50)
Event 14201.1: −0.52%




second assay:
second assay:




Event 14202.1: −1.85% (not significant, p_value = 0.29)
Event 14202.1: −16.59%




Event 14152.1: −11.40% (significant, p_value = 0.00)
Event 14152.1: −67.31%


LFS93

Aegilops

first assay:
first assay:




Event 14149.1: −2.25% (not significant, p_value = 0.28)
Event 14149.1: −18.91%




Event 14150.1: 10.52% (significant, p_value = 0.00)
Event 14150.1: 166.28%




Event 14108.1: −1.06% (not significant, p_value = 0.38)
Event 14108.1: −9.38%




Event 14147.1: −5.21% (significant, p_value = 0.07)
Event 14147.1: −38.44%




Event 14156.1: 8.77% (significant, p_value = 0.01)
Event 14156.1: 126.22%




Event 14157.1: 7.58% (significant, p_value = 0.01)
Event 14157.1: 102.48%




second assay:
second assay:




Event 14147.1: −3.07% (not significant, p_value = 0.14)
Event 14147.1: −25.88%


LFS94

Aegilops

first assay:
first assay:




Event 14165.1: 4.96% (significant, p_value = 0.07)
Event 14165.1: 49.56%




Event 14164.1: 2.87% (not significant, p_value = 0.19)
Event 14164.1: 26.25%




Event 14162.1: 0.03% (not significant, p_value = 0.50)
Event 14162.1: 0.23%




Event 14171.1: 8.46% (significant, p_value = 0.01)
Event 14171.1: 98.73%




Event 14172.1: 8.30% (significant, p_value = 0.01)
Event 14172.1: 96.09%




Event 14168.1: 3.84% (not significant, p_value = 0.12)
Event 14168.1: 36.52%


LFS95

Aegilops

first assay:
first assay:




Event 14213.1: 9.19% (significant, p_value = 0.01)
Event 14213.1: 135.70%




Event 14212.1: 4.42% (not significant, p_value = 0.12)
Event 14212.1: 51.10%




Event 14208.1: 5.86% (significant, p_value = 0.05)
Event 14208.1: 72.71%




Event 14205.1: 0.68% (not significant, p_value = 0.43)
Event 14205.1: 6.50%




Event 14210.1: 6.97% (significant, p_value = 0.03)
Event 14210.1: 91.54%




Event 14214.1: −5.60% (significant, p_value = 0.06)
Event 14214.1: −40.68%




second assay:
second assay:




Event 14214.1: 6.12% (significant, p_value = 0.04)
Event 14214.1: 82.23%


LFS96

Aegilops

first assay:
first assay:




Event 14017.34: −10.34% (significant, p_value = 0.00)
Event 14017.34: −63.12%




Event 14018.29: −7.66% (significant, p_value = 0.01)
Event 14018.29: −52.23%




Event 14014.38: −18.53% (significant, p_value = 0.00)
Event 14014.38: −83.26%




Event 14015.40: −2.21% (not significant, p_value = 0.25)
Event 14015.40: −19.17%




Event 14019.34: −8.21% (significant, p_value = 0.01)
Event 14019.34: −54.70%




Event 14020.36: −5.72% (significant, p_value = 0.04)
Event 14020.36: −42.40%




second assay:
second assay:




Event 14017.34: −1.34% (not significant, p_value = 0.28)
Event 14017.34: −13.44%




Event 14018.29: 2.48% (not significant, p_value = 0.15)
Event 14018.29: 30.61%




Event 14014.38: −2.69% (not significant, p_value = 0.13)
Event 14014.38: −25.17%




Event 14019.34: −2.40% (not significant, p_value = 0.15)
Event 14019.34: −22.82%




Event 14020.36: −0.85% (not significant, p_value = 0.36)
Event 14020.36: −8.74%


LFS97

Aegilops

first assay:
first assay:




Event 13932.1: −1.43% (not significant, p_value = 0.28)
Event 13932.1: −12.81%




Event 13938.1: −2.60% (not significant, p_value = 0.15)
Event 13938.1: −22.09%




Event 13936.1: −1.96% (not significant, p_value = 0.22)
Event 13936.1: −17.12%




Event 13937.1: −5.62% (significant, p_value = 0.01)
Event 13937.1: −41.63%




Event 13939.1: 1.19% (not significant, p_value = 0.32)
Event 13939.1: 12.11%




Event 13933.1: −0.78% (not significant, p_value = 0.38)
Event 13933.1: −7.17%




second assay:
second assay:




Event 13937.1: 3.78% (not significant, p_value = 0.13)
Event 13937.1: 41.87%




Event 13937.1: −6.16% (significant, p_value = 0.00)
Event 13937.1: −48.55%


LFS98

Aegilops

first assay:
first assay:




Event 13911.1: −0.83% (not significant, p_value = 0.42)
Event 13911.1: −7.43%




Event 13917.1: −2.20% (not significant, p_value = 0.22)
Event 13917.1: −18.51%




Event 13910.1: −5.18% (significant, p_value = 0.03)
Event 13910.1: −38.17%




Event 13909.1: −0.66% (not significant, p_value = 0.43)
Event 13909.1: −5.97%




Event 13915.1: 8.77% (significant, p_value = 0.00)
Event 13915.1: 125.80%




Event 13919.1: 6.10% (significant, p_value = 0.01)
Event 13919.1: 76.17%




second assay:
second assay:




Event 13910.1: −4.21% (not significant, p_value = 0.12)
Event 13910.1: −34.81%


LFS99

Aegilops

first assay:
first assay:




Event 14138.1: 12.65% (significant, p_value = 0.00)
Event 14138.1: 158.53%




Event 14137.1: 6.25% (significant, p_value = 0.02)
Event 14137.1: 59.92%




Event 14140.1: 12.90% (significant, p_value = 0.00)
Event 14140.1: 163.35%




Event 14134.1: 8.96% (significant, p_value = 0.00)
Event 14134.1: 95.88%




Event 14135.1: −4.95% (significant, p_value = 0.05)
Event 14135.1: −31.05%




Event 14142.1: 24.44% (significant, p_value = 0.00)
Event 14142.1: 526.43%




second assay:
second assay:




Event 14138.1: −2.38% (not significant, p_value = 0.23)
Event 14138.1: −20.09%




Event 14137.1: 13.83% (significant, p_value = 0.00)
Event 14137.1: 268.06%




Event 14140.1: 9.65% (significant, p_value = 0.00)
Event 14140.1: 148.18%




Event 14135.1: 0.57% (not significant, p_value = 0.46)
Event 14135.1: 5.54%




Event 14141.1: −0.95% (not significant, p_value = 0.35)
Event 14141.1: −8.60%




Event 14142.1: 5.81% (significant, p_value = 0.06)
Event 14142.1: 72.84%


LFS100

Aegilops

first assay:
first assay:




Event 13984.27: 8.05% (significant, p_value = 0.01)
Event 13984.27: 125.39%




Event 14070.35: −2.27% (not significant, p_value = 0.27)
Event 14070.35: −20.44%




Event 14052.30: −3.67% (not significant, p_value = 0.14)
Event 14052.30: −30.97%




Event 14051.31: −2.02% (not significant, p_value = 0.30)
Event 14051.31: −18.41%




Event 14077.38: −4.53% (significant, p_value = 0.10)
Event 14077.38: −36.71%




Event 13987.36: −4.48% (not significant, p_value = 0.10)
Event 13987.36: −36.40%




second assay:
second assay:




Event 14077.38: −1.98% (not significant, p_value = 0.27)
Event 14077.38: −18.25%


LFS104

Aegilops

first assay:
first assay:




Event 13943.39: −0.15% (not significant, p_value = 0.47)
Event 13943.39: −1.54%




Event 14068.24: −2.02% (not significant, p_value = 0.29)
Event 14068.24: −18.43%




Event 14069.22: 2.54% (not significant, p_value = 0.27)
Event 14069.22: 29.23%




Event 14022.32: −11.15% (significant, p_value = 0.00)
Event 14022.32: −67.60%




Event 14023.24: −9.31% (significant, p_value = 0.02)
Event 14023.24: −60.99%




Event 14067.17: 4.44% (not significant, p_value = 0.14)
Event 14067.17: 56.62%




second assay:
second assay:




Event 14022.32: −19.32% (significant, p_value = 0.00)
Event 14022.32: −86.85%




Event 14023.24: −20.99% (significant, p_value = 0.00)
Event 14023.24: −88.96%


LFS105

Aegilops

first assay:
first assay:




Event 14026.1: −8.60% (significant, p_value = 0.00)
Event 14026.1: −56.52%




Event 14035.1: −1.32% (not significant, p_value = 0.29)
Event 14035.1: −12.04%




Event 14031.1: −3.52% (significant, p_value = 0.09)
Event 14031.1: −28.86%




Event 14025.1: −2.91% (not significant, p_value = 0.12)
Event 14025.1: −24.57%




Event 14034.1: −2.54% (not significant, p_value = 0.16)
Event 14034.1: −21.80%




Event 14029.1: −2.34% (not significant, p_value = 0.17)
Event 14029.1: −20.30%




second assay:
second assay:




Event 14026.1: −4.00% (significant, p_value = 0.08)
Event 14026.1: −32.37%




Event 14031.1: 2.04% (not significant, p_value = 0.23)
Event 14031.1: 22.08%


LFS106

Aegilops

first assay:
first assay:




Event 13993.33: 6.18% (significant, p_value = 0.02)
Event 13993.33: 80.50%




Event 13996.23: 8.88% (significant, p_value = 0.00)
Event 13996.23: 133.43%




Event 14000.41: 7.34% (significant, p_value = 0.01)
Event 14000.41: 101.56%




Event 14003.37: 2.66% (not significant, p_value = 0.21)
Event 14003.37: 28.96%




Event 13999.42: 2.15% (not significant, p_value = 0.24)
Event 13999.42: 22.80%




Event 14001.37: 11.19% (significant, p_value = 0.00)
Event 14001.37: 191.28%




second assay:
second assay:




Event 13993.33: −7.16% (significant, p_value = 0.05)
Event 13993.33: −52.83%




Event 13996.23: −5.75% (significant, p_value = 0.09)
Event 13996.23: −45.32%




Event 14000.41: −2.56% (not significant, p_value = 0.27)
Event 14000.41: −23.57%




Event 14001.37: −4.58% (not significant, p_value = 0.14)
Event 14001.37: −38.15%


LFS107

Aegilops

first assay:
first assay:




Event 13951.32: 10.34% (significant, p_value = 0.00)
Event 13951.32: 153.93%




Event 13948.31: 11.64% (significant, p_value = 0.00)
Event 13948.31: 185.33%




Event 13946.32: 8.99% (significant, p_value = 0.00)
Event 13946.32: 124.83%




Event 13956.45: 11.85% (significant, p_value = 0.00)
Event 13956.45: 190.80%




Event 13960.33: 4.67% (significant, p_value = 0.06)
Event 13960.33: 52.37%




second assay:
second assay:




Event 13951.32: 1.24% (not significant, p_value = 0.34)
Event 13951.32: 11.52%




Event 13948.31: 3.36% (significant, p_value = 0.09)
Event 13948.31: 34.45%




Event 13946.32: 1.90% (not significant, p_value = 0.21)
Event 13946.32: 18.24%




Event 13956.45: 4.76% (significant, p_value = 0.02)
Event 13956.45: 52.17%


LFS109

Aegilops

first assay:
first assay:




Event 14193.1: −5.39% (significant, p_value = 0.00)
Event 14193.1: −39.83%




Event 14194.1: 1.98% (not significant, p_value = 0.14)
Event 14194.1: 20.52%




Event 14197.1: 0.05% (not significant, p_value = 0.48)
Event 14197.1: 0.51%




Event 14192.1: 0.49% (not significant, p_value = 0.38)
Event 14192.1: 4.67%




Event 14198.1: −5.18% (significant, p_value = 0.00)
Event 14198.1: −38.60%




Event 14191.1: −1.98% (not significant, p_value = 0.15)
Event 14191.1: −16.99%




second assay:
second assay:




Event 14193.1: 2.69% (not significant, p_value = 0.16)
Event 14193.1: 30.08%


LFS110

Aegilops

first assay:
first assay:




Event 14254.1: −3.90% (significant, p_value = 0.07)
Event 14254.1: −33.07%




Event 14283.1: 0.61% (not significant, p_value = 0.40)
Event 14283.1: 6.45%




Event 14255.1: 4.83% (significant, p_value = 0.03)
Event 14255.1: 64.37%




Event 14284.1: −6.43% (significant, p_value = 0.01)
Event 14284.1: −48.43%




Event 14250.1: 0.60% (not significant, p_value = 0.41)
Event 14250.1: 6.40%




Event 14252.1: 0.17% (not significant, p_value = 0.47)
Event 14252.1: 1.81%




second assay:
second assay:




Event 14254.1: 0.22% (not significant, p_value = 0.44)
Event 14254.1: 2.24%




Event 14284.1: −0.97% (not significant, p_value = 0.32)
Event 14284.1: −9.28%


LFS111

Aegilops

first assay:
first assay:




Event 14064.1: 17.84% (significant, p_value = 0.00)
Event 14064.1: 433.55%




Event 14058.1: −9.20% (significant, p_value = 0.00)
Event 14058.1: −57.84%




Event 14060.1: −8.68% (significant, p_value = 0.00)
Event 14060.1: −55.73%




Event 14073.1: 10.65% (significant, p_value = 0.00)
Event 14073.1: 171.77%




Event 14057.1: −5.24% (significant, p_value = 0.03)
Event 14057.1: −38.82%




Event 14059.1: 13.43% (significant, p_value = 0.00)
Event 14059.1: 252.78%




second assay:
second assay:




Event 14058.1: −2.16% (not significant, p_value = 0.20)
Event 14058.1: −17.33%




Event 14057.1: 3.96% (not significant, p_value = 0.17)
Event 14057.1: 42.13%




Event 14060.1: −0.35% (not significant, p_value = 0.45)
Event 14060.1: −3.33%


LFS112

Aegilops

first assay:
first assay:




Event 14307.1: −5.30% (significant, p_value = 0.01)
Event 14307.1: −40.36%




Event 14309.1: −1.49% (not significant, p_value = 0.25)
Event 14309.1: −13.54%




Event 14298.1: 0.65% (not significant, p_value = 0.40)
Event 14298.1: 6.53%




Event 14301.1: −4.38% (significant, p_value = 0.03)
Event 14301.1: −34.78%




Event 14302.1: −9.07% (significant, p_value = 0.00)
Event 14302.1: −58.75%




Event 14305.1: −1.56% (not significant, p_value = 0.24)
Event 14305.1: −14.12%




second assay:
second assay:




Event 14307.1: 0.90% (not significant, p_value = 0.33)
Event 14307.1: 9.44%




Event 14301.1: −3.62% (significant, p_value = 0.04)
Event 14301.1: −30.53%




Event 14302.1: −4.12% (significant, p_value = 0.02)
Event 14302.1: −33.92%


LFS113

Aegilops

first assay:
first assay:




Event 13974.29: 4.15% (significant, p_value = 0.05)
Event 13974.29: 44.75%




Event 13979.31: 4.39% (significant, p_value = 0.04)
Event 13979.31: 47.89%


LFS114

Aegilops

first assay:
first assay:




Event 14179.1: −14.48% (significant, p_value = 0.00)
Event 14179.1: −75.12%




Event 14181.1: −4.51% (significant, p_value = 0.03)
Event 14181.1: −35.14%




Event 14175.1: −8.02% (significant, p_value = 0.00)
Event 14175.1: −53.70%




Event 14184.1: −7.70% (significant, p_value = 0.00)
Event 14184.1: −52.26%




Event 14174.1: −20.43% (significant, p_value = 0.00)
Event 14174.1: −85.95%




Event 14177.1: −9.03% (significant, p_value = 0.00)
Event 14177.1: −58.00%




second assay:
second assay:




Event 14179.1: −1.72% (not significant, p_value = 0.28)
Event 14179.1: −15.43%




Event 14181.1: −1.90% (not significant, p_value = 0.26)
Event 14181.1: −16.90%




Event 14175.1: 0.26% (not significant, p_value = 0.46)
Event 14175.1: 2.59%




Event 14184.1: −1.44% (not significant, p_value = 0.31)
Event 14184.1: −13.09%




Event 14174.1: −3.09% (not significant, p_value = 0.14)
Event 14174.1: −25.97%




Event 14177.1: −2.13% (not significant, p_value = 0.23)
Event 14177.1: −18.75%


LFS115

Aegilops

first assay:
first assay:




Event 14260.1: 1.19% (not significant, p_value = 0.34)
Event 14260.1: 12.64%




Event 14261.1: −0.84% (not significant, p_value = 0.39)
Event 14261.1: −8.09%




Event 14268.1: 4.73% (significant, p_value = 0.05)
Event 14268.1: 60.69%




Event 14259.1: 0.10% (not significant, p_value = 0.49)
Event 14259.1: 0.97%




Event 14263.1: −8.76% (significant, p_value = 0.00)
Event 14263.1: −58.48%




Event 14262.1: 4.26% (significant, p_value = 0.08)
Event 14262.1: 53.37%




second assay:
second assay:




Event 14263.1: −4.66% (significant, p_value = 0.01)
Event 14263.1: −37.40%


LFS116

Aegilops

first assay:
first assay:




Event 13962.41: −2.38% (not significant, p_value = 0.30)
Event 13962.41: −20.21%




Event 13898.1: −2.67% (not significant, p_value = 0.27)
Event 13898.1: −22.39%




Event 13904.1: −7.55% (significant, p_value = 0.04)
Event 13904.1: −51.20%




Event 13961.30: −4.26% (not significant, p_value = 0.17)
Event 13961.30: −33.27%




Event 13902.1: −3.32% (not significant, p_value = 0.23)
Event 13902.1: −27.05%




Event 13903.1: −7.12% (significant, p_value = 0.06)
Event 13903.1: −49.13%




second assay:
second assay:




Event 13904.1: −1.77% (not significant, p_value = 0.29)
Event 13904.1: −16.48%




Event 13903.1: −0.56% (not significant, p_value = 0.44)
Event 13903.1: −5.52%


LFS117

Aegilops

first assay:
first assay:




Event 14246.1: −5.66% (significant, p_value = 0.05)
Event 14246.1: −42.61%




Event 14203.1: 3.03% (not significant, p_value = 0.19)
Event 14203.1: 34.55%




Event 14245.1: −9.05% (significant, p_value = 0.00)
Event 14245.1: −58.81%




second assay:
second assay:




Event 14246.1: 3.99% (not significant, p_value = 0.14)
Event 14246.1: 44.59%




Event 14245.1: −1.48% (not significant, p_value = 0.34)
Event 14245.1: −12.76%


LFS122
Foxtail
first assay:
first assay:



millet
Event 13926.1: 10.35% (significant, p_value = 0.00)
Event 13926.1: 160.27%




Event 13880.1: 14.50% (significant, p_value = 0.00)
Event 13880.1: 281.82%




Event 13879.1: 12.68% (significant, p_value = 0.00)
Event 13879.1: 222.64%




Event 13908.1: 6.91% (significant, p_value = 0.01)
Event 13908.1: 89.30%




Event 13906.1: 0.21% (not significant, p_value = 0.47)
Event 13906.1: 1.99%




Event 13907.1: −3.63% (significant, p_value = 0.09)
Event 13907.1: −28.52%




second assay:
second assay:




Event 13907.1: −9.24% (significant, p_value = 0.00)
Event 13907.1: −60.88%


LFS123
Foxtail
first assay:
first assay:



millet
Event 14127.1: 3.23% (not significant, p_value = 0.11)
Event 14127.1: 37.61%




Event 14130.1: −0.64% (not significant, p_value = 0.40)
Event 14130.1: −6.18%




Event 14125.1: −13.66% (significant, p_value = 0.00)
Event 14125.1: −74.11%




Event 14121.1: −5.44% (significant, p_value = 0.02)
Event 14121.1: −41.63%




Event 14122.1: 7.25% (significant, p_value = 0.00)
Event 14122.1: 104.81%




Event 14131.1: 1.34% (not significant, p_value = 0.30)
Event 14131.1: 14.13%




second assay:
second assay:




Event 14125.1: 0.49% (not significant, p_value = 0.47)
Event 14125.1: 4.42%




Event 14121.1: −0.47% (not significant, p_value = 0.39)
Event 14121.1: −4.06%


LFS125
Foxtail
first assay:
first assay:



millet
Event 14043.1: 0.01% (not significant, p_value = 0.49)
Event 14043.1: 0.05%




Event 14056.1: −6.59% (significant, p_value = 0.02)
Event 14056.1: −46.26%




Event 14044.1: −6.57% (significant, p_value = 0.02)
Event 14044.1: −46.15%




Event 14048.1: 1.23% (not significant, p_value = 0.34)
Event 14048.1: 12.26%




Event 14042.1: 1.20% (not significant, p_value = 0.34)
Event 14042.1: 11.95%




Event 14053.1: −4.57% (significant, p_value = 0.08)
Event 14053.1: −34.98%




second assay:
second assay:




Event 14056.1: −6.84% (significant, p_value = 0.02)
Event 14056.1: −50.22%




Event 14044.1: −6.73% (significant, p_value = 0.02)
Event 14044.1: −49.65%




Event 14053.1: −3.49% (not significant, p_value = 0.15)
Event 14053.1: −29.92%


LFS126
Foxtail
first assay:
first assay:



millet
Event 14081.1: −4.75% (significant, p_value = 0.02)
Event 14081.1: −37.78%




Event 14089.1: −9.88% (significant, p_value = 0.00)
Event 14089.1: −62.77%




Event 14085.1: 6.71% (significant, p_value = 0.00)
Event 14085.1: 95.51%




Event 14082.1: −0.40% (not significant, p_value = 0.43)
Event 14082.1: −3.88%




Event 14086.1: −10.60% (significant, p_value = 0.00)
Event 14086.1: −65.34%




Event 14087.1: 3.21% (significant, p_value = 0.08)
Event 14087.1: 37.82%




second assay:
second assay:




Event 14081.1: −5.25% (significant, p_value = 0.06)
Event 14081.1: −41.44%




Event 14089.1: 2.31% (not significant, p_value = 0.23)
Event 14089.1: 26.61%




Event 14086.1: −4.17% (significant, p_value = 0.10)
Event 14086.1: −34.67%


LFS127
Foxtail
first assay:
first assay:



millet
Event 14102.1: −1.42% (not significant, p_value = 0.32)
Event 14102.1: −14.65%




Event 14104.1: −6.45% (significant, p_value = 0.01)
Event 14104.1: −51.20%




Event 14101.1: −3.76% (significant, p_value = 0.08)
Event 14101.1: −34.14%




Event 14096.1: −4.07% (significant, p_value = 0.07)
Event 14096.1: −36.39%




Event 14098.1: −7.82% (significant, p_value = 0.00)
Event 14098.1: −58.07%




Event 14100.1: −9.83% (significant, p_value = 0.00)
Event 14100.1: −66.48%




second assay:
second assay:




Event 14104.1: 2.31% (not significant, p_value = 0.29)
Event 14104.1: 24.66%




Event 14101.1: −0.33% (not significant, p_value = 0.47)
Event 14101.1: −3.06%




Event 14096.1: 7.82% (significant, p_value = 0.03)
Event 14096.1: 110.92%




Event 14098.1: −1.41% (not significant, p_value = 0.37)
Event 14098.1: −12.56%




Event 14100.1: 2.72% (not significant, p_value = 0.26)
Event 14100.1: 29.66%


LFS128
Foxtail
first assay:
first assay:



millet
Event 14222.1: −0.79% (not significant, p_value = 0.40)
Event 14222.1: −7.05%




Event 14223.1: −2.67% (not significant, p_value = 0.19)
Event 14223.1: −21.93%




Event 14225.1: −1.43% (not significant, p_value = 0.32)
Event 14225.1: −12.39%




Event 14226.1: −2.56% (not significant, p_value = 0.17)
Event 14226.1: −21.10%




Event 14229.1: −0.93% (not significant, p_value = 0.38)
Event 14229.1: −8.23%




Event 14220.1: 2.35% (not significant, p_value = 0.22)
Event 14220.1: 24.38%





“CFU” = Colony Forming Unit






An increase in the resistance of plants to fungal diseases may be achieved by two genes stack expression, which are addressing the same or different mode of action.


Two different combinations of gene stacking were expressed in Brachypodium plants. The combination of LFS57 (disclosed in WO 2018/131037), a gene linked to programmed cell death, and LFSS86 disclosed in the present invention, a gene related to plant defense signaling activation, addresses the effect of stacking two different mode of actions directed to increase resistance.


The combination of LFS46 (disclosed in WO 2018/131037) and LFS84 disclosed in the present invention addresses the effect of stacking two genes coding for enzymes processing the same metabolite involved in defense response signaling therefore increasing the efficacy of the reaction.


Table 13 hereinbelow summarizes the reduction of fungi burden in the transgenic Brachypodium plants comprising the stacked genes infected with Fusarium verticilloides.









TABLE 13







Validation results (stacked genes): reduction of fungi burden in



Brachypodium plant infected with Fusarium verticilloides












Construct
Origin
Origin
% change of logCFU relative to control
% change of CFU


Name
gene 1
gene 2
(p-value)
relative to control





Stack: LFS57 & LFS86
Sorghum
Maize
first assay:
first assay:





Event 13342.1: −11.07% (significant,
Event 13342.1: −66.14%





p_value = 0.00)
Event 13345.1: −41.40%





Event 13345.1: −5.46% (significant,
Event 13339.1: −74.12%





p_value = 0.04)
Event 13344.1: 93.66%





Event 13339.1: −13.82% (significant,
Event 13331.1: 3.85%





p_value = 0.00)
Event 13340.1: −56.20%





Event 13344.1: 6.76% (significant,






p_value = 0.01)






Event 13331.1: 0.39% (not significant,






p_value = 0.45)






Event 13340.1: −8.44% (significant,






p value = 0.00)






second assay:
second assay:





Event 13342.1: −2.88% (not significant,
Event 13342.1: −23.99%





p_value = 0.13)
Event 13345.1: −22.59%





Event 13345.1: −2.69% (not significant,
Event 13339.1: −30.57%





p_value = 0.15)






Event 13339.1: −3.83% (significant,






p_value = 0.07)



Stack: LFS46 & LFS84
Sorghum
Wheat
first assay:
first assay:





Event 13859.1: −7.47% (significant,
Event 13859.1: −52.31%





p_value = 0.01)
Event 13852.1: −0.79%





Event 13852.1: −0.08% (not significant,
Event 13848.1: −26 99%





p_value = 0.49)






Event 13848.1: −3.18% (not significant,






p_value = 0.17)
Event 13850.1: −72.23%





Event 13850.1: −12.93% (significant,
Event 13851.1: 53.34%





p_value = 0.00)






Event 13851.1: 4.31% (not significant,






p_value = 0.11)






second assay:
second assay:





Event 13859.1: −4.10% (not significant,
Event 13859.1: −31.75%





p_value = 0.13)
Event 13850.1: −74.82%





Event 13850.1: −14.82% (significant,






p_value = 0.00)





“CFU” = Colony Forming Unit






Example 8: Overexpression of a Polypeptide by Genome Editing

Over-expression of a polypeptide according to certain embodiments of the present invention can be achieved using methods of gene editing. One example of such approach includes editing a selected genomic region as to express the polypeptide of interest. In the current example, the target genomic region is the maize locus GRMZM2G069095 (based on genome version Zea mays AGPv3) and the polypeptide to be over-expressed is the maize LFS127_H4 comprising the amino acid sequence set forth in SEQ ID NO:495 encoded by the polynucleotide having the nucleic acid sequence set forth in SEQ ID NO:289. It is to be explicitly understood that other genome loci can be used as targets for genome editing for over-expressing other polypeptides of the invention based on the same principles.



FIG. 2A depicts the sequence of the endogenous 5′ upstream flanking region of the genomic sequence GRMZM2G069095 (SEQ ID NO:12) and FIG. 2B depicts the sequence of the endogenous 3′-downstream flanking region of this genomic locus (SEQ ID NO:13). FIG. 2C depicts the sequence of the 5′-UTR gRNA (SEQ ID NO:10) and FIG. 2D depicts the sequence of the 5′-UTR gRNA without NGG nucleotides following the 3 nucleotides after the Cas9 cutting (SEQ ID NO:14). FIG. 2E depicts the sequence of the 3′-UTR gRNA (SEQ ID NO:11) and FIG. 2F depicts the sequence of the 3′-UTR gRNA after cut (SEQ ID NO:15). FIG. 2G depicts the coding sequence (from the “ATG” start codon to the “TAG” termination codon, marked by bold and underlined) of the desired LFS127_H4 sequence (SEQ ID NO:289) encoding the polypeptide set forth by SEQ ID NO:495.


The complete exemplary repair template (SEQ ID NO:16) is depicted in FIG. 2H. The repair template includes: (1) the upstream flanking region (1 kbp) sequence including part of the gRNA after cutting (SEQ ID NO:14; shown in bold and italics); (2) 5′ UTR of genomic DNA from Cas9 cutting site to ATG; (3) the coding sequence (CDS) of the desired LFS127_H4 sequence (SEQ ID NO:17) marked in lower case with the start (ATG) and the stop (TGA) codons marked in bold and underlined; (4) 3′ UTR of genomic DNA from the stop codon to Cas9 cutting site including the predicted part of the gRNA after cutting (SEQ ID NO:15), shown in bold and underlined and (5) the downstream flanking region (1kbp) sequence.


The repair template is delivered into the cell type of interest along with the 5′ and 3′guide RNA sequences (SEQ ID NO: 10 and SEQ ID NO:11, respectively).


Example 9: Knock Out of a Polypeptide by Genome Editing

Knock-out of a polypeptide according to certain embodiments of the present invention can be achieved using methods of gene editing.


In the current example, the target genomic region is the maize (Zea mays) LFS132 comprising the amino acid sequence set forth in SEQ ID NO:348 encoded by the polynucleotide having the nucleic acid sequence set forth in SEQ ID NO:62. It is to be explicitly understood that other genome loci can be used as targets for genome editing for over-expressing other polypeptides of the invention based on the same principles.


The Crisper/CAS9 or similar systems can generate double stranded breaks (DSBs) at any genomic locus under the guidance of an engineered single-guide RNA when delivered into the cell type of interest. Non-homologous end-joining (NHEJ) in the absence of DNA template accompanied with the modification of target genomic repair the DSBs but tends to be prone to insertion and/or deletion (indel) mutations at the junctional site therefore causing framshifts mutations that disrupt the targeted gene. (FIG. 3E)



FIG. 3A depicts the sequence of the KO gRNA (SEQ ID NO:51); FIG. 3C depicts the coding sequence (from the “ATG” start codon to the “TGA” termination codon, marked by bold and underlined) of the desired LFS132 sequence (SEQ ID NO:53); FIG. 3B depicts the sequence of the KO gRNA after cut (SEQ ID NO:52); FIG. 3D (targeted region in bold) and FIG. 3E depict the anticipated change in the coding sequence of the exemplified KO gene (SEQ ID NO:54).


Section II: Bacterial Genes Associated with Fungal Resistance
Example 10: Isolating Soil Bacteria with Potential Antifungal Activity

Bacteria were isolated from soil in five different locations in Israel with agriculture history for Fusaria spp. related diseases, by a two-layer functional assay against Fusarium verticillioides (Fv). In brief, fungal spores were plated on PDA agar medium with a thin R2A medium (Reasoner D J & Geldreich A new medium for the enumeration and subculture of bacteria from potable water. Appl Environ Microbiol 1985 49:1-7) overlay containing different dilutions of soil washes. Active bacteria were isolated from lytic zones and further characterized for the antifungal activity in dual-culture confrontation assays against Fv on R2A agar plates. Table 14 presents a list of the bacteria isolated. In parenthesis is the percentage of similarity between the isolate and the known genus or species.









TABLE 14







Bacterial isolates having antifungal activity








Isolate
Taxa Isolate


No.
(Percentage of similarity)











1

Arthrobacter_globiformis (31.4)



2

Bacillus (95.0)



3

Pseudomonas (66.2)



4

Erwinia (17.8)



5

Arthrobacter (33.1)



6

Bacillus (75.5)



7

Bacillus (94.8)



8

Bacillus (94.8)



9

Pseudomonas (43.6) | Massilia (45.6)



10

Pseudomonas (43.6) |Massilia (45.6)



11

Pseudomonas (44.0)



12

Pseudomonas (61.4)



13

Pseudomonas (61.4)



14

Pseudomonas (61.4)



15

Pseudomonas (52.7) |Bacillus subtilis (80.1)



16

Bacillus (78.4)



17

Bacillus (95.2)



18

Bacillus (95.1)



19

Bacillus (93.5)



20

Pseudomonas (50.6)



21

Erwinia (17.9)



22

Lysobacter_capsici (85.2) | Erwinia (18.0)



23

Arthrobacter (36.1)



24

Arthrobacter_globiformis (29.2)



25

Pseudomonas (63.8) | Lysobacter (22.5)



26

Pseudomonas (63.8) | Lysobacter (22.5)



27

Pseudomonas (59.2)



28

Pseudomonas (59.2)



29

Pseudomonas (59.2)



30

Chryseobacterium_daeguense (28.1)



31

Chryseobacterium_daeguense (28.1)



32

Pseudomonas (44.0)



33

Pseudomonas (44.0)



34

Pseudomonas (61.4)



35

Pseudomonas (60.9)



36

Pseudomonas (60.9)



37

Pseudomonas (58.9) | Bacillus (77.2)



38

Bacillus (74.0)



39

Arthrobacter (19.8)



40

Pantoea_agglomerans (35.7)



41

Curtobaderium_UNCCL17(82.3) | Pantoea_agglomerans




(91.6)


42

Pantoea (83.6)



43

Pantoea (83.6)



44

Pseudomonas alcaligenes (38.6)



45

Pseudomonas (50.2)



46

Pseudomonas (41.2)



47

Pseudomonas (85.4)



48

Leifsonia (11.9) | Clavibacter michiganensis (9.3)



49

Microbacterium (19.8)



50

Microbacterium (19.8)



51

Pseudomonas (60.1)



52

Bacillus (94.5)



53

Pseudomonas s (50.1)



54

Pseudomonas (40.5)



55

Pseudomonas (40.5)



56

Pseudomonas (59.1)



57

Pseudomonas s (59.1)



58

Bacillus (92.4)



59

Pseudomonas (44.0)



60

Bacillus (92.4)



61

Pseudomonas (44.0)



62

Pseudomonas (56.0)



63

Bacillus (95.0)



64

Bacillus (74.0)



65

Pseudomonas (41.1)



66

Pseudomonas (41.1)



67

Pseudomonas (41.1)



68

Pseudomonas (60.1)



69

Pseudomonas (60.1)



70

Pseudomonas (60.1)



71

Pantoea (100);



72

Bacillus (100);



73

Arthrobacter (100); unclassified;



74

Paenibacillus (100); unclassified;



75

Paenibacillus (100); unclassified;



76

Pseudomonas (100); Pseudomonas frederiksbergensis (98)



77

Erwinia (91); unclassified;



78

Bacillus (100)



79

Pseudomonas chloraphis



80

Alcaligenaceae (100); unclassified



81

Bacillus_axarquiensis



82

Pseudomonas spp.



83

Pantoea agglomerans



84

Chryseobacterium_ginsenosidimutans



85

Bacillus_altitudinis










Example 11: Identifying Fungicidal Genes Eligible for Disease Control

The present inventors have identified 199 polynucleotides of bacterial origin that encode for proteins with potential antifungal activity. The antifungal activity of successfully purified soluble proteins was tested in an in vitro spore assay against Fusarium verticilloides. The antifungal activity is further examined by expressing the bacterial genes in planta, and evaluating fungal resistance conferred by the exogenous polynucleotides.


The polynucleotides and polypeptides of some embodiments of the invention having the antifungal activity originate from bacterial isolate proprietary genomes as listed in Table 14 and public bacterial genomes that were incorporated into a unified database, also containing gene expression data produced ad hoc, as well as gene phylogeny, protein annotation, enzymatic function and pathway.


Genomic and Transcriptomic Profiling of Bacterial Isolates for Gene Discovery
Genomic Profiling

Genome assembly—genomes of all isolates were assembled using a proprietary pipeline.


Gene prediction—gene prediction was performed using Prokaryotic Dynamic Programming Genefinding Algorithm [Prodigal—BMC Bioinformatics. 2010 Mar. 8; 11(1):119].


Gene annotation—Predicted genes and proteins were annotated using BLAST™ search [blast.ncbi.nlm.nih.gov/Blast.cgi] against NCBI nr (non-redundant protein sequence database) and by further analysis by InterPro [ebi.ac.uk/interpro/].


Transcriptome Profiling

Eleven (11) bacterial strains (Serial Number 71, 72, 73, 74, 75, 77, 78, 79, 80, 83 of Table 14) were striped on a R2A medium in Petri dishes on the two opposite sides of a Fusarium verticillioides strip culture. Bacterial samples were collected after 3 days of co-culture after a clear inhibition zone was defined and the collected bacteria were used for transcriptomics analysis. Total RNA was extracted and submitted to Omega Bioservices contract lab for RNA sequencing. Transcriptomics data were evaluated for identification of highly expressed potential antifungal genes.


Differential Expression Data Analysis

RNA-seq uses next-generation sequencing (NGS) methods to sequence cDNA that has been derived from an RNA sample, and hence produces millions of short reads. These reads were then mapped onto a reference genome and the number of reads mapping within a genomic feature of interest (such as a gene) was used as a measure of the abundance of the feature in the analyzed sample, resulting in gene expression profile. According to the gene expression profile, a differential analysis was performed to identify genes with higher expression during fungal co-culture compared to pure bacteria cultures that are further associated with antifungal phenotypes.


Identification of Antifungal Genes from Bacterial Isolates


The 199 genes listed in Table 15 below were identified as potential antifungal and may confer fungal-resistance traits in planta. The Table shows the identified genes, their curated polynucleotide and polypeptide sequences.









TABLE 15







Identified antifungal genes from bacterial isolates














Polyn.
Polyp.


Gene


SEQ ID
SEQ ID


Name
Gene Description
Isolate Name
NO
NO














BLFS1
antifungal protein

Streptomyces clavuligerus ATCC

592
2444




27064




BLFS2
Hypothetical protein
15627EM_META_Alkanindiges1|
593
2445


BLFS3
Hypothetical protein
15627EM_META_
594
2446





Enterococcus10|16v1|scaffold580442







G95




BLFS4
Hypothetical protein
15627EM_META_Klebsiella4
595
2447


BLFS5
Hypothetical protein

Actinosporangium_NRRL_B_3428

596
2448


BLFS6
Hypothetical protein

Actinosynnema mirum

597
2449


BLFS7
peptidase C11

Ammonifex_degensii_KC4

598
2450



clostripain





BLFS8
Hypothetical protein

Amycolatopsis azurea

599
2451


BLFS9
oxidoreductase

Amycolatopsis azurea

600
2452


BLFS10
oxidoreductase

Amycolatopsis azurea DSM 43854

601
2453


BLFS11
Hypothetical protein

Arthrobacter spp.

602
2454


BLFS12
Hypothetical protein

Arthrobacter spp.

603
2455


BLFS13
Hypothetical protein

Arthrobacter spp.

604
2456


BLFS14
alpha integrin

Arthrobacter spp.

605
2457


BLFS15
alpha integrin

Arthrobacter_161MFSha2_1

606
2458


BLFS16
lysozyme

Arthrobacter_161MFSha2_1

607
2459


BLFS17
D-alanyl-D-alanine

Arthrobacter_161MFSha2_1

608
2460



carboxypeptidase





BLFS18
Hypothetical protein

Bacillus spp.

609
2461


BLFS19
peptidase p60

Bacillus spp.

610
2462


BLFS20
Cell division

Bacillus spp.

611
2463



suppressor protein





BLFS21
spoivb peptidase

Bacillus spp.

612
2464


BLFS22
Hypothetical protein

Bacillus spp.

613
2465


BLFS23
glycosyl hydrolase

Bacillus spp.

614
2466



family 18





BLFS24
Hypothetical protein

Bacillus spp.

615
2467


BLFS25
chitooligosaccharide

Bacillus spp.

616
2468



deacetylase





BLFS26
Hypothetical protein

Bacillus spp.

617
2469


BLFS27
large protein of pyocin

Bacillus spp.

618
2470



ap41





BLFS29
Hypothetical protein

Bacillus_marmarensis_DSM_21297

619
2471


BLFS30
epsilon-toxin type b

Bacillus_thuringiensis_serovar_indiana

620
2472


BLFS31
Hypothetical protein

Bacillus velezensis

621
2473


BLFS32
Protease HtpX [htpX].

Bacillus velezensis

622
2474


BLFS33
zinc metalloprotease

Bacillus velezensis

623
2475


BLFS34
Cell division

Bacillus velezensis

624
2476



suppressor protein





BLFS35
Hypothetical protein

Bradyrhizobium japonicum

625
2477


BLFS36
Hypothetical protein

Bradyrhizobium yuanmingense

626
2478


BLFS37
beta/gamma crystallin

Brevibacillus laterosporus

627
2479



family protein





BLFS38
jacalin-like lectin

Brevibacillus laterosporus

628
2480



domain protein





BLFS39
toxin ETX

Brevibacillus laterosporus

629
2481


BLFS41
chitobiase

Burkholderia gladioli

630
2482


BLFS42
chitin-binding protein

Burkholderia gladioli

631
2483


BLFS43
carboxypeptidase

Burkholderia gladioli

632
2484



regulatory-like domain






protein





BLFS44
ricin-type beta-trefoil

Burkholderia gladioli

633
2485



lectin domain protein





BLFS45
peptidase

Burkholderia gladioli

634
2486


BLFS46
lysm domain protein

Burkholderia gladioli

635
2487


BLFS47
Hypothetical protein

Cellulosimicrobium spp.

636
2488


BLFS48
peptidase m48

Collimonas arenae

637
2489


BLFS49
Hypothetical protein

Collimonas arenae

638
2490


BLFS50
chitin-binding protein

Collimonas_arenae

639
2491


BLFS51
endoglucanase

Collimonas_arenae

640
2492


BLFS52
Ricin B lectin

Dactylosporangium aurantiacum

641
2493


BLFS53
Etx/MtX1 toxin

Dickeya_dadantii

642
2494


BLFS55
beta/gamma crystallin

Duganella_HH101

643
2495



family protein





BLFS56
aminopeptidase

Duganella_HH101

644
2496


BLFS57
glycoside hydrolase

Duganella_HH101

645
2497



family 12





BLFS58
17 kda surface antigen

Duganella_HH101

646
2498


BLFS59
Hypothetical protein

Duganella_violaceinigra

647
2499


BLFS60
Hypothetical protein

Erwinia spp.

648
2500


BLFS61
type i secretion target

Erwinia spp.

649
2501


BLFS62
murein hydrolase

Erwinia spp.

650
2502



activator nlpd





BLFS63
n-acetylmuramoyl-1-

Erwinia spp.

651
2503



alanine amidase amib





BLFS64
NLP/P60 protein

Geobacillus_Y4_1MC1

652
2504


BLFS65
extracellular repeat

Janthinobacterium_HH010

653
2505



containing protein






HAF family





BLFS67
Beta/gamma crystallin

Janthinobacterium_HH103

654
2506


BLFS68
putative exported

Janthinobacterium MP5059B

655
2507



protein





BLFS69
heme adhesion protein

Janthinobacterium_Marseille

656
2508


BLFS70
Hypothetical protein

Janthinobacterium_agaricidamnosum

657
2509


BLFS72
Carboxypeptidase

Lysobacter spp.

658
2510


BLFS73
Hypothetical protein

Paenibacillus spp.

659
2511


BLFS74
m1-852

Paenibacillus spp.

660
2512


BLFS75
Serine hydrolase

Paenibacillus spp.

661
2513


BLFS76
Hypothetical protein

Paenibacillus spp.

662
2514


BLFS77
glycoside hydrolase

Paenibacillus spp.

663
2515



family 5





BLFS79
Hypothetical protein

Paenibacillus spp.

664
2516


BLFS80
Hypothetical protein

Paenibacillus spp.

665
2517


BLFS82
sspe

Paenibacillus spp.

666
2518


BLFS83
exoglucanase a

Paenibacillus spp.

667
2519


BLFS84
alpha/beta hydrolase

Paenibacillus spp.

668
2520


BLFS85
alpha-amylase

Paenibacillus spp.

669
2521


BLFS86
hypothetical protein

Paenibacillus spp.

670
2522


BLFS87
endoglucanase b

Paenibacillus spp.

671
2523


BLFS88
Hypothetical protein

Paenibacillus spp.

672
2524


BLFS89
endoglucanase

Paenibacillus spp.

673
2525


BLFS90
wgr domain-

Paenibacillus spp.

674
2526



containing protein





BLFS91
Hypothetical protein

Paenibacillus spp.

675
2527


BLFS92
spoivb peptidase

Paenibacillus spp.

676
2528


BLFS93
arabinogalactan endo-

Paenibacillus spp.

677
2529



1,4-beta-galactosidase





BLFS94
rhamnogalacturonan

Paenibacillus spp.

678
2530



endolyase





BLFS95
Hypothetical protein

Paenibacillus spp.

679
2531


BLFS96
Hypothetical protein

Paenibacillus curdlanolyticus

680
2532


BLFS97
Hypothetical protein

Paenibacillus mucilaginosus

681
2533


BLFS98
Hypothetical protein

Pantoea spp.

682
2534


BLFS99
uncharacterized

Pantoea spp.

683
2535



protein yhcn





BLFS100
uncharacterized

Pantoea spp.

684
2536



protein yhcn





BLFS101
metalloprotease loiP

Pantoea spp.

685
2537


BLFS102
Hypothetical protein

Pantoea spp.

686
2538


BLFS103
toxin tccc3

Pantoea spp.

687
2539


BLFS104
Hypothetical protein

Pantoea spp.

688
2540


BLFS105
protein of unknown

Pantoea spp.

689
2541



function (DUF1311}





BLFS106
uncharacterized

Pantoea spp.

690
2542



protein yqjd





BLFS107
polyketide synthase

Pantoea spp.

691
2543


BLFS108
toxin relE

Pantoea spp.

692
2544


BLFS109
hemolysin

Pantoea spp.

693
2545


BLFS110
hypothetical protein

Pedobacter V48

694
2546


BLFS111
hypothetical protein

Pseudogulbenkiania_ferrooxidans

695
2547




EGD HP2




BLFS112
Hypothetical protein

Pseudomonas spp.

696
2548


BLFS113
transpeptidase

Pseudomonas spp

697
2549


BLFS114
ATP-dependent zinc

Pseudomonas spp

698
2550



protease





BLFS115
probable periplasmic

Pseudomonas spp

699
2551



serine endoprotease






DegP-like





BLFS116
lysophospholipase

Pseudomonas spp

700
2552


BLFS118
peptidoglycan-binding

Pseudomonas spp

701
2553



protein





BLFS119
Hypothetical protein

Pseudomonas spp

702
2554


BLFS120
supernuclease toxin 2

Pseudomonas spp.

703
2555



family protein





BLFS121
glutamate

Pseudomonas spp

704
2556



carboxypeptidase





BLFS122
hpr

Pseudomonas spp

705
2557


BLFS123
peptidase c1

Pseudomonas spp

706
2558


BLFS124
tigr02448 family

Pseudomonas spp

707
2559



protein





BLFS125
peptidase

Pseudomonas spp

708
2560


BLFS126
Hypothetical protein

Pseudomonas spp

709
2561


BLFS127
Hypothetical protein

Pseudomonas spp

710
2562


BLFS128
peptidoglycan

Pseudomonas spp

711
2563



hydrolase flgj





BLFS129
alginate lyase

Pseudomonas spp

712
2564


BLFS130
leucine-rich repeat-

Pseudomonas spp..

713
2565



containing protein





BLFS131
uncharacterized

Pseudomonas chlororaphis

714
2566



protein ycfj





BLFS132
Hypothetical protein

Pseudomonas chlororaphis

715
2567


BLFS133
Hypothetical protein

Pseudomonas chlororaphis

716
2568


BLFS134
Protein of unknown

Pseudomonas chlororaphis

717
2569



function DUF2782





BLFS135
multidrug transporter

Pseudomonas chlororaphis

718
2570


BLFS136
Hypothetical protein

Pseudoxanthomonas J31

719
2571


BLFS137
membrane protein

Rhodococcus KB6

720
2572


BLFS138
uncharacterized

Salinibacillus spp.

721
2573



peptidase





BLFS139
peptidoglycan

Salinibacillus spp.

722
2574



endopeptidase





BLFS140
uncharacterized

Salinibacillus spp.

723
2575



lipoprotein ybbd





BLFS141
mannan endo-1,4-beta-

Salinibacillus spp.

724
2576



mannosidase





BLFS142
uncharacterized

Salinibacillus spp.

725
2577



protein ykvt





BLFS143
putative signal peptide

Salinibacillus spp.

726
2578



peptidase sppa





BLFS144
thermolysin

Salinibacillus spp.

727
2579


BLFS145
d-alanyl-d-alanine

Salinibacillus spp.

728
2580



carboxypeptidase dacb





BLFS146
l-ala--d-glu

Salinibacillus spp.

729
2581



endopeptidase





BLFS147
beta-n-

Salinibacillus spp.

730
2582



acetylglucosaminidase





BLFS148
metalloprotease loip

Serratia_fonticola_AU_P3

731
2583


BLFS149
chitobiase

Serratia_fonticola_LMG_7882

732
2584


BLFS150
lytic enzyme

Stenotrophomonas_maltophilia_ATCC_19867

733
2585


BLFS151
peptidase S33 family

Stenotrophomonas_maltophilia_S028

734
2586



protein





BLFS152
lipase

Streptomyces_NRRL_F_4335

735
2587


BLFS153
peptidase

Streptomyces albus

736
2588


BLFS154
peptidase s1

Streptomyces_clavuligerus_ATCC_27064

737
2589


BLFS155
mannose-binding

Streptomyces griseorubens

738
2590



protein





BLFS156
hypothetical protein

Streptomyces_mobaraensis_NBRC_13819

739
2591


BLFS157
hypothetical protein

Thalassobacter_arenae_DSM_19593

740
2592


BLFS158
bicupin, oxalate

Virgibacillus_Vm_5

741
2593



decarboxylase family





BLFS159
large Ala/Gln-rich

Xanthomonas_hortorum_pv_carotae_M081

742
2594



protein





PUB6
Hypothetical protein

Bacillus_weihenstephanensis

743
2595


PUB23
Hypothetical protein

Bacillus cereus

744
2596


PUB31
Hypothetical protein

Bacillus_thuringiensis_serovar_andalousiensis

745
2597


PUB65
putative extracellular

Variovorax_paradoxus_B4

746
2598



carbohydrate-






recognition protein





ZLFS1
Arthropod defensin

Actinomyces_oral_taxon_171

747
2599


ZLFS2
peptidase m14

Arthrobacter spp.

748
2600


ZLFS3
probable

Bacillus_amyloliquefaciens_plantarum

749
2601



peptidoglycan






endopeptidase lyte





ZLFS5
Hypothetical protein

Burkholderia_A1

750
2602


ZLFS6
type vi secretion

Collimonas_fungivorans_Ter331

751
2603



protein





ZLFS7
glycosyl hydrolase

Collimonas_fungivorans_Ter331

752
2604


ZLFS8
chitin-binding protein

Collimonas_fungivorans_Ter331

753
2605


ZLFS9
polyketide synthase-

Collimonas_fungivorans_Ter331

754
2606



like protein





ZLFS10
Hypothetical protein

Collimonas_fungivorans_Ter331

755
2607


ZLFS11
patatin

Collimonas_fungivorans_Ter331

756
2608


ZLFS12
pseudomonalisin

Collimonas_fungivorans_Ter331

757
2609


ZLFS13
Hypothetical protein

Collimonas_fungivorans_Ter331

758
2610


ZLFS14
Hypothetical protein

Collimonas_fungivorans_Ter331

759
2611


ZLFS15
Beta-barrel assembly-

Collimonas_fungivorans_Ter331

760
2612



enhancing protease






[bepA].





ZLFS16
d-alanyl-d-alanine

Collimonas_fungivorans_Ter331

761
2613



carboxypeptidase dacd





ZLFS17
Antimicrobial protein

Herbidospora cretacea

762
2614



MiAMP1





ZLFS18
peptidoglycan-binding

Lysobacter enzymogenes

763
2615



protein





ZLFS19
lipoprotein nlpd/lppb

Lysobacter gummosus

764
2616



homolog





ZLFS21
chitosanase

Paenibacillus spp.

765
2617


ZLFS22
Hypothetical protein

Paenibacillus spp.

766
2618


ZLFS23
beta-glucanase

Pantoea spp.

767
2619


ZLFS24
lipase 1

Pantoea spp.

768
2620


ZLFS25
SGNH hydrolase-type

Pantoea spp.

769
2621



esterase domain





ZLFS27
lytic transglycosylase

Pseudomonas_In5

770
2622


ZLFS28
hydrolase

Pseudomonas_In5

771
2623


ZLFS29
Beta-barrel assembly-

Pseudomonas_In5

772
2624



enhancing protease






[bepA].





ZLFS30
dipeptidase

Pseudomonas_In5

773
2625


ZLFS31
lipase 1

Serratia_plymuthica_S13

774
2626


ZLFS32
endoglucanase

Serratia_plymuthica_S13

775
2627


ZLFS33
chitin-binding protein

Serratia_plymuthica_S13

776
2628


ZLFS34
peptidyl-dipeptidase

Serratia_plymuthica_S13

777
2629



dcp





ZLFS35
Hypothetical protein

Serratia_plymuthica_S13

778
2630


ZLFS36
Calcium-mediated

Serratia_plymuthica_S13

779
2631



lectin





ZLFS37
upf0339 protein yegp

Serratia_plymuthica_S13

780
2632


ZLFS38
bacterioferritin

Serratia_plymuthica_S13

781
2633


ZLFS39
Glycogen debranching

Serratia_plymuthica_S13

782
2634



enzyme [glgX].





ZLFS42
killer toxin like

Streptomyces_HPH0547

783
2635


ZLFS44
glycoside hydrolase

Streptomyces_M10

784
2636


ZLFS45
Oxidoreductase

Streptomyces_M10

785
2637


ZLFS47
Antimicrobial protein

Streptomyces_scopuliridis_RB72

786
2638



MiAMP1





ZLFS48
Antimicrobial protein

Streptosporangium_amethystogenes

787
2639



MiAMP1





ZLFS49
Hypothetical protein

Curtobacterium spp.

788
2640


ZLFS51
Glycosyl hydrolase

Bacillus_subtilis_subtilis_OH_131_1

789
2641





“polyn.” = polynucleotide;


“polyp.” = polypeptide.






Example 12: Identification of Orthologous Sequences of Antifungal Proteins

Orthologues and paralogues constitute two major types of homologues: The first evolved from a common ancestor by specialization, and the latter are related by duplication events. It is assumed that paralogues arising from ancient duplication events are likely to have diverged in function while true orthologues are more likely to retain identical function over evolutionary time. Orthologues of the discovered antifungal genes are not only likely to be antifungal by themselves but also may hold improved potency or target different fungi spectra.


The search and identification of homologous genes involves the screening of sequence information available, for example, public databases such as the GenBank, and the European Molecular Biology Laboratory Nucleic Acid Sequence Database (EMBL).


Polynucleotides and polypeptides with significant homology to the identified genes described in Table 15 (Example 11) were identified from the databases using BLAST™ software with the Blastp and tBlastn algorithms as filters for the first stage, and the needle (EMBOSS package) or Frame+algorithm alignment for the second stage. Local identity (BLAST™ alignments) was defined with a very permissive cutoff—40% Identity on a span of 40% of the sequences lengths because it is used only as a filter for the global alignment stage. The default filtering of the BLAST™ package was not utilized (by setting the parameter “−F F”).


In the second stage, homologs were defined based on a global identity of at least 70% to the core gene polypeptide sequence. Two distinct forms for finding the optimal global alignment for protein or nucleotide sequences were used in this application:


1.Between two proteins (following the BLASTP filter):


EMBOSS-6.0.1 Needleman-Wunsch algorithm with the following modified parameters: gapopen=8 gapextend=2. The rest of the parameters were unchanged from the default options described hereinabove.


2.Between a protein sequence and a nucleotide sequence (following the TBLASTN filter): GenCore 6.0 OneModel application utilizing the Frame+algorithm with the following parameters: model=frame+_p2n.model mode=qglobal -q=protein.sequence -db=nucleotide.sequence. The rest of the parameters are unchanged from the default options described hereinabove.


Homology was calculated as % of identity over the aligned sequences. The query sequences were the polypeptide sequences depicted in Table 15 (Example 11). The subject sequences are protein sequences identified in the database based on greater than 80% global identity to the predicted translated sequences of the query nucleotide sequences or to the polypeptide sequences. The identified orthologous and homologous sequences having at least 70% global sequence identity to the sequences are provided in Table 16, below.









TABLE 16







Homologues (e.g., orthologues) of the identified antifungal genes/polypeptides













Polyn.
Hom. to

Polyp.
Hom. to
%



SEQ ID
Gene

SEQ ID
SEQ ID
glob.



NO
Name
Organism
NO
NO
Iden.
Algor.
















1187
BLFS87

Paenibacilhis sp.

3024
2523
96.2
globlastp


1188
BLFS87

Paenibacilhis sp.

3025
2523
95.5
globlastp


1189
BLFS87

Paenibacilhis sp.

3026
2523
94.8
globlastp


1190
BLFS87

Paenibacillus sp.

3027
2523
94.1
globlastp


1191
BLFS87

Paenibacillus sp.

3028
2523
93.5
globlastp


1192
BLFS87

Paenibacillus sp.

3029
2523
92
globlastp


1193
BLFS87

Paenibacillus sp.

3030
2523
91.1
globlastp


1194
BLFS87

Paenibacillus sp.

3031
2523
90.2
globlastp


1195
BLFS87

Paenibacillus sp.

3032
2523
89.4
globlastp


1196
BLFS87

Paenibacillus sp.

3033
2523
88.3
globlastp


1197
BLFS87

Paenibacillus sp.

3034
2523
71
globlastp


2052
ZLFS3

Bacillus sp.

3866
2601
98.5
globlastp


2053
ZLFS3

Bacillus sp.

3867
2601
97.8
globlastp


2054
ZLFS3

Bacillus sp.

3868
2601
97.4
globlastp


2055
ZLFS3

Bacillus sp.

3869
2601
94.9
globlastp


2056
ZLFS3

Bacillus sp.

3870
2601
94.6
globlastp


2057
ZLFS3

Bacillus sp.

3871
2601
94.2
globlastp


2058
ZLFS3

Bacillus sp.

3872
2601
93.8
globlastp


2059
ZLFS3

Bacillus sp.

3873
2601
84.1
globlastp


2060
ZLFS3

Bacillus sp.

3874
2601
83.8
globlastp


2061
ZLFS3

Bacillus sp.

3875
2601
76.1
globlastp


2062
ZLFS3

Bacillus sp.

3876
2601
75.7
globlastp


2063
ZLFS3

Bacillus sp.

3877
2601
75.5
globlastp


2064
ZLFS3

Bacillus sp.

3878
2601
75.4
globlastp


2065
ZLFS3

Bacillus sp.

3879
2601
75.2
globlastp


2066
ZLFS3

Bacillus sp.

3880
2601
74.9
globlastp


2067
ZLFS3

Bacillus sp.

3881
2601
74.5
globlastp


2068
ZLFS3

Bacillus sp.

3882
2601
74.1
globlastp


2069
ZLFS3

Bacillus sp.

3883
2601
73
globlastp


949
BLFS34

Bacillus sp.

2799
2476
99
globlastp


950
BLFS34

Bacillus sp.

2800
2476
98.1
globlastp


951
BLFS34

Bacillus sp.

2801
2476
95.1
globlastp


1013
BLFS56

Janthinobacterium sp.

2859
2496
98.7
globlastp


1014
BLFS56

Oxalobacteraceae sp.

2860
2496
93.2
globlastp


1015
BLFS56

Janthinobacterium sp.

2861
2496
72.5
globlastp


1016
BLFS56

Janthinobacterium sp.

2862
2496
72.2
globlastp


1017
BLFS56

Janthinobacterium sp.

2863
2496
71.6
globlastp


1018
BLFS56

Massilia sp.

2864
2496
71.4
globlastp


1019
BLFS56

Massilia sp.


2496
71.02
glotblastn


1020
BLFS56

Massilia sp.


2496
70.7
glotblastn


1021
BLFS56

Duganella sp.


2496
70.04
glotblastn


1207
BLFS89

Paenibacillus sp.

3044
2525
99.2
globlastp


1208
BLFS89

Paenibacillus sp.

3045
2525
98.7
globlastp


1209
BLFS89

Paenibacillus sp.

3046
2525
97
globlastp


1210
BLFS89

Paenibacillus sp.

3047
2525
96.7
globlastp


1211
BLFS89

Paenibacillus sp.

3048
2525
95
globlastp


1212
BLFS89

Paenibacillus sp.

3049
2525
94.2
globlastp


1213
BLFS89

Paenibacillus sp.

3050
2525
93.4
globlastp


1214
BLFS89

Paenibacillus sp.

3051
2525
92.2
globlastp


1215
BLFS89

Paenibacillus sp.

3052
2525
91.9
globlastp


1216
BLFS89

Paenibacillus sp.


2525
71.86
glotblastn


1217
BLFS89

Paenibacillus sp.


2525
70.07
glotblastn


884
BLFS23

Bacillus sp.

2734
2466
96.4
globlastp


885
BLFS23

Bacillus sp.

2735
2466
95.2
globlastp


886
BLFS23

Bacillus sp.

2736
2466
94
globlastp


887
BLFS23

Bacillus sp.

2737
2466
93.1
globlastp


888
BLFS23

Bacillus sp.

2738
2466
93
globlastp


889
BLFS23

Bacillus sp.

2739
2466
92.4
globlastp


890
BLFS23

Bacillus sp.

2740
2466
92
globlastp


891
BLFS23

Bacillus sp.

2741
2466
91.8
globlastp


892
BLFS23

Bacillus sp.

2742
2466
91.2
globlastp


1062
BLFS64

Geobacillus sp.

2904
2504
98.8
globlastp


2194
ZLFS28

Pseudomonas sp.

4008
2623
94.1
globlastp


2195
ZLFS28

Pseudomonas sp.

4009
2623
93.8
globlastp


2196
ZLFS28

Pseudomonas sp.

4010
2623
92.2
globlastp


2197
ZLFS28

Pseudomonas sp.

4011
2623
91.4
globlastp


2198
ZLFS28

Pseudomonas sp.

4012
2623
89.5
globlastp


2199
ZLFS28

Pseudomonas sp.

4013
2623
88.3
globlastp


2200
ZLFS28

Pseudomonas sp.

4014
2623
87.9
globlastp


2201
ZLFS28

Pseudomonas sp.

4015
2623
87.5
globlastp


2202
ZLFS28

Pseudomonas sp.

4016
2623
86.7
globlastp


2203
ZLFS28

Pseudomonas sp.

4017
2623
86.3
globlastp


2204
ZLFS28

Pseudomonas sp.

4018
2623
85.9
globlastp


2205
ZLFS28

Pseudomonas sp.

4019
2623
85.5
globlastp


2206
ZLFS28

Pseudomonas sp.

4020
2623
82.7
globlastp


2207
ZLFS28

Pseudomonas sp.

4021
2623
80.1
globlastp


2208
ZLFS28

Collimonas sp.

4022
2623
72.1
globlastp


908
BLFS29

Bacillus sp.

2758
2471
75.3
globlastp


2104
ZLFS19
Environmental sample
3918
2616
84.6
globlastp


2105
ZLFS19

Lysobacter sp.

3919
2616
84.3
globlastp


2106
ZLFS19

Lysobacter sp.

3920
2616
83.9
globlastp


2107
ZLFS19

Lysobacter sp.

3921
2616
83.4
globlastp


2108
ZLFS19

Lysobacter sp.

3922
2616
82.4
globlastp


2109
ZLFS19
Environmental sample
3923
2616
81.9
globlastp


2110
ZLFS19
Environmental sample
3924
2616
81.5
globlastp


2111
ZLFS19
Environmental sample
3925
2616
80.6
globlastp


2112
ZLFS19

Lysobacter sp.

3926
2616
79.7
globlastp


2113
ZLFS19

Lysobacter sp.

3927
2616
76.9
globlastp


845
BLFS18

Bacillus sp.

2697
2461
95.5
globlastp


846
BLFS18

Bacillus sp.

2698
2461
84.6
globlastp


847
BLFS18

Bacillus sp.

2699
2461
82.1
globlastp


848
BLFS18

Bacillus sp.

2700
2461
81.9
globlastp


849
BLFS18

Bacillus sp.

2701
2461
80.1
globlastp


850
BLFS18

Bacillus sp.

2702
2461
78.1
globlastp


851
BLFS18

Bacillus sp.

2703
2461
77
globlastp


852
BLFS18

Bacillus sp.

2704
2461
76.3
globlastp


853
BLFS18

Bacillus sp.

2705
2461
75
globlastp


854
BLFS18

Bacillus sp.

2706
2461
74.2
globlastp


855
BLFS18

Bacillus sp.

2707
2461
73
globlastp


856
BLFS18

Kurthia sp.

2708
2461
72
globlastp


857
BLFS18

Bacillus sp.

2709
2461
71
globlastp


858
BLFS18

Anoxybacillus sp.

2710
2461
70.3
globlastp


1997
BLFS155

Streptomyces sp.

3811
2590
88.1
globlastp


1998
BLFS155

Streptomyces sp.

3812
2590
70.2
globlastp


592
BLFS156

Streptomyces sp.


2591
71.07
glotblastn


1696
BLFS130

Pseudomonas sp.

3520
2565
98.8
globlastp


1697
BLFS130

Pseudomonas sp.

3521
2565
93.9
globlastp


1698
BLFS130

Pseudomonas sp.

3522
2565
81.2
globlastp


1699
BLFS130

Pseudomonas sp.

3523
2565
72.9
globlastp


1700
BLFS130

Pseudomonas sp.

3524
2565
70
globlastp


2426
ZLFS51

Bacillus sp.

4235
2641
99.1
globlastp


2427
ZLFS51

Bacillus sp.

4236
2641
98.1
globlastp


2428
ZLFS51

Bacillus sp.

4237
2641
97.2
globlastp


2429
ZLFS51

Bacillus sp.

4238
2641
96
globlastp


2430
ZLFS51

Salinibacillus sp.

4239
2641
95.3
globlastp


2431
ZLFS51

Bacillus sp.

4240
2641
94.6
globlastp


2432
ZLFS51

Bacillus sp.

4241
2641
93.9
globlastp


2433
ZLFS51

Bacillus sp.

4242
2641
87.6
globlastp


2434
ZLFS51

Bacillus sp.

4243
2641
86.2
globlastp


2435
ZLFS51

Bacillus sp.

4244
2641
85.7
globlastp


2436
ZLFS51

Bacillus sp.

4245
2641
78
globlastp


2437
ZLFS51

Bacillus sp.

4246
2641
77.1
globlastp


2438
ZLFS51

Bacillus sp.

4247
2641
76
globlastp


2439
ZLFS51

Bacillus sp.

4248
2641
75.2
globlastp


2440
ZLFS51

Bacillus sp.

4249
2641
74
globlastp


2441
ZLFS51

Bacillus sp.

4250
2641
73.8
globlastp


2442
ZLFS51

Bacillus sp.

4251
2641
71.1
globlastp


2443
ZLFS51

Bacillus sp.

4252
2641
70
globlastp


2389
ZLFS42

Streptomyces sp.

4202
2635
80.3
globlastp


2390
ZLFS42

Streptomyces sp.

4203
2635
78.6
globlastp


2391
ZLFS42

Streptomyces sp.

4204
2635
78.6
globlastp


2392
ZLFS42

Streptomyces sp.

4205
2635
77.8
globlastp


785
ZLFS42

Streptomyces sp.

2637
2635
77.8
globlastp


2393
ZLFS42

Streptomyces sp.

4206
2635
76.9
globlastp


2394
ZLFS42

Streptomyces sp.

4207
2635
76.1
globlastp


2395
ZLFS42

Streptomyces sp.

4207
2635
76.1
globlastp


2396
ZLFS42

Streptomyces sp.

4208
2635
75.2
globlastp


2397
ZLFS42

Streptomyces sp.

4209
2635
73.5
globlastp


2398
ZLFS42

Streptomyces sp.

4210
2635
73.5
globlastp


808
ZLFS42

Streptomyces sp.

2660
2635
70.1
globlastp


1792
BLFS136

Pseudoxanthomonas sp.

3616
2571
81
globlastp


1163
BLFS84

Paenibacillus sp.

3000
2520
98.3
globlastp


1164
BLFS84

Paenibacillus sp.

3001
2520
95.9
globlastp


1165
BLFS84

Paenibacillus sp.

3002
2520
95.1
globlastp


1166
BLFS84

Paenibacillus sp.

3003
2520
92.4
globlastp


1167
BLFS84

Paenibacillus sp.

3004
2520
89.5
globlastp


1168
BLFS84

Paenibacillus sp.

3005
2520
88.1
globlastp


1169
BLFS84

Paenibacillus sp.

3006
2520
87.5
globlastp


1129
BLFS79

Paenibacillus sp.

2966
2516
99.2
globlastp


1130
BLFS79

Paenibacillus sp.

2967
2516
98.5
globlastp


1131
BLFS79

Paenibacillus sp.

2968
2516
97
globlastp


1132
BLFS79

Paenibacillus sp.

2969
2516
96.6
globlastp


1133
BLFS79

Paenibacillus sp.

2970
2516
95.1
globlastp


1134
BLFS79

Paenibacillus sp.

2971
2516
94
globlastp


1135
BLFS79

Paenibacillus sp.

2972
2516
93.6
globlastp


1136
BLFS79

Paenibacillus sp.

2973
2516
93.2
globlastp


1137
BLFS79

Paenibacillus sp.

2974
2516
92.5
globlastp


1138
BLFS79

Paenibacillus sp.

2975
2516
91.7
globlastp


2235
ZLFS30

Pseudomonas sp.

4049
2625
94.6
globlastp


2236
ZLFS30

Pseudomonas sp.

4050
2625
90.5
globlastp


2237
ZLFS30

Pseudomonas sp.

4051
2625
89.1
globlastp


2238
ZLFS30

Pseudomonas sp.

4052
2625
88.3
globlastp


2239
ZLFS30

Pseudomonas sp.

4053
2625
87.2
globlastp


2240
ZLFS30

Pseudomonas sp.

4054
2625
86
globlastp


2241
ZLFS30

Pseudomonas sp.

4055
2625
85
globlastp


2242
ZLFS30

Erwinia sp.

4056
2625
84.1
globlastp


2243
ZLFS30

Pseudomonas sp.

4057
2625
83.1
globlastp


2244
ZLFS30

Pseudomonas sp.

4058
2625
82
globlastp


2245
ZLFS30

Pseudomonas sp.

4059
2625
81
globlastp


2246
ZLFS30

Pseudomonas sp.

4060
2625
80.1
globlastp


2247
ZLFS30

Pseudomonas sp.

4061
2625
79.1
globlastp


2248
ZLFS30

Pseudomonas sp.

4062
2625
78.1
globlastp


2249
ZLFS30

Pseudomonas sp.

4063
2625
77.2
globlastp


2250
ZLFS30

Pseudomonas sp.

4064
2625
76.7
globlastp


2251
ZLFS30

Pseudomonas sp.

4065
2625
75.5
globlastp


2252
ZLFS30
Environmental sample
4066
2625
72
globlastp


2253
ZLFS30

Pseudomonas sp.

4067
2625
71
globlastp


2254
ZLFS30
Environmental sample
4068
2625
70.1
globlastp


2033
ZLFS2

Arthrobacter sp.

3847
2600
99.4
globlastp


2034
ZLFS2

Arthrobacter sp.

3848
2600
96.3
globlastp


2035
ZLFS2

Arthrobacter sp.

3849
2600
94.6
globlastp


2036
ZLFS2
Environmental sample
3850
2600
92.7
globlastp


2037
ZLFS2
Environmental sample
3851
2600
92.1
globlastp


2038
ZLFS2

Arthrobacter sp.

3852
2600
90.1
globlastp


2039
ZLFS2

Arthrobacter sp.

3853
2600
89.8
globlastp


2040
ZLFS2
Environmental sample
3854
2600
89.3
globlastp


2041
ZLFS2

Arthrobacter sp.

3855
2600
88.9
globlastp


2042
ZLFS2

Arthrobacter sp.

3856
2600
87.3
globlastp


2043
ZLFS2

Arthrobacter sp.

3857
2600
86.2
globlastp


2044
ZLFS2

Arthrobacter sp.

3858
2600
73.7
globlastp


2045
ZLFS2

Arthrobacter sp.

3859
2600
73.2
globlastp


2046
ZLFS2

Sporosarcina sp.

3860
2600
72.6
globlastp


2047
ZLFS2

Bhargavaea sp.

3861
2600
72.1
globlastp


2048
ZLFS2

Arthrobacter sp.

3862
2600
71.5
globlastp


2049
ZLFS2

Arthrobacter sp.

3863
2600
71.2
globlastp


2050
ZLFS2

Sporosarcina sp.

3864
2600
70.6
globlastp


2051
ZLFS2

Planomicrobium sp.

3865
2600
70.5
globlastp


1071
BLFS72

Lysobacter sp.

2913
2510
95
globlastp


1124
BLFS77

Paenibacillus sp.

2963
2515
98.1
globlastp


1125
BLFS77

Paenibacillus sp.

2964
2515
96.6
globlastp


1126
BLFS77

Paenibacillus sp.

2965
2515
81.1
globlastp


1127
BLFS77

Paenibacillus sp.


2515
73.95
glotblastn


1128
BLFS77

Paenibacillus sp.


2515
73.24
glotblastn


1599
BLFS122

Pseudomonas sp.

3424
2557
97.7
globlastp


1600
BLFS122

Pseudomonas sp.

3425
2557
96.9
globlastp


1601
BLFS122

Pseudomonas sp.

3426
2557
96.2
globlastp


1602
BLFS122

Pseudomonas sp.

3427
2557
93.1
globlastp


1603
BLFS122

Pseudomonas sp.

3428
2557
79.4
globlastp


1604
BLFS122

Pseudomonas sp.


2557
72.18
glotblastn


1605
BLFS122

Pseudomonas sp.

3429
2557
71.1
globlastp


1606
BLFS122

Pseudomonas sp.

3430
2557
70.9
globlastp


970
BLFS38

Brevibacillus sp.

2818
2480
81.2
globlastp


1139
BLFS82

Paenibacillus sp.

2976
2518
99
globlastp


1140
BLFS82

Paenibacillus sp.

2977
2518
98
globlastp


1141
BLFS82

Paenibacillus sp.

2978
2518
97.1
globlastp


1142
BLFS82

Paenibacillus sp.

2979
2518
96.1
globlastp


1143
BLFS82

Paenibacillus sp.

2980
2518
94.1
globlastp


1144
BLFS82

Paenibacillus sp.

2981
2518
93.1
globlastp


1145
BLFS82

Paenibacillus sp.

2982
2518
92.2
globlastp


1146
BLFS82

Paenibacillus sp.

2983
2518
89.4
globlastp


1147
BLFS82

Paenibacillus sp.

2984
2518
88.2
globlastp


1148
BLFS82

Paenibacillus sp.

2985
2518
86.3
globlastp


1149
BLFS82

Paenibacillus sp.

2986
2518
85.3
globlastp


1150
BLFS82

Paenibacillus sp.

2987
2518
84.3
globlastp


1151
BLFS82

Paenibacillus sp.

2988
2518
82.4
globlastp


2075
ZLFS6

Collimonas sp.

3889
2603
95.7
globlastp


2076
ZLFS6

Herbaspirillum sp.

3890
2603
72
globlastp


2077
ZLFS6

Janthinobacterium sp.

3891
2603
71.3
globlastp


2078
ZLFS6

Janthinobacterium sp.

3892
2603
70.7
globlastp


2079
ZLFS6

Oxalobacteraceae sp.

3893
2603
70.6
globlastp


812
BLFS11

Arthrobacter sp.

2664
2454
96.3
globlastp


813
BLFS11

Arthrobacter sp.

2665
2454
80.4
globlastp


814
BLFS11

Arthrobacter sp.

2666
2454
79.9
globlastp


815
BLFS11
Environmental sample
2667
2454
75.7
globlastp


816
BLFS11

Arthrobacter sp.

2668
2454
73
globlastp


817
BLFS11

Arthrobacter sp.

2669
2454
71.2
globlastp


818
BLFS11

Arthrobacter sp.

2670
2454
70.1
globlastp


2114
ZLFS21

Paenibacillus sp.

3928
2617
99.1
globlastp


2115
ZLFS21

Paenibacillus sp.

3929
2617
96.7
globlastp


2116
ZLFS21

Paenibacillus sp.

3930
2617
96.4
globlastp


2117
ZLFS21

Paenibacillus sp.

3931
2617
94.5
globlastp


2118
ZLFS21

Paenibacillus sp.

3932
2617
94.2
globlastp


2119
ZLFS21

Paenibacillus sp.

3933
2617
91.3
globlastp


2120
ZLFS21

Paenibacillus sp.

3934
2617
85
globlastp


2121
ZLFS21

Paenibacillus sp.

3935
2617
84.5
globlastp


2122
ZLFS21

Paenibacillus sp.

3936
2617
84.3
globlastp


2123
ZLFS21

Paenibacillus sp.

3937
2617
82.6
globlastp


2124
ZLFS21

Paenibacillus sp.

3938
2617
81.9
globlastp


2125
ZLFS21

Paenibacillus sp.

3939
2617
81.6
globlastp


2126
ZLFS21

Paenibacillus sp.

3940
2617
72.3
globlastp


839
BLFS17
Multievos46 sp.
2691
2460
96
globlastp


840
BLFS17
Environmental sample
2692
2460
86.4
globlastp


841
BLFS17

Arthrobacter sp.

2693
2460
80
globlastp


842
BLFS17

Arthrobacter sp.

2694
2460
79
globlastp


843
BLFS17

Arthrobacter sp.

2695
2460
78.6
globlastp


844
BLFS17

Arthrobacter sp.

2696
2460
77.5
globlastp


1450
BLFS113

Pseudomonas sp.

3276
2549
99.4
globlastp


1451
BLFS113

Pseudomonas sp.

3277
2549
98.8
globlastp


1452
BLFS113

Pseudomonas sp.

3278
2549
97.6
globlastp


1453
BLFS113

Pseudomonas sp.

3279
2549
97
globlastp


1454
BLFS113

Pseudomonas sp.

3280
2549
96.4
globlastp


1455
BLFS113

Pseudomonas sp.

3281
2549
94.6
globlastp


1456
BLFS113

Pseudomonas sp.

3282
2549
92.2
globlastp


1457
BLFS113

Pseudomonas sp.

3283
2549
91
globlastp


1458
BLFS113

Pseudomonas sp.

3284
2549
90.4
globlastp


1459
BLFS113

Pseudomonas sp.

3285
2549
89.2
globlastp


1460
BLFS113

Pseudomonas sp.

3286
2549
88
globlastp


1461
BLFS113

Erwinia sp.

3287
2549
87.4
globlastp


1462
BLFS113

Pseudomonas sp.

3288
2549
86.2
globlastp


1463
BLFS113

Pseudomonas sp.

3289
2549
85
globlastp


1464
BLFS113

Pseudomonas sp.

3290
2549
84.2
globlastp


1465
BLFS113

Pseudomonas sp.

3291
2549
83
globlastp


1466
BLFS113

Pseudomonas sp.

3292
2549
82
globlastp


1467
BLFS113

Pseudomonas sp.

3293
2549
81.4
globlastp


1468
BLFS113

Pseudomonas sp.

3294
2549
80.1
globlastp


1469
BLFS113

Pseudomonas sp.

3295
2549
79
globlastp


1470
BLFS113

Pseudomonas sp.

3296
2549
78.4
globlastp


1471
BLFS113

Pseudomonas sp.

3297
2549
77.8
globlastp


1472
BLFS113
Environmental sample
3298
2549
76.6
globlastp


1473
BLFS113

Pseudomonas sp.

3299
2549
74.9
globlastp


1474
BLFS113

Pseudomonas sp.

3300
2549
71.4
globlastp


817
BLFS13

Arthrobacter sp.

2669
2456
99.4
globlastp


815
BLFS13
Environmental sample
2667
2456
73.3
globlastp


821
BLFS13

Arthrobacter sp.

2673
2456
71.4
globlastp


822
BLFS13

Arthrobacter sp.

2674
2456
70.3
globlastp


1611
BLFS124

Pseudomonas sp.

3435
2559
99
globlastp


1612
BLFS124

Pseudomonas sp.

3436
2559
93.3
globlastp


1613
BLFS124

Pseudomonas sp.

3437
2559
92.4
globlastp


1614
BLFS124

Pseudomonas sp.

3438
2559
83.8
globlastp


1615
BLFS124

Pseudomonas sp.

3439
2559
82.9
globlastp


1616
BLFS124

Pseudomonas sp.

3440
2559
81
globlastp


1617
BLFS124

Pseudomonas sp.

3441
2559
80
globlastp


1618
BLFS124

Pseudomonas sp.

3442
2559
79
globlastp


1619
BLFS124

Pseudomonas sp.

3443
2559
78.1
globlastp


1620
BLFS124

Pseudomonas sp.

3444
2559
77.3
globlastp


1621
BLFS124
Environmental sample
3445
2559
76.2
globlastp


1622
BLFS124

Pseudomonas sp.

3446
2559
75.2
globlastp


1623
BLFS124

Pseudomonas sp.

3447
2559
74.5
globlastp


1624
BLFS124
Environmental sample
3448
2559
73.3
globlastp


1625
BLFS124
Environmental sample
3449
2559
72.4
globlastp


1626
BLFS124
Environmental sample
3450
2559
71.4
globlastp


1627
BLFS124

Pseudomonas sp.

3451
2559
70.5
globlastp


2147
ZLFS24

Meta sp.

3961
2620
98.8
globlastp


2148
ZLFS24
Environmental sample
3962
2620
98.6
globlastp


2149
ZLFS24
Environmental sample
3963
2620
98.3
globlastp


2150
ZLFS24

Pantoea sp.

3964
2620
98.2
globlastp


2151
ZLFS24
Environmental sample
3965
2620
98
globlastp


2152
ZLFS24
Environmental sample
3966
2620
97.9
globlastp


2153
ZLFS24

Pantoea sp.

3967
2620
93.6
globlastp


2154
ZLFS24

Pantoea sp.

3968
2620
93.3
globlastp


2155
ZLFS24
Environmental sample
3969
2620
92.8
globlastp


2156
ZLFS24
Environmental sample
3970
2620
92.7
globlastp


2157
ZLFS24
Environmental sample
3971
2620
88.7
globlastp


2158
ZLFS24

Pantoea sp.

3972
2620
88.6
globlastp


2159
ZLFS24

Pantoea sp.

3973
2620
87.7
globlastp


2160
ZLFS24
Environmental sample
3974
2620
80.3
globlastp


2161
ZLFS24

Pantoea sp.

3975
2620
80
globlastp


2162
ZLFS24

Pantoea sp.

3976
2620
79.4
globlastp


2084
ZLFS11

Collimonas sp.

3898
2608
81.5
globlastp


2085
ZLFS11

Collimonas sp.

3899
2608
79
globlastp


825
BLFS15
Multievos46 sp.
2677
2458
93.2
globlastp


826
BLFS15
Environmental sample
2678
2458
89.2
globlastp


827
BLFS15

Arthrobacter sp.

2679
2458
84.4
globlastp


828
BLFS15

Arthrobacter sp.

2680
2458
83.6
globlastp


829
BLFS15

Arthrobacter sp.

2681
2458
83.4
globlastp


830
BLFS15

Arthrobacter sp.

2682
2458
83.2
globlastp


831
BLFS15

Arthrobacter sp.

2683
2458
76.9
globlastp


832
BLFS15

Arthrobacter sp.

2684
2458
76.7
globlastp


833
BLFS15

Arthrobacter sp.

2685
2458
76
globlastp


1753
BLFS134

Pseudomonas sp.

3577
2569
98.9
globlastp


1754
BLFS134

Pseudomonas sp.

3578
2569
96.8
globlastp


1755
BLFS134

Pseudomonas sp.

3579
2569
95.8
globlastp


1756
BLFS134

Pseudomonas sp.

3580
2569
93.7
globlastp


1757
BLFS134

Pseudomonas sp.

3581
2569
92.6
globlastp


1758
BLFS134

Pseudomonas sp.

3582
2569
91.6
globlastp


1759
BLFS134

Pseudomonas sp.

3583
2569
90.5
globlastp


1760
BLFS134

Pseudomonas sp.

3584
2569
89.5
globlastp


1761
BLFS134
Environmental sample
3585
2569
88.4
globlastp


1762
BLFS134
Environmental sample
3586
2569
87.4
globlastp


1763
BLFS134
Environmental sample
3587
2569
86.3
globlastp


1764
BLFS134

Pseudomonas sp.

3588
2569
85.3
globlastp


1765
BLFS134

Pseudomonas sp.

3589
2569
84.2
globlastp


1766
BLFS134

Pseudomonas sp.

3590
2569
83.2
globlastp


1767
BLFS134

Pseudomonas sp.

3591
2569
82.1
globlastp


1768
BLFS134
Environmental sample
3592
2569
81.1
globlastp


1769
BLFS134
Environmental sample
3593
2569
80
globlastp


1770
BLFS134

Pseudomonas sp.

3594
2569
79.2
globlastp


1771
BLFS134
Environmental sample
3595
2569
78.1
globlastp


1772
BLFS134

Pseudomonas sp.

3596
2569
77.1
globlastp


1773
BLFS134

Pseudomonas sp.

3597
2569
76
globlastp


1774
BLFS134

Pseudomonas sp.

3598
2569
75
globlastp


1775
BLFS134

Pseudomonas sp.

3599
2569
74
globlastp


1776
BLFS134
Environmental sample
3600
2569
73.7
globlastp


1777
BLFS134

Pseudomonas sp.

3601
2569
72.4
globlastp


1778
BLFS134
Environmental sample
3602
2569
71.6
globlastp


1779
BLFS134

Azotobacter sp.

3603
2569
70.5
globlastp


2163
ZLFS25

Curtobacterium sp.

3977
2621
95.4
globlastp


2164
ZLFS25
Environmental sample
3978
2621
91.3
globlastp


2165
ZLFS25

Curtobacterium sp.

3979
2621
90.1
globlastp


2166
ZLFS25

Curtobacterium sp.

3980
2621
89.9
globlastp


2167
ZLFS25
Environmental sample
3981
2621
76.6
globlastp


2168
ZLFS25
Environmental sample
3982
2621
74.4
globlastp


2169
ZLFS25
Environmental sample
3983
2621
73.8
globlastp


834
BLFS16
Environmental sample
2686
2459
94.7
globlastp


835
BLFS16

Arthrobacter sp.

2687
2459
78.1
globlastp


836
BLFS16

Arthrobacter sp.

2688
2459
78
globlastp


837
BLFS16

Arthrobacter sp.

2689
2459
77.7
globlastp


838
BLFS16

Arthrobacter sp.

2690
2459
77
globlastp


984
BLFS43

Burkholderia sp.

2832
2484
99.3
globlastp


985
BLFS43

Burkholderia sp.

2833
2484
76.8
globlastp


986
BLFS43

Burkholderia sp.

2834
2484
71.4
globlastp


987
BLFS43

Burkholderia sp.

2835
2484
70.7
globlastp


2127
ZLFS22

Paenibacillus sp.

3941
2618
99.1
globlastp


2128
ZLFS22

Paenibacillus sp.

3942
2618
98.2
globlastp


2129
ZLFS22

Paenibacillus sp.

3943
2618
97.7
globlastp


2130
ZLFS22

Paenibacillus sp.

3944
2618
96.4
globlastp


2131
ZLFS22

Paenibacillus sp.

3945
2618
95.9
globlastp


2132
ZLFS22

Paenibacillus sp.

3946
2618
95.5
globlastp


2133
ZLFS22

Paenibacillus sp.

3947
2618
95
globlastp


2134
ZLFS22

Paenibacillus sp.

3948
2618
93.7
globlastp


2135
ZLFS22

Paenibacillus sp.

3949
2618
93.2
globlastp


2136
ZLFS22

Paenibacillus sp.

3950
2618
91.9
globlastp


2137
ZLFS22

Paenibacillus sp.

3951
2618
85.6
globlastp


2138
ZLFS22

Paenibacillus sp.

3952
2618
84.7
globlastp


2139
ZLFS22

Paenibacillus sp.

3953
2618
83.8
globlastp


998
BLFS47

Salinibacillus sp.

2846
2488
90.9
globlastp


999
BLFS47

Cellulosimicrobium sp.

2847
2488
77.3
globlastp


1000
BLFS47

Cellulosimicrobium sp.

2848
2488
71.8
globlastp


1001
BLFS47
Multievos46 sp.
2849
2488
70.2
globlastp


1258
BLFS94

Paenibacillus sp.

3093
2530
99
globlastp


1259
BLFS94

Paenibacillus sp.

3094
2530
98.1
globlastp


1260
BLFS94

Paenibacillus sp.

3095
2530
96.3
globlastp


1261
BLFS94

Paenibacillus sp.

3096
2530
95
globlastp


1262
BLFS94

Paenibacillus sp.

3097
2530
94
globlastp


1263
BLFS94

Paenibacillus sp.

3098
2530
93.1
globlastp


1264
BLFS94

Paenibacillus sp.

3099
2530
74.6
globlastp


1265
BLFS94

Paenibacillus sp.

3100
2530
73.4
globlastp


1266
BLFS94

Paenibacillus sp.

3101
2530
71.7
globlastp


1104
BLFS75

Paenibacillus sp.

2944
2513
99.4
globlastp


1105
BLFS75

Paenibacillus sp.

2945
2513
98.8
globlastp


1106
BLFS75

Paenibacillus sp.

2946
2513
98.6
globlastp


1107
BLFS75

Paenibacillus sp.

2947
2513
98.3
globlastp


1108
BLFS75

Paenibacillus sp.

2948
2513
98
globlastp


1109
BLFS75

Paenibacillus sp.

2949
2513
97.7
globlastp


1110
BLFS75

Paenibacillus sp.

2950
2513
97.1
globlastp


1111
BLFS75

Paenibacillus sp.

2951
2513
92.5
globlastp


1112
BLFS75

Paenibacillus sp.

2952
2513
92.2
globlastp


1113
BLFS75

Paenibacillus sp.

2953
2513
91.9
globlastp


1114
BLFS75

Paenibacillus sp.

2954
2513
88.7
globlastp


1725
BLFS132

Pseudomonas sp.

3549
2567
98.5
globlastp


1726
BLFS132

Pseudomonas sp.

3550
2567
97.1
globlastp


1727
BLFS132

Pseudomonas sp.

3551
2567
95.6
globlastp


1728
BLFS132

Pseudomonas sp.

3552
2567
94.1
globlastp


1729
BLFS132

Pseudomonas sp.

3553
2567
92.6
globlastp


1730
BLFS132

Pseudomonas sp.

3554
2567
91.4
globlastp


1731
BLFS132

Pseudomonas sp.

3555
2567
88.6
globlastp


1732
BLFS132

Pseudomonas sp.

3556
2567
87
globlastp


1733
BLFS132

Pseudomonas sp.

3557
2567
85.5
globlastp


1734
BLFS132

Pseudomonas sp.

3558
2567
84.1
globlastp


1735
BLFS132
Environmental sample
3559
2567
83.1
globlastp


1736
BLFS132

Pseudomonas sp.

3560
2567
82.6
globlastp


1737
BLFS132

Pseudomonas sp.

3561
2567
81.2
globlastp


1738
BLFS132

Pseudomonas sp.

3562
2567
80
globlastp


1739
BLFS132
Environmental sample
3563
2567
79.4
globlastp


1740
BLFS132

Pseudomonas sp.

3564
2567
78.3
globlastp


1741
BLFS132

Pseudomonas sp.

3565
2567
77.9
globlastp


1742
BLFS132

Pseudomonas sp.

3566
2567
76.5
globlastp


1743
BLFS132

Pseudomonas sp.

3567
2567
75
globlastp


1744
BLFS132

Pseudomonas sp.

3568
2567
74
globlastp


1745
BLFS132

Pseudomonas sp.

3569
2567
73
globlastp


1746
BLFS132

Pseudomonas sp.

3570
2567
72
globlastp


1747
BLFS132

Pseudomonas sp.

3571
2567
71
globlastp


1748
BLFS132

Pseudomonas sp.

3572
2567
70.7
globlastp


809
BLFS10

Amycolatopsis sp.

2661
2453
97.4
globlastp


810
BLFS10

Amycolatopsis sp.

2662
2453
94.9
globlastp


804
BLFS10

Amycolatopsis sp.

2656
2453
94
globlastp


600
BLFS10

Amycolatopsis sp.

2452
2453
75.2
globlastp


803
BLFS10

Amycolatopsis sp.

2655
2453
74.4
globlastp


811
BLFS10

Streptomyces sp.

2663
2453
70.9
globlastp


2312
ZLFS37

Serratia sp.

4126
2632
99.1
globlastp


2313
ZLFS37
Environmental sample
4127
2632
97.3
globlastp


2314
ZLFS37

Serratia sp.

4128
2632
96.4
globlastp


2315
ZLFS37

Serratia sp.

4129
2632
95.5
globlastp


2316
ZLFS37

Serratia sp.

4130
2632
94.6
globlastp


2317
ZLFS37

Serratia sp.

4131
2632
93.8
globlastp


2318
ZLFS37

Serratia sp.

4132
2632
92.9
globlastp


2319
ZLFS37

Serratia sp.

4133
2632
91.1
globlastp


2320
ZLFS37

Serratia sp.

4134
2632
84.8
globlastp


2321
ZLFS37

Serratia sp.

4135
2632
83.9
globlastp


2322
ZLFS37

Serratia sp.

4136
2632
83
globlastp


2323
ZLFS37

Yersinia sp.

4137
2632
82.1
globlastp


2324
ZLFS37

Serratia sp.

4138
2632
81.6
globlastp


2325
ZLFS37

Yersinia sp.

4139
2632
81.2
globlastp


2326
ZLFS37

Hafnia sp.

4140
2632
80.5
globlastp


2327
ZLFS37

Yersinia sp.

4141
2632
80.4
globlastp


2328
ZLFS37

Yersinia sp.

4142
2632
79.5
globlastp


2329
ZLFS37

Yersinia sp.

4143
2632
78.6
globlastp


2330
ZLFS37

Enterobacteriaceae sp.

4144
2632
77.9
globlastp


2331
ZLFS37

Rahnella sp.

4145
2632
77.7
globlastp


2332
ZLFS37

Ewinyella sp.

4146
2632
76.8
globlastp


2333
ZLFS37

Rahnella sp.

4147
2632
76.5
globlastp


2334
ZLFS37

Rahnella sp.

4148
2632
75.7
globlastp


2335
ZLFS37

Yersinia sp.

4149
2632
74
globlastp


2336
ZLFS37

Candidatus sp.

4150
2632
71.3
globlastp


2337
ZLFS37

Serratia sp.


2632
70.54
glotblastn


2338
ZLFS37

Serratia sp.

4151
2632
70.5
globlastp


2339
ZLFS37

Candidatus sp.

4152
2632
70.4
globlastp


2340
ZLFS37
gamma sp.
4153
2632
70.2
globlastp


1038
BLFS62

Erwinia sp.

2881
2502
83.2
globlastp


1039
BLFS62

Pantoea sp.

2882
2502
81.5
globlastp


1040
BLFS62
Environmental sample
2883
2502
79.2
globlastp


1041
BLFS62
Environmental sample
2884
2502
78.2
globlastp


1042
BLFS62
Environmental sample
2885
2502
77
globlastp


1043
BLFS62
Environmental sample
2886
2502
76.2
globlastp


1044
BLFS62

Pantoea sp.

2887
2502
75.2
globlastp


1045
BLFS62

Pantoea sp.

2888
2502
74.1
globlastp


1046
BLFS62

Pantoea sp.

2889
2502
73.5
globlastp


1047
BLFS62

Pantoea sp.

2890
2502
71.5
globlastp


1048
BLFS62

Enterobacter sp.


2502
70.51
glotblastn


1006
BLFS51

Herbaspirillum sp.


2492
76.51
glotblastn


1007
BLFS51

Collimonas sp.

2854
2492
73.7
globlastp


1008
BLFS51

Collimonas sp.


2492
72.89
glotblastn


1009
BLFS51

Herbaspirillum sp.

2855
2492
70.3
globlastp


1701
BLFS131

Pseudomonas sp.

3525
2566
98.4
globlastp


1702
BLFS131

Pseudomonas sp.

3526
2566
94.5
globlastp


1703
BLFS131

Pseudomonas sp.

3527
2566
91.2
globlastp


1704
BLFS131

Pseudomonas sp.

3528
2566
90.1
globlastp


1705
BLFS131

Pseudomonas sp.

3529
2566
89
globlastp


1706
BLFS131

Pseudomonas sp.

3530
2566
88
globlastp


1707
BLFS131

Pseudomonas sp.

3531
2566
87
globlastp


1708
BLFS131

Pseudomonas sp.

3532
2566
86.3
globlastp


1709
BLFS131
Environmental sample
3533
2566
85.2
globlastp


1710
BLFS131
Environmental sample
3534
2566
84.1
globlastp


1711
BLFS131
Environmental sample
3535
2566
83
globlastp


1712
BLFS131

Pseudomonas sp.

3536
2566
82.2
globlastp


1713
BLFS131

Pseudomonas sp.

3537
2566
81.1
globlastp


1714
BLFS131

Pseudomonas sp.

3538
2566
80
globlastp


1715
BLFS131
Environmental sample
3539
2566
79.1
globlastp


1716
BLFS131

Pseudomonas sp.

3540
2566
78.1
globlastp


1717
BLFS131

Pseudomonas sp.

3541
2566
77
globlastp


1718
BLFS131

Pseudomonas sp.

3542
2566
76
globlastp


1719
BLFS131

Pseudomonas sp.

3543
2566
75.3
globlastp


1720
BLFS131
Environmental sample
3544
2566
74.2
globlastp


1721
BLFS131

Pseudomonas sp.

3545
2566
73.1
globlastp


1722
BLFS131

Pseudomonas sp.

3546
2566
72
globlastp


1723
BLFS131

Pseudomonas sp.

3547
2566
71.4
globlastp


1724
BLFS131

Pseudomonas sp.

3548
2566
70.3
globlastp


997
BLFS46

Burkholderia sp.

2845
2487
84.9
globlastp


1072
BLFS73

Paenibacillus sp.

2914
2511
99.2
globlastp


1073
BLFS73

Paenibacillus sp.

2915
2511
98.4
globlastp


1074
BLFS73

Paenibacillus sp.

2916
2511
96.7
globlastp


1075
BLFS73

Paenibacillus sp.

2917
2511
95.9
globlastp


1076
BLFS73

Paenibacillus sp.

2918
2511
95.1
globlastp


1077
BLFS73

Paenibacillus sp.

2919
2511
84.6
globlastp


1078
BLFS73

Paenibacillus sp.


2511
82.93
glotblastn


1079
BLFS73

Paenibacillus sp.

2920
2511
82.1
globlastp


1080
BLFS73

Paenibacillus sp.

2921
2511
81.3
globlastp


1081
BLFS73

Paenibacillus sp.

2922
2511
78.9
globlastp


1082
BLFS73

Paenibacillus sp.

2923
2511
78
globlastp


1083
BLFS73

Paenibacillus sp.

2924
2511
77.2
globlastp


1084
BLFS73
Environmental sample
2925
2511
76.4
globlastp


1085
BLFS73

Paenibacillus sp.

2926
2511
75
globlastp


1086
BLFS73

Paenibacillus sp.

2927
2511
74
globlastp


1087
BLFS73

Paenibacillus sp.


2511
73.17
glotblastn


1088
BLFS73

Paenibacillus sp.

2928
2511
72.4
globlastp


1089
BLFS73

Paenibacillus sp.

2929
2511
71.2
globlastp


1652
BLFS127

Pseudomonas sp.

3476
2562
98.3
globlastp


1653
BLFS127

Pseudomonas sp.

3477
2562
98.1
globlastp


1654
BLFS127

Pseudomonas sp.

3478
2562
97.7
globlastp


1655
BLFS127

Pseudomonas sp.

3479
2562
97.4
globlastp


1656
BLFS127

Pseudomonas sp.

3480
2562
96.7
globlastp


1657
BLFS127

Pseudomonas sp.

3481
2562
96
globlastp


1658
BLFS127

Pseudomonas sp.

3482
2562
95.8
globlastp


1659
BLFS127

Pseudomonas sp.

3483
2562
95.2
globlastp


1660
BLFS127

Pseudomonas sp.

3484
2562
86.2
globlastp


1661
BLFS127

Pseudomonas sp.

3485
2562
75.5
globlastp


1662
BLFS127

Pseudomonas sp.

3486
2562
75.3
globlastp


1663
BLFS127

Pseudomonas sp.

3487
2562
75.2
globlastp


1664
BLFS127

Pseudomonas sp.

3488
2562
75.1
globlastp


1665
BLFS127

Pseudomonas sp.

3489
2562
75
globlastp


1666
BLFS127

Pseudomonas sp.

3490
2562
74.8
globlastp


1667
BLFS127

Pseudomonas sp.

3491
2562
73.3
globlastp


1067
BLFS68

Janthinobacterium sp.

2909
2507
98.7
globlastp


1068
BLFS68

Janthinobacterium sp.

2910
2507
90.3
globlastp


1069
BLFS68

Janthinobacterium sp.

2911
2507
89.6
globlastp


1070
BLFS68

Janthinobacterium sp.

2912
2507
88.5
globlastp


1049
BLFS63

Erwinia sp.

2891
2503
85.7
globlastp


1050
BLFS63

Erwinia sp.

2892
2503
83.8
globlastp


1051
BLFS63
Environmental sample
2893
2503
82.9
globlastp


1052
BLFS63
Environmental sample
2894
2503
81
globlastp


1053
BLFS63
Environmental sample
2895
2503
80.3
globlastp


1054
BLFS63

Erwinia sp.

2896
2503
79.4
globlastp


1055
BLFS63

Erwinia sp.

2897
2503
78
globlastp


1056
BLFS63

Pantoea sp.

2898
2503
76.8
globlastp


1057
BLFS63

Meta sp.

2899
2503
75
globlastp


1058
BLFS63

Plautia sp.

2900
2503
74
globlastp


1059
BLFS63

Pantoea sp.

2901
2503
73.2
globlastp


1060
BLFS63
Environmental sample
2902
2503
72
globlastp


1061
BLFS63
Environmental sample
2903
2503
71.8
globlastp


1002
BLFS48

Collimonas sp.

2850
2489
98.9
globlastp


1003
BLFS48

Collimonas sp.

2851
2489
78.9
globlastp


1004
BLFS48

Collimonas sp.

2852
2489
76.5
globlastp


2080
ZLFS7

Collimonas sp.

3894
2604
70.4
globlastp


937
BLFS33

Bacillus sp.

2787
2475
99.1
globlastp


938
BLFS33

Bacillus sp.

2788
2475
98.1
globlastp


939
BLFS33

Bacillus sp.

2789
2475
96.4
globlastp


940
BLFS33

Bacillus sp.

2790
2475
94
globlastp


941
BLFS33

Bacillus sp.

2791
2475
93.1
globlastp


942
BLFS33

Bacillus sp.

2792
2475
92.1
globlastp


943
BLFS33

Bacillus sp.

2793
2475
90.5
globlastp


944
BLFS33

Bacillus sp.

2794
2475
80
globlastp


945
BLFS33

Bacillus sp.

2795
2475
78.1
globlastp


946
BLFS33

Bacillus sp.

2796
2475
76.7
globlastp


947
BLFS33

Bacillus sp.

2797
2475
76
globlastp


948
BLFS33

Bacillus sp.

2798
2475
75.4
globlastp


1431
BLFS110
Environmental sample
3257
2546
83.8
globlastp


1227
BLFS91

Paenibacillus sp.

3062
2527
99.2
globlastp


1228
BLFS91

Paenibacillus sp.

3063
2527
96.8
globlastp


1229
BLFS91

Paenibacillus sp.

3064
2527
96
globlastp


1230
BLFS91

Paenibacillus sp.

3065
2527
95.2
globlastp


1231
BLFS91

Paenibacillus sp.

3066
2527
93.6
globlastp


1232
BLFS91

Paenibacillus sp.

3067
2527
88.8
globlastp


1233
BLFS91

Paenibacillus sp.

3068
2527
87.2
globlastp


1234
BLFS91

Paenibacillus sp.

3069
2527
86.4
globlastp


1235
BLFS91

Paenibacillus sp.

3070
2527
84.8
globlastp


1236
BLFS91

Paenibacillus sp.

3071
2527
81.6
globlastp


1237
BLFS91

Paenibacillus sp.

3072
2527
80.8
globlastp


2255
ZLFS31

Serratia sp.

4069
2626
99.2
globlastp


2256
ZLFS31

Serratia sp.

4070
2626
98.8
globlastp


2257
ZLFS31

Serratia sp.

4071
2626
97
globlastp


2258
ZLFS31
Multievos46 sp.
4072
2626
92.1
globlastp


2259
ZLFS31

Serratia sp.

4073
2626
91.3
globlastp


2260
ZLFS31

Serratia sp.

4074
2626
89
globlastp


2261
ZLFS31

Serratia sp.

4075
2626
88.2
globlastp


2262
ZLFS31

Serratia sp.

4076
2626
83.1
globlastp


2263
ZLFS31

Serratia sp.

4077
2626
82.1
globlastp


2264
ZLFS31

Serratia sp.

4078
2626
81
globlastp


2265
ZLFS31

Yersinia sp.

4079
2626
75.3
globlastp


2266
ZLFS31

Yersinia sp.

4080
2626
74.8
globlastp


2267
ZLFS31

Yersinia sp.

4081
2626
73
globlastp


903
BLFS27

Bacillus sp.

2753
2470
89.4
globlastp


904
BLFS27

Bacillus sp.

2754
2470
87.6
globlastp


905
BLFS27

Bacillus sp.

2755
2470
86.6
globlastp


906
BLFS27

Bacillus sp.

2756
2470
78.6
globlastp


907
BLFS27

Bacillus sp.

2757
2470
75.1
globlastp


1475
BLFS114

Pseudomonas sp.

3301
2550
93.6
globlastp


1476
BLFS114

Pseudomonas sp.

3302
2550
93.1
globlastp


1477
BLFS114

Pseudomonas sp.

3303
2550
92.5
globlastp


1478
BLFS114

Pseudomonas sp.

3304
2550
91
globlastp


1479
BLFS114
Environmental sample
3305
2550
90.2
globlastp


1480
BLFS114
Environmental sample
3306
2550
89
globlastp


1481
BLFS114
Environmental sample
3307
2550
88.4
globlastp


1482
BLFS114

Pseudomonas sp.

3308
2550
87.3
globlastp


1483
BLFS114

Pseudomonas sp.

3309
2550
86.2
globlastp


1484
BLFS114

Pseudomonas sp.

3310
2550
85
globlastp


1485
BLFS114

Pseudomonas sp.

3311
2550
84.4
globlastp


1486
BLFS114

Erwinia sp.

3312
2550
83.2
globlastp


1487
BLFS114

Pseudomonas sp.

3313
2550
82.1
globlastp


1488
BLFS114

Pseudomonas sp.

3314
2550
81
globlastp


1489
BLFS114

Pseudomonas sp.

3315
2550
80.3
globlastp


1490
BLFS114
Environmental sample
3316
2550
79.2
globlastp


1491
BLFS114
Environmental sample
3317
2550
78
globlastp


1492
BLFS114

Pseudomonas sp.

3318
2550
77.3
globlastp


1493
BLFS114

Pseudomonas sp.

3319
2550
76
globlastp


1494
BLFS114
Environmental sample
3320
2550
75.1
globlastp


1495
BLFS114
Environmental sample
3321
2550
74
globlastp


1496
BLFS114
Environmental sample
3322
2550
73.4
globlastp


1497
BLFS114

Pseudomonas sp.

3323
2550
72
globlastp


1498
BLFS114
Environmental sample
3324
2550
71.1
globlastp


1499
BLFS114
Environmental sample
3325
2550
70.5
globlastp


1030
BLFS61

Pseudomonas sp.

2873
2501
99
globlastp


1031
BLFS61

Pseudomonas sp.

2874
2501
96
globlastp


1032
BLFS61

Pseudomonas sp.

2875
2501
95.7
globlastp


1033
BLFS61
Environmental sample
2876
2501
92.5
globlastp


1034
BLFS61

Pseudomonas sp.

2877
2501
85.5
globlastp


1035
BLFS61
Environmental sample
2878
2501
79.2
globlastp


1036
BLFS61

Pseudomonas sp.

2879
2501
78.2
globlastp


1037
BLFS61

Pseudomonas sp.

2880
2501
74.5
globlastp


1577
BLFS121

Pseudomonas sp.

3402
2556
99
globlastp


1578
BLFS121

Pseudomonas sp.

3403
2556
98.8
globlastp


1579
BLFS121

Pseudomonas sp.

3404
2556
98.3
globlastp


1580
BLFS121

Pseudomonas sp.

3405
2556
97.8
globlastp


1581
BLFS121

Pseudomonas sp.

3406
2556
93.2
globlastp


1582
BLFS121

Pseudomonas sp.

3407
2556
92.3
globlastp


1583
BLFS121

Pseudomonas sp.

3408
2556
87
globlastp


1584
BLFS121

Pseudomonas sp.

3409
2556
86
globlastp


1585
BLFS121

Pseudomonas sp.

3410
2556
85
globlastp


1586
BLFS121
Environmental sample
3411
2556
84.1
globlastp


1587
BLFS121

Pseudomonas sp.

3412
2556
83.1
globlastp


1588
BLFS121
Environmental sample
3413
2556
82.1
globlastp


1589
BLFS121

Pseudomonas sp.

3414
2556
81.4
globlastp


1590
BLFS121

Pseudomonas sp.

3415
2556
80
globlastp


1591
BLFS121

Pseudomonas sp.

3416
2556
79.2
globlastp


1592
BLFS121
Environmental sample
3417
2556
78
globlastp


1593
BLFS121

Pseudomonas sp.

3418
2556
77.5
globlastp


1594
BLFS121

Pseudomonas sp.

3419
2556
76.1
globlastp


1595
BLFS121

Polaromonas sp.

3420
2556
75.7
globlastp


1596
BLFS121

Pseudomonas sp.

3421
2556
74.2
globlastp


1597
BLFS121

Pseudomonas sp.

3422
2556
72
globlastp


1598
BLFS121

Pseudomonas sp.

3423
2556
71
globlastp


2092
ZLFS15

Collimonas sp.

3906
2612
89.2
globlastp


2093
ZLFS15

Collimonas sp.

3907
2612
87.2
globlastp


2094
ZLFS15

Collimonas sp.

3908
2612
85.6
globlastp


1115
BLFS76

Paenibacillus sp.

2955
2514
95.4
globlastp


1116
BLFS76

Paenibacillus sp.

2956
2514
94.6
globlastp


1117
BLFS76

Paenibacillus sp.

2957
2514
93.4
globlastp


1118
BLFS76

Paenibacillus sp.

2958
2514
92.7
globlastp


1119
BLFS76

Paenibacillus sp.

2959
2514
90
globlastp


1120
BLFS76

Paenibacillus sp.

2960
2514
82.6
globlastp


1121
BLFS76

Paenibacillus sp.

2961
2514
72.3
globlastp


1122
BLFS76

Paenibacillus sp.


2514
71.04
glotblastn


1123
BLFS76

Paenibacillus sp.

2962
2514
70.1
globlastp


1530
BLFS116

Pseudomonas sp.

3355
2552
95.8
globlastp


1531
BLFS116

Pseudomonas sp.

3356
2552
90.2
globlastp


1532
BLFS116

Pseudomonas sp.

3357
2552
89.2
globlastp


1533
BLFS116
Environmental sample
3358
2552
88.1
globlastp


1534
BLFS116

Pseudomonas sp.

3359
2552
87.8
globlastp


1535
BLFS116

Pseudomonas sp.

3360
2552
86.3
globlastp


1536
BLFS116

Pseudomonas sp.

3361
2552
85.5
globlastp


1537
BLFS116

Pseudomonas sp.

3362
2552
84.8
globlastp


1538
BLFS116

Pseudomonas sp.

3363
2552
83
globlastp


1539
BLFS116

Pseudomonas sp.

3364
2552
82.1
globlastp


1540
BLFS116

Pseudomonas sp.

3365
2552
81
globlastp


1541
BLFS116

Pseudomonas sp.

3366
2552
80.1
globlastp


1542
BLFS116

Pseudomonas sp.

3367
2552
79.1
globlastp


1543
BLFS116

Pseudomonas sp.

3368
2552
78
globlastp


1544
BLFS116

Pseudomonas sp.

3369
2552
77
globlastp


1545
BLFS116

Pseudomonas sp.

3370
2552
76.4
globlastp


1546
BLFS116

Pseudomonas sp.

3371
2552
75.1
globlastp


1547
BLFS116

Pseudomonas sp.

3372
2552
74
globlastp


1548
BLFS116

Pseudomonas sp.

3373
2552
73.2
globlastp


1549
BLFS116

Pseudomonas sp.

3374
2552
72.3
globlastp


1550
BLFS116

Pseudomonas sp.

3375
2552
71
globlastp


1551
BLFS116
Environmental sample
3376
2552
70.2
globlastp


1344
BLFS103

Pantoea sp.

3179
2539
94.1
globlastp


1345
BLFS103
unknown sp.
3180
2539
89.2
globlastp


1346
BLFS103

Pantoea sp.

3181
2539
86.5
globlastp


1347
BLFS103

Photorhabdus sp.


2539
74.65
glotblastn


1348
BLFS103

Yersinia sp.


2539
74.31
glotblastn


1349
BLFS103

Serratia sp.


2539
73.96
glotblastn


1350
BLFS103

Salmonella sp.


2539
73.61
glotblastn


1351
BLFS103

Salmonella sp.


2539
73.26
glotblastn


1352
BLFS103

Serratia sp.


2539
73.1
glotblastn


1353
BLFS103

Yersinia sp.

3182
2539
71.9
globlastp


1354
BLFS103

Yersinia sp.


2539
71.53
glotblastn


1355
BLFS103

Serratia sp.


2539
71.18
glotblastn


1356
BLFS103

Pantoea sp.


2539
70.14
glotblastn


1668
BLFS128

Pseudomonas sp.

3492
2563
99.3
globlastp


1669
BLFS128

Pseudomonas sp.

3493
2563
95.2
globlastp


1670
BLFS128
Environmental sample
3494
2563
94.3
globlastp


1671
BLFS128

Pseudomonas sp.

3495
2563
93.3
globlastp


1672
BLFS128

Pseudomonas sp.

3496
2563
92.9
globlastp


1673
BLFS128

Pseudomonas sp.

3497
2563
91.7
globlastp


1674
BLFS128

Pseudomonas sp.

3498
2563
89.2
globlastp


1675
BLFS128

Pseudomonas sp.

3499
2563
80.1
globlastp


1676
BLFS128

Pseudomonas sp.

3500
2563
79.5
globlastp


1677
BLFS128

Pseudomonas sp.

3501
2563
78
globlastp


1678
BLFS128

Pseudomonas sp.

3502
2563
77
globlastp


1679
BLFS128

Pseudomonas sp.

3503
2563
76
globlastp


1680
BLFS128

Pseudomonas sp.

3504
2563
75.7
globlastp


1681
BLFS128

Erwinia sp.

3505
2563
73
globlastp


1682
BLFS128

Pseudomonas sp.

3506
2563
72
globlastp


1683
BLFS128

Pseudomonas sp.

3507
2563
71
globlastp


1684
BLFS128

Pseudomonas sp.

3508
2563
70
globlastp


1937
BLFS148

Serratia sp.

3760
2583
99.2
globlastp


1938
BLFS148

Serratia sp.

3761
2583
98.4
globlastp


1939
BLFS148

Serratia sp.

3762
2583
95.2
globlastp


1940
BLFS148

Serratia sp.

3763
2583
89.2
globlastp


1941
BLFS148

Serratia sp.

3764
2583
87.2
globlastp


1942
BLFS148

Serratia sp.

3765
2583
86.4
globlastp


1943
BLFS148

Serratia sp.

3766
2583
85.6
globlastp


1944
BLFS148

Yersinia sp.

3767
2583
84
globlastp


1945
BLFS148

Yersinia sp.

3768
2583
83.2
globlastp


1946
BLFS148

Pantoea sp.

3769
2583
82
globlastp


1947
BLFS148

Yersinia sp.

3770
2583
81.2
globlastp


1948
BLFS148

Pectobacterium sp.

3771
2583
80
globlastp


1949
BLFS148

Pectobacterium sp.

3772
2583
79.2
globlastp


1950
BLFS148

Erwinia sp.

3773
2583
78
globlastp


1951
BLFS148
Environmental sample
3774
2583
77.3
globlastp


1952
BLFS148

Pantoea sp.

3775
2583
76
globlastp


1953
BLFS148

Pantoea sp.

3776
2583
75
globlastp


1954
BLFS148

Buttiauxella sp.

3777
2583
74.2
globlastp


1955
BLFS148

Salmonella sp.

3778
2583
73
globlastp


1956
BLFS148

Proteus sp.

3779
2583
72
globlastp


1957
BLFS148

Enterobacter.2 sp.

3780
2583
71
globlastp


1958
BLFS148
Environmental sample
3781
2583
70.2
globlastp


1959
BLFS148

Citrobacter sp.


2583
70
glotblastn


859
BLFS19

Bacillus sp.

2711
2462
96.4
globlastp


860
BLFS19

Bacillus sp.

2712
2462
96.1
globlastp


861
BLFS19

Bacillus sp.

2713
2462
95.8
globlastp


862
BLFS19

Bacillus sp.

2714
2462
95.5
globlastp


863
BLFS19

Bacillus sp.

2715
2462
94.7
globlastp


864
BLFS19

Bacillus sp.

2716
2462
92
globlastp


865
BLFS19

Bacillus sp.

2717
2462
91.7
globlastp


988
BLFS44

Burkholderia sp.

2836
2485
99.4
globlastp


2095
ZLFS16

Collimonas sp.

3909
2613
94.5
globlastp


2096
ZLFS16

Collimonas sp.

3910
2613
85.1
globlastp


2097
ZLFS16

Herminiimonas sp.

3911
2613
74
globlastp


2098
ZLFS16

Duganella sp.

3912
2613
73
globlastp


2099
ZLFS16

Janthinobacterium sp.

3913
2613
72
globlastp


2100
ZLFS16

Massilia sp.

3914
2613
71.2
globlastp


2101
ZLFS16

Oxalobacteraceae sp.

3915
2613
70
globlastp


1893
BLFS145

Bacillus sp.

3716
2580
99.2
globlastp


1894
BLFS145

Bacillus sp.

3717
2580
97.9
globlastp


1895
BLFS145

Bacillus sp.

3718
2580
94
globlastp


1896
BLFS145

Bacillus sp.

3719
2580
93.2
globlastp


1897
BLFS145

Bacillus sp.

3720
2580
92.7
globlastp


1898
BLFS145

Bacillus sp.

3721
2580
92.1
globlastp


1899
BLFS145

Bacillus sp.

3722
2580
91.1
globlastp


1900
BLFS145

Bacillus sp.

3723
2580
84.3
globlastp


1901
BLFS145

Bacillus sp.

3724
2580
78.1
globlastp


1902
BLFS145

Bacillus sp.

3725
2580
77.5
globlastp


1903
BLFS145

Bacillus sp.

3726
2580
74.2
globlastp


1904
BLFS145

Bacillus sp.

3727
2580
74
globlastp


1905
BLFS145

Bacillus sp.

3728
2580
73.2
globlastp


1906
BLFS145

Bacillus sp.

3729
2580
71
globlastp


1500
BLFS115

Pseudomonas sp.

3326
2551
98.5
globlastp


1501
BLFS115

Pseudomonas sp.

3327
2551
98.3
globlastp


1502
BLFS115

Pseudomonas sp.

3328
2551
98.1
globlastp


1503
BLFS115

Pseudomonas sp.

3329
2551
97.7
globlastp


1504
BLFS115

Pseudomonas sp.

3330
2551
95.2
globlastp


1505
BLFS115

Pseudomonas sp.

3331
2551
94.1
globlastp


1506
BLFS115

Pseudomonas sp.

3332
2551
93.1
globlastp


1507
BLFS115

Pseudomonas sp.

3333
2551
92
globlastp


1508
BLFS115
Environmental sample
3334
2551
91
globlastp


1509
BLFS115

Pseudomonas sp.

3335
2551
90
globlastp


1510
BLFS115

Pseudomonas sp.

3336
2551
89.7
globlastp


1511
BLFS115

Pseudomonas sp.

3337
2551
88.1
globlastp


1512
BLFS115

Pseudomonas sp.

3338
2551
87.9
globlastp


1513
BLFS115

Pseudomonas sp.

3339
2551
86.2
globlastp


1514
BLFS115

Pseudomonas sp.

3340
2551
85.2
globlastp


1515
BLFS115

Pseudomonas sp.

3341
2551
85.1
globlastp


1516
BLFS115

Pseudomonas sp.

3342
2551
84
globlastp


1517
BLFS115

Pseudomonas sp.

3343
2551
83.4
globlastp


1518
BLFS115

Pseudomonas sp.

3344
2551
82.8
globlastp


1519
BLFS115
Environmental sample
3345
2551
80
globlastp


1520
BLFS115
Environmental sample
3346
2551
79
globlastp


1521
BLFS115

Pseudomonas sp.

3347
2551
78
globlastp


1522
BLFS115

Pseudomonas sp.

3348
2551
77
globlastp


1523
BLFS115

Pseudomonas sp.

3349
2551
76
globlastp


1524
BLFS115

Pseudomonas sp.

3350
2551
75.2
globlastp


1525
BLFS115
Environmental sample
3351
2551
74.2
globlastp


1526
BLFS115
Environmental sample
3352
2551
73
globlastp


1527
BLFS115

Azotobacter sp.

3353
2551
72.1
globlastp


1528
BLFS115

Pseudomonas sp.

3354
2551
71
globlastp


1529
BLFS115

Pseudomonas sp.


2551
70.48
glotblastn


1780
BLFS135

Erwinia sp.

3604
2570
97.6
globlastp


1781
BLFS135

Pseudomonas sp.

3605
2570
97.2
globlastp


1782
BLFS135

Pseudomonas sp.

3606
2570
97
globlastp


1783
BLFS135

Pseudomonas sp.

3607
2570
96.6
globlastp


1784
BLFS135

Pseudomonas sp.

3608
2570
96.4
globlastp


1785
BLFS135

Pseudomonas sp.

3609
2570
95.2
globlastp


1786
BLFS135
Environmental sample
3610
2570
94.6
globlastp


1787
BLFS135

Pseudomonas sp.

3611
2570
88.6
globlastp


1788
BLFS135

Pseudomonas sp.

3612
2570
87.3
globlastp


1789
BLFS135

Pseudomonas sp.

3613
2570
73
globlastp


1790
BLFS135

Pseudomonas sp.

3614
2570
72.8
globlastp


1791
BLFS135

Pseudomonas sp.

3615
2570
70.9
globlastp


1882
BLFS144

Salinibacillus sp.

3705
2579
99.1
globlastp


1883
BLFS144

Bacillus sp.

3706
2579
96.9
globlastp


1884
BLFS144

Bacillus sp.

3707
2579
95.2
globlastp


1885
BLFS144

Bacillus sp.

3708
2579
94.1
globlastp


1886
BLFS144

Bacillus sp.

3709
2579
88
globlastp


1887
BLFS144

Bacillus sp.

3710
2579
87
globlastp


1888
BLFS144

Bacillus sp.

3711
2579
86.7
globlastp


1889
BLFS144

Bacillus sp.

3712
2579
85.4
globlastp


1890
BLFS144

Bacillus sp.

3713
2579
85
globlastp


1891
BLFS144

Bacillus sp.

3714
2579
84.4
globlastp


1892
BLFS144

Bacillus sp.

3715
2579
84.1
globlastp


1063
BLFS65

Duganella sp.

2905
2505
84.5
globlastp


2424
ZLFS49

Curtobacterium sp.

4233
2640
93.2
globlastp


2425
ZLFS49

Pantoea sp.

4234
2640
85.7
globlastp


1811
BLFS139

Salinibacillus sp.

3634
2574
99.4
globlastp


1812
BLFS139

Salinibacillus sp.

3635
2574
98.8
globlastp


1813
BLFS139

Bacillus sp.

3636
2574
97.9
globlastp


1814
BLFS139

Bacillus sp.

3637
2574
93.8
globlastp


1815
BLFS139

Bacillus sp.

3638
2574
89.5
globlastp


1816
BLFS139

Bacillus sp.

3639
2574
88
globlastp


1817
BLFS139

Bacillus sp.

3640
2574
87.3
globlastp


1818
BLFS139

Bacillus sp.

3641
2574
86.4
globlastp


1819
BLFS139

Bacillus sp.

3642
2574
85.8
globlastp


1820
BLFS139

Bacillus sp.

3643
2574
83.9
globlastp


1821
BLFS139

Bacillus sp.

3644
2574
77.3
globlastp


1822
BLFS139

Bacillus sp.

3645
2574
76.3
globlastp


1823
BLFS139

Bacillus sp.

3646
2574
75.1
globlastp


823
BLFS14
Environmental sample
2675
2457
95.7
globlastp


824
BLFS14

Arthrobacter sp.

2676
2457
84
globlastp


1628
BLFS125

Pseudomonas sp.

3452
2560
96.6
globlastp


1629
BLFS125

Pseudomonas sp.

3453
2560
87.6
globlastp


1630
BLFS125

Pseudomonas sp.

3454
2560
86.5
globlastp


1631
BLFS125

Pseudomonas sp.

3455
2560
85.4
globlastp


1632
BLFS125

Pseudomonas sp.

3456
2560
84.3
globlastp


1633
BLFS125
Environmental sample
3457
2560
83.1
globlastp


1634
BLFS125

Meta sp.

3458
2560
82
globlastp


1635
BLFS125

Pseudomonas sp.

3459
2560
80.9
globlastp


1636
BLFS125

Pseudomonas sp.

3460
2560
79.8
globlastp


1637
BLFS125

Pseudomonas sp.

3461
2560
78.8
globlastp


1638
BLFS125

Pseudomonas sp.

3462
2560
78.7
globlastp


1639
BLFS125
Environmental sample
3463
2560
77.5
globlastp


1640
BLFS125
Environmental sample
3464
2560
76.4
globlastp


1641
BLFS125
Environmental sample
3465
2560
75.3
globlastp


1642
BLFS125
Environmental sample
3466
2560
74.2
globlastp


1643
BLFS125
Environmental sample
3467
2560
73
globlastp


1181
BLFS86

Paenibacillus sp.

3018
2522
95.1
globlastp


1182
BLFS86

Paenibacillus sp.

3019
2522
92.7
globlastp


1183
BLFS86

Paenibacillus sp.

3020
2522
89.2
globlastp


1184
BLFS86

Paenibacillus sp.

3021
2522
89
globlastp


1185
BLFS86

Paenibacillus sp.

3022
2522
87.8
globlastp


1186
BLFS86

Paenibacillus sp.

3023
2522
84.7
globlastp


1336
BLFS102
Environmental sample
3171
2538
98.5
globlastp


1337
BLFS102
Environmental sample
3172
2538
97.1
globlastp


1338
BLFS102

Pantoea sp.

3173
2538
88.2
globlastp


1339
BLFS102
Environmental sample
3174
2538
86.8
globlastp


1340
BLFS102

Pantoea sp.

3175
2538
85.3
globlastp


1341
BLFS102

Pantoea sp.

3176
2538
80.9
globlastp


1342
BLFS102
Environmental sample
3177
2538
76.5
globlastp


1343
BLFS102

Pantoea sp.

3178
2538
72.1
globlastp


909
BLFS31

Bacillus sp.

2759
2473
99.2
globlastp


910
BLFS31

Bacillus sp.

2760
2473
98.4
globlastp


911
BLFS31

Streptococcus sp.

2761
2473
97.7
globlastp


912
BLFS31

Bacillus sp.

2762
2473
96.9
globlastp


913
BLFS31

Bacillus sp.

2763
2473
96.1
globlastp


914
BLFS31

Bacillus sp.

2764
2473
95.3
globlastp


915
BLFS31

Bacillus sp.

2765
2473
93
globlastp


916
BLFS31

Bacillus sp.

2766
2473
89.1
globlastp


917
BLFS31

Bacillus sp.

2767
2473
86.9
globlastp


918
BLFS31

Bacillus sp.

2768
2473
86.2
globlastp


919
BLFS31

Bacillus sp.

2769
2473
85.4
globlastp


1437
BLFS112

Pseudomonas sp.

3263
2548
98.8
globlastp


1438
BLFS112

Pseudomonas sp.

3264
2548
96.8
globlastp


1439
BLFS112

Pseudomonas sp.

3265
2548
94.7
globlastp


1440
BLFS112

Pseudomonas sp.

3266
2548
94.3
globlastp


1441
BLFS112

Pseudomonas sp.

3267
2548
93.1
globlastp


1442
BLFS112

Pseudomonas sp.

3268
2548
91.8
globlastp


1443
BLFS112

Pseudomonas sp.

3269
2548
91.4
globlastp


1444
BLFS112

Pseudomonas sp.

3270
2548
90.2
globlastp


1445
BLFS112

Pseudomonas sp.

3271
2548
89.4
globlastp


1446
BLFS112

Pseudomonas sp.

3272
2548
89
globlastp


1447
BLFS112

Pseudomonas sp.

3273
2548
74.8
globlastp


1448
BLFS112

Pseudomonas sp.

3274
2548
72.9
globlastp


1449
BLFS112

Pseudomonas sp.

3275
2548
71.4
globlastp


2268
ZLFS32

Serratia sp.

4082
2627
99.2
globlastp


2269
ZLFS32

Serratia sp.

4083
2627
97
globlastp


2270
ZLFS32

Serratia sp.

4084
2627
96.2
globlastp


2271
ZLFS32

Serratia sp.

4085
2627
88.1
globlastp


2272
ZLFS32

Serratia sp.

4086
2627
87
globlastp


2273
ZLFS32

Serratia sp.

4087
2627
86.1
globlastp


2274
ZLFS32

Serratia sp.

4088
2627
85
globlastp


2275
ZLFS32

Serratia sp.

4089
2627
84.6
globlastp


2276
ZLFS32

Serratia sp.

4090
2627
81.1
globlastp


2277
ZLFS32

Serratia sp.

4091
2627
80.9
globlastp


2278
ZLFS32

Serratia sp.

4092
2627
79.5
globlastp


2279
ZLFS32

Serratia sp.

4093
2627
78.6
globlastp


2280
ZLFS32

Serratia sp.

4094
2627
74.1
globlastp


2281
ZLFS32

Serratia sp.

4095
2627
73.7
globlastp


2282
ZLFS32

Yersinia sp.

4096
2627
72.4
globlastp


2283
ZLFS32

Pectobacterium sp.

4097
2627
70.1
globlastp


1793
BLFS137
Environmental sample
3617
2572
98.2
globlastp


1794
BLFS137

Rhodococcus sp.

3618
2572
94.7
globlastp


1795
BLFS137

Rhodococcus sp.

3619
2572
94.3
globlastp


1796
BLFS137
Environmental sample
3620
2572
81.1
globlastp


1797
BLFS137
Environmental sample
3621
2572
80.8
globlastp


1292
BLFS99
Environmental sample
3127
2535
98.9
globlastp


1293
BLFS99

Pantoea sp.

3128
2535
97.7
globlastp


1294
BLFS99
Environmental sample
3129
2535
96.6
globlastp


1295
BLFS99

Pantoea sp.

3130
2535
93.2
globlastp


1296
BLFS99

Pantoea sp.

3131
2535
90.9
globlastp


1297
BLFS99

Pantoea sp.

3132
2535
81.1
globlastp


1298
BLFS99
Environmental sample
3133
2535
80
globlastp


1299
BLFS99

Pantoea sp.

3134
2535
75
globlastp


1300
BLFS99
Environmental sample
3135
2535
73.3
globlastp


1301
BLFS99

Pantoea sp.

3136
2535
72.2
globlastp


1302
BLFS99

Pantoea sp.

3137
2535
71.6
globlastp


1303
BLFS99

Pantoea sp.

3138
2535
70.5
globlastp


1027
BLFS60

Streptomyces sp.

2870
2500
75.2
globlastp


1028
BLFS60

Streptomyces sp.

2871
2500
72.3
globlastp


1029
BLFS60

Streptomyces sp.

2872
2500
71.8
globlastp


2102
ZLFS17
Environmental sample
3916
2614
73.5
globlastp


971
BLFS39

Brevibacillus sp.

2819
2481
98.4
globlastp


972
BLFS39

Brevibacillus sp.

2820
2481
98.1
globlastp


973
BLFS39

Brevibacillus sp.

2821
2481
97.8
globlastp


2364
ZLFS39

Serratia sp.

4177
2634
98.8
globlastp


2365
ZLFS39

Serratia sp.

4178
2634
98.5
globlastp


2366
ZLFS39

Serratia sp.

4179
2634
98.3
globlastp


2367
ZLFS39

Serratia sp.

4180
2634
92.6
globlastp


2368
ZLFS39

Serratia sp.

4181
2634
91.4
globlastp


2369
ZLFS39
Multievos46 sp.
4182
2634
91.2
globlastp


2370
ZLFS39

Serratia sp.

4183
2634
91.1
globlastp


2371
ZLFS39
Environmental sample
4184
2634
90.9
globlastp


2372
ZLFS39

Serratia sp.

4185
2634
87.3
globlastp


2373
ZLFS39

Serratia sp.

4186
2634
87.1
globlastp


2374
ZLFS39

Serratia sp.

4187
2634
87
globlastp


2375
ZLFS39

Serratia sp.

4188
2634
86.8
globlastp


2376
ZLFS39

Serratia sp.

4189
2634
86.7
globlastp


2377
ZLFS39

Serratia sp.

4190
2634
86.5
globlastp


2378
ZLFS39
Environmental sample
4191
2634
86.4
globlastp


2379
ZLFS39

Serratia sp.

4192
2634
86.2
globlastp


2380
ZLFS39

Serratia sp.

4193
2634
86.1
globlastp


2381
ZLFS39

Serratia sp.

4194
2634
85.8
globlastp


2382
ZLFS39

Serratia sp.

4195
2634
85.6
globlastp


2383
ZLFS39

Serratia sp.

4196
2634
85.2
globlastp


2384
ZLFS39

Serratia sp.

4197
2634
85
globlastp


2385
ZLFS39

Serratia sp.

4198
2634
84.9
globlastp


2386
ZLFS39

Serratia sp.

4199
2634
84.4
globlastp


2387
ZLFS39

Serratia sp.

4200
2634
79.6
globlastp


2388
ZLFS39

Serratia sp.

4201
2634
79.2
globlastp


2209
ZLFS29

Pseudomonas sp.

4023
2624
95
globlastp


2210
ZLFS29

Pseudomonas sp.

4024
2624
94.1
globlastp


2211
ZLFS29

Pseudomonas sp.

4025
2624
93.1
globlastp


2212
ZLFS29

Pseudomonas sp.

4026
2624
92.2
globlastp


2213
ZLFS29

Pseudomonas sp.

4027
2624
91.6
globlastp


2214
ZLFS29

Pseudomonas sp.

4028
2624
90.8
globlastp


2215
ZLFS29

Pseudomonas sp.

4029
2624
89.1
globlastp


2216
ZLFS29
Environmental sample
4030
2624
88.1
globlastp


2217
ZLFS29

Pseudomonas sp.

4031
2624
87
globlastp


2218
ZLFS29

Pseudomonas sp.

4032
2624
86.4
globlastp


2219
ZLFS29

Pseudomonas sp.

4033
2624
85.7
globlastp


2220
ZLFS29

Pseudomonas sp.

4034
2624
84.1
globlastp


2221
ZLFS29

Pseudomonas sp.

4035
2624
83
globlastp


2222
ZLFS29
Environmental sample
4036
2624
82
globlastp


2223
ZLFS29
Environmental sample
4037
2624
81
globlastp


2224
ZLFS29

Pseudomonas sp.

4038
2624
80.3
globlastp


2225
ZLFS29
Environmental sample
4039
2624
79.1
globlastp


2226
ZLFS29

Pseudomonas sp.

4040
2624
78
globlastp


2227
ZLFS29
Environmental sample
4041
2624
77.4
globlastp


2228
ZLFS29

Pseudomonas sp.

4042
2624
76.1
globlastp


2229
ZLFS29

Pseudomonas sp.

4043
2624
75.3
globlastp


2230
ZLFS29

Pseudomonas sp.

4044
2624
74.4
globlastp


2231
ZLFS29

Pseudomonas sp.

4045
2624
73.4
globlastp


2232
ZLFS29

Pseudomonas sp.

4046
2624
72.1
globlastp


2233
ZLFS29

Pseudomonas sp.

4047
2624
71.1
globlastp


2234
ZLFS29

Pseudomonas sp.

4048
2624
70.2
globlastp


1986
BLFS153

Streptomyces sp.

3803
2588
98.6
globlastp


1987
BLFS153

Streptomyces sp.

3804
2588
93.3
globlastp


1988
BLFS153

Streptomyces sp.

3805
2588
71.7
globlastp


1989
BLFS153

Streptomyces sp.

3806
2588
70.9
globlastp


1990
BLFS153

Streptomyces sp.


2588
70.56
glotblastn


1991
BLFS153

Streptomyces sp.

3807
2588
70.4
globlastp


1992
BLFS153

Streptomyces sp.


2588
70.36
glotblastn


1993
BLFS153

Streptomyces sp.

3808
2588
70.1
globlastp


1994
BLFS153

Streptomyces sp.


2588
70
glotblastn


2086
ZLFS12

Collimonas sp.

3900
2609
81.4
globlastp


2087
ZLFS12

Collimonas sp.

3901
2609
81
globlastp


2088
ZLFS12

Collimonas sp.

3902
2609
73.7
globlastp


1574
BLFS120

Pseudomonas sp.

3399
2555
97.2
globlastp


1575
BLFS120

Pseudomonas sp.

3400
2555
84.8
globlastp


1576
BLFS120

Pseudomonas sp.

3401
2555
75.6
globlastp


2391
ZLFS45

Streptomyces sp.

4204
2637
98.2
globlastp


2392
ZLFS45

Streptomyces sp.

4205
2637
97.3
globlastp


2390
ZLFS45

Streptomyces sp.

4203
2637
96.4
globlastp


2389
ZLFS45

Streptomyces sp.

4202
2637
94
globlastp


2397
ZLFS45

Streptomyces sp.

4209
2637
90.2
globlastp


2394
ZLFS45

Streptomyces sp.

4207
2637
90.2
globlastp


2395
ZLFS45

Streptomyces sp.

4207
2637
90.2
globlastp


2393
ZLFS45

Streptomyces sp.

4206
2637
80.4
globlastp


2423
ZLFS45

Streptomyces sp.

4232
2637
73.2
globlastp


2398
ZLFS45

Streptomyces sp.

4210
2637
72.6
globlastp


2396
ZLFS45

Streptomyces sp.

4208
2637
72.6
globlastp


2399
ZLFS44

Streptomyces sp.

4211
2636
98.7
globlastp


2400
ZLFS44

Streptomyces sp.

4212
2636
98.1
globlastp


2401
ZLFS44

Streptomyces sp.

4213
2636
96.8
globlastp


2402
ZLFS44

Streptomyces sp.

4214
2636
75.8
globlastp


2403
ZLFS44

Streptomyces sp.

4215
2636
75.2
globlastp


2404
ZLFS44

Microtetraspora sp.

4216
2636
74.7
globlastp


2405
ZLFS44

Streptomyces sp.

4217
2636
74.5
globlastp


2406
ZLFS44

Streptomyces sp.

4218
2636
74.1
globlastp


2407
ZLFS44

Streptomyces sp.

4219
2636
73.9
globlastp


2408
ZLFS44

Streptomyces sp.


2636
73.89
glotblastn


2409
ZLFS44

Streptomyces sp.

4220
2636
73.4
globlastp


2410
ZLFS44

Streptomyces sp.

4221
2636
73.2
globlastp


2411
ZLFS44

Streptomyces sp.

4222
2636
72.8
globlastp


2412
ZLFS44

Streptomyces sp.

4223
2636
72.6
globlastp


2413
ZLFS44

Streptomyces sp.

4224
2636
72.2
globlastp


2414
ZLFS44
Environmental sample
4225
2636
72
globlastp


2415
ZLFS44

Streptomyces sp.


2636
71.52
glotblastn


2416
ZLFS44

Streptomyces sp.

4226
2636
71.5
globlastp


2417
ZLFS44

Streptomyces sp.

4227
2636
71.3
globlastp


2418
ZLFS44

Streptomyces sp.

4228
2636
71.1
globlastp


2419
ZLFS44

Streptomyces sp.

4229
2636
70.8
globlastp


2420
ZLFS44

Streptomyces sp.

4230
2636
70.7
globlastp


2421
ZLFS44

Streptomyces sp.

4231
2636
70.1
globlastp


2422
ZLFS44

Streptomyces sp.


2636
70.06
glotblastn


1022
BLFS57

Duganella sp.

2865
2497
99
globlastp


1023
BLFS57

Janthinobacterium sp.

2866
2497
98
globlastp


1024
BLFS57

Oxalobacteraceae sp.

2867
2497
85.3
globlastp


1368
BLFS105

Pantoea sp.

3194
2541
96.2
globlastp


1369
BLFS105
Environmental sample
3195
2541
92.4
globlastp


1370
BLFS105
Environmental sample
3196
2541
91.7
globlastp


1371
BLFS105
Environmental sample
3197
2541
87.9
globlastp


1372
BLFS105
Environmental sample
3198
2541
87.1
globlastp


1373
BLFS105
Environmental sample
3199
2541
86.4
globlastp


1374
BLFS105
Environmental sample
3200
2541
85.7
globlastp


1375
BLFS105
Environmental sample
3201
2541
84.8
globlastp


1376
BLFS105
Environmental sample
3202
2541
84.1
globlastp


1377
BLFS105

Pantoea sp.

3203
2541
82.6
globlastp


1378
BLFS105

Pantoea sp.

3204
2541
79.5
globlastp


1379
BLFS105

Erwinia sp.

3205
2541
75.8
globlastp


1380
BLFS105

Ewingella sp.

3206
2541
74.2
globlastp


1267
BLFS95

Paenibacillus sp.

3102
2531
97.4
globlastp


1268
BLFS95

Paenibacillus sp.

3103
2531
96.1
globlastp


1269
BLFS95

Paenibacillus sp.

3104
2531
90.8
globlastp


1270
BLFS95

Paenibacillus sp.

3105
2531
90.2
globlastp


1271
BLFS95

Paenibacillus sp.

3106
2531
89.5
globlastp


1272
BLFS95

Paenibacillus sp.

3107
2531
88.2
globlastp


1273
BLFS95

Paenibacillus sp.

3108
2531
78.4
globlastp


1274
BLFS95

Paenibacillus sp.

3109
2531
77.8
globlastp


1275
BLFS95

Paenibacillus sp.

3110
2531
75.2
globlastp


967
BLFS37

Brevibacillus sp.

2816
2479
99.1
globlastp


968
BLFS37

Brevibacillus sp.

2817
2479
98.2
globlastp


969
BLFS37

Brevibacillus sp.


2479
97.35
glotblastn


1570
BLFS119

Pseudomonas sp.

3395
2554
96.2
globlastp


1571
BLFS119

Pseudomonas sp.

3396
2554
95
globlastp


1572
BLFS119

Pseudomonas sp.

3397
2554
92.5
globlastp


1573
BLFS119

Pseudomonas sp.

3398
2554
91.2
globlastp


2284
ZLFS33

Serratia sp.

4098
2628
99.2
globlastp


2285
ZLFS33

Serratia sp.

4099
2628
96.4
globlastp


1025
BLFS58

Janthinobacterium sp.

2868
2498
97.4
globlastp


1026
BLFS58

Oxalobacteraceae sp.

2869
2498
83.5
globlastp


2002
PUB6

Bacillus sp.

3816
2595
98.6
globlastp


2003
PUB6

Bacillus sp.

3817
2595
98.5
globlastp


2004
PUB6

Bacillus sp.

3818
2595
98.4
globlastp


2005
PUB6

Bacillus sp.

3819
2595
97.9
globlastp


2006
PUB6

Bacillus sp.

3820
2595
96.1
globlastp


2007
PUB6

Bacillus sp.

3821
2595
94.7
globlastp


2008
PUB6

Bacillus sp.

3822
2595
94.2
globlastp


2009
PUB6

Bacillus sp.

3823
2595
93.2
globlastp


2010
PUB6

Bacillus sp.

3824
2595
92.6
globlastp


2011
PUB6

Bacillus sp.

3825
2595
92.1
globlastp


792
BLFS5

Streptomyces sp.

2644
2448
76.4
globlastp


793
BLFS5

Streptomyces sp.

2645
2448
75.5
globlastp


794
BLFS5

Kitasatospora sp.

2646
2448
74.5
globlastp


795
BLFS5

Kitasatospora sp.

2647
2448
73.8
globlastp


796
BLFS5

Streptomyces sp.

2648
2448
73.2
globlastp


797
BLFS5

Kitasatospora sp.

2649
2448
72
globlastp


798
BLFS5

Streptomyces sp.

2650
2448
71.1
globlastp


799
BLFS5

Streptomyces sp.

2651
2448
70.1
globlastp


989
BLFS45

Burkholderia sp.

2837
2486
97.9
globlastp


990
BLFS45
Environmental sample
2838
2486
82.3
globlastp


991
BLFS45

Burkholderia sp.

2839
2486
81.1
globlastp


992
BLFS45

Burkholderia sp.

2840
2486
80.2
globlastp


993
BLFS45

Burkholderia sp.

2841
2486
79.9
globlastp


994
BLFS45

Burkholderia sp.

2842
2486
76.1
globlastp


995
BLFS45

Burkholderia sp.

2843
2486
75.2
globlastp


996
BLFS45

Burkholderia sp.

2844
2486
74.6
globlastp


1304
BLFS100
Environmental sample
3139
2536
98.9
globlastp


1305
BLFS100
Environmental sample
3140
2536
97.7
globlastp


1306
BLFS100

Pantoea sp.

3141
2536
96.6
globlastp


1307
BLFS100
Environmental sample
3142
2536
95.4
globlastp


1308
BLFS100

Pantoea sp.

3143
2536
94.3
globlastp


1309
BLFS100

Pantoea sp.

3144
2536
88.5
globlastp


1310
BLFS100
Environmental sample
3145
2536
87.4
globlastp


1311
BLFS100

Pantoea sp.

3146
2536
85.1
globlastp


1312
BLFS100

Pantoea sp.

3147
2536
83.9
globlastp


1313
BLFS100

Pantoea sp.

3148
2536
81.8
globlastp


1314
BLFS100

Pantoea sp.

3149
2536
80.7
globlastp


1315
BLFS100

Pantoea sp.

3150
2536
78.4
globlastp


1316
BLFS100

Pantoea sp.

3151
2536
76.1
globlastp


1317
BLFS100

Pantoea sp.

3152
2536
74.4
globlastp


1318
BLFS100

Plautia sp.

3153
2536
71
globlastp


1319
BLFS100

Erwinia sp.

3154
2536
70.1
globlastp


1218
BLFS90

Paenibacillus sp.

3053
2526
99.3
globlastp


1219
BLFS90

Paenibacillus sp.

3054
2526
98.9
globlastp


1220
BLFS90

Paenibacillus sp.

3055
2526
96.1
globlastp


1221
BLFS90

Paenibacillus sp.

3056
2526
95.4
globlastp


1222
BLFS90

Paenibacillus sp.

3057
2526
93.6
globlastp


1223
BLFS90

Paenibacillus sp.

3058
2526
92.1
globlastp


1224
BLFS90

Paenibacillus sp.

3059
2526
91.4
globlastp


1225
BLFS90

Paenibacillus sp.

3060
2526
90.7
globlastp


1226
BLFS90

Paenibacillus sp.

3061
2526
72.9
globlastp


1011
BLFS55

Janthinobacterium sp.

2857
2495
95.5
globlastp


1012
BLFS55

Duganella sp.

2858
2495
78.5
globlastp


2306
ZLFS35

Serratia sp.

4120
2630
99
globlastp


2307
ZLFS35

Serratia sp.

4121
2630
98.8
globlastp


2308
ZLFS35

Serratia sp.

4122
2630
98.1
globlastp


2309
ZLFS35

Serratia sp.

4123
2630
88.3
globlastp


2310
ZLFS35

Serratia sp.

4124
2630
73.2
globlastp


1152
BLFS83

Paenibacillus sp.

2989
2519
99
globlastp


1153
BLFS83

Paenibacillus sp.

2990
2519
97.4
globlastp


1154
BLFS83

Paenibacillus sp.

2991
2519
97
globlastp


1155
BLFS83

Paenibacillus sp.

2992
2519
96.4
globlastp


1156
BLFS83

Paenibacillus sp.

2993
2519
95
globlastp


1157
BLFS83

Paenibacillus sp.

2994
2519
94.3
globlastp


1158
BLFS83

Paenibacillus sp.

2995
2519
93.2
globlastp


1159
BLFS83

Paenibacillus sp.

2996
2519
92.3
globlastp


1160
BLFS83

Paenibacillus sp.

2997
2519
80.6
globlastp


1161
BLFS83

Paenibacillus sp.

2998
2519
80
globlastp


1162
BLFS83

Paenibacillus sp.

2999
2519
79.6
globlastp


870
BLFS21

Bacillus sp.

2722
2464
99.3
globlastp


871
BLFS21

Bacillus sp.

2723
2464
83.1
globlastp


872
BLFS21

Bacillus sp.

2724
2464
79.7
globlastp


873
BLFS21

Bacillus sp.

2725
2464
71.3
globlastp


874
BLFS21

Bacillus sp.

2726
2464
70.5
globlastp


875
BLFS21

Bacillus sp.


2464
70.23
glotblastn


876
BLFS21

Bacillus sp.

2727
2464
70.2
globlastp


877
BLFS21

Bacillus sp.


2464
70.14
glotblastn


1685
BLFS129

Pseudomonas sp.

3509
2564
95.4
globlastp


1686
BLFS129

Pseudomonas sp.

3510
2564
95
globlastp


1687
BLFS129

Pseudomonas sp.

3511
2564
94.5
globlastp


1688
BLFS129

Pseudomonas sp.

3512
2564
93.1
globlastp


1689
BLFS129

Pseudomonas sp.

3513
2564
91.3
globlastp


1690
BLFS129

Pseudomonas sp.

3514
2564
90.8
globlastp


1691
BLFS129
Environmental sample
3515
2564
87.6
globlastp


1692
BLFS129

Pseudomonas sp.

3516
2564
83.9
globlastp


1693
BLFS129

Pseudomonas sp.

3517
2564
79.8
globlastp


1694
BLFS129

Pseudomonas sp.

3518
2564
76.6
globlastp


1695
BLFS129

Pseudomonas sp.

3519
2564
76.1
globlastp


920
BLFS32

Bacillus sp.

2770
2474
99
globlastp


921
BLFS32

Bacillus sp.

2771
2474
98
globlastp


922
BLFS32

Bacillus sp.

2772
2474
97.6
globlastp


923
BLFS32

Bacillus sp.

2773
2474
90.3
globlastp


924
BLFS32

Bacillus sp.

2774
2474
89.3
globlastp


925
BLFS32

Salinibacillus sp.

2775
2474
88.3
globlastp


926
BLFS32

Bacillus sp.

2776
2474
87.6
globlastp


927
BLFS32

Bacillus sp.

2777
2474
81.3
globlastp


928
BLFS32

Bacillus sp.

2778
2474
80.2
globlastp


929
BLFS32

Bacillus sp.

2779
2474
79.5
globlastp


930
BLFS32

Bacillus sp.

2780
2474
76.8
globlastp


931
BLFS32

Bacillus sp.

2781
2474
75.2
globlastp


932
BLFS32

Bacillus sp.

2782
2474
74.7
globlastp


933
BLFS32

Bacillus sp.

2783
2474
73.2
globlastp


934
BLFS32

Cohnella sp.

2784
2474
72.2
globlastp


935
BLFS32

Paenibacillus sp.

2785
2474
71.5
globlastp


936
BLFS32

Bacillus sp.

2786
2474
70.1
globlastp


1798
BLFS138

Bacillus sp.

3622
2573
98.7
globlastp


1799
BLFS138

Bacillus sp.

3623
2573
82.2
globlastp


1800
BLFS138

Bacillus sp.

3624
2573
81
globlastp


1801
BLFS138

Bacillus sp.

3625
2573
80.1
globlastp


1802
BLFS138

Bacillus sp.

3626
2573
79.4
globlastp


1803
BLFS138

Bacillus sp.

3627
2573
78.2
globlastp


1804
BLFS138

Bacillus sp.

3628
2573
77.1
globlastp


1805
BLFS138

Bacillus sp.

3629
2573
76.1
globlastp


1806
BLFS138

Bacillus sp.

3630
2573
74.3
globlastp


1807
BLFS138

Bacillus sp.

3631
2573
73.6
globlastp


1808
BLFS138

Bacillus sp.

3632
2573
72.5
globlastp


1809
BLFS138

Bacillus sp.

3633
2573
71.1
globlastp


1810
BLFS138

Bacillus sp.


2573
70
glotblastn


2103
ZLFS18
Environmental sample
3917
2615
75.5
globlastp


1999
BLFS158

Bacillus sp.

3813
2593
73.8
globlastp


1857
BLFS142

Bacillus sp.

3680
2577
98.1
globlastp


1858
BLFS142

Bacillus sp.

3681
2577
96.7
globlastp


1859
BLFS142

Bacillus sp.

3682
2577
89
globlastp


1860
BLFS142

Bacillus sp.

3683
2577
88.5
globlastp


1861
BLFS142

Bacillus sp.

3684
2577
85.2
globlastp


1862
BLFS142

Bacillus sp.

3685
2577
84.2
globlastp


1863
BLFS142

Bacillus sp.

3686
2577
83.3
globlastp


1864
BLFS142

Bacillus sp.

3687
2577
82.3
globlastp


1865
BLFS142

Bacillus sp.

3688
2577
80.4
globlastp


1866
BLFS142

Bacillus sp.

3689
2577
79.3
globlastp


1867
BLFS142

Bacillus sp.

3690
2577
77.1
globlastp


1247
BLFS93

Paenibacillus sp.

3082
2529
98.9
globlastp


1248
BLFS93

Paenibacillus sp.

3083
2529
98.3
globlastp


1249
BLFS93

Paenibacillus sp.

3084
2529
97.7
globlastp


1250
BLFS93

Paenibacillus sp.

3085
2529
96
globlastp


1251
BLFS93

Paenibacillus sp.

3086
2529
95.4
globlastp


1252
BLFS93

Paenibacillus sp.

3087
2529
94.9
globlastp


1253
BLFS93

Paenibacillus sp.

3088
2529
93.1
globlastp


1254
BLFS93

Paenibacillus sp.

3089
2529
77
globlastp


1255
BLFS93

Paenibacillus sp.

3090
2529
75.9
globlastp


1256
BLFS93

Paenibacillus sp.

3091
2529
74.1
globlastp


1257
BLFS93

Paenibacillus sp.

3092
2529
72.6
globlastp


2341
ZLFS38

Serratia sp.

4154
2633
98.7
globlastp


2342
ZLFS38

Serratia sp.

4155
2633
96.2
globlastp


2343
ZLFS38

Serratia sp.

4156
2633
95
globlastp


2344
ZLFS38

Serratia sp.

4157
2633
94.3
globlastp


2345
ZLFS38

Serratia sp.

4158
2633
93.1
globlastp


2346
ZLFS38

Serratia sp.

4159
2633
92.5
globlastp


2347
ZLFS38

Pectobacterium sp.

4160
2633
89.3
globlastp


2348
ZLFS38

Hafnia sp.

4161
2633
88.1
globlastp


2349
ZLFS38

Dickeya sp.

4162
2633
87.4
globlastp


2350
ZLFS38

Dickeya sp.

4163
2633
86.2
globlastp


2351
ZLFS38

Enterobacter sp.

4164
2633
85
globlastp


2352
ZLFS38

Achromobacter sp.

4165
2633
84.3
globlastp


2353
ZLFS38
Environmental sample
4166
2633
83
globlastp


2354
ZLFS38

Serratia sp.

4167
2633
82.2
globlastp


2355
ZLFS38
Environmental sample
4168
2633
81.1
globlastp


2356
ZLFS38
Environmental sample
4169
2633
80
globlastp


2357
ZLFS38

Cronobacter sp.

4170
2633
79.5
globlastp


2358
ZLFS38

Enterobacter sp.

4171
2633
78.1
globlastp


2359
ZLFS38

Arsenophonus sp.

4172
2633
77.4
globlastp


2360
ZLFS38

Pantoea sp.

4173
2633
76.1
globlastp


2361
ZLFS38

Vibrio sp.

4174
2633
72
globlastp


2362
ZLFS38

Sodalis sp.

4175
2633
71
globlastp


2363
ZLFS38

Vibrio sp.

4176
2633
70
globlastp


803
BLFS9

Amycolatopsis sp.

2655
2452
92.3
globlastp


804
BLFS9

Amycolatopsis sp.

2656
2452
78.6
globlastp


805
BLFS9

Streptomyces sp.

2657
2452
71.8
globlastp


806
BLFS9

Streptomyces sp.

2658
2452
71.4
globlastp


807
BLFS9

Streptomyces sp.

2659
2452
70.9
globlastp


808
BLFS9

Streptomyces sp.

2660
2452
70.1
globlastp


1090
BLFS74

Paenibacillus sp.

2930
2512
99.4
globlastp


1091
BLFS74

Paenibacillus sp.

2931
2512
99.1
globlastp


1092
BLFS74

Paenibacillus sp.

2932
2512
98.6
globlastp


1093
BLFS74

Paenibacillus sp.

2933
2512
97.4
globlastp


1094
BLFS74

Paenibacillus sp.

2934
2512
97.1
globlastp


1095
BLFS74

Paenibacillus sp.

2935
2512
96
globlastp


1096
BLFS74

Paenibacillus sp.

2936
2512
95.7
globlastp


1097
BLFS74

Paenibacillus sp.

2937
2512
95.4
globlastp


1098
BLFS74

Paenibacillus sp.

2938
2512
92.2
globlastp


1099
BLFS74

Paenibacillus sp.

2939
2512
91.7
globlastp


1100
BLFS74

Paenibacillus sp.

2940
2512
90.5
globlastp


1101
BLFS74

Paenibacillus sp.

2941
2512
89.7
globlastp


1102
BLFS74

Paenibacillus sp.

2942
2512
89.1
globlastp


1103
BLFS74

Paenibacillus sp.

2943
2512
87.1
globlastp


2140
ZLFS23

Curtobacterium sp.

3954
2619
95.8
globlastp


2141
ZLFS23

Curtobacterium sp.

3955
2619
95.4
globlastp


2142
ZLFS23

Curtobacterium sp.

3956
2619
90.5
globlastp


2143
ZLFS23

Curtobacterium sp.

3957
2619
80.5
globlastp


2144
ZLFS23
Environmental sample
3958
2619
76.3
globlastp


2145
ZLFS23
Environmental sample
3959
2619
74.5
globlastp


2146
ZLFS23
Environmental sample
3960
2619
71.8
globlastp


1420
BLFS108

Pantoea sp.

3246
2544
99.1
globlastp


1421
BLFS108
Environmental sample
3247
2544
98.3
globlastp


1422
BLFS108

Klebsiella sp.

3248
2544
76.9
globlastp


1423
BLFS108

Citrobacter sp.

3249
2544
76.1
globlastp


1424
BLFS108

Meta sp.

3250
2544
75.2
globlastp


1425
BLFS108
Environmental sample
3251
2544
74.4
globlastp


1426
BLFS108

Salmonella sp.

3252
2544
73.5
globlastp


1427
BLFS108

Escherichia sp.

3253
2544
72
globlastp


1428
BLFS108

Kluyvera sp.

3254
2544
71.8
globlastp


2311
ZLFS36

Serratia sp.

4125
2631
99.2
globlastp


2170
ZLFS27

Pseudomonas sp.

3984
2622
93
globlastp


2171
ZLFS27

Pseudomonas sp.

3985
2622
92
globlastp


2172
ZLFS27

Pseudomonas sp.

3986
2622
91
globlastp


2173
ZLFS27

Pseudomonas sp.

3987
2622
90.8
globlastp


2174
ZLFS27

Pseudomonas sp.

3988
2622
89
globlastp


2175
ZLFS27

Pseudomonas sp.

3989
2622
88
globlastp


2176
ZLFS27

Pseudomonas sp.

3990
2622
87.1
globlastp


2177
ZLFS27

Pseudomonas sp.

3991
2622
86.1
globlastp


2178
ZLFS27

Pseudomonas sp.

3992
2622
85.1
globlastp


2179
ZLFS27

Pseudomonas sp.

3993
2622
84.1
globlastp


2180
ZLFS27

Pseudomonas sp.

3994
2622
83.1
globlastp


2181
ZLFS27

Pseudomonas sp.

3995
2622
82.4
globlastp


2182
ZLFS27

Pseudomonas sp.

3996
2622
81.9
globlastp


2183
ZLFS27

Pseudomonas sp.

3997
2622
79.1
globlastp


2184
ZLFS27

Pseudomonas sp.

3998
2622
78
globlastp


2185
ZLFS27

Pseudomonas sp.

3999
2622
77.1
globlastp


2186
ZLFS27

Pseudomonas sp.

4000
2622
75.9
globlastp


2187
ZLFS27

Pseudomonas sp.

4001
2622
75.3
globlastp


2188
ZLFS27

Pseudomonas sp.

4002
2622
74.2
globlastp


2189
ZLFS27

Pseudomonas sp.

4003
2622
73.8
globlastp


2190
ZLFS27
Environmental sample
4004
2622
72
globlastp


2191
ZLFS27

Pseudomonas sp.

4005
2622
71.5
globlastp


2192
ZLFS27

Pseudomonas sp.

4006
2622
70.3
globlastp


2193
ZLFS27

Pseudomonas sp.

4007
2622
70.1
globlastp


1824
BLFS140

Salinibacillus sp.

3647
2575
99.4
globlastp


1825
BLFS140

Bacillus sp.

3648
2575
98.4
globlastp


1826
BLFS140

Bacillus sp.

3649
2575
95.6
globlastp


1827
BLFS140

Bacillus sp.

3650
2575
88
globlastp


1828
BLFS140

Bacillus sp.

3651
2575
87.1
globlastp


1829
BLFS140

Bacillus sp.

3652
2575
86.1
globlastp


1830
BLFS140

Bacillus sp.

3653
2575
85.2
globlastp


1831
BLFS140

Bacillus sp.

3654
2575
85
globlastp


1832
BLFS140

Bacillus sp.

3655
2575
84.9
globlastp


1833
BLFS140

Bacillus sp.

3656
2575
84.7
globlastp


1834
BLFS140

Bacillus sp.

3657
2575
81.6
globlastp


1835
BLFS140

Bacillus sp.

3658
2575
81.5
globlastp


1836
BLFS140

Bacillus sp.

3659
2575
74
globlastp


1837
BLFS140

Bacillus sp.

3660
2575
73.8
globlastp


2102
ZLFS48
Environmental sample
3916
2639
79.4
globlastp


982
BLFS42

Burkholderia sp.

2830
2483
97.6
globlastp


983
BLFS42

Burkholderia sp.

2831
2483
97.2
globlastp


1927
BLFS147

Salinibacillus sp.

3750
2582
98.9
globlastp


1928
BLFS147

Bacillus sp.

3751
2582
97.7
globlastp


1929
BLFS147

Bacillus sp.

3752
2582
95.7
globlastp


1930
BLFS147

Bacillus sp.

3753
2582
88
globlastp


1931
BLFS147

Bacillus sp.

3754
2582
87.3
globlastp


1932
BLFS147

Bacillus sp.

3755
2582
86
globlastp


1933
BLFS147

Bacillus sp.

3756
2582
85
globlastp


1934
BLFS147

Bacillus sp.

3757
2582
84.8
globlastp


1935
BLFS147

Bacillus sp.

3758
2582
74.1
globlastp


1936
BLFS147

Bacillus sp.

3759
2582
73.9
globlastp


1170
BLFS85

Paenibacillus sp.

3007
2521
99.4
globlastp


1171
BLFS85

Paenibacillus sp.

3008
2521
98.3
globlastp


1172
BLFS85

Paenibacillus sp.

3009
2521
97.6
globlastp


1173
BLFS85

Paenibacillus sp.

3010
2521
97
globlastp


1174
BLFS85

Paenibacillus sp.

3011
2521
96.6
globlastp


1175
BLFS85

Paenibacillus sp.

3012
2521
95.3
globlastp


1176
BLFS85

Paenibacillus sp.

3013
2521
93.2
globlastp


1177
BLFS85

Paenibacillus sp.

3014
2521
92.7
globlastp


1178
BLFS85

Paenibacillus sp.

3015
2521
91
globlastp


1179
BLFS85

Paenibacillus sp.

3016
2521
90.4
globlastp


1180
BLFS85

Paenibacillus sp.

3017
2521
89.1
globlastp


1552
BLFS118

Pseudomonas sp.

3377
2553
98.8
globlastp


1553
BLFS118

Pseudomonas sp.

3378
2553
98.5
globlastp


1554
BLFS118

Pseudomonas sp.

3379
2553
98.2
globlastp


1555
BLFS118

Pseudomonas sp.

3380
2553
97.7
globlastp


1556
BLFS118

Pseudomonas sp.

3381
2553
97.1
globlastp


1557
BLFS118

Pseudomonas sp.

3382
2553
96.8
globlastp


1558
BLFS118

Pseudomonas sp.

3383
2553
96.2
globlastp


1559
BLFS118

Pseudomonas sp.

3384
2553
88
globlastp


1560
BLFS118

Pseudomonas sp.

3385
2553
85.9
globlastp


1561
BLFS118

Pseudomonas sp.

3386
2553
78
globlastp


1562
BLFS118

Pseudomonas sp.

3387
2553
77.1
globlastp


1563
BLFS118

Pseudomonas sp.

3388
2553
76
globlastp


1564
BLFS118
Environmental sample
3389
2553
75.1
globlastp


1565
BLFS118

Pseudomonas sp.

3390
2553
74
globlastp


1566
BLFS118

Pseudomonas sp.

3391
2553
73
globlastp


1567
BLFS118

Pseudomonas sp.

3392
2553
72.1
globlastp


1568
BLFS118
Environmental sample
3393
2553
71
globlastp


1569
BLFS118

Pseudomonas sp.

3394
2553
70.1
globlastp


1357
BLFS104

Pantoea sp.

3183
2540
99.2
globlastp


1358
BLFS104

Pantoea sp.

3184
2540
98.3
globlastp


1359
BLFS104
Environmental sample
3185
2540
95
globlastp


1360
BLFS104

Pantoea sp.

3186
2540
94.1
globlastp


1361
BLFS104

Pantoea sp.

3187
2540
93.3
globlastp


1362
BLFS104

Pantoea sp.

3188
2540
90.8
globlastp


1363
BLFS104
Environmental sample
3189
2540
89.1
globlastp


1364
BLFS104

Pantoea sp.

3190
2540
84.9
globlastp


1365
BLFS104

Pantoea sp.

3191
2540
78.2
globlastp


1366
BLFS104
Environmental sample
3192
2540
77.3
globlastp


1367
BLFS104

Pantoea sp.

3193
2540
73.9
globlastp


1381
BLFS106
Environmental sample
3207
2542
99
globlastp


1382
BLFS106

Pantoea sp.

3208
2542
98
globlastp


1383
BLFS106

Pantoea sp.

3209
2542
97
globlastp


1384
BLFS106

Pantoea sp.

3210
2542
94.4
globlastp


1385
BLFS106
Environmental sample
3211
2542
92.1
globlastp


1386
BLFS106

Pantoea sp.

3212
2542
88.1
globlastp


1387
BLFS106

Pantoea sp.

3213
2542
86.9
globlastp


1388
BLFS106

Pantoea sp.

3214
2542
83.2
globlastp


1389
BLFS106

Pantoea sp.

3215
2542
82.2
globlastp


1390
BLFS106

Pantoea sp.

3216
2542
80.2
globlastp


1391
BLFS106
Environmental sample
3217
2542
79.2
globlastp


1392
BLFS106

Pantoea sp.

3218
2542
78.2
globlastp


1393
BLFS106

Plautia sp.

3219
2542
78.1
globlastp


1394
BLFS106

Pantoea sp.

3220
2542
77.2
globlastp


1395
BLFS106

Cedecea sp.

3221
2542
76.2
globlastp


1396
BLFS106

Enterobacter sp.

3222
2542
75.2
globlastp


1397
BLFS106
Environmental sample
3223
2542
74.3
globlastp


1398
BLFS106
Environmental sample
3224
2542
73.3
globlastp


1399
BLFS106
Environmental sample
3225
2542
72.3
globlastp


1400
BLFS106

Citrobacter sp.

3226
2542
71.3
globlastp


1401
BLFS106

Enterobacter sp.

3227
2542
70.3
globlastp


1238
BLFS92

Paenibacillus sp.

3073
2528
99.2
globlastp


1239
BLFS92

Paenibacillus sp.

3074
2528
97.7
globlastp


1240
BLFS92

Paenibacillus sp.

3075
2528
97.2
globlastp


1241
BLFS92

Paenibacillus sp.

3076
2528
96.6
globlastp


1242
BLFS92

Paenibacillus sp.

3077
2528
96.1
globlastp


1243
BLFS92

Paenibacillus sp.

3078
2528
94.3
globlastp


1244
BLFS92

Paenibacillus sp.

3079
2528
94
globlastp


1245
BLFS92

Paenibacillus sp.

3080
2528
93.5
globlastp


1246
BLFS92

Paenibacillus sp.

3081
2528
92
globlastp


1868
BLFS143

Salinibacillus sp.

3691
2578
99.4
globlastp


1869
BLFS143

Bacillus sp.

3692
2578
98.2
globlastp


1870
BLFS143

Bacillus sp.

3693
2578
96.1
globlastp


1871
BLFS143

Bacillus sp.

3694
2578
95.2
globlastp


1872
BLFS143

Bacillus sp.

3695
2578
94.6
globlastp


1873
BLFS143

Bacillus sp.

3696
2578
87.5
globlastp


1874
BLFS143

Bacillus sp.

3697
2578
86.9
globlastp


1875
BLFS143

Bacillus sp.

3698
2578
79.1
globlastp


1876
BLFS143

Bacillus sp.

3699
2578
78.5
globlastp


1877
BLFS143

Bacillus sp.

3700
2578
74.1
globlastp


1878
BLFS143

Bacillus sp.

3701
2578
72.9
globlastp


1879
BLFS143

Bacillus sp.

3702
2578
71.3
globlastp


1880
BLFS143

Bacillus sp.

3703
2578
70.4
globlastp


1881
BLFS143

Bacillus sp.

3704
2578
70.1
globlastp


800
BLFS8

Amycolatopsis sp.

2652
2451
98.3
globlastp


801
BLFS8

Amycolatopsis sp.

2653
2451
95.7
globlastp


802
BLFS8

Amycolatopsis sp.

2654
2451
77.1
globlastp


961
BLFS36

Bradyrhizobium sp.

2811
2478
82.9
globlastp


962
BLFS36

Bradyrhizobium sp.

2812
2478
78.3
globlastp


963
BLFS36

Bradyrhizobium sp.


2478
76.83
glotblastn


964
BLFS36

Bradyrhizobium sp.

2813
2478
76.8
globlastp


965
BLFS36

Bradyrhizobium sp.

2814
2478
75.6
globlastp


966
BLFS36

Bradyrhizobium sp.

2815
2478
74.4
globlastp


1402
BLFS107
Environmental sample
3228
2543
97.9
globlastp


1403
BLFS107

Pantoea sp.

3229
2543
97.4
globlastp


1404
BLFS107

Pantoea sp.

3230
2543
96.8
globlastp


1405
BLFS107
Environmental sample
3231
2543
95.8
globlastp


1406
BLFS107

Pantoea sp.

3232
2543
94.2
globlastp


1407
BLFS107

Pantoea sp.

3233
2543
88.4
globlastp


1408
BLFS107
Environmental sample
3234
2543
86.3
globlastp


1409
BLFS107
Environmental sample
3235
2543
85.8
globlastp


1410
BLFS107
Environmental sample
3236
2543
85.7
globlastp


1411
BLFS107
Environmental sample
3237
2543
85.3
globlastp


1412
BLFS107

Pantoea sp.

3238
2543
84.8
globlastp


1413
BLFS107

Pantoea sp.

3239
2543
84.7
globlastp


1414
BLFS107

Pantoea sp.

3240
2543
78.3
globlastp


1415
BLFS107

Erwinia sp.

3241
2543
74.6
globlastp


1416
BLFS107

Erwinia sp.

3242
2543
73.5
globlastp


1417
BLFS107

Erwinia sp.

3243
2543
72.3
globlastp


1418
BLFS107

Pantoea sp.

3244
2543
72
globlastp


1419
BLFS107
Environmental sample
3245
2543
70.4
globlastp


866
BLFS20

Bacillus sp.

2718
2463
99.1
globlastp


867
BLFS20

Bacillus sp.

2719
2463
98.2
globlastp


868
BLFS20

Bacillus sp.

2720
2463
97.2
globlastp


869
BLFS20

Bacillus sp.

2721
2463
96.3
globlastp


2012
PUB65

Variovorax sp.

3826
2598
98.9
globlastp


2013
PUB65

Variovorax sp.

3827
2598
94.8
globlastp


2014
PUB65

Variovorax sp.

3828
2598
94.5
globlastp


2015
PUB65

Variovorax sp.

3829
2598
91.4
globlastp


2016
PUB65

Variovorax sp.

3830
2598
91.3
globlastp


2017
PUB65

Variovorax sp.

3831
2598
90.6
globlastp


2018
PUB65

Variovorax sp.

3832
2598
90.5
globlastp


2019
PUB65

Variovorax sp.

3833
2598
90.3
globlastp


2020
PUB65
Environmental sample
3834
2598
90.1
globlastp


2021
PUB65
Environmental sample
3835
2598
87.1
globlastp


2022
PUB65

Variovorax sp.

3836
2598
84.5
globlastp


2023
PUB65

Variovorax sp.

3837
2598
84.3
globlastp


2024
PUB65
Environmental sample
3838
2598
78.4
globlastp


2025
PUB65
Environmental sample
3839
2598
77.9
globlastp


2026
PUB65

Variovorax sp.

3840
2598
77.4
globlastp


2027
PUB65

Acidovorax sp.

3841
2598
75.5
globlastp


2028
PUB65
Environmental sample
3842
2598
75.4
globlastp


2029
PUB65

Acidovorax sp.

3843
2598
75.2
globlastp


2030
PUB65
Environmental sample
3844
2598
74.5
globlastp


2031
PUB65
Environmental sample
3845
2598
72.3
globlastp


790
BLFS1

Streptomyces sp.

2642
2444
72.8
globlastp


1976
BLFS150

Pseudomonas sp.


2585
95.9
glotblastn


1977
BLFS150

Xanthomonas sp.


2585
77.05
glotblastn


1978
BLFS150

Xanthomonas sp.


2585
75.41
glotblastn


1979
BLFS150

Xanthomonas sp.


2585
72.95
glotblastn


1980
BLFS150

Xanthomonas sp.


2585
71.31
glotblastn


2081
ZLFS8

Collimonas sp.

3895
2605
78.5
globlastp


2082
ZLFS8

Collimonas sp.

3896
2605
78.3
globlastp


952
BLFS35

Bradyrhizohium sp.

2802
2477
96.3
globlastp


953
BLFS35

Mesorhizohium sp.

2803
2477
89.8
globlastp


954
BLFS35

Bradyrhizohium sp.

2804
2477
87
globlastp


955
BLFS35

Bradyrhizohium sp.

2805
2477
86
globlastp


956
BLFS35

Bradyrhizohium sp.

2806
2477
76.8
globlastp


957
BLFS35

Bradyrhizohium sp.

2807
2477
75.5
globlastp


958
BLFS35

Bradyrhizohium sp.

2808
2477
75
globlastp


959
BLFS35

Bradyrhizohium sp.

2809
2477
71.2
globlastp


960
BLFS35

Bradyrhizohium sp.

2810
2477
70.2
globlastp


1429
BLFS109

Pantoea sp.

3255
2545
89.2
globlastp


1430
BLFS109

Pantoea sp.

3256
2545
76.2
globlastp


1320
BLFS101
Environmental sample
3155
2537
95.2
globlastp


1321
BLFS101

Pantoea sp.

3156
2537
94
globlastp


1322
BLFS101

Pantoea sp.

3157
2537
87.3
globlastp


1323
BLFS101
Environmental sample
3158
2537
86.1
globlastp


1324
BLFS101
Environmental sample
3159
2537
85.3
globlastp


1325
BLFS101

Pantoea sp.

3160
2537
84.1
globlastp


1326
BLFS101

Erwinia sp.

3161
2537
79.3
globlastp


1327
BLFS101

Serratia sp.

3162
2537
78.1
globlastp


1328
BLFS101

Erwinia sp.

3163
2537
77.3
globlastp


1329
BLFS101
Environmental sample
3164
2537
76.1
globlastp


1330
BLFS101

Klebsiella sp.

3165
2537
75
globlastp


1331
BLFS101

Yersinia sp.

3166
2537
74.1
globlastp


1332
BLFS101
Environmental sample
3167
2537
73
globlastp


1333
BLFS101

Rahnella sp.

3168
2537
72.1
globlastp


1334
BLFS101

Enterobacter sp.

3169
2537
71
globlastp


1335
BLFS101

Xenorhabdus sp.

3170
2537
70.1
globlastp


1607
BLFS123

Pseudomonas sp.

3431
2558
96.2
globlastp


1608
BLFS123

Pseudomonas sp.

3432
2558
95.9
globlastp


1609
BLFS123

Pseudomonas sp.

3433
2558
79.8
globlastp


1610
BLFS123

Pseudomonas sp.

3434
2558
79.5
globlastp


974
BLFS41

Burkholderia sp.

2822
2482
99.2
globlastp


975
BLFS41

Burkholderia sp.

2823
2482
98.9
globlastp


976
BLFS41

Burkholderia sp.

2824
2482
76.3
globlastp


977
BLFS41
Environmental sample
2825
2482
75.1
globlastp


978
BLFS41

Burkholderia sp.

2826
2482
74
globlastp


979
BLFS41

Burkholderia sp.

2827
2482
73.1
globlastp


980
BLFS41

Burkholderia sp.

2828
2482
72.9
globlastp


981
BLFS41

Burkholderia sp.

2829
2482
71.3
globlastp


1064
BLFS67

Janthinobacterium sp.

2906
2506
98.6
globlastp


1065
BLFS67

Janthinobacterium sp.

2907
2506
93.8
globlastp


1066
BLFS67

Janthinobacterium sp.

2908
2506
92.5
globlastp


1276
BLFS98

Pantoea sp.

3111
2534
96.3
globlastp


1277
BLFS98
Environmental sample
3112
2534
95.3
globlastp


1278
BLFS98

Pantoea sp.

3113
2534
93
globlastp


1279
BLFS98
Environmental sample
3114
2534
91.4
globlastp


1280
BLFS98
Environmental sample
3115
2534
89.8
globlastp


1281
BLFS98

Pantoea sp.

3116
2534
83.3
globlastp


1282
BLFS98
Environmental sample
3117
2534
82.9
globlastp


1283
BLFS98

Pantoea sp.

3118
2534
81.5
globlastp


1284
BLFS98
Environmental sample
3119
2534
79.4
globlastp


1285
BLFS98

Pantoea sp.

3120
2534
77.2
globlastp


1286
BLFS98

Pantoea sp.

3121
2534
76.1
globlastp


1287
BLFS98
Environmental sample
3122
2534
75.3
globlastp


1288
BLFS98

Pantoea sp.

3123
2534
74
globlastp


1289
BLFS98
Environmental sample
3124
2534
72.6
globlastp


1290
BLFS98

Erwinia sp.

3125
2534
71.6
globlastp


1291
BLFS98
Environmental sample
3126
2534
70.2
globlastp


898
BLFS26

Bacillus sp.

2748
2469
99
globlastp


899
BLFS26

Bacillus sp.

2749
2469
98.4
globlastp


900
BLFS26

Bacillus sp.

2750
2469
97.9
globlastp


901
BLFS26

Bacillus sp.

2751
2469
96.3
globlastp


902
BLFS26

Bacillus sp.

2752
2469
72.3
globlastp


1838
BLFS141

Salinibacillus sp.

3661
2576
98.1
globlastp


1839
BLFS141

Bacillus sp.

3662
2576
97.5
globlastp


1840
BLFS141

Bacillus sp.

3663
2576
95.9
globlastp


1841
BLFS141

Bacillus sp.

3664
2576
90.1
globlastp


1842
BLFS141

Bacillus sp.

3665
2576
89
globlastp


1843
BLFS141

Bacillus sp.

3666
2576
88.1
globlastp


1844
BLFS141

Bacillus sp.

3667
2576
87.2
globlastp


1845
BLFS141

Bacillus sp.

3668
2576
86.6
globlastp


1846
BLFS141

Bacillus sp.

3669
2576
86.4
globlastp


1847
BLFS141

Bacillus sp.

3670
2576
83.7
globlastp


1848
BLFS141

Bacillus sp.

3671
2576
82.6
globlastp


1849
BLFS141

Bacillus sp.

3672
2576
77.1
globlastp


1850
BLFS141

Bacillus sp.

3673
2576
76
globlastp


1851
BLFS141

Bacillus sp.

3674
2576
75.6
globlastp


1852
BLFS141

Bacillus sp.

3675
2576
74
globlastp


1853
BLFS141

Bacillus sp.

3676
2576
73.2
globlastp


1854
BLFS141

Bacillus sp.

3677
2576
72.4
globlastp


1855
BLFS141

Bacillus sp.

3678
2576
71.5
globlastp


1856
BLFS141

Paenibacillus sp.

3679
2576
70.4
globlastp


1198
BLFS88

Paenibacillus sp.

3035
2524
99.2
globlastp


1199
BLFS88

Paenibacillus sp.

3036
2524
96.9
globlastp


1200
BLFS88

Paenibacillus sp.

3037
2524
95.3
globlastp


1201
BLFS88

Paenibacillus sp.

3038
2524
95
globlastp


1202
BLFS88

Paenibacillus sp.

3039
2524
94.4
globlastp


1203
BLFS88

Paenibacillus sp.

3040
2524
93.6
globlastp


1204
BLFS88

Paenibacillus sp.

3041
2524
92.5
globlastp


1205
BLFS88

Paenibacillus sp.

3042
2524
88.3
globlastp


1206
BLFS88

Paenibacillus sp.

3043
2524
72.9
globlastp


1010
BLFS53

Dickeya sp.

2856
2494
99
globlastp


878
BLFS22

Bacillus sp.

2728
2465
99.3
globlastp


879
BLFS22

Bacillus sp.

2729
2465
97.8
globlastp


880
BLFS22

Bacillus sp.

2730
2465
97.1
globlastp


881
BLFS22

Bacillus sp.

2731
2465
96.4
globlastp


882
BLFS22

Bacillus sp.

2732
2465
95.6
globlastp


883
BLFS22

Bacillus sp.

2733
2465
94.9
globlastp


819
BLFS12

Arthrobacter sp.

2671
2455
98.9
globlastp


820
BLFS12

Arthrobacter sp.

2672
2455
92.3
globlastp


1960
BLFS149

Serratia sp.

3782
2584
98.8
globlastp


1961
BLFS149

Serratia sp.

3783
2584
97
globlastp


1962
BLFS149

Serratia sp.

3784
2584
93.8
globlastp


1963
BLFS149

Serratia sp.

3785
2584
91.7
globlastp


1964
BLFS149

Serratia sp.

3786
2584
84
globlastp


1965
BLFS149

Serratia sp.

3787
2584
83.8
globlastp


1966
BLFS149

Serratia sp.

3788
2584
82
globlastp


1967
BLFS149

Serratia sp.

3789
2584
81.3
globlastp


1968
BLFS149

Serratia sp.

3790
2584
78
globlastp


1969
BLFS149

Serratia sp.

3791
2584
77.8
globlastp


1970
BLFS149

Yersinia sp.

3792
2584
76
globlastp


1971
BLFS149

Yersinia sp.

3793
2584
75
globlastp


1972
BLFS149

Yersinia sp.

3794
2584
74
globlastp


1973
BLFS149

Yersinia sp.

3795
2584
73.1
globlastp


1974
BLFS149

Edwardsiella sp.

3796
2584
72
globlastp


1975
BLFS149
Environmental sample
3797
2584
71.2
globlastp


1984
BLFS152

Streptomyces sp.

3801
2587
82.9
globlastp


1985
BLFS152

Streptomyces sp.

3802
2587
78.9
globlastp


1995
BLFS154

Streptomyces sp.

3809
2589
98.2
globlastp


1996
BLFS154

Streptomyces sp.

3810
2589
72.2
globlastp


1432
BLFS111

Chromobacterium sp.

3258
2547
97
globlastp


1433
BLFS111

Chromobacterium sp.

3259
2547
89.9
globlastp


1434
BLFS111

Chromobacterium sp.

3260
2547
84.4
globlastp


1435
BLFS111

Chromobacterium sp.

3261
2547
83.4
globlastp


1436
BLFS111

Paludibacterium sp.

3262
2547
71.4
globlastp


791
BLFS2

Alkanindiges sp.

2643
2445
91.7
globlastp


1644
BLFS126

Pseudomonas sp.

3468
2561
98.3
globlastp


1645
BLFS126

Pseudomonas sp.

3469
2561
96.7
globlastp


1646
BLFS126

Pseudomonas sp.

3470
2561
92.6
globlastp


1647
BLFS126

Pseudomonas sp.

3471
2561
91.7
globlastp


1648
BLFS126

Pseudomonas sp.

3472
2561
90.9
globlastp


1649
BLFS126

Pseudomonas sp.

3473
2561
80.3
globlastp


1650
BLFS126

Pseudomonas sp.

3474
2561
75.6
globlastp


1651
BLFS126

Pseudomonas sp.

3475
2561
73.2
globlastp


2286
ZLFS34

Serratia sp.

4100
2629
99
globlastp


2287
ZLFS34

Serratia sp.

4101
2629
98.4
globlastp


2288
ZLFS34
Multievos46 sp.
4102
2629
97
globlastp


2289
ZLFS34

Serratia sp.

4103
2629
96
globlastp


2290
ZLFS34

Serratia sp.

4104
2629
95.9
globlastp


2291
ZLFS34

Serratia sp.

4105
2629
93
globlastp


2292
ZLFS34

Serratia sp.

4106
2629
92.1
globlastp


2293
ZLFS34

Serratia sp.

4107
2629
91.4
globlastp


2294
ZLFS34

Serratia sp.

4108
2629
88.1
globlastp


2295
ZLFS34

Serratia sp.

4109
2629
87
globlastp


2296
ZLFS34

Serratia sp.

4110
2629
84.3
globlastp


2297
ZLFS34

Xenorhabdus sp.

4111
2629
83.5
globlastp


2298
ZLFS34

Xenorhabdus sp.

4112
2629
82
globlastp


2299
ZLFS34

Xenorhabdus sp.

4113
2629
80.6
globlastp


2300
ZLFS34

Yersinia sp.

4114
2629
78.8
globlastp


2301
ZLFS34

Yersinia sp.

4115
2629
75
globlastp


2302
ZLFS34
Environmental sample
4116
2629
74.5
globlastp


2303
ZLFS34

Rahnella sp.

4117
2629
73.1
globlastp


2304
ZLFS34

Ewingella sp.

4118
2629
71.5
globlastp


2305
ZLFS34

Rhodanobacter sp.

4119
2629
70.1
globlastp


1907
BLFS146

Bacillus sp.

3730
2581
98.8
globlastp


1908
BLFS146

Bacillus sp.

3731
2581
97.2
globlastp


1909
BLFS146

Bacillus sp.

3732
2581
96
globlastp


1910
BLFS146

Bacillus sp.

3733
2581
95.4
globlastp


1911
BLFS146

Bacillus sp.

3734
2581
94
globlastp


1912
BLFS146

Bacillus sp.

3735
2581
93.4
globlastp


1913
BLFS146

Bacillus sp.

3736
2581
92.9
globlastp


1914
BLFS146

Bacillus sp.

3737
2581
91.7
globlastp


1915
BLFS146

Bacillus sp.

3738
2581
89.1
globlastp


1916
BLFS146

Bacillus sp.

3739
2581
85
globlastp


1917
BLFS146

Bacillus sp.

3740
2581
84.7
globlastp


1918
BLFS146

Bacillus sp.

3741
2581
84
globlastp


1919
BLFS146

Bacillus sp.

3742
2581
81.2
globlastp


1920
BLFS146

Bacillus sp.

3743
2581
78.2
globlastp


1921
BLFS146

Bacillus sp.

3744
2581
77.9
globlastp


1922
BLFS146

Bacillus sp.

3745
2581
76.4
globlastp


1923
BLFS146

Bacillus sp.

3746
2581
74
globlastp


1924
BLFS146

Bacillus sp.

3747
2581
73.1
globlastp


1925
BLFS146

Bacillus sp.

3748
2581
71.1
globlastp


1926
BLFS146

Bacillus sp.

3749
2581
70.8
globlastp


1749
BLFS133

Pseudomonas sp.

3573
2568
93.2
globlastp


1750
BLFS133

Pseudomonas sp.

3574
2568
91.8
globlastp


1751
BLFS133

Pseudomonas sp.

3575
2568
79.5
globlastp


1752
BLFS133

Pseudomonas sp.

3576
2568
78.1
globlastp


893
BLFS25

Bacillus sp.

2743
2468
99.2
globlastp


894
BLFS25

Bacillus sp.

2744
2468
98.8
globlastp


895
BLFS25

Bacillus sp.

2745
2468
98.4
globlastp


896
BLFS25

Bacillus sp.

2746
2468
98
globlastp


897
BLFS25

Bacillus sp.

2747
2468
70.8
globlastp


2070
ZLFS5

Burkholderia sp.

3884
2602
98.4
globlastp


2071
ZLFS5

Burkholderia sp.

3885
2602
97.7
globlastp


2072
ZLFS5

Burkholderia sp.

3886
2602
96.1
globlastp


2073
ZLFS5

Burkholderia sp.

3887
2602
88.3
globlastp


2074
ZLFS5

Burkholderia sp.

3888
2602
75
globlastp


1981
BLFS151

Stenotrophomonas sp.

3798
2586
78.1
globlastp


1982
BLFS151

Stenotrophomonas sp.

3799
2586
75.7
globlastp


1983
BLFS151

Stenotrophomonas sp.

3800
2586
73.5
globlastp


2000
BLFS159

Xanthomonas sp.

3814
2594
79.9
globlastp


2001
BLFS159

Xanthomonas sp.

3815
2594
76.8
globlastp


2089
ZLFS13

Collimonas sp.

3903
2610
96.7
globlastp


2090
ZLFS13

Collimonas sp.

3904
2610
80.3
globlastp


2032
ZLFS1

Actinomyces sp.

3846
2599
73.5
globlastp


1005
BLFS50

Collimonas sp.

2853
2491
85.1
globlastp


2091
ZLFS14

Collimonas sp.

3905
2611
75
globlastp


2083
ZLFS9

Collimonas sp.

3897
2606
98.6
globlastp





“Polyn.” = polynucleotide; “Polyp.” = polypeptide; “Algor.” = algorithm (used for sequence alignment and determination of percent homology); “Hom.”—homology; “iden.”—identity; “glob.”—global. Environmental sample = unidentified organisms obtained from the sample.







The output of the functional genomics approach described herein is a set of genes highly predicted to improve plant resistance to fungal infection.


Example 13: Identification of Domains Comprised within Polypeptides Encoded by the Identified Genes

Polypeptide domains were identified as described in Example 4 hereinabove.


The polypeptides of some embodiments of the invention, the expression of which in a may result in fungal control by invoking antifungal effects, can be characterized by specific amino acid domains. According to certain embodiments of the invention, particular domains are conserved within a family of polypeptides as described in Table 17 hereinbelow. Without wishing to be bound by specific theory or mechanism of action, the conserved domain may indicate common function of the polypeptides comprising same. The domains are presented by an arbitrary identifier (*ID). Table 18 provides the details of each domain according to the InterPro Entry.


Table 17 summarizes the domains in each of the “core” polypeptides (e.g., the polypeptides from Table 15) identified by the present inventors as being capable of conferring resistant to fungi and/or Oomycetes infection when its expression is modified in a plant (e.g. enhanced or down-regulated), wherein each of the listed domains is conserved in the representative homologous polypeptides identified by the present inventors exhibiting at least 70% global identity to the “core” polypeptides (as detailed in Table 16 in Example 12 above). As explained above, each domain received an arbitrary ID number wherein description of these arbitrary domain IDs according to the InterPro database is provided in Table 18 below. In addition, the start and end position of each of the domains with respect to the amino acid sequence of the “core” polypeptide was identified, as well as the E-values for each of the conserved domains as indicated by the domain tool used for analyzing these sequences, as part of interproscan programs, e.g., SMART, prosite scans patterns and profiles (not shown). For example, in the case of the Prosite search, the Prosite profiles report normalized scores instead of E-values, which are defined as the base 10 logarithm of the size (in residues) of the database in which one false positive match is expected to occur by chance. The normalized score is independent of the size of the databases searched. The so-called bit scores reported by other database-search programs have a distinct meaning but are also independent of the size of the database searched


For example, for SEQ ID NO: 9060, the domain ID “1” appears at amino acid positions 37 through 124. For some domains the e-value was not specified. In these cases, the presence of the domain was verified by ScanRegExp, which is able to verify PROSITE matches using corresponding statistically-significant CONFIRM patterns. The CONFIRM patterns were generated based on the true positive SWISS-PROT PROSITE matches using eMOTIF software with a stringency of 10e-9 P-value. Further details can be found in hypertext transfer protocol://computing.bio.cam.ac.uk/local/doc/iprscan.html.









TABLE 17







Core and homologous polypeptides comprising the same domains










Core





Polypeptide
Gene

Homologous Polypeptides


(SEQ ID
Name
Characteristic Domains
Comprising the Domains


NO)
(Core)
(Domain identifier)
(SEQ ID NOs)













2444
BLFS1
1; 2; 3 in core and homologs
2642


2445
BLFS2
4; 5; 6 in core and homologs
2643


2446
BLFS3
7; 8; 9; 10; 11; 12 in core



2447
BLFS4
6 in core



2448
BLFS5
13; 14 in core and homologs
2644; 2645; 2646; 2647; 2648; 2649;





2650; 2651


2449
BLFS6
2; 3 in core



2450
BLFS7
15; 16 in core



2451
BLFS8
1; 2; 3 in core and homologs
2652; 2653; 2654


2452
BLFS9
2; 1; 3 in core and homologs
2655; 2656; 2657; 2658; 2659; 2660


2453
BLFS10
1; 2; 3 in core and homologs
2452; 2655; 2656; 2661; 2662; 2663


2454
BLFS11
no domains in core



2455
BLFS12
no domains in core



2456
BLFS13
no domains in core



2457
BLFS14
17 in core and homologs
2675; 2676


2458
BLFS15
18; 19; 20 in core and homologs
2677; 2678; 2679; 2680; 2681; 2682;





2683; 2684; 2685


2459
BLFS16
21; 22; 23; 24 in core and homologs
2686; 2687; 2688; 2689; 2690


2460
BLFS17
25; 26 in core and homologs
2691; 2692; 2693; 2694; 2695; 2696


2461
BLFS18
27; 28; 29 in core and homologs
2697; 2698; 2699; 2700; 2701; 2702;





2703; 2704; 2705; 2706; 2707; 2708;





2709; 2710


2462
BLFS19
8; 30 in core and homologs
2711; 2712; 2713; 2714; 2715; 2716;





2717


2463
BLFS20
31; 32; 33 in core and homologs
2718; 2719; 2720; 2721


2464
BLFS21
34; 35; 36; 37; 38 in core and
2722; 2723; 2724; 2725; 2726; 2727




homologs



2465
BLFS22
no domains in core



2466
BLFS23
39; 31; 40; 41; 42; 32; 24 in core and
2734; 2735; 2736; 2737; 2738; 2739;




homologs
2740; 2741; 2742


2467
BLFS24
no domains in core



2468
BLFS25
43; 44 in core and homologs
2743; 2744; 2745; 2746; 2747


2469
BLFS26
31; 45; 32 in core and homologs
2748; 2749; 2750; 2751; 2752


2470
BLFS27
46; 47; 48 in core and homologs
2753; 2754; 2755; 2756; 2757


2471
BLFS29
8; 49; 30 in core and homologs
2758


2472
BLFS30
6 in core



2473
BLFS31
50 in core and homologs
2759; 2760; 2761; 2762; 2763; 2764;





2765; 2766; 2767; 2768; 2769


2474
BLFS32
52 in core and homologs
2770; 2771; 2772; 2773; 2774; 2775;





2776; 2777; 2778; 2779; 2780; 2781;





2782; 2783; 2784; 2785; 2786


2475
BLFS33
53; 35; 54 in core and homologs
2787; 2788; 2789; 2790; 2791; 2792;





2793; 2794; 2795; 2796; 2797; 2798


2476
BLFS34
31; 32; 33 in core and homologs
2799; 2800; 2801


2477
BLFS35
55 in core and homologs
2802; 2803; 2804; 2805; 2806; 2807;





2808; 2809; 2810


2478
BLFS36
no domains in core



2479
BLFS37
1; 56; 57 in core and homologs
2816; 2817


2480
BLFS38
58; 59 in core and homologs
2818


2481
BLFS39
6 in core and homologs
2819; 2820; 2821


2482
BLFS41
60; 61; 62; 63; 24; 64; 65; 66; 67 in core
2822; 2823; 2824; 2825; 2826; 2827;




and homologs
2828; 2829


2483
BLFS42
60; 68 in core and homologs
2830; 2831


2484
BLFS43
51 in core and homologs
2832; 2833; 2834; 2835


2485
BLFS44
69; 70; 71; 7; 10; 24 in core and
2836




homologs



2486
BLFS45
73 in core and homologs
2837; 2838; 2839; 2840; 2841; 2842;





2843; 2844


2487
BLFS46
31; 74; 32; 75 in core and homologs
2845


2488
BLFS47
1 in core and homologs
2846; 2847; 2848; 2849


2489
BLFS48
35; 52; 37 in core and homologs
2850; 2851; 2852


2490
BLFS49
7; 10 in core



2491
BLFS50
60; 68 in core and homologs
2853


2492
BLFS51
76; 24 in core and homologs
2854; 2855


2493
BLFS52
7; 10 in core



2494
BLFS53
no domains in core



2495
BLFS55
1; 57 in core and homologs
2857; 2858


2496
BLFS56
77; 17; 79 in core and homologs
2859; 2860; 2861; 2862; 2863; 2864


2497
BLFS57
80; 81; 7; 10; 82 in core and
2865; 2866; 2867




homologs



2498
BLFS58
1; 83; 57 in core and homologs
2868; 2869


2499
BLFS59
1; 57 in core



2500
BLFS60
1 in core and homologs
2870; 2871; 2872


2501
BLFS61
84; 85; 86; 87; 88 in core and
2873; 2874; 2875; 2876; 2877; 2878;




homologs
2879; 2880


2502
BLFS62
31; 74; 32; 75 in core and homologs
2881; 2882; 2883; 2884; 2885; 2886;





2887; 2888; 2889; 2890


2503
BLFS63
89; 90; 31; 32 in core and homologs
2891; 2892; 2893; 2894; 2895; 2896;





2897; 2898; 2899; 2900; 2901; 2902;





2903


2504
BLFS64
8; 49; 30 in core and homologs
2904


2505
BLFS65
91 in core and homologs
2905


2506
BLFS67
1; 57 in core and homologs
2906; 2907; 2908


2507
BLFS68
92 in core and homologs
2909; 2910; 2911; 2912


2508
BLFS69
93; 94; 95; 96; 97 in core



2509
BLFS70
98 in core



2510
BLFS72
99; 100; 101; 102 in core and
2913




homologs



2511
BLFS73
1 in core and homologs
2914; 2915; 2916; 2917; 2918; 2919;





2920; 2921; 2922; 2923; 2924; 2925;





2926; 2927; 2928; 2929


2512
BLFS74
34 in core and homologs
2930; 2931; 2932; 2933; 2934; 2935;





2936; 2937; 2938; 2939; 2940; 2941;





2942; 2943


2513
BLFS75
103; 104 in core and homologs
2944; 2945; 2946; 2947; 2948; 2949;





2950; 2951; 2952; 2953; 2954


2514
BLFS76
no domains in core



2515
BLFS77
17; 7; 78; 79; 10; 24; 76 in core and
2963; 2964; 2965




homologs



2516
BLFS79
43; 44; 105 in core and homologs
2966; 2967; 2968; 2969; 2970; 2971;





2972; 2973; 2974; 2975


2517
BLFS80
17; 78; 19 in core



2518
BLFS82
no domains in core



2519
BLFS83
106; 79; 107; 108; 17; 66; 78; 109 in
2989; 2990; 2991; 2992; 2993; 2994;




core and homologs
2995; 2996; 2997; 2998; 2999


2520
BLFS84
86; 110 in core and homologs
3000; 3001; 3002; 3003; 3004; 3005; 3006


2521
BLFS85
71; 24; 111 in core and homologs
3007; 3008; 3009; 3010; 3011; 3012;





3013; 3014; 3015; 3016; 3017


2522
BLFS86
no domains in core



2523
BLFS87
17; 60; 112; 76; 24; 113; 114 in core
3024; 3025; 3026; 3027; 3028; 3029;




and homologs
3030; 3031; 3032; 3033; 3034


2524
BLFS88
no domains in core



2525
BLFS89
24; 113; 76 in core and homologs
3044; 3045; 3046; 3047; 3048; 3049;





3050; 3051; 3052


2526
BLFS90
115; 116 in core and homologs
3053; 3054; 3055; 3056; 3057; 3058;





3059; 3060; 3061


2527
BLFS91
no domains in core



2528
BLFS92
34; 35; 36; 37; 38 in core and
3073; 3074; 3075; 3076; 3077; 3078;




homologs
3079; 3080; 3081


2529
BLFS93
117; 24 in core and homologs
3082; 3083; 3084; 3085; 3086; 3087;





3088; 3089; 3090; 3091; 3092


2530
BLFS94
17; 118 in core and homologs
3093; 3094; 3095; 3096; 3097; 3098;





3099; 3100; 3101


2531
BLFS95
no domains in core



2532
BLFS96
7; 10; 6 in core



2533
BLFS97
119 in core



2534
BLFS98
34 in core and homologs
3111; 3112; 3113; 3114; 3115; 3116;





3117; 3118; 3119; 3120; 3121; 3122;





3123; 3124; 3125; 3126


2535
BLFS99
120; 121; 122 in core and homologs
3127; 3128; 3129; 3130; 3131; 3132;





3133; 3134; 3135; 3136; 3137; 3138


2536
BLFS100
120; 121; 122 in core and homologs
3139; 3140; 3141; 3142; 3143; 3144;





3145; 3146; 3147; 3148; 3149; 3150;





3151; 3152; 3153; 3154


2537
BLFS101
52 in core and homologs
3155; 3156; 3157; 3158; 3159; 3160;





3161; 3162; 3163; 3164; 3165; 3166;





3167; 3168; 3169; 3170


2538
BLFS102
no domains in core



2539
BLFS103
no domains in core



2540
BLFS104
no domains in core



2541
BLFS105
123 in core and homologs
3194; 3195; 3196; 3197; 3198; 3199;





3200; 3201; 3202; 3203; 3204; 3205; 3206


2542
BLFS106
124 in core and homologs
3207; 3208; 3209; 3210; 3211; 3212;





3213; 3214; 3215; 3216; 3217; 3218;





3219; 3220; 3221; 3222; 3223; 3224;





3225; 3226; 3227


2543
BLFS107
125 in core and homologs
3228; 3229; 3230; 3231; 3232; 3233;





3234; 3235; 3236; 3237; 3238; 3239;





3240; 3241; 3242; 3243; 3244; 3245


2544
BLFS108
126 in core and homologs
3246; 3247; 3248; 3249; 3250; 3251;





3252; 3253; 3254


2545
BLFS109
127 in core and homologs
3255; 3256


2546
BLFS110
128 in core and homologs
3257


2547
BLFS111
129 in core and homologs
3258; 3259; 3260; 3261; 3262


2548
BLFS112
no domains in core



2549
BLFS113
130; 131 in core and homologs
3276; 3277; 3278; 3279; 3280; 3281;





3282; 3283; 3284; 3285; 3286; 3287;





3288; 3289; 3290; 3291; 3292; 3293;





3294; 3295; 3296; 3297; 3298; 3299; 3300


2550
BLFS114
132 in core and homologs
3301; 3302; 3303; 3304; 3305; 3306;





3307; 3308; 3309; 3310; 3311; 3312;





3313; 3314; 3315; 3316; 3317; 3318;





3319; 3320; 3321; 3322; 3323; 3324; 3325


2551
BLFS115
34; 133; 35; 37 in core and homologs
3326; 3327; 3328; 3329; 3330; 3331;





3332; 3333; 3334; 3335; 3336; 3337;





3338; 3339; 3340; 3341; 3342; 3343;





3344; 3345; 3346; 3347; 3348; 3349; 3350;





3351; 3352; 3353; 3354


2552
BLFS116
86; 110 in core and homologs
3355; 3356; 3357; 3358; 3359; 3360;





3361; 3362; 3363; 3364; 3365; 3366;





3367; 3368; 3369; 3370; 3371; 3372;





3373; 3374; 3375; 3376


2553
BLFS118
31; 32 in core and homologs
3377; 3378; 3379; 3380; 3381; 3382;





3383; 3384; 3385; 3386; 3387; 3388;





3389; 3390; 3391; 3392; 3393; 3394


2554
BLFS119
no domains in core



2555
BLFS120
134; 135 in core and homologs
3399; 3400; 3401


2556
BLFS121
136; 137; 138; 139; 140 in core and
3402; 3403; 3404; 3405; 3406; 3407;




homologs
3408; 3409; 3410; 3411; 3412; 3413;





3414; 3415; 3416; 3417; 3418; 3419;





3420; 3421; 3422; 3423


2557
BLFS122
no domains in core



2558
BLFS123
141; 8 in core and homologs
3431; 3432; 3433; 3434


2559
BLFS124
142 in core and homologs
3435; 3436; 3437; 3438; 3439; 3440;





3441; 3442; 3443; 3444; 3445; 3446;





3447; 3448; 3449; 3450; 3451


2560
BLFS125
72 in core and homologs
3452; 3453; 3454; 3455; 3456; 3457;





3458; 3459; 3460; 3461; 3462; 3463;





3464; 3465; 3466; 3467


2561
BLFS126
no domains in core



2562
BLFS127
24 in core and homologs
3476; 3477; 3478; 3479; 3480; 3481;





3482; 3483; 3484; 3485; 3486; 3487;





3488; 3489; 3490; 3491


2563
BLFS128
143; 144; 145 in core and homologs
3492; 3493; 3494; 3495; 3496; 3497;





3498; 3499; 3500; 3501; 3502; 3503;





3504; 3505; 3506; 3507; 3508


2564
BLFS129
146; 82 in core and homologs
3509; 3510; 3511; 3512; 3513; 3514;





3515; 3516; 3517; 3518; 3519


2565
BLFS130
147; 148; 115; 116 in core and
3520; 3521; 3522; 3523; 3524




homologs



2566
BLFS131
83 in core and homologs
3525; 3526; 3527; 3528; 3529; 3530;





3531; 3532; 3533; 3534; 3535; 3536;





3537; 3538; 3539; 3540; 3541; 3542;





3543; 3544; 3545; 3546; 3547; 3548


2567
BLFS132
no domains in core



2568
BLFS133
no domains in core



2569
BLFS134
149 in core and homologs
3577; 3578; 3579; 3580; 3581; 3582;





3583; 3584; 3585; 3586; 3587; 3588;





3589; 3590; 3591; 3592; 3593; 3594;





3595; 3596; 3597; 3598; 3599; 3600;





3601; 3602; 3603


2570
BLFS135
60; 68 in core and homologs
3604; 3605; 3606; 3607; 3608; 3609;





3610; 3611; 3612; 3613; 3614; 3615


2571
BLFS136
150; 151 in core and homologs
3616


2572
BLFS137
152; 153 in core and homologs
3617; 3618; 3619; 3620; 3621


2573
BLFS138
154; 155 in core and homologs
3622; 3623; 3624; 3625; 3626; 3627;





3628; 3629; 3630; 3631; 3632; 3633


2574
BLFS139
31; 32; 8; 30 in core and homologs
3634; 3635; 3636; 3637; 3638; 3639;





3640; 3641; 3642; 3643; 3644; 3645; 3646


2575
BLFS140
156; 157; 158; 159; 24; 160 in core
3647; 3648; 3649; 3650; 3651; 3652;




and homologs
3653; 3654; 3655; 3656; 3657; 3658;





3659; 3660


2576
BLFS141
161; 162; 163; 24 in core and
3661; 3662; 3663; 3664; 3665; 3666;




homologs
3667; 3668; 3669; 3670; 3671; 3672;





3673; 3674; 3675; 3676; 3677; 3678; 3679


2577
BLFS142
45 in core and homologs
3680; 3681; 3682; 3683; 3684; 3685;





3686; 3687; 3688; 3689; 3690


2578
BLFS143
164; 165; 166 in core and homologs
3691; 3692; 3693; 3694; 3695; 3696;





3697; 3698; 3699; 3700; 3701; 3702;





3703; 3704


2579
BLFS144
72; 167; 73; 168; 169; 170 in core and
3705; 3706; 3707; 3708; 3709; 3710;




homologs
3711; 3712; 3713; 3714; 3715


2580
BLFS145
103; 171; 172; 173 in core and
3716; 3717; 3718; 3719; 3720; 3721;




homologs
3722; 3723; 3724; 3725; 3726; 3727;





3728; 3729


2581
BLFS146
74; 75 in core and homologs
3730; 3731; 3732; 3733; 3734; 3735;





3736; 3737; 3738; 3739; 3740; 3741;





3742; 3743; 3744; 3745; 3746; 3747;





3748; 3749


2582
BLFS147
174; 143; 39 in core and homologs
3750; 3751; 3752; 3753; 3754; 3755;





3756; 3757; 3758; 3759


2583
BLFS148
52 in core and homologs
3760; 3761; 3762; 3763; 3764; 3765;





3766; 3767; 3768; 3769; 3770; 3771;





3772; 3773; 3774; 3775; 3776; 3777;





3778; 3779; 3780; 3781


2584
BLFS149
175; 60; 61; 62; 63;
3782; 3783; 3784; 3785; 3786; 3787;




24; 64; 17; 65; 66; 67
3788; 3789; 3790; 3791; 3792; 3793;




in core and homologs
3794; 3795; 3796; 3797


2585
BLFS150
no domains in core



2586
BLFS151
86; 110 in core and homologs
3798; 3799; 3800


2587
BLFS152
176; 177 in core and homologs
3801; 3802


2588
BLFS153
34; 178; 179; 180; 181 in core and
3803; 3804; 3805; 3806; 3807; 3808




homologs



2589
BLFS154
182 in core and homologs
3809; 3810


2590
BLFS155
4; 5 in core and homologs
3811; 3812


2591
BLFS156
1; 2; 3 in core



2592
BLFS157
99; 100; 183; 184; 6; 102 in core



2593
BLFS158
185; 186; 187 in core and homologs
3813


2594
BLFS159
no domains in core



2595
PUB6
188; 7; 10; 189 in core and homologs
3816; 3817; 3818; 3819; 3820; 3821;





3822; 3823; 3824; 3825


2596
PUB23
190; 191; 7; 10; 192; 193; 194; 19 in





core



2597
PUB31
195; 196; 7; 10; 19 in core



2598
PUB65
197; 15; 7; 198; 10; 82; 199; 200; 201;
3826; 3827; 3828; 3829; 3830; 3831;




202 in core and homologs
3832; 3833; 3834; 3835; 3836; 3837;





3838; 3839; 3840; 3841; 3842; 3843;





3844; 3845


2599
ZLFS1
203; 204; 205 in core and homologs
3846


2600
ZLFS2
206 in core and homologs
3847; 3848; 3849; 3850; 3851; 3852;





3853; 3854; 3855; 3856; 3857; 3858;





3859; 3860; 3861; 3862; 3863; 3864; 3865


2601
ZLFS3
31; 32; 8; 30 in core and homologs
3866; 3867; 3868; 3869; 3870; 3871;





3872; 3873; 3874; 3875; 3876; 3877;





3878; 3879; 3880; 3881; 3882; 3883


2602
ZLFS5
123 in core and homologs
3884; 3885; 3886; 3887; 3888


2603
ZLFS6
207; 208 in core and homologs
3889; 3890; 3891; 3892; 3893


2604
ZLFS7
209 in core and homologs
3894


2605
ZLFS8
210; 211 in core and homologs
3895; 3896


2606
ZLFS9
113; 24 in core and homologs
3897


2607
ZLFS10
no domains in core



2608
ZLFS11
212; 213; 214 in core and homologs
3898; 3899


2609
ZLFS12
215; 216; 217; 218 in core and
3900; 3901; 3902




homologs



2610
ZLFS13
no domains in core



2611
ZLFS14
126 in core and homologs
3905


2612
ZLFS15
219; 52 in core and homologs
3906; 3907; 3908


2613
ZLFS16
220; 221; 103; 171; 173; 172 in core
3909; 3910; 3911; 3912; 3913; 3914;




and homologs
3915


2614
ZLFS17
2; 1; 222 in core and homologs
3916


2615
ZLFS18
99; 100; 102 in core and homologs
3917


2616
ZLFS19
31; 74; 32; 75 in core and homologs
3918; 3919; 3920; 3921; 3922; 3923;





3924; 3925; 3926; 3927


2617
ZLFS21
223; 101; 224 in core and homologs
3928; 3929; 3930; 3931; 3932; 3933;





3934; 3935; 3936; 3937; 3938; 3939; 3940


2618
ZLFS22
225; 31; 32; 226 in core and
3941; 3942; 3943; 3944; 3945; 3946;




homologs
3947; 3948; 3949; 3950; 3951; 3952; 3953


2619
ZLFS23
82; 92; 227 in core and homologs
3954; 3955; 3956; 3957; 3958; 3959; 3960


2620
ZLFS24
176; 228; 229; 230; 231 in core and
3961; 3962; 3963; 3964; 3965; 3966;




homologs
3967; 3968; 3969; 3970; 3971; 3972;





3973; 3974; 3975; 3976


2621
ZLFS25
176; 232; 177 in core and homologs
3977; 3978; 3979; 3980; 3981; 3982; 3983


2622
ZLFS27
31; 233; 32; 101; 234 in core and
3984; 3985; 3986; 3987; 3988; 3989;




homologs
3990; 3991; 3992; 3993; 3994; 3995;





3996; 3997; 3998; 3999; 4000; 4001;





4002; 4003; 4004; 4005; 4006; 4007


2623
ZLFS28
86; 110 in core and homologs
4008; 4009; 4010; 4011; 4012; 4013;





4014; 4015; 4016; 4017; 4018; 4019;





4020; 4021; 4022


2624
ZLFS29
235; 219; 52 in core and homologs
4023; 4024; 4025; 4026; 4027; 4028;





4029; 4030; 4031; 4032; 4033; 4034;





4035; 4036; 4037; 4038; 4039; 4040;





4041; 4042; 4043; 4044; 4045; 4046; 4047; 4048


2625
ZLFS30
136; 140; 236 in core and homologs
4049; 4050; 4051; 4052; 4053; 4054;





4055; 4056; 4057; 4058; 4059; 4060;





4061; 4062; 4063; 4064; 4065; 4066;





4067; 4068


2626
ZLFS31
228; 230; 229; 231 in core and
4069; 4070; 4071; 4072; 4073; 4074;




homologs
4075; 4076; 4077; 4078; 4079; 4080; 4081


2627
ZLFS32
237; 238; 239 in core and homologs
4082; 4083; 4084; 4085; 4086; 4087;





4088; 4089; 4090; 4091; 4092; 4093;





4094; 4095; 4096; 4097


2628
ZLFS33
60; 68 in core and homologs
4098; 4099


2629
ZLFS34
240; 241; 242 in core and homologs
4100; 4101; 4102; 4103; 4104; 4105;





4106; 4107; 4108; 4109; 4110; 4111;





4112; 4113; 4114; 4115; 4116; 4117;





4118; 4119


2630
ZLFS35
no domains in core



2631
ZLFS36
no domains in core



2632
ZLFS37
243; 244 in core and homologs
4126; 4127; 4128; 4129; 4130; 4131;





4132; 4133; 4134; 4135; 4136; 4137;





4138; 4139; 4140; 4141; 4142; 4143;





4144; 4145; 4146; 4147; 4148; 4149; 4150;





4151; 4152; 4153


2633
ZLFS38
245; 246; 247; 248; 249 in core and
4154; 4155; 4156; 4157; 4158; 4159;




homologs
4160; 4161; 4162; 4163; 4164; 4165;





4166; 4167; 4168; 4169; 4170; 4171;





4172; 4173; 4174; 4175; 4176


2634
ZLFS39
250; 60; 251; 24; 71; 17; 252; 111 in
4177; 4178; 4179; 4180; 4181; 4182;




core and homologs
4183; 4184; 4185; 4186; 4187; 4188;





4189; 4190; 4191; 4192; 4193; 4194;





4195; 4196; 4197; 4198; 4199; 4200; 4201


2635
ZLFS42
1; 2; 3 in core and homologs
2637; 2660; 4202; 4203; 4204; 4205;





4206; 4207; 4208; 4209; 4210


2636
ZLFS44
8; 30 in core and homologs
4211; 4212; 4213; 4214; 4215; 4216;





4217; 4218; 4219; 4220; 4221; 4222;





4223; 4224; 4225; 4226; 4227; 4228;





4229; 4230; 4231


2637
ZLFS45
2; 1; 3 in core and homologs
4202; 4203; 4204; 4205; 4206; 4207;





4208; 4209; 4210; 4232


2638
ZLFS47
2; 1; 222; 92 in core



2639
ZLFS48
2; 1; 222 in core and homologs
3916


2640
ZLFS49
4; 74; 5 in core and homologs
4233; 4234


2641
ZLFS51
31; 40; 41; 42; 32; 24 in core and
4235; 4236; 4237; 4238; 4239; 4240;




homologs
4241; 4242; 4243; 4244; 4245; 4246;





4247; 4248; 4249; 4250; 4251; 4252
















TABLE 18







Details of identified domains presented in Table 17










Domain
InterPro




Identifier
No.
Accession No.
Description













1
IPR011024
SSF49695
Gamma-crystallin-like


2
IPR015791
G3DSA:2.60.20.30
Antimicrobial/protein inhibitor, gamma-





crystallin-like


3
IPR015161
PF09076
Beta/Gamma crystallin Streptomyces killer





toxin-like, beta/gamma crystallin


4
IPR001480
PS50927
Bulb-type lectin domain profile. Bulb-type lectin





domain


5
IPR036426
G3DSA:2.90.10.10
Bulb-type lectin domain superfamily


6
IPR004991
PF03318
Clostridium epsilon toxin ETX/Bacillus





mosquitocidal toxin MTX2 Aerolysin-like toxin


7
IPR035992
SSF50370
Ricin B-like lectins


8
IPR038765
SSF54001
Papain-like cysteine peptidase superfamily


9
IPR013688
PF08481
GBS Bsp-like repeat GBS Bsp-like


10
IPR000772
cd00161
RICIN Ricin B, lectin domain


11
IPR008999
SSF50405
Actin-crosslinking


12
IPR039564
PF13529
Peptidase C39 like family Peptidase C39-like


13
IPR001322
PF00932
Lamin Tail Domain Lamin tail domain


14
IPR036415
SSF74853
Lamin tail domain superfamily


15
IPR032812
PF13205
Bacterial Ig-like domain SbsA, Ig-like domain


16
IPR005077
PF03415
Clostripain family Peptidase C11, clostripain


17
IPR013783
G3DSA:2.60.40.10
Immunoglobulin-like fold


18
IPR003737
PF02585
GlcNAc-PI de-N-acetylase N-





acetylglucosaminyl phosphatidylinositol





deacetylase-related


19
IPR008979
G3DSA:2.60.120.260
Galactose-binding-like domain superfamily


20
IPR024078
SSF102588
Putative deacetylase LmbE-like domain





superfamily


21
IPR008270
PS00953
Glycosyl hydrolases family 25 active sites





signature. Glycoside hydrolase, family 25, active





site


22
IPR002053
PF01183
Glycosyl hydrolases family 25 Glycoside





hydrolase, family 25


23
IPR018077
SM00641
Glycoside hydrolase, family 25 subgroup


24
IPR017853
SSF51445
Glycoside hydrolase superfamily


25
IPR009045
G3DSA:3.30.1380.10
Hedgehog signalling/DD-peptidase zinc-





binding domain superfamily


26
IPR003709
PF02557
D-alanyl-D-alanine carboxypeptidase Peptidase





M15B


27
IPR012340
SSF50249
Nucleic acid-binding, OB-fold


28
IPR003029
PS50126
S1 domain profile. S1 domain


29
IPR022967
SM00316
RNA-binding domain, S1


30
IPR000064
PF00877
NlpC/P60 family Endopeptidase, NLPC/P60





domain


31
IPR018392
SM00257
LysM domain


32
IPR036779
G3DSA:3.10.350.10
LysM domain superfamily


33
IPR022887
MF_02014
Cell division suppressor protein YneA [yneA],





Cell division suppressor protein, YneA


34
IPR009003
SSF50494
Peptidase S1, PA clan


35
IPR036034
SSF50156
PDZ superfamily


36
IPR008763
PF05580
SpoIVB peptidase S55 Peptidase S55, SpoIVB


37
IPR001478
PS50106
PDZ domain profile. PDZ domain


38
IPR014219
TIGR02860
spore_IV_B: stage IV sporulation protein B





Peptidase S55, sporulation stage IV, protein B


39
IPR003646
PF08239
Bacterial SH3 domain SH3-like domain,





bacterial-type


40
IPR001223
PF00704
Glycosyl hydrolases family 18 Glycoside





hydrolase family 18, catalytic domain


41
IPR011583
SM00636
Chitinase II


42
IPR029070
G3DSA:3.10.50.10
Chitinase insertion domain superfamily


43
IPR011330
SSF88713
Glycoside hydrolase/deacetylase, beta/alpha-





barrel


44
IPR002509
PS51677
NodB homology domain profile. NodB





homology domain


45
IPR011105
PF07486
Cell Wall Hydrolase Cell wall hydrolase, SleB


46
IPR010310
PF06013
Proteins of 100 residues with WXG Type VII





secretion system ESAT-6-like


47
IPR037146
G3DSA:3.90.540.10
Colicin/pyocin, DNase domain superfamily


48
IPR036689
SSF140453
ESAT-6-like superfamily


49
IPR001119
PF00395
S-layer homology domain S-layer homology





domain


50
IPR019730
PF10794
Protein of unknown function (DUF2606) Protein





of unknown function DUF2606


51
IPR013784
SSF49452
Carbohydrate-binding-like fold


52
IPR001915
PF01435
Peptidase family M48 Peptidase M48


53
IPR008915
PF02163
Peptidase family M50 Peptidase M50


54
IPR004387
TIGR00054
TIGR00054: RIP metalloprotease RseP





Peptidase M50, putative membrane-associated





zinc metallopeptidase


55
IPR025493
PF14326
Domain of unknown function (DUF4384)





Domain of unknown function DUF4384


56
IPR015059
PF08964
Beta/Gamma crystallin Calcium-dependent cell





adhesion molecule, N-terminal


57
IPR001064
PS50915
Crystallins beta and gamma ‘Greek key’ motif





profile. Beta/gamma crystallin


58
IPR036404
G3DSA:2.100.10.30
Jacalin-like lectin domain superfamily


59
IPR001229
PS51752
Jacalin-type lectin domain profile. Jacalin-like





lectin domain


60
IPR014756
SSF81296
Immunoglobulin E-set


61
IPR015883
PF00728
Glycosyl hydrolase family 20, catalytic domain





Glycoside hydrolase family 20, catalytic domain


62
IPR015882
PF02838
Glycosyl hydrolase family 20, domain 2 Beta-





hexosaminidase, bacterial type, N-terminal


63
IPR012291
G3DSA:2.60.40.290
CBM2, carbohydrate-binding domain





superfamily


64
IPR029018
G3DSA:3.30.379.10
Beta-hexosaminidase-like, domain 2


65
IPR025705
PR00738
Glycosyl hydrolase family 20 signature Beta-





hexosaminidase


66
IPR008965
SSF49384
CBM2/CBM3, carbohydrate-binding domain





superfamily


67
IPR004866
PF03173
Putative carbohydrate binding domain





Chitobiase/beta-hexosaminidases, N-terminal





domain


68
IPR004302
PF03067
Lytic polysaccharide mono-oxygenase,





cellulose-degrading Cellulose/chitin-binding





protein, N-terminal


69
IPR013785
G3DSA:3.20.20.70
Aldolase-type TIM barrel


70
IPR002241
PF16499
Alpha galactosidase A Glycoside hydrolase,





family 27


71
IPR013780
G3DSA:2.60.40.1180
Glycosyl hydrolase, all-beta


72
IPR025711
PF13670
Peptidase propeptide and YPEB domain PepSY





domain


73
IPR011096
PF07504
Fungalysin/Thermolysin Propeptide Motif FTP





domain


74
IPR011055
G3DSA:2.70.70.10
Duplicated hybrid motif


75
IPR016047
PF01551
Peptidase family M23 Peptidase M23


76
IPR001547
PF00150
Cellulase (glycosyl hydrolase family 5)





Glycoside hydrolase, family 5


77
IPR007484
PF04389
Peptidase family M28 Peptidase M28


78
IPR036116
SSF49265
Fibronectin type III superfamily


79
IPR003961
cd00063
FN3 Fibronectin type III


80
IPR002594
PF01670
Glycosyl hydrolase family 12 Glycoside





hydrolase family 12


81
IPR013319
G3DSA:2.60.120.180
Glycoside hydrolase family 11/12


82
IPR013320
SSF49899
Concanavalin A-like lectin/glucanase domain





superfamily


83
IPR008816
PF05433
Glycine zipper 2TM domain Glycine zipper





2TM domain


84
IPR011049
SSF51120
Serralysin-like metalloprotease, C-terminal


85
IPR001343
PF00353
RTX calcium-binding nonapeptide repeat (4





copies) RTX calcium-binding nonapeptide





repeat


86
IPR029058
G3DSA:3.40.50.1820
Alpha/Beta hydrolase fold


87
IPR003995
PR01488
Gram-negative bacterial RTX toxin determinant





A family signature RTX toxin determinant A


88
IPR018511
PS00330
Hemolysin-type calcium-binding region





signature. Hemolysin-type calcium-binding





conserved site


89
IPR021731
PF11741
AMIN domain AMIN domain


90
IPR002508
cd02696
MurNAc-LAA N-acetylmuramoyl-L-alanine





amidase, catalytic domain


91
IPR014262
TIGR02913
HAF_rpt: probable extracellular repeat, HAF





family Extracellular HAF


92
IPR006311
PS51318
Twin arginine translocation (Tat) signal profile.





Twin-arginine translocation pathway, signal





sequence


93
IPR011050
SSF51126
Pectin lyase fold/virulence factor


94
IPR024973
PF13018
Extended Signal Peptide of Type V secretion





system ESPR domain


95
IPR008638
TIGR01901
adhes_NPXG: filamentous hemagglutinin





family N-terminal domain Filamentous





haemagglutinin, N-terminal


96
IPR011493
PF07581
The GLUG motif GLUG


97
IPR012334
G3DSA:2.160.20.10
Pectin lyase fold


98
IPR003540
PF03496
ADP-ribosyltransferase exoenzyme ADP





ribosyltransferase


99
IPR036365
SSF47090
PGBD-like superfamily


100
IPR002477
PF01471
Putative peptidoglycan binding domain





Peptidoglycan binding-like


101
IPR023346
SSF53955
Lysozyme-like domain superfamily


102
IPR036366
G3DSA:1.10.101.10
PGBD superfamily


103
IPR012338
SSF56601
Beta-lactamase/transpeptidase-like


104
IPR001466
PF00144
Beta-lactamase Beta-lactamase-related


105
IPR014235
cd10948
CE4_BsPdaA_like Peptidoglycan-N-





acetylmuramic acid deacetylase PdaA


106
IPR036434
SSF51989
1,4-beta cellobiohydrolase superfamily


107
IPR016288
PF01341
Glycosyl hydrolases family 6 1,4-beta





cellobiohydrolase


108
IPR036966
G3DSA:2.60.40.710
Carbohydrate-binding module 3 superfamily


109
IPR001956
PS51172
CBM3 (carbohydrate binding type-3) domain





profile. Carbohydrate-binding module 3


110
IPR000073
PF12697
Alpha/beta hydrolase family Alpha/beta





hydrolase fold-1


111
IPR006047
SM00642
Glycosyl hydrolase, family 13, catalytic domain


112
IPR016282
PIRSF001043
Glycoside hydrolase, family 5, endoglucanase B


113
IPR018087
PS00659
Glycosyl hydrolases family 5 signature.





Glycoside hydrolase, family 5, conserved site


114
IPR005102
PF03442
Carbohydrate binding domain X2 Carbohydrate





binding X2 domain


115
IPR001611
PS51450
Leucine-rich repeat profile. Leucine-rich repeat


116
IPR032675
G3DSA:3.80.10.10
Leucine-rich repeat domain superfamily


117
IPR011683
PF07745
Glycosyl hydrolase family 53 Glycosyl





hydrolase family 53


118
IPR034641
cd10318
RGL11 Polysaccharide lyase family 11


119
IPR020864
PF01823
MAC/Perforin domain Membrane attack





complex component/perforin (MACPF) domain


120
IPR010854
PF07338
Protein of unknown function (DUF1471)





Domain of unknown function DUF1471


121
IPR036275
SSF159871
YdgH-like superfamily


122
IPR025543
G3DSA:3.30.1660.10
Dodecin-like


123
IPR009739
PF07007
Lysozyme inhibitor LprI Lysozyme inhibitor





LprI, N-terminal


124
IPR010279
PF05957
Bacterial protein of unknown function





(DUF883) Inner membrane protein YqjD/ElaB


125
IPR007893
SM00972
Spore coat protein U


126
IPR009241
PF05973
Phage derived protein Gp49-like (DUF891)





Toxin HigB-like


127
IPR006914
PF04829
Pre-toxin domain with VENN motif VENN





motif-containing domain


128
IPR013728
PF08522
Domain of unknown function (DUF1735)





Domain of unknown function DUF1735


129
IPR003848
cd06259
YdcF-like Domain of unknown function





DUF218


130
IPR005490
PF03734
L,D-transpeptidase catalytic domain L,D-





transpeptidase catalytic domain


131
IPR038063
SSF141523
L,D-transpeptidase catalytic domain-like


132
IPR021109
SSF50630
Aspartic peptidase domain superfamily


133
IPR001940
PR00834
HtrA/DegQ protease family signature Peptidase





S1C


134
IPR025425
PF13665
Domain of unknown function (DUF4150)





Domain of unknown function DUF4150


135
IPR028917
PF15635
GHH signature containing HNH/Endo VII





superfamily nuclease toxin 2 Tox-GHH2





domain


136
IPR002933
PF01546
Peptidase family M20/M25/M40 Peptidase M20


137
IPR001261
PS00758
ArgE/dapE/ACY1/CPG2/yscS family





signature 1. ArgE/DapE/ACY1/CPG2/YscS.





conserved site


138
IPR017150
cd03885
M20_CPDG2 Peptidase M20, glutamate





carboxypeptidase


139
IPR011650
PF07687
Peptidase dimerisation domain Peptidase M20,





dimerisation domain


140
IPR036264
SSF55031
Bacterial exopeptidase dimerisation domain


141
IPR000668
PF00112
Papain family cysteine protease Peptidase C1A,





papain C-terminal


142
IPR012661
PF09498
Protein of unknown function (DUF2388)





Conserved hypothetical protein CHP02448


143
IPR002901
PF01832
Mannosyl-glycoprotein endo-beta-N-





acetylglucosaminidase Mannosyl-glycoprotein





endo-beta-N-acetylglucosamidase-like domain


144
IPR013377
TIGR02541
flagell_FlgJ: flagellar rod assembly





protein/muramidase FlgJ Peptidoglycan





hydrolase FlgJ


145
IPR019301
PF10135
Rod binding protein Flagellar protein FlgJ, N-





terminal


146
IPR014895
PF08787
Alginate lyase Alginate lyase 2


147
IPR029487
PF14496
C-terminal novel E3 ligase, LRR-interacting





Novel E3 ligase domain


148
IPR003591
SM00369
Leucine-rich repeat, typical subtype


149
IPR021357
PF11191
Protein of unknown function (DUF2782) Protein





of unknown function DUF2782


150
IPR007055
PS50914
BON domain profile. BON domain


151
IPR014004
SM00749
Transport-associated and nodulation domain,





bacteria


152
IPR015286
PF09203
MspA Porin family, mycobacterial-type


153
IPR036435
SSF56959
Leukocidin/porin MspA superfamily


154
IPR005320
cd03146
GAT1_Peptidase_E Peptidase S51


155
IPR029062
G3DSA:3.40.50.880
Class I glutamine amidotransferase-like


156
IPR001764
PF00933
Glycosyl hydrolase family 3 N terminal domain





Glycoside hydrolase, family 3, N-terminal


157
IPR002772
PF01915
Glycosyl hydrolase family 3 C-terminal domain





Glycoside hydrolase family 3 C-terminal





domain


158
IPR036962
G3DSA:3.20.20.300
Glycoside hydrolase, family 3, N-terminal





domain superfamily


159
IPR019800
PS00775
Glycosyl hydrolases family 3 active site.





Glycoside hydrolase, family 3, active site


160
IPR036881
SSF52279
Glycoside hydrolase family 3 C-terminal





domain superfamily


161
IPR016714
PIRSF018168
Mannan endo-1,4-beta-mannosidase


162
IPR000805
PR00739
Glycosyl hydrolase family 26 signature





Glycoside hydrolase family 26


163
IPR022790
PF02156
Glycosyl hydrolase family 26 Glycosyl





hydrolase family 26 domain


164
IPR029045
SSF52096
ClpP/crotonase-like domain superfamily


165
IPR002142
PF01343
Peptidase family S49 Peptidase S49


166
IPR004635
TIGR00706
SppA_dom: signal peptide peptidase SppA, 36K





type Peptidase S49, SppA


167
IPR027268
G3DSA:1.10.390.10
Peptidase M4/M1, CTD superfamily


168
IPR001570
PF02868
Thermolysin metallopeptidase, alpha-helical





domain Peptidase M4, C-terminal


169
IPR013856
PF01447
Thermolysin metallopeptidase, catalytic domain





Peptidase M4 domain


170
IPR023612
PR00730
Thermolysin metalloprotease (M4) family





signature Peptidase M4


171
IPR012907
PF07943
Penicillin-binding protein 5, C-terminal domain





Peptidase S11, D-Ala-D-Alacarboxypeptidase





A, C-terminal


172
IPR018044
PR00725
D-Ala-D-Alacarboxypeptidase 1 (S11) family





signature Peptidase S11, D-alanyl-D-alanine





carboxypeptidase A


173
IPR001967
PF00768
D-alanyl-D-alanine carboxypeptidase Peptidase





S11, D-alanyl-D-alanine carboxypeptidase A, N-





terminal


174
IPR007730
PS51724
SPOR domain profile. Sporulation-like domain


175
IPR004867
cd02847
E_set_Chitobiase_C Chitobiase C-terminal





domain


176
IPR036514
G3DSA:3.40.50.1110
SGNH hydrolase superfamily


177
IPR013830
PF13472
GDSL-like Lipase/Acylhydrolase family SGNH





hydrolase-type esterase domain


178
IPR001316
PR00861
Alpha-lytic endopeptidase serine protease (S2A)





signature Peptidase S1A, streptogrisin


179
IPR033116
PS00135
Serine proteases, trypsin family, serine active





site. Serine proteases, trypsin family, serine





active site


180
IPR004236
PF02983
Alpha-lytic protease prodomain Peptidase S1A,





alpha-lytic prodomain


181
IPR001254
PF00089
Trypsin Serine proteases, trypsin domain


182
IPR007921
PF05257
CHAP domain CHAP domain


183
IPR006616
SM00696
DM9 repeat


184
IPR024518
PF11901
Protein of unknown function (DUF3421)





Domain of unknown function DUF3421


185
IPR011051
SSF51182
RmlC-like cupin domain superfamily


186
IPR006045
PF00190
Cupin Cupin 1


187
IPR014710
G3DSA:2.60.120.10
RmlC-like jelly roll fold


188
IPR001434
PF01345
Domain of unknown function DUF11 Domain





of unknown function DUF11


189
IPR008966
SSF49401
Adhesion domain superfamily


190
IPR036399
G3DSA:2.100.10.10
Pesticidal crystal protein, central domain





superfamily


191
IPR005639
PF03945
delta endotoxin, N-terminal domain Pesticidal





crystal protein, N-terminal


192
IPR001178
PF00555
delta endotoxin Pesticidal crystal protein, central





domain


193
IPR005638
PF03944
delta endotoxin Pesticidal crystal protein, C-





terminal


194
IPR036716
SSF56849
Pesticidal crystal protein, N-terminal domain





superfamily


195
IPR000909
PF00388
Phosphatidylinositol-specific phospholipase C,





X domain Phosphatidylinositol-specific





phospholipase C, X domain


196
IPR017946
SSF51695
PLC-like phosphodiesterase, TIM beta/alpha-





barrel domain superfamily


197
IPR022409
SM00089
PKD/Chitinase domain


198
IPR000601
PF00801
PKD domain PKD domain


199
IPR036909
SSF46626
Cytochrome c-like domain superfamily


200
IPR015943
G3DSA:2.130.10.10
WD40/YVTN repeat-like-containing domain





superfamily


201
IPR009056
PS51007
Cytochrome c family profile. Cytochrome c-like





domain


202
IPR035986
SSF49299
PKD domain superfamily


203
IPR036574
G3DSA:3.30.30.10
Knottin, scorpion toxin-like superfamily


204
IPR003614
cd00107
Knot1 Knottin, scorpion toxin-like


205
IPR001542
PF01097
Arthropod defensin Defensin,





invertebrate/fungal


206
IPR000834
PS00132
Zinc carboxypeptidases, zinc-binding region 1





signature. Peptidase M14, carboxypeptidase A


207
IPR007048
PF04965
Gene 25-like lysozyme GpW/Gp25/IraD


208
IPR017737
TIGR03357
VI_zyme: type VI secretion system lysozyme-





like protein Type VI secretion system-related





protein


209
IPR021459
PF11308
Glycosyl hydrolases related to GH101 family,





GH129 Glycosyl hydrolases related to GH101





family, GHL1-GHL3


210
IPR003610
SM00495
Carbohydrate-binding module family 5/12


211
IPR036573
SSF51055
Carbohydrate-binding module superfamily 5/12


212
IPR000184
PF01103
Surface antigen Bacterial surface antigen (D15)


213
IPR016035
SSF52151
Acyl transferase/acyl





hydrolase/lysophospholipase


214
IPR002641
PS51635
Patatin-like phospholipase (PNPLA) domain





profile. Patatin-like phospholipase domain


215
IPR036852
SSF52743
Peptidase S8/S53 domain superfamily


216
IPR030400
PS51695
Sedolisin domain profile. Sedolisin domain


217
IPR023828
PS00138
Serine proteases, subtilase family, serine active





site. Peptidase S8, subtilisin, Ser-active site


218
IPR015366
PF09286
Pro-kumamolisin, activation domain Peptidase





S53, activation domain


219
IPR011990
SSF48452
Tetratricopeptide-like helical domain





superfamily


220
IPR037167
G3DSA:2.60.410.10
D-Ala-D-Ala carboxypeptidase, C-terminal





domain superfamily


221
IPR015956
SSF69189
Penicillin-binding protein, C-terminal domain





superfamily


222
IPR015201
PD068438
ANTIMICROBIAL PEPTIDE FUNGICIDE





3D-STRUCTURE DEFENSE SEQUENCING





DIRECT AMP1 PLANT PRECURSOR





Antimicrobial protein MiAMP1


223
IPR023099
G3DSA:3.30.386.10
Glycoside hydrolase, family 46, N-terminal


224
IPR000400
PS60000
Chitosanases families 46 and 80 active sites





signature. Glycoside hydrolase, family 46


225
IPR036908
SSF50685
RlpA-like domain superfamily


226
IPR010611
PF06725
3D domain 3D domain


227
IPR000757
PF00722
Glycosyl hydrolases family 16 Glycoside





hydrolase family 16


228
IPR005546
PS51208
Autotransporter beta-domain profile.





Autotransporter beta-domain


229
IPR017186
PIRSF037375
Lipase, autotransporter EstA


230
IPR036709
SSF103515
Autotransporter beta-domain superfamily


231
IPR001087
PF00657
GDSL-like Lipase/Acylhydrolase GDSL





lipase/esterase


232
IPR037460
cd01823
SEST like Streptomyces scabies esterase-like


233
IPR008258
PF01464
Transglycosylase SLT domain Transglycosylase





SLT domain 1


234
IPR000189
PS00922
Prokaryotic transglycosylases signature.





Prokaryotic transglycosylase, active site


235
IPR030873
MF_00997
Beta-barrel assembly-enhancing protease





[bepA]. Beta-barrel assembly-enhancing





protease


236
IPR010964
TIGR01887
dipeptidaselike: putative dipeptidase Peptidase





M20A, peptidase V-related


237
IPR012341
G3DSA:1.50.10.10
Six-hairpin glycosidase-like superfamily


238
IPR008928
SSF48208
Six-hairpin glycosidase superfamily


239
IPR002037
PR00735
Glycosyl hydrolase family 8 signature Glycoside





hydrolase, family 8


240
IPR024077
G3DSA:1.10.1370.10
Neurolysin/Thimet oligopeptidase, domain 2


241
IPR001567
PF01432
Peptidase family M3 Peptidase M3A/M3B





catalytic domain


242
IPR034005
cd06456
M3A_DCP Peptidyl-dipeptidase DCP


243
IPR036913
SSF160113
YegP-like superfamily


244
IPR010879
PF07411
Domain of unknown function (DUF1508)





Domain of unknown function DUF1508


245
IPR012347
G3DSA:1.20.1260.10
Ferritin-like


246
IPR002024
cd00907
Bacterioferritin Bacterioferritin


247
IPR009040
PS50905
Ferritin-like diiron domain profile. Ferritin-like





diiron domain


248
IPR009078
SSF47240
Ferritin-like superfamily


249
IPR008331
PF00210
Ferritin-like domain Ferritin/DPS protein





domain


250
IPR011837
TIGR02100
glgX_debranch: glycogen debranching enzyme





GlgX Glycogen debranching enzyme, GlgX type


251
IPR004193
PF02922
Carbohydrate-binding module 48 (Isoamylase





N-terminal domain) Glycoside hydrolase, family





13, N-terminal


252
IPR022844
MF_01248
Glycogen debranching enzyme [glgX].





Glycogen debranching enzyme, bacterial









Example 14: Cloning of Resistance-Conferring Bacterial Genes for Expression in E. coli

The fungicidal effect of isolated bacterial polypeptides was examined by expressing the polypeptides in E. coli and purifying the expressed proteins. For expression in E. coli, selected genes were synthesized by Genscript. The original bacterial sequences were modified such that the codons were optimized for protein expression in E. coli (further details are available at genscript.com/tools/codon-frequency-table), and a 6-Histidine sequence tag was inserted at either the 5′ or the 3′ end.


In cases where the original bacterial sequences included a nucleic acid sequence encoding a native signal peptide, it was removed up to the native cleavage site separating the encoded mature protein (i.e., the protein not including the signal peptide) from the native signal peptide. The polynucleotide encoding the mature protein was further modified by adding an artificial initiator Methionine codon immediately after the cleavage site.


All optimized genes were synthesized with 5′ NcoI and 3′ EcoRI restrictions sites, and in some of the genes as a result of the addition of the restriction site an additional glycine residue codon was added at the 2nd position (after the initiator Methionine codon) in order to maintain the reading frame of the coding sequence.


Genes lacking an original (native) signal peptide were cloned into pET22bd (a modified version of pET22B+ in which the periplasmic signal peptide PelB SEQ ID NO:5 was removed).


Genes having an original (native) signal peptide that was replaced with an artificial signal peptide were cloned into either the pET22bd and/or the pET22B+(purchased from Merck Millipore, merckmillipore.com/INTL/en/product/pET-22b %28%2B %29-DNA---Novagen, EMD_BIO-69744?ReferrerURL=%3A%2F%2Fwww.google.co(dot)il%2F&bd=1#anchor_Description) by digesting the gene and the vector with NcoI and EcoRI.


The sequence of each gene was verified by Sanger sequencing in each expression vector. All aforementioned modifications are summarized in Table 19 below.









TABLE 19







Sequences synthesized for cloning in E. coli













Native
Derived
Synthesized
Synthesized



Gene
Polyp.
Polyp.
Polyn.
Polyp.



Name
SEQ ID NO
SEQ ID NO
SEQ ID NO
SEQ ID NO
Modifications





BLFS1
2444
4776
4420
4617
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS2
2445

4442
4639
Gly & 3′ His-tag added


BLFS3
2446
4698
4316
4513
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS4
2447

4392
4589
3′ His-tag added


BLFS5
2448
4744
4373
4570
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS6
2449
4682
4298
4495
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS7
2450
4753
4386
4583
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS8
2451
4771
4414
4611
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS9
2452
4757
4394
4591
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS10
2453
4690
4306
4503
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS11
2454
4669
4284
4481
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS12
2455
4788
4435
4632
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS13
2456
4673
4288
4485
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS14
2457
4722
4344
4541
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS15
2458
4678
4294
4491
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS16
2459
4681
4297
4494
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS17
2460
4671
4286
4483
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS18
2461

4265
4462
Gly & 3′ His-tag added


BLFS19
2462
4712
4334
4531
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS20
2463
4774
4417
4614
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS21
2464
4751
4382
4579
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS22
2465
4787
4434
4631
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS23
2466
4653
4259
4456
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS24
2467

4395
4592
3′ His-tag added


BLFS25
2468
4799
4449
4646
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS26
2469
4784
4430
4627
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS29
2471
4657
4263
4460
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS30
2472

4408
4605
3′ His-tag added


BLFS31
2473
4726
4348
4545
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS32
2474
4752
4384
4581
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS33
2475
4701
4319
4516
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS34
2476
4650
4256
4453
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS35
2477
4778
4423
4620
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS36
2478
4772
4415
4612
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS37
2479

4368
4565
Gly & 3′ His-tag added


BLFS38
2480

4281
4478
Gly & 3′ His-tag added


BLFS39
2481
4732
4356
4553
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS41
2482
4781
4427
4624
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS42
2483
4763
4404
4601
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS43
2484
4683
4299
4496
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS44
2485
4713
4335
4532
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS45
2486
4745
4374
4571
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS46
2487
4694
4311
4508
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS47
2488
4686
4302
4499
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS48
2489
4699
4317
4514
Native lipid anchor removed;







added MetGly; 3′ His-tag


BLFS49
2490
4790
4438
4635
Native lipid anchor removed;







added MetGly; 3′ His-tag


BLFS50
2491
4754
4388
4585
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS51
2492
4692
4309
4506
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS52
2493
4737
4364
4561
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS53
2494

4433
4630
Gly & 3′ His-tag added


BLFS55
2495
4748
4378
4575
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS56
2496
4651
4257
4454
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS57
2497
4738
4365
4562
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS58
2498
4743
4371
4568
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS59
2499
4674
4290
4487
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS60
2500
4730
4353
4550
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS61
2501

4325
4522
3′ His-tag added


BLFS62
2502
4691
4308
4505
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS63
2503
4697
4315
4512
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS64
2504
4654
4260
4457
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS65
2505
4719
4341
4538
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS67
2506
4782
4428
4625
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS68
2507
4696
4314
4511
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS69
2508
4655
4261
4458
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS70
2509

4269
4466
3′ His-tag added


BLFS72
2510

4278
4475
Gly & 3′ His-tag added


BLFS73
2511

4312
4509
3′ His-tag added


BLFS74
2512
4758
4396
4593
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS75
2513
4688
4304
4501
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS76
2514
4710
4329
4526
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS77
2515
4668
4279
4476
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS79
2516
4665
4275
4472
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS80
2517
4709
4328
4525
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS82
2518

4282
4479
Gly & 3′ His-tag added


BLFS83
2519
4750
4380
4577
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS84
2520
4663
4273
4470
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS85
2521
4765
4406
4603
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS86
2522
4724
4346
4543
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS87
2523
4648
4254
4451
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS88
2524
4786
4432
4629
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS89
2525
4652
4258
4455
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS90
2526

4376
4573
3′ His-tag added


BLFS91
2527
4703
4321
4518
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS92
2528
4769
4412
4609
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS93
2529
4756
4391
4588
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS94
2530
4687
4303
4500
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS95
2531
4740
4367
4564
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS96
2532

4419
4616
3′ His-tag added


BLFS97
2533

4443
4640
Gly & 3′ His-tag added


BLFS98
2534
4783
4429
4626
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS99
2535
4729
4352
4549
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS100
2536
4746
4375
4572
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS101
2537
4780
4425
4622
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS102
2538
4725
4347
4544
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS103
2539

4331
4528
Gly & 3′ His-tag added


BLFS104
2540
4768
4410
4607
Native lipid anchor removed;







added MetGly; 3′ His-tag


BLFS105
2541
4739
4366
4563
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS106
2542

4411
4608
Gly & 3′ His-tag added


BLFS107
2543
4773
4416
4613
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS108
2544

4399
4596
Gly & 3′ His-tag added


BLFS109
2545
4779
4424
4621
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS110
2546
4702
4320
4517
Native lipid anchor removed;







added MetGly; 3′ His-tag


BLFS111
2547
4793
4441
4638
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS112
2548

4349
4546
Gly & 3′ His-tag added


BLFS113
2549
4672
4287
4484
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS114
2550
4705
4323
4520
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS115
2551
4716
4338
4535
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS116
2552

4330
4527
3′ His-tag added


BLFS118
2553
4767
4409
4606
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS119
2554
4741
4369
4566
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS120
2555

4361
4558
3′ His-tag added


BLFS121
2556
4707
4326
4523
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS122
2557

4280
4477
Gly & 3′ His-tag added


BLFS123
2558

4426
4623
3′ His-tag added


BLFS124
2559
4675
4291
4488
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS125
2560
4723
4345
4542
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS126
2561
4795
4445
4642
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS127
2562
4695
4313
4510
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS128
2563

4332
4529
3′ His-tag added


BLFS129
2564

4383
4580
Gly & 3′ His-tag added


BLFS130
2565

4268
4465
Gly & 3′ His-tag added


BLFS131
2566
4693
4310
4507
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS132
2567
4689
4305
4502
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS133
2568
4798
4448
4645
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS134
2569
4679
4295
4492
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS135
2570
4717
4339
4536
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS136
2571
4662
4272
4469
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS137
2572
4728
4351
4548
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS138
2573

4385
4582
Gly & 3′ His-tag added


BLFS139
2574
4721
4343
4540
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS140
2575
4761
4402
4599
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS141
2576
4785
4431
4628
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS142
2577
4755
4390
4587
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS143
2578
4770
4413
4610
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS144
2579
4718
4340
4537
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS145
2580
4715
4337
4534
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS146
2581
4797
4447
4644
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS147
2582
4764
4405
4602
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS148
2583
4711
4333
4530
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS149
2584
4789
4437
4634
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS150
2585

4421
4618
Gly & 3′ His-tag added


BLFS151
2586
4664
4274
4471
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS152
2587
4791
4439
4636
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS153
2588
4734
4359
4556
Native signal peptide







removed; MetGly & 3′ His-







tag added


BLFS154
2589
4792
4440
4637
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS155
2590
4659
4266
4463
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS156
2591
4660
4267
4464
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS157
2592
4747
4377
4574
Native signal peptide







removed; Met & 3′ His-tag







added


BLFS158
2593

4389
4586
Gly & 3′ His-tag added


BLFS159
2594

4289
4486
Gly & 3′ His-tag added


PUB6
2595

4372
4569
3′ His-tag added


PUB23
2596

4355
4552
Gly & 3′ His-tag added


PUB31
2597

4398
4595
3′ His-tag added


PUB65
2598
4775
4418
4615
Native signal peptide







removed; Met & 3′ His-tag







added


ZLFS1
2599

4381
4578
3′ His-tag added


ZLFS2
2600
4667
4277
4474
Native signal peptide







removed; Met & 3′ His-tag







added


ZLFS3
2601
4649
4255
4452
Native signal peptide







removed; Met & 3′ His-tag







added


ZLFS5
2602
4647
4253
4450
Native signal peptide







removed; Met & 3′ His-tag







added


ZLFS6
2603

4283
4480
Gly & 3′ His-tag added


ZLFS7
2604
4700
4318
4515
Native signal peptide







removed; Met & 3′ His-tag







added


ZLFS8
2605
4777
4422
4619
Native lipid anchor removed;







added MetGly; 3′ His-tag


ZLFS9
2606
4794
4444
4641
Native signal peptide







removed; MetGly & 3′ His-







tag added


ZLFS10
2607
4766
4407
4604
Native signal peptide







removed; Met & 3′ His-tag







added


ZLFS11
2608
4677
4293
4490
Native signal peptide







removed; Met & 3′ His-tag







added


ZLFS12
2609

4360
4557
Gly & 3′ His-tag added


ZLFS13
2610
4706
4324
4521
Native signal peptide







removed; Met & 3′ His-tag







added


ZLFS14
2611

4436
4633
Gly & 3′ His-tag added


ZLFS15
2612
4708
4327
4524
Native signal peptide







removed; Met & 3′ His-tag







added


ZLFS16
2613
4714
4336
4533
Native signal peptide







removed; MetGly & 3′ His-







tag added


ZLFS17
2614
4731
4354
4551
Native signal peptide







removed; MetGly & 3′ His-







tag added


ZLFS18
2615

4387
4584
3′ His-tag added


ZLFS19
2616
4658
4264
4461
Native lipid anchor removed;







added MetGly; 3′ His-tag


ZLFS21
2617
4670
4285
4482
Native signal peptide







removed; Met & 3′ His-tag







added


ZLFS22
2618
4685
4301
4498
Native signal peptide







removed; Met & 3′ His-tag







added


ZLFS23
2619
4759
4397
4594
Native signal peptide







removed; Met & 3′ His-tag







added


ZLFS24
2620
4676
4292
4489
Native signal peptide







removed; MetGly & 5' His-







tag added


ZLFS25
2621
4680
4296
4493
Native signal peptide







removed; Met & 3′ His-tag







added


ZLFS27
2622
4760
4401
4598
Native lipid anchor removed;







added MetGly; 3′ His-tag


ZLFS28
2623
4656
4262
4459
Native signal peptide







removed; Met & 3′ His-tag







added


ZLFS29
2624
4733
4358
4555
Native signal peptide







removed; Met & 3′ His-tag







added


ZLFS30
2625
4666
4276
4473
Native signal peptide







removed; MetGly & 3′ His-







tag added


ZLFS31
2626
4704
4322
4519
Native signal peptide







removed; MetGly & 5' His-







tag added


ZLFS32
2627
4727
4350
4547
Native lipid anchor removed;







added Met; 3′ His-tag


ZLFS33
2628
4742
4370
4567
Native signal peptide







removed; Met & 3′ His-tag







added


ZLFS34
2629
4796
4446
4643
Native signal peptide







removed; Met & 3′ His-tag







added


ZLFS35
2630
4749
4379
4576
Native signal peptide







removed; Met & 3′ His-tag







added


ZLFS36
2631

4400
4597
Gly & 3′ His-tag added


ZLFS37
2632

4307
4504
3′ His-tag added


ZLFS38
2633

4393
4590
Gly & 3′ His-tag added


ZLFS39
2634

4357
4554
Gly & 3′ His-tag added


ZLFS42
2635
4661
4271
4468
Native signal peptide







removed; MetGly & 3′ His-







tag added


ZLFS44
2636
4736
4363
4560
Native signal peptide







removed; Met & 3′ His-tag







added


ZLFS45
2637
4735
4362
4559
Native signal peptide







removed; MetGly & 3′ His-







tag added


ZLFS47
2638
4684
4300
4497
Native signal peptide







removed; MetGly & 3′ His-







tag added


ZLFS48
2639
4762
4403
4600
Native signal peptide







removed; MetGly & 3′ His-







tag added


ZLFS49
2640
4720
4342
4539
Native signal peptide







removed; Met & 3′ His-tag







added


ZLFS51
2641

4270
4467
3′ His-tag added





“Polyp.”—Polypeptide. “Polyn.”—Polynucleotide






In most cases, following the optimization, the synthesized sequences exhibit at least 70% global identity to the curated sequences from which they were obtained. In several cases replacing the native signal peptide resulted in a polynucleotide and/or polypeptide having less than 7000 global identity to their parent sequence. For the genes presented in Table 20, the optimized synthesized polypeptide sequences exhibit less than 70% global identity to the curated sequences from which they were obtained, due to a replacement of relatively long signal sequence. For example, the polypeptide of SEQ ID NO:4502 exhibits only 48.65% global identity to SEQ ID NO:2567 which is the original curated sequence of BLFS132. These polypeptides, however, comprise the core amino acid sequence of the parent polypeptides.









TABLE 20







Optimized polypeptides having less that 70% global


identity to the parent not optimized polypeptide












Native Polypeptide
Synthesized Polypeptide



Gene Name
SEQ ID NO.
SEQ ID NO.







ZLFS42
2635
4468



ZLFS47
2638
4497



BLFS132
2567
4502



BLFS10
2453
4503



BLFS86
2522
4543



BLFS102
2538
4544



BLFS52
2493
4561



BLFS119
2554
4566



BLFS9
2452
4591



BLFS36
2478
4612



BLFS20
2463
4614



BLFS133
2568
4645










Example 15: Purification of Proteins Expressed in Bacterial Cells

Transformation of Bacterial Cells with the Polynucleotides Encoding the Polypeptides Having the Ability to Kill or Inhibit the Development of Fusarium


Genes encoding unknown toxin candidate proteins of the present invention were cloned in pET22/T7-lac promoter-based vector and coding DNA sequence was confirmed by sequencing. pET-based expression vectors were transformed into BL21(DE3) E. coli host using heat shock method. After overnight growth in Terrific Broth (TB) medium at 37° C. in the presence of Ampicillin (100 μg/mL), 5 mL starter cultures were used to inoculate 100 mL TB culture at OD600 0.03 in 1 L flat bottom flask. The cultures were allowed to grow until OD600-0.5 (2-3 hours at 37° C. with 250 rpm). The incubator shaker temperature was reduced to 22° C. and Isopropyl R-D-1-thiogalactopyranoside (IPTG) was added at a final concentration of 1 mM. The cultures were incubated further for 15 to 18 hours for target protein expression and cells were harvested by centrifuging at 4,000 rpm/4° C./10 minutes. The cell pellet was washed with cold water and stored at −80° C. until used for protein purification.


Bacterial cell pellet was lysed using B-PER™ Bacterial Protein Extraction Reagent (Thermo Fisher Scientific, catalog number: 78248) at room temperature for 1 hour. The supernatant fraction (containing soluble protein) was clarified by centrifugation at 16000 rcf/4° C./20 minutes.


Purification of Expressed Recombinant Polypeptides:

Soluble fractions—The supernatant fraction containing soluble protein was incubated with Ni-NTA beads (washed with binding buffer prior to addition of supernatant fraction: 50 mM potassium phosphate pH 8.0, 600 mM NaCl and 20 mM imidazole) for 1 hour at 4° C. on a rotatory shaker with gentle shaker speed. The Ni-NTA-protein bound beads were collected by centrifugation at 800 rcf/4° C./3 minutes. The Ni-NTA-protein bound beads were washed with washing buffer (50 mM potassium phosphate pH 8.0, 600 mM NaCl, 0.5% Triton and 20 mM imidazole) for 3 times. The bound proteins were eluted with elution buffer (50 mM potassium phosphate pH 8.0, 600 mM NaCl and 250 mM imidazole). The salts in the eluted proteins were removed by dialysis. Samples were loaded on a Pur-A+Lyzer tubes (catalog number: PurR35100) and placed in a beaker containing 0.5 liter of sterile dialysis buffer (16.5 mM Potassium phosphate buffer pH 5.0) for 1 night/weekend in the refrigerator at 8° C. SDS-PAGE analysis was used to quantify protein using known concentrations of bovine serum albumin (BSA) as standard. The known concentrations of toxin candidates were used for bioassay.


Example 16: Effect of Isolated Proteins on Fungal Growth

The spore assay is an in vitro bioassay in a 96 wells plate format, implementing spores from a Fusarium verticillioides (Fv) GFP-labeled strain and the candidate protein purified from bacterial cells as described hereinabove. Mycelia growth of fungi treated with the protein was compared to the growth of untreated fungi and growth inhibition was quantified by detection of GFP fluorescence (475 nm excitation, 509 nm emission). Optical Density (OD) of 620 nm is also measured for QA purposes.


Fresh spores were prepared, quantified and dispensed in each well (500 spores) in 50 μl 2×PDB and incubated for 2-3 hours at 25° C. before protein addition.


Purified clean proteins prepared as described hereinabove were quantified and diluted with buffer phosphate pH 5.0 at the desired concentration and added to each assay well (up to 40 μM) for a final reaction volume of 100 μl. The plates are incubated for 72 hr at 25° C. and responses were measured every 24 hr.


Each protein treatment in each plate was compared to the corresponding spore growth in untreated control by t-test α=0.05 one tailed


Table 21 summarizes the results of two independent assays using purified proteins from bacterial cells over-expressing the polypeptides of some embodiments of the invention.









TABLE 21







Inhibition of Fusarium mycelia growth by proteins


of the invention isolated from bacteria












First assay
Second assay


Gene Name
General annotation
% Inhibition **
% Inhibition **





ZLFS42
Killer toxin like
90% (72 hr)
86% (72 hr)




20 μM
25 μM


ZLFS48
Antimicrobial protein
56% (48 hr)
30% (72 hr)



MiAMP1
25 μM
20 μM


BLFS129
Alginate lyase
85% (48 hr)
69% (72 hr)




6.4 μM 
 4 μM


BLFS139
Peptidoglycan endopeptidase
72% (48 hr)
64% (48 hr)




14 μM
 6 μM


BLFS128
Peptidoglycan hydrolase flgj
37% (48 hr)
52% (48 hr)




14 μM
 3 μM


BLFS70
Hypothetical protein
36% (48 hr)
52% (48 hr)




 2 μM
 3 μM


BLFS111
Hypothetical protein)
20% (72 hr)
63% (48 hr)




4.5 μM 
10 μM


BLFS157
Hypothetical protein
37% (48 hr)
41% (48 hr)




12 μM
10 μM


BLFS101
Metalloprotease loiP
47% (24 hr)
70% (48 hr)




25 μM
25 μM


BLFS63
n-acetylmuramoyl-1-alanine
44% (48 hr)
55% (48 hr)



amidase amib
39 μM
 2 μM


ZLFS1
Arthropod defensin
29% (48 hr)
32% (48 hr)




13 μM
27 μM


BLFS155
Mannose-binding protein
45% (48 hr)
70% (48 hr)




25 μM
44 μM


BLFS7
Peptidase C11 clostripain
65% (48 hr)
36% (48 hr)




<1 μM
 5 μM


BLFS140
Uncharacterized lipoprotein
75% (48 hr)
63% (48 hr)



ybbd
 9 μM
40 μM


BLFS103
Toxin tccc3
67% (48 hr)
56% (48 hr)




3 μM
3.75 μM  


BLFS46
Lysm domain protein
46% (48 hr)
32% (48 hr)




18 μM
40 μM


BLFS115
Probable periplasmic serine
39% (48 hr)
39% (48 hr)



endoprotease DegP-like
23 μM
40 μM


BLFS47
Hypothetical protein
11% (72 hr)
25% (48 hr)




40 μM
0.5 μM


BLFS87
Endoglucanase b
25% (48 hr)
27% (48 hr)




40 μM
40 μM


BLFS102
Hypothetical protein
30% (48 hr)
44% (48 hr)




40 μM
40 μM


BLFS42
Chitin-binding protein
21% (72 hr)
60% (48 hr)




25 μM
40 μM


ZLFS29
Beta-barrel assembly-
36.5% (48 hr)  
44% (48 hr)



enhancing protease [bepA].
40 μM
40 μM


BLFS154
Peptidase s1
52% (48 hr)
33% (48 hr)




40 μM
40 μM


BLFS62
Murein hydrolase activator
53% (24 hr)
27% (48 hr)



nlpd
40 μM
40 μM


BLFS19
Peptidase p60
31% (48 hr)
29% (48 hr)




40 μM
40 μM


ZLFS16
d-alanyl-d-alanine
11% (24 hr)
36% (24 hr)



carboxypeptidase dacd
41 μM
38 μM


ZLFS19
Lipoprotein nlpd/
21% (48 hr)
33% (24 hr)



lppb homolog
38 μM
37 μM


BLFS1
Antifungal protein
57% (48 hr)
31% (48 hr)




40 μM
40 μM


BLFS9
Oxidoreductase
21% (48 hr)
15% (48 hr)




25 μM
18 μM


BLFS80
Hypothetical protein
33% (48 hr)
43% (48 hr)




40 μM
40 μM


BLFS123
Peptidase c1
17% (72 hr)
 1% (48 hr)




20 μM
20 μM


BLFS113
Transpeptidase
11% (72 hr)
19% (48 hr)




40 μM
25 μM


BLFS121
Glutamate carboxypeptidase
26% (48 hr)
14% (24 hr)




40 μM
25 μM


BLFS142
Uncharacterized protein ykvt
34% (48 hr)
31% (48 hr)




21 μM
42 μM


ZLFS5
Hypothetical protein
29% (48 hr)
15% (48 hr)




20 μM
40 μM


ZLFS32
Endoglucanase
29% (48 hr)
16% (48 hr)




20 μM
40 μM


ZLFS22
Hypothetical protein
95% (72 hr)
11% (24 hr)




40 μM
40 μM


ZLFS51
Glycosyl hydrolase
36% (24 hr)
 3% (24 hr)




40 μM
40 μM


ZLFS27
Lytic transglycosylase
28% (48 hr)
87% (48 hr)




40 μM
20 μM


ZLFS47
Antimicrobial protein
 5% (48 hr)
56% (48 hr)



MiAMP1
20 μM
25 μM


ZLFS45
Oxidoreductase
63% (24 hr)
81% (72 hr)




20 μM
40 μM





** = Best inhibition value scored at defined time and concentration; α = 0.05 and % inhibition > 10%






The foregoing description of the specific embodiments will so fully reveal the general nature of the invention that others can, by applying current knowledge, readily modify and/or adapt for various applications such specific embodiments without undue experimentation and without departing from the generic concept, and, therefore, such adaptations and modifications should and are intended to be comprehended within the meaning and range of equivalents of the disclosed embodiments. It is to be understood that the phraseology or terminology employed herein is for the purpose of description and not of limitation. The means, materials, and steps for carrying out various disclosed functions may take a variety of alternative forms without departing from the invention.

Claims
  • 1. A method for enhancing the resistance of a plant or a part thereof to at least one pathogenic fungus and/or oomycete, comprising introducing into at least one cell of the plant or part thereof at least one exogenous polynucleotide encoding at least one polypeptide at least 70% identical to a polypeptide having the amino acid sequence set forth in SEQ ID NO:2618, a fragment or a variant thereof, thereby enhancing the resistance of said plant or part thereof to the at least one pathogenic fungus and/or oomycete compared to the resistance of a control plant.
  • 2. The method of claim 1, wherein the at least one polypeptide at least 70% identical to the polypeptide having the amino acid sequence set forth in SEQ ID NO:2618 comprises an amino acid sequence selected from the group consisting of SEQ ID NOs:3941-3953, a fragment of said polypeptide comprises the amino acid sequence set forth in SEQ ID NO:4685, and a variant of said polypeptide comprises the amino acid sequence set forth in SEQ ID NO:4498.
  • 3. The method of claim 1, wherein the at least one polynucleotide comprises a nucleic acid sequence at least 70% identical to the nucleic acid sequence set forth in SEQ ID NO:766, a fragment or a variant thereof.
  • 4. The method of claim 3, wherein the at least one polynucleotide at least 70% identical to the polynucleotide having the nucleic acid sequence set forth in SEQ ID NO:766 comprises a nucleic acid sequence selected from the group consisting of SEQ ID NOs:2127-2139, and a variant of said polypeptide comprises the amino acid sequence set forth in SEQ ID NO:4301.
  • 5. The method of claim 1, wherein introducing the exogenous polynucleotide into the at least one cell of the plant or part thereof comprises transforming said polynucleotide or a construct comprising same into said at least one cell.
  • 6. The method of claim 1, wherein introducing the exogenous polynucleotide into the at least one cell of the plant or part thereof comprises subjecting the at least one cell to genome editing using artificially engineered nucleases.
  • 7. A method for producing a population of plants each having an enhanced resistance to at least one pathogenic fungus and/or oomycete, comprising the steps of: a) introducing into at least one cell of each plant of the plant population at least one polynucleotide encoding at least one polypeptide at least 70% identical to a polypeptide having the amino acid sequence set forth in SEQ ID NO:2618, a fragment or a variant thereof as to produce a genetically engineered plant population;b) inoculating each plant of the genetically engineered plant population with the at least one pathogenic fungus or oomycete; andc) selecting plants showing an enhanced resistance to said at least one pathogenic fungus or oomycete compared to a control plant or to a pre-determined resistance score value, thereby producing a population of genetically engineered plants having enhanced resistance to said at least one pathogenic fungus and/or oomycete.
  • 8. The method of any one of claim 7, wherein the at least one polypeptide at least 70% identical to the polypeptide having the amino acid sequence set forth in SEQ ID NO:2618 comprises an amino acid sequence selected from the group consisting of SEQ ID NOs:3941-3953, a fragment of said polypeptide comprises the amino acid sequence set fort SEQ ID NO:4685, and a variant of said polypeptide comprises the amino acid sequence set forth in SEQ ID NO:4498.
  • 9. A method for selecting a plant having an enhanced resistance to at least one pathogenic fungus and/or oomycete, comprising the steps of: a) providing a plurality of plants each comprising at least one cell with enhanced expression and/or activity of a polypeptide at least 70% identical to a polypeptide having the amino acid sequence set forth in SEQ ID NO:2618, a fragment or a variant thereof;b) inoculating the plurality of plants with the at least one pathogenic fungus and/or oomycete; andc) selecting plants showing an enhanced resistance to said at least one pathogenic fungus and/or oomycete compared to a control plant or to a pre-determined resistance score value, thereby selecting a plant having enhanced resistance to said at least one pathogenic fungus.
  • 10. The method of claim 9, wherein the at least one polypeptide at least 70% identical to the polypeptide having the amino acid sequence set forth in SEQ ID NO:2618 comprises an amino acid sequence selected from the group consisting of SEQ ID NOs:3941-3953, a fragment of said polypeptide comprises the amino acid sequence set fort SEQ ID NO:4685, and a variant of said polypeptide comprises the amino acid sequence set forth in SEQ ID NO:4498.
  • 11. A genetically engineered plant having enhanced resistance to at least one fungus and/or oomycete compared to a non-engineered control plant, the genetically engineered plant comprises at least one cell having enhanced expression and/or activity of at least one polypeptide at least 70% identical to a polypeptide having the amino acid sequence set forth in SEQ ID NO:2618, a fragment or a variant thereof compared to the polypeptide expression and/or activity in the non-engineered control plant.
  • 12. The genetically engineered plant of claim 11, wherein the at least one polypeptide at least 70% identical to the polypeptide having the amino acid sequence set forth in SEQ ID NO:2618 comprises an amino acid sequence selected from the group consisting of SEQ ID NOs:3941-3953, a fragment of said polypeptide comprises the amino acid sequence set fort SEQ ID NO:4685, and a variant of said polypeptide comprises the amino acid sequence set forth in SEQ ID NO:4498.
  • 13. A nucleic acid construct comprising a polynucleotide comprising a nucleic acid sequence encoding a polypeptide comprising an amino acid sequence at least 70% identical to the amino acid sequence set forth in SEQ ID NO:2618, a fragment or a variant thereof, wherein the polynucleotide further comprises at least one regulatory element for directing the expression of said polynucleotide within a plant cell, and wherein the polypeptide, when expressed in the plant cell, is capable of enhancing the resistance of the plant to at least one pathogenic fungus and/or oomycete.
  • 14. A genetically engineered plant cell expressing the construct according to claim 13.
  • 15. A fungicidal composition comprising an effective amount of at least one polypeptide at least 70% identical to a polypeptide having the amino acid sequence set forth in SEQ ID NO:2618, a fragment and/or a variant thereof, wherein the fungicidal composition is effective in inhibiting the growth and/or development of at least one plant pathogenic fungi and/or oomycetes.
  • 16. The fungicidal composition of claim 15, wherein the at least one polypeptide at least 70% identical to the polypeptide having the amino acid sequence set forth in SEQ ID NO:2618 comprises an amino acid sequence selected from the group consisting of SEQ ID NOs:3941-3953, a fragment of said polypeptide comprises the amino acid sequence set fort SEQ ID NO:4685, and a variant of said polypeptide comprises the amino acid sequence set forth in SEQ ID NO:4498.
  • 17. A plurality of bacteria, each bacterium comprising a recombinant polynucleotide optimized for expression of at least one recombinant polypeptide in the bacterium, wherein the at least one expressed recombinant polypeptide is at least 70% identical to a polypeptide having the amino acid sequence set forth in SEQ ID NO:2618, a fragment or a variant thereof, wherein said at least one expressed recombinant polypeptide is effective in inhibiting the growth and/or development of at least one plant pathogenic fungi and/or oomycetes.
  • 18. The plurality of bacteria of claim 17, wherein the at least one polypeptide at least 70% identical to the polypeptide having the amino acid sequence set forth in SEQ ID NO:2618 comprises an amino acid sequence selected from the group consisting of SEQ ID NOs:3941-3953, a fragment of said polypeptide comprises the amino acid sequence set fort SEQ ID NO:4685, and a variant of said polypeptide comprises the amino acid sequence set forth in SEQ ID NO:4498.
  • 19. A fungicidal composition comprising the plurality of bacteria of claim 18.
  • 20. The fungicidal composition of claim 19, wherein said fungicidal composition further comprises at least one agriculturally compatible agent selected from the group consisting of a carrier, a stabilizer, a diluent, a surfactant, a mineral and an adjuvant.
CROSS REFERENCE TO RELATED APPLICATIONS

This application is a Divisional application of U.S. application Ser. No. 17/046,110, filed Oct. 8, 2020; which is a national phase of PCT International Application No. PCT/IL2019/050399, filed on Apr. 10, 2019; which claims priority to U.S. Application No. 62/656,377, filed on Apr. 12, 2018, which are hereby incorporated by reference in their entirety.

Provisional Applications (1)
Number Date Country
62656377 Apr 2018 US
Divisions (1)
Number Date Country
Parent 17046110 Oct 2020 US
Child 18360543 US