Agricultural compositions comprising remodeled nitrogen fixing microbes

Information

  • Patent Grant
  • 11963530
  • Patent Number
    11,963,530
  • Date Filed
    Wednesday, June 26, 2019
    4 years ago
  • Date Issued
    Tuesday, April 23, 2024
    18 days ago
  • CPC
    • A01N63/20
  • Field of Search
    • US
    • NON E00000
  • International Classifications
    • A01N63/20
Abstract
The present disclosure provides non-intergeneric remodeled microbes that are able to fix atmospheric nitrogen and deliver such to plants in a targeted, efficient, and environmentally sustainable manner. The utilization of the taught microbial products will enable farmers to realize more productive and predictable crop yields without the nutrient degradation, leaching, or toxic runoff associated with traditional synthetically derived nitrogen fertilizer. The remodeled microbes taught herein are able to be combined with leading agricultural chemistry and elite germplasm. Furthermore, the disclosure provides seed treatments comprising remodeled microbes.
Description
STATEMENT REGARDING SEQUENCE LISTING

The contents of the text file submitted electronically herewith are incorporated herein by reference in their entirety: A computer readable format copy of the Sequence Listing filename: PIVO_004_02WO_SeqList_ST25.txt, date created, Jun. 18, 2019, file size ≈578 kilobytes.


BACKGROUND OF THE DISCLOSURE

By 2050 the United Nations' Food and Agriculture Organization projects that total food production must increase by 70% to meet the needs of a growing population, a challenge that is exacerbated by numerous factors, including: diminishing freshwater resources, increasing competition for arable land, rising energy prices, increasing input costs, and the likely need for crops to adapt to the pressures of a drier, hotter, and more extreme global climate.


Current agricultural practices are not well equipped to meet this growing demand for food production, while simultaneously balancing the environmental impacts that result from increased agricultural intensity.


One of the major agricultural inputs needed to satisfy global food demand is nitrogen fertilizer. However, the current industrial standard utilized to produce nitrogen fertilizer, is an artificial nitrogen fixation method called the Haber-Bosch process, which converts atmospheric nitrogen (N2) to ammonia (NH3) by a reaction with hydrogen (H2) using a metal catalyst under high temperatures and pressures. This process is resource intensive and deleterious to the environment.


In contrast to the synthetic Haber-Bosch process, certain biological systems have evolved to fix atmospheric nitrogen. These systems utilize an enzyme called nitrogenase that catalyzes the reaction between N2 and H2, and results in nitrogen fixation. For example, rhizobia are diazotrophic bacteria that fix nitrogen after becoming established inside root nodules of legumes. An important goal of nitrogen fixation research is the extension of this phenotype to non-leguminous plants, particularly to important agronomic grasses such as wheat, rice, and corn. However, despite the significant progress made in understanding the development of the nitrogen-fixing symbiosis between rhizobia and legumes, the path to use that knowledge to induce nitrogen-fixing nodules on non-leguminous crops is still not clear.


Consequently, the vast majority of modern row crop agriculture utilizes nitrogen fertilizer that is produced via the resource intensive and environmentally deleterious Haber-Bosch process. For instance, the USDA indicates that the average U.S. corn farmer typically applies between 130 and 200 lb. of nitrogen per acre (146 to 224 kg/ha). This nitrogen is not only produced in a resource intensive synthetic process, but is applied by heavy machinery crossing/impacting the field's soil, burning petroleum, and requiring hours of human labor.


Furthermore, the nitrogen fertilizer produced by the industrial Haber-Bosch process is not well utilized by the target crop. Rain, runoff, heat, volatilization, and the soil microbiome degrade the applied chemical fertilizer. This equates to not only wasted money, but also adds to increased pollution instead of harvested yield. To this end, the United Nations has calculated that nearly 80% of fertilizer is lost before a crop can utilize it. Consequently, modern agricultural fertilizer production and delivery is not only deleterious to the environment, but it is extremely inefficient.


In order to meet the world's growing food supply needs—while also balancing resource utilization and providing minimal impacts upon environmental systems—a better approach to nitrogen fixation and delivery to plants is urgently needed.


SUMMARY OF THE DISCLOSURE

In some aspects, the disclosure is generally drawn to a seed treatment composition, comprising: (a) a plurality of non-intergeneric remodeled bacteria that have an average colonization ability per unit of plant root tissue of at least about 1.0×104 bacterial cells per gram of fresh weight of plant root tissue and produce fixed N of at least about 1×10−17 mmol N per bacterial cell per hour; and (b) at least one pesticide.


In some aspects, the pesticide is a fungicide. In some aspects, the pesticide is a fungicide selected from the group consisting of: fludioxonil, metalaxyl, mefenoxam, azoxystrobin, thiabendazole, ipconazole, tebuconazole, prothioconazole, and combinations thereof.


In some aspects, the pesticide is an insecticide. In some aspects, the pesticide is a neonicotinoid insecticide. In some aspects, the pesticide is an insecticide selected from the group consisting of: imidacloprid, clothianidin, thiamethoxam, chlorantraniliprole, and combinations thereof.


In some aspects, the at least one pesticide is a fungicide and an insecticide combination. In some aspects, the pesticide is a nematicide. In some aspects, the pesticide is an herbicide. In some aspects, the pesticide is selected from those in Table 13.


In some aspects, the non-intergeneric remodeled bacteria and pesticide exhibit a synergistic effect.


In some aspects, the seed treatment is disposed onto a seed. In some aspects, the seed treatment is disposed onto a seed from the family Poaceae. In some aspects, the seed treatment is disposed onto a cereal seed. In some aspects, the seed treatment is disposed onto a corn, rice, wheat, barley, Sorghum, millet, oat, rye, or triticale seed. In some aspects, the seed treatment is disposed onto a corn seed. In some aspects, the seed treatment is disposed onto a genetically modified corn seed.


In some aspects, the seed treatment is disposed onto a genetically modified corn seed, wherein said corn comprises an herbicide tolerant trait. In some aspects, the seed treatment is disposed onto a genetically modified corn seed, wherein said corn comprises an insect resistant trait. In some aspects, the seed treatment is disposed onto a genetically modified corn seed, wherein said corn comprises an herbicide tolerant trait and an insect resistance trait. In some aspects, the seed treatment is disposed onto a genetically modified corn seed, wherein said corn comprises a trait listed in Table 19.


In some aspects, the seed treatment is disposed onto a non-genetically modified corn seed. In some aspects, the seed treatment is disposed onto a sweet corn, flint corn, popcorn, dent corn, pod corn, or flour corn.


In some aspects, the plurality of non-intergeneric remodeled bacteria produce 1% or more of the fixed nitrogen in a plant exposed thereto. In some aspects, the non-intergeneric remodeled bacteria are capable of fixing atmospheric nitrogen in the presence of exogenous nitrogen.


In some aspects, each member of the plurality of non-intergeneric remodeled bacteria comprises at least one genetic variation introduced into at least one gene, or non-coding polynucleotide, of the nitrogen fixation or assimilation genetic regulatory network. In some aspects, each member of the plurality of non-intergeneric remodeled bacteria comprises an introduced control sequence operably linked to at least one gene of the nitrogen fixation or assimilation genetic regulatory network. In some aspects, each member of the plurality of non-intergeneric remodeled bacteria comprises a heterologous promoter operably linked to at least one gene of the nitrogen fixation or assimilation genetic regulatory network.


In some aspects, each member of the plurality of non-intergeneric remodeled bacteria comprises at least one genetic variation introduced into a member selected from the group consisting of nifA, nifL, ntrB, ntrC, polynucleotide encoding glutamine synthetase, glnA, glnB, glnK, drat, amtB, polynucleotide encoding glutaminase, glnD, glnE, nifJ, nifH, nifD, nifK, nifY, nifE, nifN, nifJ, nifS, nifV, nifW, nifZ, nifM, nifF, nifB, nifQ, a gene associated with biosynthesis of a nitrogenase enzyme, or combinations thereof.


In some aspects, each member of the plurality of non-intergeneric remodeled bacteria comprises at least one genetic variation introduced into at least one gene, or non-coding polynucleotide, of the nitrogen fixation or assimilation genetic regulatory network that results in one or more of: increased expression or activity of NifA or glutaminase; decreased expression or activity of NifL, NtrB, glutamine synthetase, GlnB, GlnK, DraT, AmtB; decreased adenylyl-removing activity of GlnE; or decreased uridylyl-removing activity of GlnD.


In some aspects, each member of the plurality of non-intergeneric remodeled bacteria comprises a mutated nifL gene that has been altered to comprise a heterologous promoter inserted into said nifL gene. In some aspects, each member of the plurality of non-intergeneric remodeled bacteria comprises a mutated glnE gene that results in a truncated GlnE protein lacking an adenylyl-removing (AR) domain. In some aspects, each member of the plurality of non-intergeneric remodeled bacteria comprises a mutated amtB gene that results in the lack of expression of said amtB gene.


In some aspects, each member of the plurality of non-intergeneric remodeled bacteria comprises at least one of: a mutated nifL gene that has been altered to comprise a heterologous promoter inserted into said nifL gene; a mutated glnE gene that results in a truncated GlnE protein lacking an adenylyl-removing (AR) domain; a mutated amtB gene that results in the lack of expression of said amtB gene; and combinations thereof. In some aspects, each member of the plurality of non-intergeneric remodeled bacteria comprises a mutated nifL gene that has been altered to comprise a heterologous promoter inserted into said nifL gene and a mutated glnE gene that results in a truncated GlnE protein lacking an adenylyl-removing (AR) domain. In some aspects, each member of the plurality of non-intergeneric remodeled bacteria comprises a mutated nifL gene that has been altered to comprise a heterologous promoter inserted into said nifL gene and a mutated glnE gene that results in a truncated GlnE protein lacking an adenylyl-removing (AR) domain and a mutated amtB gene that results in the lack of expression of said amtB gene.


In some aspects, the plurality of non-intergeneric remodeled bacteria are present at a concentration of about 1×105 to about 1×107 cfu per seed. In some aspects, the plurality of non-intergeneric remodeled bacteria comprise at least two different species of bacteria. In some aspects, the plurality of non-intergeneric remodeled bacteria comprise at least two different strains of the same species of bacteria.


In some aspects, the plurality of non-intergeneric remodeled bacteria comprise bacteria selected from: Rahnella aquatilis, Klebsiella variicola, Achromobacter spiritinus, Achromobacter marplatensis, Microbacterium murale, Kluyvera intermedia, Kosakonia pseudosacchari, Enterobacter sp., Azospirillum lipoferum, Kosakonia sacchari, and combinations thereof.


In some aspects, the plurality of non-intergeneric remodeled bacteria are endophytic, epiphytic, or rhizospheric.


In some aspects, the plurality of non-intergeneric remodeled bacteria comprise bacteria selected from: a bacteria deposited as NCMA 201701002, a bacteria deposited as NCMA 201708004, a bacteria deposited as NCMA 201708003, a bacteria deposited as NCMA 201708002, a bacteria deposited as NCMA 201712001, a bacteria deposited as NCMA 201712002, and combinations thereof.


In some aspects, the plurality of non-intergeneric remodeled bacteria comprise bacteria with a nucleic acid sequence that shares at least about 90% sequence identity to a nucleic acid sequence selected from SEQ ID NOs: 177-260, 296-303. In some aspects, the plurality of non-intergeneric remodeled bacteria comprise bacteria with a nucleic acid sequence that shares at least about 95% sequence identity to a nucleic acid sequence selected from SEQ ID NOs: 177-260, 296-303. In some aspects, the plurality of non-intergeneric remodeled bacteria comprise bacteria with a nucleic acid sequence that shares at least about 99% sequence identity to a nucleic acid sequence selected from SEQ ID NOs: 177-260, 296-303. In some aspects, the plurality of non-intergeneric remodeled bacteria comprise bacteria with a nucleic acid sequence selected from SEQ ID NOs: 177-260, 296-303.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1A depicts an overview of the guided microbial remodeling process, in accordance with embodiments.



FIG. 1B depicts an expanded view of the measurement of microbiome composition as shown in FIG. 1A.



FIG. 1C depicts a problematic “traditional bioprospecting” approach, which has several drawbacks compared to the taught guided microbial remodeling (GMR) platform.



FIG. 1D depicts a problematic “field-first approach to bioprospecting” system, which has several drawbacks compared to the taught guided microbial remodeling (GMR) platform.



FIG. 1E depicts the time period in the corn growth cycle, at which nitrogen is needed most by the plant.



FIG. 1F depicts an overview of a field development process for a remodeled microbe.



FIG. 1G depicts an overview of a guided microbial remodeling platform embodiment.



FIG. 1H depicts an overview of a computationally-guided microbial remodeling platform.



FIG. 1I depicts the use of field data combined with modeling in aspects of the guided microbial remodeling platform.



FIG. 1J depicts five properties that can be possessed by remodeled microbes of the present disclosure.



FIG. 1K depicts a schematic of a remodeling approach for a microbe, PBC6.1.



FIG. 1L depicts decoupled nifA expression from endogenous nitrogen regulation in remodeled microbes.



FIG. 1M depicts improved assimilation and excretion of fixed nitrogen by remodeled microbes.



FIG. 1N depicts corn yield improvement attributable to remodeled microbes.



FIG. 1O illustrates the inefficiency of current nitrogen delivery systems, which result in underfertilized fields, over fertilized fields, and environmentally deleterious nitrogen runoff



FIG. 2 illustrates PBC6.1 colonization to nearly 21% abundance of the root-associated microbiota in corn roots. Abundance data is based on 16S amplicon sequencing of the rhizosphere and endosphere of corn plants inoculated with PBC6.1 and grown in greenhouse conditions.



FIGS. 3A-3E illustrate derivative microbes that fix and excrete nitrogen in vitro under conditions similar to high nitrate agricultural soils. FIG. 3A illustrates the regulatory network controlling nitrogen fixation and assimilation in PBC6.1 is shown, including the key nodes NifL, NifA, GS, GlnE depicted as the two-domain ATase-AR enzyme, and AmtB. FIG. 3B illustrates the genome of Kosakonia sacchari isolate PBC6.1 is shown. The three tracks circumscribing the genome convey transcription data from PBC6.1, PBC6.38, and the differential expression between the strains respectively. FIG. 3C illustrates the nitrogen fixation gene cluster and transcription data is expanded for finer detail. FIG. 3D illustrates nitrogenase activity under varying concentrations of exogenous nitrogen is measured with the acetylene reduction assay. The wild type strain exhibits repression of nitrogenase activity as glutamine concentrations increase, while derivative strains show varying degrees of robustness. In the line graph, triangles represent strain PBC6.22; circles represent strain PBC6.1; squares represent strain PBC6.15; and diamonds represent strain PBC6.14. Error bars represent standard error of the mean of at least three biological replicates. FIG. 3E illustrates temporal excretion of ammonia by derivative strains is observed at mM concentrations. Wild type strains are not observed to excrete fixed nitrogen, and negligible ammonia accumulates in the media. Error bars represent standard error of the mean.



FIG. 4 illustrates transcriptional rates of nifA in derivative strains of PBC6.1 correlated with acetylene reduction rates. An ARA assay was performed as described in the Methods, after which cultures were sampled and subjected to qPCR analysis to determine nifA transcript levels. Error bars show standard error of the mean of at least three biological replicates in each measure.



FIGS. 5A-5C illustrate greenhouse experiments that demonstrate microbial nitrogen fixation in corn. FIG. 5A illustrates microbe colonization six weeks after inoculation of corn plants by PBC6.1 derivative strains. Error bars show standard error of the mean of at least eight biological replicates. FIG. 5B illustrates in planta transcription of nifH measured by extraction of total RNA from roots and subsequent Nanostring analysis. Only derivative strains show nifH transcription in the root environment. Error bars show standard error of the mean of at least three biological replicates. FIG. 5C illustrates microbial nitrogen fixation measured by the dilution of isotopic tracer in plant tissues. Derivative microbes exhibit substantial transfer of fixed nitrogen to the plant. Error bars show standard error of the mean of at least ten biological replicates.



FIG. 6 depicts the lineage of modified strains that were derived from strain CI006.



FIG. 7 depicts the lineage of modified strains that were derived from strain CI019.



FIG. 8 depicts a heatmap of the pounds of nitrogen delivered per acre-season by microbes of the present disclosure recorded as a function of microbes per g-fresh weight by mmol of nitrogen/microbe-hr. Below the thin line that transects the larger image are the microbes that deliver less than one pound of nitrogen per acre-season, and above the line are the microbes that deliver greater than one pound of nitrogen per acre-season. The table below the heatmap gives the precise value of mmol N produced per microbe per hour (mmol N/Microbe hr) along with the precise CFU per gram of fresh weight (CFU/g fw) for each microbe shown in the heatmap. The microbes utilized in the heatmap were assayed for N production in corn. For the WT strains CI006 and CI019, corn root colonization data was taken from a single field site. For the remaining strains, colonization was assumed to be the same as the WT field level. N-fixation activity was determined using an in vitro ARA assay at 5 mM glutamine.



FIG. 9 depicts the plant yield of plants having been exposed to strain CI006. The area of the circles corresponds to the relative yield, while the shading corresponds to the particular MRTN treatment. The x-axis is the p value and the y-axis is the win rate.



FIG. 10 depicts the plant yield of plants having been exposed to strain CM029. The area of the circles corresponds to the relative yield, while the shading corresponds to the particular MRTN treatment. The x-axis is the p value and the y-axis is the win rate.



FIG. 11 depicts the plant yield of plants having been exposed to strain CM038. The area of the circles corresponds to the relative yield, while the shading corresponds to the particular MRTN treatment. The x-axis is the p value and the y-axis is the win rate.



FIG. 12 depicts the plant yield of plants having been exposed to strain CI019. The area of the circles corresponds to the relative yield, while the shading corresponds to the particular MRTN treatment. The x-axis is the p value and the y-axis is the win rate.



FIG. 13 depicts the plant yield of plants having been exposed to strain CM081. The area of the circles corresponds to the relative yield, while the shading corresponds to the particular MRTN treatment. The x-axis is the p value and the y-axis is the win rate.



FIG. 14 depicts the plant yield of plants having been exposed to strains CM029 and CM081. The area of the circles corresponds to the relative yield, while the shading corresponds to the particular MRTN treatment. The x-axis is the p value and the y-axis is the win rate.



FIG. 15 depicts the plant yield of plants as the aggregated bushel gain/loss. The area of the circles corresponds to the relative yield, while the shading corresponds to the particular MRTN treatment. The x-axis is the p value and the y-axis is the win rate.



FIG. 16 illustrates results from a summer 2017 field testing experiment. The yield results obtained demonstrate that the microbes of the disclosure can serve as a potential fertilizer replacement. For instance, the utilization of a microbe of the disclosure (i.e. 6-403) resulted in a higher yield than the wild type strain (WT) and a higher yield than the untreated control (UTC). The “−25 lbs N” treatment utilizes 25 lbs less N per acre than standard agricultural practices of the region. The “100% N” UTC treatment is meant to depict standard agricultural practices of the region, in which 100% of the standard utilization of N is deployed by the farmer. The microbe “6-403” was deposited as NCMA 201708004 and can be found in Table 1. This is a mutant Kosakonia sacchari (also called CM037) and is a progeny mutant strain from CI006 WT.



FIG. 17 illustrates results from a summer 2017 field testing experiment. The yield results obtained demonstrate that the microbes of the disclosure perform consistently across locations. Furthermore, the yield results demonstrate that the microbes of the disclosure perform well in both a nitrogen stressed environment, as well as an environment that has sufficient supplies of nitrogen. The microbe “6-881” (also known as CM094, PBC6.94), and which is a progeny mutant Kosakonia sacchari strain from CI006 WT, was deposited as NCMA 201708002 and can be found in Table 1. The microbe “137-1034,” which is a progeny mutant Klebsiella variicola strain from CI137 WT, was deposited as NCMA 201712001 and can be found in Table 1. The microbe “137-1036,” which is a progeny mutant Klebsiella variicola strain from CI137 WT, was deposited as NCMA 201712002 and can be found in Table 1. The microbe “6-404” (also known as CM38, PBC6.38), and which is a progeny mutant Kosakonia sacchari strain from CI006 WT, was deposited as NCMA 201708003 and can be found in Table 1. The “Nutrient Stress” condition corresponds to the 0% nitrogen regime. The “Sufficient Fertilizer” condition corresponds to the 100% nitrogen regime.



FIG. 18 depicts the lineage of modified strains that were derived from strain CI006 (also termed “6”, Kosakonia sacchari WT).



FIG. 19 depicts the lineage of modified strains that were derived from strain CI019 (also termed “19”, Rahnella aquatilis WT).



FIG. 20 depicts the lineage of modified strains that were derived from strain CI137 (also termed (“137”, Klebsiella variicola WT).



FIG. 21 depicts the lineage of modified strains that were derived from strain 1021 (Kosakonia pseudosacchari WT).



FIG. 22 depicts the lineage of modified strains that were derived from strain 910 (Kluyvera intermedia WT).



FIG. 23 depicts the lineage of modified strains that were derived from strain 63 (Rahnella aquatilis WT).



FIG. 24 depicts a heatmap of the pounds of nitrogen delivered per acre-season by microbes of the present disclosure recorded as a function of microbes per g-fresh weight by mmol of nitrogen/microbe-hr. Below the thin line that transects the larger image are the microbes that deliver less than one pound of nitrogen per acre-season, and above the line are the microbes that deliver greater than one pound of nitrogen per acre-season. The Table 28 in Example 5 gives the precise value of mmol N produced per microbe per hour (mmol N/Microbe hr) along with the precise CFU per gram of fresh weight (CFU/g fw) for each microbe shown in the heatmap. The data in FIG. 24 is derived from microbial strains assayed for N production in corn in field conditions. Each point represents lb N/acre produced by a microbe using corn root colonization data from a single field site. N-fixation activity was determined using in vitro ARA assay at 5 mM N in the form of glutamine or ammonium phosphate.



FIG. 25 depicts a heatmap of the pounds of nitrogen delivered per acre-season by microbes of the present disclosure recorded as a function of microbes per g-fresh weight by mmol of nitrogen/microbe-hr. Below the thin line that transects the larger image are the microbes that deliver less than one pound of nitrogen per acre-season, and above the line are the microbes that deliver greater than one pound of nitrogen per acre-season. The Table 29 in Example 5 gives the precise value of mmol N produced per microbe per hour (mmol N/Microbe hr) along with the precise CFU per gram of fresh weight (CFU/g fw) for each microbe shown in the heatmap. The data in FIG. 25 is derived from microbial strains assayed for N production in corn in laboratory and greenhouse conditions. Each point represents lb N/acre produced by a single strain. White points represent strains in which corn root colonization data was gathered in greenhouse conditions. Black points represent mutant strains for which corn root colonization levels are derived from average field corn root colonization levels of the wild-type parent strain. Hatched points represent the wild type parent strains at their average field corn root colonization levels. In all cases, N-fixation activity was determined by in vitro ARA assay at 5 mM N in the form of glutamine or ammonium phosphate.





DETAILED DESCRIPTION OF THE DISCLOSURE

While various embodiments of the disclosure have been shown and described herein, it will be obvious to those skilled in the art that such embodiments are provided by way of example only. Numerous variations, changes, and substitutions may occur to those skilled in the art without departing from the disclosure. It should be understood that various alternatives to the embodiments of the disclosure described herein may be employed.


Increased fertilizer utilization brings with it environmental concerns and is also likely not possible for many economically stressed regions of the globe. Furthermore, many industry players in the microbial arena are focused on creating intergeneric microbes. However, there is a heavy regulatory burden placed on engineered microbes that are characterized/classified as intergeneric. These intergeneric microbes face not only a higher regulatory burden, which makes widespread adoption and implementation difficult, but they also face a great deal of public perception scrutiny.


Currently, there are no engineered microbes on the market that are non-intergeneric and that are capable of increasing nitrogen fixation in non-leguminous crops. This dearth of such a microbe is a missing element in helping to usher in a truly environmentally friendly and more sustainable 21st century agricultural system.


The present disclosure solves the aforementioned problems and provides a non-intergeneric microbe that has been engineered to readily fix nitrogen in crops. These microbes are not characterized/classified as intergeneric microbes and thus will not face the steep regulatory burdens of such. Further, the taught non-intergeneric microbes will serve to help 21st century farmers become less dependent upon utilizing ever increasing amounts of exogenous nitrogen fertilizer.


Definitions

The use of the terms “a” and “an” and “the” and similar referents in the context of describing the disclosure (especially in the context of the following claims) are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context. The terms “comprising,” “having,” “including,” and “containing” are to be construed as open-ended terms (i.e., meaning “including, but not limited to,”) unless otherwise noted. Recitation of ranges of values herein are merely intended to serve as a shorthand method of referring individually to each separate value falling within the range, unless otherwise indicated herein, and each separate value is incorporated into the specification as if it were individually recited herein. For example, if the range 10-15 is disclosed, then 11, 12, 13, and 14 are also disclosed. All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g., “such as”) provided herein, is intended merely to better illuminate the disclosure and does not pose a limitation on the scope of the disclosure unless otherwise claimed. No language in the specification should be construed as indicating any non-claimed element as essential to the practice of the disclosure.


The terms “polynucleotide”, “nucleotide”, “nucleotide sequence”, “nucleic acid” and “oligonucleotide” are used interchangeably. They refer to a polymeric form of nucleotides of any length, either deoxyribonucleotides or ribonucleotides, or analogs thereof. Polynucleotides may have any three dimensional structure, and may perform any function, known or unknown. The following are non-limiting examples of polynucleotides: coding or non-coding regions of a gene or gene fragment, loci (locus) defined from linkage analysis, exons, introns, messenger RNA (mRNA), transfer RNA (tRNA), ribosomal RNA (rRNA), short interfering RNA (siRNA), short-hairpin RNA (shRNA), micro-RNA (miRNA), ribozymes, cDNA, recombinant polynucleotides, branched polynucleotides, plasmids, vectors, isolated DNA of any sequence, isolated RNA of any sequence, nucleic acid probes, and primers. A polynucleotide may comprise one or more modified nucleotides, such as methylated nucleotides and nucleotide analogs. If present, modifications to the nucleotide structure may be imparted before or after assembly of the polymer. The sequence of nucleotides may be interrupted by non-nucleotide components. A polynucleotide may be further modified after polymerization, such as by conjugation with a labeling component.


“Hybridization” refers to a reaction in which one or more polynucleotides react to form a complex that is stabilized via hydrogen bonding between the bases of the nucleotide residues. The hydrogen bonding may occur by Watson Crick base pairing, Hoogstein binding, or in any other sequence specific manner according to base complementarity. The complex may comprise two strands forming a duplex structure, three or more strands forming a multi stranded complex, a single self-hybridizing strand, or any combination of these. A hybridization reaction may constitute a step in a more extensive process, such as the initiation of PCR, or the enzymatic cleavage of a polynucleotide by an endonuclease. A second sequence that is complementary to a first sequence is referred to as the “complement” of the first sequence. The term “hybridizable” as applied to a polynucleotide refers to the ability of the polynucleotide to form a complex that is stabilized via hydrogen bonding between the bases of the nucleotide residues in a hybridization reaction.


“Complementarity” refers to the ability of a nucleic acid to form hydrogen bond(s) with another nucleic acid sequence by either traditional Watson-Crick or other non-traditional types. A percent complementarity indicates the percentage of residues in a nucleic acid molecule which can form hydrogen bonds (e.g., Watson-Crick base pairing) with a second nucleic acid sequence (e.g., 5, 6, 7, 8, 9, 10 out of 10 being 50%, 60%, 70%, 80%, 90%, and 100% complementary, respectively). “Perfectly complementary” means that all the contiguous residues of a nucleic acid sequence will hydrogen bond with the same number of contiguous residues in a second nucleic acid sequence. “Substantially complementary” as used herein refers to a degree of complementarity that is at least 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98%, 99%, or 100% over a region of 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30, 35, 40, 45, 50, or more nucleotides, or refers to two nucleic acids that hybridize under stringent conditions. Sequence identity, such as for the purpose of assessing percent complementarity, may be measured by any suitable alignment algorithm, including but not limited to the Needleman-Wunsch algorithm (see e.g. the EMBOSS Needle aligner available at www.ebi.ac.uk/Tools/psa/emboss_needle/nucleotide.html, optionally with default settings), the BLAST algorithm (see e.g. the BLAST alignment tool available at blast.ncbi.nlm.nih.gov/Blast.cgi, optionally with default settings), or the Smith-Waterman algorithm (see e.g. the EMBOSS Water aligner available at www.ebi.ac.uk/Tools/psa/emboss_water/nucleotide.html, optionally with default settings). Optimal alignment may be assessed using any suitable parameters of a chosen algorithm, including default parameters.


In general, “stringent conditions” for hybridization refer to conditions under which a nucleic acid having complementarity to a target sequence predominantly hybridizes with a target sequence, and substantially does not hybridize to non-target sequences. Stringent conditions are generally sequence-dependent and vary depending on a number of factors. In general, the longer the sequence, the higher the temperature at which the sequence specifically hybridizes to its target sequence. Non-limiting examples of stringent conditions are described in detail in Tijssen (1993), Laboratory Techniques In Biochemistry And Molecular Biology-Hybridization With Nucleic Acid Probes Part I, Second Chapter “Overview of principles of hybridization and the strategy of nucleic acid probe assay”, Elsevier, N.Y.


As used herein, “expression” refers to the process by which a polynucleotide is transcribed from a DNA template (such as into and mRNA or other RNA transcript) and/or the process by which a transcribed mRNA is subsequently translated into peptides, polypeptides, or proteins. Transcripts and encoded polypeptides may be collectively referred to as “gene product.” If the polynucleotide is derived from genomic DNA, expression may include splicing of the mRNA in a eukaryotic cell.


The terms “polypeptide”, “peptide” and “protein” are used interchangeably herein to refer to polymers of amino acids of any length. The polymer may be linear or branched, it may comprise modified amino acids, and it may be interrupted by non-amino acids. The terms also encompass an amino acid polymer that has been modified; for example, disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation, such as conjugation with a labeling component. As used herein the term “amino acid” includes natural and/or unnatural or synthetic amino acids, including glycine and both the D or L optical isomers, and amino acid analogs and peptidomimetics.


As used herein, the term “about” is used synonymously with the term “approximately.” Illustratively, the use of the term “about” with regard to an amount indicates that values slightly outside the cited values, e.g., plus or minus 0.1% to 10%.


The term “biologically pure culture” or “substantially pure culture” refers to a culture of a bacterial species described herein containing no other bacterial species in quantities sufficient to interfere with the replication of the culture or be detected by normal bacteriological techniques.


“Plant productivity” refers generally to any aspect of growth or development of a plant that is a reason for which the plant is grown. For food crops, such as grains or vegetables, “plant productivity” can refer to the yield of grain or fruit harvested from a particular crop. As used herein, improved plant productivity refers broadly to improvements in yield of grain, fruit, flowers, or other plant parts harvested for various purposes, improvements in growth of plant parts, including stems, leaves and roots, promotion of plant growth, maintenance of high chlorophyll content in leaves, increasing fruit or seed numbers, increasing fruit or seed unit weight, reducing NO2 emission due to reduced nitrogen fertilizer usage and similar improvements of the growth and development of plants.


Microbes in and around food crops can influence the traits of those crops. Plant traits that may be influenced by microbes include: yield (e.g., grain production, biomass generation, fruit development, flower set); nutrition (e.g., nitrogen, phosphorus, potassium, iron, micronutrient acquisition); abiotic stress management (e.g., drought tolerance, salt tolerance, heat tolerance); and biotic stress management (e.g., pest, weeds, insects, fungi, and bacteria). Strategies for altering crop traits include: increasing key metabolite concentrations; changing temporal dynamics of microbe influence on key metabolites; linking microbial metabolite production/degradation to new environmental cues; reducing negative metabolites; and improving the balance of metabolites or underlying proteins.


As used herein, a “control sequence” refers to an operator, promoter, silencer, or terminator.


As used herein, “in planta” may refer to in the plant, on the plant, or intimately associated with the plant, depending upon context of usage (e.g. endophytic, epiphytic, or rhizospheric associations). The plant may comprise plant parts, tissue, leaves, roots, root hairs, rhizomes, stems, seed, ovules, pollen, flowers, fruit, etc.


In some embodiments, native or endogenous control sequences of genes of the present disclosure are replaced with one or more intrageneric control sequences.


As used herein, “introduced” refers to the introduction by means of modern biotechnology, and not a naturally occurring introduction.


In some embodiments, the bacteria of the present disclosure have been modified such that they are not naturally occurring bacteria.


In some embodiments, the bacteria of the present disclosure are present in the plant in an amount of at least 103 cfu, 104 cfu, 105 cfu, 106 cfu, 107 cfu, 108 cfu, 109 cfu, 1010 cfu, 1011 cfu, or 1012 cfu per gram of fresh or dry weight of the plant. In some embodiments, the bacteria of the present disclosure are present in the plant in an amount of at least about 103 cfu, about 104 cfu, about 105 cfu, about 106 cfu, about 107 cfu, about 108 cfu, about 109 cfu, about 1010 cfu, about 1011 cfu, or about 1012 cfu per gram of fresh or dry weight of the plant. In some embodiments, the bacteria of the present disclosure are present in the plant in an amount of at least 103 to 109, 103 to 107, 103 to 105, 105 to 109, 105 to 107, 106 to 1010, 106 to 107 cfu per gram of fresh or dry weight of the plant.


Fertilizers and exogenous nitrogen of the present disclosure may comprise the following nitrogen-containing molecules: ammonium, nitrate, nitrite, ammonia, glutamine, etc. Nitrogen sources of the present disclosure may include anhydrous ammonia, ammonia sulfate, urea, diammonium phosphate, urea-form, monoammonium phosphate, ammonium nitrate, nitrogen solutions, calcium nitrate, potassium nitrate, sodium nitrate, etc.


As used herein, “exogenous nitrogen” refers to non-atmospheric nitrogen readily available in the soil, field, or growth medium that is present under non-nitrogen limiting conditions, including ammonia, ammonium, nitrate, nitrite, urea, uric acid, ammonium acids, etc.


As used herein, “non-nitrogen limiting conditions” refers to non-atmospheric nitrogen available in the soil, field, media at concentrations greater than about 4 mM nitrogen, as disclosed by Kant et al. (2010. J. Exp. Biol. 62(4):1499-1509), which is incorporated herein by reference.


As used herein, an “intergeneric microorganism” is a microorganism that is formed by the deliberate combination of genetic material originally isolated from organisms of different taxonomic genera. An “intergeneric mutant” can be used interchangeably with “intergeneric microorganism”. An exemplary “intergeneric microorganism” includes a microorganism containing a mobile genetic element which was first identified in a microorganism in a genus different from the recipient microorganism. Further explanation can be found, inter alia, in 40 C.F.R. § 725.3.


In aspects, microbes taught herein are “non-intergeneric,” which means that the microbes are not intergeneric.


As used herein, an “intrageneric microorganism” is a microorganism that is formed by the deliberate combination of genetic material originally isolated from organisms of the same taxonomic genera. An “intrageneric mutant” can be used interchangeably with “intrageneric microorganism”.


As used herein, “introduced genetic material” means genetic material that is added to, and remains as a component of, the genome of the recipient.


As used herein, in the context of non-intergeneric microorganisms, the term “remodeled” is used synonymously with the term “engineered”. Consequently, a “non-intergeneric remodeled microorganism” has a synonymous meaning to “non-intergeneric engineered microorganism,” and will be utilized interchangeably. Further, the disclosure may refer to an “engineered strain” or “engineered derivative” or “engineered non-intergeneric microbe,” these terms are used synonymously with “remodeled strain” or “remodeled derivative” or “remodeled non-intergeneric microbe.”


In some embodiments, the nitrogen fixation and assimilation genetic regulatory network comprises polynucleotides encoding genes and non-coding sequences that direct, modulate, and/or regulate microbial nitrogen fixation and/or assimilation and can comprise polynucleotide sequences of the nif cluster (e.g., nifA, nifB, nifC . . . nifZ), polynucleotides encoding nitrogen regulatory protein C, polynucleotides encoding nitrogen regulatory protein B, polynucleotide sequences of the gln cluster (e.g. glnA and glnD), draT, and ammonia transporters/permeases. In some cases, the Nif cluster may comprise NifB, NifH, NifD, NifK, NifE, NifN, NifX, hesa, and NifV. In some cases, the Nif cluster may comprise a subset of NifB, NifH, NifD, NifK, NifE, NifN, NifX, hesa, and NifV.


In some embodiments, fertilizer of the present disclosure comprises at least 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 21%, 22%, 23%, 24%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 41%, 42%, 43%, 44%, 45%, 46%, 47%, 48%, 49%, 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59%, 60%, 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% nitrogen by weight.


In some embodiments, fertilizer of the present disclosure comprises at least about 5%, about 6%, about 7%, about 8%, about 9%, about 10%, about 11%, about 12%, about 13%, about 14%, about 15%, about 16%, about 17%, about 18%, about 19%, about 20%, about 21%, about 22%, about 23%, about 24%, about 25%, about 26%, about 27%, about 28%, about 29%, about 30%, about 31%, about 32%, about 33%, about 34%, about 35%, about 36%, about 37%, about 38%, about 39%, about 40%, about 41%, about 42%, about 43%, about 44%, about 45%, about 46%, about 47%, about 48%, about 49%, about 50%, about 51%, about 52%, about 53%, about 54%, about 55%, about 56%, about 57%, about 58%, about 59%, about 60%, about 61%, about 62%, about 63%, about 64%, about 65%, about 66%, about 67%, about 68%, about 69%, about 70%, about 71%, about 72%, about 73%, about 74%, about 75%, about 76%, about 77%, about 78%, about 79%, about 80%, about 81%, about 82%, about 83%, about 84%, about 85%, about 86%, about 87%, about 88%, about 89%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, or about 99% nitrogen by weight.


In some embodiments, fertilizer of the present disclosure comprises about 5% to 50%, about 5% to 75%, about 10% to 50%, about 10% to 75%, about 15% to 50%, about 15% to 75%, about 20% to 50%, about 20% to 75%, about 25% to 50%, about 25% to 75%, about 30% to 50%, about 30% to 75%, about 35% to 50%, about 35% to 75%, about 40% to 50%, about 40% to 75%, about 45% to 50%, about 45% to 75%, or about 50% to 75% nitrogen by weight.


In some embodiments, the increase of nitrogen fixation and/or the production of 1% or more of the nitrogen in the plant are measured relative to control plants, which have not been exposed to the bacteria of the present disclosure. All increases or decreases in bacteria are measured relative to control bacteria. All increases or decreases in plants are measured relative to control plants.


As used herein, a “constitutive promoter” is a promoter, which is active under most conditions and/or during most development stages. There are several advantages to using constitutive promoters in expression vectors used in biotechnology, such as: high level of production of proteins used to select transgenic cells or organisms; high level of expression of reporter proteins or scorable markers, allowing easy detection and quantification; high level of production of a transcription factor that is part of a regulatory transcription system; production of compounds that requires ubiquitous activity in the organism; and production of compounds that are required during all stages of development. Non-limiting exemplary constitutive promoters include, CaMV 35S promoter, opine promoters, ubiquitin promoter, alcohol dehydrogenase promoter, etc.


As used herein, a “non-constitutive promoter” is a promoter which is active under certain conditions, in certain types of cells, and/or during certain development stages. For example, tissue specific, tissue preferred, cell type specific, cell type preferred, inducible promoters, and promoters under development control are non-constitutive promoters. Examples of promoters under developmental control include promoters that preferentially initiate transcription in certain tissues.


As used herein, “inducible” or “repressible” promoter is a promoter which is under chemical or environmental factors control. Examples of environmental conditions that may affect transcription by inducible promoters include anaerobic conditions, certain chemicals, the presence of light, acidic or basic conditions, etc.


As used herein, a “tissue specific” promoter is a promoter that initiates transcription only in certain tissues. Unlike constitutive expression of genes, tissue-specific expression is the result of several interacting levels of gene regulation. As such, in the art sometimes it is preferable to use promoters from homologous or closely related species to achieve efficient and reliable expression of transgenes in particular tissues. This is one of the main reasons for the large amount of tissue-specific promoters isolated from particular tissues found in both scientific and patent literature.


As used herein, the term “operably linked” refers to the association of nucleic acid sequences on a single nucleic acid fragment so that the function of one is regulated by the other. For example, a promoter is operably linked with a coding sequence when it is capable of regulating the expression of that coding sequence (i.e., that the coding sequence is under the transcriptional control of the promoter). Coding sequences can be operably linked to regulatory sequences in a sense or antisense orientation. In another example, the complementary RNA regions of the disclosure can be operably linked, either directly or indirectly, 5′ to the target mRNA, or 3′ to the target mRNA, or within the target mRNA, or a first complementary region is 5′ and its complement is 3′ to the target mRNA.


In aspects, “applying to the plant a plurality of non-intergeneric bacteria,” includes any means by which the plant (including plant parts such as a seed, root, stem, tissue, etc.) is made to come into contact (i.e. exposed) with said bacteria at any stage of the plant's life cycle. Consequently, “applying to the plant a plurality of non-intergeneric bacteria,” includes any of the following means of exposing the plant (including plant parts such as a seed, root, stem, tissue, etc.) to said bacteria: spraying onto plant, dripping onto plant, applying as a seed coat, applying to a field that will then be planted with seed, applying to a field already planted with seed, applying to a field with adult plants, etc.


As used herein “MRTN” is an acronym for maximum return to nitrogen and is utilized as an experimental treatment in the Examples. MRTN was developed by Iowa State University and information can be found at: http://cnrc.agron.iastate.edu/ The MRTN is the nitrogen rate where the economic net return to nitrogen application is maximized. The approach to calculating the MRTN is a regional approach for developing corn nitrogen rate guidelines in individual states. The nitrogen rate trial data was evaluated for Illinois, Iowa, Michigan, Minnesota, Ohio, and Wisconsin where an adequate number of research trials were available for corn plantings following soybean and corn plantings following corn. The trials were conducted with spring, sidedress, or split preplant/sidedress applied nitrogen, and sites were not irrigated except for those that were indicated for irrigated sands in Wisconsin. MRTN was developed by Iowa State University due to apparent differences in methods for determining suggested nitrogen rates required for corn production, misperceptions pertaining to nitrogen rate guidelines, and concerns about application rates. By calculating the MRTN, practitioners can determine the following: (1) the nitrogen rate where the economic net return to nitrogen application is maximized, (2) the economic optimum nitrogen rate, which is the point where the last increment of nitrogen returns a yield increase large enough to pay for the additional nitrogen, (3) the value of corn grain increase attributed to nitrogen application, and the maximum yield, which is the yield where application of more nitrogen does not result in a corn yield increase. Thus the MRTN calculations provide practitioners with the means to maximize corn crops in different regions while maximizing financial gains from nitrogen applications.


The term mmol is an abbreviation for millimole, which is a thousandth (10−3) of a mole, abbreviated herein as mol.


As used herein the terms “microorganism” or “microbe” should be taken broadly. These terms, used interchangeably, include but are not limited to, the two prokaryotic domains, Bacteria and Archaea. The term may also encompass eukaryotic fungi and protists.


The term “microbial consortia” or “microbial consortium” refers to a subset of a microbial community of individual microbial species, or strains of a species, which can be described as carrying out a common function, or can be described as participating in, or leading to, or correlating with, a recognizable parameter, such as a phenotypic trait of interest.


The term “microbial community” means a group of microbes comprising two or more species or strains. Unlike microbial consortia, a microbial community does not have to be carrying out a common function, or does not have to be participating in, or leading to, or correlating with, a recognizable parameter, such as a phenotypic trait of interest.


As used herein, “isolate,” “isolated,” “isolated microbe,” and like terms, are intended to mean that the one or more microorganisms has been separated from at least one of the materials with which it is associated in a particular environment (for example soil, water, plant tissue, etc.). Thus, an “isolated microbe” does not exist in its naturally occurring environment; rather, it is through the various techniques described herein that the microbe has been removed from its natural setting and placed into a non-naturally occurring state of existence. Thus, the isolated strain or isolated microbe may exist as, for example, a biologically pure culture, or as spores (or other forms of the strain). In aspects, the isolated microbe may be in association with an acceptable carrier, which may be an agriculturally acceptable carrier.


In certain aspects of the disclosure, the isolated microbes exist as “isolated and biologically pure cultures.” It will be appreciated by one of skill in the art that an isolated and biologically pure culture of a particular microbe, denotes that said culture is substantially free of other living organisms and contains only the individual microbe in question. The culture can contain varying concentrations of said microbe. The present disclosure notes that isolated and biologically pure microbes often “necessarily differ from less pure or impure materials.” See, e.g. In re Bergstrom, 427 F.2d 1394, (CCPA 1970)(discussing purified prostaglandins), see also, In re Bergy, 596 F.2d 952 (CCPA 1979)(discussing purified microbes), see also, Parke-Davis & Co. v. HK Mulford & Co., 189 F. 95 (S.D.N.Y. 1911) (Learned Hand discussing purified adrenaline), aff'd in part, rev'd in part, 196 F. 496 (2d Cir. 1912), each of which are incorporated herein by reference. Furthermore, in some aspects, the disclosure provides for certain quantitative measures of the concentration, or purity limitations, that must be found within an isolated and biologically pure microbial culture. The presence of these purity values, in certain embodiments, is a further attribute that distinguishes the presently disclosed microbes from those microbes existing in a natural state. See, e.g., Merck & Co. v. Olin Mathieson Chemical Corp., 253 F.2d 156 (4th Cir. 1958) (discussing purity limitations for vitamin B12 produced by microbes), incorporated herein by reference.


As used herein, “individual isolates” should be taken to mean a composition, or culture, comprising a predominance of a single genera, species, or strain, of microorganism, following separation from one or more other microorganisms.


Microbes of the present disclosure may include spores and/or vegetative cells. In some embodiments, microbes of the present disclosure include microbes in a viable but non-culturable (VBNC) state. As used herein, “spore” or “spores” refer to structures produced by bacteria and fungi that are adapted for survival and dispersal. Spores are generally characterized as dormant structures; however, spores are capable of differentiation through the process of germination. Germination is the differentiation of spores into vegetative cells that are capable of metabolic activity, growth, and reproduction. The germination of a single spore results in a single fungal or bacterial vegetative cell. Fungal spores are units of asexual reproduction, and in some cases are necessary structures in fungal life cycles. Bacterial spores are structures for surviving conditions that may ordinarily be nonconducive to the survival or growth of vegetative cells.


As used herein, “microbial composition” refers to a composition comprising one or more microbes of the present disclosure. In some embodiments, a microbial composition is administered to plants (including various plant parts) and/or in agricultural fields.


As used herein, “carrier,” “acceptable carrier,” or “agriculturally acceptable carrier” refers to a diluent, adjuvant, excipient, or vehicle with which the microbe can be administered, which does not detrimentally effect the microbe.


Regulation of Nitrogen Fixation


In some cases, nitrogen fixation pathway may act as a target for genetic engineering and optimization. One trait that may be targeted for regulation by the methods described herein is nitrogen fixation. Nitrogen fertilizer is the largest operational expense on a farm and the biggest driver of higher yields in row crops like corn and wheat. Described herein are microbial products that can deliver renewable forms of nitrogen in non-leguminous crops. While some endophytes have the genetics necessary for fixing nitrogen in pure culture, the fundamental technical challenge is that wild-type endophytes of cereals and grasses stop fixing nitrogen in fertilized fields. The application of chemical fertilizers and residual nitrogen levels in field soils signal the microbe to shut down the biochemical pathway for nitrogen fixation.


Changes to the transcriptional and post-translational levels of components of the nitrogen fixation regulatory network may be beneficial to the development of a microbe capable of fixing and transferring nitrogen to corn in the presence of fertilizer. To that end, described herein is Host-Microbe Evolution (HoME) technology to precisely evolve regulatory networks and elicit novel phenotypes. Also described herein are unique, proprietary libraries of nitrogen-fixing endophytes isolated from corn, paired with extensive omics data surrounding the interaction of microbes and host plant under different environmental conditions like nitrogen stress and excess. In some embodiments, this technology enables precision evolution of the genetic regulatory network of endophytes to produce microbes that actively fix nitrogen even in the presence of fertilizer in the field. Also described herein are evaluations of the technical potential of evolving microbes that colonize corn root tissues and produce nitrogen for fertilized plants and evaluations of the compatibility of endophytes with standard formulation practices and diverse soils to determine feasibility of integrating the microbes into modern nitrogen management strategies.


In order to utilize elemental nitrogen (N) for chemical synthesis, life forms combine nitrogen gas (N2) available in the atmosphere with hydrogen in a process known as nitrogen fixation. Because of the energy-intensive nature of biological nitrogen fixation, diazotrophs (bacteria and archaea that fix atmospheric nitrogen gas) have evolved sophisticated and tight regulation of the nif gene cluster in response to environmental oxygen and available nitrogen. Nif genes encode enzymes involved in nitrogen fixation (such as the nitrogenase complex) and proteins that regulate nitrogen fixation. Shamseldin (2013. Global J. Biotechnol. Biochem. 8(4):84-94) discloses detailed descriptions of nif genes and their products, and is incorporated herein by reference. Described herein are methods of producing a plant with an improved trait comprising isolating bacteria from a first plant, introducing a genetic variation into a gene of the isolated bacteria to increase nitrogen fixation, exposing a second plant to the variant bacteria, isolating bacteria from the second plant having an improved trait relative to the first plant, and repeating the steps with bacteria isolated from the second plant.


In Proteobacteria, regulation of nitrogen fixation centers around the σ54-dependent enhancer-binding protein NifA, the positive transcriptional regulator of the nif cluster. Intracellular levels of active NifA are controlled by two key factors: transcription of the nifLA operon, and inhibition of NifA activity by protein-protein interaction with NifL. Both of these processes are responsive to intracelluar glutamine levels via the PII protein signaling cascade. This cascade is mediated by GlnD, which directly senses glutamine and catalyzes the uridylylation or deuridylylation of two PII regulatory proteins—GlnB and GlnK—in response the absence or presence, respectively, of bound glutamine. Under conditions of nitrogen excess, unmodified GlnB signals the deactivation of the nifLA promoter. However, under conditions of nitrogen limitation, GlnB is post-translationally modified, which inhibits its activity and leads to transcription of the nifLA operon. In this way, nifLA transcription is tightly controlled in response to environmental nitrogen via the PII protein signaling cascade. On the post-translational level of NifA regulation, GlnK inhibits the NifL/NifA interaction in a matter dependent on the overall level of free GlnK within the cell.


NifA is transcribed from the nifLA operon, whose promoter is activated by phosphorylated NtrC, another σ54-dependent regulator. The phosphorylation state of NtrC is mediated by the histidine kinase NtrB, which interacts with deuridylylated GlnB but not uridylylated GlnB. Under conditions of nitrogen excess, a high intracellular level of glutamine leads to deuridylylation of GlnB, which then interacts with NtrB to deactivate its phosphorylation activity and activate its phosphatase activity, resulting in dephosphorylation of NtrC and the deactivation of the nifLA promoter. However, under conditions of nitrogen limitation, a low level of intracellular glutamine results in uridylylation of GlnB, which inhibits its interaction with NtrB and allows the phosphorylation of NtrC and transcription of the nifLA operon. In this way, nifLA expression is tightly controlled in response to environmental nitrogen via the PII protein signaling cascade. nifA, ntrB, ntrC, and glnB, are all genes that can be mutated in the methods described herein. These processes may also be responsive to intracellular or extracellular levels of ammonia, urea or nitrates.


The activity of NifA is also regulated post-translationally in response to environmental nitrogen, most typically through NifL-mediated inhibition of NifA activity. In general, the interaction of NifL and NifA is influenced by the PII protein signaling cascade via GlnK, although the nature of the interactions between GlnK and NifL/NifA varies significantly between diazotrophs. In Klebsiella pneumoniae, both forms of GlnK inhibit the NifL/NifA interaction, and the interaction between GlnK and NifL/NifA is determined by the overall level of free GlnK within the cell. Under nitrogen-excess conditions, deuridylylated GlnK interacts with the ammonium transporter AmtB, which serves to both block ammonium uptake by AmtB and sequester GlnK to the membrane, allowing inhibition of NifA by NifL. On the other hand, in Azotobacter vinelandii, interaction with deuridylylated GlnK is required for the NifL/NifA interaction and NifA inhibition, while uridylylation of GlnK inhibits its interaction with NifL. In diazotrophs lacking the nifL gene, there is evidence that NifA activity is inhibited directly by interaction with the deuridylylated forms of both GlnK and GlnB under nitrogen-excess conditions. In some bacteria the Nif cluster may be regulated by glnR, and further in some cases this may comprise negative regulation. Regardless of the mechanism, post-translational inhibition of NifA is an important regulator of the nif cluster in most known diazotrophs. Additionally, nifL, amtB, glnK, and glnR are genes that can be mutated in the methods described herein.


In addition to regulating the transcription of the nif gene cluster, many diazotrophs have evolved a mechanism for the direct post-translational modification and inhibition of the nitrogenase enzyme itself, known as nitrogenase shutoff. This is mediated by ADP-ribosylation of the Fe protein (NifH) under nitrogen-excess conditions, which disrupts its interaction with the MoFe protein complex (NifDK) and abolishes nitrogenase activity. DraT catalyzes the ADP-ribosylation of the Fe protein and shutoff of nitrogenase, while DraG catalyzes the removal of ADP-ribose and reactivation of nitrogenase. As with nifLA transcription and NifA inhibition, nitrogenase shutoff is also regulated via the PII protein signaling cascade. Under nitrogen-excess conditions, deuridylylated GlnB interacts with and activates DraT, while deuridylylated GlnK interacts with both DraG and AmtB to form a complex, sequestering DraG to the membrane. Under nitrogen-limiting conditions, the uridylylated forms of GlnB and GlnK do not interact with DraT and DraG, respectively, leading to the inactivation of DraT and the diffusion of DraG to the Fe protein, where it removes the ADP-ribose and activates nitrogenase. The methods described herein also contemplate introducing genetic variation into the nifH, nifD, nifK, and draT genes.


Although some endophytes have the ability to fix nitrogen in vitro, often the genetics are silenced in the field by high levels of exogenous chemical fertilizers. One can decouple the sensing of exogenous nitrogen from expression of the nitrogenase enzyme to facilitate field-based nitrogen fixation. Improving the integral of nitrogenase activity across time further serves to augment the production of nitrogen for utilization by the crop. Specific targets for genetic variation to facilitate field-based nitrogen fixation using the methods described herein include one or more genes selected from the group consisting of nifA, nifL, ntrB, ntrC, glnA, glnB, glnK, draT, amtB, glnD, glnE, nifJ, nifH, nifD, nifK, nifY, nifE, nifN, nifU, nifS, nifV, nifW, nifZ, nifM, nifF, nifB, and nifQ.


An additional target for genetic variation to facilitate field-based nitrogen fixation using the methods described herein is the NifA protein. The NifA protein is typically the activator for expression of nitrogen fixation genes. Increasing the production of NifA (either constitutively or during high ammonia condition) circumvents the native ammonia-sensing pathway. In addition, reducing the production of NifL proteins, a known inhibitor of NifA, also leads to an increased level of freely active NifA. In addition, increasing the transcription level of the nifAL operon (either constitutively or during high ammonia condition) also leads to an overall higher level of NifA proteins. Elevated level of nifAL expression is achieved by altering the promoter itself or by reducing the expression of NtrB (part of ntrB and ntrC signaling cascade that originally would result in the shutoff of nifAL operon during high nitrogen condition). High level of NifA achieved by these or any other methods described herein increases the nitrogen fixation activity of the endophytes.


Another target for genetic variation to facilitate field-based nitrogen fixation using the methods described herein is the GlnD/GlnB/GlnK PII signaling cascade. The intracellular glutamine level is sensed through the GlnD/GlnB/GlnK PII signaling cascade. Active site mutations in GlnD that abolish the uridylyl-removing activity of GlnD disrupt the nitrogen-sensing cascade. In addition, reduction of the GlnB concentration short circuits the glutamine-sensing cascade. These mutations “trick” the cells into perceiving a nitrogen-limited state, thereby increasing the nitrogen fixation level activity. These processes may also be responsive to intracellular or extracellular levels of ammonia, urea or nitrates.


The amtB protein is also a target for genetic variation to facilitate field-based nitrogen fixation using the methods described herein. Ammonia uptake from the environment can be reduced by decreasing the expression level of amtB protein. Without intracellular ammonia, the endophyte is not able to sense the high level of ammonia, preventing the down-regulation of nitrogen fixation genes. Any ammonia that manages to get into the intracellular compartment is converted into glutamine. Intracellular glutamine level is the major currency of nitrogen sensing. Decreasing the intracellular glutamine level prevents the cells from sensing high ammonium levels in the environment. This effect can be achieved by increasing the expression level of glutaminase, an enzyme that converts glutamine into glutamate. In addition, intracellular glutamine can also be reduced by decreasing glutamine synthase (an enzyme that converts ammonia into glutamine). In diazotrophs, fixed ammonia is quickly assimilated into glutamine and glutamate to be used for cellular processes. Disruptions to ammonia assimilation may enable diversion of fixed nitrogen to be exported from the cell as ammonia. The fixed ammonia is predominantly assimilated into glutamine by glutamine synthetase (GS), encoded by glnA, and subsequently into glutamine by glutamine oxoglutarate aminotransferase (GOGAT). In some examples, glnS encodes a glutamine synthetase. GS is regulated post-translationally by GS adenylyl transferase (GlnE), a bi-functional enzyme encoded by glnE that catalyzes both the adenylylation and de-adenylylation of GS through activity of its adenylyl-transferase (AT) and adenylyl-removing (AR) domains, respectively. Under nitrogen limiting conditions, glnA is expressed, and GlnE's AR domain de-adynylylates GS, allowing it to be active. Under conditions of nitrogen excess, glnA expression is turned off, and GlnE's AT domain is activated allosterically by glutamine, causing the adenylylation and deactivation of GS.


Furthermore, the draT gene may also be a target for genetic variation to facilitate field-based nitrogen fixation using the methods described herein. Once nitrogen fixing enzymes are produced by the cell, nitrogenase shut-off represents another level in which cell downregulates fixation activity in high nitrogen condition. This shut-off could be removed by decreasing the expression level of DraT.


Methods for imparting new microbial phenotypes can be performed at the transcriptional, translational, and post-translational levels. The transcriptional level includes changes at the promoter (such as changing sigma factor affinity or binding sites for transcription factors, including deletion of all or a portion of the promoter) or changing transcription terminators and attenuators. The translational level includes changes at the ribosome binding sites and changing mRNA degradation signals. The post-translational level includes mutating an enzyme's active site and changing protein-protein interactions. These changes can be achieved in a multitude of ways. Reduction of expression level (or complete abolishment) can be achieved by swapping the native ribosome binding site (RBS) or promoter with another with lower strength/efficiency. ATG start sites can be swapped to a GTG, TTG, or CTG start codon, which results in reduction in translational activity of the coding region. Complete abolishment of expression can be done by knocking out (deleting) the coding region of a gene. Frameshifting the open reading frame (ORF) likely will result in a premature stop codon along the ORF, thereby creating a non-functional truncated product. Insertion of in-frame stop codons will also similarly create a non-functional truncated product. Addition of a degradation tag at the N or C terminal can also be done to reduce the effective concentration of a particular gene.


Conversely, expression level of the genes described herein can be achieved by using a stronger promoter. To ensure high promoter activity during high nitrogen level condition (or any other condition), a transcription profile of the whole genome in a high nitrogen level condition could be obtained and active promoters with a desired transcription level can be chosen from that dataset to replace the weak promoter. Weak start codons can be swapped out with an ATG start codon for better translation initiation efficiency. Weak ribosomal binding sites (RBS) can also be swapped out with a different RBS with higher translation initiation efficiency. In addition, site specific mutagenesis can also be performed to alter the activity of an enzyme.


Increasing the level of nitrogen fixation that occurs in a plant can lead to a reduction in the amount of chemical fertilizer needed for crop production and reduce greenhouse gas emissions (e.g., nitrous oxide).


Generation of Bacterial Populations


Isolation of Bacteria


Microbes useful in methods and compositions disclosed herein can be obtained by extracting microbes from surfaces or tissues of native plants. Microbes can be obtained by grinding seeds to isolate microbes. Microbes can be obtained by planting seeds in diverse soil samples and recovering microbes from tissues. Additionally, microbes can be obtained by inoculating plants with exogenous microbes and determining which microbes appear in plant tissues. Non-limiting examples of plant tissues may include a seed, seedling, leaf, cutting, plant, bulb, or tuber.


A method of obtaining microbes may be through the isolation of bacteria from soils. Bacteria may be collected from various soil types. In some example, the soil can be characterized by traits such as high or low fertility, levels of moisture, levels of minerals, and various cropping practices. For example, the soil may be involved in a crop rotation where different crops are planted in the same soil in successive planting seasons. The sequential growth of different crops on the same soil may prevent disproportionate depletion of certain minerals. The bacteria can be isolated from the plants growing in the selected soils. The seedling plants can be harvested at 2-6 weeks of growth. For example, at least 400 isolates can be collected in a round of harvest. Soil and plant types reveal the plant phenotype as well as the conditions, which allow for the downstream enrichment of certain phenotypes.


Microbes can be isolated from plant tissues to assess microbial traits. The parameters for processing tissue samples may be varied to isolate different types of associative microbes, such as rhizopheric bacteria, epiphytes, or endophytes. The isolates can be cultured in nitrogen-free media to enrich for bacteria that perform nitrogen fixation. Alternatively, microbes can be obtained from global strain banks.


In planta analytics are performed to assess microbial traits. In some embodiments, the plant tissue can be processed for screening by high throughput processing for DNA and RNA. Additionally, non-invasive measurements can be used to assess plant characteristics, such as colonization. Measurements on wild microbes can be obtained on a plant-by-plant basis. Measurements on wild microbes can also be obtained in the field using medium throughput methods. Measurements can be done successively over time. Model plant system can be used including, but not limited to, Setaria.


Microbes in a plant system can be screened via transcriptional profiling of a microbe in a plant system. Examples of screening through transcriptional profiling are using methods of quantitative polymerase chain reaction (qPCR), molecular barcodes for transcript detection, Next Generation Sequencing, and microbe tagging with fluorescent markers. Impact factors can be measured to assess colonization in the greenhouse including, but not limited to, microbiome, abiotic factors, soil conditions, oxygen, moisture, temperature, inoculum conditions, and root localization. Nitrogen fixation can be assessed in bacteria by measuring 15N gas/fertilizer (dilution) with IRMS or NanoSIMS as described herein NanoSIMS is high-resolution secondary ion mass spectrometry. The NanoSIMS technique is a way to investigate chemical activity from biological samples. The catalysis of reduction of oxidation reactions that drive the metabolism of microorganisms can be investigated at the cellular, subcellular, molecular and elemental level. NanoSIMS can provide high spatial resolution of greater than 0.1 μm. NanoSIMS can detect the use of isotope tracers such as 13C, 15N, and 18O. Therefore, NanoSIMS can be used to the chemical activity nitrogen in the cell.


Automated greenhouses can be used for planta analytics. Plant metrics in response to microbial exposure include, but are not limited to, biomass, chloroplast analysis, CCD camera, volumetric tomography measurements.


One way of enriching a microbe population is according to genotype. For example, a polymerase chain reaction (PCR) assay with a targeted primer or specific primer. Primers designed for the nifH gene can be used to identity diazotrophs because diazotrophs express the nifH gene in the process of nitrogen fixation. A microbial population can also be enriched via single-cell culture-independent approaches and chemotaxis-guided isolation approaches. Alternatively, targeted isolation of microbes can be performed by culturing the microbes on selection media. Premeditated approaches to enriching microbial populations for desired traits can be guided by bioinformatics data and are described herein.


Enriching for Microbes with Nitrogen Fixation Capabilities Using Bioinformatics


Bioinformatic tools can be used to identify and isolate plant growth promoting rhizobacteria (PGPRs), which are selected based on their ability to perform nitrogen fixation. Microbes with high nitrogen fixing ability can promote favorable traits in plants. Bioinformatic modes of analysis for the identification of PGPRs include, but are not limited to, genomics, metagenomics, targeted isolation, gene sequencing, transcriptome sequencing, and modeling.


Genomics analysis can be used to identify PGPRs and confirm the presence of mutations with methods of Next Generation Sequencing as described herein and microbe version control.


Metagenomics can be used to identify and isolate PGPR using a prediction algorithm for colonization. Metadata can also be used to identify the presence of an engineered strain in environmental and greenhouse samples.


Transcriptomic sequencing can be used to predict genotypes leading to PGPR phenotypes. Additionally, transcriptomic data is used to identify promoters for altering gene expression. Transcriptomic data can be analyzed in conjunction with the Whole Genome Sequence (WGS) to generate models of metabolism and gene regulatory networks.


Domestication of Microbes


Microbes isolated from nature can undergo a domestication process wherein the microbes are converted to a form that is genetically trackable and identifiable. One way to domesticate a microbe is to engineer it with antibiotic resistance. The process of engineering antibiotic resistance can begin by determining the antibiotic sensitivity in the wild type microbial strain. If the bacteria are sensitive to the antibiotic, then the antibiotic can be a good candidate for antibiotic resistance engineering. Subsequently, an antibiotic resistant gene or a counterselectable suicide vector can be incorporated into the genome of a microbe using recombineering methods. A counterselectable suicide vector may consist of a deletion of the gene of interest, a selectable marker, and the counterselectable marker sacB. Counterselection can be used to exchange native microbial DNA sequences with antibiotic resistant genes. A medium throughput method can be used to evaluate multiple microbes simultaneously allowing for parallel domestication. Alternative methods of domestication include the use of homing nucleases to prevent the suicide vector sequences from looping out or from obtaining intervening vector sequences.


DNA vectors can be introduced into bacteria via several methods including electroporation and chemical transformations. A standard library of vectors can be used for transformations. An example of a method of gene editing is CRISPR preceded by Cas9 testing to ensure activity of Cas9 in the microbes.


Non-Transgenic Engineering of Microbes


A microbial population with favorable traits can be obtained via directed evolution. Direct evolution is an approach wherein the process of natural selection is mimicked to evolve proteins or nucleic acids towards a user-defined goal. An example of direct evolution is when random mutations are introduced into a microbial population, the microbes with the most favorable traits are selected, and the growth of the selected microbes is continued. The most favorable traits in growth promoting rhizobacteria (PGPRs) may be in nitrogen fixation. The method of directed evolution may be iterative and adaptive based on the selection process after each iteration.


Plant growth promoting rhizobacteria (PGPRs) with high capability of nitrogen fixation can be generated. The evolution of PGPRs can be carried out via the introduction of genetic variation. Genetic variation can be introduced via polymerase chain reaction mutagenesis, oligonucleotide-directed mutagenesis, saturation mutagenesis, fragment shuffling mutagenesis, homologous recombination, CRISPR/Cas9 systems, chemical mutagenesis, and combinations thereof. These approaches can introduce random mutations into the microbial population. For example, mutants can be generated using synthetic DNA or RNA via oligonucleotide-directed mutagenesis. Mutants can be generated using tools contained on plasmids, which are later cured. Genes of interest can be identified using libraries from other species with improved traits including, but not limited to, improved PGPR properties, improved colonization of cereals, increased oxygen sensitivity, increased nitrogen fixation, and increased ammonia excretion. Intrageneric genes can be designed based on these libraries using software such as Geneious or Platypus design software. Mutations can be designed with the aid of machine learning. Mutations can be designed with the aid of a metabolic model. Automated design of the mutation can be done using a la Platypus and will guide RNAs for Cas-directed mutagenesis.


The intra-generic genes can be transferred into the host microbe. Additionally, reporter systems can also be transferred to the microbe. The reporter systems characterize promoters, determine the transformation success, screen mutants, and act as negative screening tools.


The microbes carrying the mutation can be cultured via serial passaging. A microbial colony contains a single variant of the microbe. Microbial colonies are screened with the aid of an automated colony picker and liquid handler. Mutants with gene duplication and increased copy number express a higher genotype of the desired trait.


Selection of Plant Growth Promoting Microbess Based on Nitrogen Fixation


The microbial colonies can be screened using various assays to assess nitrogen fixation. One way to measure nitrogen fixation is via a single fermentative assay, which measures nitrogen excretion. An alternative method is the acetylene reduction assay (ARA) with in-line sampling over time. ARA can be performed in high throughput plates of microtube arrays. ARA can be performed with live plants and plant tissues. The media formulation and media oxygen concentration can be varied in ARA assays. Another method of screening microbial variants is by using biosensors. The use of NanoSIMS and Raman microspectroscopy can be used to investigate the activity of the microbes. In some cases, bacteria can also be cultured and expanded using methods of fermentation in bioreactors. The bioreactors are designed to improve robustness of bacteria growth and to decrease the sensitivity of bacteria to oxygen. Medium to high TP plate-based microfermentors are used to evaluate oxygen sensitivity, nutritional needs, nitrogen fixation, and nitrogen excretion. The bacteria can also be co-cultured with competitive or beneficial microbes to elucidate cryptic pathways. Flow cytometry can be used to screen for bacteria that produce high levels of nitrogen using chemical, colorimetric, or fluorescent indicators. The bacteria may be cultured in the presence or absence of a nitrogen source. For example, the bacteria may be cultured with glutamine, ammonia, urea or nitrates.


Guided Microbial Remodeling—an Overview


Guided microbial remodeling is a method to systematically identify and improve the role of species within the crop microbiome. In some aspects, and according to a particular methodology of grouping/categorization, the method comprises three steps: 1) selection of candidate species by mapping plant-microbe interactions and predicting regulatory networks linked to a particular phenotype, 2) pragmatic and predictable improvement of microbial phenotypes through intra-species crossing of regulatory networks and gene clusters within a microbe's genome, and 3) screening and selection of new microbial genotypes that produce desired crop phenotypes.


To systematically assess the improvement of strains, a model is created that links colonization dynamics of the microbial community to genetic activity by key species. The model is used to predict genetic targets for non-intergeneric genetic remodeling (i.e. engineering the genetic architecture of the microbe in a non-transgentic fashion). See, FIG. 1A for a graphical representation of an embodiment of the process.


As illustrated in FIG. 1A, rational improvement of the crop microbiome may be used to increase soil biodiversity, tune impact of keystone species, and/or alter timing and expression of important metabolic pathways.


To this end, the inventors have developed a platform to identify and improve the role of strains within the crop microbiome. In some aspects, the inventors call this process microbial breeding.


The aforementioned “Guided Microbial Remodeling” process will be further elaborated upon in the Examples, for instance in Example 1, entitled: “Guided Microbial Remodeling—A Platform for the Rational Improvement of Microbial Species for Agriculture.”


Serial Passage


Production of bacteria to improve plant traits (e.g., nitrogen fixation) can be achieved through serial passage. The production of this bacteria can be done by selecting plants, which have a particular improved trait that is influenced by the microbial flora, in addition to identifying bacteria and/or compositions that are capable of imparting one or more improved traits to one or more plants. One method of producing a bacteria to improve a plant trait includes the steps of: (a) isolating bacteria from tissue or soil of a first plant; (b) introducing a genetic variation into one or more of the bacteria to produce one or more variant bacteria; (c) exposing a plurality of plants to the variant bacteria; (d) isolating bacteria from tissue or soil of one of the plurality of plants, wherein the plant from which the bacteria is isolated has an improved trait relative to other plants in the plurality of plants; and (e) repeating steps (b) to (d) with bacteria isolated from the plant with an improved trait (step (d)). Steps (b) to (d) can be repeated any number of times (e.g., once, twice, three times, four times, five times, ten times, or more) until the improved trait in a plant reaches a desired level. Further, the plurality of plants can be more than two plants, such as 10 to 20 plants, or 20 or more, 50 or more, 100 or more, 300 or more, 500 or more, or 1000 or more plants.


In addition to obtaining a plant with an improved trait, a bacterial population comprising bacteria comprising one or more genetic variations introduced into one or more genes (e.g., genes regulating nitrogen fixation) is obtained. By repeating the steps described above, a population of bacteria can be obtained that include the most appropriate members of the population that correlate with a plant trait of interest. The bacteria in this population can be identified and their beneficial properties determined, such as by genetic and/or phenotypic analysis. Genetic analysis may occur of isolated bacteria in step (a). Phenotypic and/or genotypic information may be obtained using techniques including: high through-put screening of chemical components of plant origin, sequencing techniques including high throughput sequencing of genetic material, differential display techniques (including DDRT-PCR, and DD-PCR), nucleic acid microarray techniques, RNA-sequencing (Whole Transcriptome Shotgun Sequencing), and qRT-PCR (quantitative real time PCR). Information gained can be used to obtain community profiling information on the identity and activity of bacteria present, such as phylogenetic analysis or microarray-based screening of nucleic acids coding for components of rRNA operons or other taxonomically informative loci. Examples of taxonomically informative loci include 16S rRNA gene, 23S rRNA gene, 5S rRNA gene, 5.8S rRNA gene, 12S rRNA gene, 18S rRNA gene, 28S rRNA gene, gyrB gene, rpoB gene, fusA gene, recA gene, cox1 gene, nifD gene. Example processes of taxonomic profiling to determine taxa present in a population are described in US20140155283. Bacterial identification may comprise characterizing activity of one or more genes or one or more signaling pathways, such as genes associated with the nitrogen fixation pathway. Synergistic interactions (where two components, by virtue of their combination, increase a desired effect by more than an additive amount) between different bacterial species may also be present in the bacterial populations.


Genetic Variation—Locations and Sources of Genomic Alteration


The genetic variation may be a gene selected from the group consisting of: nifA, nifL, ntrB, ntrC, glnA, glnB, glnK, draT, amtB, glnD, glnE, nifJ, nifH, nifD, nifK, nifY, nifE, nifN, nifU, nifS, nifV, nifW, nifZ, nifM, nifF, nifB, and nifQ. The genetic variation may be a variation in a gene encoding a protein with functionality selected from the group consisting of: glutamine synthetase, glutaminase, glutamine synthetase adenylyltransferase, transcriptional activator, anti-transcriptional activator, pyruvate flavodoxin oxidoreductase, flavodoxin, or NAD+-dinitrogen-reductase aDP-D-ribosyltransferase. The genetic variation may be a mutation that results in one or more of: increased expression or activity of NifA or glutaminase; decreased expression or activity of NifL, NtrB, glutamine synthetase, GlnB, GlnK, DraT, AmtB; decreased adenylyl-removing activity of GlnE; or decreased uridylyl-removing activity of GlnD. Introducing a genetic variation may comprise insertion and/or deletion of one or more nucleotides at a target site, such as 1, 2, 3, 4, 5, 10, 25, 50, 100, 250, 500, or more nucleotides. The genetic variation introduced into one or more bacteria of the methods disclosed herein may be a knock-out mutation (e.g. deletion of a promoter, insertion or deletion to produce a premature stop codon, deletion of an entire gene), or it may be elimination or abolishment of activity of a protein domain (e.g. point mutation affecting an active site, or deletion of a portion of a gene encoding the relevant portion of the protein product), or it may alter or abolish a regulatory sequence of a target gene. One or more regulatory sequences may also be inserted, including heterologous regulatory sequences and regulatory sequences found within a genome of a bacterial species or genus corresponding to the bacteria into which the genetic variation is introduced. Moreover, regulatory sequences may be selected based on the expression level of a gene in a bacterial culture or within a plant tissue. The genetic variation may be a pre-determined genetic variation that is specifically introduced to a target site. The genetic variation may be a random mutation within the target site. The genetic variation may be an insertion or deletion of one or more nucleotides. In some cases, a plurality of different genetic variations (e.g. 2, 3, 4, 5, 10, or more) are introduced into one or more of the isolated bacteria before exposing the bacteria to plants for assessing trait improvement. The plurality of genetic variations can be any of the above types, the same or different types, and in any combination. In some cases, a plurality of different genetic variations are introduced serially, introducing a first genetic variation after a first isolation step, a second genetic variation after a second isolation step, and so forth so as to accumulate a plurality of genetic variations in bacteria imparting progressively improved traits on the associated plants.


Genetic Variation—Methods of Introducing Genomic Alteration


In general, the term “genetic variation” refers to any change introduced into a polynucleotide sequence relative to a reference polynucleotide, such as a reference genome or portion thereof, or reference gene or portion thereof. A genetic variation may be referred to as a “mutation,” and a sequence or organism comprising a genetic variation may be referred to as a “genetic variant” or “mutant”. Genetic variations can have any number of effects, such as the increase or decrease of some biological activity, including gene expression, metabolism, and cell signaling. Genetic variations can be specifically introduced to a target site, or introduced randomly. A variety of molecular tools and methods are available for introducing genetic variation. For example, genetic variation can be introduced via polymerase chain reaction mutagenesis, oligonucleotide-directed mutagenesis, saturation mutagenesis, fragment shuffling mutagenesis, homologous recombination, recombineering, lambda red mediated recombination, CRISPR/Cas9 systems, chemical mutagenesis, and combinations thereof. Chemical methods of introducing genetic variation include exposure of DNA to a chemical mutagen, e.g., ethyl methanesulfonate (EMS), methyl methanesulfonate (MMS), N-nitrosourea (EN U), N-methyl-N-nitro-N′-nitrosoguanidine, 4-nitroquinoline N-oxide, diethylsulfate, benzopyrene, cyclophosphamide, bleomycin, triethylmelamine, acrylamide monomer, nitrogen mustard, vincristine, diepoxyalkanes (for example, diepoxybutane), ICR-170, formaldehyde, procarbazine hydrochloride, ethylene oxide, dimethylnitrosamine, 7,12 dimethylbenz(a)anthracene, chlorambucil, hexamethylphosphoramide, bisulfan, and the like. Radiation mutation-inducing agents include ultraviolet radiation, γ-irradiation, X-rays, and fast neutron bombardment. Genetic variation can also be introduced into a nucleic acid using, e.g., trimethylpsoralen with ultraviolet light. Random or targeted insertion of a mobile DNA element, e.g., a transposable element, is another suitable method for generating genetic variation. Genetic variations can be introduced into a nucleic acid during amplification in a cell-free in vitro system, e.g., using a polymerase chain reaction (PCR) technique such as error-prone PCR. Genetic variations can be introduced into a nucleic acid in vitro using DNA shuffling techniques (e.g., exon shuffling, domain swapping, and the like). Genetic variations can also be introduced into a nucleic acid as a result of a deficiency in a DNA repair enzyme in a cell, e.g., the presence in a cell of a mutant gene encoding a mutant DNA repair enzyme is expected to generate a high frequency of mutations (i.e., about 1 mutation/100 genes-1 mutation/10,000 genes) in the genome of the cell. Examples of genes encoding DNA repair enzymes include but are not limited to Mut H, Mut S, Mut L, and Mut U, and the homologs thereof in other species (e.g., MSH 1 6, PMS 1 2, MLH 1, GTBP, ERCC-1, and the like). Example descriptions of various methods for introducing genetic variations are provided in e.g., Stemple (2004) Nature 5:1-7; Chiang et al. (1993) PCR Methods Appl 2(3): 210-217; Stemmer (1994) Proc. Natl. Acad. Sci. USA 91:10747-10751; and U.S. Pat. Nos. 6,033,861, and 6,773,900.


Genetic variations introduced into microbes may be classified as transgenic, cisgenic, intragenomic, intrageneric, intergeneric, synthetic, evolved, rearranged, or SNPs.


Genetic variation may be introduced into numerous metabolic pathways within microbes to elicit improvements in the traits described above. Representative pathways include sulfur uptake pathways, glycogen biosynthesis, the glutamine regulation pathway, the molybdenum uptake pathway, the nitrogen fixation pathway, ammonia assimilation, ammonia excretion or secretion, nNitrogen uptake, glutamine biosynthesis, annamox, phosphate solubilization, organic acid transport, organic acid production, agglutinins production, reactive oxygen radical scavenging genes, Indole Acetic Acid biosynthesis, trehalose biosynthesis, plant cell wall degrading enzymes or pathways, root attachment genes, exopolysaccharide secretion, glutamate synthase pathway, iron uptake pathways, siderophore pathway, chitinase pathway, ACC deaminase, glutathione biosynthesis, phosphorous signalig genes, quorum quenching pathway, cytochrome pathways, hemoglobin pathway, bacterial hemoglobin-like pathway, small RNA rsmZ, rhizobitoxine biosynthesis, lapA adhesion protein, AHL quorum sensing pathway, phenazine biosynthesis, cyclic lipopeptide biosynthesis, and antibiotic production.


CRISPR/Cas9 (Clustered regularly interspaced short palindromic repeats)/CRISPR-associated (Cas) systems can be used to introduce desired mutations. CRISPR/Cas9 provide bacteria and archaea with adaptive immunity against viruses and plasmids by using CRISPR RNAs (crRNAs) to guide the silencing of invading nucleic acids. The Cas9 protein (or functional equivalent and/or variant thereof, i.e., Cas9-like protein) naturally contains DNA endonuclease activity that depends on the association of the protein with two naturally occurring or synthetic RNA molecules called crRNA and tracrRNA (also called guide RNAs). In some cases, the two molecules are covalently link to form a single molecule (also called a single guide RNA (“sgRNA”). Thus, the Cas9 or Cas9-like protein associates with a DNA-targeting RNA (which term encompasses both the two-molecule guide RNA configuration and the single-molecule guide RNA configuration), which activates the Cas9 or Cas9-like protein and guides the protein to a target nucleic acid sequence. If the Cas9 or Cas9-like protein retains its natural enzymatic function, it will cleave target DNA to create a double-stranded break, which can lead to genome alteration (i.e., editing: deletion, insertion (when a donor polynucleotide is present), replacement, etc.), thereby altering gene expression. Some variants of Cas9 (which variants are encompassed by the term Cas9-like) have been altered such that they have a decreased DNA cleaving activity (in some cases, they cleave a single strand instead of both strands of the target DNA, while in other cases, they have severely reduced to no DNA cleavage activity). Further exemplary descriptions of CRISPR systems for introducing genetic variation can be found in, e.g. U.S. Pat. No. 8,795,965.


As a cyclic amplification technique, polymerase chain reaction (PCR) mutagenesis uses mutagenic primers to introduce desired mutations. PCR is performed by cycles of denaturation, annealing, and extension. After amplification by PCR, selection of mutated DNA and removal of parental plasmid DNA can be accomplished by: 1) replacement of dCTP by hydroxymethylated-dCTP during PCR, followed by digestion with restriction enzymes to remove non-hydroxymethylated parent DNA only; 2) simultaneous mutagenesis of both an antibiotic resistance gene and the studied gene changing the plasmid to a different antibiotic resistance, the new antibiotic resistance facilitating the selection of the desired mutation thereafter; 3) after introducing a desired mutation, digestion of the parent methylated template DNA by restriction enzyme Dpn1 which cleaves only methylated DNA, by which the mutagenized unmethylated chains are recovered; or 4) circularization of the mutated PCR products in an additional ligation reaction to increase the transformation efficiency of mutated DNA. Further description of exemplary methods can be found in e.g. U.S. Pat. Nos. 7,132,265, 6,713,285, 6,673,610, 6,391,548, 5,789,166, 5,780,270, 5,354,670, 5,071,743, and US20100267147.


Oligonucleotide-directed mutagenesis, also called site-directed mutagenesis, typically utilizes a synthetic DNA primer. This synthetic primer contains the desired mutation and is complementary to the template DNA around the mutation site so that it can hybridize with the DNA in the gene of interest. The mutation may be a single base change (a point mutation), multiple base changes, deletion, or insertion, or a combination of these. The single-strand primer is then extended using a DNA polymerase, which copies the rest of the gene. The gene thus copied contains the mutated site, and may then be introduced into a host cell as a vector and cloned. Finally, mutants can be selected by DNA sequencing to check that they contain the desired mutation.


Genetic variations can be introduced using error-prone PCR. In this technique the gene of interest is amplified using a DNA polymerase under conditions that are deficient in the fidelity of replication of sequence. The result is that the amplification products contain at least one error in the sequence. When a gene is amplified and the resulting product(s) of the reaction contain one or more alterations in sequence when compared to the template molecule, the resulting products are mutagenized as compared to the template. Another means of introducing random mutations is exposing cells to a chemical mutagen, such as nitrosoguanidine or ethyl methanesulfonate (Nestmann, Mutat Res 1975 June; 28(3):323-30), and the vector containing the gene is then isolated from the host.


Saturation mutagenesis is another form of random mutagenesis, in which one tries to generate all or nearly all possible mutations at a specific site, or narrow region of a gene. In a general sense, saturation mutagenesis is comprised of mutagenizing a complete set of mutagenic cassettes (wherein each cassette is, for example, 1-500 bases in length) in defined polynucleotide sequence to be mutagenized (wherein the sequence to be mutagenized is, for example, from 15 to 100,000 bases in length). Therefore, a group of mutations (e.g. ranging from 1 to 100 mutations) is introduced into each cassette to be mutagenized. A grouping of mutations to be introduced into one cassette can be different or the same from a second grouping of mutations to be introduced into a second cassette during the application of one round of saturation mutagenesis. Such groupings are exemplified by deletions, additions, groupings of particular codons, and groupings of particular nucleotide cassettes.


Fragment shuffling mutagenesis, also called DNA shuffling, is a way to rapidly propagate beneficial mutations. In an example of a shuffling process, DNAse is used to fragment a set of parent genes into pieces of e.g. about 50-100 bp in length. This is then followed by a polymerase chain reaction (PCR) without primers—DNA fragments with sufficient overlapping homologous sequence will anneal to each other and are then be extended by DNA polymerase. Several rounds of this PCR extension are allowed to occur, after some of the DNA molecules reach the size of the parental genes. These genes can then be amplified with another PCR, this time with the addition of primers that are designed to complement the ends of the strands. The primers may have additional sequences added to their 5′ ends, such as sequences for restriction enzyme recognition sites needed for ligation into a cloning vector. Further examples of shuffling techniques are provided in US20050266541.


Homologous recombination mutagenesis involves recombination between an exogenous DNA fragment and the targeted polynucleotide sequence. After a double-stranded break occurs, sections of DNA around the 5′ ends of the break are cut away in a process called resection. In the strand invasion step that follows, an overhanging 3′ end of the broken DNA molecule then “invades” a similar or identical DNA molecule that is not broken. The method can be used to delete a gene, remove exons, add a gene, and introduce point mutations. Homologous recombination mutagenesis can be permanent or conditional. Typically, a recombination template is also provided. A recombination template may be a component of another vector, contained in a separate vector, or provided as a separate polynucleotide. In some embodiments, a recombination template is designed to serve as a template in homologous recombination, such as within or near a target sequence nicked or cleaved by a site-specific nuclease. A template polynucleotide may be of any suitable length, such as about or more than about 10, 15, 20, 25, 50, 75, 100, 150, 200, 500, 1000, or more nucleotides in length. In some embodiments, the template polynucleotide is complementary to a portion of a polynucleotide comprising the target sequence. When optimally aligned, a template polynucleotide might overlap with one or more nucleotides of a target sequences (e.g. about or more than about 1, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100 or more nucleotides). In some embodiments, when a template sequence and a polynucleotide comprising a target sequence are optimally aligned, the nearest nucleotide of the template polynucleotide is within about 1, 5, 10, 15, 20, 25, 50, 75, 100, 200, 300, 400, 500, 1000, 5000, 10000, or more nucleotides from the target sequence. Non-limiting examples of site-directed nucleases useful in methods of homologous recombination include zinc finger nucleases, CRISPR nucleases, TALE nucleases, and meganuclease. For a further description of the use of such nucleases, see e.g. U.S. Pat. No. 8,795,965 and US20140301990.


Mutagens that create primarily point mutations and short deletions, insertions, transversions, and/or transitions, including chemical mutagens or radiation, may be used to create genetic variations. Mutagens include, but are not limited to, ethyl methanesulfonate, methylmethane sulfonate, N-ethyl-N-nitrosurea, triethylmelamine, N-methyl-N-nitrosourea, procarbazine, chlorambucil, cyclophosphamide, diethyl sulfate, acrylamide monomer, melphalan, nitrogen mustard, vincristine, dimethylnitrosamine, N-methyl-N′-nitro-Nitrosoguanidine, nitrosoguanidine, 2-aminopurine, 7,12 dimethyl-benz(a)anthracene, ethylene oxide, hexamethylphosphoramide, bisulfan, diepoxyalkanes (diepoxyoctane, diepoxybutane, and the like), 2-methoxy-6-chloro-9[3-(ethyl-2-chloro-ethyl)aminopropylamino]acridine dihydrochloride and formaldehyde.


Introducing genetic variation may be an incomplete process, such that some bacteria in a treated population of bacteria carry a desired mutation while others do not. In some cases, it is desirable to apply a selection pressure so as to enrich for bacteria carrying a desired genetic variation. Traditionally, selection for successful genetic variants involved selection for or against some functionality imparted or abolished by the genetic variation, such as in the case of inserting antibiotic resistance gene or abolishing a metabolic activity capable of converting a non-lethal compound into a lethal metabolite. It is also possible to apply a selection pressure based on a polynucleotide sequence itself, such that only a desired genetic variation need be introduced (e.g. without also requiring a selectable marker). In this case, the selection pressure can comprise cleaving genomes lacking the genetic variation introduced to a target site, such that selection is effectively directed against the reference sequence into which the genetic variation is sought to be introduced. Typically, cleavage occurs within 100 nucleotides of the target site (e.g. within 75, 50, 25, 10, or fewer nucleotides from the target site, including cleavage at or within the target site). Cleaving may be directed by a site-specific nuclease selected from the group consisting of a Zinc Finger nuclease, a CRISPR nuclease, a TALE nuclease (TALEN), or a meganuclease. Such a process is similar to processes for enhancing homologous recombination at a target site, except that no template for homologous recombination is provided. As a result, bacteria lacking the desired genetic variation are more likely to undergo cleavage that left unrepaired, results in cell death. Bacteria surviving selection may then be isolated for use in exposing to plants for assessing conferral of an improved trait.


A CRISPR nuclease may be used as the site-specific nuclease to direct cleavage to a target site. An improved selection of mutated microbes can be obtained by using Cas9 to kill non-mutated cells. Plants are then inoculated with the mutated microbes to re-confirm symbiosis and create evolutionary pressure to select for efficient symbionts. Microbes can then be re-isolated from plant tissues. CRISPR nuclease systems employed for selection against non-variants can employ similar elements to those described above with respect to introducing genetic variation, except that no template for homologous recombination is provided. Cleavage directed to the target site thus enhances death of affected cells.


Other options for specifically inducing cleavage at a target site are available, such as zinc finger nucleases, TALE nuclease (TALEN) systems, and meganuclease. Zinc-finger nucleases (ZFNs) are artificial DNA endonucleases generated by fusing a zinc finger DNA binding domain to a DNA cleavage domain. ZFNs can be engineered to target desired DNA sequences and this enables zinc-finger nucleases to cleave unique target sequences. When introduced into a cell, ZFNs can be used to edit target DNA in the cell (e.g., the cell's genome) by inducing double stranded breaks. Transcription activator-like effector nucleases (TALENs) are artificial DNA endonucleases generated by fusing a TAL (Transcription activator-like) effector DNA binding domain to a DNA cleavage domain. TALENS can be quickly engineered to bind practically any desired DNA sequence and when introduced into a cell, TALENs can be used to edit target DNA in the cell (e.g., the cell's genome) by inducing double strand breaks. Meganucleases (homing endonuclease) are endodeoxyribonucleases characterized by a large recognition site (double-stranded DNA sequences of 12 to 40 base pairs. Meganucleases can be used to replace, eliminate or modify sequences in a highly targeted way. By modifying their recognition sequence through protein engineering, the targeted sequence can be changed. Meganucleases can be used to modify all genome types, whether bacterial, plant or animal and are commonly grouped into four families: the LAGLIDADG family (SEQ ID NO: 1), the GIY-YIG family, the His-Cyst box family and the HNH family. Exemplary homing endonucleases include I-SceI, I-CeuI, PI-PspI, PI-Sce, I-SceIV, I-CsmI, I-PanI, I-SceII, I-PpoI, I-SceIII, I-CreI, I-TevI, I-TevII and I-TevIII.


Genetic Variation—Methods of Identification


The microbes of the present disclosure may be identified by one or more genetic modifications or alterations, which have been introduced into said microbe. One method by which said genetic modification or alteration can be identified is via reference to a SEQ ID NO that contains a portion of the microbe's genomic sequence that is sufficient to identify the genetic modification or alteration.


Further, in the case of microbes that have not had a genetic modification or alteration (e.g. a wild type, WT) introduced into their genomes, the disclosure can utilize 16S nucleic acid sequences to identify said microbes. A 16S nucleic acid sequence is an example of a “molecular marker” or “genetic marker,” which refers to an indicator that is used in methods for visualizing differences in characteristics of nucleic acid sequences. Examples of other such indicators are restriction fragment length polymorphism (RFLP) markers, amplified fragment length polymorphism (AFLP) markers, single nucleotide polymorphisms (SNPs), insertion mutations, microsatellite markers (SSRs), sequence-characterized amplified regions (SCARs), cleaved amplified polymorphic sequence (CAPS) markers or isozyme markers or combinations of the markers described herein which defines a specific genetic and chromosomal location. Markers further include polynucleotide sequences encoding 16S or 18S rRNA, and internal transcribed spacer (ITS) sequences, which are sequences found between small-subunit and large-subunit rRNA genes that have proven to be especially useful in elucidating relationships or distinctions when compared against one another. Furthermore, the disclosure utilizes unique sequences found in genes of interest (e.g. nifH,D,K,L,A, glnE, amtB, etc.) to identify microbes disclosed herein.


The primary structure of major rRNA subunit 16S comprise a particular combination of conserved, variable, and hypervariable regions that evolve at different rates and enable the resolution of both very ancient lineages such as domains, and more modern lineages such as genera. The secondary structure of the 16S subunit include approximately 50 helices which result in base pairing of about 67% of the residues. These highly conserved secondary structural features are of great functional importance and can be used to ensure positional homology in multiple sequence alignments and phylogenetic analysis. Over the previous few decades, the 16S rRNA gene has become the most sequenced taxonomic marker and is the cornerstone for the current systematic classification of bacteria and archaea (Yarza et al. 2014. Nature Rev. Micro. 12:635-45).


Thus, in certain aspects, the disclosure provides for a sequence, which shares at least about 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to any sequence in Tables 23, 24, 30, 31, and 32.


Thus, in certain aspects, the disclosure provides for a microbe that comprises a sequence, which shares at least about 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NOs: 62-303. These sequences and their associated descriptions can be found in Tables 31 and 32.


In some aspects, the disclosure provides for a microbe that comprises a 16S nucleic acid sequence, which shares at least about 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NOs: 85, 96, 111, 121, 122, 123, 124, 136, 149, 157, 167, 261, 262, 269, 277-283. These sequences and their associated descriptions can be found in Table 32.


In some aspects, the disclosure provides for a microbe that comprises a nucleic acid sequence, which shares at least about 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NOs: 86-95, 97-110, 112-120, 125-135, 137-148, 150-156, 158-166, 168-176, 263-268, 270-274, 275, 276, 284-295. These sequences and their associated descriptions can be found in Table 32.


In some aspects, the disclosure provides for a microbe that comprises a nucleic acid sequence, which shares at least about 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NOs: 177-260, 296-303. These sequences and their associated descriptions can be found in Table 32.


In some aspects, the disclosure provides for a microbe that comprises, or primer that comprises, or probe that comprises, or non-native junction sequence that comprises, a nucleic acid sequence, which shares at least about 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NOs: 304-424. These sequences and their associated descriptions can be found in Table 30.


In some aspects, the disclosure provides for a microbe that comprises a non-native junction sequence that shares at least about 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NOs: 372-405. These sequences and their associated descriptions can be found in Table 30.


In some aspects, the disclosure provides for a microbe that comprises an amino acid sequence, which shares at least about 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NOs: 77, 78, 81, 82, or 83. These sequences and their associated descriptions can be found in Table 31.


Genetic Variation—Methods of Detection: Primers, Probes, and Assays


The present disclosure teaches primers, probes, and assays that are useful for detecting the microbes taught herein. In some aspects, the disclosure provides for methods of detecting the WT parental strains. In other aspects, the disclosure provides for methods of detecting the non-intergeneric engineered microbes derived from the WT strains. In aspects, the present disclosure provides methods of identifying non-intergeneric genetic alterations in a microbe.


In aspects, the genomic engineering methods of the present disclosure lead to the creation of non-natural nucleotide “junction” sequences in the derived non-intergeneric microbes. These non-naturally occurring nucleotide junctions can be used as a type of diagnostic that is indicative of the presence of a particular genetic alteration in a microbe taught herein.


The present techniques are able to detect these non-naturally occurring nucleotide junctions via the utilization of specialized quantitative PCR methods, including uniquely designed primers and probes. In some aspects, the probes of the disclosure bind to the non-naturally occurring nucleotide junction sequences. In some aspects, traditional PCR is utilized. In other aspects, real-time PCR is utilized. In some aspects, quantitative PCR (qPCR) is utilized.


Thus, the disclosure can cover the utilization of two common methods for the detection of PCR products in real-time: (1) non-specific fluorescent dyes that intercalate with any double-stranded DNA, and (2) sequence-specific DNA probes consisting of oligonucleotides that are labelled with a fluorescent reporter which permits detection only after hybridization of the probe with its complementary sequence. In some aspects, only the non-naturally occurring nucleotide junction will be amplified via the taught primers, and consequently can be detected via either a non-specific dye, or via the utilization of a specific hybridization probe. In other aspects, the primers of the disclosure are chosen such that the primers flank either side of a junction sequence, such that if an amplification reaction occurs, then said junction sequence is present.


Aspects of the disclosure involve non-naturally occurring nucleotide junction sequence molecules per se, along with other nucleotide molecules that are capable of binding to said non-naturally occurring nucleotide junction sequences under mild to stringent hybridization conditions. In some aspects, the nucleotide molecules that are capable of binding to said non-naturally occurring nucleotide junction sequences under mild to stringent hybridization conditions are termed “nucleotide probes.”


In aspects, genomic DNA can be extracted from samples and used to quantify the presence of microbes of the disclosure by using qPCR. The primers utilized in the qPCR reaction can be primers designed by Primer Blast (https://www.ncbi.nlm.nih.gov/tools/primer-blast/) to amplify unique regions of the wild-type genome or unique regions of the engineered non-intergeneric mutant strains. The qPCR reaction can be carried out using the SYBR GreenER qPCR SuperMix Universal (Thermo Fisher P/N 11762100) kit, using only forward and reverse amplification primers; alternatively, the Kapa Probe Force kit (Kapa Biosystems P/N KK4301) can be used with amplification primers and a TaqMan probe containing a FAM dye label at the 5′ end, an internal ZEN quencher, and a minor groove binder and fluorescent quencher at the 3′ end (Integrated DNA Technologies).


Certain primer, probe, and non-native junction sequences are listed in Table 30. qPCR reaction efficiency can be measured using a standard curve generated from a known quantity of gDNA from the target genome. Data can be normalized to genome copies per g fresh weight using the tissue weight and extraction volume.


Quantitative polymerase chain reaction (qPCR) is a method of quantifying, in real time, the amplification of one or more nucleic acid sequences. The real time quantification of the PCR assay permits determination of the quantity of nucleic acids being generated by the PCR amplification steps by comparing the amplifying nucleic acids of interest and an appropriate control nucleic acid sequence, which may act as a calibration standard.


TaqMan probes are often utilized in qPCR assays that require an increased specificity for quantifying target nucleic acid sequences. TaqMan probes comprise a oligonucleotide probe with a fluorophore attached to the 5′ end and a quencher attached to the 3′ end of the probe. When the TaqMan probes remain as is with the 5′ and 3′ ends of the probe in close contact with each other, the quencher prevents fluorescent signal transmission from the fluorophore. TaqMan probes are designed to anneal within a nucleic acid region amplified by a specific set of primers. As the Taq polymerase extends the primer and synthesizes the nascent strand, the 5′ to 3′ exonuclease activity of the Taq polymerase degrades the probe that annealed to the template. This probe degradation releases the fluorophore, thus breaking the close proximity to the quencher and allowing fluorescence of the fluorophore. Fluorescence detected in the qPCR assay is directly proportional to the fluorophore released and the amount of DNA template present in the reaction.


The features of qPCR allow the practitioner to eliminate the labor-intensive post-amplification step of gel electrophoresis preparation, which is generally required for observation of the amplified products of traditional PCR assays. The benefits of qPCR over conventional PCR are considerable, and include increased speed, ease of use, reproducibility, and quantitative ability.


Improvement of Traits


Methods of the present disclosure may be employed to introduce or improve one or more of a variety of desirable traits. Examples of traits that may introduced or improved include: root biomass, root length, height, shoot length, leaf number, water use efficiency, overall biomass, yield, fruit size, grain size, photosynthesis rate, tolerance to drought, heat tolerance, salt tolerance, resistance to nematode stress, resistance to a fungal pathogen, resistance to a bacterial pathogen, resistance to a viral pathogen, level of a metabolite, and proteome expression. The desirable traits, including height, overall biomass, root and/or shoot biomass, seed germination, seedling survival, photosynthetic efficiency, transpiration rate, seed/fruit number or mass, plant grain or fruit yield, leaf chlorophyll content, photosynthetic rate, root length, or any combination thereof, can be used to measure growth, and compared with the growth rate of reference agricultural plants (e.g., plants without the improved traits) grown under identical conditions.


A preferred trait to be introduced or improved is nitrogen fixation, as described herein. In some cases, a plant resulting from the methods described herein exhibits a difference in the trait that is at least about 5% greater, for example at least about 5%, at least about 8%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 75%, at least about 80%, at least about 80%, at least about 90%, or at least 100%, at least about 200%, at least about 300%, at least about 400% or greater than a reference agricultural plant grown under the same conditions in the soil. In additional examples, a plant resulting from the methods described herein exhibits a difference in the trait that is at least about 5% greater, for example at least about 5%, at least about 8%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 75%, at least about 80%, at least about 80%, at least about 90%, or at least 100%, at least about 200%, at least about 300%, at least about 400% or greater than a reference agricultural plant grown under similar conditions in the soil.


The trait to be improved may be assessed under conditions including the application of one or more biotic or abiotic stressors. Examples of stressors include abiotic stresses (such as heat stress, salt stress, drought stress, cold stress, and low nutrient stress) and biotic stresses (such as nematode stress, insect herbivory stress, fungal pathogen stress, bacterial pathogen stress, and viral pathogen stress).


The trait improved by methods and compositions of the present disclosure may be nitrogen fixation, including in a plant not previously capable of nitrogen fixation. In some cases, bacteria isolated according to a method described herein produce 1% or more (e.g. 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 15%, 20%, or more) of a plant's nitrogen, which may represent an increase in nitrogen fixation capability of at least 2-fold (e.g. 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 20-fold, 50-fold, 100-fold, 1000-fold, or more) as compared to bacteria isolated from the first plant before introducing any genetic variation. In some cases, the bacteria produce 5% or more of a plant's nitrogen. The desired level of nitrogen fixation may be achieved after repeating the steps of introducing genetic variation, exposure to a plurality of plants, and isolating bacteria from plants with an improved trait one or more times (e.g. 1, 2, 3, 4, 5, 10, 15, 25, or more times). In some cases, enhanced levels of nitrogen fixation are achieved in the presence of fertilizer supplemented with glutamine, ammonia, or other chemical source of nitrogen. Methods for assessing degree of nitrogen fixation are known, examples of which are described herein.


Microbe breeding is a method to systematically identify and improve the role of species within the crop microbiome. The method comprises three steps: 1) selection of candidate species by mapping plant-microbe interactions and predicting regulatory networks linked to a particular phenotype, 2) pragmatic and predictable improvement of microbial phenotypes through intra-species crossing of regulatory networks and gene clusters, and 3) screening and selection of new microbial genotypes that produce desired crop phenotypes. To systematically assess the improvement of strains, a model is created that links colonization dynamics of the microbial community to genetic activity by key species. The model is used to predict genetic targets for breeding and improve the frequency of selecting improvements in microbiome-encoded traits of agronomic relevance.


Measuring Nitrogen Delivered in an Agriculturally Relevant Field Context


In the field, the amount of nitrogen delivered can be determined by the function of colonization multiplied by the activity.







Nitrogen


delivered

=





Time

&



Space



Colonization

×

Activitiy






The above equation requires (1) the average colonization per unit of plant tissue, and (2) the activity as either the amount of nitrogen fixed or the amount of ammonia excreted by each microbial cell. To convert to pounds of nitrogen per acre, corn growth physiology is tracked over time, e.g., size of the plant and associated root system throughout the maturity stages.


The pounds of nitrogen delivered to a crop per acre-season can be calculated by the following equation:

Nitrogen delivered=∫Plant Tissue(t)×Colonization(t)×Activity(t)dt


The Plant Tissue(t) is the fresh weight of corn plant tissue over the growing time (t). Values for reasonably making the calculation are described in detail in the publication entitled Roots, Growth and Nutrient Uptake (Mengel. Dept. of Agronomy Pub. #AGRY-95-08 (Rev. May-95. p. 1-8.).


The Colonization (t) is the amount of the microbes of interest found within the plant tissue, per gram fresh weight of plant tissue, at any particular time, t, during the growing season. In the instance of only a single timepoint available, the single timepoint is normalized as the peak colonization rate over the season, and the colonization rate of the remaining timepoints are adjusted accordingly.


Activity (t) is the rate of which N is fixed by the microbes of interest per unit time, at any particular time, t, during the growing season. In the embodiments disclosed herein, this activity rate is approximated by in vitro acetylene reduction assay (ARA) in ARA media in the presence of 5 mM glutamine or Ammonium excretion assay in ARA media in the presence of 5 mM ammonium ions.


The Nitrogen delivered amount is then calculated by numerically integrating the above function. In cases where the values of the variables described above are discretely measured at set timepoints, the values in between those timepoints are approximated by performing linear interpolation.


Nitrogen Fixation


Described herein are methods of increasing nitrogen fixation in a plant, comprising exposing the plant to bacteria comprising one or more genetic variations introduced into one or more genes regulating nitrogen fixation, wherein the bacteria produce 1% or more of nitrogen in the plant (e.g. 2%, 5%, 10%, or more), which may represent a nitrogen-fixation capability of at least 2-fold as compared to the plant in the absence of the bacteria. The bacteria may produce the nitrogen in the presence of fertilizer supplemented with glutamine, urea, nitrates or ammonia. Genetic variations can be any genetic variation described herein, including examples provided above, in any number and any combination. The genetic variation may be introduced into a gene selected from the group consisting of nifA, nifL, ntrB, ntrC, glutamine synthetase, glnA, glnB, glnK, draT, amtB, glutaminase, glnD, glnE, nifJ, nifH, nifD, nifK, nifY, nifE, nifN, nifU, nifS, nifV, nifW, nifZ, nifM, nifF, nifB, and nifQ. The genetic variation may be a mutation that results in one or more of: increased expression or activity of nifA or glutaminase; decreased expression or activity of nifL, ntrB, glutamine synthetase, glnB, glnK, draT, amtB; decreased adenylyl-removing activity of GlnE; or decreased uridylyl-removing activity of GlnD. The genetic variation introduced into one or more bacteria of the methods disclosed herein may be a knock-out mutation or it may abolish a regulatory sequence of a target gene, or it may comprise insertion of a heterologous regulatory sequence, for example, insertion of a regulatory sequence found within the genome of the same bacterial species or genus. The regulatory sequence can be chosen based on the expression level of a gene in a bacterial culture or within plant tissue. The genetic variation may be produced by chemical mutagenesis. The plants grown in step (c) may be exposed to biotic or abiotic stressors.


The amount of nitrogen fixation that occurs in the plants described herein may be measured in several ways, for example by an acetylene-reduction (AR) assay. An acetylene-reduction assay can be performed in vitro or in vivo. Evidence that a particular bacterium is providing fixed nitrogen to a plant can include: 1) total plant N significantly increases upon inoculation, preferably with a concomitant increase in N concentration in the plant; 2) nitrogen deficiency symptoms are relieved under N-limiting conditions upon inoculation (which should include an increase in dry matter); 3) N2 fixation is documented through the use of an 15N approach (which can be isotope dilution experiments, 15N2 reduction assays, or 15N natural abundance assays); 4) fixed N is incorporated into a plant protein or metabolite; and 5) all of these effects are not be seen in non-inoculated plants or in plants inoculated with a mutant of the inoculum strain.


The wild-type nitrogen fixation regulatory cascade can be represented as a digital logic circuit where the inputs O2 and NH4+ pass through a NOR gate, the output of which enters an AND gate in addition to ATP. In some embodiments, the methods disclosed herein disrupt the influence of NH4+ on this circuit, at multiple points in the regulatory cascade, so that microbes can produce nitrogen even in fertilized fields. However, the methods disclosed herein also envision altering the impact of ATP or O2 on the circuitry, or replacing the circuitry with other regulatory cascades in the cell, or altering genetic circuits other than nitrogen fixation. Gene clusters can be re-engineered to generate functional products under the control of a heterologous regulatory system. By eliminating native regulatory elements outside of, and within, coding sequences of gene clusters, and replacing them with alternative regulatory systems, the functional products of complex genetic operons and other gene clusters can be controlled and/or moved to heterologous cells, including cells of different species other than the species from which the native genes were derived. Once re-engineered, the synthetic gene clusters can be controlled by genetic circuits or other inducible regulatory systems, thereby controlling the products' expression as desired. The expression cassettes can be designed to act as logic gates, pulse generators, oscillators, switches, or memory devices. The controlling expression cassette can be linked to a promoter such that the expression cassette functions as an environmental sensor, such as an oxygen, temperature, touch, osmotic stress, membrane stress, or redox sensor.


As an example, the nifL, nifA, nifT, and nifX genes can be eliminated from the nif gene cluster. Synthetic genes can be designed by codon randomizing the DNA encoding each amino acid sequence. Codon selection is performed, specifying that codon usage be as divergent as possible from the codon usage in the native gene. Proposed sequences are scanned for any undesired features, such as restriction enzyme recognition sites, transposon recognition sites, repetitive sequences, sigma 54 and sigma 70 promoters, cryptic ribosome binding sites, and rho independent terminators. Synthetic ribosome binding sites are chosen to match the strength of each corresponding native ribosome binding site, such as by constructing a fluorescent reporter plasmid in which the 150 bp surrounding a gene's start codon (from −60 to +90) is fused to a fluorescent gene. This chimera can be expressed under control of the Ptac promoter, and fluorescence measured via flow cytometry. To generate synthetic ribosome binding sites, a library of reporter plasmids using 150 bp (−60 to +90) of a synthetic expression cassette is generated. Briefly, a synthetic expression cassette can consist of a random DNA spacer, a degenerate sequence encoding an RBS library, and the coding sequence for each synthetic gene. Multiple clones are screened to identify the synthetic ribosome binding site that best matched the native ribosome binding site. Synthetic operons that consist of the same genes as the native operons are thus constructed and tested for functional complementation. A further exemplary description of synthetic operons is provided in US20140329326.


Bacterial Species


Microbes useful in the methods and compositions disclosed herein may be obtained from any source. In some cases, microbes may be bacteria, archaea, protozoa or fungi. The microbes of this disclosure may be nitrogen fixing microbes, for example a nitrogen fixing bacteria, nitrogen fixing archaea, nitrogen fixing fungi, nitrogen fixing yeast, or nitrogen fixing protozoa. Microbes useful in the methods and compositions disclosed herein may be spore forming microbes, for example spore forming bacteria. In some cases, bacteria useful in the methods and compositions disclosed herein may be Gram positive bacteria or Gram negative bacteria. In some cases, the bacteria may be an endospore forming bacteria of the Firmicute phylum. In some cases, the bacteria may be a diazatroph. In some cases, the bacteria may not be a diazotroph.


The methods and compositions of this disclosure may be used with an archaea, such as, for example, Methanothermobacter thermoautotrophicus.


In some cases, bacteria which may be useful include, but are not limited to, Agrobacterium radiobacter, Bacillus acidocaldarius, Bacillus acidoterrestris, Bacillus agri, Bacillus aizawai, Bacillus albolactis, Bacillus alcalophilus, Bacillus alvei, Bacillus aminoglucosidicus, Bacillus aminovorans, Bacillus amylolyticus (also known as Paenibacillus amylolyticus) Bacillus amyloliquefaciens, Bacillus aneurinolyticus, Bacillus atrophaeus, Bacillus azotoformans, Bacillus badius, Bacillus cereus (synonyms: Bacillus endorhythmos, Bacillus medusa), Bacillus chitinosporus, Bacillus circulans, Bacillus coagulans, Bacillus endoparasiticus Bacillus fastidiosus, Bacillus firmus, Bacillus kurstaki, Bacillus lacticola, Bacillus lactimorbus, Bacillus lactis, Bacillus laterosporus (also known as Brevibacillus laterosporus), Bacillus lautus, Bacillus lentimorbus, Bacillus lentus, Bacillus licheniformis, Bacillus maroccanus, Bacillus megaterium, Bacillus metiens, Bacillus mycoides, Bacillus natto, Bacillus nematocida, Bacillus nigrificans, Bacillus nigrum, Bacillus pantothenticus, Bacillus popillae, Bacillus psychrosaccharolyticus, Bacillus pumilus, Bacillus siamensis, Bacillus smithii, Bacillus sphaericus, Bacillus subtilis, Bacillus thuringiensis, Bacillus uniflagellatus, Bradyrhizobium japonicum, Brevibacillus brevis Brevibacillus laterosporus (formerly Bacillus laterosporus), Chromobacterium subtsugae, Delftia acidovorans, Lactobacillus acidophilus, Lysobacter antibioticus, Lysobacter enzymogenes, Paenibacillus alvei, Paenibacillus polymyxa, Paenibacillus popilliae (formerly Bacillus popilliae), Pantoea agglomerans, Pasteuria penetrans (formerly Bacillus penetrans), Pasteuria usgae, Pectobacterium carotovorum (formerly Erwinia carotovora), Pseudomonas aeruginosa, Pseudomonas aureofaciens, Pseudomonas cepacia (formerly known as Burkholderia cepacia), Pseudomonas chlororaphis, Pseudomonas fluorescens, Pseudomonas proradix, Pseudomonas putida, Pseudomonas syringae, Serratia entomophila, Serratia marcescens, Streptomyces colombiensis, Streptomyces galbus, Streptomyces goshikiensis, Streptomyces griseoviridis, Streptomyces lavendulae, Streptomyces prasinus, Streptomyces saraceticus, Streptomyces venezuelae, Xanthomonas campestris, Xenorhabdus luminescens, Xenorhabdus nematophila, Rhodococcus globerulus AQ719 (NRRL Accession No. B-21663), Bacillus sp. AQ175 (ATCC Accession No. 55608), Bacillus sp. AQ 177 (ATCC Accession No. 55609), Bacillus sp. AQ178 (ATCC Accession No. 53522), and Streptomyces sp. strain NRRL Accession No. B-30145. In some cases the bacterium may be Azotobacter chroococcum, Methanosarcina barkeri, Klebsiella pneumoniae, Azotobacter vinelandii, Rhodobacter sphaeroides, Rhodobacter capsulatus, Rhodobacter palustris, Rhodospirillum rubrum, Rhizobium leguminosarum or Rhizobium etli.


In some cases the bacterium may be a species of Clostridium, for example Clostridium pasteurianum, Clostridium beijerinckii, Clostridium perfringens, Clostridium tetani, Clostridium acetobutylicum.


In some cases, bacteria used with the methods and compositions of the present disclosure may be cyanobacteria. Examples of cyanobacterial genuses include Anabaena (for example Anagaena sp. PCC7120), Nostoc (for example Nostoc punctiforme), or Synechocystis (for example Synechocystis sp. PCC6803).


In some cases, bacteria used with the methods and compositions of the present disclosure may belong to the phylum Chlorobi, for example Chlorobium tepidum.


In some cases, microbes used with the methods and compositions of the present disclosure may comprise a gene homologous to a known NifH gene. Sequences of known NifH genes may be found in, for example, the Zehr lab NifH database, (https://wwwzehr.pmc.ucsc.edu/nifH_Database_Public/, Apr. 4, 2014), or the Buckley lab NifH database (http://www.css.cornell.edu/faculty/buckley/nifh.htm, and Gaby, John Christian, and Daniel H. Buckley. “A comprehensive aligned nifH gene database: a multipurpose tool for studies of nitrogen-fixing bacteria.” Database 2014 (2014): bau001.). In some cases, microbes used with the methods and compositions of the present disclosure may comprise a sequence which encodes a polypeptide with at least 60%, 70%, 80%, 85%, 90%, 95%, 96%, 96%, 98%, 99% or more than 99% sequence identity to a sequence from the Zehr lab NifH database, (https://wwwzehr.pmc.ucsc.edu/nifH_Database_Public/, Apr. 4, 2014). In some cases, microbes used with the methods and compositions of the present disclosure may comprise a sequence which encodes a polypeptide with at least 60%, 70%, 80%, 85%, 90%, 95%, 96%, 96%, 98%, 99% or more than 99% sequence identity to a sequence from the Buckley lab NifH database, (Gaby, John Christian, and Daniel H. Buckley. “A comprehensive aligned nifH gene database: a multipurpose tool for studies of nitrogen-fixing bacteria.” Database 2014 (2014): bau001.).


Microbes useful in the methods and compositions disclosed herein can be obtained by extracting microbes from surfaces or tissues of native plants; grinding seeds to isolate microbes; planting seeds in diverse soil samples and recovering microbes from tissues; or inoculating plants with exogenous microbes and determining which microbes appear in plant tissues. Non-limiting examples of plant tissues include a seed, seedling, leaf, cutting, plant, bulb, tuber, root, and rhizomes. In some cases, bacteria are isolated from a seed. The parameters for processing samples may be varied to isolate different types of associative microbes, such as rhizospheric, epiphytes, or endophytes. Bacteria may also be sourced from a repository, such as environmental strain collections, instead of initially isolating from a first plant. The microbes can be genotyped and phenotyped, via sequencing the genomes of isolated microbes; profiling the composition of communities in planta; characterizing the transcriptomic functionality of communities or isolated microbes; or screening microbial features using selective or phenotypic media (e.g., nitrogen fixation or phosphate solubilization phenotypes). Selected candidate strains or populations can be obtained via sequence data; phenotype data; plant data (e.g., genome, phenotype, and/or yield data); soil data (e.g., pH, N/P/K content, and/or bulk soil biotic communities); or any combination of these.


The bacteria and methods of producing bacteria described herein may apply to bacteria able to self-propagate efficiently on the leaf surface, root surface, or inside plant tissues without inducing a damaging plant defense reaction, or bacteria that are resistant to plant defense responses. The bacteria described herein may be isolated by culturing a plant tissue extract or leaf surface wash in a medium with no added nitrogen. However, the bacteria may be unculturable, that is, not known to be culturable or difficult to culture using standard methods known in the art. The bacteria described herein may be an endophyte or an epiphyte or a bacterium inhabiting the plant rhizosphere (rhizospheric bacteria). The bacteria obtained after repeating the steps of introducing genetic variation, exposure to a plurality of plants, and isolating bacteria from plants with an improved trait one or more times (e.g. 1, 2, 3, 4, 5, 10, 15, 25, or more times) may be endophytic, epiphytic, or rhizospheric. Endophytes are organisms that enter the interior of plants without causing disease symptoms or eliciting the formation of symbiotic structures, and are of agronomic interest because they can enhance plant growth and improve the nutrition of plants (e.g., through nitrogen fixation). The bacteria can be a seed-borne endophyte. Seed-borne endophytes include bacteria associated with or derived from the seed of a grass or plant, such as a seed-borne bacterial endophyte found in mature, dry, undamaged (e.g., no cracks, visible fungal infection, or prematurely germinated) seeds. The seed-borne bacterial endophyte can be associated with or derived from the surface of the seed; alternatively, or in addition, it can be associated with or derived from the interior seed compartment (e.g., of a surface-sterilized seed). In some cases, a seed-borne bacterial endophyte is capable of replicating within the plant tissue, for example, the interior of the seed. Also, in some cases, the seed-borne bacterial endophyte is capable of surviving desiccation.


The bacterial isolated according to methods of the disclosure, or used in methods or compositions of the disclosure, can comprise a plurality of different bacterial taxa in combination. By way of example, the bacteria may include Proteobacteria (such as Pseudomonas, Enterobacter, Stenotrophomonas, Burkholderia, Rhizobium, Herbaspirillum, Pantoea, Serratia, Rahnella, Azospirillum, Azorhizobium, Azotobacter, Duganella, Delftia, Bradyrhizobium, Sinorhizobium and Halomonas), Firmicutes (such as Bacillus, Paenibacillus, Lactobacillus, Mycoplasma, and Acetobacterium), and Actinobacteria (such as Streptomyces, Rhodococcus, Microbacterium, and Curtobacterium). The bacteria used in methods and compositions of this disclosure may include nitrogen fixing bacterial consortia of two or more species. In some cases, one or more bacterial species of the bacterial consortia may be capable of fixing nitrogen. In some cases, one or more species of the bacterial consortia may facilitate or enhance the ability of other bacteria to fix nitrogen. The bacteria which fix nitrogen and the bacteria which enhance the ability of other bacteria to fix nitrogen may be the same or different. In some examples, a bacterial strain may be able to fix nitrogen when in combination with a different bacterial strain, or in a certain bacterial consortia, but may be unable to fix nitrogen in a monoculture. Examples of bacterial genuses which may be found in a nitrogen fixing bacterial consortia include, but are not limited to, Herbaspirillum, Azospirillum, Enterobacter, and Bacillus.


Bacteria that can be produced by the methods disclosed herein include Azotobacter sp., Bradyrhizobium sp., Klebsiella sp., and Sinorhizobium sp. In some cases, the bacteria may be selected from the group consisting of: Azotobacter vinelandii, Bradyrhizobium japonicum, Klebsiella pneumoniae, and Sinorhizobium meliloti. In some cases, the bacteria may be of the genus Enterobacter or Rahnella. In some cases, the bacteria may be of the genus Frankia, or Clostridium. Examples of bacteria of the genus Clostridium include, but are not limited to, Clostridium acetobutylicum, Clostridium pasteurianum, Clostridium beijerinckii, Clostridium perfringens, and Clostridium tetani. In some cases, the bacteria may be of the genus Paenibacillus, for example Paenibacillus azotofixans, Paenibacillus borealis, Paenibacillus durus, Paenibacillus macerans, Paenibacillus polymyxa, Paenibacillus alvei, Paenibacillus amylolyticus, Paenibacillus campinasensis, Paenibacillus chibensis, Paenibacillus glucanolyticus, Paenibacillus illinoisensis, Paenibacillus larvae subsp. larvae, Paenibacillus larvae subsp. pulvifaciens, Paenibacillus lautus, Paenibacillus macerans, Paenibacillus macquariensis, Paenibacillus macquariensis, Paenibacillus pabuli, Paenibacillus peoriae, or Paenibacillus polymyxa.


In some examples, bacteria isolated according to methods of the disclosure can be a member of one or more of the following taxa: Achromobacter, Acidithiobacillus, Acidovorax, Acidovoraz, Acinetobacter, Actinoplanes, Adlercreutzia, Aerococcus, Aeromonas, Afipia, Agromyces, Ancylobacter, Arthrobacter, Atopostipes, Azospirillum, Bacillus, Bdellovibrio, Beijerinckia, Bosea, Bradyrhizobium, Brevibacillus, Brevundimonas, Burkholderia, Candidatus Haloredivivus, Caulobacter, Cellulomonas, Cellvibrio, Chryseobacterium, Citrobacter, Clostridium, Coraliomargarita, Corynebacterium, Cupriavidus, Curtobacterium, Curvibacter, Deinococcus, Delftia, Desemzia, Devosia, Dokdonella, Dyella, Enhydrobacter, Enterobacter, Enterococcus, Erwinia, Escherichia, Escherichia/Shigella, Exiguobacterium, Ferroglobus, Filimonas, Finegoldia, Flavisolibacter, Flavobacterium, Frigoribacterium, Gluconacetobacter, Hafnia, Halobaculum, Halomonas, Halosimplex, Herbaspirillum, Hymenobacter, Klebsiella, Kocuria, Kosakonia, Lactobacillus, Leclercia, Lentzea, Luteibacter, Luteimonas, Massilia, Mesorhizobium, Methylobacterium, Microbacterium, Micrococcus, Microvirga, Mycobacterium, Neisseria, Nocardia, Oceanibaculum, Ochrobactrum, Okibacterium, Oligotropha, Oryzihumus, Oxalophagus, Paenibacillus, Panteoa, Pantoea, Pelomonas, Perlucidibaca, Plantibacter, Polynucleobacter, Propionibacterium, Propioniciclava, Pseudoclavibacter, Pseudomonas, Pseudonocardia, Pseudoxanthomonas, Psychrobacter, Rahnella, Ralstonia, Rheinheimera, Rhizobium, Rhodococcus, Rhodopseudomonas, Roseateles, Ruminococcus, Sebaldella, Sediminibacillus, Sediminibacterium, Serratia, Shigella, Shinella, Sinorhizobium, Sinosporangium, Sphingobacterium, Sphingomonas, Sphingopyxis, Sphingosinicella, Staphylococcus, 25 Stenotrophomonas, Strenotrophomonas, Streptococcus, Streptomyces, Stygiolobus, Sulfurisphaera, Tatumella, Tepidimonas, Thermomonas, Thiobacillus, Variovorax, WPS-2 genera incertae sedis, Xanthomonas, and Zimmermannella.


In some cases, a bacterial species selected from at least one of the following genera are utilized: Enterobacter, Klebsiella, Kosakonia, and Rahnella. In some cases, a combination of bacterial species from the following genera are utilized: Enterobacter, Klebsiella, Kosakonia, and Rahnella. In some cases, the species utilized can be one or more of: Enterobacter sacchari, Klebsiella variicola, Kosakonia sacchari, and Rahnella aquatilis.


In some cases, a Gram positive microbe may have a Molybdenum-Iron nitrogenase system comprising: nifH, nifD, nifK, nifB, nifE, nifN, nifX hesA, nifV nifW, nifU, nifS, nifI1, and nifI2. In some cases, a Gram positive microbe may have a vanadium nitrogenase system comprising: vnfDG, vnfK, vnfE, vnfN, vupC, vupB, vupA, vnfV, vnfR1, vnfH, vnfR2, vnfA (transcriptional regulator). In some cases, a Gram positive microbe may have an iron-only nitrogenase system comprising: anfK, anfG, anfD, anfH, anfA (transcriptional regulator). In some cases a Gram positive microbe may have a nitrogenase system comprising glnB, and glnK (nitrogen signaling proteins). Some examples of enzymes involved in nitrogen metabolism in Gram positive microbes include glnA (glutamine synthetase), gdh (glutamate dehydrogenase), bdh (3-hydroxybutyrate dehydrogenase), glutaminase, gltAB/gltB/gltS (glutamate synthase), asnA/asnB (aspartate-ammonia ligase/asparagine synthetase), and ansA/ansZ (asparaginase). Some examples of proteins involved in nitrogen transport in Gram positive microbes include amtB (ammonium transporter), glnK (regulator of ammonium transport), glnPHQ/glnQHMP (ATP-dependent glutamine/glutamate transporters), glnT/alsT/yrbD/yflA (glutamine-like proton symport transporters), and gltP/gltT/yhcl/nqt (glutamate-like proton symport transporters).


Examples of Gram positive microbes which may be of particular interest include Paenibacillus polymixa, Paenibacillus riograndensis, Paenibacillus sp., Frankia sp., Heliobacterium sp., Heliobacterium chlorum, Heliobacillus sp., Heliophilum sp., Heliorestis sp., Clostridium acetobutylicum, Clostridium sp., Mycobacterium flaum, Mycobacterium sp., Arthrobacter sp., Agromyces sp., Corynebacterium autitrophicum, Corynebacterium sp., Micromonspora sp., Propionibacteria sp., Streptomyces sp., and Microbacterium sp.


Some examples of genetic alterations which may be made in Gram positive microbes include: deleting glnR to remove negative regulation of BNF in the presence of environmental nitrogen, inserting different promoters directly upstream of the nif cluster to eliminate regulation by GlnR in response to environmental nitrogen, mutating glnA to reduce the rate of ammonium assimilation by the GS-GOGAT pathway, deleting amtB to reduce uptake of ammonium from the media, mutating glnA so it is constitutively in the feedback-inhibited (FBI-GS) state, to reduce ammonium assimilation by the GS-GOGAT pathway.


In some cases, glnR is the main regulator of N metabolism and fixation in Paenibacillus species. In some cases, the genome of a Paenibacillus species may not contain a gene to produce glnR. In some cases, the genome of a Paenibacillus species may not contain a gene to produce glnE or glnD. In some cases, the genome of a Paenibacillus species may contain a gene to produce glnB or glnK. For example, Paenibacillus sp. WLY78 doesn't contain a gene for glnB, or its homologs found in the archaeon Methanococcus maripaludis, nifI1 and nifI2. In some cases, the genomes of Paenibacillus species may be variable. For example, Paenibacillus polymixa E681 lacks glnK and gdh, has several nitrogen compound transporters, but only amtB appears to be controlled by GlnR. In another example, Paenibacillus sp. JDR2 has glnK, gdh and most other central nitrogen metabolism genes, has many fewer nitrogen compound transporters, but does have glnPHQ controlled by GlnR. Paenibacillus riograndensis SBR5 contains a standard glnRA operon, an fdx gene, a main nif operon, a secondary nif operon, and an anf operon (encoding iron-only nitrogenase). Putative glnR/tnrA sites were found upstream of each of these operons. GlnR may regulate all of the above operons, except the anf operon. GlnR may bind to each of these regulatory sequences as a dimer.



Paenibacillus N-fixing strains may fall into two subgroups: Subgroup I, which contains only a minimal nif gene cluster and subgroup II, which contains a minimal cluster, plus an uncharacterized gene between mifX and hesA, and often other clusters duplicating some of the nif genes, such as nifH, nifHDK, nifBEN, or clusters encoding vanadaium nitrogenase (vnf) or iron-only nitrogenase (anf) genes.


In some cases, the genome of a Paenibacillus species may not contain a gene to produce glnB or glnK. In some cases, the genome of a Paenibacillus species may contain a minimal nif cluster with 9 genes transcribed from a sigma-70 promoter. In some cases, a Paenibacillus nif cluster may be negatively regulated by nitrogen or oxygen. In some cases, the genome of a Paenibacillus species may not contain a gene to produce sigma-54. For example, Paenibacillus sp. WLY78 does not contain a gene for sigma-54. In some cases, a nif cluster may be regulated by glnR, and/or TnrA. In some cases, activity of a nif cluster may be altered by altering activity of glnR, and/or TnrA.


In Bacilli, glutamine synthetase (GS) is feedback-inhibited by high concentrations of intracellular glutamine, causing a shift in confirmation (referred to as FBI-GS). Nif clusters contain distinct binding sites for the regulators GlnR and TnrA in several Bacilli species. GlnR binds and represses gene expression in the presence of excess intracellular glutamine and AMP. A role of GlnR may be to prevent the influx and intracellular production of glutamine and ammonium under conditions of high nitrogen availability. TnrA may bind and/or activate (or repress) gene expression in the presence of limiting intracellular glutamine, and/or in the presence of FBI-GS. In some cases the activity of a Bacilli nif cluster may be altered by altering the activity of GlnR.


Feedback-inhibited glutamine synthetase (FBI-GS) may bind GlnR and stabilize binding of GlnR to recognition sequences. Several bacterial species have a GlnR/TnrA binding site upstream of the nif cluster. Altering the binding of FBI-GS and GlnR may alter the activity of the nif pathway.


Sources of Microbes


The bacteria (or any microbe according to the disclosure) may be obtained from any general terrestrial environment, including its soils, plants, fungi, animals (including invertebrates) and other biota, including the sediments, water and biota of lakes and rivers; from the marine environment, its biota and sediments (for example, sea water, marine muds, marine plants, marine invertebrates (for example, sponges), marine vertebrates (for example, fish)); the terrestrial and marine geosphere (regolith and rock, for example, crushed subterranean rocks, sand and clays); the cryosphere and its meltwater; the atmosphere (for example, filtered aerial dusts, cloud and rain droplets); urban, industrial and other man-made environments (for example, accumulated organic and mineral matter on concrete, roadside gutters, roof surfaces, and road surfaces).


The plants from which the bacteria (or any microbe according to the disclosure) are obtained may be a plant having one or more desirable traits, for example a plant which naturally grows in a particular environment or under certain conditions of interest. By way of example, a certain plant may naturally grow in sandy soil or sand of high salinity, or under extreme temperatures, or with little water, or it may be resistant to certain pests or disease present in the environment, and it may be desirable for a commercial crop to be grown in such conditions, particularly if they are, for example, the only conditions available in a particular geographic location. By way of further example, the bacteria may be collected from commercial crops grown in such environments, or more specifically from individual crop plants best displaying a trait of interest amongst a crop grown in any specific environment: for example the fastest-growing plants amongst a crop grown in saline-limiting soils, or the least damaged plants in crops exposed to severe insect damage or disease epidemic, or plants having desired quantities of certain metabolites and other compounds, including fiber content, oil content, and the like, or plants displaying desirable colors, taste or smell. The bacteria may be collected from a plant of interest or any material occurring in the environment of interest, including fungi and other animal and plant biota, soil, water, sediments, and other elements of the environment as referred to previously.


The bacteria (or any microbe according to the disclosure) may be isolated from plant tissue. This isolation can occur from any appropriate tissue in the plant, including for example root, stem and leaves, and plant reproductive tissues. By way of example, conventional methods for isolation from plants typically include the sterile excision of the plant material of interest (e.g. root or stem lengths, leaves), surface sterilization with an appropriate solution (e.g. 2% sodium hypochlorite), after which the plant material is placed on nutrient medium for microbial growth. Alternatively, the surface-sterilized plant material can be crushed in a sterile liquid (usually water) and the liquid suspension, including small pieces of the crushed plant material spread over the surface of a suitable solid agar medium, or media, which may or may not be selective (e.g. contain only phytic acid as a source of phosphorus). This approach is especially useful for bacteria which form isolated colonies and can be picked off individually to separate plates of nutrient medium, and further purified to a single species by well-known methods. Alternatively, the plant root or foliage samples may not be surface sterilized but only washed gently thus including surface-dwelling epiphytic microorganisms in the isolation process, or the epiphytic microbes can be isolated separately, by imprinting and lifting off pieces of plant roots, stem or leaves onto the surface of an agar medium and then isolating individual colonies as above. This approach is especially useful for bacteria, for example. Alternatively, the roots may be processed without washing off small quantities of soil attached to the roots, thus including microbes that colonize the plant rhizosphere. Otherwise, soil adhering to the roots can be removed, diluted and spread out onto agar of suitable selective and non-selective media to isolate individual colonies of rhizospheric bacteria.


Budapest Treaty on the International Recognition of the Deposit of Microorganisms for the Purpose of Patent Procedures


The microbial deposits of the present disclosure were made under the provisions of the Budapest Treaty on the International Recognition of the Deposit of Microorganisms for the Purpose of Patent Procedure (Budapest Treaty).


Applicants state that pursuant to 37 C.F.R. § 1.808(a)(2) “all restrictions imposed by the depositor on the availability to the public of the deposited material will be irrevocably removed upon the granting of the patent.” This statement is subject to paragraph (b) of this section (i.e. 37 C.F.R. § 1.808(b)).


The Enterobacter sacchari has now been reclassified as Kosakonia sacchari, the name for the organism may be used interchangeably throughout the manuscript.


Many microbes of the present disclosure are derived from two wild-type strains, as depicted in FIG. 6 and FIG. 7. Strain CI006 is a bacterial species previously classified in the genus Enterobacter (see aforementioned reclassification into Kosakonia), and FIG. 6 identifies the lineage of the mutants that have been derived from CI006. Strain CI019 is a bacterial species classified in the genus Rahnella, and FIG. 7 identifies the lineage of the mutants that have been derived from CI019. With regard to FIG. 6 and FIG. 7, it is noted that strains comprising CM in the name are mutants of the strains depicted immediately to the left of said CM strain. The deposit information for the CI006 Kosakonia wild type (WT) and CI019 Rahnella WT are found in the below Table 1.


Some microorganisms described in this application were deposited on Jan. 6, 2017 or Aug. 11, 2017 with the Bigelow National Center for Marine Algae and Microbiota (NCMA), located at 60 Bigelow Drive, East Boothbay, Maine 04544, USA. As aforementioned, all deposits were made under the terms of the Budapest Treaty on the International Recognition of the Deposit of Microorganisms for the Purposes of Patent Procedure. The Bigelow National Center for Marine Algae and Microbiota accession numbers and dates of deposit for the aforementioned Budapest Treaty deposits are provided in Table 1.


Biologically pure cultures of Kosakonia sacchari (WT), Rahnella aquatilis (WT), and a variant/remodeled Kosakonia sacchari strain were deposited on Jan. 6, 2017 with the Bigelow National Center for Marine Algae and Microbiota (NCMA), located at 60 Bigelow Drive, East Boothbay, Maine 04544, USA, and assigned NCMA Patent Deposit Designation numbers 201701001, 201701003, and 201701002, respectively. The applicable deposit information is found below in Table 1.


Biologically pure cultures of variant/remodeled Kosakonia sacchari strains were deposited on Aug. 11, 2017 with the Bigelow National Center for Marine Algae and Microbiota (NCMA), located at 60 Bigelow Drive, East Boothbay, Maine 04544, USA, and assigned NCMA Patent Deposit Designation numbers 201708004, 201708003, and 201708002, respectively. The applicable deposit information is found below in Table 1.


A biologically pure culture of Klebsiella variicola (WT) was deposited on Aug. 11, 2017 with the Bigelow National Center for Marine Algae and Microbiota (NCMA), located at 60 Bigelow Drive, East Boothbay, Maine 04544, USA, and assigned NCMA Patent Deposit Designation number 201708001. Biologically pure cultures of two Klebsiella variicola variants/remodeled strains were deposited on Dec. 20, 2017 with the Bigelow National Center for Marine Algae and Microbiota (NCMA), located at 60 Bigelow Drive, East Boothbay, Maine 04544, USA, and assigned NCMA Patent Deposit Designation numbers 201712001 and 201712002, respectively. The applicable deposit information is found below in Table 1.









TABLE 1







Microorganisms Deposited under the Budapest Treaty












Pivot Strain






Designation






(some strains





De-
have multiple

Accession
Date of


pository
designations)
Taxonomy
Number
Deposit





NCMA
CI006,

Kosakonia

201701001
Jan. 6, 2017



PBC6.1,

sacchari (WT)






6





NCMA
CI019,

Rahnella

201701003
Jan. 6, 2017



19

aquatilis (WT)





NCMA
CM029,

Kosakonia sacchari

201701002
Jan. 6, 2017



6-412





NCMA
6-403

Kosakonia sacchari

201708004
Aug. 11, 2017



CM037





NCMA
6-404,

Kosokonia sacchari

201708003
Aug. 11, 2017



CM38,






PBC6.38





NCMA
CM094,

Kosakonia sacchari

201708002
Aug. 11, 2017



6-881,






PBC6.94





NCMA
CI137, 137,

Klebsiella

201708001
Aug. 11, 2017



PB137

variicola (WT)





NCMA
137-1034

Klebsiella variicola

201712001
Dec. 20, 2017


NCMA
137-1036

Klebsiella variicola

201712002
Dec. 20, 2017










Isolated and Biologically Pure Microorganisms


The present disclosure, in certain embodiments, provides isolated and biologically pure microorganisms that have applications, inter alia, in agriculture. The disclosed microorganisms can be utilized in their isolated and biologically pure states, as well as being formulated into compositions (see below section for exemplary composition descriptions). Furthermore, the disclosure provides microbial compositions containing at least two members of the disclosed isolated and biologically pure microorganisms, as well as methods of utilizing said microbial compositions. Furthermore, the disclosure provides for methods of modulating nitrogen fixation in plants via the utilization of the disclosed isolated and biologically pure microbes.


In some aspects, the isolated and biologically pure microorganisms of the disclosure are those from Table 1. In other aspects, the isolated and biologically pure microorganisms of the disclosure are derived from a microorganism of Table 1. For example, a strain, child, mutant, or derivative, of a microorganism from Table 1 are provided herein. The disclosure contemplates all possible combinations of microbes listed in Table 1, said combinations sometimes forming a microbial consortia. The microbes from Table 1, either individually or in any combination, can be combined with any plant, active molecule (synthetic, organic, etc.), adjuvant, carrier, supplement, or biological, mentioned in the disclosure.


In some aspects, the disclosure provides microbial compositions comprising species as grouped in Tables 2-8. In some aspects, these compositions comprising various microbial species are termed a microbial consortia or consortium.


With respect to Tables 2-8, the letters A through I represent a non-limiting selection of microorganisms of the present disclosure, defined as:

    • A=Microbe with accession number 201701001 identified in Table 1;
    • B=Microbe with accession number 201701003 identified in Table 1;
    • C=Microbe with accession number 201701002 identified in Table 1;
    • D=Microbe with accession number 201708004 identified in Table 1;
    • E=Microbe with accession number 201708003 identified in Table 1;
    • F=Microbe with accession number 201708002 identified in Table 1;
    • G=Microbe with accession number 201708001 identified in Table 1;
    • H=Microbe with accession number 201712001 identified in Table 1; and
    • I=Microbe with accession number 201712002 identified in Table 1.









TABLE 2





Eight and Nine Strain Compositions




















A, B, C, D, E, F, G, H
A, B, C, D, E, F, G, I
A, B, C, D, E, F, H, I
A, B, C, D, E, G, H, I
A, B, C, D, F, G, H, I
A, B, C, E, F, G, H, I


A, B, D, E, F, G, H, I
A, C, D, E, F, G, H, I
B, C, D, E, F, G, H, I
A, B, C, D, E, F, G, H, I
















TABLE 3





Seven Strain Compositions




















A, B, C, D, E, F, G
A, B, C, D, E, F, H
A, B, C, D, E, F, I
A, B, C, D, E, G, H
A, B, C, D, E, G, I
A, B, C, D, E, H, I


A, B, C, D, F, G, H
A, B, C, D, F, G, I
A, B, C, D, F, H, I
A, B, C, D, G, H, I
A, B, C, E, F, G, H
A, B, C, E, F, G, I


A, B, C, E, F, H, I
A, B, C, E, G, H, I
A, B, C, F, G, H, I
A, B, D, E, F, G, H
A, B, D, E, F, G, I
A, B, D, E, F, H, I


A, B, D, E, G, H, I
A, B, D, F, G, H, I
A, B, E, F, G, H, I
A, C, D, E, F, G, H
A, C, D, E, F, G, I
A, C, D, E, F, H, I


A, C, D, E, G, H, I
A, C, D, F, G, H, I
A, C, E, F, G, H, I
A, D, E, F, G, H, I
B, C, D, E, F, G, H
B, C, D, E, F, G, I


B, C, D, E, F, H, I
B, C, D, E, G, H, I
B, C, D, F, G, H, I
B, C, E, F, G, H, I
B, D, E, F, G, H, I
C, D, E, F, G, H, I
















TABLE 4





Six Strain Compositions





















A,B,C,D,E,F
A,B,C,D,E,G
A,B,C,D,E,H
A,B,C,D,E,I
A,B,C,D,F,G
A,B,C,D,F,H
A,B,C,D,F,I


A,B,C,D,G,H
A,B,C,D,G,I
A,B,C,D,H,I
A,B,C,E,F,G
A,B,C,E,F,H
A,B,C,E,F,I
A,B,C,E,G,H


A,B,C,E,G,I
A,B,C,E,H,I
A,B,C,F,G,H
A,B,C,F,G,I
A,B,C,F,H,I
A,B,C,G,H,I
A,B,D,E,F,G


A,B,D,E,F,H
A,B,D,E,F,I
A,B,D,E,G,H
A,B,D,E,G,I
A,B,D,E,H,I
A,B,D,F,G,H
A,B,D,F,G,I


D,E,F,G,H,I
C,E,F,G,H,I
A,B,D,F,H,I
A,B,D,G,H,I
A,B,E,F,G,H
A,B,E,F,G,I
A,B,E,F,H,I


A,B,E,G,H,I
A,B,F,G,H,I
A,C,D,E,F,G
A,C,D,E,F,H
A,C,D,E,F,I
A,C,D,E,G,H
A,C,D,E,G,I


A,C,D,E,H,I
A,C,D,F,G,H
A,C,D,F,G,I
A,C,D,F,H,I
A,C,D,G,H,I
A,C,E,F,G,H
A,C,E,F,G,I


A,C,E,F,H,I
A,C,E,G,H,I
A,C,F,G,H,I
A,D,E,F,G,H
A,D,E,F,G,I
A,D,E,F,H,I
A,D,E,G,H,I


A,D,F,G,H,I
A,E,F,G,H,I
B,C,D,E,F,G
B,C,D,E,F,H
B,C,D,E,F,I
B,C,D,E,G,H
B,C,D,E,G,I


B,C,D,E,H,I
B,C,D,F,G,H
B,C,D,F,G,I
B,C,D,F,H,I
B,C,D,G,H,I
B,C,E,F,G,H
B,C,E,F,G,I


B,C,E,F,H,I
B,C,E,G,H,I
B,C,F,G,H,I
B,D,E,F,G,H
B,D,E,F,G,I
B,D,E,F,H,I
B,D,E,G,H,I


B,D,F,G,H,I
B,E,F,G,H,I
C,D,E,F,G,H
C,D,E,F,G,I
C,D,E,F,H,I
C,D,E,G,H,I
C,D,F,G,H,I
















TABLE 5





Five Strain Compositions






















A,B,C,D,E
A,B,C,D,F
A,B,C,D,G
A,B,C,D,H
A,B,C,D,I
A,B,C,E,F
A,B,C,E,G
A,B,C,E,H


A,B,C,F,H
A,B,C,F,G
A,B,C,F,I
A,B,C,G,H
A,B,C,G,I
A,B,C,H,I
A,B,D,E,F
A,B,D,E,G


A,B,D,E,I
A,B,D,F,G
A,B,D,F,H
A,B,D,F,I
A,B,D,G,H
A,B,D,G,I
A,B,D,H,I
A,B,E,F,G


A,B,E,F,I
A,B,E,G,H
A,B,E,G,I
A,B,E,H,I
A,B,F,G,H
A,B,F,G,I
A,B,F,H,I
A,B,G,H,I


A,C,D,E,G
A,C,D,E,H
A,C,D,E,I
A,C,D,F,G
A,C,D,F,H
A,C,D,F,I
A,C,D,G,H
A,C,D,G,I


A,C,E,F,G
A,C,E,F,H
A,C,E,F,I
A,C,E,G,H
A,C,E,G,I
A,C,E,H,I
A,C,F,G,H
A,C,F,G,I


A,C,G,H,I
A,D,E,F,G
A,D,E,F,H
A,D,E,F,I
A,D,E,G,H
A,D,E,G,I
A,D,E,H,I
A,D,F,G,H


A,D,F,H,I
A,D,G,H,I
A,E,F,G,H
A,E,F,G,I
A,E,F,H,I
A,E,G,H,I
A,F,G,H,I
B,C,D,E,F


B,C,D,E,H
B,C,D,E,I
B,C,D,F,G
B,C,D,F,H
B,C,D,F,I
B,C,D,G,H
B,C,D,G,I
B,C,D,H,I


B,C,E,F,H
B,C,E,F,I
B,C,E,G,H
B,C,E,G,I
B,C,E,H,I
B,C,F,G,H
B,C,F,G,I
B,C,F,H,I


B,D,E,F,G
B,D,E,F,H
B,D,E,F,I
B,D,E,G,H
B,D,E,G,I
B,D,E,H,I
B,D,F,G,H
B,D,F,G,I


B,D,G,H,I
B,E,F,G,H
B,E,F,G,I
B,E,F,H,I
B,E,G,H,I
B,F,G,H,I
C,D,E,F,G
C,D,E,F,H


C,D,E,G,H
C,D,E,G,I
C,D,E,H,I
C,D,F,G,H
C,D,F,G,I
C,D,F,H,I
C,D,G,H,I
C,E,F,G,H


C,E,F,H,I
C,E,G,H,I
C,F,G,H,I
D,E,F,G,H
D,E,F,G,I
D,E,F,H,I
D,E,G,H,I
D,F,G,H,I


A,B,C,E,I
A,B,D,E,H
A,B,E,F,H
A,C,D,E,F
A,C,D,H,I
A,C,F,H,I
A,D,F,G,I
B,C,D,E,G


B,C,E,F,G
B,C,G,H,I
B,D,F,H,I
C,D,E,F,I
C,E,F,G,I
E,F,G,H,I
















TABLE 6





Four Strain Compositions























A,B,C,D
A,B,C,E
A,B,C,F
A,B,C,G
A,B,C,H
A,B,C,I
A,B,D,E
A,B,D,F
D,G,H,I


A,B,D,G
A,B,D,H
A,B,D,I
A,B,E,F
A,B,E,G
A,B,E,H
A,B,E,I
A,B,F,G
E,F,G,H


A,B,F,H
A,D,F,H
A,D,F,I
A,D,G,H
A,D,G,I
A,D,H,I
A,E,F,G
A,E,F,H
E,F,G,I


A,B,F,I
A,B,G,H
A,B,G,I
A,B,H,I
A,C,D,E
A,C,D,F
A,C,D,G
A,C,D,H
E,F,H,I


A,C,D,I
A,C,E,F
A,C,E,G
A,C,E,H
A,C,E,I
A,C,F,G
A,C,F,H
A,C,F,I
E,G,H,I


A,C,G,H
A,C,G,I
A,C,H,I
A,D,E,F
A,D,E,G
A,D,E,H
A,D,E,I
A,D,F,G
F,G,H,I


A,E,F,I
A,E,G,H
A,E,G,I
A,E,H,I
A,F,G,H
A,F,G,I
A,F,H,I
A,G,H,I
D,E,F,H


B,C,D,E
B,C,D,F
B,C,D,G
B,C,D,H
B,C,D,I
B,C,E,F
B,C,E,G
B,C,E,H
D,E,F,I


B,C,E,I
B,C,F,G
B,C,F,H
B,C,F,I
B,C,G,H
B,C,G,I
B,C,H,I
B,D,E,F
D,E,G,H


B,D,E,G
B,D,E,H
B,D,E,I
B,D,F,G
B,D,F,H
B,D,F,I
B,D,G,H
B,D,G,I
D,E,G,I


B,D,H,I
B,E,F,G
B,E,F,H
B,E,F,I
B,E,G,H
B,E,G,I
B,E,H,I
B,F,G,H
D,E,H,I


B,F,G,I
B,F,H,I
B,G,H,I
C,D,E,F
C,D,E,G
C,D,E,H
C,D,E,I
C,D,F,G
D,F,G,H


C,D,F,H
C,D,F,I
C,D,G,H
C,D,G,I
C,D,H,I
C,E,F,G
C,E,F,H
C,E,F,I
D,F,G,I


C,E,G,H
C,E,G,I
C,E,H,I
C,F,G,H
C,F,G,I
C,F,H,I
C,G,H,I
D,E,F,G
D,F,H,I
















TABLE 7





Three Strain Compositions

























A,B,C
A,B,D
A,B,E
A,B,F
A,B,G
A,B,H
A,B,I
A,C,D
A,C,E
G,H,I
E,F,H


A,C,F
A,C,G
A,C,H
A,C,I
A,D,E
A,D,F
A,D,G
A,D,H
A,D,I
F,H,I
E,F,G


A,E,F
A,E,G
A,E,H
A,E,I
A,F,G
A,F,H
A,F,I
A,G,H
A,G,I
F,G,I
D,H,I


A,H,I
B,C,D
B,C,E
B,C,F
B,C,G
B,C,I
B,C,I
B,D,E
B,D,F
F,G,H
D,G,I


B,D,G
B,D,H
B,D,I
B,E,F
B,E,G
B,E,H
B,E,I
B,F,G
B,F,H
E,H,I
E,F,I


B,F,I
B,G,H
B,G,I
B,H,I
C,D,E
C,D,F
C,D,G
C,D,H
C,D,I
E,G,I
D,G,H


C,E,F
C,E,G
C,E,H
C,E,I
C,F,G
C,F,H
C,F,I
C,G,H
C,G,I
E,G,H
D,F,I


C,H,I
D,E,F
D,E,G
D,E,H
D,E,I
D,F,G
D,F,H
















TABLE 8





Two strain Compositions






























A,B
A,C
A,D
A,E
A,F
A,G
A,H
A,I
B,C
B,D
B,E
B,F
B,G
B,H
B,I
C,D


C,E
C,F
C,G
C,H
C,I
D,E
D,F
D,G
D,H
D,I
E,F
EG
E,H
E,I
F,G
F,H


F,I
G,H
G,I
H,I









In some embodiments, microbial compositions may be selected from any member group from Tables 2-8.


Agricultural Compositions


Compositions comprising bacteria or bacterial populations produced according to methods described herein and/or having characteristics as described herein can be in the form of a liquid, a foam, or a dry product. Compositions comprising bacteria or bacterial populations produced according to methods described herein and/or having characteristics as described herein may also be used to improve plant traits. In some examples, a composition comprising bacterial populations may be in the form of a dry powder, a slurry of powder and water, or a flowable seed treatment. The compositions comprising bacterial populations may be coated on a surface of a seed, and may be in liquid form.


The composition can be fabricated in bioreactors such as continuous stirred tank reactors, batch reactors, and on the farm. In some examples, compositions can be stored in a container, such as a jug or in mini bulk. In some examples, compositions may be stored within an object selected from the group consisting of a bottle, jar, ampule, package, vessel, bag, box, bin, envelope, carton, container, silo, shipping container, truck bed, and/or case.


Compositions may also be used to improve plant traits. In some examples, one or more compositions may be coated onto a seed. In some examples, one or more compositions may be coated onto a seedling. In some examples, one or more compositions may be coated onto a surface of a seed. In some examples, one or more compositions may be coated as a layer above a surface of a seed. In some examples, a composition that is coated onto a seed may be in liquid form, in dry product form, in foam form, in a form of a slurry of powder and water, or in a flowable seed treatment. In some examples, one or more compositions may be applied to a seed and/or seedling by spraying, immersing, coating, encapsulating, and/or dusting the seed and/or seedling with the one or more compositions. In some examples, multiple bacteria or bacterial populations can be coated onto a seed and/or a seedling of the plant. In some examples, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, or more than ten bacteria of a bacterial combination can be selected from one of the following genera: Acidovorax, Agrobacterium, Bacillus, Burkholderia, Chryseobacterium, Curtobacterium, Enterobacter, Escherichia, Methylobacterium, Paenibacillus, Pantoea, Pseudomonas, Ralstonia, Saccharibacillus, Sphingomonas, and Stenotrophomonas.


In some examples, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, or more than ten bacteria and bacterial populations of an endophytic combination are selected from one of the following families: Bacillaceae, Burkholderiaceae, Comamonadaceae, Enterobacteriaceae, Flavobacteriaceae, Methylobacteriaceae, Microbacteriaceae, Paenibacillileae, Pseudomonnaceae, Rhizobiaceae, Sphingomonadaceae, Xanthomonadaceae, Cladosporiaceae, Gnomoniaceae, Incertae sedis, Lasiosphaeriaceae, Netriaceae, and Pleosporaceae.


In some examples, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least night, at least ten, or more than ten bacteria and bacterial populations of an endophytic combination are selected from one of the following families: Bacillaceae, Burkholderiaceae, Comamonadaceae, Enterobacteriaceae, Flavobacteriaceae, Methylobacteriaceae, Microbacteriaceae, Paenibacillileae, Pseudomonnaceae, Rhizobiaceae, Sphingomonadaceae, Xanthomonadaceae, Cladosporiaceae, Gnomoniaceae, Incertae sedis, Lasiosphaeriaceae, Netriaceae, Pleosporaceae.


Examples of compositions may include seed coatings for commercially important agricultural crops, for example, Sorghum, canola, tomato, strawberry, barley, rice, maize, and wheat. Examples of compositions can also include seed coatings for corn, soybean, canola, Sorghum, potato, rice, vegetables, cereals, and oilseeds. Seeds as provided herein can be genetically modified organisms (GMO), non-GMO, organic, or conventional. In some examples, compositions may be sprayed on the plant aerial parts, or applied to the roots by inserting into furrows in which the plant seeds are planted, watering to the soil, or dipping the roots in a suspension of the composition. In some examples, compositions may be dehydrated in a suitable manner that maintains cell viability and the ability to artificially inoculate and colonize host plants. The bacterial species may be present in compositions at a concentration of between 108 to 1010 CFU/ml. In some examples, compositions may be supplemented with trace metal ions, such as molybdenum ions, iron ions, manganese ions, or combinations of these ions. The concentration of ions in examples of compositions as described herein may between about 0.1 mM and about 50 mM. Some examples of compositions may also be formulated with a carrier, such as beta-glucan, carboxylmethyl cellulose (CMC), bacterial extracellular polymeric substance (EPS), sugar, animal milk, or other suitable carriers. In some examples, peat or planting materials can be used as a carrier, or biopolymers in which a composition is entrapped in the biopolymer can be used as a carrier. The compositions comprising the bacterial populations described herein can improve plant traits, such as promoting plant growth, maintaining high chlorophyll content in leaves, increasing fruit or seed numbers, and increasing fruit or seed unit weight.


The compositions comprising the bacterial populations described herein may be coated onto the surface of a seed. As such, compositions comprising a seed coated with one or more bacteria described herein are also contemplated. The seed coating can be formed by mixing the bacterial population with a porous, chemically inert granular carrier. Alternatively, the compositions may be inserted directly into the furrows into which the seed is planted or sprayed onto the plant leaves or applied by dipping the roots into a suspension of the composition. An effective amount of the composition can be used to populate the sub-soil region adjacent to the roots of the plant with viable bacterial growth, or populate the leaves of the plant with viable bacterial growth. In general, an effective amount is an amount sufficient to result in plants with improved traits (e.g. a desired level of nitrogen fixation).


Bacterial compositions described herein can be formulated using an agriculturally acceptable carrier. The formulation useful for these embodiments may include at least one member selected from the group consisting of a tackifier, a microbial stabilizer, a fungicide, an antibacterial agent, a preservative, a stabilizer, a surfactant, an anti-complex agent, an herbicide, a nematicide, an insecticide, a plant growth regulator, a fertilizer, a rodenticide, a dessicant, a bactericide, a nutrient, a hormone, or any combination thereof. In some examples, compositions may be shelf-stable. For example, any of the compositions described herein can include an agriculturally acceptable carrier (e.g., one or more of a fertilizer such as a non-naturally occurring fertilizer, an adhesion agent such as a non-naturally occurring adhesion agent, and a pesticide such as a non-naturally occurring pesticide). A non-naturally occurring adhesion agent can be, for example, a polymer, copolymer, or synthetic wax. For example, any of the coated seeds, seedlings, or plants described herein can contain such an agriculturally acceptable carrier in the seed coating. In any of the compositions or methods described herein, an agriculturally acceptable carrier can be or can include a non-naturally occurring compound (e.g., a non-naturally occurring fertilizer, a non-naturally occurring adhesion agent such as a polymer, copolymer, or synthetic wax, or a non-naturally occurring pesticide). Non-limiting examples of agriculturally acceptable carriers are described below. Additional examples of agriculturally acceptable carriers are known in the art.


In some cases, bacteria are mixed with an agriculturally acceptable carrier. The carrier can be a solid carrier or liquid carrier, and in various forms including microspheres, powders, emulsions and the like. The carrier may be any one or more of a number of carriers that confer a variety of properties, such as increased stability, wettability, or dispersability. Wetting agents such as natural or synthetic surfactants, which can be nonionic or ionic surfactants, or a combination thereof can be included in the composition. Water-in-oil emulsions can also be used to formulate a composition that includes the isolated bacteria (see, for example, U.S. Pat. No. 7,485,451). Suitable formulations that may be prepared include wettable powders, granules, gels, agar strips or pellets, thickeners, and the like, microencapsulated particles, and the like, liquids such as aqueous flowables, aqueous suspensions, water-in-oil emulsions, etc. The formulation may include grain or legume products, for example, ground grain or beans, broth or flour derived from grain or beans, starch, sugar, or oil.


In some embodiments, the agricultural carrier may be soil or a plant growth medium. Other agricultural carriers that may be used include water, fertilizers, plant-based oils, humectants, or combinations thereof. Alternatively, the agricultural carrier may be a solid, such as diatomaceous earth, loam, silica, alginate, clay, bentonite, vermiculite, seed cases, other plant and animal products, or combinations, including granules, pellets, or suspensions. Mixtures of any of the aforementioned ingredients are also contemplated as carriers, such as but not limited to, pesta (flour and kaolin clay), agar or flour-based pellets in loam, sand, or clay, etc. Formulations may include food sources for the bacteria, such as barley, rice, or other biological materials such as seed, plant parts, sugar cane bagasse, hulls or stalks from grain processing, ground plant material or wood from building site refuse, sawdust or small fibers from recycling of paper, fabric, or wood.


For example, a fertilizer can be used to help promote the growth or provide nutrients to a seed, seedling, or plant. Non-limiting examples of fertilizers include nitrogen, phosphorous, potassium, calcium, sulfur, magnesium, boron, chloride, manganese, iron, zinc, copper, molybdenum, and selenium (or a salt thereof). Additional examples of fertilizers include one or more amino acids, salts, carbohydrates, vitamins, glucose, NaCl, yeast extract, NH4H2PO4, (NH4)2SO4, glycerol, valine, L-leucine, lactic acid, propionic acid, succinic acid, malic acid, citric acid, KH tartrate, xylose, lyxose, and lecithin. In one embodiment, the formulation can include a tackifier or adherent (referred to as an adhesive agent) to help bind other active agents to a substance (e.g., a surface of a seed). Such agents are useful for combining bacteria with carriers that can contain other compounds (e.g., control agents that are not biologic), to yield a coating composition. Such compositions help create coatings around the plant or seed to maintain contact between the microbe and other agents with the plant or plant part. In one embodiment, adhesives are selected from the group consisting of: alginate, gums, starches, lecithins, formononetin, polyvinyl alcohol, alkali formononetinate, hesperetin, polyvinyl acetate, cephalins, Gum Arabic, Xanthan Gum, Mineral Oil, Polyethylene Glycol (PEG), Polyvinyl pyrrolidone (PVP), Arabino-galactan, Methyl Cellulose, PEG 400, Chitosan, Polyacrylamide, Polyacrylate, Polyacrylonitrile, Glycerol, Triethylene glycol, Vinyl Acetate, Gellan Gum, Polystyrene, Polyvinyl, Carboxymethyl cellulose, Gum Ghatti, and polyoxyethylene-polyoxybutylene block copolymers.


In some embodiments, the adhesives can be, e.g. a wax such as carnauba wax, beeswax, Chinese wax, shellac wax, spermaceti wax, candelilla wax, castor wax, ouricury wax, and rice bran wax, a polysaccharide (e.g., starch, dextrins, maltodextrins, alginate, and chitosans), a fat, oil, a protein (e.g., gelatin and zeins), gum arables, and shellacs. Adhesive agents can be non-naturally occurring compounds, e.g., polymers, copolymers, and waxes. For example, non-limiting examples of polymers that can be used as an adhesive agent include: polyvinyl acetates, polyvinyl acetate copolymers, ethylene vinyl acetate (EVA) copolymers, polyvinyl alcohols, polyvinyl alcohol copolymers, celluloses (e.g., ethylcelluloses, methylcelluloses, hydroxymethylcelluloses, hydroxypropylcelluloses, and carboxymethylcelluloses), polyvinylpyrolidones, vinyl chloride, vinylidene chloride copolymers, calcium lignosulfonates, acrylic copolymers, polyvinylacrylates, polyethylene oxide, acylamide polymers and copolymers, polyhydroxyethyl acrylate, methylacrylamide monomers, and polychloroprene.


In some examples, one or more of the adhesion agents, anti-fungal agents, growth regulation agents, and pesticides (e.g., insecticide) are non-naturally occurring compounds (e.g., in any combination). Additional examples of agriculturally acceptable carriers include dispersants (e.g., polyvinylpyrrolidone/vinyl acetate PVPIVA S-630), surfactants, binders, and filler agents.


The formulation can also contain a surfactant. Non-limiting examples of surfactants include nitrogen-surfactant blends such as Prefer 28 (Cenex), Surf-N(US), Inhance (Brandt), P-28 (Wilfarm) and Patrol (Helena); esterified seed oils include Sun-It II (AmCy), MSO (UAP), Scoil (Agsco), Hasten (Wilfarm) and Mes-100 (Drexel); and organo-silicone surfactants include Silwet L77 (UAP), Silikin (Terra), Dyne-Amic (Helena), Kinetic (Helena), Sylgard 309 (Wilbur-Ellis) and Century (Precision). In one embodiment, the surfactant is present at a concentration of between 0.01% v/v to 10% v/v. In another embodiment, the surfactant is present at a concentration of between 0.1% v/v to 1% v/v.


In certain cases, the formulation includes a microbial stabilizer. Such an agent can include a desiccant, which can include any compound or mixture of compounds that can be classified as a desiccant regardless of whether the compound or compounds are used in such concentrations that they in fact have a desiccating effect on a liquid inoculant. Such desiccants are ideally compatible with the bacterial population used, and should promote the ability of the microbial population to survive application on the seeds and to survive desiccation. Examples of suitable desiccants include one or more of trehalose, sucrose, glycerol, and Methylene glycol. Other suitable desiccants include, but are not limited to, non reducing sugars and sugar alcohols (e.g., mannitol or sorbitol). The amount of desiccant introduced into the formulation can range from about 5% to about 50% by weight/volume, for example, between about 10% to about 40%, between about 15% to about 35%, or between about 20% to about 30%. In some cases, it is advantageous for the formulation to contain agents such as a fungicide, an antibacterial agent, an herbicide, a nematicide, an insecticide, a plant growth regulator, a rodenticide, bactericide, or a nutrient. In some examples, agents may include protectants that provide protection against seed surface-borne pathogens. In some examples, protectants may provide some level of control of soil-borne pathogens. In some examples, protectants may be effective predominantly on a seed surface.


In some examples, a fungicide may include a compound or agent, whether chemical or biological, that can inhibit the growth of a fungus or kill a fungus. In some examples, a fungicide may include compounds that may be fungistatic or fungicidal. In some examples, fungicide can be a protectant, or agents that are effective predominantly on the seed surface, providing protection against seed surface-borne pathogens and providing some level of control of soil-borne pathogens. Non-limiting examples of protectant fungicides include captan, maneb, thiram, or fludioxonil.


In some examples, fungicide can be a systemic fungicide, which can be absorbed into the emerging seedling and inhibit or kill the fungus inside host plant tissues. Systemic fungicides used for seed treatment include, but are not limited to the following: azoxystrobin, carboxin, mefenoxam, metalaxyl, thiabendazole, trifloxystrobin, and various triazole fungicides, including difenoconazole, ipconazole, tebuconazole, and triticonazole. Mefenoxam and metalaxyl are primarily used to target the water mold fungi Pythium and Phytophthora. Some fungicides are preferred over others, depending on the plant species, either because of subtle differences in sensitivity of the pathogenic fungal species, or because of the differences in the fungicide distribution or sensitivity of the plants. In some examples, fungicide can be a biological control agent, such as a bacterium or fungus. Such organisms may be parasitic to the pathogenic fungi, or secrete toxins or other substances which can kill or otherwise prevent the growth of fungi. Any type of fungicide, particularly ones that are commonly used on plants, can be used as a control agent in a seed composition.


In some examples, the seed coating composition comprises a control agent which has antibacterial properties. In one embodiment, the control agent with antibacterial properties is selected from the compounds described herein elsewhere. In another embodiment, the compound is Streptomycin, oxytetracycline, oxolinic acid, or gentamicin. Other examples of antibacterial compounds which can be used as part of a seed coating composition include those based on dichlorophene and benzylalcohol hemi formal (Proxel® from ICI or Acticide® RS from Thor Chemie and Kathon® MK 25 from Rohm & Haas) and isothiazolinone derivatives such as alkylisothiazolinones and benzisothiazolinones (Acticide® MBS from Thor Chemie).


In some examples, growth regulator is selected from the group consisting of: Abscisic acid, amidochlor, ancymidol, 6-benzylaminopurine, brassinolide, butralin, chlormequat (chlormequat chloride), choline chloride, cyclanilide, daminozide, dikegulac, dimethipin, 2,6-dimethylpuridine, ethephon, flumetralin, flurprimidol, fluthiacet, forchlorfenuron, gibberellic acid, inabenfide, indole-3-acetic acid, maleic hydrazide, mefluidide, mepiquat (mepiquat chloride), naphthaleneacetic acid, N-6-benzyladenine, paclobutrazol, prohexadione phosphorotrithioate, 2,3,5-tri-iodobenzoic acid, trinexapac-ethyl and uniconazole. Additional non-limiting examples of growth regulators include brassinosteroids, cytokinines (e.g., kinetin and zeatin), auxins (e.g., indolylacetic acid and indolylacetyl aspartate), flavonoids and isoflavanoids (e.g., formononetin and diosmetin), phytoaixins (e.g., glyceolline), and phytoalexin-inducing oligosaccharides (e.g., pectin, chitin, chitosan, polygalacuronic acid, and oligogalacturonic acid), and gibellerins. Such agents are ideally compatible with the agricultural seed or seedling onto which the formulation is applied (e.g., it should not be deleterious to the growth or health of the plant). Furthermore, the agent is ideally one which does not cause safety concerns for human, animal or industrial use (e.g., no safety issues, or the compound is sufficiently labile that the commodity plant product derived from the plant contains negligible amounts of the compound).


Some examples of nematode-antagonistic biocontrol agents include ARF18; 30 Arthrobotrys spp.; Chaetomium spp.; Cylindrocarpon spp.; Exophilia spp.; Fusarium spp.; Gliocladium spp.; Hirsutella spp.; Lecanicillium spp.; Monacrosporium spp.; Myrothecium spp.; Neocosmospora spp.; Paecilomyces spp.; Pochonia spp.; Stagonospora spp.; vesicular-arbuscular mycorrhizal fungi, Burkholderia spp.; Pasteuria spp., Brevibacillus spp.; Pseudomonas spp.; and Rhizobacteria. Particularly preferred nematode-antagonistic biocontrol agents include ARF18, Arthrobotrys oligospora, Arthrobotrys dactyloides, Chaetomium globosum, Cylindrocarpon heteronema, Exophilia jeanselmei, Exophilia pisciphila, Fusarium aspergillus, Fusarium solani, Gliocladium catenulatum, Gliocladium roseum, Gliocladium vixens, Hirsutella rhossiliensis, Hirsutella minnesotensis, Lecanicillium lecanii, Monacrosporium drechsleri, Monacrosporium gephyropagum, Myrotehcium verrucaria, Neocosmospora vasinfecta, Paecilomyces lilacinus, Pochonia chlamydosporia, Stagonospora heteroderae, Stagonospora phaseoli, vesicular-arbuscular mycorrhizal fungi, Burkholderia cepacia, Pasteuria penetrans, Pasteuria thomei, Pasteuria nishizawae, Pasteuria ramosa, Pasteuria usage, Brevibacillus laterosporus strain G4, Pseudomonas fluorescens and Rhizobacteria.


Some examples of nutrients can be selected from the group consisting of a nitrogen fertilizer including, but not limited to Urea, Ammonium nitrate, Ammonium sulfate, Non-pressure nitrogen solutions, Aqua ammonia, Anhydrous ammonia, Ammonium thiosulfate, Sulfur-coated urea, Urea-formaldehydes, IBDU, Polymer-coated urea, Calcium nitrate, Ureaform, and Methylene urea, phosphorous fertilizers such as Diammonium phosphate, Monoammonium phosphate, Ammonium polyphosphate, Concentrated superphosphate and Triple superphosphate, and potassium fertilizers such as Potassium chloride, Potassium sulfate, Potassium-magnesium sulfate, Potassium nitrate. Such compositions can exist as free salts or ions within the seed coat composition. Alternatively, nutrients/fertilizers can be complexed or chelated to provide sustained release over time.


Some examples of rodenticides may include selected from the group of substances consisting of 2-isovalerylindan-1,3-dione, 4-(quinoxalin-2-ylamino) benzenesulfonamide, alpha-chlorohydrin, aluminum phosphide, antu, arsenous oxide, barium carbonate, bisthiosemi, brodifacoum, bromadiolone, bromethalin, calcium cyanide, chloralose, chlorophacinone, cholecalciferol, coumachlor, coumafuryl, coumatetralyl, crimidine, difenacoum, difethialone, diphacinone, ergocalciferol, flocoumafen, fluoroacetamide, flupropadine, flupropadine hydrochloride, hydrogen cyanide, iodomethane, lindane, magnesium phosphide, methyl bromide, norbormide, phosacetim, phosphine, phosphorus, pindone, potassium arsenite, pyrinuron, scilliroside, sodium arsenite, sodium cyanide, sodium fluoroacetate, strychnine, thallium sulfate, warfarin and zinc phosphide.


In the liquid form, for example, solutions or suspensions, bacterial populations can be mixed or suspended in water or in aqueous solutions. Suitable liquid diluents or carriers include water, aqueous solutions, petroleum distillates, or other liquid carriers.


Solid compositions can be prepared by dispersing the bacterial populations in and on an appropriately divided solid carrier, such as peat, wheat, bran, vermiculite, clay, talc, bentonite, diatomaceous earth, fuller's earth, pasteurized soil, and the like. When such formulations are used as wettable powders, biologically compatible dispersing agents such as non-ionic, anionic, amphoteric, or cationic dispersing and emulsifying agents can be used.


The solid carriers used upon formulation include, for example, mineral carriers such as kaolin clay, pyrophyllite, bentonite, montmorillonite, diatomaceous earth, acid white soil, vermiculite, and pearlite, and inorganic salts such as ammonium sulfate, ammonium phosphate, ammonium nitrate, urea, ammonium chloride, and calcium carbonate. Also, organic fine powders such as wheat flour, wheat bran, and rice bran may be used. The liquid carriers include vegetable oils such as soybean oil and cottonseed oil, glycerol, ethylene glycol, polyethylene glycol, propylene glycol, polypropylene glycol, etc.


Pests


Agricultural compositions of the disclosure, which may comprise any microbe taught herein, are sometimes combined with one or more pesticides.


The pesticides that are combined with the microbes of the disclosure may target any of the pests mentioned below.


“Pest” includes but is not limited to, insects, fungi, bacteria, nematodes, mites, ticks and the like. Insect pests include insects selected from the orders Coleoptera, Diptera, Hymenoptera, Lepidoptera, Mallophaga, Homoptera, Hemiptera Orthoptera, Thysanoptera, Dermaptera, Isoptera, Anoplura, Siphonaptera, Trichoptera, etc., particularly Lepidoptera and Coleoptera.


Those skilled in the art will recognize that not all compounds are equally effective against all pests. Compounds that may be combined with microbes of the disclosure may display activity against insect pests, which may include economically important agronomic, forest, greenhouse, nursery ornamentals, food and fiber, public and animal health, domestic and commercial structure, household and stored product pests.


As aforementioned, the agricultural compositions of the disclosure (which may comprise any microbe taught herein) are in embodiments combined with one or more pesticides. These pesticides may be active against any of the following pests:


Larvae of the order Lepidoptera include, but are not limited to, armyworms, cutworms, loopers and heliothines in the family Noctuidae Spodoptera frugiperda J E Smith (fall armyworm); S. exigua Hubner (beet armyworm); S. litura Fabricius (tobacco cutworm, cluster caterpillar); Mamestra configurata Walker (bertha armyworm); M. brassicae Linnaeus (cabbage moth); Agrotis ipsilon Hufnagel (black cutworm); A. orthogonia Morrison (western cutworm); A. subterranea Fabricius (granulate cutworm); Alabama argillacea Hubner (cotton leaf worm); Trichoplusia ni Hubner (cabbage looper); Pseudoplusia includens Walker (soybean looper); Anticarsia gemmatalis Hubner (velvet bean caterpillar); Hypena scabra Fabricius (green clover worm); Heliothis virescens Fabricius (tobacco budworm); Pseudaletia unipuncta Haworth (armyworm); Athetis mindara Barnes and Mcdunnough (rough skinned cutworm); Euxoa messoria Harris (darksided cutworm); Earias insulana Boisduval (spiny bollworm); E. vittella Fabricius (spotted bollworm); Helicoverpa armigera Hubner (American bollworm); H. zea Boddie (corn earworm or cotton bollworm); Melanchra picta Harris (zebra caterpillar); Egira (Xylomyges) curialis Grote (citrus cutworm); borers, case bearers, webworms, coneworms, and skeletonizers from the family Pyralidae Ostrinia nubilalis Hubner (European corn borer); Amyelois transitella Walker (naval orangeworm); Anagasta kuehniella Zeller (Mediterranean flour moth); Cadra cautella Walker (almond moth); Chilo suppressalis Walker (rice stem borer); C. partellus, (Sorghum borer); Corcyra cephalonica Stainton (rice moth); Crambus caliginosellus Clemens (corn root webworm); C. teterrellus Zincken (bluegrass webworm); Cnaphalocrocis medinalis Guenee (rice leaf roller); Desmia funeralis Hubner (grape leaffolder); Diaphania hyalinata Linnaeus (melon worm); D. nitidalis Stoll (pickleworm); Diatraea grandiosella Dyar (southwestern corn borer), D. saccharalis Fabricius (surgarcane borer); Eoreuma loftini Dyar (Mexican rice borer); Ephestia elutella Hubner (tobacco (cacao) moth); Galleria mellonella Linnaeus (greater wax moth); Herpetogramma licarsisalis Walker (sod webworm); Homoeosoma electellum Hulst (sunflower moth); Elasmopalpus lignosellus Zeller (lesser cornstalk borer); Achroia grisella Fabricius (lesser wax moth); Loxostege sticticalis Linnaeus (beet webworm); Orthaga thyrisalis Walker (tea tree web moth); Maruca testulalis Geyer (bean pod borer); Plodia interpunctella Hubner (Indian meal moth); Scirpophaga incertulas Walker (yellow stem borer); Udea rubigalis Guenee (celery leaftier); and leafrollers, budworms, seed worms and fruit worms in the family Tortricidae Acleris gloverana Walsingham (Western blackheaded budworm); A. variana Fernald (Eastern blackheaded budworm); Archips argyrospila Walker (fruit tree leaf roller); A. rosana Linnaeus (European leaf roller); and other Archips species, Adoxophyes orana Fischer von Rosslerstamm (summer fruit tortrix moth); Cochylis hospes Walsingham (banded sunflower moth); Cydia latiferreana Walsingham (filbertworm); C. pomonella Linnaeus (colding moth); Platynota flavedana Clemens (variegated leafroller); P. stultana Walsingham (omnivorous leafroller); Lobesia botrana Denis & Schiffermuller (European grape vine moth); Spilonota ocellana Denis & Schiffermuller (eyespotted bud moth); Endopiza viteana Clemens (grape berry moth); Eupoecilia ambiguella Hubner (vine moth); Bonagota salubricola Meyrick (Brazilian apple leafroller); Grapholita molesta Busck (oriental fruit moth); Suleima helianthana Riley (sunflower bud moth); Argyrotaenia spp.; Choristoneura spp.


Selected other agronomic pests in the order Lepidoptera include, but are not limited to, Alsophila pometaria Harris (fall cankerworm); Anarsia lineatella Zeller (peach twig borer); Anisota senatoria J. E. Smith (orange striped oakworm); Antheraea pernyi Guerin-Meneville (Chinese Oak Tussah Moth); Bombyx mori Linnaeus (Silkworm); Bucculatrix thurberiella Busck (cotton leaf perforator); Colias eurytheme Boisduval (alfalfa caterpillar); Datana integerrima Grote & Robinson (walnut caterpillar); Dendrolimus sibiricus Tschetwerikov (Siberian silk moth), Ennomos subsignaria Hubner (elm spanworm); Erannis tiliaria Harris (linden looper); Euproctis chrysorrhoea Linnaeus (browntail moth); Harrisina americana Guerin-Meneville (grapeleaf skeletonizer); Hemileuca oliviae Cockrell (range caterpillar); Hyphantria cunea Drury (fall web-worm); Keiferia lycopersicella Walsingham (tomato pinworm); Lambdina fiscellaria fiscellaria Hulst (Eastern hemlock looper); L. fiscellaria lugubrosa Hulst (Western hemlock looper); Leucoma salicis Linnaeus (satin moth); Lymantria dispar Linnaeus (gypsy moth); Manduca quinquemaculata Haworth (five spotted hawk moth, tomato homworm); M. sexta Haworth (tomato homworm, tobacco hornworm); Operophtera brumata Linnaeus (winter moth); Paleacrita vernata Peck (spring cankerworm); Papilio cresphontes Cramer (giant swallowtail orange dog); Phryganidia californica Packard (California oakworm); Phyllocnistis citrella Stainton (citrus leafminer); Phyllonorycter blancardella Fabricius (spotted tentiform leafminer); Pieris brassicae Linnaeus (large white butterfly); P. rapae Linnaeus (small white butterfly); P. napi Linnaeus (green veined white butterfly); Platyptilia carduidactyla Riley (artichoke plume moth); Plutella xylostella Linnaeus (diamondback moth); Pectinophora gossypiella Saunders (pink bollworm); Pontia protodice Boisduval and Leconte (Southern cabbage-worm); Sabulodes aegrotata Guenee (omnivorous looper); Schizura concinna J. E. Smith (red humped caterpillar); Sitotroga cerealella Olivier (Angoumois grain moth); Thaumetopoea pityocampa Schiffermuller (pine processionary caterpillar); Tineola bisselliella Hummel (webbing clothes moth); Tuta absoluta Meyrick (tomato leafminer); Yponomeuta padella Linnaeus (ermine moth); Heliothis subflexa Guenee; Malacosoma spp. and Orgyia spp.; Ostrinia nubilalis (European corn borer); seed corn maggot; Agrotis ipsilon (black cutworm).


Larvae and adults of the order Coleoptera including weevils from the families Anthribidae, Bruchidae and Curculionidae (including, but not limited to: Anthonomus grandis Boheman (boll weevil); Lissorhoptrus oryzophilus Kuschel (rice water weevil); Sitophilus granarius Linnaeus (granary weevil); S. oryzae Linnaeus (rice weevil); Hypera punctata Fabricius (clover leaf weevil); Cylindrocopturus adspersus LeConte (sunflower stem weevil); Smicronyx fulvus LeConte (red sunflower seed weevil); S. sordidus LeConte (gray sunflower seed weevil); Sphenophorus maidis Chittenden (maize billbug)); flea beetles, cucumber beetles, rootworms, leaf beetles, potato beetles and leafminers in the family Chrysomelidae (including, but not limited to: Leptinotarsa decemlineata Say (Colorado potato beetle); Diabrotica virgifera virgifera LeConte (western corn rootworm); D. barberi Smith and Lawrence (northern corn rootworm); D. undecimpunctata howardi Barber (southern corn rootworm); Chaetocnema pulicaria Melsheimer (corn flea beetle); Phyllotreta cruciferae Goeze (Crucifer flea beetle); Phyllotreta striolata (stripped flea beetle); Colaspis brunnea Fabricius (grape Colaspis); Oulema melanopus Linnaeus (cereal leaf beetle); Zygogramma exclamationis Fabricius (sunflower beetle)); beetles from the family Coccinellidae (including, but not limited to: Epilachna varivestis Mulsant (Mexican bean beetle)); chafers and other beetles from the family Scarabaeidae (including, but not limited to: Popillia japonica Newman (Japanese beetle); Cyclocephala borealis Arrow (northern masked chafer, white grub); C. immaculata Olivier (southern masked chafer, white grub); Rhizotrogus majalis Razoumowsky (European chafer); Phyllophaga crinita Burmeister (white grub); Ligyrus gibbosus De Geer (carrot beetle)); carpet beetles from the family Dermestidae; wireworms from the family Elateridae, Eleodes spp., Melanotus spp.; Conoderus spp.; Limonius spp.; Agriotes spp.; Ctenicera spp.; Aeolus spp.; bark beetles from the family Scolytidae and beetles from the family Tenebrionidae; Cerotoma trifurcate (bean leaf beetle); and wireworm.


Adults and immatures of the order Diptera, including leafminers Agromyza parvicornis Loew (corn blotch leafminer); midges (including, but not limited to: Contarinia sorghicola Coquillett (Sorghum midge); Mayetiola destructor Say (Hessian fly); Sitodiplosis mosellana Gehin (wheat midge); Neolasioptera murtfeldtiana Felt, (sunflower seed midge)); fruit flies (Tephritidae), Oscinella frit Linnaeus (fruit flies); maggots (including, but not limited to: Delia platura Meigen (seedcorn maggot); D. coarctata Fallen (wheat bulb fly) and other Delia spp., Meromyza americana Fitch (wheat stem maggot); Musca domestica Linnaeus (house flies); Fannia canicularis Linnaeus, F. femoralis Stein (lesser house flies); Stomoxys calcitrans Linnaeus (stable flies)); face flies, hom flies, blow flies, Chrysomya spp.; Phormia spp. and other muscoid fly pests, horse flies Tabanus spp.; bot flies Gastrophilus spp.; Oestrus spp.; cattle grubs Hypoderma spp.; deer flies Chrysops spp.; Melophagus ovinus Linnaeus (keds) and other Brachycera, mosquitoes Aedes spp.; Anopheles spp.; Culex spp.; black flies Prosimulium spp.; Simulium spp.; biting midges, sand flies, sciarids, and other Nematocera.


Adults and nymphs of the orders Hemiptera and Homoptera such as, but not limited to, adelgids from the family Adelgidae, plant bugs from the family Miridae, cicadas from the family Cicadidae, leafhoppers, Empoasca spp.; from the family Cicadellidae, planthoppers from the families Cixiidae, Flatidae, Fulgoroidea, Issidae and Delphacidae, treehoppers from the family Membracidae, psyllids from the family Psyllidae, whiteflies from the family Aleyrodidae, aphids from the family Aphididae, Phylloxera from the family Phylloxeridae, mealybugs from the family Pseudococcidae, scales from the families Asterolecanidae, Coccidae, Dactylopiidae, Diaspididae, Eriococcidae Ortheziidae, Phoenicococcidae and Margarodidae, lace bugs from the family Tingidae, stink bugs from the family Pentatomidae, cinch bugs, Blissus spp.; and other seed bugs from the family Lygaeidae, spittlebugs from the family Cercopidae squash bugs from the family Coreidae and red bugs and cotton stainers from the family Pyrrhocoridae.


Agronomically important members from the order Homoptera further include, but are not limited to: Acyrthisiphon pisum Harris (pea aphid); Aphis craccivora Koch (cowpea aphid); A. fabae Scopoli (black bean aphid); A. gossypii Glover (cotton aphid, melon aphid); A. maidiradicis Forbes (corn root aphid); A. pomi De Geer (apple aphid); A. spiraecola Patch (spirea aphid); Aulacorthum solani Kaltenbach (foxglove aphid); Chaetosiphon fragaefolii Cockerell (strawberry aphid); Diuraphis noxia Kurdjumov/Mordvilko (Russian wheat aphid); Dysaphis plantaginea Paaserini (rosy apple aphid); Eriosoma lanigerum Hausmann (woolly apple aphid); Brevicoryne brassicae Linnaeus (cabbage aphid); Hyalopterus pruni Geoffroy (mealy plum aphid); Lipaphis erysimi Kaltenbach (turnip aphid); Metopolophium dirrhodum Walker (cereal aphid); Macrosiphum euphorbiae Thomas (potato aphid); Myzus persicae Sulzer (peach potato aphid, green peach aphid); Nasonovia ribisnigri Mosley (lettuce aphid); Pemphigus spp. (root aphids and gall aphids); Rhopalosiphum maidis Fitch (corn leaf aphid); R. padi Linnaeus (bird cherry-oat aphid); Schizaphis graminum Rondani (greenbug); Sipha flava Forbes (yellow sugarcane aphid); Sitobion avenae Fabricius (English grain aphid); Therioaphis maculata Buckton (spotted alfalfa aphid); Toxoptera aurantii Boyer de Fonscolombe (black citrus aphid) and T. citricida Kirkaldy (brown citrus aphid); Melanaphis sacchari (sugarcane aphid); Adelges spp. (adelgids); Phylloxera devastatrix Pergande (pecan Phylloxera); Bemisia tabaci Gennadius (tobacco whitefly, sweetpotato whitefly); B. argentifolii Bellows & Perring (silverleaf whitefly); Dialeurodes citri Ashmead (citrus whitefly); Trialeurodes abutiloneus (bandedwinged whitefly) and T. vaporariorum Westwood (greenhouse whitefly); Empoasca fabae Harris (potato leafhopper); Laodelphax striatellus Fallen (smaller brown planthopper); Macrolestes quadrilineatus Forbes (aster leafhopper); Nephotettix cinticeps Uhler (green leafhopper); N. nigropictus Stal (rice leafhopper); Nilaparvata lugens Stal (brown planthopper); Peregrinus maidis Ashmead (corn planthopper); Sogatella furcifera Horvath (white backed planthopper); Sogatodes orizicola Muir (rice delphacid); Typhlocyba pomaria McAtee (white apple leafhopper); Erythroneoura spp. (grape leafhoppers); Magicicada septendecim Linnaeus (periodical cicada); Icerya purchasi Maskell (cottony cushion scale); Quadraspidiotus perniciosus Comstock (San Jose scale); Planococcus citri Risso (citrus mealybug); Pseudococcus spp. (other mealybug complex); Cacopsylla pyricola Foerster (pear psylla); Trioza diospyri Ashmead (persimmon psylla).


Species from the order Hemiptera include, but are not limited to: Acrosternum hilare Say (green stink bug); Anasa tristis De Geer (squash bug); Blissus leucopterus leucopterus Say (chinch bug); Corythuca gossypii Fabricius (cotton lace bug); Cyrtopeltis modesta Distant (tomato bug); Dysdercus suturellus Herrich-Schaffer (cotton stainer); Euschistus servus Say (brown stink bug); E. variolarius Palisot de Beauvais (one spotted stink bug); Graptostethus spp. (complex of seed bugs); Leptoglossus corculus Say (leaf footed pine seed bug); Lygus lineolaris Palisot de Beauvais (tarnished plant bug); L. Hesperus Knight (Western tarnished plant bug); L. pratensis Linnaeus (common meadow bug); L. rugulipennis Poppius (European tarnished plant bug); Lygocoris pabulinus Linnaeus (common green capsid); Nezara viridula Linnaeus (southern green stink bug); Oebalus pugnax Fabricius (rice stink bug); Oncopeltus fasciatus Dallas (large milk-weed bug); Pseudatomoscelis seriatus Reuter (cotton flea hopper).


Hemiptera such as, Calocoris norvegicus Gmelin (strawberry bug); Orthops campestris Linnaeus; Plesiocoris rugicollis Fallen (apple capsid); Cyrtopeltis modestus Distant (tomato bug); Cyrtopeltis notatus Distant (suckfly); Spanagonicus albofasciatus Reuter (whitemarked fleahopper); Diaphnocoris chlorionis Say (honeylocust plant bug); Labopidicola ali Knight (onion plant bug); Pseudatomoscelis seriatus Reuter (cotton fleahopper); Adelphocoris rapidus Say (rapid plant bug); Poecilocapsus lineatus Fabricius (four lined plant bug); Nysius ericae Schilling (false chinch bug); Nysius raphanus Howard (false chinch bug); Nezara viridula Linnaeus (Southern green stink bug); Eurygaster spp.; Coreidae spp.; Pyrrhocoridae spp.; Tinidae spp.; Blostomatidae spp.; Reduviidae spp. and Cimicidae spp.


Adults and larvae of the order Acari (mites) such as Aceria tosichella Keifer (wheat curl mite); Petrobia latens Muller (brown wheat mite); spider mites and red mites in the family Tetranychidae, Panonychus ulmi Koch (European red mite); Tetranychus urticae Koch (two spotted spider mite); (T. mcdanieli McGregor (McDaniel mite); T. cinnabarinus Boisduval (carmine spider mite); T. turkestani Ugarov & Nikolski (strawberry spider mite); flat mites in the family Tenuipalpidae, Brevipalpus lewisi McGregor (citrus flat mite); rust and bud mites in the family Eriophyidae and other foliar feeding mites and mites important in human and animal health, i.e., dust mites in the family Epidermoptidae, follicle mites in the family Demodicidae, grain mites in the family Glycyphagidae, ticks in the order Ixodidae. Ixodes scapularis Say (deer tick); I. holocyclus Neumann (Australian paralysis tick); Dermacentor variabilis Say (American dog tick); Amblyomma americanum Linnaeus (lone star tick) and scab and itch mites in the families Psoroptidae, Pyemotidae and Sarcoptidae.


Insect pests of the order Thysanura, such as Lepisma saccharina Linnaeus (silverfish); Thermobia domestica Packard (firebrat).


Additional arthropod pests include: spiders in the order Araneae such as Loxosceles reclusa Gertsch and Mulaik (brown recluse spider) and the Latrodectus mactans Fabricius (black widow spider) and centipedes in the order Scutigeromorpha such as Scutigera coleoptrata Linnaeus (house centipede).


Superfamily of stink bugs and other related insects including but not limited to species belonging to the family Pentatomidae (Nezara viridula, Halyomorpha halys, Piezodorus guildini, Euschistus servus, Acrosternum hilare, Euschistus heros, Euschistus tristigmus, Acrosternum hilare, Dichelops furcatus, Dichelops melacanthus, and Bagrada hilaris (Bagrada Bug)), the family Plataspidae (Megacopta cribraria—Bean plataspid) and the family Cydnidae (Scaptocoris castanea—Root stink bug) and Lepidoptera species including but not limited to: diamond-back moth, e.g., Helicoverpa zea Boddie; soybean looper, e.g., Pseudoplusia includens Walker and velvet bean caterpillar e.g., Anticarsia gemmatalis Huber.


Nematodes include parasitic nematodes such as root-knot, cyst and lesion nematodes, including Heterodera spp., Meloidogyne spp. and Globodera spp.; particularly members of the cyst nematodes, including, but not limited to, Heterodera glycines (soybean cyst nematode); Heterodera schachtii (beet cyst nematode); Heterodera avenae (cereal cyst nematode) and Globodera rostochiensis and Globodera pailida (potato cyst nematodes). Lesion nematodes include Pratylenchus spp.


Pesticidal Compositions Comprising a Pesticide and Microbe of the Disclosure


As aforementioned, agricultural compositions of the disclosure, which may comprise any microbe taught herein, are sometimes combined with one or more pesticides. Pesticides can include herbicides, insecticides, fungicides, nematicides, etc.


In some embodiments the pesticides/microbial combinations can be applied in the form of compositions and can be applied to the crop area or plant to be treated, simultaneously or in succession, with other compounds. These compounds can be fertilizers, weed killers, cryoprotectants, surfactants, detergents, pesticidal soaps, dormant oils, polymers, and/or time release or biodegradable carrier formulations that permit long term dosing of a target area following a single application of the formulation. They can also be selective herbicides, chemical insecticides, virucides, microbicides, amoebicides, pesticides, fungicides, bacteriocides, nematicides, molluscicides or mixtures of several of these preparations, if desired, together with further agriculturally acceptable carriers, surfactants or application promoting adjuvants customarily employed in the art of formulation. Suitable carriers (i.e. agriculturally acceptable carriers) and adjuvants can be solid or liquid and correspond to the substances ordinarily employed in formulation technology, e.g. natural or regenerated mineral substances, solvents, dispersants, wetting agents, sticking agents, tackifiers, binders or fertilizers. Likewise the formulations may be prepared into edible baits or fashioned into pest traps to permit feeding or ingestion by a target pest of the pesticidal formulation.


Exemplary chemical compositions, which may be combined with the microbes of the disclosure, include:

    • Fruits/Vegetables Herbicides: Atrazine, Bromacil, Diuron, Glyphosate, Linuron, Metribuzin, Simazine, Trifluralin, Fluazifop, Glufosinate, Halo sulfuron Gowan, Paraquat, Propyzamide, Sethoxydim, Butafenacil, Halosulfuron, Indaziflam; Fruits/Vegetables Insecticides: Aldicarb, Bacillus thuringiensis, Carbaryl, Carbofuran, Chlorpyrifos, Cypermethrin, Deltamethrin, Diazinon, Malathion, Abamectin, Cyfluthrin/betacyfluthrin, Esfenvalerate, Lambda-cyhalothrin, Acequinocyl, Bifenazate, Methoxyfenozide, Novaluron, Chromafenozide, Thiacloprid, Dinotefuran, FluaCrypyrim, Tolfenpyrad, Clothianidin, Spirodiclofen, Gamma-cyhalothrin, Spiromesifen, Spinosad, Rynaxypyr, Cyazypyr, Spinoteram, Triflumuron, Spirotetramat, Imidacloprid, Flubendiamide, Thiodicarb, Metaflumizone, Sulfoxaflor, Cyflumetofen, Cyanopyrafen, Imidacloprid, Clothianidin, Thiamethoxam, Spinotoram, Thiodicarb, Flonicamid, Methiocarb, Emamectin benzoate, Indoxacarb, Forthiazate, Fenamiphos, Cadusaphos, Pyriproxifen, Fenbutatin oxide, Hexthiazox, Methomyl, 4-[[(6-Chlorpyridin-3-yl)methyl](2,2-difluorethyl)amino]furan-2(5H)-on; Fruits Vegetables Fungicides: Carbendazim, Chlorothalonil, EBDCs, Sulphur, Thiophanate-methyl, Azoxystrobin, Cymoxanil, Fluazinam, Fosetyl, Iprodione, Kresoxim-methyl, Metalaxyl/mefenoxam, Trifloxystrobin, Ethaboxam, Iprovalicarb, Trifloxystrobin, Fenhexamid, Oxpoconazole fumarate, Cyazofamid, Fenamidone, Zoxamide, Picoxystrobin, Pyraclostrobin, Cyflufenamid, Boscalid;
    • Cereals Herbicides: Isoproturon, Bromoxynil, loxynil, Phenoxies, Chlorsulfuron, Clodinafop, Diclofop, Diflufenican, Fenoxaprop, Florasulam, Fluoroxypyr, Metsulfuron, Triasulfuron, Flucarbazone, lodosulfuron, Propoxycarbazone, Picolin-afen, Mesosulfuron, Beflubutamid, Pinoxaden, Amidosulfuron, Thifensulfuron Methyl, Tribenuron, Flupyrsulfuron, Sulfosulfuron, Pyrasulfotole, Pyroxsulam, Flufenacet, Tralkoxydim, Pyroxasulfon; Cereals Fungicides: Carbendazim, Chlorothalonil, Azoxystrobin, Cyproconazole, Cyprodinil, Fenpropimorph, Epoxiconazole, Kresoxim-methyl, Quinoxyfen, Tebuconazole, Trifloxystrobin, Simeconazole, Picoxystrobin, Pyraclostrobin, Dimoxystrobin, Prothioconazole, Fluoxastrobin; Cereals Insecticides: Dimethoate, Lambda-cyhalothrin, Deltamethrin, alpha-Cypermethrin, β-cyfluthrin, Bifenthrin, Imidacloprid, Clothianidin, Thiamethoxam, Thiacloprid, Acetamiprid, Dinetofuran, Clorphyriphos, Metamidophos, Oxidemethon methyl, Pirimicarb, Methiocarb;
    • Maize Herbicides: Atrazine, Alachlor, Bromoxynil, Acetochlor, Dicamba, Clopyralid, S-Dimethenamid, Glufosinate, Glyphosate, Isoxaflutole, S-Metolachlor, Mesotrione, Nicosulfuron, Primisulfuron, Rimsulfuron, Sulcotrione, Foramsulfuron, Topramezone, Tembotrione, Saflufenacil, Thiencarbazone, Flufenacet, Pyroxasulfon; Maize Insecticides: Carbofuran, Chlorpyrifos, Bifenthrin, Fipronil, Imidacloprid, Lambda-Cyhalothrin, Tefluthrin, Terbufos, Thiamethoxam, Clothianidin, Spiromesifen, Flubendiamide, Triflumuron, Rynaxypyr, Deltamethrin, Thiodicarb, O-Cyfluthrin, Cypermethrin, Bifenthrin, Lufenuron, Triflumoron, Tefluthrin, Tebupirim-phos, Ethiprole, Cyazypyr, Thiacloprid, Acetamiprid, Dinetofuran, Avermectin, Methiocarb, Spirodiclofen, Spirotetramat; Maize Fungicides: Fenitropan, Thiram, Prothioconazole, Tebuconazole, Trifloxystrobin;
    • Rice Herbicides: Butachlor, Propanil, Azimsulfuron, Bensulfuron, Cyhalo-fop, Daimuron, Fentrazamide, Imazosulfuron, Mefenacet, Oxaziclomefone, Pyrazosulfuron, Pyributicarb, Quinclorac, Thiobencarb, Indanofan, Flufenacet, Fentrazamide, Halosulfuron, Oxaziclomefone, Benzobicyclon, Pyriftalid, Penoxsulam, Bispyribac, Oxadiargyl, Ethoxysulfuron, Pretilachlor, Mesotrione, Tefuryltrione, Oxadiazone, Fenoxaprop, Pyrimisulfan; Rice Insecticides: Diazinon, Fenitro-thion, Fenobucarb, Monocrotophos, Benfuracarb, Buprofezin, Dinotefuran, Fipronil, Imidacloprid, Isoprocarb, Thiacloprid, Chromafenozide, Thiacloprid, Dinotefuran, Clothianidin, Ethiprole, Flubendiamide, Rynaxypyr, Deltamethrin, Acetamiprid, Thiamethoxam, Cyazypyr, Spinosad, Spinotoram, Emamectin-Benzoate, Cypermethrin, Chlorpyriphos, Cartap, Methamidophos, Etofen-prox, Triazophos, 4-[[(6-Chlorpyridin-3-yl)methyl](2,2-difluorethyl)amino]furan-2(5H)-on, Carbofuran, Benfuracarb; Rice Fungicides: Thiophanate-methyl, Azoxystrobin, Carpropamid, Edifenphos, Ferimzone, Iprobenfos, Isoprothiolane, Pencycuron, Probenazole, Pyroquilon, Tricyclazole, Trifloxystrobin, Diclocymet, Fenoxanil, Simeconazole, Tiadinil;
    • Cotton Herbicides: Diuron, Fluometuron, MSMA, Oxyfluorfen, Prometryn, Trifluralin, Carfentrazone, Clethodim, Fluazifop-butyl, Glyphosate, Norflurazon, Pendimethalin, Pyrithiobac-sodium, Trifloxysulfuron, Tepraloxydim, Glufosinate, Flumioxazin, Thidiazuron; Cotton Insecticides: Acephate, Aldicarb, Chlorpyrifos, Cypermethrin, Deltamethrin, Malathion, Monocrotophos, Abamectin, Acetamiprid, Emamectin Benzoate, Imidacloprid, Indoxacarb, Lambda-Cyhalothrin, Spinosad, Thiodicarb, Gamma-Cyhalothrin, Spiromesifen, Pyridalyl, Flonicamid, Flubendiamide, Triflumuron, Rynaxypyr, Beta-Cyfluthrin, Spirotetramat, Clothianidin, Thiamethoxam, Thiacloprid, Dinetofuran, Flubendiamide, Cyazypyr, Spinosad, Spinotoram, gamma Cyhalothrin, 4-[[(6-Chlorpyridin-3-yl) methyl](2,2-difluorethyl)amino]furan-2(5H)-on, Thiodicarb, Avermectin, Flonicamid, Pyridalyl, Spiromesifen, Sulfoxaflor, Profenophos, Thriazophos, Endosulfan; Cotton Fungicides: Etridiazole, Metalaxyl, Quintozene;
    • Soybean Herbicides: Alachlor, Bentazone, Trifluralin, Chlorimuron-Ethyl, Cloransulam-Methyl, Fenoxaprop, Fomesafen, Flu-azifop, Glyphosate, Imazamox, Imazaquin, Imazethapyr, (S-)Metolachlor, Metribuzin, Pendimethalin, Tepraloxydim, Glufosinate; Soybean Insecticides: Lambda-cyhalothrin, Methomyl, Parathion, Thiocarb, Imidacloprid, Clothianidin, Thiamethoxam, Thiacloprid, Acetamiprid, Dinetofuran, Flubendiamide, Rynaxypyr, Cyazypyr, Spinosad, Spinotoram, Emamectin-Benzoate, Fipronil, Ethiprole, Deltamethrin, β-Cyfluthrin, gamma and lambda Cyhalothrin, 4-[[(6-Chlorpyridin-3-yl)methyl] (2,2-difluorethyl)amino]furan-2(5H)-on, Spirotetramat, Spinodiclofen, Triflumuron, Flonicamid, Thiodicarb, beta-Cyfluthrin; Soybean Fungicides: Azoxystrobin, Cyproconazole, Epoxiconazole, Flutriafol, Pyraclostrobin, Tebuconazole, Trifloxystrobin, Prothioconazole, Tetraconazole;
    • Sugarbeet Herbicides: Chloridazon, Desmedipham, Ethofumesate, Phenmedipham, Triallate, Clopyralid, Fluazifop, Lenacil, Metamitron, Quinmerac, Cycloxydim, Triflusulfuron, Tepral-oxydim, Quizalofop; Sugarbeet Insecticides: Imidacloprid, Clothianidin, Thiamethoxam, Thiacloprid, Acetamiprid, Dinetofuran, Deltamethrin, β-Cyfluthrin, gamma/lambda Cyhalothrin, 4-[[(6-Chlorpyridin-3-yl)methyl](2,2-difluor-ethyl)amino]furan-2(5H)-on, Tefluthrin, Rynaxypyr, Cyaxypyr, Fipronil, Carbofuran;
    • Canola Herbicides: Clopyralid, Diclofop, Fluazifop, Glufosinate, Glyphosate, Metazachlor, Trifluralin Ethametsulfuron, Quinmerac, Quizalofop, Clethodim, Tepraloxydim; Canola Fungicides: Azoxystrobin, Carbendazim, Fludioxonil, Iprodione, Prochloraz, Vinclozolin; Canola Insecticides: Carbofuran organophos-phates, Pyrethroids, Thiacloprid, Deltamethrin, Imidacloprid, Clothianidin, Thiamethoxam, Acetamiprid, Dineto-furan, 0-Cyfluthrin, gamma and lambda Cyhalothrin, tau-Fluvaleriate, Ethiprole, Spinosad, Spinotoram, Flubendiamide, Rynaxypyr, Cyazypyr, 4-[[(6-Chlorpyridin-3-yl)methyl] (2,2-difluorethyl)amino] furan-2(5H)-on.


      Insecticidal Compositions Comprising an Insecticide and Microbe of the Disclosure


As aforementioned, agricultural compositions of the disclosure, which may comprise any microbe taught herein, are sometimes combined with one or more insecticides.


In some embodiments, insecticidal compositions may be included in the compositions set forth herein, and can be applied to a plant(s) or a part(s) thereof simultaneously or in succession, with other compounds. Insecticides include ammonium carbonate, aqueous potassium silicate, boric acid, copper sulfate, elemental sulfur, lime sulfur, sucrose octanoate esters, 4-[[(6-Chlorpyridin-3-yl)methyl](2,2-difluorethyl)amino]furan-2(5H)-on, abamectin, notenone, fenazaquin, fenpyroximate, pyridaben, pyrimedifen, tebufenpyrad, tolfenpyrad, acephate, emamectin benzoate, lepimectin, milbemectin, hdroprene, kinoprene, methoprene, fenoxycarb, pyriproxyfen, methryl bromide and other alkyl halides, fulfuryl fluoride, chloropicrin, borax, disodium octaborate, sodium borate, sodium metaborate, tartar emetic, dazomet, metam, pymetrozine, pyrifluquinazon, flofentezine, diflovidazin, hexythiazox, bifenazate, thiamethoxam, imidacloprid, fenpyroximate, azadirachtin, permethrin, esfenvalerate, acetamiprid, bifenthrin, indoxacarb, azadirachtin, pyrethrin, imidacloprid, beta-cyfluthrin, sulfotep, tebupirimfos, temephos, terbufos, tetrachlorvinphos, thiometon, triazophos, alanycarb, aldicarb, bendiocarb, benfluracarb, butocarboxim, butoxycarboxim, carbaryl, carbofuran, carbosulfan, ethiofencarb, fenobucarb, formetanate, furathiocarb, isoprocarb, methiocarb, methymyl, metolcarb, oxamyl, primicarb, propoxur, thiodicarb, thiofanox, triazamate, trimethacarb, XMC, xylylcarb, acephate, azamethiphos, azinphos-ethyl, azinphos-methyl, cadusafos, chlorethoxyfox, trichlorfon, vamidothion, chlordane, endosulfan, ethiprole, fipronil, acrinathrin, allethrin, bifenthrin, bioallethrin, bioalletherin X-cyclopentenyl, bioresmethrin, cyclorothrin, cyfluthrin, cyhalothrin, cypermethrin, cyphenothrin [(1R)-trans-isomers], deltamethrin, empenthrin [(EZ)-(1R)-isomers], esfenvalerate, etofenprox, fenpropathrin, fenvalerate, flucythrinate, flumethrin, halfenprox, kadathrin, phenothrin [(1R)-trans-isomer] prallethrin, pyrethrins (pyrethrum), resmethrin, silafluofen, tefluthrin, tetramethrin, tetramethrin [(1R)-isomers], tralomethrin, transfluthrin, alpha-cypermethrin, beta-cyfluthrin, beta-cypermethrin, d-cis-trans allethrin, d-trans allethrin, gamma-cyhalothrin, lamda-cyhalothrin, tau-fluvalinate, theta-cypermethrin, zeta-cypermethrin, methoxychlor, nicotine, sulfoxaflor, acetamiprid, clothianidin, dinotefuran, imidacloprid, nitenpyram, thiacloprid, thiamethoxan, tebuprimphos, beta-cyfluthrin, clothianidin, flonicamid, hydramethylnon, amitraz, flubendiamide, blorantraniliprole, lambda cyhalothrin, spinosad, gamma cyhalothrin, Beauveria bassiana, capsicum oleoresin extract, garlic oil, carbaryl, chlorpyrifos, sulfoxaflor, lambda cyhalothrin, Chlorfenvinphos, Chlormephos, Chlorpyrifos, Chlorpyrifos-methyl, Coumaphos, Cyanophos, Demeton-S-methyl, Diazinon, Dichlorvos/DDVP, Dicrotophos, Dimethoate, Dimethylvinphos, Disulfoton, EPN, Ethion, Ethoprophos, Famphur, Fenamiphos, Fenitrothion, Fenthion, Fosthiazate, Heptenophos, Imicyafos, Isofenphos, Isopropyl O-(methoxyaminothio-phosphoryl) salicylate, Isoxathion, Malathion, Mecarbam, Methamidophos, Methidathion, Mevinphos, Monocrotophos, Naled, Omethoate, Oxydemeton-methyl, Parathion, Parathion-methyl, Phenthoate, Phorate, Phosalone, Phosmet, Phosphamidon, Phoxim, Pirimiphos-methyl, Profenofos, Propetamphos, Prothiofos, Pyraclofos, Pyridaphenthion, Quinalphosfluacrypyrim, tebufenozide, chlorantraniliprole, Bacillus thuringiensis subs. Kurstaki, terbufos, mineral oil, fenpropathrin, metaldehyde, deltamethrin, diazinon, dimethoate, diflubenzuron, pyriproxyfen, rosemary oil, peppermint oil, geraniol, azadirachtin, piperonyl butoxide, cyantraniliprole, alpha cypermethrin, tefluthrin, pymetrozine, malathion, Bacillus thuringiensis subsp. israelensis, dicofol, bromopropylate, benzoximate, azadirachtin, flonicamid, soybean oil, Chromobacterium subtsugae strain PRAA4-1, zeta cypermethrin, phosmet, methoxyfenozide, paraffinic oil, spirotetramat, methomyl, Metarhizium anisopliae strain F52, ethoprop, tetradifon, propargite, fenbutatin oxide, azocyclotin, cyhexatin, diafenthiuron, Bacillus sphaericus, etoxazole, flupyradifurone, azadirachtin, Beauveria bassiana, cyflumetofen, azadirachtin, chinomethionat, acephate, Isaria fumosorosea Apopka strain 97, sodium tetraborohydrate decahydrate, emamectin benzoate, cryolite, spinetoram, Chenopodium ambrosioides extract, novaluron, dinotefuran, carbaryl, acequinocyl, flupyradifurone, iron phosphate, kaolin, buprofezin, cyromazine, chromafenozide, halofenozide, methoxyfenozide, tebufenozide, bistrifluron, chlorfluazuron, diflubenzuron, flucycloxuron, flufenoxuron, hexaflumuron, lufenuron, nocaluron, noviflumuron, teflubenzuron, triflumuron, bensultap, cartap hydrochloride, thiocyclam, thiosultap-sodium, DNOC, chlorfenapyr, sulfuramid, phorate, tolfenpyrad, sulfoxaflor, neem oil, Bacillus thuringiensis subsp. tenebrionis strain SA-10, cyromazine, heat-killed Burkholderia spp., cyantraniliprole, cyenopyrafen, cyflumetofen, sodium cyanide, potassium cyanide, calcium cyanide, aluminum phosphide, calcium phosphide, phosphine, zinc phosphide, spriodiclofen, spiromesifen, spirotetramat, metaflumizone, flubendiamide, pyflubumide, oxamyl, Bacillus thuringiensis subsp. aizawai, etoxazole, and esfenvalerate









TABLE 9







Exemplary insecticides associated with various modes of action,


which can be combined with microbes of the disclosure













Physio-





logical





func-



Compound

tion(s)


Mode of Action
class
Exemplary insecticides
affected





acetyl-
carbamates
Alanycarb, Aldicarb,
Nerve


cholinesterase

Bendiocarb, Benfuracarb,
and


(AChE)

Butocarboxim,
muscle


inhibitors

Butoxycarboxim, Carbaryl,





Carbofuran, Carbosulfan,





Ethiofencarb, Fenobucarb,





Formetanate, Furathiocarb,





Isoprocarb, Methiocarb,





Methomyl, Metolcarb,





Oxamyl, Pirimicarb, Propoxur,





Thiodicarb, Thiofanox,





Triazamate, Trimethacarb,





XMC, Xylylcarb



acetyl-
organo-
Acephate, Azamethiphos,
Nerve


cholinesterase
phosphates
Azinphos-ethyl, Azinphos-
and


(AChE)

methyl, Cadusafos,
muscle


inhibitors

Chlorethoxyfos,





Chlorfenvinphos,





Chlormephos, Chlorpyrifos,





Chlorpyrifos-methyl,





Coumaphos, Cyanophos,





Demeton-S-methyl, Diazinon,





Dichlorvos/DDVP,





Dicrotophos, Dimethoate,





Dimethylvinphos, Disulfoton,





EPN, Ethion, Ethoprophos,





Famphur, Fenamiphos,





Fenitrothion, Fenthion,





Fosthiazate, Heptenophos,





Imicyafos, Isofenphos,





Isopropyl O-





(methoxyaminothio-





phosphoryl) salicylate,





Isoxathion, Malathion,





Mecarbam, Methamidophos,





Methidathion, Mevinphos,





Monocrotophos, Naled,





Omethoate, Oxydemeton-





methyl, Parathion, Parathion-





methyl, Phenthoate, Phorate,





Phosalone, Phosmet,





Phosphamidon, Phoxim,





Pirimiphos-methyl,





Profenofos, Propetamphos,





Prothiofos, Pyraclofos,





Pyridaphenthion, Quinalphos,





Sulfotep, Tebupirimfos,





Temephos, Terbufos,





Tetrachlorvinphos, Thiometon,





Triazophos, Trichlorfon,





Vamidothion



GABA-gated
cyclodiene
Chlordane, Endosulfan
Nerve


chloride channel
organo-

and


blockers
chlorines

muscle


GABA-gated
phenyl-
Ethiprole, Fipronil
Nerve


chloride channel
pyrazoles

and


blockers
(Fiproles)

muscle


sodium channel
pyrethroids,
Acrinathrin, Allethrin,
Nerve


modulators
pyrethrins
Bifenthrin, Bioallethrin,
and




Bioallethrin S-cyclopentenyl,
muscle




Bioresmethrin, Cycloprothrin,





Cyfluthrin, Cyhalothrin,





Cypermethrin, Cyphenothrin





[(1R)-trans-isomers],





Deltamethrin, Empenthrin





[(EZ)-(1R)-isomers],





Esfenvalerate, Etofenprox,





Fenpropathrin, Fenvalerate,





Flucythrinate, Flumethrin,





Halfenprox, Kadathrin,





Phenothrin [(1R)-trans-





isomer], Prallethrin, Pyrethrins





(pyrethrum), Resmethrin,





Silafluofen, Tefluthrin,





Tetramethrin, Tetramethrin





[(1R)-isomers], Tralomethrin,





Transfluthrin, alpha-





Cypermethrin, beta-Cyfluthrin,





beta-Cypermethrin, d-cis-trans





Allethrin, d-trans Allethrin,





gamma-Cyhalothrin, lambda-





Cyhalothrin, tau-Fluvalinate,





theta-Cypermethrin, zeta-





Cypermethrin



sodium channel
DDT,
DDT, methoxychlor
Nerve


modulators
methoxychlor

and





muscle


nicotinic
neo-
Acetamiprid, Clothianidin,
Nerve


acetylcholine
nicotinoids
Dinotefuran, Imidacloprid,
and


receptor

Nitenpyram, Thiacloprid,
muscle


(nAChR)

Thiamethoxam



competitive





modulators





nicotinic
nicotine
nicotine
Nerve


acetylcholine


and


receptor


muscle


(nAChR)





competitive





modulators





nicotinic
sulfoximines
sulfoxaflor
Nerve


acetylcholine


and


receptor


muscle


(nAChR)





competitive





modulators





nicotinic
butenolides
Flupyradifurone
Nerve


acetylcholine


and


receptor


muscle


(nAChR)





competitive





modulators





nicotinic
spinosyns
Spinetoram, Spinosad
Nerve


acetylcholine


and


receptor


muscle


(nAChR)





allosteric





modulators





Glutamate-gated
avermectins,
Abamectin, Emamectin
Nerve


chloride channel
milbemycins
benzoate, Lepimectin,
and


(GluCl)

Milbemectin
muscle


allosteric





modulators





juvenile
juvenile
Hydroprene, Kinoprene,
Growth


hormone
hormone
Methoprene



mimics
analogues




juvenile
Fenoxycarb
Fenoxycarb
Growth


hormone





mimics





juvenile
Pyriproxyfen
Pyriproxyfen
Growth


hormone





mimics





miscellaneous
alkyl halides
Methyl bromide and other
Un-


non-specific

alkyl halides
known


(multi-site)


or non-


inhibitors


specific


miscellaneous
Chloropicrin
Chloropicrin
Un-


non-specific


known


(multi-site)


or non-


inhibitors


specific


miscellaneous
fluorides
Cryolite, sulfuryl fluoride
Un-


non-specific


known


(multi-site)


or non-


inhibitors


specific


miscellaneous
borates
Borax, Boric acid, Disodium
Un-


non-specific

octaborate, Sodium borate,
known


(multi-site)

Sodium metaborate
or non-


inhibitors


specific


miscellaneous
tartar emetic
tartar emetic
Un-


non-specific


known


(multi-site)


or non-


inhibitors


specific


miscellaneous
methyl
Dazomet, Metam
Un-


non-specific
iso-

known


(multi-site)
thiocyanate

or non-


inhibitors
generators

specific


modulators of
Pyridine
Pymetrozine, Pyrifluquinazon
Nerve


chordotonal
azomethine

and


organs
derivatives

muscle


mite growth
Clofentezine,
Clofentezine, Diflovidazin,
Growth


inhibitors
Diflovidazin,
Hexythiazox




Hexythiazox




mite growth
Etoxazole
Etoxazole
Growth


inhibitors





microbial

Bacillus


Bt var. aizawai, Bt var.

Midgut


disruptors of

thuringiensis


israelensis, Bt var. kurstaki,




insect midgut
and the

Bt var. tenebrionensis




membranes
insecticidal





proteins they





produce




microbial

Bacillus


Bacillus sphaericus

Midgut


disruptors of

sphaericus





insect midgut





membranes





inhibitors of
Diafenthiuron
Diafenthiuron
Res-


mitochondrial


piration


ATP synthase





inhibitors of
organotin
Azocyclotin, Cyhexatin,
Res-


mitochondrial
miticides
Fenbutatin oxide
piration


ATP synthase





inhibitors of
Propargite
Propargite
Res-


mitochondrial


piration


ATP synthase





inhibitors of
Tetradifon
Tetradifon
Res-


mitochondrial


piration


ATP synthase





uncouplers of
Chlorfenapyr,
Chlorfenapyr, DNOC,
Res-


oxidative
DNOC,
Sulfuramid
piration


phosphorylation
Sulfuramid




via disruption





of the proton





gradient





Nicotinic
nereistoxin
Bensultap, Cartap
Nerve


acetylcholine
analogues
hydrochloride, Thiocyclam,
and


receptor

Thiosultap-sodium
muscle


(nAChR)





channel





blockers





inhibitors of
benzoylureas
Bistrifluron, Chlorfluazuron,
Growth


chitin

Diflubenzuron, Flucycloxuron,



biosynthesis,

Flufenoxuron, Hexaflumuron,



type 0

Lufenuron, Novaluron,





Noviflumuron, Teflubenzuron,





Triflumuron



inhibitors of
Buprofezin
Buprofezin
Growth


chitin





biosynthesis,





type 1





moulting
Cyromazine
Cyromazine
Growth


disruptor,





Dipteran





ecdysone
diacyl-
Chromafenozide,
Growth


receptor
hydrazines
Halofenozide,



agonists

Methoxyfenozide,





Tebufenozide



octopamine
Amitraz
Amitraz
Nerve


receptor


and


agonists


muscle


mitochondrial
Hydra-
Hydramethylnon
Res-


complex III
methylnon

piration


electron trans-





port inhibitors





mitochondrial
Acequinocyl
Acequinocyl
Res-


complex III


piration


electron trans-





port inhibitors





mitochondrial
Fluacrypyrim
Fluacrypyrim
Res-


complex III


piration


electron trans-





port inhibitors





mitochondrial
Bifenazate
Bifenazate
Res-


complex III


piration


electron trans-





port inhibitors





mitochondrial
Meti
Fenazaquin, Fenpyroximate,
Res-


complex I
acaricides and
Pyridaben, Pyrimidifen,
piration


electron trans-
insecticides
Tebufenpyrad, Tolfenpyrad



port inhibitors





mitochondrial
Rotenone
Rotenone
Res-


complex I


piration


electron trans-





port inhibitors





voltage-
oxadiazines
Indoxacarb
Nerve


dependent


and


sodium channel


muscle


blockers





voltage-
semi-
Metaflumizone
Nerve


dependent
carbazones

and


sodium channel


muscle


blockers





inhibitors of
tetronic and
Spirodiclofen, Spiromesifen,
Growth


acetyl CoA
tetramic acid
Spirotetramat



carboxylase
derivatives




mitochondrial
phosphides
Aluminium phosphide,
Res-


complex IV

Calcium phosphide,
piration


electron trans-

Phosphine, Zinc phosphide



port inhibitors





mitochondrial
cyanides
Calcium cyanide, Potassium
Res-


complex IV

cyanide, Sodium cyanide
piration


electron trans-





port inhibitors





mitochondrial
beta-
Cyenopyrafen, Cyflumetafen
Res-


complex II
ketonitrile

piration


electron trans-
derivatives




port inhibitors





mitochondrial
carboxanilides
Pyflubumide
Res-


complex II


piration


electron trans-





port inhibitors





ryanodine
diamides
Chlorantraniliprole,
Nerve


receptor

Cyantraniliprole,
and


modulators

Flubendiamide
muscle


Chordotonal
Flonicamid
Flonicamid
Nerve


organ


and


modulators-


muscle


undefined target





site





compounds of
Azadirachtin
Azadirachtin
Un-


unknown or


known


uncertain mode





of action





compounds of
Benzoximate
Benzoximate
Un-


unknown or


known


uncertain mode





of action





compounds of
Bromo-
Bromopropylate
Un-


unknown or
propylate

known


uncertain mode





of action





compounds of
Chino-
Chinomethionat
Un-


unknown or
methionat

known


uncertain mode





of action





compounds of
Dicofol
Dicofol
Un-


unknown or


known


uncertain mode





of action





compounds of
lime sulfur
lime sulfur
Un-


unknown or


known


uncertain mode





of action





compounds of
Pyridalyl
Pyridalyl
Un-


unknown or


known


uncertain mode





of action





compounds of
sulfur
sulfur
Un-


unknown or


known


uncertain mode





of action
















TABLE 10







Exemplary list of pesticides, which can be combined with microbes


of the disclosure








Category
Compounds





INSECTICIDES



arsenical insecticides
calcium arsenate



copper acetoarsenite



copper arsenate



lead arsenate



potassium arsenite



sodium arsenite


botanical insecticides
allicin



anabasine



azadirachtin



carvacrol



d-limonene



matrine



nicotine



nornicotine



oxymatrine



pyrethrins



cinerins



cinerin I



cinerin II



jasmolin I



jasmolin II



pyrethrin I



pyrethrin II



quassia



rhodojaponin-III



rotenone



ryania



sabadilla



sanguinarine



triptolide


carbamate insecticides
bendiocarb



carbaryl


benzofuranyl methylcarbamate
benfuracarb


insecticides
carbofuran



carbosulfan



decarbofuran



furathiocarb


dimethylcarbamate insecticides
dimetan



dimetilan



hyquincarb



isolan



pirimicarb



pyramat



pyrolan


oxime carbamate insecticides
alanycarb



aldicarb



aldoxycarb



butocarboxim



butoxycarboxim



methomyl



nitrilacarb



oxamyl



tazimcarb



thiocarboxime



thiodicarb



thiofanox


phenyl methylcarbamate insecticides
allyxycarb



aminocarb



bufencarb



butacarb



carbanolate



cloethocarb



CPMC



dicresyl



dimethacarb



dioxacarb



EMPC



ethiofencarb



fenethacarb



fenobucarb



isoprocarb



methiocarb



metolcarb



mexacarbate



promacyl



promecarb



propoxur



trimethacarb



XMC



xylylcarb


diamide insecticides
broflanilide



chlorantraniliprole



cyantraniliprole



cyclaniliprole



cyhalodiamide



flubendiamide



tetraniliprole


dinitrophenol insecticides
dinex



dinoprop



dinosam



DNOC


fluorine insecticides
barium hexafluorosilicate



cryolite



flursulamid



sodium fluoride



sodium hexafluorosilicate



sulfluramid


formamidine insecticides
amitraz



chlordimeform



formetanate



formparanate



medimeform



semiamitraz


fumigant insecticides
acrylonitrile



carbon disulfide



carbon tetrachloride



carbonyl sulfide



chloroform



chloropicrin



cyanogen



para-dichlorobenzene



1,2-dichloropropane



dithioether



ethyl formate



ethylene dibromide



ethylene dichloride



ethylene oxide



hydrogen cyanide



methyl bromide



methyl iodide



methylchloroform



methylene chloride



naphthalene



phosphine



sodium tetrathiocarbonate



sulfuryl fluoride



tetrachloroethane


inorganic insecticides
borax



boric acid



calcium polysulfide



copper oleate



diatomaceous earth



mercurous chloride



potassium thiocyanate



silica gel



sodium thiocyanate


insect growth regulators



chitin synthesis inhibitors
buprofezin



cyromazine


benzoylphenylurea chitin synthesis
bistrifluron


inhibitors
chlorbenzuron



chlorfluazuron



dichlorbenzuron



diflubenzuron



flucycloxuron



flufenoxuron



hexaflumuron



lufenuron



novaluron



novifiumuron



penfluron



teflubenzuron



triflumuron


juvenile hormone mimics
dayoutong



epofenonane



fenoxycarb



hydroprene



kinoprene



methoprene



pyriproxyfen



triprene


juvenile hormones
juvenile hormone I



juvenile hormone II



juvenile hormone III


moulting hormone agonists
chromafenozide



furan tebufenozide



halofenozide



methoxyfenozide



tebufenozide



yishijing


moulting hormones
α-ecdysone



ecdysterone


moulting inhibitors
diofenolan


precocenes
precocene I



precocene II



precocene III


unclassified insect growth regulators
dicyclanil


macrocyclic lactone insecticides



avermectin insecticides
abamectin



doramectin



emamectin



eprinomectin



ivermectin



selamectin


milbemycin insecticides
lepimectin



milbemectin



milbemycin oxime



moxidectin


spinosyn insecticides
spinetoram



spinosad


neonicotinoid insecticides



nitroguanidine neonicotinoid
clothianidin


insecticides
dinotefuran



imidacloprid



imidaclothiz



thiamethoxam


nitromethylene neonicotinoid
nitenpyram


insecticides
nithiazine


pyridylmethylamine neonicotinoid
acetamiprid


insecticides
imidacloprid



nitenpyram



paichongding



thiacloprid


nereistoxin analogue insecticides
bensultap



cartap



polythialan



thiocyclam



thiosultap


organochlorine insecticides
bromo-DDT



camphechlor



DDT



pp′-DDT



ethyl-DDD



HCH



gamma-HCH



lindane



methoxychlor



pentachlorophenol



TDE


cyclodiene insecticides
aldrin



bromocyclen



chlorbicyclen



chlordane



chlordecone



dieldrin



dilor



endosulfan



alpha-endosulfan



endrin



HEOD



heptachlor



HHDN



isobenzan



isodrin



kelevan



mirex


organophosphorus insecticides



organophosphate insecticides
bromfenvinfos



calvinphos



chlorfenvinphos



crotoxyphos



dichlorvos



dicrotophos



dimethylvinphos



fospirate



heptenophos



methocrotophos



mevinphos



monocrotophos



naled



naftalofos



phosphamidon



propaphos



TEPP



tetrachlorvinphos


organothiophosphate
dioxabenzofos


insecticides
fosmethilan



phenthoate


aliphatic
acethion


organothiophosphate
acetophos


insecticides
amiton



cadusafos



chlorethoxyfos



chlormephos



demephion



demephion-O



demephion-S



demeton



demeton-O



demeton-S



demeton-methyl



demeton-O-methyl



demeton-S-methyl



demeton-S-methylsulphon



disulfoton



ethion



ethoprophos



IPSP



isothioate



malathion



methacrifos



methylacetophos



oxydemeton-methyl



oxydeprofos



oxydisulfoton



phorate



sulfotep



terbufos



thiometon


aliphatic amide
amidithion


organothiophosphate insecticides
cyanthoate



dimethoate



ethoate-methyl



formothion



mecarbam



omethoate



prothoate



sophamide



vamidothion


oxime organothiophosphate
chlorphoxim


insecticides
phoxim



phoxim-methyl


heterocyclic organothiophosphate
azamethiphos


insecticides
colophonate



coumaphos



coumithoate



dioxathion



endothion



menazon



morphothion



phosalone



pyraclofos



pyrazothion



pyridaphenthion



quinothion


benzothiopyran
dithicrofos


organothiophosphate insecticides
thicrofos


benzotriazine organothiophosphate
azinphos-ethyl


insecticides
azinphos-methyl


isoindole organothiophosphate
dialifos


insecticides
phosmet


isoxazole organothiophosphate
isoxathion


insecticides
zolaprofos


pyrazolopyrimidine
chlorprazophos


organothiophosphate insecticides
pyrazophos


pyridine organothiophosphate
chlorpyrifos


insecticides
chlorpyrifos-methyl


pyrimidine organothiophosphate
butathiofos


insecticides
diazinon



etrimfos



lirimfos



pirimioxyphos



pirimiphos-ethyl



pirimiphos-methyl



primidophos



pyrimitate



tebupirimfos


quinoxaline organothiophosphate
quinalphos


insecticides
quinalphos-methyl


thiadiazole organothiophosphate
athidathion


insecticides
lythidathion



methidathion



prothidathion


triazole organothiophosphate
isazofos


insecticides
triazophos


phenyl organothiophosphate
azothoate


insecticides
bromophos



bromophos-ethyl



carbophenothion



chlorthiophos



cyanophos



cythioate



dicapthon



dichlofenthion



etaphos



famphur



fenchlorphos



fenitrothion



fensulfothion



fenthion



fenthion-ethyl



heterophos



jodfenphos



mesulfenfos



parathion



parathion-methyl



phenkapton



phosnichlor



profenofos



prothiofos



sulprofos



temephos



trichlormetaphos-3



trifenofos



xiaochongliulin


phosphonate insecticides
butonate



trichlorfon


phosphonothioate insecticides
mecarphon


phenyl ethylphosphonothioate
fonafos


insecticides
trichloronat


phenyl phenylphosphonothioate
cyanofenphos


insecticides
EPN



leptophos


phosphoramidate insecticides
crufomate



fenamiphos



fosthietan



mephosfolan



phosfolan



phosfolan-methyl



pirimetaphos


phosphoramidothioate insecticides
acephate



chloramine phosphorus



isocarbophos



isofenphos



isofenphos-methyl



methamidophos



phosglycin



propetamphos


phosphorodiamide insecticides
dimefox



mazidox



mipafox



schradan


oxadiazine insecticides
indoxacarb


oxadiazolone insecticides
metoxadiazone


phthalimide insecticides
dialifos



phosmet



tetramethrin


physical insecticides
maltodextrin


desiccant insecticides
boric acid



diatomaceous earth



silica gel


pyrazole insecticides
chlorantraniliprole



cyantraniliprole



cyclaniliprole



dimetilan



isolan



tebufenpyrad



tetraniliprole



tolfenpyrad


phenylpyrazole insecticides
acetoprole



ethiprole



fipronil



flufiprole



pyraclofos



pyrafluprole



pyriprole



pyrolan



vaniliprole


pyrethroid insecticides



pyrethroid ester insecticides
acrinathrin



allethrin



bioallethrin



esdépalléthrine



barthrin



bifenthrin



kappa-bifenthrin



bioethanomethrin



brofenvalerate



brofluthrinate



bromethrin



butethrin



chlorempenthrin



cyclethrin



cycloprothrin



cyfluthrin



beta-cyfluthrin



cyhalothrin



gamma-cyhalothrin



lambda-cyhatothrin



cypermethrin



alpha-cypermethrin



beta-cypermethrin



theta-cypermethrin



zeta-cypermethrin



cyphenothrin



deltamethrin



dimefluthrin



dimethrin



empenthrin



d-fanshiluquebingjuzhi



chloroprallethrin



fenfluthrin



fenpirithrin



fenpropathrin



fenvalerate



esfenvalerate



flucythrinate



fluvalinate



tau-fluvalinate



furamethrin



furethrin



heptafluthrin



imiprothrin



japothrins



kadethrin



methothrin



metofluthrin



epsilon-metofluthrin



momfluorothrin



epsilon-momfluorothrin



pentmethrin



permethrin



biopemethrin



transpermethrin



phenothrin



prallethrin



profluthrin



proparthrin



pyresmethrin



renofluthrin



meperfluthrin



resmethrin



bioresmethrin



cismethrin



tefluthrin



kappa-tefluthrin



terallethrin



tetramethrin



tetramethylfluthrin



tralocythrin



tralomethrin



transfluthrin



valerate


pyrethroid ether insecticides
etofenprox



flufenprox



halfenprox



protrifenbute



silafluofen


pyrethroid oxime insecticides
sulfoxime



thiofluoximate


pyrimidinamine insecticides
flufenerim



pyrimidifen


pyrrole insecticides
chlorfenapyr


quaternary ammonium insecticides
sanguinarine


sulfoximine insecticides
sulfoxaflor


tetramic acid insecticides
spirotetramat


tetronic acid insecticides
spiromesifen


thiazole insecticides
clothianidin



imidaclothiz



thiamethoxam



thiapronil


thiazolidine insecticides
tazimcarb



thiacloprid


thiourea insecticides
diafenthiuron


urea insecticides
flucofuron



sulcofuron


zwitterionic insecticides
dicloromezotiaz



triflumezopyrim


unclassified insecticides
afidopyropen



afoxolaner



allosamidin



closantel



copper naphthenate



crotamiton



EXD



fenazaflor



fenoxacrim



flometoquin



flonicamid



fluhexafon



flupyradifurone



fluralaner



fluxametamide



hydramethylnon



isoprothiolane



jiahuangchongzong



malonoben



metaflumizone



nifluridide



plifenate



pyridaben



pyridalyl



pyrifluquinazon



rafoxanide



thuringiensin



triarathene



triazamate


ACARICIDES



botanical acaricides
carvacrol



sanguinarine


bridged diphenyl acaricides
azobenzene



benzoximate



benzyl benzoate



bromopropylate



chlorbenside



chlorfenethol



chlorfenson



chlorfensulphide



chlorobenzilate



chloropropylate



cyflumetofen



DDT



dicofol



diphenyl sulfone



dofenapyn



fenson



fentrifanil



fluorbenside



genit



hexachlorophene



phenproxide



proclonol



tetradifon



tetrasul


carbamate acaricides
benomyl



carbanolate



carbaryl



carbofuran



methiocarb



metolcarb



promacyl



propoxur


oxime carbamate acaricides
aldicarb



butocarboxim



oxamyl



thiocarboxime



thiofanox


carbazate acaricides
bifenazate


dinitrophenol acaricides
binapacryl



dinex



dinobuton



dinocap



dinocap-4



dinocap-6



dinocton



dinopenton



dinosulfon



dinoterbon



DNOC


formamidine acaricides
amitraz



chlordimeform



chloromebuform



formetanate



formparanate



medimeform



semiamitraz


macrocyclic lactone acaricides
tetranactin


avermectin acaricides
abamectin



doramectin



eprinomectin



ivermectin



selamectin


milbemycin acaricides
milbemectin



milbemycin oxime



moxidectin


mite growth regulators
clofentezine



cyromazine



diflovidazin



dofenapyn



fluazuron



flubenzimine



flucycloxuron



flufenoxuron



hexythiazox


organochlorine acaricides
bromocyclen



camphechlor



DDT



dienochlor



endosulfan



lindane


organophosphorus acaricides



organophosphate acaricides
chlorfenvinphos



crotoxyphos



dichlorvos



heptenophos



mevinphos



monocrotophos



naled



TEPP



tetrachlorvinphos


organothiophosphate acaricides
amidithion



amiton



azinphos-ethyl



azinphos-methyl



azothoate



benoxafos



bromophos



bromophos-ethyl



carbophenothion



chlorpyrifos



chlorthiophos



coumaphos



cyanthoate



demeton



demeton-O



demeton-S



demeton-methyl



demeton-O-methyl



demeton-S-methyl



demeton-S-methylsulphon



dialifos



diazinon



dimethoate



dioxathion



disulfoton



endothion



ethion



ethoate-methyl



formothion



malathion



mecarbam



methacrifos



omethoate



oxydeprofos



oxydisulfoton



parathion



phenkapton



phorate



phosalone



phosmet



phostin



phoxim



pirimiphos-methyl



prothidathion



prothoate



pyrimitate



quinalphos



quiniofos



sophamide



sulfotep



thiometon



triazophos



trifenofos



vamidothion


phosphonate acaricides
trichlorfon


phosphoramidothioate acaricides
isocarbophos



methamidophos



propetamphos


phosphorodiamide acaricides
dimefox



mipafox



schradan


organotin acaricides
azocyclotin



cyhexatin



fenbutatin oxide



phostin


phenylsulfamide acaricides
dichlofluanid


phthalimide acaricides
dialifos



phosmet


pyrazole acaricides
cyenopyrafen



fenpyroximate



pyflubumide



tebufenpyrad


phenylpyrazole acaricides
acetoprole



fipronil



vaniliprole


pyrethroid acaricides



pyrethroid ester acaricides
acrinathrin



bifenthrin



brofluthrinate



cyhalothrin



cypermethrin



alpha-cypermethrin



fenpropathrin



fenvalerate



flucythrinate



flumethrin



fluvalinate



tau-fluvalnate



permethrin


pyrethroid ether acaricides
halfenprox


pyrimidinamine acaricides
pyrimidifen


pyrrole acaricides
chlorfenapyr


quaternary ammonium acaricides
sanguinarine


quinoxaline acaricides
chinomethionat



thioquinox


strobilurin acaricides



methoxyacrylate
bifujunzhi


strobilurin acaricides
fluacrypyrim



flufenoxystrobin



pyriminostrobin


sulfite ester acaricides
aramite



propargite


tetronic acid acaricides
spirodiclofen


tetrazine acaricides
clofentezine



diflovidazin


thiazolidine acaricides
flubenzimine



hexythiazox


thiocarbamate acaricides
fenothiocarb


thiourea acaricides
chloromethiuron



diafenthiuron


unclassified acaricides
acequinocyl



afoxolaner



amidoflumet



arsenous oxide



clenpirin



closantel



crotamiton



cycloprate



cymiazole



disulfiram



etoxazole



fenazaflor



fenazaquin



fluenetil



fluralaner



mesulfen



MNAF



nifluridide



nikkomycins



pyridaben



sulfiram



sulfluramid



sulfur



thuringiensin



triarathene


CHEMOSTERILANTS




apholate



bisazir



busulfan



diflubenzuron



dimatif



hemel



hempa



metepa



methiotepa



methyl apholate



morzid



penfluron



tepa



thiohempa



thiotepa



tretamine



uredepa


INSECT REPELLENTS




acrep



butopyronoxyl



camphor



d-camphor



carboxide



dibutyl phthalate



diethyltoluamide



dimethyl carbate



dimethyl phthalate



dibutyl succinate



ethohexadiol



hexamide



icaridin



methoquin-butyl



methylneodecanamide



2-(octylthio)ethanol



oxamate



quwenzhi



quyingding



rebemide



zengxiaoan


NEMATICIDES



avermectin nematicides
abamectin


botanical nematicides
carvacrol


carbamate nematicides
benomyl



carbofuran



carbosulfan



cloethocarb


oxime carbamate nematicides
alanycarb



aldicarb



aldoxycarb



oxamyl



tirpate


fumigant nematicides
carbon disulfide



cyanogen



1,2-dichloropropane



1,3-dichloropropene



dithioether



methyl bromide



methyl iodide



sodium tetrathiocarbonate


organophosphorus nematicides



organophosphate nematicides
diamidafos



fenamiphos



fosthietan



phosphamidon


organothiophosphate nematicides
cadusafos



chlorpyrifos



dichlofenthion



dimethoate



ethoprophos



fensulfothion



fosthiazate



heterophos



isamidofos



isazofos



phorate



phosphocarb



terbufos



thionazin



triazophos


phosphonothioate nematicides
imicyafos



mecarphon


unclassified nematicides
acetoprole



benclothiaz



chloropicrin



dazomet



DBCP



DCIP



fluazaindolizine



fluensulfone



furfural



metam



methyl isothiocyanate



tioxazafen



xylenols









Insecticides also include synergists or activators that are not in themselves considered toxic or insecticidal, but are materials used with insecticides to synergize or enhance the activity of the insecticides. Synergists or activators include piperonyl butoxide.


Biorational Pesticides


Insecticides can be biorational, or can also be known as biopesticides or biological pesticides. Biorational refers to any substance of natural origin (or man-made substances resembling those of natural origin) that has a detrimental or lethal effect on specific target pest(s), e.g., insects, weeds, plant diseases (including nematodes), and vertebrate pests, possess a unique mode of action, are non-toxic to man, domestic plants and animals, and have little or no adverse effects on wildlife and the environment.


Biorational insecticides (or biopesticides or biological pesticides) can be grouped as: (1) biochemicals (hormones, enzymes, pheromones and natural agents, such as insect and plant growth regulators), (2) microbial (viruses, bacteria, fungi, protozoa, and nematodes), or (3) Plant-Incorporated protectants (PIPs)—primarily transgenic plants, e.g., Bt corn.


Biopesticides, or biological pesticides, can broadly include agents manufactured from living microorganisms or a natural product and sold for the control of plant pests. Biopesticides can be: microorganisms, biochemicals, and semiochemicals. Biopesticides can also include peptides, proteins and nucleic acids such as double-stranded DNA, single-stranded DNA, double-stranded RNA, single-stranded RNA and hairpin DNA or RNA.


Bacteria, fungi, oomycetes, viruses and protozoa are all used for the biological control of insect pests. The most widely used microbial biopesticide is the insect pathogenic bacteria Bacillus thuringiensis (Bt), which produces a protein crystal (the Bt δ-endotoxin) during bacterial spore formation that is capable of causing lysis of gut cells when consumed by susceptible insects. Microbial Bt biopesticides consist of bacterial spores and δ-endotoxin crystals mass-produced in fermentation tanks and formulated as a sprayable product. Bt does not harm vertebrates and is safe to people, beneficial organisms and the environment. Thus, Bt sprays are a growing tactic for pest management on fruit and vegetable crops where their high level of selectivity and safety are considered desirable, and where resistance to synthetic chemical insecticides is a problem. Bt sprays have also been used on commodity crops such as maize, soybean and cotton, but with the advent of genetic modification of plants, farmers are increasingly growing Bt transgenic crop varieties.


Other microbial insecticides include products based on entomopathogenic baculoviruses. Baculoviruses that are pathogenic to arthropods belong to the virus family and possess large circular, covalently closed, and double-stranded DNA genomes that are packaged into nucleocapsids. More than 700 baculoviruses have been identified from insects of the orders Lepidoptera, Hymenoptera, and Diptera. Baculoviruses are usually highly specific to their host insects and thus, are safe to the environment, humans, other plants, and beneficial organisms. Over 50 baculovirus products have been used to control different insect pests worldwide. In the US and Europe, the Cydia pomonella granulovirus (CpGV) is used as an inundative biopesticide against codlingmoth on apples. Washington State, as the biggest apple producer in the US, uses CpGV on 13% of the apple crop. In Brazil, the nucleopolyhedrovirus of the soybean caterpillar Anticarsia gemmatalis was used on up to 4 million ha (approximately 35%) of the soybean crop in the mid-1990s. Viruses such as Gemstar® (Certis USA) are available to control larvae of Heliothis and Helicoverpa species.


At least 170 different biopesticide products based on entomopathogenic fungi have been developed for use against at least five insect and acarine orders in glasshouse crops, fruit and field vegetables as well as commodity crops. The majority of products are based on the ascomycetes Beauveria bassiana or Metarhizium anisopliae. M anisopliae has also been developed for the control of locust and grasshopper pests in Africa and Australia and is recommended by the Food and Agriculture Organization of the United Nations (FAO) for locust management.


A number of microbial pesticides registered in the United States are listed in Table 16 of Kabaluk et al. 2010 (Kabaluk, J. T. et al. (ed.). 2010. The Use and Regulation of Microbial Pesticides in Representative Jurisdictions Worldwide. IOBC Global. 99pp.) and microbial pesticides registered in selected countries are listed in Annex 4 of Hoeschle-Zeledon et al. 2013 (Hoeschle-Zeledon, I., P. Neuenschwander and L. Kumar. (2013). Regulatory Challenges for biological control. SP-IPM Secretariat, International Institute of Tropical Agriculture (IITA), Ibadan, Nigeria. 43 pp.), each of which is incorporated herein in its entirety.


Plants produce a wide variety of secondary metabolites that deter herbivores from feeding on them. Some of these can be used as biopesticides. They include, for example, pyrethrins, which are fast-acting insecticidal compounds produced by Chrysanthemum cinerariaefolium. They have low mammalian toxicity but degrade rapidly after application. This short persistence prompted the development of synthetic pyrethrins (pyrethroids). The most widely used botanical compound is neem oil, an insecticidal chemical extracted from seeds of Azadirachta indica. Two highly active pesticides are available based on secondary metabolites synthesized by soil actinomycetes, but they have been evaluated by regulatory authorities as if they were synthetic chemical pesticides. Spinosad is a mixture of two macrolide compounds from Saccharopolyspora spinosa. It has a very low mammalian toxicity and residues degrade rapidly in the field. Farmers and growers used it widely following its introduction in 1997 but resistance has already developed in some important pests such as western flower thrips. Abamectin is a macrocyclic lactone compound produced by Streptomyces avermitilis. It is active against a range of pest species but resistance has developed to it also, for example, in tetranychid mites.


Peptides and proteins from a number of organisms have been found to possess pesticidal properties. Perhaps most prominent are peptides from spider venom (King, G. F. and Hardy, M. C. (2013) Spider-venom peptides: structure, pharmacology, and potential for control of insect pests. Annu. Rev. Entomol. 58: 475-496). A unique arrangement of disulfide bonds in spider venom peptides render them extremely resistant to proteases. As a result, these peptides are highly stable in the insect gut and hemolymph and many of them are orally active. The peptides target a wide range of receptors and ion channels in the insect nervous system. Other examples of insecticidal peptides include: sea anemone venom that act on voltage-gated Na+ channels (Bosmans, F. and Tytgat, J. (2007) Sea anemone venom as a source of insecticidal peptides acting on voltage-gated Na+ channels. Toxicon. 49(4): 550-560); the PAlb (Pea Albumin 1, subunit b) peptide from Legume seeds with lethal activity on several insect pests, such as mosquitoes, some aphids and cereal weevils (Eyraud, V. et al. (2013) Expression and Biological Activity of the Cystine Knot Bioinsecticide PAlb (Pea Albumin 1 Subunit b). PLoS ONE 8(12): e81619); and an internal 10 kDa peptide generated by enzymatic hydrolysis of Canavalia ensiformis (jack bean) urease within susceptible insects (Martinelli, A. H. S., et al. (2014) Structure-function studies on jaburetox, a recombinant insecticidal peptide derived from jack bean (Canavalia ensiformis) urease. Biochimica et Biophysica Acta 1840: 935-944). Examples of commercially available peptide insecticides include Spear™-T for the treatment of thrips in vegetables and ornamentals in greenhouses, Spear™-P to control the Colorado Potato Beetle, and Spear™-C to protect crops from lepidopteran pests (Vestaron Corporation, Kalamazoo, MI). A novel insecticidal protein from Bacillus bombysepticus, called parasporal crystal toxin (PC), shows oral pathogenic activity and lethality towards silkworms and Cry1Ac-resistant Helicoverpa armigera strains (Lin, P. et al. (2015) PC, a novel oral insecticidal toxin from Bacillus bombysepticus involved in host lethality via APN and BtR-175. Sci. Rep. 5: 11101).


A semiochemical is a chemical signal produced by one organism that causes a behavioral change in an individual of the same or a different species. The most widely used semiochemicals for crop protection are insect sex pheromones, some of which can now be synthesized and are used for monitoring or pest control by mass trapping, lure-and-kill systems and mating disruption. Worldwide, mating disruption is used on over 660,000 ha and has been particularly useful in orchard crops.


As used herein, “transgenic insecticidal trait” refers to a trait exhibited by a plant that has been genetically engineered to express a nucleic acid or polypeptide that is detrimental to one or more pests. In one embodiment, the plants of the present disclosure are resistant to attach and/or infestation from any one or more of the pests of the present disclosure. In one embodiment, the trait comprises the expression of vegetative insecticidal proteins (VIPs) from Bacillus thuringiensis, lectins and proteinase inhibitors from plants, terpenoids, cholesterol oxidases from Streptomyces spp., insect chitinases and fungal chitinolytic enzymes, bacterial insecticidal proteins and early recognition resistance genes. In another embodiment, the trait comprises the expression of a Bacillus thuringiensis protein that is toxic to a pest. In one embodiment, the Bt protein is a Cry protein (crystal protein). Bt crops include Bt corn, Bt cotton and Bt soy. Bt toxins can be from the Cry family (see, for example, Crickmore et al., 1998, Microbiol. Mol. Biol. Rev. 62: 807-812), which are particularly effective against Lepidoptera, Coleoptera and Diptera.


Bt Cry and Cyt toxins belong to a class of bacterial toxins known as pore-forming toxins (PFT) that are secreted as water-soluble proteins undergoing conformational changes in order to insert into, or to translocate across, cell membranes of their host. There are two main groups of PFT: (i) the α-helical toxins, in which α-helix regions form the trans-membrane pore, and (ii) the β-barrel toxins, that insert into the membrane by forming a β-barrel composed of Psheet hairpins from each monomer. See, Parker M W, Feil S C, “Pore-forming protein toxins: from structure to function,” Prog. Biophys. Mol. Biol. 2005 May; 88(1):91-142. The first class of PFT includes toxins such as the colicins, exotoxin A, diphtheria toxin and also the Cry three-domain toxins. On the other hand, aerolysin, α-hemolysin, anthrax protective antigen, cholesterol-dependent toxins as the perfringolysin O and the Cyt toxins belong to the β-barrel toxins. Id. In general, PFT producing-bacteria secrete their toxins and these toxins interact with specific receptors located on the host cell surface. In most cases, PFT are activated by host proteases after receptor binding inducing the formation of an oligomeric structure that is insertion competent. Finally, membrane insertion is triggered, in most cases, by a decrease in pH that induces a molten globule state of the protein. Id.


The development of transgenic crops that produce Bt Cry proteins has allowed the substitution of chemical insecticides by environmentally friendly alternatives. In transgenic plants the Cry toxin is produced continuously, protecting the toxin from degradation and making it reachable to chewing and boring insects. Cry protein production in plants has been improved by engineering cry genes with a plant biased codon usage, by removal of putative splicing signal sequences and deletion of the carboxy-terminal region of the protoxin. See, Schuler T H, et al., “Insect-resistant transgenic plants,” Trends Biotechnol. 1998; 16:168-175. The use of insect resistant crops has diminished considerably the use of chemical pesticides in areas where these transgenic crops are planted. See, Qaim M, Zilberman D, “Yield effects of genetically modified crops in developing countries,” Science. 2003 Feb. 7; 299(5608):900-2.


Known Cry proteins include: δ-endotoxins including but not limited to: the Cry1, Cry2, Cry3, Cry4, Cry5, Cry6, Cry7, Cry8, Cry9, Cry10, Cry11, Cry12, Cry13, Cry14, Cry15, Cry16, Cry17, Cry18, Cry19, Cry20, Cry21, Cry22, Cry23, Cry24, Cry25, Cry26, Cry27, Cry 28, Cry 29, Cry 30, Cry31, Cry32, Cry33, Cry34, Cry35, Cry36, Cry37, Cry38, Cry39, Cry40, Cry41, Cry42, Cry43, Cry44, Cry45, Cry 46, Cry47, Cry49, Cry 51, Cry52, Cry 53, Cry 54, Cry55, Cry56, Cry57, Cry58, Cry59. Cry60, Cry61, Cry62, Cry63, Cry64, Cry65, Cry66, Cry67, Cry68, Cry69, Cry70 and Cry71 classes of δ-endotoxin genes and the B. thuringiensis cytolytic cyt1 and cyt2 genes.


Members of these classes of B. thuringiensis insecticidal proteins include, but are not limited to: Cry1Aa1 (Accession #AAA22353); Cry1Aa2 (Accession #Accession #AAA22552); Cry1Aa3 (Accession #BAA00257); Cry1Aa4 (Accession #CAA31886); Cry1Aa5 (Accession #BAA04468); Cry1Aa6 (Accession #AAA86265); Cry1Aa7 (Accession #AAD46139); Cry1Aa8 (Accession #126149); Cry1Aa9 (Accession #BAA77213); Cry1Aa10 (Accession #AAD55382); Cry1Aa11 (Accession #CAA70856); Cry1Aa12 (Accession #AAP80146); Cry1Aa13 (Accession #AAM44305); Cry1Aa14 (Accession #AAP40639); Cry1Aa15 (Accession #AAY66993); Cry1Aa16 (Accession #HQ439776); Cry1Aa17 (Accession #HQ439788); Cry1Aa18 (Accession #HQ439790); Cry1Aa19 (Accession #HQ685121); Cry1Aa20 (Accession #JF340156); Cry1Aa21 (Accession #JN651496); Cry1Aa22 (Accession #KC158223); Cry1Ab1 (Accession #AAA22330); Cry1Ab2 (Accession #AAA22613); Cry1Ab3 (Accession #AAA22561); Cry1Ab4 (Accession #BAA00071); Cry1Ab5 (Accession #CAA28405); Cry1Ab6 (Accession #AAA22420); Cry1Ab7 (Accession #CAA31620); Cry1Ab8 (Accession #AAA22551); Cry1Ab9 (Accession #CAA38701); Cry1Ab10 (Accession #A29125); Cry1Ab11 (Accession #112419); Cry1Ab12 (Accession #AAC64003); Cry1Ab13 (Accession #AAN76494); Cry1Ab14 (Accession #AAG16877); Cry1Ab15 (Accession #AA013302); Cry1Ab16 (Accession #AAK55546); Cry1Ab17 (Accession #AAT46415); Cry1Ab18 (Accession #AAQ88259); Cry1Ab19 (Accession #AAW31761); Cry1Ab20 (Accession #ABB72460); Cry1Ab21 (Accession #ABS18384); Cry1Ab22 (Accession #ABW87320); Cry1Ab23 (Accession #HQ439777); Cry1Ab24 (Accession #HQ439778); Cry1Ab25 (Accession #HQ685122); Cry1Ab26 (Accession #HQ847729); Cry1Ab27 (Accession #JN135249); Cry1Ab28 (Accession #JN135250); Cry1Ab29 (Accession #JN135251); Cry1Ab30 (Accession #JN135252); Cry1Ab31 (Accession #JN135253); Cry1Ab32 (Accession #JN135254); Cry1Ab33 (Accession #AAS93798); Cry1Ab34 (Accession #KC156668); Cry1Ab-like (Accession #AAK14336); Cry1Ab-like (Accession #AAK14337); Cry1Ab-like (Accession #AAK14338); Cry1Ab-like (Accession #ABG88858); Cry1Ac1 (Accession #AAA22331); Cry1Ac2 (Accession #AAA22338); Cry1Ac3 (Accession #CAA38098); Cry1Ac4 (Accession #AAA73077); Cry1Ac5 (Accession #AAA22339); Cry1Ac6 (Accession #AAA86266); Cry1Ac7 (Accession #AAB46989); Cry1Ac8 (Accession #AAC44841); Cry1Ac9 (Accession #AAB49768); Cry1Ac10 (Accession #CAA05505); Cry1Ac11 (Accession #CAA10270); Cry1Ac12 (Accession #112418); Cry1Ac13 (Accession #AAD38701); Cry1Ac14 (Accession #AAQ06607); Cry1Ac15 (Accession #AAN07788); Cry1Ac16 (Accession #AAU87037); Cry1Ac17 (Accession #AAX18704); Cry1Ac18 (Accession #AAY88347); Cry1Ac19 (Accession #ABD37053); Cry1Ac20 (Accession #ABB89046); Cry1Ac21 (Accession #AAY66992); Cry1Ac22 (Accession #ABZ01836); Cry1Ac23 (Accession #CAQ30431); Cry1Ac24 (Accession #ABL01535); Cry1Ac25 (Accession #FJ513324); Cry1Ac26 (Accession #FJ617446); Cry1Ac27 (Accession #FJ617447); Cry1Ac28 (Accession #ACM90319); Cry1Ac29 (Accession #DQ438941); Cry1Ac30 (Accession #GQ227507); Cry1Ac31 (Accession #GU446674); Cry1Ac32 (Accession #HM061081); Cry1Ac33 (Accession #GQ866913); Cry1Ac34 (Accession #HQ230364); Cry1Ac35 (Accession #JF340157); Cry1Ac36 (Accession #JN387137); Cry1Ac37 (Accession #JQ317685); Cry1Ad1 (Accession #AAA22340); Cry1Ad2 (Accession #CAA01880); Cry1Ae1 (Accession #AAA22410); Cry1Af1 (Accession #AAB82749); Cry1Ag1 (Accession #AAD46137); Cry1Ah1 (Accession #AAQ14326); Cry1Ah2 (Accession #ABB76664); Cry1Ah3 (Accession #HQ439779); Cry1Ai1 (Accession #AA039719); Cry1Ai2 (Accession #HQ439780); Cry1A-like (Accession #AAK14339); Cry1Ba1 (Accession #CAA29898); Cry1Ba2 (Accession #CAA65003); Cry1Ba3 (Accession #AAK63251); Cry1Ba4 (Accession #AAK51084); Cry1Ba5 (Accession #AB020894); Cry1Ba6 (Accession #ABL60921); Cry1Ba7 (Accession #HQ439781); Cry1Bb1 (Accession #AAA22344); Cry1Bb2 (Accession #HQ439782); Cry1Bc1 (Accession #CAA86568); Cry1Bd1 (Accession #AAD10292); Cry1Bd2 (Accession #AAM93496); Cry1Be1 (Accession #AAC32850); Cry1Be2 (Accession #AAQ52387); Cry1Be3 (Accession #ACV96720); Cry1Be4 (Accession #HM070026); Cry1Bf1 (Accession #CAC50778); Cry1Bf2 (Accession #AAQ52380); Cry1Bg1 (Accession #AA039720); Cry1Bh1 (Accession #HQ589331); Cry1Bi1 (Accession #KC156700); Cry1Ca1 (Accession #CAA30396); Cry1Ca2 (Accession #CAA31951); Cry1Ca3 (Accession #AAA22343); Cry1Ca4 (Accession #CAA01886); Cry1Ca5 (Accession #CAA65457); Cry1Ca6 [1] (Accession #AAF37224); Cry1Ca7 (Accession #AAG50438); Cry1Ca8 (Accession #AAM00264); Cry1Ca9 (Accession #AAL79362); Cry1Ca10 (Accession #AAN16462); Cry1Ca11 (Accession #AAX53094); Cry1Ca12 (Accession #HM070027); Cry1Ca13 (Accession #HQ412621); Cry1Ca14 (Accession #JN651493); Cry1Cb1 (Accession #M97880); Cry1Cb2 (Accession #AAG35409); Cry1Cb3 (Accession #ACD50894); Cry1Cb-like (Accession #AAX63901); Cry1Da1 (Accession #CAA38099); Cry1Da2 (Accession #I76415); Cry1Da3 (Accession #HQ439784); Cry1 db1 (Accession #CAA80234); Cry1 db2 (Accession #AAK48937); Cry1 Dc1 (Accession #ABK35074); Cry1Ea1 (Accession #CAA37933); Cry1Ea2 (Accession #CAA39609); Cry1Ea3 (Accession #AAA22345); Cry1Ea4 (Accession #AAD04732); Cry1Ea5 (Accession #A15535); Cry1Ea6 (Accession #AAL50330); Cry1Ea7 (Accession #AAW72936); Cry1Ea8 (Accession #ABX11258); Cry1Ea9 (Accession #HQ439785); Cry1Ea10 (Accession #ADR00398); Cry1Ea11 (Accession #JQ652456); Cry1Eb1 (Accession #AAA22346); Cry1Fa1 (Accession #AAA22348); Cry1Fa2 (Accession #AAA22347); Cry1Fa3 (Accession #HM070028); Cry1Fa4 (Accession #HM439638); Cry1 Fb1 (Accession #CAA80235); Cry1Fb2 (Accession #BAA25298); Cry1Fb3 (Accession #AAF21767); Cry1Fb4 (Accession #AAC10641); Cry1Fb5 (Accession #AA013295); Cry1Fb6 (Accession #ACD50892); Cry1Fb7 (Accession #ACD50893); Cry1Ga1 (Accession #CAA80233); Cry1Ga2 (Accession #CAA70506); Cry1Gb1 (Accession #AAD10291); Cry1Gb2 (Accession #AA013756); Cry1Ge1 (Accession #AAQ52381); Cry1Ha1 (Accession #CAA80236); Cry1Hb1 (Accession #AAA79694); Cry1Hb2 (Accession #HQ439786); Cry1H-like (Accession #AAF01213); Cry1Ia1 (Accession #CAA44633); Cry1Ia2 (Accession #AAA22354); Cry1Ia3 (Accession #AAC36999); Cry1Ia4 (Accession #AAB00958); Cry1Ia5 (Accession #CAA70124); Cry1Ia6 (Accession #AAC26910); Cry1Ia7 (Accession #AAM73516); Cry1Ia8 (Accession #AAK66742); Cry1Ia9 (Accession #AAQ08616); Cry1Ia10 (Accession #AAP86782); Cry1Ia11 (Accession #CAC85964); Cry1Ia12 (Accession #AAV53390); Cry1Ia13 (Accession #ABF83202); Cry1Ia14 (Accession #ACG63871); Cry1Ia15 (Accession #FJ617445); Cry1Ia16 (Accession #FJ617448); Cry1Ia17 (Accession #GU989199); Cry1Ia18 (Accession #ADK23801); Cry1Ia19 (Accession #HQ439787); Cry1Ia20 (Accession #JQ228426); Cry1Ia21 (Accession #JQ228424); Cry1Ia22 (Accession #JQ228427); Cry1Ia23 (Accession #JQ228428); Cry1Ia24 (Accession #JQ228429); Cry1Ia25 (Accession #JQ228430); Cry1Ia26 (Accession #JQ228431); Cry1Ia27 (Accession #JQ228432); Cry1Ia28 (Accession #JQ228433); Cry1Ia29 (Accession #JQ228434); Cry1Ia30 (Accession #JQ317686); Cry1Ia31 (Accession #JX944038); Cry1Ia32 (Accession #JX944039); Cry1Ia33 (Accession #JX944040); Cry1Ib1 (Accession #AAA82114); Cry1Ib2 (Accession #ABW88019); Cry1Ib3 (Accession #ACD75515); Cry1Ib4 (Accession #HM051227); Cry1Ib5 (Accession #HM070028); Cry1Ib6 (Accession #ADK38579); Cry1Ib7 (Accession #JN571740); Cry1Ib8 (Accession #JN675714); Cry1Ib9 (Accession #JN675715); Cry1Ib10 (Accession #JN675716); Cry1Ib11 (Accession #JQ228423); Cry1Ic1 (Accession #AAC62933); Cry1Ic2 (Accession #AAE71691); Cry1Id1 (Accession #AAD44366); Cry1Id2 (Accession #JQ228422); Cry1Ie1 (Accession #AAG43526); Cry1Ie2 (Accession #HM439636); Cry1Ie3 (Accession #KC156647); Cry1Ie4 (Accession #KC156681); Cry1If1 (Accession #AAQ52382); Cry1Ig1 (Accession #KC156701); Cry1I-like (Accession #AAC31094); Cry1I-like (Accession #ABG88859); Cry1Ja1 (Accession #AAA22341); Cry1Ja2 (Accession #HM070030); Cry1Ja3 (Accession #JQ228425); Cry1Jb1 (Accession #AAA98959); Cry1Jc1 (Accession #AAC31092); Cry1Jc2 (Accession #AAQ52372); Cry1Jd1 (Accession #CAC50779); Cry1Ka1 (Accession #AAB00376); Cry1Ka2 (Accession #HQ439783); Cry1La1 (Accession #AAS60191); Cry1La2 (Accession #HM070031); Cry1Ma1 (Accession #FJ884067); Cry1Ma2 (Accession #KC156659); Cry1Na1 (Accession #KC156648); Cry1Nb1 (Accession #KC156678); Cry1-like (Accession #AAC31091); Cry2Aa1 (Accession #AAA22335); Cry2Aa2 (Accession #AAA83516); Cry2Aa3 (Accession #D86064); Cry2Aa4 (Accession #AAC04867); Cry2Aa5 (Accession #CAA10671); Cry2Aa6 (Accession #CAA10672); Cry2Aa7 (Accession #CAA10670); Cry2Aa8 (Accession #AA013734); Cry2Aa9 (Accession #AA013750); Cry2Aa1 O (Accession #AAQ04263); Cry2Aa1 1 (Accession #AAQ52384); Cry2Aa12 (Accession #AB183671); Cry2Aa13 (Accession #ABL01536); Cry2Aa14 (Accession #ACF04939); Cry2Aa15 (Accession #JN426947); Cry2Ab1 (Accession #AAA22342); Cry2Ab2 (Accession #CAA39075); Cry2Ab3 (Accession #AAG36762); Cry2Ab4 (Accession #AA013296); Cry2Ab5 (Accession #AAQ04609); Cry2Ab6 (Accession #AAP59457); Cry2Ab7 (Accession #AAZ66347); Cry2Ab8 (Accession #ABC95996); Cry2Ab9 (Accession #ABC74968); Cry2Ab10 (Accession #EF157306); Cry2Ab11 (Accession #CAM84575); Cry2Ab12 (Accession #ABM21764); Cry2Ab13 (Accession #ACG76120); Cry2Ab14 (Accession #ACG76121); Cry2Ab15 (Accession #HM037126); Cry2Ab16 (Accession #GQ866914); Cry2Ab1 7 (Accession #HQ439789); Cry2Ab18 (Accession #JN135255); Cry2Ab19 (Accession #JN135256); Cry2Ab20 (Accession #JN135257); Cry2Ab21 (Accession #JN135258); Cry2Ab22 (Accession #JN135259); Cry2Ab23 (Accession #JN135260); Cry2Ab24 (Accession #JN135261); Cry2Ab25 (Accession #JN415485); Cry2Ab26 (Accession #JN426946); Cry2Ab27 (Accession #JN415764); Cry2Ab28 (Accession #JN651494); Cry2Ac1 (Accession #CAA40536); Cry2Ac2 (Accession #AAG35410); Cry2Ac3 (Accession #AAQ52385); Cry2Ac4 (Accession #ABC95997); Cry2Ac5 (Accession #ABC74969); Cry2Ac6 (Accession #ABC74793); Cry2Ac7 (Accession #CAL18690); Cry2Ac8 (Accession #CAM09325); Cry2Ac9 (Accession #CAM09326); Cry2Ac10 (Accession #ABN15104); Cry2Ac11 (Accession #CAM83895); Cry2Ac12 (Accession #CAM83896); Cry2Ad1 (Accession #AAF09583); Cry2Ad2 (Accession #ABC86927); Cry2Ad3 (Accession #CAK29504); Cry2Ad4 (Accession #CAM32331); Cry2Ad5 (Accession #CA078739); Cry2Ae1 (Accession #AAQ52362); Cry2Af1 (Accession #AB030519); Cry2Af2 (Accession #GQ866915); Cry2Ag1 (Accession #ACH91610); Cry2Ah1 (Accession #EU939453); Cry2Ah2 (Accession #ACL80665); Cry2Ah3 (Accession #GU073380); Cry2Ah4 (Accession #KC156702); Cry2Ai1 (Accession #FJ788388); Cry2Aj (Accession #); Cry2Ak1 (Accession #KC156660); Cry2Ba1 (Accession #KC156658); Cry3Aa1 (Accession #AAA22336); Cry3Aa2 (Accession #AAA22541); Cry3Aa3 (Accession #CAA68482); Cry3Aa4 (Accession #AAA22542); Cry3Aa5 (Accession #AAA50255); Cry3Aa6 (Accession #AAC43266); Cry3Aa7 (Accession #CAB41411); Cry3Aa8 (Accession #AAS79487); Cry3Aa9 (Accession #AAW05659); Cry3Aa10 (Accession #AAU29411); Cry3Aa11 (Accession #AAW82872); Cry3Aa12 (Accession #ABY49136); Cry3Ba1 (Accession #CAA34983); Cry3Ba2 (Accession #CAA00645); Cry3Ba3 (Accession #JQ397327); Cry3Bb1 (Accession #AAA22334); Cry3Bb2 (Accession #AAA74198); Cry3Bb3 (Accession #115475); Cry3Ca1 (Accession #CAA42469); Cry4Aa1 (Accession #CAA68485); Cry4Aa2 (Accession #BAAOO1 79); Cry4Aa3 (Accession #CAD30148); Cry4Aa4 (Accession #AFB18317); Cry4A-like (Accession #AAY96321); Cry4Ba1 (Accession #CAA30312); Cry4Ba2 (Accession #CAA30114); Cry4Ba3 (Accession #AAA22337); Cry4Ba4 (Accession #BAAOO1 78); Cry4Ba5 (Accession #CAD30095); Cry4Ba-like (Accession #ABC47686); Cry4Ca1 (Accession #EU646202); Cry4Cb1 (Accession #FJ403208); Cry4Cb2 (Accession #FJ597622); Cry4Cc1 (Accession #FJ403207); Cry5Aa1 (Accession #AAA67694); Cry5Ab1 (Accession #AAA67693); Cry5Ac1 (Accession #I34543); Cry5Ad1 (Accession #ABQ82087); Cry5Ba1 (Accession #AAA68598); Cry5Ba2 (Accession #ABW88931); Cry5Ba3 (Accession #AFJ04417); Cry5Ca1 (Accession #HM461869); Cry5Ca2 (Accession #ZP_04123426); Cry5Da1 (Accession #HM461870); Cry5Da2 (Accession #ZP_04123980); Cry5Ea1 (Accession #HM485580); Cry5Ea2 (Accession #ZP_04124038); Cry6Aa1 (Accession #AAA22357); Cry6Aa2 (Accession #AAM46849); Cry6Aa3 (Accession #ABH03377); Cry6Ba1 (Accession #AAA22358); Cry7 Aa1 (Accession #AAA22351); Cry7Ab1 (Accession #AAA21120); Cry7Ab2 (Accession #AAA21121); Cry7Ab3 (Accession #ABX24522); Cry7 Ab4 (Accession #EU380678); Cry7 Ab5 (Accession #ABX79555); Cry7 Ab6 (Accession #ACI44005); Cry7 Ab7 (Accession #ADB89216); Cry7 Ab8 (Accession #GU145299); Cry7Ab9 (Accession #ADD92572); Cry7Ba1 (Accession #ABB70817); Cry7Bb1 (Accession #KC156653); Cry7Ca1 (Accession #ABR67863); Cry7Cb1 (Accession #KC156698); Cry7Da1 (Accession #ACQ99547); Cry7Da2 (Accession #HM572236); Cry7Da3 (Accession #KC156679); Cry7Ea1 (Accession #HM035086); Cry7Ea2 (Accession #HM132124); Cry7Ea3 (Accession #EEM19403); Cry7Fa1 (Accession #HM035088); Cry7Fa2 (Accession #EEM19090); Cry7Fb1 (Accession #HM572235); Cry7Fb2 (Accession #KC156682); Cry7Ga1 (Accession #HM572237); Cry7Ga2 (Accession #KC156669); Cry7Gb1 (Accession #KC156650); Cry7Gc1 (Accession #KC156654); Cry7Gd1 (Accession #KC156697); Cry7Ha1 (Accession #KC156651); Cry7Ia1 (Accession #KC156665); Cry7Ja1 (Accession #KC156671); Cry7Ka1 (Accession #KC156680); Cry7Kb1 (Accession #BAM99306); Cry7La1 (Accession #BAM99307); Cry8Aa1 (Accession #AAA21117); Cry8Ab1 (Accession #EU044830); Cry8Ac1 (Accession #KC156662); Cry8Ad1 (Accession #KC156684); Cry8Ba1 (Accession #AAA21118); Cry8Bb1 (Accession #CAD57542); Cry8Bc1 (Accession #CAD57543); Cry8Ca1 (Accession #AAA21119); Cry8Ca2 (Accession #AAR98783); Cry8Ca3 (Accession #EU625349); Cry8Ca4 (Accession #ADB54826); Cry8Da1 (Accession #BAC07226); Cry8Da2 (Accession #BD133574); Cry8Da3 (Accession #BD133575); Cry8db1 (Accession #BAF93483); Cry8Ea1 (Accession #AAQ73470); Cry8Ea2 (Accession #EU047597); Cry8Ea3 (Accession #KC855216); Cry8Fa1 (Accession #AAT48690); Cry8Fa2 (Accession #HQ1 74208); Cry8Fa3 (Accession #AFH78109); Cry8Ga1 (Accession #AAT46073); Cry8Ga2 (Accession #ABC42043); Cry8Ga3 (Accession #FJ198072); Cry8Ha1 (Accession #AAW81032); Cry8Ia1 (Accession #EU381044); Cry8Ia2 (Accession #GU073381); Cry8Ia3 (Accession #HM044664); Cry8Ia4 (Accession #KC156674); Cry8Ib1 (Accession #GU325772); Cry8Ib2 (Accession #KC156677); Cry8Ja1 (Accession #EU625348); Cry8Ka1 (Accession #FJ422558); Cry8Ka2 (Accession #ACN87262); Cry8Kb1 (Accession #HM123758); Cry8Kb2 (Accession #KC156675); Cry8La1 (Accession #GU325771); Cry8Ma1 (Accession #HM044665); Cry8Ma2 (Accession #EEM86551); Cry8Ma3 (Accession #HM210574); Cry8Na1 (Accession #HM640939); Cry8Pa1 (Accession #HQ388415); Cry8Qa1 (Accession #HQ441166); Cry8Qa2 (Accession #KC152468); Cry8Ra1 (Accession #AFP87548); Cry8Sa1 (Accession #JQ740599); Cry8Ta1 (Accession #KC156673); Cry8-like (Accession #FJ770571); Cry8-like (Accession #ABS53003); Cry9Aa1 (Accession #CAA41122); Cry9Aa2 (Accession #CAA41425); Cry9Aa3 (Accession #GQ249293); Cry9Aa4 (Accession #GQ249294); Cry9Aa5 (Accession #JX1 74110); Cry9Aa like (Accession #AAQ52376); Cry9Ba1 (Accession #CAA52927); Cry9Ba2 (Accession #GU299522); Cry9Bb1 (Accession #AAV28716); Cry9Ca1 (Accession #CAA85764); Cry9Ca2 (Accession #AAQ52375); Cry9Da1 (Accession #BAA1 9948); Cry9Da2 (Accession #AAB97923); Cry9Da3 (Accession #GQ249293); Cry9Da4 (Accession #GQ249297); Cry9db1 (Accession #AAX78439); Cry9Dc1 (Accession #KC1 56683); Cry9Ea1 (Accession #BAA34908); Cry9Ea2 (Accession #AA012908); Cry9Ea3 (Accession #ABM21765); Cry9Ea4 (Accession #ACE88267); Cry9Ea5 (Accession #ACF04743); Cry9Ea6 (Accession #ACG63872); Cry9Ea7 (Accession #FJ380927); Cry9Ea8 (Accession #GQ249292); Cry9Ea9 (Accession #JN651495); Cry9Eb1 (Accession #CAC50780); Cry9Eb2 (Accession #GQ249298); Cry9Eb3 (Accession #KC156646); Cry9Ec1 (Accession #AAC63366); Cry9Ed1 (Accession #AAX78440); Cry9Ee1 (Accession #GQ249296); Cry9Ee2 (Accession #KC156664); Cry9Fa1 (Accession #KC156692); Cry9Ga1 (Accession #KC156699); Cry9-like (Accession #AAC63366); Cry1OAa1 (Accession #AAA22614); Cry10Aa2 (Accession #E00614); Cry10Aa3 (Accession #CAD30098); Cry10Aa4 (Accession #AFB18318); Cry1OA-like (Accession #DQ167578); Cry1 1Aa1 (Accession #AAA22352); Cry1 1Aa2 (Accession #AAA22611); Cry11Aa3 (Accession #CAD30081); Cry11Aa4 (Accession #AFB18319); Cry11Aa-like (Accession #DQ166531); Cry11Ba1 (Accession #CAA60504); Cry11Bb1 (Accession #AAC97162); Cry11Bb2 (Accession #HM068615); Cry12Aa1 (Accession #AAA22355); Cry13Aa1 (Accession #AAA22356); Cry14Aa1 (Accession #AAA21516); Cry14Ab1 (Accession #KC156652); Cry15Aa1 (Accession #AAA22333); Cry16Aa1 (Accession #CAA63860); Cry17Aa1 (Accession #CAA67841); Cry18Aa1 (Accession #CAA67506); Cry18Ba1 (Accession #AAF89667); Cry18Ca1 (Accession #AAF89668); Cry19Aa1 (Accession #CAA68875); Cry19Ba1 (Accession #BAA32397); Cry19Ca1 (Accession #AFM37572); Cry20Aa1 (Accession #AAB93476); Cry20Ba1 (Accession #ACS93601); Cry20Ba2 (Accession #KC156694); Cry20-like (Accession #GQ144333); Cry21Aa1 (Accession #I32932); Cry21Aa2 (Accession #I66477); Cry21Ba1 (Accession #BAC06484); Cry21Ca1 (Accession #JF521577); Cry21Ca2 (Accession #KC156687); Cry21Da1 (Accession #JF521578); Cry22Aa1 (Accession #I34547); Cry22Aa2 (Accession #CAD43579); Cry22Aa3 (Accession #ACD93211); Cry22Ab1 (Accession #AAK50456); Cry22Ab2 (Accession #CAD43577); Cry22Ba1 (Accession #CAD43578); Cry22Bb1 (Accession #KC156672); Cry23Aa1 (Accession #AAF76375); Cry24Aa1 (Accession #AAC61891); Cry24Ba1 (Accession #BAD32657); Cry24Ca1 (Accession #CAJ43600); Cry25Aa1 (Accession #AAC61892); Cry26Aa1 (Accession #AAD25075); Cry27Aa1 (Accession #BAA82796); Cry28Aa1 (Accession #AAD24189); Cry28Aa2 (Accession #AAG00235); Cry29Aa1 (Accession #CAC80985); Cry30Aa1 (Accession #CAC80986); Cry30Ba1 (Accession #BAD00052); Cry30Ca1 (Accession #BAD67157); Cry30Ca2 (Accession #ACU24781); Cry30Da1 (Accession #EF095955); Cry30db1 (Accession #BAE80088); Cry30Ea1 (Accession #ACC95445); Cry30Ea2 (Accession #FJ499389); Cry30Fa1 (Accession #ACI22625); Cry30Ga1 (Accession #ACG60020); Cry30Ga2 (Accession #HQ638217); Cry31Aa1 (Accession #BAB11 757); Cry31Aa2 (Accession #AAL87458); Cry31Aa3 (Accession #BAE79808); Cry31Aa4 (Accession #BAF32571); Cry31Aa5 (Accession #BAF32572); Cry31Aa6 (Accession #BA144026); Cry31Ab1 (Accession #BAE79809); Cry31Ab2 (Accession #BAF32570); Cry31Ac1 (Accession #BAF34368); Cry31Ac2 (Accession #AB731600); Cry31Ad1 (Accession #BA144022); Cry32Aa1 (Accession #AAG36711); Cry32Aa2 (Accession #GU063849); Cry32Ab1 (Accession #GU063850); Cry32Ba1 (Accession #BAB78601); Cry32Ca1 (Accession #BAB78602); Cry32Cb1 (Accession #KC156708); Cry32Da1 (Accession #BAB78603); Cry32Ea1 (Accession #GU324274); Cry32Ea2 (Accession #KC156686); Cry32Eb1 (Accession #KC156663); Cry32Fa1 (Accession #KC156656); Cry32Ga1 (Accession #KC156657); Cry32Ha1 (Accession #KC156661); Cry32Hb1 (Accession #KC156666); Cry32Ia1 (Accession #KC1 56667); Cry32Ja1 (Accession #KC1 56685); Cry32Ka1 (Accession #KC1 56688); Cry32La1 (Accession #KC156689); Cry32Ma1 (Accession #KC156690); Cry32Mb1 (Accession #KC156704); Cry32Na1 (Accession #KC156691); Cry32Oa1 (Accession #KC156703); Cry32Pa1 (Accession #KC156705); Cry32Qa1 (Accession #KC156706); Cry32Ra1 (Accession #KC156707); Cry32Sa1 (Accession #KC156709); Cry32Ta1 (Accession #KC156710); Cry32Ua1 (Accession #KC156655); Cry33Aa1 (Accession #AAL26871); Cry34Aa1 (Accession #AAG50341); Cry34Aa2 (Accession #AAK64560); Cry34Aa3 (Accession #AAT29032); Cry34Aa4 (Accession #AAT29030); Cry34Ab1 (Accession #AAG41671); Cry34Ac1 (Accession #AAG50118); Cry34Ac2 (Accession #AAK64562); Cry34Ac3 (Accession #AAT29029); Cry34Ba1 (Accession #AAK64565); Cry34Ba2 (Accession #AAT29033); Cry34Ba3 (Accession #AAT29031); Cry35Aa1 (Accession #AAG50342); Cry35Aa2 (Accession #AAK64561); Cry35Aa3 (Accession #AAT29028); Cry35Aa4 (Accession #AAT29025); Cry35Ab1 (Accession #AAG41672); Cry35Ab2 (Accession #AAK64563); Cry35Ab3 (Accession #AY536891); Cry35Ac1 (Accession #AAG50117); Cry35Ba1 (Accession #AAK64566); Cry35Ba2 (Accession #AAT29027); Cry35Ba3 (Accession #AAT29026); Cry36Aa1 (Accession #AAK64558); Cry37 Aa1 (Accession #AAF76376); Cry38Aa1 (Accession #AAK64559); Cry39Aa1 (Accession #BAB72016); Cry40Aa1 (Accession #BAB72018); Cry40Ba1 (Accession #BAC77648); Cry40Ca1 (Accession #EU381045); Cry40Da1 (Accession #ACF15199); Cry41Aa1 (Accession #BAD35157); Cry41Ab1 (Accession #BAD35163); Cry41Ba1 (Accession #HM461871); Cry41Ba2 (Accession #ZP_04099652); Cry42Aa1 (Accession #BAD35166); Cry43Aa1 (Accession #BAD15301); Cry43Aa2 (Accession #BAD95474); Cry43Ba1 (Accession #BAD15303); Cry43Ca1 (Accession #KC156676); Cry43Cb1 (Accession #KC156695); Cry43Cc1 (Accession #KC156696); Cry43-like (Accession #BAD15305); Cry44Aa (Accession #BAD08532); Cry45Aa (Accession #BAD22577); Cry46Aa (Accession #BAC79010); Cry46Aa2 (Accession #BAG68906); Cry46Ab (Accession #BAD35170); Cry47 Aa (Accession #AAY24695); Cry48Aa (Accession #CAJ18351); Cry48Aa2 (Accession #CAJ86545); Cry48Aa3 (Accession #CAJ86546); Cry48Ab (Accession #CAJ86548); Cry48Ab2 (Accession #CAJ86549); Cry49Aa (Accession #CAH56541); Cry49Aa2 (Accession #CAJ86541); Cry49Aa3 (Accession #CAJ86543); Cry49Aa4 (Accession #CAJ86544); Cry49Ab1 (Accession #CAJ86542); Cry50Aa1 (Accession #BAE86999); Cry50Ba1 (Accession #GU446675); Cry50Ba2 (Accession #GU446676); Cry51Aa1 (Accession #AB114444); Cry51Aa2 (Accession #GU570697); Cry52Aa1 (Accession #EF613489); Cry52Ba1 (Accession #FJ361760); Cry53Aa1 (Accession #EF633476); Cry53Ab1 (Accession #FJ361759); Cry54Aa1 (Accession #ACA52194); Cry54Aa2 (Accession #GQ140349); Cry54Ba1 (Accession #GU446677); Cry55Aa1 (Accession #ABW88932); Cry54Ab1 (Accession #JQ916908); Cry55Aa2 (Accession #AAE33526); Cry56Aa1 (Accession #ACU57499); Cry56Aa2 (Accession #GQ483512); Cry56Aa3 (Accession #JX025567); Cry57Aa1 (Accession #ANC87261); Cry58Aa1 (Accession #ANC87260); Cry59Ba1 (Accession #JN790647); Cry59Aa1 (Accession #ACR43758); Cry60Aa1 (Accession #ACU24782); Cry60Aa2 (Accession #EA057254); Cry60Aa3 (Accession #EEM99278); Cry60Ba1 (Accession #GU810818); Cry60Ba2 (Accession #EA057253); Cry60Ba3 (Accession #EEM99279); Cry61Aa1 (Accession #HM035087); Cry61Aa2 (Accession #HM132125); Cry61Aa3 (Accession #EEM19308); Cry62Aa1 (Accession #HM054509); Cry63Aa1 (Accession #BA144028); Cry64Aa1 (Accession #BAJ05397); Cry65Aa1 (Accession #HM461868); Cry65Aa2 (Accession #ZP_04123838); Cry66Aa1 (Accession #HM485581); Cry66Aa2 (Accession #ZP_04099945); Cry67Aa1 (Acces-sion #HM485582); Cry67Aa2 (Accession #ZP_04148882); Cry68Aa1 (Accession #HQ113114); Cry69Aa1 (Accession #HQ401006); Cry69Aa2 (Accession #JQ821388); Cry69Ab1 (Accession #JN209957); Cry70Aa1 (Accession #JN646781); Cry70Ba1 (Accession #AD051070); Cry70Bb1 (Accession #EEL67276); Cry71Aa1 (Accession #JX025568); Cry72Aa1 (Accession #JX025569); Cyt1Aa (GenBank Accession Number X03182); Cyt1Ab (GenBank Accession Number X98793); Cyt1B (GenBank Accession Number U37196); Cyt2A (GenBank Accession Number Z14147); and Cyt2B (GenBank Accession Number U52043).


Examples of δ-endotoxins also include but are not limited to Cry1A proteins of U.S. Pat. Nos. 5,880,275, 7,858,849 8,530,411, 8,575,433, and 8,686,233; a DIG-3 or DIG-11 toxin (N-terminal deletion of α-helix 1 and/or α-helix 2 variants of cry proteins such as Cry1A, Cry3A) of U.S. Pat. Nos. 8,304,604, 8,304,605 and 8,476,226; Cry1B of U.S. patent application Ser. No. 10/525,318; Cry1C of U.S. Pat. No. 6,033,874; Cry1F of U.S. Pat. Nos. 5,188,960 and 6,218,188; Cry1A/F chimeras of U.S. Pat. Nos. 7,070,982; 6,962,705 and 6,713,063); a Cry2 protein such as Cry2Ab protein of U.S. Pat. No. 7,064,249); a Cry3A protein including but not limited to an engineered hybrid insecticidal protein (eHIP) created by fusing unique combinations of variable regions and conserved blocks of at least two different Cry proteins (US Patent Application Publication Number 2010/0017914); a Cry4 protein; a Cry5 protein; a Cry6 protein; Cry8 proteins of U.S. Pat. Nos. 7,329,736, 7,449,552, 7,803,943, 7,476,781, 7,105,332, 7,378,499 and 7,462,760; a Cry9 protein such as such as members of the Cry9A, Cry9B, Cry9C, Cry9D, Cry9E and Cry9F families, including but not limited to the Cry9D protein of U.S. Pat. No. 8,802,933 and the Cry9B protein of U.S. Pat. No. 8,802,934; a Cry15 protein of Naimov, et al., (2008), “Applied and Environmental Microbiology,” 74:7145-7151; a Cry22, a Cry34Ab1 protein of U.S. Pat. Nos. 6,127,180, 6,624,145 and 6,340,593; a CryET33 and cryET34 protein of U.S. Pat. Nos. 6,248,535, 6,326,351, 6,399,330, 6,949,626, 7,385,107 and 7,504,229; a CryET33 and CryET34 homologs of US Patent Publication Number 2006/0191034, 2012/0278954, and PCT Publication Number WO 2012/139004; a Cry35Ab1 protein of U.S. Pat. Nos. 6,083,499, 6,548,291 and 6,340,593; a Cry46 protein, a Cry 51 protein, a Cry binary toxin; a TIC901 or related toxin; TIC807 of US Patent Application Publication Number 2008/0295207; ET29, ET37, TIC809, TIC810, TIC812, TIC127, TIC128 of PCT US 2006/033867; TIC853 toxins of U.S. Pat. No. 8,513,494, AXMI-027, AXMI-036, and AXMI-038 of U.S. Pat. No. 8,236,757; AXMI-031, AXMI-039, AXMI-040, AXMI-049 of U.S. Pat. No. 7,923,602; AXMI-018, AXMI-020 and AXMI-021 of WO 2006/083891; AXMI-010 of WO 2005/038032; AXMI-003 of WO 2005/021585; AXMI-008 of US Patent Application Publication Number 2004/0250311; AXMI-006 of US Patent Application Publication Number 2004/0216186; AXMI-007 of US Patent Applica-tion Publication Number 2004/0210965; AXMI-009 of US Patent Application Number 2004/0210964; AXMI-014 of US Patent Application Publication Number 2004/0197917; AXMI-004 of US Patent Application Publication Number 2004/0197916; AXMI-028 and AXMI-029 of WO 2006/119457; AXMI-007, AXMI-008, AXMI-0080rf2, AXMI-009, AXMI-014 and AXMI-004 of WO 2004/074462; AXMI-150 of U.S. Pat. No. 8,084,416; AXMI-205 of US Patent Application Publication Number 2011/0023184; AXMI-011, AXMI-012, AXMI-013, AXMI-015, AXMI-019, AXMI-044, AXMI-037, AXMI-043, AXMI-033, AXMI-034, AXMI-022, AXMI-023, AXMI-041, AXMI-063 and AXMI-064 of US Patent Application Publication Number 2011/0263488; AXMI-R1 and related proteins of US Patent Application Publication Number 2010/0197592; AXMI221Z, AXMI222z, AXMI223z, AXMI224z and AXMI225z of WO 2011/103248; AXMI218, AXMI219, AXMI220, AXMI226, AXMI227, AXMI228, AXMI229, AXMI230 and AXMI231 of WO 2011/103247 and U.S. Pat. No. 8,759,619; AXMI-115, AXMI-113, AXMI-005, AXMI-163 and AXMI-184 of U.S. Pat. No. 8,334,431; AXMI-001, AXMI-002, AXMI-030, AXMI-035 and AXMI-045 of US Patent Application Publication Number 2010/0298211; AXMI-066 and AXMI-076 of US Patent Application Publication Number 2009/0144852; AXMI128, AXMI130, AXMI131, AXMI133, AXMI140, AXMI141, AXMI142, AXMI143, AXMI144, AXMI146, AXMI148, AXMI149, AXMI152, AXMI153, AXMI154, AXMI155, AXMI156, AXMI157, AXMI158, AXMI162, AXMI165, AXMI166, AXMI167, AXMI168, AXMI169, AXMI170, AXMI171, AXMI172, AXMI173, AXMI174, AXMI175, AXMI176, AXMI177, AXMI178, AXMI1179, AXMI180, AXMI181, AXMI182, AXMI185, AXMI186, AXMI187, AXMI188, AXMI189 of U.S. Pat. No. 8,318,900; AXMI079, AXMI080, AXMI081, AXMI082, AXMI091, AXMI092, AXMI096, AXMI097, AXMI098, AXMI099, AXMI100, AXMI101, AXMI102, AXMI103, AXMI104, AXMI107, AXMI108, AXMI109, AXMI110, AXMI111, AXMI1112, AXMI114, AXMI116, AXMI117, AXMI118, AXMI119, AXMI120, AXMI121, AXMI122, AXMI123, AXMI124, AXMI1257, AXMI1268, AXMI127, AXMI1129, AXMI1164, AXMI151, AXMI161, AXMI183, AXMI132, AXMI138, AXMI137 of US Patent Application Publication Number 2010/0005543, AXMI270 of US Patent Application Publication US20140223598, AXMI279 of US Patent Application Publication US20140223599, cry proteins such as Cry1A and Cry3A having modified proteolytic sites of U.S. Pat. No. 8,319,019; a Cry1Ac, Cry2Aa and Cry1Ca toxin protein from Bacillus thuringiensis strain VBTS 2528 of US Patent Application Publication Number 2011/0064710. Other Cry proteins are well known to one skilled in the art. See, N. Crickmore, et al., “Revision of the Nomenclature for the Bacillus thuringiensis Pesticidal Crystal Proteins,” Microbiology and Molecular Biology Reviews,” (1998) Vol 62: 807-813; see also, N. Crickmore, et al., “Bacillus thuringiensis toxin nomenclature” (2016), at http://www.btnomenclature.info/.


The use of Cry proteins as transgenic plant traits is well known to one skilled in the art and Cry-transgenic plants including but not limited to plants expressing Cry1Ac, Cry1Ac+Cry2Ab, Cry1Ab, Cry1A.105, Cry1F, Cry1Fa2, Cry1F+Cry1Ac, Cry2Ab, Cry3A, mCry3A, Cry3Bb1, Cry34Ab1, Cry35Ab1, Vip3A, mCry3A, Cry9c and CBI-Bt have received regulatory approval. See, Sanahuja et al., “Bacillus thuringiensis: a century of research, development and commercial applications,” (2011) Plant Biotech Journal, April 9(3):283-300 and the CERA (2010) GM Crop Database Center for Environmental Risk Assessment (CERA), ILSI Research Foundation, Washington D.C. at cera-gmc.org/index.php?action=gm_crop_database, which can be accessed on the world-wide web using the “www” prefix). More than one pesticidal proteins well known to one skilled in the art can also be expressed in plants such as Vip3Ab & Cry1Fa (US2012/0317682); Cry1BE & Cry1F (US2012/0311746); Cry1CA & Cry1AB (US2012/0311745); Cry1F & CryCa (US2012/0317681); Cry1DA& Cry1BE (US2012/0331590); Cry1DA & Cry1Fa (US2012/0331589); Cry1AB & Cry1BE (US2012/0324606); Cry1Fa & Cry2Aa and Cry11 & Cry1E (US2012/0324605); Cry34Ab/35Ab and Cry6Aa (US20130167269); Cry34Ab/VCry35Ab & Cry3Aa (US20130167268); Cry1Ab & Cry1F (US20140182018); and Cry3A and Cry1Ab or Vip3Aa (US20130116170). Pesticidal proteins also include insecticidal lipases including lipid acyl hydrolases of U.S. Pat. No. 7,491,869, and cholesterol oxidases such as from Streptomyces (Purcell et al. (1993) Biochem Biophys Res Commun 15:1406-1413).


Pesticidal proteins also include VIP (vegetative insecticidal proteins) toxins. Entomopathogenic bacteria produce insecticidal proteins that accumulate in inclusion bodies or parasporal crystals (such as the aforementioned Cry and Cyt proteins), as well as insecticidal proteins that are secreted into the culture medium. Among the latter are the Vip proteins, which are divided into four families according to their amino acid identity. The Vip1 and Vip2 proteins act as binary toxins and are toxic to some members of the Coleoptera and Hemiptera. The Vip1 component is thought to bind to receptors in the membrane of the insect midgut, and the Vip2 component enters the cell, where it displays its ADP-ribosyltransferase activity against actin, preventing microfilament formation. Vip3 has no sequence similarity to Vip1 or Vip2 and is toxic to a wide variety of members of the Lepidoptera. Its mode of action has been shown to resemble that of the Cry proteins in terms of proteolytic activation, binding to the midgut epithelial membrane, and pore formation, although Vip3A proteins do not share binding sites with Cry proteins. The latter property makes them good candidates to be combined with Cry proteins in transgenic plants (Bacillus thuringiensis-treated crops [Bt crops]) to prevent or delay insect resistance and to broaden the insecticidal spectrum. There are commercially grown varieties of Bt cotton and Bt maize that express the Vip3Aa protein in combination with Cry proteins. For the most recently reported Vip4 family, no target insects have been found yet. See, Chakroun et al., “Bacterial Vegetative Insecticidal Proteins (Vip) from Entomopathogenic Bacteria,” Microbiol Mol Biol Rev. 2016 Mar. 2; 80(2):329-50. VIPs can be found in U.S. Pat. Nos. 5,877,012, 6,107,279 6,137,033, 7,244,820, 7,615,686, and 8,237,020 and the like. Other VIP proteins are well known to one skilled in the art (see, lifesci.sussex.ac.uk/home/Neil_Crickmore/Bt/vip.html, which can be accessed on the world-wide web using the “www” prefix).


Pesticidal proteins also include toxin complex (TC) proteins, obtainable from organisms such as Xenorhabdus, Photorhabdus and Paenibacillus (see, U.S. Pat. Nos. 7,491,698 and 8,084,418). Some TC proteins have “stand alone” insecticidal activity and other TC proteins enhance the activity of the stand-alone toxins produced by the same given organism. The toxicity of a “stand-alone” TC protein (from Photorhabdus, Xenorhabdus or Paenibacillus, for example) can be enhanced by one or more TC protein “potentiators” derived from a source organism of a different genus. There are three main types of TC proteins. As referred to herein, Class A proteins (“Protein A”) are stand-alone toxins. Class B proteins (“Protein B”) and Class C proteins (“Protein C”) enhance the toxicity of Class A proteins. Examples of Class A proteins are TcbA, TcdA, XptA1 and XptA2. Examples of Class B proteins are TcaC, TcdB, XptB1Xb and XptC1 Wi. Examples of Class C proteins are TccC, XptC1Xb and XptB1 Wi. Pesticidal proteins also include spider, snake and scorpion venom proteins. Examples of spider venom peptides include, but are not limited to lycotoxin-1 peptides and mutants thereof (U.S. Pat. No. 8,334,366). 03321 Some currently registered PIPs are listed in Table 11. Transgenic plants have also been engineered to express dsRNA directed against insect genes (Baum, J. A. et al. (2007) Control of coleopteran insect pests through RNA interference. Nature Biotechnology 25: 1322-1326; Mao, Y. B. et al. (2007) Silencing a cotton bollworm P450 monooxygenase gene by plant-mediated RNAi impairs larval tolerance of gossypol. Nature Biotechnology 25: 1307-1313). RNA interference can be triggered in the pest by feeding of the pest on the transgenic plant. Pest feeding thus causes injury or death to the pest.









TABLE 11







List of exemplary Plant-incorporated Protectants, which


can be combined with microbes of the disclosure











Pesticide



Company and Trade
Registration


Plant-Incorporated Protectants (PIPs)
Names
Numbers





Potato
Potato



Cry3A Potato PC Code 006432
Naturemark
524-474



New Leaf Monsanto



Cry3A & PLRV Potato
Monsanto
524-498


PC Codes 006432, 006469
New Leaf Plus



Corn




Cry1Ab Corn Event 176 PC Code 006458
Mycogen Seeds/Dow
68467-1  



Agro
66736-1  



Syngenta Seeds



Cry1Ab Corn Event Bt11 EPA PC Code
Agrisure CB (with
67979-1  


006444 OECD Unique Identifier SYN-
Yieldgard)
65268-1  


BTØ11-1,
Attribute Insect




Protected Sweet Corn




Syngenta Seeds



Cry1Ab Corn Event MON 801
Monsanto
524-492


Cry1Ab corn Event MON 810 PC Code
Monsanto
524-489


006430 OECD Unique Identifier MON-




ØØ81Ø-6




Cry1Ac Corn PC Code 006463
Dekalb Genetics c/o
69575-2  



Monsanto




BT-XTRA



Cry1F corn Event TC1507 PC Code
Mycogen Seeds/Dow
68467-2  


006481 OECD Unique Identifier DAS-
Agro
29964-3  


Ø15Ø7-1
Pioneer Hi-




Bred/Dupont



moCry1F corn Event DAS-Ø6275-8 PC
Mycogen Seeds/Dow
68467-4  


Code 006491 OECD Unique Identifier
Agro



DAS-Ø6275-8




Cry9C Corn
Aventis
264-669



StarLink



Cry3Bb1 corn Event MON863 PC Code
Monsanto
524-528


006484
YielGard RW



OECD Unique Identifier MON-ØØ863-5




Cry3Bb1 corn Event MON 88017 PC
Monsanto
524-551


Code 006498
YieldGrad VT



OECD Unique Identifier MON-88Ø17-3
Rootworm



Cry34Ab1/Cry35Ab1 corn Event DAS-
Mycogen Seeds/Dow
68467-5  


591227-7
Agro
29964-4  


PC Code 006490
Pioneer Hi-



OECD Unique Identifier DAS-59122-7
Bred/Dupont Herculex




Rootworm



Cry34Ab1/Cry35Ab1 and Cry1F corn
Pioneer Hi-
29964-17  


Event 4114
Bred/Dupont



PC Codes 006555, 006556




mCry3A corn Event MIR 604
Syngenta Seeds
67979-5  


PC Code 006509 OECD Unique Identifier
Agrisure RW



SYN-IR604-8




Cry1A.105 and Cry2Ab2 corn Event
Monsanto
524-575


MON 89034 PC Codes 006515 and
Genuity VT Double



006514
Pro



Vip3Aa20 corn Event MIR 162
Syngenta Seeds
67979-14  


PC Code 006599 OECD Unique Identifier
Agrisure Viptera



SYN-IR162-4




eCry3.1Ab corn in Event 5307 PC Code
Syngenta
67979-22  


016483 OECD Unique Identifier SYN-




Æ53Æ7-1




Stacked Events and Seed Blend Corn




MON863 × MON810 with Cry3Bb1 +
Monsanto YieldGard
524-545


Cry1Ab
Plus



DAS-59122-7 × TC1507 with
Mycogen Seeds/Dow
68467-6  


Cr34Ab1/Cry35Ab1 + Cry1F
Agro Pioneer Hi-
29964-5  



Bred/Dupont




Herculex Xtra



MON 88017 × MON 810 with Cry1AB +
Monsanto
524-552


Cry3Bb
YieldGard VT Triple




YieldGard VT Plus



MIR 604 × Bt11 with mCry3A +Cry1Ab
Syngenta
67979-8  



Agrisure CB/RW




Agrisure 3000GT



Mon 89034 × Mon 88017 with Cry1A.105 +
Monsanto
524-576


Cry2Ab2 +Cry3Bb1
Genuity VT Triple




PRO



Bt11 × MIR 162 with Cry1Ab +Vip3Aa
Syngenta Seeds
67979-12  


20
Agrisure 2100



Bt11 × MIR 162 × MIR 604 with Cry1Ab +
Syngenta Seeds
67979-13  


Vip3Aa20 + mCry3A
Agrisure 3100



MON 89034 × TC1507 × MON 88017 ×
Monsanto Company
524-581


DAS-59122-7 with Cry1A.105 +
Mycogen Seeds/Dow
68467-7  


Cry2Ab2 + Cry1F + Cry3Bb1 +
Agro



Cry34Ab1/Cry35Ab1
Genuity SmartStax




SmartStax



MON 89034 × TC1507 × MON 88017 ×
Monsanto Company
524-595


DAS-59122-7 Seed Blend
Mycogen Seeds/Dow
68467-16  



Agro




Genuity SmartStax




RIB Complete




SmartStax Refuge




Advanced; Refuge




Advanced Powered by




SmartStax



Seed Blend of Herculex Xtra + Herculex I
Pioneer Hi-
29964-6  



Bred/Dupont




Optimum AcreMax1




Insect Protection



Seed Blend of Herculex RW + Non-Bt
Pioneer Hi-
29964-10  


corn
Bred/Dupont




Optimum AcreMax




RW



(Cry1F × Cry34/35 × Cry1Ab)-seed
Pioneer Hi-
29964-11  


blend
Bred/Dupont




Optimum AcreMax




Xtra



(Cry1F × Cry1Ab)-seed blend
Pioneer Hi-
29964-12  



Bred/Dupont




Optimum AcreMax




Insect Protection



(Cry1F × mCry3A)
Pioneer Hi-
29964-13  



Bred/Dupont




Optimum Trisect



(Cry1F × Cry34/35 × Cry1Ab × mCry3A)
Pioneer Hi-
29964-14  



Bred/Dupont




Optimum Intrasect




Xtreme



59122 × MON 810 × MIR 604 (C34/35 ×
Pioneer Hi-
29964-15  


Cry1Ab × mCry3A)
Bred/Dupont



Optimum AcreMax Xtreme (Cry1F ×
Pioneer Hi-
29964-16  


Cry34/35 × Cry1Ab × mCry3A)-seed
Bred/Dupont



blend
Optimum AcreMax




Xtreme (seed blend)



MON 810 × MIR 604 (Cry1Ab ×
Pioneer Hi-
29964-18  


mCry3A)
Bred/Dupont



1507 × MON810 × MIR 162 (Cry1F ×
Pioneer Hi-
29964-19  


Cry1Ab × Vip 3Aa20)
Bred/Dupont




Optimum Intrasect




Leptra



1507 × MIR 162 (Cry1F × Vip30Aa20)
Pioneer Hi-
29964-20  



Bred/Dupont



4114 × MON 810 × MIR 604 (Cry34/35 ×
Pioneer Hi-
29964-21  


Cry1F × Cry1Ab × mCry3A)-seed blend
Bred/Dupont



4114 × MON810 × MIR 604 (Cry34/35 ×
Pioneer Hi-
29964-22  


Cry1F × Cry1Ab × mCry3A)
Bred/Dupont



1507 × MON810 × MIR604 (Cry1F ×
Pioneer Hi-
29964-23  


Cry1Ab × mCry3A)-seed blend
Bred/Dupont




Optimum AcreMax




Trisect



1507 × MON810 × MIR 604 (Cry1F ×
Pioneer Hi-
29964-24  


Cry1Ab × mCry3A)
Bred/Dupont




Optimum Intrasect




Trisect



4114 × MON 810 (Cry34/35 × Cry1F ×
Pioneer Hi-
29964-25  


Cry1Ab)
Bred/Dupont



1507 × MON810 × MIR 162 (Cry1F ×
Pioneer Hi-
29964-26  


Cry1Ab × Vip 3Aa20)-seed blend
Bred/Dupont




Optimum AcreMax




Leptra



SmartStax intermediates (8 products)
Monsanto
524-583, 524-584,




524-586, 524-587,




524-588, 524-589,




524-590


MON 89034 × 1507 (Cry1A.105 ×
Monsanto
524-585


Cry2Ab2 × Cry1F)
Genuity PowerCore



MON 89034 (Cry1A.105 × Cry2Ab2)-
Monsanto
524-597


seed blend
Genuity VT Double




PRO RIB Complete



MON 89034 × 88017 R113 Complete
Monsanto
524-606


(Cry1A.105 × Cry2Ab2 × Cry3Bb1)-
Genuity VT Triple



seed blend
PRO RIB Complete



MON 89034 × 1507 (Cry1A.105 ×
Monsanto
524-612


Cry2Ab2 × Cry1F)-seed blend
Genuity PowerCore




RIB Complete



Bt11 × MIR162 × 1507 (Cry1Ab ×
Syngenta Seeds
67979-15  


Vip3Aa20 × Cry1F)
Agrisure Viptera 3220




Refuge Renew



Bt11 × 59122-7 × MIR 604 × 1507
Syngenta Seeds
67979-17  


(Cry1Ab × Cry34/35 × mCry3A × Cry1F)
Agrisure 3122



Bt11 × MIR162 × TC1507 (Cry1Ab ×
Syngenta Seeds
67979-19  


Vip3Aa20 × Cry1F)-seed blend
Agrisure Viptera 3220




(E-Z Refuge) (Refuge




Advanced)



Bt11 × DAS 59122-7 × MIR604 ×
Syngenta Seeds
67979-20  


TC1507 (Cry1Ab × Cry34/35 × mCry3A ×
Agrisure Viptera 3122



Cry1F)-seed blend
(E-Z Refuge) (Refuge




Advanced)



Bt11 × MIR 162 × MIR 604 × TC1507 ×
Syngenta Seeds
67979-23  


5307 (Cry1Ab × Vip3Aa20 × mCry3A ×
Agrisure Duracade



Cry1F × eCry3.1Ab)
(Refuge Renew) 5222



Bt11 × MIR 604 × TC1507 × 5307
Syngenta Seeds
67979-24  


(Cry1Ab × mCry3A × Cry1F ×
Agrisure Duracade



eCry3.1Ab)
(Refuge Renew) 5122



Bt11 × MIR 604 × TC1507 × 5307
Syngenta Seeds
67979-25  


(Cry1Ab × mCry3A × Cry1F ×
Agrisure Duracade



eCry3.1Ab)-seed blend
5122 E-Z Refuge



Bt11 × MIR 162 × MIR 604 × TC1507 ×
Syngenta Seeds
67979-26  


5307 (Cry1Ab × Vip3Aa20 × mCry3A ×
Agrisure Duracade



Cry1F × eCry3.1Ab)-seed blend
5222 E-Z Refuge



Bt11 × MIR 162 × MIR 604 × TC1507 ×
Syngenta Seeds
67979-27  


5307 (Cry1Ab × Vip3Aa20 × mCry3A ×
Agrisure Duracade



Cry1F × eCry3.1Ab)
(Refuge Renew) 5022



MIR604 × DAS-59122-7 × TC1507
Syngenta Seeds
67979-29  


(mCry3A × Cry34/35 × Cry1F)




SmartStax Intermediates (8 products)
Mycogen Seeds/Dow
68467-8, 68467-9,



Agro
68467-10, 68467-11,




68467-13, 68467-14,




68467-15  


MON 89034 × 1507 (Cry1A.105 ×
Mycogen Seeds/Dow
68467-12  


Cry2Ab2 × Cry1F)
Agro




PowerCore;




PowerCore Enlist



MON 89034 × 1507 (Cry1A.105 ×
Mycogen Seeds/Dow
68467-21  


Cry2Ab2 × Cry1F)-seed blend
Agro




PowerCore Refuge




Advanced; Refuge




Advanced Powered)




PowerCore



1507 × MON810
Pioneer Hi-
29964-7  



Bred/Dupont




Optimum Intrasect



59122 × 1507 × MON810
Pioneer Hi-
29964-8  



Bred/Dupont



59122 × MON810
Pioneer Hi-
29964-9  



Bred/Dupont



Cotton




Cry1Ac Cotton
Monsanto
524-478



BollGard



Cry1Ac and Cry2Ab2 in Event 15985
Monsanto
524-522


Cotton PC Codes 006445, 006487
BollGard II



Bt cotton Event MON531 with Cry1Ac
Monsanto
524-555


(breeding nursery use only)




Bt cotton Event MON15947 with
Monsanto
524-556


Cry2Ab2 (breeding nursery use only)




COT102 × MON 15985 (Vip3Aa19 ×
Monsanto
524-613


Cry1Ac × Cry2Ab2)
Bollgard II



Cry1F and Cry1Ac (Events DAS-21023-5 ×
Mycogen Seeds/Dow
68467-3  


DAS-24236-5) Cotton PC Codes
Agro



006512, 006513
Widestrike



Event 3006-210-23 (Cry1Ac)
Mycogen Seeds/Dow
68467-17  



Agro



Event 281-24-236 (Cry1F)
Mycogen Seeds/Dow
68467-18  



Agro



WideStrike × COT102 (Cry1F × Cry1Ac ×
Mycogen Seeds/Dow
68467-19  


Vip3Aa19)
Agro




Wide Strike 3



Vip3Aa19 and FLCry1Ab (Events
Syngenta Seeds
67979-9  


Cot102 × Cot67B) Cotton PC Codes
(Formally VipCot)



016484, 016486 OECD Unique Identifier




SYN-IR102-7 X SYN-IR67B-1




COT102 (Vip3Aa19)
Syngenta Seeds
67979-18  


COT67B (FLCry1Ab)
Syngenta Seeds
67979-21  


T304-40 (Cry1Ab)
Bayer CropScience
 264-1094


GHB119 (Cry2Ae)
Bayer CropScience
 264-1095


T304-40 × GHB 119 (Cry1Ab × Cry2Ae)
Bayer CropScience
 264-1096


OECD Unique Identifier: BCS-GHØØ4-7 ×
TwinLink



BCS-GHØØ5-8




Soybean




Cry1Ac in Event 87701 Soybean PC
Monsanto
524-594


Code 006532 OECD Unique Identifier
Inacta



Cry1A.105 and Cry2Ab2 in Event 87751
Monsanto
524-619


Soybean PC Codes 006614, 006615




OECD Unique Identifier MON-87751-7




Cry1Ac × Cry1F in Event DAS 81419
Mycogen Seeds/Dow
68467-20  


Soybean PC Codes 006527, 006528
Agro



OECD Unique Identifier




DAS 81419 (Cry1Ac × Cry1F)









In some embodiments, any one or more of the pesticides set forth herein may be utilized with any one or more of the microbes of the disclosure and can be applied to plants or parts thereof, including seeds.


Herbicides


As aforementioned, agricultural compositions of the disclosure, which may comprise any microbe taught herein, are sometimes combined with one or more herbicides.


Compositions comprising bacteria or bacterial populations produced according to methods described herein and/or having characteristics as described herein may further include one or more herbicides. In some embodiments, herbicidal compositions are applied to the plants and/or plant parts. In some embodiments, herbicidal compositions may be included in the compositions set forth herein, and can be applied to a plant(s) or a part(s) thereof simultaneously or in succession, with other compounds.


Herbicides include 2,4-D, 2,4-DB, acetochlor, acifluorfen, alachlor, ametryn, atrazine, aminopyralid, benefin, bensulfuron, bensulide, bentazon, bicyclopyrone, bromacil, bromoxynil, butylate, carfentrazone, chlorimuron, chlorsulfuron, clethodim, clomazone, clopyralid, cloransulam, cycloate, DCPA, desmedipham, dicamba, dichlobenil, diclofop, diclosulam, diflufenzopyr, dimethenamid, diquat, diuron, DSMA, endothall, EPTC, ethalfluralin, ethofumesate, fenoxaprop, fluazifop-P, flucarbazone, flufenacet, flumetsulam, flumiclorac, flumioxazin, fluometuron, fluroxypyr, fomesafen, foramsulfuron, glufosinate, glyphosate, halosulfuron, hexazinone, imazamethabenz, imazamox, imazapic, imazaquin, imazethapyr, isoxaflutole, lactofen, linuron, MCPA, MCPB, mesotrione, metolachlor-s, metribuzin, indaziflam, metsulfuron, molinate, MSMA, napropamide, naptalam, nicosulfuron, norflurazon, oryzalin, oxadiazon, oxyfluorfen, paraquat, pelargonic acid, pendimethalin, phenmedipham, picloram, primisulfuron, prodiamine, prometryn, pronamide, propanil, prosulfuron, pyrazon, pyrithioac, quinclorac, quizalofop, rimsulfuron, S-metolachlor, sethoxydim, siduron, simazine, sulfentrazone, sulfometuron, sulfosulfuron, tebuthiuron, tembotrione, terbacil, thiazopyr, thifensulfuron, thiobencarb, topramezone, tralkoxydim, triallate, triasulfuron, tribenuron, triclopyr, trifluralin, and triflusulfuron.


In some embodiments, any one or more of the herbicides set forth herein may be utilized with any one or more of the plants or parts thereof set forth herein.


Herbicidal products may include CORVUS, BALANCE FLEXX, CAPRENO, DIFLEXX, LIBERTY, LAUDIS, AUTUMN SUPER, and DIFLEXX DUO.


In some embodiments, any one or more of the herbicides set forth in the below Table 12 may be utilized with any one or more of the microbes taught herein, and can be applied to any one or more of the plants or parts thereof set forth herein.









TABLE 12







List of exemplary herbicides, which can be combined with microbes of the


disclosure











Herbicide





Group




Site of Action
Number
Chemical Family
Herbicide













ACCase
1
Cyclohexanediones
Sethoxydim (Roast,


inhibitors


Roast Plus)





Clethodim (Select,





Select Max, Arrow)




Aryloxyphenoxypropionates
Fluazifop (Fusilade DX,





component in Fusion)





Fenoxapmp (Puma,





component in Fusion)





Quizalofop (Assure II,





Targa)




Phenylpyrazolins
Pinoxaden (Axial XL)


ALS inhibitors
2
Imidazolinones
Imazethapyr (Pursuit)





Imazamox (Raptor)




Sulfonylureas
Chlorimuron (Classic)





Halosulfuron (Permit





Sandea)





Iodosulfuron





(component in Autumn





Super)





Mesosulfuron (Osprey)





Nicosulfuron (Accent Q)





Primisulfuron (Beacon)





Prosulfuron (Peak)





Rimsulfuron (Matrix,





Resolve)





Thifensulfuron





(Harmony)





Tribenuron (Express)





Triflusulfuron (UpBeet)




Triazolopyrimidine
Flumetsulam (Python)





Cloransulam-methyl





(FirstRate)





Pymxsulam (PowerFlex





HL)





Florasulam (component





in Quelex)




Sulfonylaminocarbonyltriazolin
Propoxycarbazone




ones
(Olympus)





Thiencarbazone-methyl





(component in





Capreno)


Microtubule
3
Dinitroanilines
Trifluralin (many


inhibitors (root


names)


inhibitors)


Ethaffluralin (Sonalan)





Pendimethalin





(Prowl/Prowl H2O)




Benzamide
Pronamide (Kerb)


Synthetic auxins
4
Arylpicolinate
Halauxifen (Elevore,





component in Ouelex)




Phenoxy acetic acids
2,4-D (Enlist One,





others)





2,4-DB (Butvrac 200,





Butoxone 200)





MCPA




Benzoic acids
Dicamba (Banvel,




Clarity, DiFlexx,





Engenia, XtendiMax;






component in Status)




Pyridines
Clopyralid (Stinger)





Fluroxypyr (Starane





Ultra)


Photosystem II
5
Triazines
Atrazine


inhibitors


Simazine (Princep, Sim-





Trol)




Triazinone
Metribuzin (Metribuzin,





others)





Hexazinone (Velpar)




Phenyl-carbamates
Desmedipham (Betenex)





Pherimedipham





(component in Betamix)




Uracils
Terbacil (Sinbar)



6
Benzothiadiazoles
Ben 1a7on (Basagran,





others)




Nitrites
Bromoxynit (Buctril,





Moxy, others)



7
Phenyl ureas
Linuron (Lorox, Lima)


Lipid synthesis
8
Thiocarbamates
EPTC (Eptatn)


inhibitor





EPSPS inhibitor
9
Organophosphorus
Glyphosate


Glutamine
10
Organophosphorus
Glufosinate (Liberty,


synthetase


Rely)


inhibitor





Diterpene
13
Isoxazolidinone
Clomazone (Command)


biosynthesis





inhibitor





(bleaching)





Protoporphyrinogen
14
Diphenylether
Acifluorfen (Ultra


oxidase


Blazer)


inhibitors (PPO)


Fomesafen (Flexstar,





Reflex)





Lactofen (Cobra,





Phoenix)




N-phenylphthalimide
Flumiclorac (Resource)





Flumioxazin (Valor,





Valor EZ, Rowel)




Aryl triazolinone
Sulfentrazone





(Authority Spartan)





Carfentrazone (Aim)





Fluthiacet-methyl





(Cadet)




Pyrazoles
Pyraflufen-ethyl (Vida)




Pyrimidinedione
Saflufenacil (Sharpen)


Long-chain fatty
15
Acetarnides
Acetochlor (Harness,


acid inhibitors


Surpass AXT,





Break free AXT,





Warrant)





Dimethenamid-P





(Outlook)





Metolachlor (Parallel)





Pyroxasulfone (Zidua,





Zidua SC)





s-metolachlor (Dual





Magnum, Dual II





Magnum, Cinch)





Flufenacet (Define)


Specific site
16
Benzofuranes
Ethofiuriesate (Nortrot


unknown





Auxin transport
19
Semicarbazone
diflufenzopyr


inhibitor


(component in Status)


Photosystem 1
22
Bipyridiliums
Paraquat (Gramoxone,


inhibitors


Parazone)





Diquat (Reglone)


4-HPPD
27
Isoxazole
Isoxaflutole (Balance


inhibitors

Pyrazole
Flexx)


(bleaching)

Pyrazolone
Pyrasulfotole




Triketone
(component in Huskie)





Topramezone





(Armezon/Impact)





Bicyclopyrone





(component in Acuron)





Mesotrione (Callisto)





Tembotrione (Laudis)










Fungicides


As aforementioned, agricultural compositions of the disclosure, which may comprise any microbe taught herein, are sometimes combined with one or more fungicides.


Compositions comprising bacteria or bacterial populations produced according to methods described herein and/or having characteristics as described herein may further include one or more fungicides. In some embodiments, fungicidal compositions may be included in the compositions set forth herein, and can be applied to a plant(s) or a part(s) thereof simultaneously or in succession, with other compounds. The fungicides include azoxystrobin, captan, carboxin, ethaboxam, fludioxonil, mefenoxam, fludioxonil, thiabendazole, thiabendaz, ipconazole, mancozeb, cyazofamid, zoxamide, metalaxyl, PCNB, metaconazole, pyraclostrobin, Bacillus subtilis strain QST 713, sedaxane, thiamethoxam, fludioxonil, thiram, tolclofos-methyl, trifloxystrobin, Bacillus subtilis strain MBI 600, pyraclostrobin, fluoxastrobin, Bacillus pumilus strain QST 2808, chlorothalonil, copper, flutriafol, fluxapyroxad, mancozeb, gludioxonil, penthiopyrad, triazole, propiconaozole, prothioconazole, tebuconazole, fluoxastrobin, pyraclostrobin, picoxystrobin, qols, tetraconazole, trifloxystrobin, cyproconazole, flutriafol, SDHI, EBDCs, sedaxane, MAXIM QUATTRO (gludioxonil, mefenoxam, azoxystrobin, and thiabendaz), RAXIL (tebuconazole, prothioconazole, metalaxyl, and ethoxylated tallow alkyl amines), and benzovindiflupyr.


In some embodiments, any one or more of the fungicides set forth herein may be utilized with any one or more of the plants or parts thereof set forth herein.


Hormones


As aforementioned, agricultural compositions of the disclosure, which may comprise any microbe taught herein, are sometimes combined with one or more hormones.


Compositions comprising bacteria or bacterial populations produced according to methods described herein and/or having characteristics as described herein may further include one or more hormones. In some embodiments, hormone compositions are applied to the plants and/or plant parts. In some embodiments, hormone compositions may be included in the compositions set forth herein, and can be applied to a plant(s) or a part(s) thereof simultaneously or in succession, with other compounds.


Hormones include, but are not limited to, auxins, cytokinins, gibberellins, abscisic acid, ethylene, brassinosteroids, jasmonic acid, strigolactones, and chemical mimics of strigolactone.


In some embodiments, any one or more of the hormones set forth herein may be utilized with any one or more of the plants or parts thereof set forth herein.


Strigolactones


As aforementioned, agricultural compositions of the disclosure, which may comprise any microbe taught herein, are sometimes combined with one or more strigolactone or chemical mimics of strigolactone. Such compounds are described in PCT/US2016/029080, filed Apr. 23, 2016, and entitled: Methods for Hydraulic Enhancement of Crops, which is hereby incorporated by reference. They are further described in U.S. Pat. No. 9,994,557, issued on Jun. 12, 2018, and entitled: Strigolactone Formulations and Uses Thereof, which is hereby incorporated by reference.


In some embodiments, the strigolactone is a compound of Formula (I), a salt, solvate, polymorph, stereoisomer, or isomer thereof: wherein: a, b, and c are one of the following:




embedded image




    • i. a is 0 or 2, and b and c are each independently 0, 1, or 2;

    • ii. a is 1, b is 0, and c is 0 or 2;

    • iii. a is 1, b is 1, and c is 1 or 2; or

    • iv. a is 1, b is 2, and c is 0, 1, or 2;
      • each A is independently O, or S;
      • each E is independently O, S, or —NR18.
      • each G is independently C;
      • R5, R6, R11, R12, R14, R15, and R17 are each independently H, alkyl, haloalkyl, amino, halo, or —OR18 or a lone electron pair;
      • R1 and R16 are each independently H, alkyl, haloalkyl, amino, halo, or —OR18;
      • or R4 and R13 together form a direct bond to provide a double bond;
      • Each R18 is independently H, alkyl, haloalkyl, aryl, heteroaryl, —C(O)R19 or







embedded image






      • and

      • each R19 is independently H, alkyl, haloalkyl, aryl, or heteroaryl.







In some embodiments, the strigolactone is a compound of Formula (1), a salt, solvate, or isomer, thereof, wherein:




embedded image




    • each E is independently O, S, or —NR7;

    • each G is independently C or N;

    • R1, R4, R5, and R6 are each independently H, amino, halo, substituted or unsubstituted alkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted arylalkyl, substituted or unsubstituted heteroaryl, substituted or unsubstituted heteroarylalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, —OR8, —C(O)R8,







embedded image



or a lone electron pair, wherein




embedded image



indicates a single bond;

    • R2 and R3 are each independently H, amino, halo, substituted or unsubstituted alkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted arylalkyl, substituted or unsubstituted heteroaryl, substituted or unsubstituted heteroarylalkyl, substituted or unsubstituted cycloalkyl, or substituted or unsubstituted heterocycloalkyl, or a lone electron pair; or R2 and R3 together form a bond, or form a substituted or unsubstituted aryl; and.
    • R7 and R8 are each independently H, amino, halo, substituted or unsubstituted alkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted arylalkyl, substituted or unsubstituted heteroaryl, substituted or unsubstituted heteroarylalkyl, substituted or unsubstituted cycloalkyl, or substituted or unsubstituted heterocycloalkyl.


In some embodiments, any one or more of the strigolactones or mimics of strigolactone set forth herein may be utilized with any one or more of the plants or parts thereof set forth herein.


In some embodiments of the method, the combination of agricultural compositions of the disclosure, which may comprise any microbe taught herein, with one or more strigolactone or chemical mimics of strigolactone, a yield of the contacted plant is extended as compared to an uncontacted plant, a wilting of the contacted plant is reduced or delayed as compared to an uncontacted plant, a turgidity of the contacted plant is prolonged or maintained as compared to an uncontacted plant, a loss of one or more petals of the contacted plant is reduced or delayed as compared to an uncontacted plant, a chlorophyll content of the contacted plant is maintained as compared to an uncontacted plant, a loss of the chlorophyll content of the contacted plant is reduced or delayed as compared to an uncontacted plant, a chlorophyll content of the contacted plant is increased as compared to an uncontacted plant, a salinity tolerance of the contacted plant is increased as compared to an uncontacted plant, a water consumption of the contacted plant is reduced as compared to an uncontacted plant, a drought tolerance of the contacted plant is increased as compared to an uncon-tacted plant, a pest resistance of the contacted plant is increased as compared to an uncontacted plant, a pesticides consumption of the contacted plant is reduced as compared to an uncontacted plant, or any combination thereof.


In one embodiment of the method, a yield of the contacted plant is increased as compared to an uncontacted plant.


In another embodiment of the method, a life of the contacted plant is extended as compared to an uncontacted plant.


In another embodiment of the method, a wilting of the contacted plant is reduced or delayed as compared to an uncontacted plant.


In another embodiment of the method, a wilting of the contacted plant is reduced or delayed as compared to an uncontacted plant.


In another embodiment of the method, a turgidity of the contacted plant is prolonged or maintained as compared to an uncontacted plant.


In another embodiment of the method, a loss of one or more petals of the contacted plant is reduced or delayed as compared to an uncontacted plant.


In another embodiment of the method, a chlorophyll content of the contacted plant is maintained as compared to an uncontacted plant.


In another embodiment of the method, a loss of the chlorophyll content of the contacted plant is reduced or delayed as compared to an uncontacted plant.


In another embodiment of the method, a chlorophyll content of the contacted plant is increased as compared to an uncon-tacted plant.


In another embodiment of the method, a salinity tolerance of the contacted plant is increased as compared to an uncontacted plant.


In another embodiment of the method, a water consumption of the contacted plant is reduced as compared to an uncontacted plant.


In another embodiment of the method, a drought tolerance of the contacted plant is increased as compared to an uncontacted plant.


In another embodiment of the method, a pest resistance of the contacted plant is increased as compared to an uncontacted plant.


In another embodiment of the method, a pesticides consumption of the contacted plant is reduced as compared to an uncontacted plant.


In another embodiment, when an agricultural composition of the disclosure, which may comprise any microbe taught herein, is combined with one or more strigolactone or chemical mimics of strigolactone, transpiration of the plant is increased as compared to an uncontacted plant.


In another embodiment, when an agricultural composition of the disclosure, which may comprise any microbe taught herein, is combined with one or more strigolactone or chemical mimics of strigolactone, canopy temperature of the contacted plant plant is decreased by at least about 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1.0° C. as compared to a substantially identical but uncontacted plant.


In another embodiment, when an agricultural composition of the disclosure, which may comprise any microbe taught herein, is combined with one or more strigolactone or chemical mimics of strigolactone, hydraulic enhancement of a plant is elicited upon contact with a plant wherein a permanent wilting point of the contacted plant is decreased as compared to a substantially identical but otherwise uncontacted plant.


In another embodiment, when an agricultural composition of the disclosure, which may comprise any microbe taught herein, is combined with one or more strigolactone or chemical mimics of strigolactone, transpiration of the plant is increased as compared to an uncontacted plant.


Nematicides


As aforementioned, agricultural compositions of the disclosure, which may comprise any microbe taught herein, are sometimes combined with one or more nematicides.


Compositions comprising bacteria or bacterial populations produced according to methods described herein and/or having characteristics as described herein may further include one or more nematicide. In some embodiments, nematicidal compositions may be included in the compositions set forth herein, and can be applied to a plant(s) or a part(s) thereof simultaneously or in succession, with other compounds. The nematicides may be selected from D-D, 1,3-dichloropropene, ethylene dibromide, 1,2-dibromo-3-chloropropane, methyl bromide, chloropicrin, metam sodium, dazomet, methylisothiocyanate, sodium tetrathiocarbonate, aldicarb, aldoxycarb, carbofuran, oxamyl, ethoprop, fenamiphos, cadusafos, fosthiazate, terbufos, fensulfothion, phorate, DiTera, clandosan, sincocin, methyl iodide, propargyl bromide, 2,5-dihydroxymethyl-3,4-dihydroxypyrrolidine (DMDP), any one or more of the avermectins, sodium azide, furfural, Bacillus firmus, abamectrin, thiamethoxam, fludioxonil, clothiandin, salicylic acid, and benzo-(1,2,3)-thiadiazole-7-carbothioic acid S-methyl ester.


In some embodiments, any one or more of the nematicides set forth herein may be utilized with any one or more of the plants or parts thereof set forth herein.


In some embodiments, any one or more of the nematicides, fungicides, herbicides, insecticides, and/or pesticides set forth herein may be utilized with any one or more of the plants or parts thereof set forth herein.


Fertilizers, Nitrogen Stabilizers, and Urease Inhibitors


As aforementioned, agricultural compositions of the disclosure, which may comprise any microbe taught herein, are sometimes combined with one or more of a: fertilizer, nitrogen stabilizer, or urease inhibitor.


In some embodiments, fertilizers are used in combination with the methods and bacteria of the present disclosure. Fertilizers include anhydrous ammonia, urea, ammonium nitrate, and urea-ammonium nitrate (UAN) compositions, among many others. In some embodiments, pop-up fertilization and/or starter fertilization is used in combination with the methods and bacteria of the present disclosure.


In some embodiments, nitrogen stabilizers are used in combination with the methods and bacteria of the present disclosure. Nitrogen stabilizers include nitrapyrin, 2-chloro-6-(trichloromethyl) pyridine, N-SERVE 24, INSTINCT, dicyandiamide (DCD).


In some embodiments, urease inhibitors are used in combination with the methods and bacteria of the present disclosure. Urease inhibitors include N-(n-butyl)-thiophosphoric triamide (NBPT), AGROTAIN, AGROTAIN PLUS, and AGROTAIN PLUS SC. Further, the disclosure contemplates utilization of AGROTAIN ADVANCED 1.0, AGROTAIN DRI-MAXX, and AGROTAIN ULTRA.


Further, stabilized forms of fertilizer can be used. For example, a stabilized form of fertilizer is SUPER U, containing 46% nitrogen in a stabilized, urea-based granule, SUPERU contains urease and nitrification inhibitors to guard from denitrification, leaching, and volatilization. Stabilized and targeted foliar fertilizer such as NITAMIN may also be used herein.


Pop-up fertilizers are commonly used in corn fields. Pop-up fertilization comprises applying a few pounds of nutrients with the seed at planting. Pop-up fertilization is used to increase seedling vigor.


Slow- or controlled-release fertilizer that may be used herein entails: A fertilizer containing a plant nutrient in a form which delays its availability for plant uptake and use after application, or which extends its availability to the plant significantly longer than a reference ‘rapidly available nutrient fertilizer’ such as ammonium nitrate or urea, ammonium phosphate or potassium chloride. Such delay of initial availability or extended time of continued availability may occur by a variety of mechanisms. These include controlled water solubility of the material by semi-permeable coatings, occlusion, protein materials, or other chemical forms, by slow hydrolysis of water-soluble low molecular weight compounds, or by other unknown means.


Stabilized nitrogen fertilizer that may be used herein entails: A fertilizer to which a nitrogen stabilizer has been added. A nitrogen stabilizer is a substance added to a fertilizer which extends the time the nitrogen component of the fertilizer remains in the soil in the urea-N or ammoniacal-N form.


Nitrification inhibitor that may be used herein entails: A substance that inhibits the biological oxidation of ammoniacal-N to nitrate-N. Some examples include: (1) 2-chloro-6-(trichloromethyl-pyridine), common name Nitrapyrin, manufactured by Dow Chemical; (2) 4-amino-1,2,4-6-triazole-HCl, common name ATC, manufactured by Ishihada Industries; (3) 2,4-diamino-6-trichloro-methyltriazine, common name CI-1580, manufactured by American Cyanamid; (4) Dicyandiamide, common name DCD, manufactured by Showa Denko; (5) Thiourea, common name TU, manufactured by Nitto Ryuso; (6) 1-mercapto-1,2,4-triazole, common name MT, manufactured by Nippon; (7) 2-amino-4-chloro-6-methyl-pyramidine, common name AM, manufactured by Mitsui Toatsu; (8) 3,4-dimethylpyrazole phosphate (DMPP), from BASF; (9) 1-amide-2-thiourea (ASU), from Nitto Chemical Ind.; (10) Ammoniumthiosulphate (ATS); (11) 1H-1,2,4-triazole (HPLC); (12) 5-ethylene oxide-3-trichloro-methyl 1,2,4-thiodiazole (Terrazole), from Olin Mathieson; (13) 3-methylpyrazole (3-MP); (14) 1-carbamoyle-3-methyl-pyrazole (CMP); (15) Neem; and (16) DMPP.


Urease inhibitor that may be used herein entails: A substance that inhibits hydrolytic action on urea by the enzyme urease. Thousands of chemicals have been evaluated as soil urease inhibitors (Kiss and Simihaian, 2002). However, only a few of the many compounds tested meet the necessary requirements of being non toxic, effective at low concentration, stable, and compatible with urea (solid and solutions), degradable in the soil and inexpensive. They can be classified according to their structures and their assumed interaction with the enzyme urease (Watson, 2000, 2005). Four main classes of urease inhibitors have been proposed: (a) reagents which interact with the sulphydryl groups (sulphydryl reagents), (b) hydroxamates, (c) agricultural crop protection chemicals, and (d) structural analogues of urea and related compounds. N-(n-Butyl) thiophosphoric triamide (NBPT), phenylphosphorodiamidate (PPD/PPDA), and hydroquinone are probably the most thoroughly studied urease inhibitors (Kiss and Simihaian, 2002). Research and practical testing has also been carried out with N-(2-nitrophenyl) phosphoric acid triamide (2-NPT) and ammonium thiosulphate (ATS). The organo-phosphorus compounds are structural analogues of urea and are some of the most effective inhibitors of urease activity, blocking the active site of the enzyme (Watson, 2005).


Insecticidal Seed Treatments (ISTs) for Corn


Corn seed treatments normally target three spectrums of pests: nematodes, fungal seedling diseases, and insects.


Insecticide seed treatments are usually the main component of a seed treatment package. Most corn seed available today comes with a base package that includes a fungicide and insecticide. In some aspects, the insecticide options for seed treatments include PONCHO (clothianidin), CRUISER/CRUISER EXTREME (thiamethoxam) and GAUCHO (Imidacloprid). All three of these products are neonicotinoid chemistries. CRUISER and PONCHO at the 250 (0.25 mg AI/seed) rate are some of the most common base options available for corn. In some aspects, the insecticide options for treatments include CRUISER 250 thiamethoxam, CRUISER 250 (thiamethoxam) plus LUMIVIA (chlorantraniliprole), CRUISER 500 (thiamethoxam), and PONCHO VOTIVO 1250 (Clothianidin & Bacillus firmus I-1582).


Pioneer's base insecticide seed treatment package consists of CRUISER 250 with PONCHO/VOTIVO 1250 also available. VOTIVO is a biological agent that protects against nematodes.


Monsanto's products including corn, soybeans, and cotton fall under the ACCELERON treatment umbrella. Dekalb corn seed comes standard with PONCHO 250. Producers also have the option to upgrade to PONCHO/VOTIVO, with PONCHO applied at the 500 rate.


Agrisure, Golden Harvest and Garst have a base package with a fungicide and CRUISER 250. AVICTA complete corn is also available; this includes CRUISER 500, fungicide, and nematode protection. CRUISER EXTREME is another option available as a seed treatment package, however; the amounts of CRUISER are the same as the conventional CRUISER seed treatment, i.e. 250, 500, or 1250.


Another option is to buy the minimum insecticide treatment available, and have a dealer treat the seed downstream.


Commercially available ISTs for corn are listed in the below Table 13 and can be combined with one or more of the microbes taught herein.









TABLE 13







List of exemplary seed treatments, including ISTs, which can be combined with


microbes of the disclosure










Treatment





Type
Active Ingredient(s)
Product Trade Name
Crop





F
azoxystrobin
DYNASTY
Corn, Soybean




PROTEGE FL
Corn


F

Bacillus pumilus

YIELD SHIELD
Corn, Soybean


F

Bacillus subtilis

HISTICK N/T
Soybean




VAULT HP
Corn, Soybean


F
Captan
CAPTAN 400
Corn, Soybean




CAPTAN 400-C
Corny





Soybean


F
Fludioxonil
MAXIUM 4FS
Corn, Soybean


F
Hydrogen peroxide
OXIDATE
Soybean




STOROX
Soybean


F
ipconazole
ACCELERON DC-509
Corn




RANCONA 3.8 FS
Corn, Soybean




VORTEX
Corn


F
mancozeb
BONIDE MANCOZEB w/Zinc
Corn




Concentrate





DITHANE 75D-F





RAINSHIELD
Corn




DITHANE DF RAINSHIELD
Corn




DITHANE F45 RAINSHIELD
Corn




DITHANE M45
Corn




LESCO 4 FLOWABLE
Corn




MANCOZEB





PENNCOZEB 4FL





FLOWABLE
Corn




PENNCOZEB 75DF DRY
Corn




FLOWABLE





PENNCOZEB 80WP
Corn


F
mefenoxam
APRON XL
Corn, Soybean


F
metalaxyl
ACCELERON DC-309
Corn




ACCELERON DX-309
Corn, Soybean




ACQUIRE
Corn, Soybean




AGRI STAR METALAXYL
Corn, Soybean




265 ST





ALLEGIANCE DRY
Corn, Soybean




ALLEGIANCE FL
Corn, Soybean




BELMONT 2.7 FS
Corn, Soybean




DYNA-SHIELD
Corn, Soybean




METALAXYL





SEBRING 2.65 ST
Corn, Soybean




SEBRING 318 FS
Corn, Soybean




SEBRING 480 FS
Corn, Soybean




VIREO MEC
Soybean


F
pyraclostrobin
ACCELERON DX-109
Soybean




STAMINA
Corn


F

Streptomyces

MYCOSTOP
Corn, Soybean



griseoviridis




F

Streptomyces lydicus

ACTINOGROW ST
Corn, Soybean


F
tebuconazole
AMTIDE TEBIJ 3.6F
Corn




SATIVA 309 FS
Corn




SATIVA 318 FS
Corn




TEBUSHA 3.6FL
Corn




TEBUZOL 3.6F
Corn


F
thiabendazole
MERTECT 340-F
Soybean


F
thiram
42-S THIRAM
Corn, Soybean




FLOWSAN
Corn, Soybean




SIGNET 480 FS
Corn, Soybean


F
Trichoderma
T-22 HC
Corn, Soybean



harzianum Rifai




F
trifloxystrobin
ACCELERON DX-709
Corn




TRILEX FLOWABLE
Corn, soybean


I
chlorpyrifos
LORSBAN saw in water
Corn




soluble packets



I
clothianidin
ACCELERON IC-609
Corn




NIPSIT INSIDE
Corn, Soybean




PONCHO 600
Corn


I
imidacloprid
ACCELERON IX-409
Corn




AGRI STAR MACHO 600 ST
Corn, Soybean




AGRISOLUTIONS NITRO
Corn, Soybean




SHIELD






Corn, Soybean




ATTENDANT 600
Corn, Soybean




AXCESS
Soybean




COURAZE 2F
Corn, Soybean




DYNA-SHIELD





IMIDACLOPRID 5
Corn, Soybean




GAUCHO 480 FLOWABLE
Corn, Soybean




GAUCHO 600 FLOWABLE
Corn, Soybean




GAUCHO SB FLOWABLE
Soybean




NUPRID 4.6F PRO
Corn, Soybean




SENATOR 600 FS



I
thiamethoxam
CRUISER 5FS
Corn, Soybean


N
abamectin
AVICTA 500 FS
Corn, Soybean


N

Bacillus firmus

VOTIVO FS
Soybean


P
cytokinin
SOIL X-CYTO
Soybean




X-CYTE
Soybean


P
harpin alpha beta
ACCELERON HX-209
Corn, Soy-bean



protein
N-HIBIT GOLD CST
Corn, Soybean




N-HIBIT HX-209
Corn, Soybean


P
indole butyric acid
KICKSTAND PGR
Corn, Soybean


I, N
thiamethoxam,
AVICTA DUO CORN-
Corn



abamectin
AVICTA DUO 250



I, F
clothianidin, Bacillus
PONCHO VOTIVO
Corn, Soybean




firmus





F, F
carboxin, captan
ENHANCE
Soybean


I, F
permethrin, carboxin
KERNEL GUARD SUPREME
Corn, Soybean


F, F
carboxin, thiram
VITAFLO 280
Corn, Soybean


F, F
mefenoxam,
MAXIM XL
Corn, Soybean



fludioxonil
WARDEN RTA
Soybean




APRON MAXX RFC





APRON MAXX RTA + MOLY





APRON MAXX RTA



I, F
imidacloprid,
AGRISOLUTIONS CONCUR
Corn



metalaxyl




F, F
metalaxyl, ipconazole
RANCONA SUMMIT
Soybean




RANCONA XXTRA



F, F
thiram, metalaxyl
PROTECTOR-L-
Soybean




ALLEGIANCE



F, F
trifloxystrobin,
TRILEX AL
Soybean



metalaxyl
TRILEX 2000



P, P, P
cytokinin, gibberellic
STIMULATE YIELD
Corn, Soybean



acid, indole butyric
ENHANCER ASCEND




acid




F, F, I
mefenoxam,
CRUISERMAXX PLUS
Soybean



fludioxonil,





thiamethoxam




F, F, F
captan, carboxin,
BEAN GUARD/
Soybean



metalaxyl
ALLEGIANCE



F, F, I
captan, carboxin,
ENHANCE AW
Soybean



imidacloprid




F, F, I
carboxin,
LATITUDE
Corn, Soybean



metalaxyl, imidacloprid




F, F, F
metalaxyl,
STAMINA F3 HL
Corn



pyraclostrobin,





triticonazole




F, F, F, I
azoxystrobin,
CRUISER EXTREME
Corn



fludioxonil,





mefenoxam,





thiamethoxam




F, F, F, F,
azoxystrobin,
MAXIM QUATTRO
Corn


F
fludioxonil,





mefenoxam,





thiabendazole




I
Chlorantran iliprole
LUMIVIA
Corn





F = Fungicide;


I = Insecticide;


N = Nematicide;


P = Plant Growth Regulator







Application of Bacterial Populations on Crops


The composition of the bacteria or bacterial population described herein can be applied in furrow, in talc, or as seed treatment. The composition can be applied to a seed package in bulk, mini bulk, in a bag, or in talc.


The planter can plant the treated seed and grows the crop according to conventional ways, twin row, or ways that do not require tilling. The seeds can be distributed using a control hopper or an individual hopper. Seeds can also be distributed using pressurized air or manually. Seed placement can be performed using variable rate technologies. Additionally, application of the bacteria or bacterial population described herein may be applied using variable rate technologies. In some examples, the bacteria can be applied to seeds of corn, soybean, canola, Sorghum, potato, rice, vegetables, cereals, pseudocereals, and oilseeds. Examples of cereals may include barley, fonio, oats, palmer's grass, rye, pearl millet, Sorghum, spelt, teff, triticale, and wheat. Examples of pseudocereals may include breadnut, buckwheat, cattail, chia, flax, grain amaranth, hanza, quinoa, and sesame. In some examples, seeds can be genetically modified organisms (GMO), non-GMO, organic or conventional.


Additives such as micro-fertilizer, PGR, herbicide, insecticide, and fungicide can be used additionally to treat the crops. Examples of additives include crop protectants such as insecticides, nematicides, fungicide, enhancement agents such as colorants, polymers, pelleting, priming, and disinfectants, and other agents such as inoculant, PGR, softener, and micronutrients. PGRs can be natural or synthetic plant hormones that affect root growth, flowering, or stem elongation. PGRs can include auxins, gibberellins, cytokinins, ethylene, and abscisic acid (ABA).


The composition can be applied in furrow in combination with liquid fertilizer. In some examples, the liquid fertilizer may be held in tanks. NPK fertilizers contain macronutrients of sodium, phosphorous, and potassium.


The composition may improve plant traits, such as promoting plant growth, maintaining high chlorophyll content in leaves, increasing fruit or seed numbers, and increasing fruit or seed unit weight. Methods of the present disclosure may be employed to introduce or improve one or more of a variety of desirable traits. Examples of traits that may introduced or improved include: root biomass, root length, height, shoot length, leaf number, water use efficiency, overall biomass, yield, fruit size, grain size, photosynthesis rate, tolerance to drought, heat tolerance, salt tolerance, tolerance to low nitrogen stress, nitrogen use efficiency, resistance to nematode stress, resistance to a fungal pathogen, resistance to a bacterial pathogen, resistance to a viral pathogen, level of a metabolite, modulation in level of a metabolite, proteome expression. The desirable traits, including height, overall biomass, root and/or shoot biomass, seed germination, seedling survival, photosynthetic efficiency, transpiration rate, seed/fruit number or mass, plant grain or fruit yield, leaf chlorophyll content, photosynthetic rate, root length, or any combination thereof, can be used to measure growth, and compared with the growth rate of reference agricultural plants (e.g., plants without the introduced and/or improved traits) grown under identical conditions. In some examples, the desirable traits, including height, overall biomass, root and/or shoot biomass, seed germination, seedling survival, photosynthetic efficiency, transpiration rate, seed/fruit number or mass, plant grain or fruit yield, leaf chlorophyll content, photosynthetic rate, root length, or any combination thereof, can be used to measure growth, and compared with the growth rate of reference agricultural plants (e.g., plants without the introduced and/or improved traits) grown under similar conditions.


An agronomic trait to a host plant may include, but is not limited to, the following: altered oil content, altered protein content, altered seed carbohydrate composition, altered seed oil composition, and altered seed protein composition, chemical tolerance, cold tolerance, delayed senescence, disease resistance, drought tolerance, ear weight, growth improvement, health enhancement, heat tolerance, herbicide tolerance, herbivore resistance improved nitrogen fixation, improved nitrogen utilization, improved root architecture, improved water use efficiency, increased biomass, increased root length, increased seed weight, increased shoot length, increased yield, increased yield under water-limited conditions, kernel mass, kernel moisture content, metal tolerance, number of ears, number of kernels per ear, number of pods, nutrition enhancement, pathogen resistance, pest resistance, photosynthetic capability improvement, salinity tolerance, stay-green, vigor improvement, increased dry weight of mature seeds, increased fresh weight of mature seeds, increased number of mature seeds per plant, increased chlorophyll content, increased number of pods per plant, increased length of pods per plant, reduced number of wilted leaves per plant, reduced number of severely wilted leaves per plant, and increased number of non-wilted leaves per plant, a detectable modulation in the level of a metabolite, a detectable modulation in the level of a transcript, and a detectable modulation in the proteome, compared to an isoline plant grown from a seed without said seed treatment formulation.


In some cases, plants are inoculated with bacteria or bacterial populations that are isolated from the same species of plant as the plant element of the inoculated plant. For example, an bacteria or bacterial population that is normally found in one variety of Zea mays (corn) is associated with a plant element of a plant of another variety of Zea mays that in its natural state lacks said bacteria and bacterial populations. In one embodiment, the bacteria and bacterial populations is derived from a plant of a related species of plant as the plant element of the inoculated plant. For example, an bacteria and bacterial populations that is normally found in Zea diploperennis Iltis et al., (diploperennial teosinte) is applied to a Zea mays (corn), or vice versa. In some cases, plants are inoculated with bacteria and bacterial populations that are heterologous to the plant element of the inoculated plant. In one embodiment, the bacteria and bacterial populations is derived from a plant of another species. For example, an bacteria and bacterial populations that is normally found in dicots is applied to a monocot plant (e.g., inoculating corn with a soybean-derived bacteria and bacterial populations), or vice versa. In other cases, the bacteria and bacterial populations to be inoculated onto a plant is derived from a related species of the plant that is being inoculated. In one embodiment, the bacteria and bacterial populations is derived from a related taxon, for example, from a related species. The plant of another species can be an agricultural plant. In another embodiment, the bacteria and bacterial populations is part of a designed composition inoculated into any host plant element.


In some examples, the bacteria or bacterial population is exogenous wherein the bacteria and bacterial population is isolated from a different plant than the inoculated plant. For example, in one embodiment, the bacteria or bacterial population can be isolated from a different plant of the same species as the inoculated plant. In some cases, the bacteria or bacterial population can be isolated from a species related to the inoculated plant.


In some examples, the bacteria and bacterial populations described herein are capable of moving from one tissue type to another. For example, the present disclosure's detection and isolation of bacteria and bacterial populations within the mature tissues of plants after coating on the exterior of a seed demonstrates their ability to move from seed exterior into the vegetative tissues of a maturing plant. Therefore, in one embodiment, the population of bacteria and bacterial populations is capable of moving from the seed exterior into the vegetative tissues of a plant. In one embodiment, the bacteria and bacterial populations that is coated onto the seed of a plant is capable, upon germination of the seed into a vegetative state, of localizing to a different tissue of the plant. For example, bacteria and bacterial populations can be capable of localizing to any one of the tissues in the plant, including: the root, adventitious root, seminal 5 root, root hair, shoot, leaf, flower, bud, tassel, meristem, pollen, pistil, ovaries, stamen, fruit, stolon, rhizome, nodule, tuber, trichome, guard cells, hydathode, petal, sepal, glume, rachis, vascular cambium, phloem, and xylem. In one embodiment, the bacteria and bacterial populations is capable of localizing to the root and/or the root hair of the plant. In another embodiment, the bacteria and bacterial populations is capable of localizing to the photosynthetic tissues, for example, leaves and shoots of the plant. In other cases, the bacteria and bacterial populations is localized to the vascular tissues of the plant, for example, in the xylem and phloem. In still another embodiment, the bacteria and bacterial populations is capable of localizing to the reproductive tissues (flower, pollen, pistil, ovaries, stamen, fruit) of the plant. In another embodiment, the bacteria and bacterial populations is capable of localizing to the root, shoots, leaves and reproductive tissues of the plant. In still another embodiment, the bacteria and bacterial populations colonizes a fruit or seed tissue of the plant. In still another embodiment, the bacteria and bacterial populations is able to colonize the plant such that it is present in the surface of the plant (i.e., its presence is detectably present on the plant exterior, or the episphere of the plant). In still other embodiments, the bacteria and bacterial populations is capable of localizing to substantially all, or all, tissues of the plant. In certain embodiments, the bacteria and bacterial populations is not localized to the root of a plant. In other cases, the bacteria and bacterial populations is not localized to the photosynthetic tissues of the plant.


The effectiveness of the compositions can also be assessed by measuring the relative maturity of the crop or the crop heating unit (CHU). For example, the bacterial population can be applied to corn, and corn growth can be assessed according to the relative maturity of the corn kernel or the time at which the corn kernel is at maximum weight. The crop heating unit (CHU) can also be used to predict the maturation of the corn crop. The CHU determines the amount of heat accumulation by measuring the daily maximum temperatures on crop growth.


In examples, bacterial may localize to any one of the tissues in the plant, including: the root, adventitious root, seminal root, root hair, shoot, leaf, flower, bud tassel, meristem, pollen, pistil, ovaries, stamen, fruit, stolon, rhizome, nodule, tuber, trichome, guard cells, hydathode, petal, sepal, glume, rachis, vascular cambium, phloem, and xylem. In another embodiment, the bacteria or bacterial population is capable of localizing to the photosynthetic tissues, for example, leaves and shoots of the plant. In other cases, the bacteria and bacterial populations is localized to the vascular tissues of the plant, for example, in the xylem and phloem. In another embodiment, the bacteria or bacterial population is capable of localizing to reproductive tissues (flower, pollen, pistil, ovaries, stamen, or fruit) of the plant. In another embodiment, the bacteria and bacterial populations is capable of localizing to the root, shoots, leaves and reproductive tissues of the plant. In another embodiment, the bacteria or bacterial population colonizes a fruit or seed tissue of the plant. In still another embodiment, the bacteria or bacterial population is able to colonize the plant such that it is present in the surface of the plant. In another embodiment, the bacteria or bacterial population is capable of localizing to substantially all, or all, tissues of the plant. In certain embodiments, the bacteria or bacterial population is not localized to the root of a plant. In other cases, the bacteria and bacterial populations is not localized to the photosynthetic tissues of the plant.


The effectiveness of the bacterial compositions applied to crops can be assessed by measuring various features of crop growth including, but not limited to, planting rate, seeding vigor, root strength, drought tolerance, plant height, dry down, and test weight.


Plant Species


The methods and bacteria described herein are suitable for any of a variety of plants, such as plants in the genera Hordeum, Oryza, Zea, and Triticeae. Other non-limiting examples of suitable plants include mosses, lichens, and algae. In some cases, the plants have economic, social and/or environmental value, such as food crops, fiber crops, oil crops, plants in the forestry or pulp and paper industries, feedstock for biofuel production and/or ornamental plants. In some examples, plants may be used to produce economically valuable products such as a grain, a flour, a starch, a syrup, a meal, an oil, a film, a packaging, a nutraceutical product, a pulp, an animal feed, a fish fodder, a bulk material for industrial chemicals, a cereal product, a processed human-food product, a sugar, an alcohol, and/or a protein. Non-limiting examples of crop plants include maize, rice, wheat, barley, Sorghum, millet, oats, rye triticale, buckwheat, sweet corn, sugar cane, onions, tomatoes, strawberries, and asparagus. In some embodiments, the methods and bacteria described herein are suitable for any of a variety of transgenic plants, non-transgenic plants, and hybrid plants thereof.


In some examples, plants that may be obtained or improved using the methods and composition disclosed herein may include plants that are important or interesting for agriculture, horticulture, biomass for the production of biofuel molecules and other chemicals, and/or forestry. Some examples of these plants may include pineapple, banana, coconut, lily, grasspeas and grass; and dicotyledonous plants, such as, for example, peas, alfalfa, tomatillo, melon, chickpea, chicory, clover, kale, lentil, soybean, tobacco, potato, sweet potato, radish, cabbage, rape, apple trees, grape, cotton, sunflower, thale cress, canola, citrus (including orange, mandarin, kumquat, lemon, lime, grapefruit, tangerine, tangelo, citron, and pomelo), pepper, bean, lettuce, Panicum virgatum (switch), Sorghum bicolor (Sorghum, sudan), Miscanthus giganteus (miscanthus), Saccharum sp. (energycane), Populus balsamifera (poplar), Zea mays (corn), Glycine max (soybean), Brassica napus (canola), Triticum aestivum (wheat), Gossypium hirsutum (cotton), Oryza sativa (rice), Helianthus annuus (sunflower), Medicago sativa (alfalfa), Beta vulgaris (sugarbeet), Pennisetum glaucum (pearl millet), Panicum spp. Sorghum spp., Miscanthus spp., Saccharum spp., Erianthus spp., Populus spp., Secale cereale (rye), Salix spp. (willow), Eucalyptus spp. (eucalyptus), Triticosecale spp. (Triticum-25 wheat X rye), Bamboo, Carthamus tinctorius (safflower), Jatropha curcas (Jatropha), Ricinus communis (castor), Elaeis guineensis (oil palm), Phoenix dactylifera (date palm), Archontophoenix cunninghamiana (king palm), Syagrus romanzoffiana (queen palm), Linum usitatissimum (flax), Brassica juncea, Manihot esculenta (cassaya), Lycopersicon esculentum (tomato), Lactuca saliva (lettuce), Musa paradisiaca (banana), Solanum tuberosum (potato), Brassica oleracea (broccoli, cauliflower, brussel sprouts), Camellia sinensis (tea), Fragaria ananassa (strawberry), Theobroma cacao (cocoa), Coffea arabica (coffee), Vitis vinifera (grape), Ananas comosus (pineapple), Capsicum annum (hot & sweet pepper), Allium cepa (onion), Cucumis melo (melon), Cucumis sativus (cucumber), Cucurbita maxima (squash), Cucurbita moschata (squash), Spinacea oleracea (spinach), Citrullus lanatus (watermelon), Abelmoschus esculentus (okra), Solanum melongena (eggplant), Papaver somniferum (opium poppy), Papaver orientale, Taxus baccata, Taxus brevifolia, Artemisia annua, Cannabis saliva, Camptotheca acuminate, Catharanthus roseus, Vinca rosea, Cinchona officinalis, Coichicum autumnale, Veratrum californica, Digitalis lanata, Digitalis purpurea, Dioscorea 5 spp., Andrographis paniculata, Atropa belladonna, Datura stomonium, Berberis spp., Cephalotaxus spp., Ephedra sinica, Ephedra spp., Erythroxylum coca, Galanthus womorii, Scopolia spp., Lycopodium serratum (Huperzia serrata), Lycopodium spp., Rauwolfia serpentina, Rauwolfia spp., Sanguinaria canadensis, Hyoscyamus spp., Calendula officinalis, Chrysanthemum parthenium, Coleus forskohlii, Tanacetum parthenium, Parthenium argentatum (guayule), Hevea spp. (rubber), Mentha spicata (mint), Mentha piperita (mint), Bixa orellana, Alstroemeria spp., Rosa spp. (rose), Dianthus caryophyllus (camation), Petunia spp. (petunia), Poinsettia pulcherrima (Poinsettia), Nicotiana tabacum (tobacco), Lupinus albus (lupin), Uniola paniculata (oats), Hordeum vulgare (barley), and Lolium spp. (rye).


In some examples, a monocotyledonous plant may be used. Monocotyledonous plants belong to the orders of the Alismatales, Arales, Arecales, Bromeliales, Commelinales, Cyclanthales, Cyperales, Eriocaulales, Hydrocharitales, Juncales, Lilliales, Najadales, Orchidales, Pandanales, Poales, Restionales, Triuridales, Typhales, and Zingiberales. Plants belonging to the class of the Gymnospermae are Cycadales, Ginkgoales, Gnetales, and Pinales. In some examples, the monocotyledonous plant can be selected from the group consisting of a maize, rice, wheat, barley, and sugarcane.


In some examples, a dicotyledonous plant may be used, including those belonging to the orders of the Aristochiales, Asterales, Batales, Campanulales, Capparales, Caryophyllales, Casuarinales, Celastrales, Comales, Diapensales, Dilleniales, Dipsacales, Ebenales, Ericales, Eucomiales, Euphorbiales, Fabales, Fagales, Gentianales, Geraniales, Haloragales, Hamamelidales, Middles, Juglandales, Lamiales, Laurales, Lecythidales, Leitneriales, Magniolales, Malvales, Myricales, Myrtales, Nymphaeales, Papeverales, Piperales, Plantaginales, Plumb aginales, Podostemales, Polemoniales, Polygalales, Polygonales, Primulales, Proteales, Rafflesiales, Ranunculales, Rhamnales, Rosales, Rubiales, Salicales, Santales, Sapindales, Sarraceniaceae, Scrophulariales, Theales, Trochodendrales, Umbellales, Urticales, and Violates. In some examples, the dicotyledonous plant can be selected from the group consisting of cotton, soybean, pepper, and tomato.


In some cases, the plant to be improved is not readily amenable to experimental conditions. For example, a crop plant may take too long to grow enough to practically assess an improved trait serially over multiple iterations. Accordingly, a first plant from which bacteria are initially isolated, and/or the plurality of plants to which genetically manipulated bacteria are applied may be a model plant, such as a plant more amenable to evaluation under desired conditions. Non-limiting examples of model plants include Setaria, Brachypodium, and Arabidopsis. Ability of bacteria isolated according to a method of the disclosure using a model plant may then be applied to a plant of another type (e.g. a crop plant) to confirm conferral of the improved trait.


Traits that may be improved by the methods disclosed herein include any observable characteristic of the plant, including, for example, growth rate, height, weight, color, taste, smell, changes in the production of one or more compounds by the plant (including for example, metabolites, proteins, drugs, carbohydrates, oils, and any other compounds). Selecting plants based on genotypic information is also envisaged (for example, including the pattern of plant gene expression in response to the bacteria, or identifying the presence of genetic markers, such as those associated with increased nitrogen fixation). Plants may also be selected based on the absence, suppression or inhibition of a certain feature or trait (such as an undesirable feature or trait) as opposed to the presence of a certain feature or trait (such as a desirable feature or trait).


Non-Genetically Modified Maize


The methods and bacteria described herein are suitable for any of a variety of non-genetically modified maize plants or part thereof. And in some aspects the corn is organic. Furthermore, the methods and bacteria described herein are suitable for any of the following non-genetically modified hybrids, verities, lineages, etc. In some embodiments, corn varieties generally fall under six categories: sweet corn, flint corn, popcorn, dent corn, pod corn, and flour corn.


Sweet Corn


Yellow su varieties include Earlivee, Early Sunglow, Sundance, Early Golden Bantam, Iochief, Merit, Jubilee, and Golden Cross Bantam. White su varieties include True Platinum, Country Gentleman, Silver Queen, and Stowell's Evergreen. Bicolor su varieties include Sugar & Gold, Quickie, Double Standard, Butter & Sugar, Sugar Dots, Honey & Cream. Multicolor su varieties include Hookers, Triple Play, Painted Hill, Black Mexican/Aztec.


Yellow se varieties include Buttergold, Precocious, Spring Treat, Sugar Buns, Colorow, Kandy King, Bodacious R/M, Tuxedo, Incredible, Merlin, Miracle, and Kandy Korn EH. White se varieties include Spring Snow, Sugar Pearl, Whiteout, Cloud Nine, Alpine, Silver King, and Argent. Bicolor se varieties include Sugar Baby, Fleet, Bon Jour, Trinity, Bi-Licious, Temptation, Luscious, Ambrosia, Accord, Brocade, Lancelot, Precious Gem, Peaches and Cream Mid EH, and Delectable R/M. Multicolor se varieties include Ruby Queen.


Yellow sh2 varieties include Extra Early Super Sweet, Takeoff, Early Xtra Sweet, Raveline, Summer Sweet Yellow, Krispy King, Garrison, Illini Gold, Challenger, Passion, Excel, Jubilee SuperSweet, Illini Xtra Sweet, and Crisp 'N Sweet. White sh2 varieties include Summer Sweet White, Tahoe, Aspen, Treasure, How Sweet It Is, and Camelot. Bicolor sh2 varieties include Summer Sweet Bicolor, Radiance, Honey 'N Pearl, Aloha, Dazzle, Hudson, and Phenomenal.


Yellow sy varieties include Applause, Inferno, Honeytreat, and Honey Select. White sy varieties include Silver Duchess, Cinderella, Mattapoisett, Avalon, and Captivate. Bicolor sy varieties include Pay Dirt, Revelation, Renaissance, Charisma, Synergy, Montauk, Kristine, Serendipity/Providence, and Cameo.


Yellow augmented supersweet varieties include Xtra-Tender 1ddA, Xtra-Tender 11dd, Mirai 131Y, Mirai 130Y, Vision, and Mirai 002. White augmented supersweet varieties include Xtra-Tender 3dda, Xtra-Tender 31dd, Mirai 421W, XTH 3673, and Devotion. Bicolor augmented supersweet varieties include Xtra-Tender 2dda, Xtra-Tender 21dd, Kickoff XR, Mirai 308BC, Anthem XR, Mirai 336BC, Fantastic XR, Triumph, Mirai 301BC, Stellar, American Dream, Mirai 350BC, and Obsession.


Flint Corn


Flint corn varieties include Bronze-Orange, Candy Red Flint, Floriani Red Flint, Glass Gem, Indian Ornamental (Rainbow), Mandan Red Flour, Painted Mountain, Petmecky, Cherokee White Flour,


PopCorn


Pop corn varieties include Monarch Butterfly, Yellow Butterfly, Midnight Blue, Ruby Red, Mixed Baby Rice, Queen Mauve, Mushroom Flake, Japanese Hull-less, Strawberry, Blue Shaman, Miniature Colored, Miniature Pink, Pennsylvania Dutch Butter Flavor, and Red Strawberry.


Dent Corn


Dent corn varieties include Bloody Butcher, Blue Clarage, Ohio Blue Clarage, Cherokee White Eagle, Hickory Cane, Hickory King, Jellicorse Twin, Kentucky Rainbow, Daymon Morgan's Knt. Butcher, Leaming, Leaming's Yellow, McCormack's Blue Giant, Neal Paymaster, Pungo Creek Butcher, Reid's Yellow Dent, Rotten Clarage, and Tennessee Red Cob.


In some embodiments, corn varieties include P1618W, P1306W, P1345, P1151, P1197, P0574, P0589, and P0157. W=white corn.


In some embodiments, the methods and bacteria described herein are suitable for any hybrid of the maize varieties setforth herein.


Genetically Modified Maize


The methods and bacteria described herein are suitable for any of a hybrid, variety, lineage, etc. of genetically modified maize plants or part thereof.


Furthermore, the methods and bacteria described herein are suitable for any of the following genetically modified maize events, which have been approved in one or more countries: 32138 (32138 SPT Maintainer), 3272 (ENOGEN), 3272 x Bt11, 3272 x bt11 x GA21, 3272 x Bt11 x MIR604, 3272 x Bt11 x MIR604 x GA21, 3272 x Bt11 x MIR604 x TC1507 x 5307 x GA21, 3272 x GA21, 3272 x MIR604, 3272 x MIR604 x GA21, 4114, 5307 (AGRISURE Duracade), 5307 x GA21, 5307 x MIR604 x Bt11 x TC1507 x GA21 (AGRISURE Duracade 5122), 5307 x MIR604 x Bt11 x TC1507 x GA21 x MIR162 (AGRISURE Duracade 5222), 59122 (HERCULEX RW), 59122 x DAS40278, 59122 x GA21, 59122 x MIR604, 59122 x MIR604 x GA21, 59122 x MIR604 x TC1507, 59122 x MIR604 x TC1507 x GA21, 59122 x MON810, 59122 x MON810 x MIR604, 59122 x MON810 x NK603, 59122 x MON810 x NK603 x MIR604, 59122 x MON88017, 59122 x MON88017 x DAS40278, 59122 x NK603 (Herculex RW ROUNDUP READY 2), 59122 x NK603 x MIR604, 59122 x TC1507 x GA21, 676, 678, 680, 3751 IR, 98140, 98140 x 59122, 98140 x TC1507, 98140 x TC1507 x 59122, Bt10 (Bt10), Bt11 [X4334CBR, X4734CBR] (AGRISURE CB/LL), Bt11 x 5307, Bt11 x 5307 x GA21, Bt11 x 59122 x MIR604, Br11 x 59122 x MIR604 x GA21, Bt11 x 59122 x MIR604 x TC1507, M53, M56, DAS-59122-7, Bt11 x 59122 x MIR604 x TC1507 x GA21, Bt11 x 59122 x TC1507, TC1507 x DAS-59122-7, Bt11 x 59122 x TC1507 x GA21, Bt11 x GA21 (AGRISURE GT/CB/LL), Bt11 x MIR162 (AGRISURE Viptera 2100), BT11 x MIR162 x 5307, Bt11 x MIR162 x 5307 x GA21, Bt11 x MIR162 x GA21 (AGRISURE Viptera 3110), Bt11 x MIR162 x MIR604 (AGRISURE Viptera 3100), Bt11 x MIR162 x MIR604 x 5307, Bt11 x MIR162 x MIR604 x 5307 x GA21, Bt11 x MIR162 x MIR604 x GA21 (AGRISURE Viptera 3111/AGRISURE Viptera 4), Bt1, MIR162 x MIR604 x MON89034 x 5307 x GA21, Bt11 x MIR162 x MIR604 x TC1507, Bt11 x MIR162 x MIR604 x TC1507 x 5307, Bt11 x MIR162 x MIR604 x TC1507 x GA21, Bt11 x MIR162 x MON89034, Bt11 x MIR162 x MON89034 x GA21, Bt11 x MIR162 x TC1507, Bt11 x MIR162 x TC1507 x 5307, Bt11 x MIR162 x TC1507 x 5307 x GA21, Bt11 x MR162 x TC1507 x GA21 (AGRISURE Viptera 3220), BT11 x MIR604 (Agrisure BC/LL/RW), Bt1 x MIR604 x 5307, Bt11 x MIR604 x 5307 x GA21, Bt11 x MIR604 x GA21, Bt11 x MIR604 x TC1507, Bt11 x MIR604 x TC1507 x 5307, Bt11 x MIR604 x TC1507 x GA21, Bt11 x MON89Ø34 x GA21, Bt11 x TC1507, Bt11 x TC1507 x 5307, Bt11 x TC1507 x GA21, Bt176 [176] (NaturGard KnockOut/Maximizer), BVLA430101, CBH-351 (STARLINK Maize), DAS40278 (ENLIST Maize), DAS40278 x NK603, DBT418 (Bt Xtra Maize), DLL25 [B16], GA21 (ROUNDUP READY Maize/AGRISURE GT), GA21 x MON810 (ROUNDUP READY Yieldgard Maize), GA21 x T25, HCEM485, LY038 (MAVERA Maize), LY038 x MON810 (MAVERA Yieldgard Maize), MIR162 (AGRISURE Viptera), MIR162 x 5307, MIR162 x 5307 x GA21, MIR162 x GA21, MIR162 x MIR604, MIR162 x MIR604 x 5307, MIR162 x MIR604 x 5307 x GA21, MIR162 x MIR604 x GA21, MIR162 x MIR604 x TC1507 x 5307, MIR162 x MIR604 x TC1507 x 5307 x GA21, MIR162 x MIR604 x TC1507 x GA21, MIR162 x MON89Ø34, MIR162 x NK603, MIR162 x TC1507, MIR162 x TC1507 x 5307, MIR162 x TC1507 x 5307 x GA21, MIR162 x TC1507 x GA21, MIR604 (AGRISURE RW), MIR604 x 5307, MIR604 x 5307 x GA21, MIR604 x GA21 (AGRISURE GT/RW), MIR604 x NK603, MIR604 x TC1507, MIR604 x TC1507 x 5307, MIR604 x TC1507 x 5307 xGA21, MIR604 x TC1507 x GA21, MON801 [MON80100], MON802, MON809, MON810 (YIELDGARD, MAIZEGARD), MON810 x MIR162, MON810 x MIR162 x NK603, MON810 x MIR604, MON810 x MON88017 (YIELDGARD VT Triple), MON810 x NK603 x MIR604, MON832 (ROUNDUP READY Maize), MON863 (YIELDGARD Rootworm RW, MAXGARD), MON863 x MON810 (YIELDGARD Plus), MON863 x MON810 x NK603 (YIELDGARD Plus with RR), MON863 x NK603 (YIELDGARD RW+RR), MON87403, MON87411, MON87419, MON87427 (ROUNDUP READY Maize), MON87427 x 59122, MON87427 x MON88017, MON87427 x MON88017 x 59122, MON87427 x MON89034, MON87427 x MON89034 x 59122, MON87427 x MON89034 x MIR162 x MON87411, MON87427 x MON89034 x MON88017, MON87427 x MON89034 x MON88017 x 59122, MON87427 x MON89034 x NK603, MON87427 x MON89034 x TC1507, MON87427 x MON89034 x TC1507 x 59122, MON87427 x MON89034 x TC1507 x MON87411 x 59122, MON87427 x MON89034 x TC1507 x MON87411 x 59122 x DAS40278, MON87427 x MON89034 x TC1507 x MON88017, MON87427 x MON89Ø34 x MIR162 x NK603, MON87427 x MON89Ø034 x TC15Ø7 x MON88Ø17 x 59122, MON87427 x TC1507, MON87427 x TC1507 x 59122, MON87427 x TC1507 x MON88017, MON87427 x TC1507 x MON88017 x 59122, MON87460 (GENUITY DROUGHTGARD), MON87460 x MON88017, MON87460 x MON89034 x MON88017, MON87460 x MON89034 x NK603, MON87460 x NK603, MON88017, MON88017 x DAS40278, MON89034, MON89034 x 59122, MON89034 x 59122 x DAS40278, MON89034 x 59122 x MON88017, MON89034 x 59122 x MON88017 x DAS40278, MON89034 x DAS40278, MON89034 x MON87460, MON89034 x MON88017 (GENUITY VT Triple Pro), MON89034 x MON88017 x DAS40278, MON89034 x NK603 (GENUITY VT Double Pro), MON89034 x NK603 x DAS40278, MON89034 x TC1507, MON89034 x TC1507 x 59122, MON89034 x TC1507 x 59122 x DAS40278, MON89034 x TC1507 x DAS40278, MON89034 x TC1507 x MON88017, MON89034 x TC1507 x MON88017 x 59122 (GENUITY SMARTSTAX), MON89034 x TC1507 x MON88017 x 59122 x DAS40278, MON89034 x TC1507 x MON88017 x DAS40278, MON89034 x TC1507 x NK603 (POWER CORE), MON89034 x TC1507 x NK603 x DAS40278, MON89034 x TC1507 x NK603 x MIR162, MON89034 x TC1507 x NK603 x MIR162 x DAS40278, MON89034 x GA21, MS3 (INVIGOR Maize), MS6 (INVIGOR Maize), MZHG0JG, MZIR098, NK603 (ROUNDUP READY 2 Maize), NK603 x MON810 x 4114 x MIR604, NK603 x MON810 (YIELDGARD CB+RR), NK603 x T25 (ROUNDUP READY LIBERTY LINK Maize), T14 (LIBERTY LINK Maize), T25 (LIBERTY LINK Maize), T25 x MON810 (LIBERTY LINK YIELDGARD Maize), TC1507 (HERCULEX I, HERCULEX CB), TC1507 x 59122 x MON810 x MIR604 x NK603 (OPTIMUM INTRASECT XTREME), TC1507 x MON810 x MIR604 x NK603, TC1507 x 5307, TC1507 x 5307 x GA21, TC1507 x 59122 (HERCULEX XTRA), TC1507 x 59122 x DAS40278, TC1507 x 59122 x MON810, TC1507 x 59122 x MON810 x MIR604, TC1507 x 59122 x MON810 x NK603 (OPTIMUM INTRASECT XTRA), TC1507 x 59122 x MON88017, TC1507 x 59122 x MON88017 x DAS40278, TC1507 x 59122 x NK603 (HERCULEX XTRA RR), TC1507 x 59122 x NK603 x MIR604, TC1507 x DAS40278, TC1507 x GA21, TC1507 x MIR162 x NK603, TC1507 x MIR604 x NK603 (OPTIMUM TRISECT), TC1507 x MON810, TC1507 x MON810 x MIR162, TC1507 x MON810 x MIR162 x NK603, TC1507 x MON810 x MIR604, TC1507 x MON810 x NK603 (OPTIMUM INTRASECT), TC1507 x MON810 x NK603 x MIR604, TC1507 x MON88017, TC1507 x MON88017 x DAS40278, TC1507 x NK603 (HERCULEX I RR), TC1507 x NK603 x DAS40278, TC6275, and VCO-01981-5.


Additional Genetically Modified Plants


The methods and bacteria described herein are suitable for any of a variety of genetically modified plants or part thereof.


Furthermore, the methods and bacteria described herein are suitable for any of the following genetically modified plant events which have been approved in one or more countries.









TABLE 14







Rice Traits, which can be combined


with microbes of the disclosure



Oryza
sativa Rice










Event
Company
Description





CL112 , CL141
BASF Inc.
Tolerance to the imidazolinone


CFX51

herbicide, imazethapyr, induced




by chemical mutagenesis of the




acetolactate synthase (ALS)




enzyme using ethyl




methanesulfonate (EMS).


IMINTA-1,
BASF Inc.
Tolerance to imidazolinone


IMINTA-4

herbicides induced by chemical




mutagenesis of the acetolactate




synthase (ALS) enzyme using




sodium azide.


LLRICE06,
Aventis
Glufosinate ammonium herbicide


LLRICE62
CropScience
tolerant rice produced by




inserting a modified




phosphinothricin




acetyltransferase (PAT) encoding




gene from the soil bacterium





Streptomyces
hygroscopicus).



LLRICE601
Bayer
Glufosinate ammonium herbicide



CropScience
tolerant rice produced by



Aventis
inserting a modified.



CmpScience
phosphinothricin



(AgrEvo))
acetyltransferase (PAT) encoding




gene from the soil bacterium





Streptomyces
hygroscopicus).



PWC16
BASF Inc.
Tolerance to the imidazolinone




herbicide, imazethapyr, induced




by chemical mutagenesis of the




acetolactate synthase (ALS)




enzyme using ethyl




methanesulfonate (EMS).
















TABLE 15







Alfalfa Traits, which can be combined with microbes of the disclosure



Medicago saliva Alfalfa










Event
Company
Description





J101, J163
Monsanto Company and
Glyphosate herbicide tolerant



Forage Genetics
alfalfa (lucerne) produced by



International
inserting a gene encoding the




enzyme 5-enolypyruvylshikimate-




3-phosphate synthase (EPSPS)




from the CP4 strain of





Agrobacterium tumefaciens.

















TABLE 16







Wheat Traits,


which can be combined with microbes of the disclosure



Triticum aestivum Wheat










Event
Company
Description





AP205CL
BASF Inc.
Selection for a mutagenized




version of the enzyme




acetohydroxyacid synthase




(AHAS), also known as




acetolactate synthase (ALS) or




acetolactate pyruvate-lyase.


AP602CL
BASF Inc.
Selection for a mutagenized




version of the enzyme




ace tohydroxyacid synthase




(AHAS), also known as




acetolactate synthase (ALS) or




acetolactate pyruvate-lyase.


BW255-2, BW238-3
BASF Inc.
Selection for a mutagenized




version of the enzyme




acetohydroxyacid synthase




(AHAS), also known as




acetolactate synthase (ALS) or




acetolactate pyruvate-lyase.


BW7
BASF Inc.
Tolerance to imidazolinone




herbicides induced by chemical




mutagenesis of the




acetohydroxyacid synthase




(AHAS) gene using sodium azide.


MON71800
Monsanto
Glyphosate tolerant wheat variety



Company
produced by inserting a modified




5-enolpyruvylshikimate-3-




phosphate synthase (EPSPS)




encoding gene from the soil




bacterium Agrobacterium





tumefaciens, strain CP4.



SWP965001
Cyanamid
Selection for a mutagenized



Crop
version of the enzyme



Protection
acetohydroxyacid synthase




(AHAS), also known as




acetolactate synthase (ALS) or




acetolactate pyruvate-lyase.


Teal 11A
BASF Inc.
Selection for a mutaaenized




version of the enzyme




ace tohydroxyacid synthase




(AHAS), also known as




acetolactate synthase (ALS) or




acctolactate pyruvate-lyase.
















TABLE 17







Sunflower Traits, which can be combined


with microbes of the disclosure



Helianthus
annuus Sunflower












Event
Company
Description







X81359
BASF Inc,
Tolerance to imidazolinone





herbicides by selection of a





naturally occurring mutant.

















TABLE 18







Soybean Traits, which can be combined


with microbes of the disclosure



Glycine max L. Soybean










Event
Company
Description





A2704-12,
Bayer
Glufosinate ammonium herbicide


A2704-21,
CropScience
tolerant soybean produced by


A5547-35
(Aventis
inserting a modified



CropScience
phosphinothricin acetyltransferase



(AgrEvo))
(PAT) encoding gene from the soil




bacterium Streptomyces





viridochromogenes.



A5547-127
Bayer
Glufosinate ammonium herbicide



CropScience
tolerant soybean produced by



(Aventis
inserting a modified



CropScience
phosphinothricin acetyltransferase



(AgrEvo))
(PAT) encoding gene from the soil




bacterium Streptomyces





viridochromogenes.



BPS-CV127-9
BASF Inc.
The introduced csr1-2 gene from





Arabidopsis thaliana encodes an





acetohydroxyacid synthase protein




that confers tolerance to




imidazolinone herbicides due to a




point mutation that results in a




single amino acid substitution in




which the serine residue at position




653 is replaced by asparagine




(S653N)


DP-305423
Pioneer Hi-Bred
High oleic acid soybean produced



International Inc.
by inserting additional copies of a




portion of the omega 6 desaturase




encoding gene, gm-fad2-1




resulting in silencing of the




endogenous omega-6 desaturase




gene (FAD2-1).


DP356043
Pioneer Hi-Bred
Soybean event with two herbicide



International Inc.
tolerance genes: glyphosate N-




acetlytransferase, which detoxifies




glyphosate, and a modified




acetolactate synthase (ALS) gene




which is tolerant to ALS-inhibiting




herbicides.


G94-1,
DuPont
High oleic acid soybean produced


G94-19,
Canada
by inserting a second copy of the


G168
Agricultural
fatty acid desaturase (Gm Fad2-1)



Products
encoding gene from soybean,




which resulted in “silencing” of




the endogenous host gene.


GTS 40-3-2
Monsanto
Glyphosate tolerant soybean



Company
variety produced by inserting a




modified 5-enolpyruvylshikimate-




3-phosphate synthase (EPSPS)




encoding gene from the soil




bacterium Agrobacterium





tumefaciens.



GU262
Bayer
Gluthsinate ammonium herbicide



CmpScience
tolerant soybean produced by



(Aventis
inserting a modified



CropScience
phosphinothricin acetyltransferase



(AgrEvo))
(PAT) encoding gene from the soil




bacterium Streptomyces





viridochromogenes.



MON87701
Monsanto
Resistance to Lepidopteran pests



Company
of soybean including velvetbean




caterpillar (Anticarsia gemmatalis)




and soybean looper (Pseudoplusia





includens).



MON87701 x
Monsanto
Glyphosate herbicide tolerance


MON89788
Company
through expression of the EPSPS




encoding gene from A. tumefaciens




strain CP4, and resistance to




Lepidopteran pests of soybean




including velvetbean caterpillar




(Anticarsia gemmatatis) and




soybean looper (Pseudoplusia





includens) via expression of the





Cry1Ac encoding gene from B.





thuringiensis.



MON89788
Monsanto
Glyphosate-tolerant soybean



Company
produced by inserting a modified




5-enolpyrtivylshikimate-3-




phosphate synthase (EPSPS)




encoding aroA (epsps) gene from





Agrobacterium tumefaciens CP4.



OT96-15
Agriculture &
Low linolenic acid soybean



Agri-Food
produced through traditional cross-



Canada
breeding to incorporate the novel




trait from a naturally occurring




fan1 gene mutant that was selected




for low linolenic acid.


W62, W98
Bayer
Glufosinate ammonium herbicide



CropScience
tolerant soybean produced by



(Aventis
inserting a modified



CropScience
phosphinothricin acetyltransferase



(AgrEvo))
(PAT) encoding gene from the soil




bacterium Streptomyces





hygroscopicus.

















TABLE 19







Corn Traits, which can be combined with microbes of the disclosure


Zea mays L. Maize









Event
Company
Description





176
Syngenta Seeds, Inc.
Insect-resistant maize produced by




inserting the Cry1Ab gene from





Bacillus thuringiensis subsp.






kurstaki. The genetic modification





affords resistance to attack by the




European corn borer (ECB).


3751 IR
Pioneer Hi-Bred
Selection of somaclonal variants


676, 678, 680
International Inc.
by culture of embiyos on



Pioneer Hi-Bred
imidazolinone containing media.



International Inc.
Male-sterile and gluthsinate




ammonium herbicide tolerant




maize produced by inserting genes




encoding DNA adenine m.eth.ylase




and phosphinothricin




acetyltransferase (PAT) from





Escherichia coli and Streptomyces






viridochromogenes, respectively.



B16 (DLL25)
Dekalb Genetics
Glufosinate ammonium herbicide



Corporation
tolerant maize produced by




inserting the gene encoding




phosphinothricin acetyltransferase




(PAT) from Streptomyces





hygroscopicus.



BT11 (X4334CBR,
Syngenta Seeds, Inc.
Insect-resistant and herbicide


X4734CBR)

tolerant maize produced by




inserting the Cry1Ab gene from





Bacillus thuringiensis subsp.






kurstaki, and the phosphinothricin





N-acetyltransferase (PAT)




encoding gene from S.





viriclochromogenes.



BT11 x GA21
Svngenta Seeds, Inc.
Stacked insect resistant and




herbicide tolerant maize produced




by conventional cross breeding of




parental lines BT11 (OECD unique




identifier: SYN-BTO11-1) and




GA21 (OECD unique identifier:




MON-OOO21-9).


BT11 x MIR162 x
Syngenta Seeds, Inc.
Resistance to Coleopteran pests,


MIR604 x GA21

particularly corn rootworm pests




(Diabrotica spp.) and several




Lepidopteran pests of corn,




including European corn borer




(ECB, Ostrinianubilalis), corn




carworm (CEW, Helicoverpa zea),




fall army worm (FAW, Spodoptera





frugiperda), and black cutworm





(BCW, Agrotisipsilon): tolerance




to glyphosate and glufosinate-




ammonium containing herbicides.


BT11 x MIR162
Syngenta Seeds, Inc.
Stacked insect resistant and




herbicide tolerant maize produced




by conventional cross breeding of




parental lines BT11 (OECD unique




identifier: SYN-BT011-1) and




MIR162 (OECD unique identifier:




SYN-1R162-4). Resistance to the




European Corn Borer and




tolerance to the herbicide




glufosinate ammonium (Liberty) is




derived from BT11, which




contains the Cry1Ab gene from





Bacillus thuringiensis subsp.






kurstaki, and the phosphinothricin





N-acetyltransferase (PAT)




encoding gene from S.





viridochromogenes. Resistance to





other Lepidopteran pests, including





H. zea, S. albicosta, A. ipsilon,





and S. albicosta, is derived from




MIR162, which contains the




vip3Aa gene from Bacillus





thuringiensis strain AB88.



BT11 x MIR162 x
Syngenta Seeds, Inc.

Bacillus thuringiensis Cry1Ab



MIR604

delta-endotoxin protein and the




genetic material necessary for its




production via elements of vector




pZO1502) in Event Bill corn




(OECD Unique Identifier:




SYNBTO11-1) x Bacillus





thuringiensis Vip3Aa20





insecticidal protein and the genetic




material necessary for its




production via elements of vector




pNOV1300) in Event MIR162




maize (OECD Unique Identifier:




SYN-IR162-4) x modified Cry3A




protein and the genetic material




necessary for its production via




elements of vector pZM26) in




Event MIR604 corn (OECD




Unique Identifier: SYN-1R604-5).


CBH-351
Aventis CropScience
Insect-resistant and glufosinate




ammonium herbicide tolerant




maize developed by inserting




genes encoding Cry9C protein




from Bacillus thuringiensis subsp




tolworthi and phosphinothricin




acetyltransferase (PAT) from




Streptornyces hygroscopicia.


DAS-06275-8
DOW AgroSciences LLC
Lepidopteran insect resistant and




glufosinate ammonium herbicide-




tolerant maize variety produced by




inserting the Cry1F gene from





Bacillus thuringiensis var aizawai





and the phosphinothricin




acetyltransferase (PAT) from





Streptomyces
hygroscopicus.



BT11 x MIR604
Syngenta Seeds, Inc.
Stacked insect resistant and




herbicide tolerant maize produced




by conventional cross breeding of




parental lines BT11 (OECD unique




identifier: SYN-BTO11-1) and




MIR604 (OECD unique identifier:




SYN-IR6O5-5). Resistance to the




European Corn Borer and




tolerance to the herbicide




glufosinate ammonium (Liberty) is




derived from Bill, which




contains the Cry1Ab gene from





Bacillus thuringiensis subsp.






kurstaki, and the phosphinothricin





N-acetyltransferase (PAT)




encoding gene from S.





viridochromogenes. Corn





rootworm -resistance is derived




from MIR604 which contains the




mCry3A gene from Bacillus





thuringiensis.



BT11 x MIR604 x
Syngenta Seeds, Inc.
Stacked insect resistant and


GA21

herbicide tolerant maize produced




by conventional cross breeding of




parental lines BT11 (OECD unique




identifier: SYN-BTO11-1),




MIR604 (OECD unique identifier:




SYN-1R605-5) and GA21 (OECD




unique identifier: MON-




00021-9). Resistance to the




European Corn Borer and




tolerance to the herbicide




glufosinate ammonium (Liberty) is




derived from BT11, which




contains the Cry1Ab gene from





Bacillus thuringiensis subsp.






kurstaki, and the phosphinothricin





N-acetyltransferase (PAT)




encoding gene from S.





viridochromogenes. Corn





rootworm-resistance is derived




from MIR604 which contains the




mCry3A gene from Bacillus





thuringiensis. Tolerance to





glyphosate herbicide is derived




from GA21 which contains a




modified EPSPS gene from maize.


DAS-59122-7
DOW AgroSciences LLC
Corn rootwortn-resistant maize



and Pioneer Hi-Bred
produced by inserting the



International Inc.
Cry34Ab1 and Cry35Ab1 genes




from Bacillus thuringiensis strain




PS149B1. The PAT encoding gene




from Streptomyces





viridochromogenes was introduced





as a selectable marker.


DAS-59122-7 x TC1507
DOW AgroSciences LLC
Stacked insect resistant and


x NK603
and Pioneer Hi-Bred
herbicide tolerant maize produced



International Inc.
by conventional cross breeding of




parental lines DAS-59122-7




(OECD unique identifier: DAS-




59122-7) and TC1507 (OECD




unique identifier: DAS-01507-1)




with NK603 (OECD unique




identifier: NION-00603-6). Corn




rootworm-resistance is derived




from DAS-59122-7 which




contains the Cry34Abl and




Cry35Abl genes from Bacillus





thuringiensis strain P5149B1.





Lepidopteran resistance and




tolerance to glufosinate ammonium




herbicide is derived from TC1507.




Tolerance to glyphosate herbicide




is derived from NK603.


DBT418
Dekalb Genetics
Insect-resistant and glufosina,te



Corporation
ammonium herbicide tolerant




maize developed by inserting




genes encoding Cry1AC protein




from Bacillus thuringiensis subsp





kurstaki and phosphinothricin





acetyltransferase (PAT) from





Streptomyces
hygroscopicus.



MIR604 x GA21
Syngenta Seeds, Inc.
Stacked insect resistant and




herbicide tolerant maize produced




by conventional cross breeding of




parental lines MIR604 (OECD




unique identifier: SYN-1R605-5)




and GA21 (OECD unique




identifier: MON-00021-9). Corn




rootworm-resistance is derived




from MIR604 which contains the




mCry3A gene from Bacillus





thuringiensis. Tolerance to





glyphosate herbicide is derived




from GA21.


MON80100
Monsanto Company
Insect-resistant maize produced by




inserting the Cry1Ab gene from





Bacillus thuringiensis subsp.






kurstaki. The genetic modification





affords resistance to attack by the




European corn borer (ECB).


MON802
Monsanto Company
Insect-resistant and glyphosate




herbicide tolerant maize produced




by inserting the genes encoding the




Cry1Ab protein from Bacillus





thuringiensis and the 5-





enolpyruvylshikimate-3-phosphate




synthase (EPSPS) from A.




tumefaciens strain CP4.


MON809
Pioneer Hi-Bred
Resistance to European corn borer



International Inc.
(Ostrinia nubilalis) by introduction




of a synthetic Cry lAb gene.




Glyphosate resistance via




introduction of the bacterial




version of a plant enzyme,




5-enolpynivyl shikimate-3-




phosphate synthase (EPSPS).


MON810
Monsanto Company
Insect-resistant maize produced by




inserting a truncated forril of the




Cry1Ab gene from Bacillus





thuringiensis subsp. kurstaki HD-





1. The genetic modification affords




resistance to attack by the




European corn borer (ECB).


MON810 x LY038
Monsanto Company
Stacked insect resistant and




enhanced lysine content maize




derived from conventional




crossbreeding of the parental lines




MON810 (OECD identifier:




MON-OO81O-6) and IN-038




(OECD identifier: REN-OOO38-




3).


MON810 x MON88017
Monsanto Company
Stacked insect resistant and




glyphosate tolerant maize derived




from conventional cross-breeding




of the parental lines MON810




(OECD identifier: MON-OO81O-




6) and MON88017 (OECD




identifier: MON-88017-3).




European corn borer (ECB)




resistance is derived from a




truncated form of the Cry1Ab gene




from Bacillus thuringiensis subsp.





kurstaki HD-1 present in





MON810. Corn rootworm




resistance is derived from the




Cry3Bbl gene from Bacillus





thuringiensis subspecies






kumarnotoensis strain EG4691





present in MON88017. Glyphosate




tolerance is derived from a 5-




enolpyruvylshikimate-3-phosphate




synthase (EPSPS) encoding gene




from Agrobacterium tumefaciens




strain CP4 present in MON88017.


MON832
Monsanto Company
Introduction, by particle




bombardment, of glyphosate




oxidase (GOX) and a modified 5-




enolpyruvyl shikimate-3-phosphate




synthase (EPSPS), an enzyme




involved in the shikimate




biochemical pathway for the




production of the aromatic amino




acids.


MON863
Monsanto Company
Corn rootworm resistant maize




produced by inserting the Cry3Bbl




gene from Bacillus thuringiensis




subsp. kumamotoensis.


MON863 x MON810
Monsanto Company
Stacked insect resistant corn




hybrid derived from conventional




cross-breeding of the parental lines




MON863 (OECD identifier:




MON-00863-5) and MON810




(OECD identifier: MON-00810-6)


MON863 x MON810 x
Monsanto Company
Stacked insect resistant and


Monsanto NK603

herbicide tolerant corn hybrid




derived from conventional




crossbreeding of the stacked




hybrid MON-00863-5 x MON-




00810-6 and NK603 (OECD




identifier: MON-00603-6),


MON883 x NK603
Monsanto Company
Stacked insect resistant and




herbicide tolerant corn hybrid




derived from conventional




crossbreeding of the parental lines




MON863 (OECD identifier:




MON-OO863-5) and NK603




(OECD identifier: MON-OO6O3-




6).


MON87460
Monsanto Company
MON 87460 was developed to




provide reduced yield loss under




water-limited conditions compared




to conventional maize. Efficacy in




MON 87460 is derived by




expression of the inserted Bacillus





subtilis cold shock protein B





(CspB).


MON88017
Monsanto Company
Corn rootworm-resistant maize




produced by inserting the Cry3Bbl




gene from Bacillus thuringiensis




subspecies kumamotoensis strain




EG4691, Glyphosate tolerance




derived by inserting a 5-




enolpyruvylshikimate-3-phosphate




synthase (EPSPS) encoding gene




from Agrobacterium tumefaciens




strain CP4.


MON89034
Monsanto Company
Maize event expressing two




different insecticidal proteins from





Bacillus thuringiensis providing





resistance to number of




Lepidopteran pests.


MON89034 x
Monsanto Company
Stacked insect resistant and


MON88017

glyphosate tolerant maize derived




from conventional cross-breeding




of the parental lines MON89034




(OECD identifier: MON-89O34-3)




and MON88017 (OECD identifier:




MON-88017-3). Resistance to




Lepidopteran insects is derived




from two Cry genes present in




MON89043, Corn rootworm




resistance is derived from a single




Cry genes and glyphosate




tolerance is derived from the




5-enolpyruvylshikimate-3-




phosphate synthase (EPSPS)




encoding gene from





Agrobacterium tumefaciens





present in MON88017.


MON89034 x NK603
Monsanto Company
Stacked insect resistant and




herbicide tolerant maize produced




by conventional cross breeding of




parental lines MON89034 (OECD




identifier: MON-89034-3) with




NK603 (OECD unique identifier:




MON-00603-6). Resistance to




Lepidopteran insects is derived




from two Cry genes present in




MON89043. Tolerance to




glyphosate herbicide is derived




from NK603.


NK603 x MON810
Monsanto Company
Stacked insect resistant and




herbicide tolerant corn hybrid




derived from conventional




crossbreeding of the parental lines




NK603 (OECD identifier: MON-




00603-6) and M0N810 (OECD




identifier: MON-00810-6),


MON89034 x TC1507 x
Monsanto Company and
Stacked insect resistant and


MON88017 x DAS-
Mycogen Seeds c/o Dow
herbicide tolerant maize produced


59122-7
AgroSciences LLC
by conventional cross breeding of




parental lines: MON89034,




TC1507, MON-88017, and DAS-59




122. Resistance to the above-




ground and below-ground insect




pests and tolerance to glyphosate




and glufosinate-ammonium




containing herbicides.


M53
Bayer CropScience
Male sterility caused by expression



(Aventis
of the bamase ribonuclease gene



CropScience(AgrEvo))
from Bacillus amyloliquefaciens;




PPT resistance was via PPT-




acetyltransferase (PAT).


M56
Bayer CropScience
Male sterility caused by expression



(Aventis
of the bamase ribonuclease gene



CropScience(AgrEvo)
from Bacillus amyloliquefaciens;




PPT resistance was via PPT-




acetyltransferase (PAT).


NK603
Monsanto Company
Introduction, by particle




bombardment, of a modified 5-




enolpyruvyl shikimate-3-phosphate




synthase (EPSPS), an enzyme




involved in the shikimate




biochemical pathway for the




production of the aromatic amino




acids.


NK603 x T25
Monsanto Company
Stacked glufosinate ammonium




and glyphosate herbicide tolerant




maize hybrid derived from




conventional cross-breeding of the




parental lines NK603 (OECD




identifier: MON-00603-6) and T25




(OECD identifier: ACS-ZM003-




2).


T25 x MON810
Bayer CropScience
Stacked insect resistant and



(Aventis
herbicide tolerant corn hybrid



CropScience(AgrEvo))
derived from conventional




crossbreeding of the parental lines




T25 (OECD identifier: ACS-




ZMO03-2) and MON810 (OECD




identifier: MON-OO81O-6).


TC1507
Mycogen (c/o Dow
Insect-resistant and glufosinate



AgroSciences); Pioneer
anunonium herbicide tolerant



(c/o DuPont)
maize produced by inserting the




Cry1F gene from Bacillus





thuringiensis var. aizawai and the





phosphinothricin




N-acetyltransferase encoding gene




from Streptomyces





viridochromogenes.



TC1507 x NK603
DOW AgroSciences LLC
Stacked insect resistant and




herbicide tolerant corn hybrid




derived from conventional




crossbreeding of the parental lines




1507 (OECD identifier: DAS-




O1507-1) and NK603 (OECD




identifier: MON-00603-6).


TC1507 x DAS-59122-7
DOW AgroSciences LLC
Stacked insect resistant and



and Pioneer Hi-Bred
herbicide tolerant maize produced



International Inc.
by conventional cross breeding of




parental lines TC1507 (OECD




unique identifier: DAS-01507-1)




with DAS-59122-7 (OECD unique




identifier: DAS-59122-7).




Resistance to Lepidoptemn insects




is derived from TC1507 due the




presence of the Cry1F gene from





Bacillus thuringiensis var. aizawai.





Corn rootwomi-resistance is




derived from DAS-59122-7 which




contains the Ccry34Ab1 and




Cry35Ab1 genes from Bacillus





thuringiensis strain P5149B1.





Tolerance to gluthsinate




ammonium herbicide is derived




from TC1507 from the




phosphinothricin




N-acetyltransferase encoding gene




from Streptomyces





viridochromogenes.














Event
Company
Description
Hybrid Family





P0157
Dupont Pioneer

P0157


P0157AM
Dupont Pioneer
AM LL RR2
P0157


P0157AMXT
Dupont Pioneer
ANTXT LL RR2
P0157


P0157R
Dupont Pioneer
RR2
P0157


P0339AM
Dupont Pioneer
AM LL RR2
P0339


P0339ANIXT
Dupont Pioneer
AMXT LL RR2
P0339


P0306AM
Dupont Pioneer
AM LL RR2
P0306


P0589
Dupont Pioneer

P0589


P0589AM
Dupont Pioneer
AM LL RR2
P0589


P0589AMXT
Dupont Pioneer
AMXT LL RR2
P0589


P0589R
Dupont Pioneer
RR2
P0589


P0574
Dupont Pioneer

P0574


P0574AM
Dupont Pioneer
AM LL RR2
P0574


P0574ANIXT
Dupont Pioneer
AMXT LL RR2
P0574


P0533EXR
Dupont Pioneer
HXX LL RR2
P0533


P0506AM
Dupont Pioneer
AM LL RR2
P0566


P0760AMXT
Dupont Pioneer
AMXT LL RR2
P0760


P0707AM
Dupont Pioneer
AM LL RR2
P0707


P0707AMXT
Dupont Pioneer
AMXT LL RR2
P0707


P0825AM
Dupont Pioneer
AM LL RR2
P0825


P0825AMXT
Dupont Pioneer
AMXT LL RR2
P0825


P0969AM
Dupont Pioneer
AM LL RR2
P0969


P0969AMXT
Dupont Pioneer
AMXT LL RR2
P0969


P0937AM
Dupont Pioneer
AM LL RR2
P0937


P0919AM
Dupont Pioneer
AM LL RR2
P0919


P0905EXR
Dupont Pioneer
HXX LL RR2
P0905


P1197
Dupont Pioneer

P1197


P1197AM
Dupont Pioneer
AM LL RR2
P1197


P1197AMXT
Dupont Pioneer
AMXT LL RR2
P1197


P1197R
Dupont Pioneer
RR2
P1197


P1151
Dupont Pioneer

P1151


P1151AM
Dupont Pioneer
AM LL R2
P1151


P1151R
Dupont Pioneer
RR2
P1151


P1138AM
Dupont Pioneer
AM LL RR2
P1138


P1366AM
Dupont Pioneer
AM LL RR2
P1366


P1366AMXT
Dupont Pioneer
AMXT LL RR2
P1366


P1365AMX
Dupont Pioneer
AMX LL RR2
P1365


P1353AM
Dupont Pioneer
AM LL RR2
P1353


P1345
Dupont Pioneer

P1345


P1311AMXT
Dupont Pioneer
AMXT LL RR2
P1311


P1498EHR
Dupont Pioneer
HX1 LL RR2
P1498


P1498R
Dupont Pioneer
RR2
P1498


P1443AM
Dupont Pioneer
AM LL RR2
P1443


P1555CHR
Dupont Pioneer
RW HX1 LL
P1555




RR2



P1751AMT
Dupont Pioneer
AMT LL RR2
P1751


P2089AM
Dupont Pioneer
AM LL RR2
P2089


QROME
Dupont Pioneer
Q LL RR2









The following are the definitions for the shorthand occurring in Table 19. AM-OPTIMUM ACREMAX Insect Protection system with YGCB, HX1, LL, RR2. AMT-OPTIMUM ACREMAX TRISECT Insect Protection System with RW,YGCB,HX1,LL,RR2. AMXT—(OPTIMUM ACREMAX XTreme). HXX—HERCULEX XTRA contains the Herculex I and Herculex RW genes. HX1—Contains the HERCULEX I Insect Protection gene which provides protection against European corn borer, southwestern corn borer, black cutworm, fall armyworm, western bean cutworm, lesser corn stalk borer, southern corn stalk borer, and sugarcane borer; and suppresses corn earworm. LL—Contains the LIBERTYLINK gene for resistance to LIBERTY herbicide. RR2—Contains the ROUNDUP READY Corn 2 trait that provides crop safety for over-the-top applications of labeled glyphosate herbicides when applied according to label directions. YGCB—contains the YIELDGARD Corn Borer gene offers a high level of resistance to European corn borer, southwestern corn borer, and southern cornstalk borer; moderate resistance to corn earworm and common stalk borer; and above average resistance to fall armyworm. RW—contains the AGRISURE root worm resistance trait. Q—provides protection or suppression against susceptible European corn borer, southwestern corn borer, black cutworm, fall armyworm, lesser corn stalk borer, southern corn stalk borer, stalk borer, sugarcane borer, and corn earworm; and also provides protection from larval injury caused by susceptible western corn rootworm, northern corn rootworm, and Mexican corn rootworm; contains (1) HERCULEX XTRA Insect Protection genes that produce Cry1F and Cry34ab1 and Cry35ab1 proteins, (2) AGRISURE RW trait that includes a gene that produces mCry3A protein, and (3) YIELDGARD Corn Borer gene which produces Cry1Ab protein.


Concentrations and Rates of Application of Agricultural Compositions


As aforementioned, the agricultural compositions of the present disclosure, which comprise a taught microbe, can be applied to plants in a multitude of ways. In two particular aspects, the disclosure contemplates an in-furrow treatment or a seed treatment


For seed treatment embodiments, the microbes of the disclosure can be present on the seed in a variety of concentrations. For example, the microbes can be found in a seed treatment at a cfu concentration, per seed of: 1×101, 1×102, 1×103, 1×104, 1×105, 1×106, 1×107, 1×108, 1×109, 1×1010, or more. In particular aspects, the seed treatment compositions comprise about 1×104 per to about 1×108 cfu per seed. In other particular aspects, the seed treatment compositions comprise about 1×105 to about 1×107 cfu per seed. In other aspects, the seed treatment compositions comprise about 1×106 cfu per seed.


In the United States, about 10% of corn acreage is planted at a seed density of above about 36,000 seeds per acre; ⅓ of the corn acreage is planted at a seed density of between about 33,000 to 36,000 seeds per acre; ⅓ of the corn acreage is planted at a seed density of between about 30,000 to 33,000 seeds per acre, and the remainder of the acreage is variable. See, “Corn Seeding Rate Considerations,” written by Steve Butzen, available at: https://www.pioneer.com/home/site/us/agronomy/library/corn-seeding-rate-considerations/


Table 20 below utilizes various cfu concentrations per seed in a contemplated seed treatment embodiment (rows across) and various seed acreage planting densities (1st column: 15K-41K) to calculate the total amount of cfu per acre, which would be utilized in various agricultural scenarios (i.e. seed treatment concentration per seed×seed density planted per acre). Thus, if one were to utilize a seed treatment with 1×106 cfu per seed and plant 30,000 seeds per acre, then the total cfu content per acre would be 3×1010 (i.e. 30K*1×106).









TABLE 20







Total CPU Per Acre Calculation for Seed Treatment Embodiments















Corn Population










(i.e. seeds per










acre)
1.00E+02
1.00E+03
1.00E+04
1.00E+05
1.00E+06
1.00E+07
1.00E+08
1.00E+09





15,000
1.50E+06
1.50E+07
1.50E+08
1.50E+09
1.50E+10
1.50E+11
1.50E+12
1.50E+13


16,000
1.60E+06
1.60E+07
1.60E+08
1.60E+09
1.60E+10
1.60E+11
1.60E+12
1.60E+13


17,000
1.70E+06
1.70E+07
1.70E+08
1.70E+09
1.70E+10
1.70E+11
1.70E+12
1.70E+13


18,000
1.80E+06
1.80E+07
1.80E+08
1.80E+09
1.80E+10
1.80E+11
1.80E+12
1.80E+13


19,000
1.90E+06
1.90E+07
1.90E+08
1.90E+09
1.90E+10
1.90E+11
1.90E+12
1.90E+13


20,000
2.00E+06
2.00E+07
2.00E+08
2.00E+09
2.00E+10
2.00E+11
2.00E+12
2.00E+13


21,000
2.10E+06
2.10E+07
2.10E+08
2.10E+09
2.10E+10
2.10E+11
2.10E+12
2.10E+13


22,000
2.20E+06
2.20E+07
2.20E+08
2.20E+09
2.20E+10
2.20E+11
2.20E+12
2.20E+13


23,000
2.30E+06
2.30E+07
2.30E+08
2.30E+09
2.30E+10
2.30E+11
2.30E+12
2.30E+13


24,000
2.40E+06
2.40E+07
2.40E+08
2.40E+09
2.40E+10
2.40E+11
2.40E+12
2.40E+13


25.000
2.50E+06
2.50E+07
2.50E+08
2.50E+09
2.50E+10
2.50E+11
2.50E+12
2.50E+13


26,000
2.60E+06
2.60E+07
2.60E+08
2.60E+09
2.60E+10
2.60E+11
2.60E+12
2.60E+13


27,000
2.70E+06
2.70E+07
2.70E+08
2.70E+09
2.70E+10
2.70E+11
2.70E+12
2.70E+13


28,000
2.80E+06
2.80E+07
2.80E+08
2.80E+09
2.80E+10
2.80E+11
2.80E+12
2.80E+13


29,000
2.90E+06
2.90E+07
2.90E+08
2.90E+09
2.90E+10
2.90E+11
2.90E+12
2.90E+13


30,000
3.00E+06
3.00E+07
3.00E+08
3.00E+09
3.00E+10
3.00E+11
3.00E+12
3.00E+13


31,000
3.10E406
3.10E+07
3.10E+08
3.10E+09
3.10E+10
3.10E+11
3.10E+12
3.10E+13


32,000
3.20E+06
3.20E+07
3.20E+08
3.20E+09
3.20E+10
3.20E+11
3.20E+12
3.20E+13


33,000
3.30E+06
3.30E+07
3.30E+08
3.30E+09
3.30E+10
3.30E+13
3.30E+11
3.30E+12


34,000
3.40E+06
3.40E+07
3.40E+08
3.40E+09
3.40E+10
3.40E+11
3.40E+12
3.40E+13


35,000
3.50E+06
3.50E+07
3.50E+08
3.50E+09
3.50E+10
3.50E+11
3.50E+12
3.50E+13


36,000
3.60E+06
3.60E+07
3.60E+08
3.60E+09
3.60E+10
3.60E+11
3.60E+12
3.60E+13


37,000
3.70E+06
3.70E+07
3.70E+08
3.70E+09
3.70E+10
3.70E+11
3.70E+12
3.70E+13


38,000
3.80E+06
3.80E+07
3.80E+08
3.80E+09
3.80E+10
3.80E+11
3.80E+12
3.80E+13


39,000
3.90E+06
3.90E+07
3.90E+08
3.90E+09
3.90E+10
3.90E+11
3.90E+12
3.91E+13


40,000
4.00E+06
4.00E+07
4.00E+08
4.00E+09
4.00E+10
4.00E+11
4.00E+12
4.00E+13


41,000
4.10E+06
4.10E+07
4.10E+08
4.10E+09
4.10E+10
4.10E+11
4.10E+12
4.10E+13









For in-furrow embodiments, the microbes of the disclosure can be applied at a cfu concentration per acre of: 1×106, 3.20×1010, 1.60×1011, 3.20×1011, 8.0×1011, 1.6×1012, 3.20×1012, or more. Therefore, in aspects, the liquid in-furrow compositions can be applied at a concentration of between about 1×106 to about 3×1012 cfu per acre.


In some aspects, the in-furrow compositions are contained in a liquid formulation. In the liquid in-furrow embodiments, the microbes can be present at a cfu concentration per milliliter of: 1×101, 1×102, 1×103, 1×104, 1×105, 1×106, 1×107, 1×108, 1×109, 1×1010, 1×1011, 1×1012, 1×1013, or more. In certain aspects, the liquid in-furrow compositions comprise microbes at a concentration of about 1×106 to about 1×1011 cfu per milliliter. In other aspects, the liquid in-furrow compositions comprise microbes at a concentration of about 1×107 to about 1×1010 cfu per milliliter. In other aspects, the liquid in-furrow compositions comprise microbes at a concentration of about 1×108 to about 1×109 cfu per milliliter. In other aspects, the liquid in-furrow compositions comprise microbes at a concentration of up to about 1×1013 cfu per milliliter.


Transcriptomic Profiling of Candidate Microbes


Previous work by the inventors entailed transcriptomic profiling of strain CI010 to identify promoters that are active in the presence of environmental nitrogen. Strain CI010 was cultured in a defined, nitrogen-free media supplemented with 10 mM glutamine. Total RNA was extracted from these cultures (QIAGEN RNeasy kit) and subjected to RNAseq sequencing via Illumina HiSeq (SeqMatic, Fremont CA). Sequencing reads were mapped to the CI010 genome data using Geneious, and highly expressed genes under control of proximal transcriptional promoters were identified.


Tables 21-23 lists genes and their relative expression level as measured through RNASeq sequencing of total RNA. Sequences of the proximal promoters were recorded for use in mutagenesis of nif pathways, nitrogen utilization related pathways, or other genes with a desired expression level.













TABLE 21





Name
Minimum
Maximum
Length
Direction



















murein lipoprotein CDS
2,929,898
2,930,134
237
forward


membrane protein CDS
5,217,517
5,217,843
327
forward


zinc/cadmium-binding protein
3,479,979
3,480,626
648
forward


CDS






acyl carrier protein CDS
4,563,344
4,563,580
237
reverse


ompX CDS
4,251,002
4,251,514
513
forward


DNA-binding protein HU-
375,156
375,428
273
forward


beta CDS






sspA CDS
629,998
630,636
639
reverse


tatE CDS
3,199,435
3,199,638
204
reverse


LexA repressor CDS
1,850,457
1,851,065
609
forward


hisS CDS
<3999979
4,001,223
>1245
forward






















TABLE 22










RNASeg_




Differential

RNASeq_
RNASeq_
WT -
RNASeq_



Expression
Differential
nifL - Raw
nifL - Raw
Raw
WT - Raw



Absolute
Expression
Read
Transcript
Read
Transcript


Name
Confidence
Ratio
Count
Count
Count
Count





















murein
1000
−1.8
12950.5
10078.9
5151.5
4106.8


lipoprotein








CDS








membrane
1000
−1.3
9522.5
5371.3
5400
3120


protein CDS








zinc/cadmium-
3.3
1.1
6461
1839.1
5318
1550.6


binding








protein CDS








acyl carrier
25.6
1.6
1230.5
957.6
1473.5
1174.7


protein CDS








ompX CDS
1.7
1.1
2042
734.2
1687.5
621.5


DNA-binding








protein HU-
6.9
−1.3
1305
881.7
725
501.8


beta CDS








sspA CDS
0.2
1
654
188.8
504.5
149.2


tatE CDS
1.4
1.3
131
118.4
125
115.8


LexA
0.1
−1.1
248
75.1
164
50.9


repressor CDS








hisS CDS
0
−1.1
467
69.2
325
49.3



















TABLE 23






Prm (In Forward





direction, −250 to





+10 region)
Expressed Sequence
Neighbor Sequence


Name
SEQ ID NO:
SEQ ID NO:
SEQ ID NO:







murein lipoprotein CDS
SEQ ID NO: 3
SEQ ID NO: 13
SEQ ID NO: 23


membrane protein CDS
SEQ ID NO: 4
SEQ ID NO: 14
SEQ ID NO: 24


zinc/cadmium-binding protein
SEQ ID NO: 5
SEQ ID NO: 15
SEQ ID NO: 25


CDS





acyl carrier protein CDS
SEQ ID NO: 6
SEQ ID NO: 16
SEQ ID NO: 26


ompX CDS
SEQ ID NO: 7
SEQ ID NO: 17
SEQ ID NO: 27


DNA-binding protein HU-beta
SEQ ID NO: 8
SEQ ID NO: 18
SEQ ID NO: 28


CDS





sspA CDS
SEQ ID NO: 9
SEQ ID NO: 19
SEQ ID NO: 29


tatE CDS
SEQ ID NO: 10
SEQ ID NO: 20
SEQ ID NO: 30


LexA repressor CDS
SEQ ID NO: 11
SEQ ID NO: 21
SEQ ID NO: 31


hisS CDS
SEQ ID NO: 12
SEQ ID NO: 22
SEQ ID NO: 32
















TABLE 24







Table of Strains














Mutagenic DNA

Gene 1
Gene 2


Name
Lineage
Description
Genotype
mutation
mutation





CI006
Isolated strain
None
WT





from Enterobacter







(now Kosakonia)







genera






CI008
Isolated strain
None
WT





from Burkholderia







genera






CI010
Isolated strain
None
WT





from Klebsiella







genera






CI019
Isolated strain
None
WT





from Rahnella







genera






CI028
Isolated strain
None
WT





from Enterobacter







genera






CI050
Isolated strain
None
WT





from Klebsiella







genera






CM002
Mutant of CI050
Disruption of nifL gene
ΔnifL::KanR
SEQ ID





with a kanamycin

NO: 33





resistance expression







cassette (Kara) encoding







the aminoglycoside O-







phosphotransferase gene







aph1 inserted.





CM011
Mutant of CI019
Disruption of niFL gene
ΔnifL::SpecR
SEQ ID





with a spectinomycin

NO: 34





resistance expression







cassette (SpecR) encoring







the streptomycin 3′-O-







adenylyltransferase gene







aadA inserted.





CM013
Mutant of CI006
Disruption of nifL gene
ΔnifL::KanR
SEQ ID





with a kanamycin

NO: 35





resistance expression







cassette (Kara) encoding







the aminoglycoside O-







phosphotransferase gene







aph1 inserted.





CM004
Mutant of CI010
Disruption of amtB gene
ΔamtB::KanR
SEQ ID





with a kanamycin

NO: 36





resistance expression







cassette (KanR) encoding







the aminoglycoside O-







phosphotransferase gene







aph1 inserted.





CM005
Mutant of CI010
Disruption of nifL gene
ΔnifL::KanR
SEQ ID





with a kanamycin

NO: 37





resistance expression







cassette (KanR) encoding







the aminoglycoside O-







phosphotransferase gene,







aph1 inserted.





CM015
Mutant of CI006
Disruption of nifL gene
ΔnifL::Prm5
SEQ ID





with a fragment of the

NO: 38





region upstream of the







ompX gene inserted







(Prm5).





CM021
Mutant of CI006
Disruption of nifL gene
ΔnifL::Prm2
SEQ ID





with a fragment of the

NO: 39





region upstream of an







unanotated gene and the







first 73 bp of that gene







inserted (Prm2).





CM023
Mutant of CI006
Disruption of nifL gene
ΔnifL::Prm4
SEQ ID





with a fragment of the

NO: 40





region upstream of the







acpP gene and the first







121 bp of the acpP gene







inserted (Prm4).





CM014
Mutant of CI006
Disruption of nifL gene
ΔnifL::Prm1
SEQ ID





with a fragment of the

NO: 41





region upstream of the lpp







gene and the first 29 bp of







the lpp gene inserted







(Prm1).





CM016
Mutant of CI006
Disruption of nifL gene
ΔnifL::Prm9
SEQ ID





with a fragment of the

NO: 42





region upstream of the







lexA 3 gene and the first







21 bp of the lexA 3 gene







inserted (Prm9).





CM022
Mutant of CI006
Disruption of nifL gene
ΔnifL::Prm3
SEQ ID





with a fragment of the

NO: 43





region upstream of the







mntP 1 gene and the first







53 bp of the mntP 1 gene







inserted (Prm3).





CM024
Mutant of CI006
Disruption of nifL gene
ΔnifL::Prm7
SEQ ID





with a fragment of the

NO: 44





region upstream of the







sspA gene inserted (Prm7).





CM025
Mutant of CI006
Disruption of nifL gene
ΔnifL::Prm10
SEQ ID





with a fragment of the

NO: 45





region upstream of the hisS







gene and the first 52 bp of







the hisS gene inserted







(Prm10).





CM006
Mutant of CI010
Disruption of glnB gene
ΔglnB::KanR
SEQ ID





with a kanamycin

NO: 46





resistance expression







cassette (KanR) encoding







the aminoglycoside O-







phosphotransferase gene







aph1 inserted.





CM017
Mutant of CI028
Disruption of nifL gene
ΔnifL::KanR
SEQ ID





with a kanamycin

NO: 47





resistance expression







cassette (KanR) encoding







the aminoglycoside O-







phosphotransferase gene







aph1 inserted.





CM011
Mutant of CI019
Disruption of nifL gene
ΔnifL::SpecR
SEQ ID





with a spectinomycin

NO: 48





resistance expression







cassette (SpecR) encoding







the streptomycin 3″-O







adenylyltransferase gene







aadA inserted.





CM013
Mutant of CI006
Disruption of nifL gene
ΔnifL::KanR
SEQ ID





with a kanamycin

NO: 49





resistance expression







cassette (KanR) encoding







the aminoglycoside O-







phosphotransferase gene







aph1 inserted.





CM005
Mutant of CI010
Disruption of nifL gene
ΔnifL::KanR
SEQ ID





with a kanamycin

NO: 50





resistance expression







cassette (KanR) encoding







the aminoglycoside O-







phosphotransferase gene







aph1 inserted.





CM014
Mutant of CI006
Disruption of nifL gene
ΔnifL::Prm1
SEQ ID





with a fragment of the

NO: 51





region upstream of the lpp







gene and the first 29 bp of







the lpp gene inserted







(Prm1).





CM015
Mutant of CI006
Disruption of nifL gene
ΔnifL::Prm5
SEQ ID





with a fragment of the

NO: 52





region upstream of the







ompX gene inserted







(Prm5).





CM023
Mutant of CI006
Disruption of nifL gene
ΔnifL::Prm4
SEQ ID





with a fragment of the

NO: 53





region upstream of the







acpP gene and the first







121 bp of the acpP gene







inserted (Prm4).





CM029
Mutant of CI006
Disruption of nifL gene
ΔnifL::Prm5
SEQ ID
SEQ ID




with a fragment of the
ΔglnE-
NO: 54
NO: 61




region upstream of the
AR_KO1






ompX gene inserted







(Prm5) and deletion of the







1287 bp after the start







codon of the glnE gene







containing the adenylyl-







removing domain of







glutamate-ammonia-ligase







adenylyltransferase







(ΔglnE-AR_KO1).





CM014
Mutant of CI006
Disruption of nifL gene
ΔnifL::Prm1
SEQ ID





with a fragment of the

NO: 55





region upstream of the lpp







gene and the first 29 bp of







the lpp gene inserted







(Prm1).





CM011
Mutant of C1019
Disruption of nifL gene
ΔnifL::SpecR
SEQ ID





with a spectinomycin

NO: 56





resistance expression







cassette (SpecR) encoding







the streptomycin 3′-O-







adenylyltransferase gene







aadA inserted.





CM011
Mutant of CI019
Disruption of nifL gene
ΔnifL::SpecR
SEQ ID





with a spectinomycin

NO: 57





resistance expression







cassette (SpecR) encoding







the streptomycin 3′-O -







adenylyltransferase gene







aadA inserted.





CM013
Mutant of CI006
Disruption of nifL gene
ΔnifL::KanR
SEQ ID





with a kanamycin

NO: 58





resistance expression







cassette (KanR) encoding







the aminogiycoside O-







phosphotransferase gene







aph1 inserted.





CM011
Mutant of CI019
Disruption of nifL gene
ΔnifL::SpecR
SEQ ID





with a spectinomycin

NO: 59





resistance expression







cassette (SpecR) encoding







the streptomycin 3″-O-







adenylyltransferase gene







aadA inserted.





CM011
Mutant of CI019
Disruption of nifL gene
ΔnifL::SpecR
SEQ ID





with a spectinomycin

NO: 60





resistance expression







cassette (SpecR) encoding







the streptomycin 3″-O-







adenyltransferase gene







aadA inserted.









EXAMPLES

The following examples are given for the purpose of illustrating various embodiments of the disclosure and are not meant to limit the present disclosure in any fashion. Changes therein and other uses which are encompassed within the spirit of the disclosure, as defined by the scope of the claims, will be recognized by those skilled in the art.


Example 1: Guided Microbial Remodeling—a Platform for the Rational Improvement of Microbial Species for Agriculture

An example overview of an embodiment of the Guided Microbial Remodeling (GMR) platform can be summarized in the schematic of FIG. 1A.



FIG. 1A illustrates that the composition of the microbiome can first be characterized and a species of interest is identified (e.g. to find a microbe with the appropriate colonization characteristics).


The metabolism of the species of interest can be mapped and linked to genetics. For example, the nitrogen fixation pathway of the microbe can be characterized. The pathway that is being characterized can be examined under a range of environmental conditions. For example, the microbe's ability to fix atmospheric nitrogen in the presence of various levels of exogenous nitrogen in its environment can be examined. The metabolism of nitrogen can involve the entrance of ammonia (NH4+) from the rhizosphere into the cytosol of the bacteria via the AmtB transporter. Ammonia and L-glutamate (L-Glu) are catalyzed by glutamine synthetase and ATP into glutamine. Glutamine can lead to the formation of bacterial biomass and it can also inhibit expression of the nif operon, i.e. it can be a competing force when one desires the microbe to fix atmospheric nitrogen and excrete ammonia. The nitrogen fixation pathway is characterized in great detail in earlier sections of the specification.


Afterwards, a targeted non-intergeneric genomic alteration can be introduced to the microbe's genome, using methods including, but not limited to: conjugation and recombination, chemical mutagenesis, adaptive evolution, and gene editing. The targeted non-intergeneric genomic alteration can include an insertion, disruption, deletion, alteration, perturbation, modification, etc. of the genome.


Derivative remodeled microbes, which comprise the desired phenotype resulting from the remodeled underlying genotype, are then used to inoculate crops.


The present disclosure provides, in certain embodiments, non-intergeneric remodeled microbes that are able to fix atmospheric nitrogen and supply such nitrogen to a plant. In aspects, these non-intergeneric remodeled microbes are able to fix atmospheric nitrogen, even in the presence of exogenous nitrogen.



FIG. 1B depicts an expanded view of the measurement of the microbiome step. In some embodiments, the present disclosure finds microbial species that have desired colonization characteristics, and then utilizes those species in the subsequent remodeling process.


The aforementioned Guided Microbial Remodeling (GMR) platform will now be described with more specificity.


In aspects, the GMR platform comprises the following steps:

    • A. Isolation—Obtain microbes from the soil, rhizosphere, surface, etc. of a crop plant of interest;
    • B. Characterization—Involves characterizing the isolated microbes for genotype/phenotypes of interest (e.g. genome sequence, colonization ability, nitrogen fixation activity, solubilization of P ability, excretion of a metabolite of interest, excretion of a plant promoting compound, etc.)
    • C. Domestication—Development of a molecular protocol for non-intergeneric genetic modification of the microbe;
    • D. Non-Intergeneric Engineering Campaign and Optimization—Generation of derivative non-intergeneric microbial strains with genetic modifications in key pathways (e.g. colonization associated genes, nitrogen fixation/assimilation genes, P solubilization genes);
    • E. Analytics—Evaluation of derived non-intergeneric strains for phenotypes of interest both in vitro (e.g. ARA assays) and in planta (e.g. colonization assays).
    • F. Iterate Engineering Campaign/Analytics—Iteration of steps D and E for further improvement of microbial strain.


Each of the GMR platform process steps will now be elaborated upon below.


A. Isolation of Microbes


1. Obtain a Soil Sample


Microbes will be isolated from soil and/or roots of a plant. In one example, plants will be grown in a laboratory or a greenhouse in small pots. Soil samples will be obtained from various agricultural areas. For example, soils with diverse texture characteristics can be collected, including loam (e.g. peaty clay loam, sandy loam), clay soil (e.g. heavy clay, silty clay), sandy soil, silty soil, peaty soil, chalky soil, and the like.


2. Grow Bait Plants


Seeds of a bait plant (a plant of interest) (e.g. corn, wheat, rice, Sorghum, millet, soybean, vegetables, fruits, etc.) will be planted into each soil type. In one example, different varieties of a bait plant will be planted in various soil types. For example, if the plant of interest is corn, seeds of different varieties of corn such as field corn, sweet corn, heritage corn, etc. will be planted in various soil types described above.


3. Harvest Soil and/or Root Samples and Plate on Appropriate Medium


Plants will be harvested by uprooting them after a few weeks (e.g. 2-4 weeks) of growth. Alternative to growing plants in a laboratory/greenhouse, soil and/or roots of the plant of interest can be collected directly from the fields with different soil types.


To isolate rhizosphere microbes and epiphytes, plants will be removed gently by saturating the soil with distilled water or gently loosening the soil by hand to avoid damage to the roots. If larger soil particles are present, these particles will be removed by submerging the roots in a still pool of distilled water and/or by gently shaking the roots. The root will be cut and a slurry of the soil sticking to the root will be prepared by placing the root in a plate or tube with small amount of distilled water and gently shaking the plate/tube on a shaker or centrifuging the tube at low speed. This slurry will be processed as described below.


To isolate endophytes, excess soil on root surfaces will be removed with deionized water. Following soil removal, plants will be surface sterilized and rinsed vigorously in sterile water. A cleaned, 1 cm section of root will be excised from the plant and placed in a phosphate buffered saline solution containing 3 mm steel beads. A slurry will be generated by vigorous shaking of the solution with a Qiagen TissueLyser II.


The soil and/or root slurry can be processed in various ways depending on the desired plant-beneficial trait of microbes to be isolated. For example, the soil and root slurry can be diluted and inoculated onto various types of screening media to isolate rhizospheric, endophytic, epiphytic, and other plant-associated microbes. For example, if the desired plant-beneficial trait is nitrogen fixation, then the soil/root slurry will be plated on a nitrogen free media (e.g. Nfb agar media) to isolate nitrogen fixing microbes. Similarly, to isolate phosphate solubilizing bacteria (PSB), media containing calcium phosphate as the sole source of phosphorus can be used. PSB can solubilize calcium phosphate and assimilate and release phosphorus in higher amounts. This reaction is manifested as a halo or a clear zone on the plate and can be used as an initial step for isolating PSB.


4. Pick Colonies, Purify Cultures, and Screen for the Presence of Genes of Interest


Populations of microbes obtained in step A3 are streaked to obtain single colonies (pure cultures). A part of the pure culture is resuspended in a suitable medium (e.g. a mixture of R2A and glycerol) and subjected to PCR analysis to screen for the presence of one or more genes of interest. For example, to identify nitrogen fixing bacteria (diazotrophs), purified cultures of isolated microbes can be subjected to a PCR analysis to detect the presence of nif genes that encode enzymes involved in the fixation of atmospheric nitrogen into a form of nitrogen available to living organisms.


5. Bank a Purified Culture


Purified cultures of isolated strains will be stored, for example at −80° C., for future reference and analysis.


B. Characterization of Isolated Microbes


1. Phylogenetic Characterization and Whole Genome Sequencing


Isolated microbes will be analyzed for phylogenetic characterization (assignment of genus and species) and the whole genome of the microbes will be sequenced.


For phylogenetic characterization, 16S rDNA of the isolated microbe will be sequenced using degenerate 16S rDNA primers to generate phylogenetic identity. The 16S rDNA sequence reads will be mapped to a database to initially assign the genus, species and strain name for isolated microbes. Whole genome sequencing is used as the final step to assign phylogenetic genus/species to the microbes.


The whole genome of the isolated microbes will be sequenced to identify key pathways. For the whole genome sequencing, the genomic DNA will be isolated using a genomic DNA isolation kit (e.g. QIAmp DNA mini kit from QIAGEN) and a total DNA library will be prepared using the methods known in the art. The whole genome will be sequenced using high throughput sequencing (also called Next Generation Sequencing) methods known in the art. For example, Illumina, Inc., Roche, and Pacific Biosciences provide whole genome sequencing tools that can be used to prepare total DNA libraries and perform whole genome sequencing.


The whole genome sequence for each isolated strain will be assembled; genes of interest will be identified; annotated; and noted as potential targets for remodeling. The whole genome sequences will be stored in a database.


2. Assay the Microbe for Colonization of a Host Plant in a Greenhouse


Isolated microbes will be characterized for the colonization of host plants in a greenhouse. For this, seeds of the desired host plant (e.g., corn, wheat, rice, Sorghum, soybean) will be inoculated with cultures of isolated microbes individually or in combination and planted into soil. Alternatively, cultures of isolated microbes, individually or in combination, can be applied to the roots of the host plant by inoculating the soil directly over the roots. The colonization potential of the microbes will be assayed, for example, using a quantitative PCR (qPCR) method described in a greater detail below.


3. Assay the Microbe for Colonization of the Host Plant in Small-Scale Field Trials and Isolate RNA from Colonized Root Samples (CAT Trials)


Isolated microbes will be assessed for colonization of the desired host plant in small-scale field trials. Additionally, RNA will be isolated from colonized root samples to obtain transcriptome data for the strain in a field environment. These small-scale field trials are referred to herein as CAT (Colonization and Transcript) trials, as these trials provide Colonization and Transcript data for the strain in a field environment.


For these trials, seeds of the host plant (e.g., corn, wheat, rice, Sorghum, soybean) will be inoculated using cultures of isolated microbes individually or in combination and planted into soil. Alternatively, cultures of isolated microbes, individually or in combination, can be applied to the roots of the host plant by inoculating the soil directly over the roots. The CAT trials can be conducted in a variety of soils and/or under various temperature and/or moisture conditions to assess the colonization potential and obtain transcriptome profile of the microbe in various soil types and environmental conditions.


Colonization of roots of the host plant by the inoculated microbe(s) will be assessed, for example, using a qPCR method as described below.


In one protocol, the colonization potential of isolated microbes was assessed as follows. One day after planting of corn seeds, 1 ml of microbial overnight culture (SOB media) was drenched right at the spot of where the seed was located. 1 mL of this overnight culture was roughly equivalent to about 10{circumflex over ( )}9 cfu, varying within 3-fold of each other, depending on which strain is being used. Each seedling was fertilized 3× weekly with 50 mL modified Hoagland's solution supplemented with either 2.5 mM or 0.25 mM ammonium nitrate. At four weeks after planting, root samples were collected for DNA extraction. Soil debris were washed away using pressurized water spray. These tissue samples were then homogenized using QIAGEN Tissuelyzer and the DNA was then extracted using QIAmp DNA Mini Kit (QIAGEN) according to the recommended protocol. qPCR assay was performed using Stratagene Mx3005P RT-PCR on these DNA extracts using primers that were designed (using NCBI's Primer BLAST) to be specific to a loci in each of the microbe's genome.


The presence of the genome copies of the microbe was quantified, which reflected the colonization potential of the microbe. Identity of the microbial species was confirmed by sequencing the PCR amplification products.


Additionally, RNA will be isolated from colonized root and/or soil samples and sequenced.


Unlike the DNA profile, an RNA profile varies depending on the environmental conditions. Therefore, sequencing of RNA isolated from colonized roots and/or soil will reflect the transcriptional activity of genes in planta in the rhizosphere.


RNA can be isolated from colonized root and/or soil samples at different time points to analyze the changes in the RNA profile of the colonized microbe at these time points.


For example, RNA can be isolated from colonized root and/or soil samples right after fertilization of the field and a few weeks after fertilization of the field and sequenced to generate corresponding transcriptional profile.


Similarly, RNA sequencing can be carried out under high phosphate and low phosphate conditions to understand which genes are transcriptionally active or repressed under these conditions.


Methods for transcriptomic/RNA sequencing are known in the art. Briefly, total RNA will be isolated from the purified culture of the isolated microbe; cDNA will be prepared using reverse transcriptase; and the cDNA will be sequenced using high throughput sequencing tools described above.


Sequencing reads from the transcriptome analysis can be mapped to the genomic sequence and transcriptional promoters for the genes of interest can be identified.


4. Assay the Plant-Beneficial Activity of Isolated Microbes


The plant-beneficial activity of isolated microbes will be assessed.


For example, nitrogen fixing microbes will be assayed for nitrogen fixation activity using an acetylene reduction assay (ARA) or phosphate solubilizing microbes will be assayed for phosphate solubilization. Any parameter of interest can be utilized and an appropriate assay developed for such. For instance, assays could include growth curves for colonization metrics and assays for production of phytohormones like indole acetic acid (IAA) or gibberellins. An assay for any plant-beneficial activity that is of interest can be developed.


This step will confirm the phenotype of interest and eliminate any false positives.


5. Selection of Potential Candidates from Isolated Microbes


The data generated in the above steps will be used to select microbes for further development. For example, microbes showing a desired combination of colonization potential, plant-beneficial activity, and/or relevant DNA and RNA profile will be selected for domestication and remodeling.


C. Domestication of Selected Microbes


The selected microbes will be domesticated; wherein, the microbes will be converted to a form that is genetically tractable and identifiable.


1. Test for Antibiotic Sensitivity


One way to domesticate the microbes is to engineer them with antibiotic resistance. For this, the wild type microbial strain will be tested for sensitivity to various antibiotics. If the strain is sensitive to the antibiotic, then the antibiotic can be a good candidate for use in genetic tools/vectors for remodeling the strain.


2. Design and Build a Vector


Vectors that are conditional for their replication (e.g. a suicide plasmid) will be constructed to domesticate the selected microbes (host microbes). For example, a suicide plasmid containing an appropriate antibiotic resistance marker, a counter selectable marker, an origin of replication for maintenance in a donor microbe (e.g. E. coli), a gene encoding a fluorescent protein (GFP, RFP, YFP, CFP, and the like) to screen for insertion through fluorescence, an origin of transfer for conjugation into the host microbe, and a polynucleotide sequence comprising homology arms to the host genome with a desired genetic variation will be constructed. The vector may comprise a SceI site and other additional elements.


Exemplary antibiotic resistance markers include ampicillin resistance marker, kanamycin resistance marker, tetracycline resistance marker, chloramphenicol resistance marker, erythromycin resistance marker, streptomycin resistance marker, spectinomycin resistance marker, etc. Exemplary counter selectable markers include sacB, rpsL, tetAR, pheS, thyA, lacY, gata-1, ccdB, etc.


3. Generation of Donor Microbes


In one protocol, a suicide plasmid containing an appropriate antibiotic resistance marker, a counter selectable marker, the λpir origin of replication for maintenance in E. coli ST18 containing the pir replication initiator gene, a gene encoding green fluorescent protein (GFP) to screen for insertion through fluorescence, an origin of transfer for conjugation into the host microbe, and a polynucleotide sequence comprising homology arms to the host genome with a desired genetic variation (e.g. a promoter from within the microbe's own genome for insertion into a heterologous location) will be transformed into E. coli ST18 (an auxotroph for aminolevulinic acid, ALA) to generate donor microbes.


4. Mix Donor Microbes with Host Microbes


Donor microbes will be mixed with host microbes (selected candidate microbes from step B5) to allow conjugative integration of the plasmid into the host genome. The mixture of donor and host microbes will be plated on a medium containing the antibiotic and not containing ALA. The suicide plasmid is able to replicate in donor microbes (E. coli ST18), but not in the host. Therefore, when the mixture containing donor and host microbes is plated on a medium containing the antibiotic and not containing ALA, only host cells that integrated the plasmid into its genome will be able to grow and form colonies on the medium. The donor microbes will not grow due to the absence of ALA.


5. Confirm Integration of the Vector


A proper integration of the suicide plasmid containing the fluorescent protein marker, the antibiotic resistance marker, the counter selectable marker, etc. at the intended locus of the host microbe will be confirmed through fluorescence of colonies on the plate and using colony PCR.


6. Streak Confirm Integration Colony


A second round of homologous recombination in the host microbes will loop out (remove) the plasmid backbone leaving the desired genetic variation (e.g. a promoter from within the microbe's own genome for insertion into a heterologous location) integrated into the host genome of a certain percentage of host microbes, while reverting a certain percentage back to wild type.


Colonies of host microbes that have looped out the plasmid backbone (and therefore, looped out the counter selectable marker) can be selected by growing them on an appropriate medium.


For example, if sacB is used as a counter selectable marker, loss of this marker due to the loss of the plasmid backbone will be tested by growing the colonies on a medium containing sucrose (sacB confers sensitivity to sucrose). Colonies that grow on this medium would have lost the sacB marker and the plasmid backbone and would either contain the desired genetic variation or be reverted to wild type. Also, these colonies will not fluoresce on the plate due to the loss of the fluorescent protein marker.


In some isolates, the sacB or other counterselectable markers do not confer full sensitivity to sucrose or other counterselection mechanisms, which necessitates screening large numbers of colonies to isolate a successful loop-out. In those cases, loop-out may be aided by use of a “helper plasmid” that replicates independently in the host cell and expresses a restriction endonuclease, e.g. SceI, which recognizes a site in the integrated suicide plasmid backbone. The strain with the integrated suicide plasmid is transformed with the helper plasmid containing an antibiotic resistance marker, an origin of replication compatible with the host strain, and a gene encoding a restriction endonuclease controlled by a constitutive or inducible promoter. The double-strand break induced in the integrated plasmid backbone by the restriction endonuclease promotes homologous recombination to loop-out the suicide plasmid. This increases the number of looped-out colonies on the counterselection plate and decreases the number of colonies that need to be screened to find a colony containing the desired mutation. The helper plasmid is then removed from the strain by culture and serial passaging in the absence of antibiotic selection for the plasmid. The passaged cultures are streaked for single colonies, colonies are picked and screened for sensitivity to the antibiotic used for selection of the helper plasmid, as well as absence of the plasmid confirmed by colony PCR. Finally, the genome is sequenced and the absence of helper plasmid DNA is confirmed as described in D6.


7. Confirm Integration of the Genetic Variation Through Colony PCR


The colonies that grew better on the sucrose-containing medium (or other appropriate media depending on the counter selectable marked used) will be picked and the presence of the genetic variation at the intended locus will be confirmed by screening the colonies using colony PCR.


Although this example describes one protocol for domesticating the microbe and introducing genetic variation into the microbe, one of ordinary skill in the art would understand that the genetic variation can be introduced into the selected microbes using a variety of other techniques known in the art such as: polymerase chain reaction mutagenesis, oligonucleotide-directed mutagenesis, saturation mutagenesis, fragment shuffling mutagenesis, homologous recombination, ZFN, TALENS, CRISPR systems (Cas9, Cpf1, etc.), chemical mutagenesis, and combinations thereof.


8. Iterate Upon Steps C2-C7


If any of the steps C2-C7 fail to provide the intended outcome, the steps will be repeated to design an alternative vector that may comprise different elements for facilitating incorporation of desired genetic variations and markers into the host microbe.


9. Develop a Standard Operating Procedure (SOP)


Once the steps C2-C7 can be reproduced consistently for a given strain, the steps will be used to develop a standard operating procedure (SOP) for that strain and vector. This SOP can be used to improve other plant-beneficial traits of the microbe.


D. Non-Intergeneric Engineering Campaign and Optimization


1. Identify Gene Targets for Optimization


Selected microbes will be engineered/remodeled to improve performance of the plant-beneficial activity. For this, gene targets for improving the plant-beneficial activity will be identified.


Gene targets can be identified in various ways. For example, genes of interest can be identified while annotating the genes from the whole genome sequencing of isolated microbes. They can be identified through a literature search. For example, genes involved in nitrogen fixation are known in the literature. These known genes can be used as targets for introducing genetic variations. Gene targets can also be identified based on the RNA sequencing data obtained in the step B3 (small-scale field trials for colonization) or by performing RNA sequencing described in the step below.


2. Select Promoters for Promoter Swaps


A desired genetic variation for improving the plant-beneficial activity can comprise promoter swapping, in which the native promoter for a target gene is replaced with a stronger or weaker promoter (when compared to the native promoter) from within the microbe's genome, or differently regulated promoter (e.g. a N-independent). If the expression of a target gene increases the plant-beneficial activity (e.g., nifA, the expression of which enhances nitrogen fixation in microbes), the desired promoter for promoter swapping is a stronger promoter (compared to the native promoter of the target gene) that would further increase the expression level of the target gene compared to the native promoter. If the expression of a target gene decreases the plant-beneficial activity (e.g., nifL that downregulates nitrogen fixation), the desired promoter for promoter swapping is a weak promoter (compared to the native promoter of the target gene) that would substantially decrease the expression level of the target gene compared to the native promoter. Promoters can be inserted into genes to “knock-out” a gene's expression, while at the same time upregulating the expression of a downstream gene.


Promoters for promoter swapping can be selected based on the RNA sequencing data. For example, the RNA sequencing data can be used to identify strong and weak promoters, or constitutively active vs. inducible promoters.


For example, to identify strong and weak promoters, or constitutively active vs. inducible promoters, in the nitrogen fixation pathway, selected microbes will be cultured in vitro under nitrogen-depleted and nitrogen-replete conditions; RNA of the microbe will be isolated from these cultures; and sequenced.


In one protocol, the RNA profile of the microbe under nitrogen-depleted and nitrogen-replete conditions will be compared and active promoters with a desired transcription level will be identified. These promoters can be selected to swap a weak promoter.


Promoters can also be selected using the RNA sequencing data obtained in the step B3 that reflects the RNA profile of the microbe in planta in the host plant rhizosphere.


RNA sequencing under various conditions allows for selection of promoters that: a) are active in the rhizosphere during the host plant growth cycle in fertilized field conditions, and b) are also active in relevant in vitro conditions so they can be rapidly screened.


In an exemplary protocol, in planta RNA sequencing data from colonization assays (e.g. step B3) is used to measure the expression levels of genes in isolated microbes. In one embodiment, the level of gene expression is calculated as reads per kilobase per million mapped reads (RPKM). The expression level of various genes is compared to the expression level of a target gene and at least the top 10, 20, 30, 40, 50, 60, or 70 promoters, associated with the various genes, that show the highest or lowest level of expression compared to the target gene are selected as possible candidates for promoter swapping. Thus, one looks at expression levels of various genes relative to a target gene and then selects genes that demonstrate increased expression relative to a target (or standard) gene and then find the promoters associated with said genes.


For example, if the target gene is upregulation of nifA, the first 10, 20, 30, 40, 50, or 60 promoters for genes that show the highest level of expression compared to nifA are selected as possible candidates for promoter swapping.


These candidates can be further short-listed based on in vitro RNA sequencing data. For example, for nifA as the target gene, possible promoter candidates selected based on the in planta RNA sequencing data are further selected by choosing promoters with similar or increased gene expression levels compared to nifA under in vitro nitrogen-deplete vs. nitrogen-replete conditions.


The set of promoters selected in this step are used to swap the native promoter of the target gene (e.g. nifA). Remodeled strains with swapped promoters are tested in in vitro assays; strains with lower than expected activity are eliminated; and strains with expected or higher than expected activity are tested in field. The cycle of promoter selection may be repeated on remodeled strains to further improve their plant-beneficial activity.


Described here is an exemplary promoter swap experiment that was carried out based on in planta and in vitro RNA sequencing data from Klebsiella variicola strain, CI137 to improve the nitrogen fixation trait. CI137 was analyzed in ARA assays at 0 mM and 5 mM glutamine concentration and RNA was extracted from these ARA samples. The RNA was sequenced via NextSeq and a subset of reads from one sample was mapped to the CI137 genome (in vitro RNA sequencing data). RNA was extracted from the roots of corn plants at V5 stage in the colonization and activity assay (e.g. step B3) for CI137. Samples from six plants were pooled; the RNA from the pooled sample was sequenced using NextSeq, and reads were mapped to the CI137 genome (in planta RNA sequencing data). Out of 2×108 total reads, 7×104 reads mapped to CI137. In planta RNA sequencing data was used to rank genes in order of in planta expression levels and the expression levels were compared to the native nifA expression level. The first 40 promoters that showed the highest expression level (based on gene expression) compared to the native nifA expression level were selected. These 40 promoters were further short-listed based on the in vitro RNA sequencing data, where promoters with increased or similar in vitro expression levels compared to nifA were selected. The final list of promoters included 17 promoters and two versions of most promoters were used to generate promoter swap mutants; thus a total of 30 promoters were tested. Generation of a suite of CI137 mutants where nifL was deleted partially or completely and the 30 promoters inserted (ΔnifL::Prm) was attempted. 28 out of 30 mutants were generated successfully. The ΔnifL::Prm mutants were analyzed in ARA assays at 0 mM and 5 mM glutamine concentration and RNA was extracted from these ARA samples. Several mutants showed lower than expected or decreased ARA activity compared to the WT CI137 strain. A few mutants showed higher than expected ARA activity.


A person of ordinary skill in the art would appreciate from the above example that while in planta and/or in vitro RNA sequencing data can be used to select promoters for promoter swapping, the step of promoter selection is highly unpredictable and involves many challenges.


For example, in planta RNA sequencing mainly reveals the genes that are highly expressed; however, it is difficult to detect fine differences in gene expression and/or genes with low expression levels. For instance, in some in planta RNA sequencing experiments, only about 40 out of about 5000 genes from a microbial genome were detected. Thus, in planta RNA sequencing technique is useful to identify abundantly expressed genes and their corresponding promoters; however, the technique has difficulty in identifying low expression genes and corresponding promoters and small differences between gene expression.


Furthermore, in planta RNA profile reflects the status of the genes at the time the microbes were isolated; however, a slight change in the field conditions can substantially change the RNA profile of rhizosphere/epiphytic/endophytic microbes. Therefore, it is difficult to predict in advance whether the promoters selected based on one field trial RNA sequencing data would provide desirable expression levels of the target gene when remodeled strains are tested in vitro and in field.


Additionally, in planta evaluation is time and resource-consuming; therefore, in planta experiments cannot be conducted often and/or repeated quickly or easily. On the other hand, while in vitro RNA sequencing can be conducted relatively quickly and easily, the in vitro conditions do not mimic the field conditions and promoters that may show high activity in vitro may not show comparable activity in planta.


Moreover, promoters often don't behave as predicted in a new context. Therefore, in planta and in vitro RNA sequencing data can at best serve as a starting point in the step of promoter selection; however, arriving at any particular promoter that would provide desirable expression levels of the target gene in the field is, in some instances, unpredictable.


Another limitation in the step of promoter selection is the number of available promoters. Because one of the goals of the present invention is to provide non-transgenic microbes; promoters for promoter swapping need to be selected from within the microbe's genome, or genus. Thus, unlike a transgenic approach, the present process can not merely go out into the literature and find/use a well characterized transgenic promoter from a different host organism.


Another constraint is that the promoter must be active in planta during a desired growth phase. For example, the highest requirement for nitrogen in plants is generally late in the growing season, e.g. late vegetative and early reproductive phases. For example, in corn, nitrogen uptake is the highest during V6 (six leaves) through R1 (reproductive stage 1) stages. Therefore, to increase the availability of nitrogen during V6 through R1 stages of corn, remodeled microbes must show highest nitrogen fixation activity during these stages of the corn lifecycle. Accordingly, promoters that are active in planta during the late vegetative and early reproductive stages of corn need to be selected. This constraint not only reduces the number of promoters that may be tested in promoter swapping, but also make the step of promoter selection unpredictable. As discussed above, unpredictability arises, in part, because although the RNA sequencing data from small scale field trials (e.g. step B3) may be used to identify promoters that are active in planta during a desired growth stage, the RNA data is based on the field conditions (e.g., type of soil, level of water in the soil, level of available nitrogen, etc.) at the time of sample collection. As one of ordinary skill in the art would understand, the field conditions may change over the period of time within the same field and also change substantially across various fields. Thus, the promoters selected under one field condition may not behave as expected under other field conditions. Similarly, selected promoters may not behave as expected after swapping. Therefore, it is difficult to anticipate in advance whether the selected promoters would be active in planta during a desired growth phase of a plant of interest.


3. Design Non-Intergeneric Genetic Variations


Based on steps D1 (identification of gene targets) and D2 (identification of promoters for promoter swaps), non-intergeneric genetic variations will be designed.


The term “non-intergeneric” indicates that the genetic variation to be introduced into the host does not contain a nucleic acid sequence from outside the host genus (i.e., no transgenic DNA). Although vectors and/or other genetic tools will be used to introduce the genetic variation into the host microbe, the methods of the present disclosure include steps to loop-out (remove) the backbone vector sequences or other genetic tools introduced into the host microbe leaving only the desired genetic variation into the host genome. Thus, the resulting microbe is non-transgenic.


Exemplary non-intergeneric genetic variations include a mutation in the gene of interest that may improve the function of the protein encoded by the gene; a constitutionally active promoter that can replace the endogenous promoter of the gene of interest to increase the expression of the gene; a mutation that will inactivate the gene of interest; the insertion of a promoter from within the host's genome into a heterologous location, e.g. insertion of the promoter into a gene that results in inactivation of said gene and upregulation of a downstream gene; and the like. The mutations can be point mutations, insertions, and/or deletions (full or partial deletion of the gene). For example, in one protocol, to improve the nitrogen fixation activity of the host microbe, a desired genetic variation may comprise an inactivating mutation of the nifL gene (negative regulator of nitrogen fixation pathway) and/or comprise replacing the endogenous promoter of the nifH gene (nitrogenase iron protein that catalyzes a key reaction to fix atmospheric nitrogen) with a constitutionally active promoter that will drive the expression of the nifH gene constitutively.


4. Generate Non-Intergeneric Derivative Strains


After designing the non-intergeneric genetic variations, steps C2-C7 will be carried out to generate non-intergeneric derivative strains (i.e. remodeled microbes).


5. Bank a Purified Culture of the Remodeled Microbe


A purified culture of the remodeled microbe will be preserved in a bank, so that gDNA can be extracted for whole genome sequencing described below.


6. Confirm Presence of the Desired Genetic Variation


The genomic DNA of the remodeled microbe will be extracted and the whole genome sequencing will be performed on the genomic DNA using methods described previously. The resulting reads will be mapped to the reads previously stored in LIMS to confirm: a) presence of the desired genetic variation, and b) complete absence of reads mapping to vector sequences (e.g. plasmid backbone or helper plasmid sequence) that were used to generate the remodeled microbe.


This step allows sensitive detection of non-host genus DNA (transgenic DNA) that may remain in the strain after looping out of the vector backbone (e.g. suicide plasmid) method and could provide a control for accidental off-target insertion of the genetic variation, etc.


E. Analytics Upon Remodeled Microbes


1. Analysis of the Plant-Beneficial Activity


The plant-beneficial activity and growth kinetics of the remodeled microbes will be assessed in vitro.


For example, strains remodeled for improving nitrogen fixation function will be assessed for nitrogen fixation activity and fitness through acetylene reduction assays, ammonium excretion assays, etc.


Strains remodeled for improved phosphate solubilization will be assessed for the phosphate solubilization activity.


This step allows rapid, medium to high throughput screening of remodeled strains for the phenotypes of interest.


2. Analysis of Colonization and Transcription of the Altered Genes


Remodeled strains will be assessed for colonization of the host plant either in the greenhouse or in the field using the steps described in B3. Additionally, RNA will be isolated from colonized root and/or soil samples and sequenced to analyze the transcriptional activity of target genes. Target genes comprise the genes containing the genetic variation introduced and may also comprise other genes that play a role in the plant-beneficial trait of the microbe.


For example, a cluster of genes, the nif genes, controls the nitrogen fixation activity of microbes. Using the protocol described above, a genetic variation may be introduced into one of the nif genes (e.g. a promoter insertion), whereas the other genes in the nif cluster are in their endogenous form (i.e. their gene sequence and/or the promoter region is not altered). The RNA sequencing data will be analyzed for the transcriptional activity of the nif gene containing the genetic variation and may also be analyzed for other nif genes that are not altered directly, by the inserted genetic change, but nonetheless may be influenced by the introduced genetic change.


This step allows determination of the fitness of top in vitro performing strains in the rhizosphere and allows measurement of the transcriptional activity of altered genes in planta.


F. Iterate Engineering Campaign/Analytics


The data from in vitro and in planta analytics (steps E1 and E2) will be used to iteratively stack beneficial mutations.


Furthermore, steps A-E described above may be repeated to finetune the plant-beneficial traits of the microbes. For example, plants will be inoculated using microbial strains remodeled in the first round; harvested after a few weeks of growth; and microbes from the soil and/or roots of the plants will be isolated. The functional activity (plant-beneficial trait and colonization potential) and the DNA and RNA profile of isolated microbes will be characterized, in order to select microbes with improved plant-beneficial activity and colonization potential. The selected microbes will be remodeled to further improve the plant-beneficial activity. Remodeled microbes will be screened for the functional activity (plant-beneficial trait and colonization potential) and RNA profile in vitro and in planta and the top performing strains will be selected. If desired, steps A-E can be repeated to further improve the plant-beneficial activity of the remodeled microbes from the second round. The process can be repeated for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more rounds.


The exemplary steps described above are summarized in Table A below.









TABLE A







An Overview of an Embodiment of the Guided Microbial Remodeling Platform











Steps
Contribution
Alternate Forms





A
Isolation




1
Obtain a soil sample
Provides WT soil





microbes to be isolated



2
Grow corn “bait
Allows selection of plant-
Wheat, sorghum, rice, millet,



plants” in soil sample
beneficial microbes by
soybean, etc.




rhizosphere



3
Harvest, clean and
Down-select soil microbes
Other nitrogen-free media, other



extract root sample
to those that a) colonize
selective or screening media



and plate on nitrogen-
the root and b) fix
(e.g . for phosphate



free (specifically
atmospheric nitrogen
solubilization)



NfB) media




4
Pick colonies, purify
Down-select microbes to
Degenerate primers for other



cultures and screen
those containing the nifH
genes of interest, e.g. ipdC



for presence of nifH
gene (eliminate false-
(phytohormone biosynthesis)



using degenerate
positives from media




primers
screen)



5
Bank a purified





culture of the strain




B
Characterization




1
Sequence and
Characterize genome for




assemble the genome
key pathways




of the strain using





Illumina and/or





PacBio platform




2
Assay the microbe for
Down-select for microbes
Wheat, sorghum, rice, millet,



colonization of corn
that colonize the plant well
soybean, etc., other methods for



roots in the

assaying colonization (e.g.



greenhouse (qPCR-

plating)



based method)




3
Assay the microbe for
Known internally as




colonization of corn
“CAT” trials, these
Larger field trials, other crops,



roots in a small-scale
provide Colonization And
other methods for assaying



field trials (qPCR-
Transcript data for the
colonization (e.g. plating)



based method) and
strain in a field




isolate RNA from
environment




colonized root





samples




4
Assay the microbe for
Confirm N-fixation




nitrogen fixation
phenotype of strain




activity in an





acetylene reduction





assay (ARA)




5
Use the above data to
Allows selection of




select candidate
greatest-potential




microbe for further
candidates




domestication and





optimization




C
Domestication




1
Test microbes for
Determine which




sensitivity to various
antibiotic selection




antibiotics
markers can be used to





transform genetic tools



2
Design and build a
These are the “parts”
Plasmid could contain a SceI site



suicide plasmid
necessary to maintain the
or other counter-selectable



containing an
plasmid and carry out
marker, alternate fluorescent



appropriate antibiotic
conjugation, insertion and
reporters, additional elements



resistance marker,
“loop-out” of the hose




sacB counter-
genome




selectable marker,





origin of replication





for maintenance in E.






coli, GFP to screen






for insertion through





fluorescence, origin





of transfer for





conjugation into the





host, homology arms





to the host genome,





and the desired





mutation.




3
Transform suicide
Preparation for
Could use a different donor



plasmid into E. coli
conjugation into host;
strain of E. coli or other



ST18 (an auxotroph
plasmid maintenance
microbe; different auxotrophic



for aminolevulinic

marker



acid, ALA) to





generate donor cells




4
Mix donor cells with
The suicide plasmid is able
Could use a different donor



recipient host cells to
to replicate in E. coli but
strain of E. coli or other



conjugate, and plate
not in the host. Therefore
microbe; different auxotrophic



on media selecting for
plating of the mixture on
marker



the antibiotic
such plates means that




resistance marker and
only host cells that




NOT containing ALA
received the plasmid and





experience plasmid





integration into the





chromosome will be able





to grow and form colonies.





The Ecoli ST18 is unable





to grow due to the absence





of ALA.



5
Confirm integration
Confirms proper




of the plasmid
integration of the suicide




through GFP
plasmid backbone




fluorescence, and
containing GFP, the




integration at the
antibiotic resistance




intended locus
cassette, the sacB marker,




through colony PCR
etc.



6
Streak confirmed
The sacB marker confers
Different counter selectable



integration colony on
sensitivity to sucrose;
marker, SceI-mediated loop-out,



a plate containing
colonies which have
etc.



sucrose and screen for
undergone a second round




non-fluorescent
of homologous




colonies
recombination and





“looped-out” the plasmid





will grow better and not





fluoresce on the plate.



7
Screen looped-out
Upon the second




colonies for the
homologous




intended mutation
recombination event only




using colony PCR
50% of looped out





colonies should contain





the mutation, the other





50% will be WT



8
If any of the steps 2-7
Allows iterative




fail, go back to step 2
troubleshooting of suicide




and re-design with
plasmid to develop a




alternate plasmid
working protocol




parts




9
Once steps 2-7 can be





reliably performed,





develop an SOP for





that strain/plasmid to





be used for





Optimization




D
Non-Intergeneric





Engineering





Campaign and





Optimization




1
Identify gene targets





for optimizing a





pathway, e.g. nif





genes through





literature search




2
Select promoters for
Allows for selection of
Alternate crops; alternate



promoter swaps using
promoters that a) are
RNAseq data conditions



RNAseq data
active in the rhizosphere
(greenhouse, field, in vitro,



collected both in vitro
during the corn growth
whatever's relevant for the



in N-depleted and N-
cycle in fertilized field
phenotype targeted)



replete conditions,
conditions b) are also




and in planta from
active in in vitro N-replete




the corn rhizosphere
conditions so they can be




(Collected in step B3)
rapidly screened.



3
Design non-
No DNA from outside the
Alter regulatory sequences e.g.



intergeneric
host chromosome is added,
RBS), non-coding: RNAs, etc.



mutations in key
therefore the resulting




genes: deletions (full
microbe is non-transgenic




or partial gene),





promoter swaps, or





single base pair





changes; store these





designs in our LIMS




4
Using the established
We perform this in higher




protocol, carry out
throughput than the




steps C2-7 to generate
domestication step - up to




non-intergeneric
20 or so strains at once per




derivative strains
person.




(mutants)




5
Bank a purified





culture of the strain,





extract gDNA and





conduct WGS via





Illumina




6
Map the resulting
Allows very sensitive
Suicide plasmid removal is fairly



reads to the designs
detection of non-
reliable; however use of other



stored in LIMS to
intergeneric DNA that
stable plasmids in alternate



confirm a) presence
may remain in the strain
methods necessitates this extra



of the desire mutation
after the suicide plasmid
step to ensure with complete



and b) complete
method; confirm absence
confidence that no transgenic



absence of reads
of transgenic DNA,
DNA that was previously



mapping to any
controls for accidental off-
transformed in remains in the



suicide plasmid or
target insertion of the
strain.



other plasmid
suicide plasmid, etc.




sequences used to





generate the strains




E
Analytics




1
Analyze the strains
Allow rapid, med- to high-
Any other in vitro assay, e.g.



for in vitro nitrogen
throughput screening of
phosphate solubilization, qPCR



fixation activity and
mutants for phenotypes of
for transcription of specific



fitness through ARA,
interest
genes, etc.



ammonium excretion





assays, and growth





curves




2
Analyze the strains
Measure fitness of top in




for colonization
vitro performing strains in




(gPCR) and
the rhizosphere; measure




transcription of target
transcription of promoter-




and promoter-
swapped genes in planta




swapped genes





(Nanostring) in the





plant (greenhouse or





field)




F
Iterate Engineering





Campaign/Analytics




1
Use data from in vitro





and in planta





analytics to iteratively





stack beneficial.





mutations.










Traditional Approaches to Creating Biologicals for Agriculture Suffer from Drawbacks Inherent in their Methodology


Unlike pure bioprospecting of wild-type (WI) microbes or transgenic approaches, GMR allows for non-intergeneric genetic optimization of key regulatory networks within the microbe, which improves plant-beneficial phenotypes over WI microbes, but doesn't have the risks associated with transgenic approaches (e.g. unpredictable gene function, public and regulatory concerns). See, FIG. 1C for a depiction of a problematic “traditional bioprospecting” approach, which has several drawbacks compared to the taught GMR platform.


Other methods for developing microbials for agriculture are focused on either extensive lab development, which often fails at the field scale, or extensive greenhouse or “field-first” testing without an understanding of the underlying mechanisms/plant-microbe interactions. See, FIG. 1D for a depiction of a problematic “field-first approach to bioprospecting” system, which has several drawbacks compared to the taught GMR platform.


The GMR Platform Solves these Problems in Numerous Ways


One strength of the GMR platform is the identification of active promoters, which are active at key physiologically important times for a target crop, and which are also active under particular, agriculturally relevant, environmental conditions.


As has been explained, within the context of nitrogen fixation, the GMR platform is able to identify microbial promoter sequences, which are active under environmental conditions of elevated exogenous nitrogen, which thereby allows the remodeled microbe to fix atmospheric nitrogen and deliver it to a target crop plant, under modern agricultural row crop conditions, and at a time when a plant needs the fixed nitrogen the most. See, FIG. 1E for a depiction of the time period in the corn growth cycle, at which nitrogen is needed most by the plant. The taught GMR platform is able to create remodeled microbes that supply nitrogen to a corn plant at the time period in which the nitrogen is needed, and also deliver such nitrogen even in the presence of exogenous nitrogen in the soil environment.


These promoters can be identified by rhizosphere RNA sequencing and read mapping to the microbe's genome sequence, and key pathways can be “reprogrammed” to be turned on or off during key stages of the plant growth cycle. Additionally, through whole genome sequencing of optimized microbes and mapping to previously-transformed sequences, the method has the ability to ensure that no transgenic sequences are accidentally released into the field through off-target insertion of plasmid DNA, low-level retention of plasmids not detected through PCR or antibiotic resistance, etc.


The GMR platform combines these approaches by evaluating microbes iteratively in the lab and plant environment, leading to microbes that are robust in greenhouse and field conditions rather than just in lab conditions.


Various aspects and embodiments of the taught GMR platform can be found in FIGS. 1F-1I. The GMR platform culminates in the derivation/creation/production of remodeled microbes that possess a plant-beneficial property, e.g. nitrogen fixation.


The traditional bioprospecting methods are not able to produce microbes having the aforementioned properties.


Properties of a Microbe Remodeled for Nitrogen Fixation


In the context of remodeling microbes for nitrogen fixation, there are several properties that the remodeled microbe may possess. For instance, FIG. 1J depicts five properties that can be possessed by remodeled microbes of the present disclosure.


Furthermore, as can be seen in Example 2, the present inventors have utilized the GMR platform to produce remodeled non-intergeneric bacteria (i.e. Kosakonia sacchari) capable of fixing atmospheric nitrogen and delivering said nitrogen to a corn plant, even under conditions in which exogenous nitrogen is present in the environment. See, FIG. 1K-M, which illustrate that the remodeling process successfully: (1) decoupled nifA expression from endogenous nitrogen regulation; and (2) improved the assimilation and excretion of fixed nitrogen.


These remodeled microbes ultimately result in corn yield improvement, when applied to corn crops. See, FIG. 1N.


The GMR Platform Provides an Approach to Nitrogen Fixation and Delivery that Solves Pressing Environmental Concerns


As explained previously, the nitrogen fertilizer produced by the industrial Haber-Bosch process is not well utilized by the target crop. Rain, runoff, heat, volatilization, and the soil microbiome degrade the applied chemical fertilizer. This equates to not only wasted money, but also adds to increased pollution instead of harvested yield. To this end, the United Nations has calculated that nearly 80% of fertilizer is lost before a crop can utilize it. Consequently, modern agricultural fertilizer production and delivery is not only deleterious to the environment, but it is extremely inefficient. See, FIG. 1O, illustrating the inefficiency of current nitrogen delivery systems, which result in underfertilized fields, over fertilized fields, and environmentally deleterious nitrogen runoff.


The current GMR platform, and resulting remodeled microbes, provide a better approach to nitrogen fixation and delivery to plants. As will be seen in the below Examples, the non-intergeneric remodeled microbes of the disclosure are able to colonize the roots of a corn plant and spoon feed said corn plants with fixed atmospheric nitrogen, even in the presence of exogenous nitrogen. This system of nitrogen fixation and delivery-enabled by the taught GMR platform-will help transform modern agricultural to a more environmentally sustainable system.


Example 2: Guided Microbial Remodeling—an Example Embodiment for the Rational Improvement of Nitrogen Fixation

A diversity of nitrogen fixing bacteria can be found in nature, including in agricultural soils. However, the potential of a microbe to provide sufficient nitrogen to crops to allow decreased fertilizer use may be limited by repression of nitrogenase genes in fertilized soils as well as low abundance in close association with crop roots. Identification, isolation and breeding of microbes that closely associate with key commercial crops might disrupt and improve the regulatory networks linking nitrogen sensing and nitrogen fixation and unlock significant nitrogen contributions by crop-associated microbes. To this end, nitrogen fixing microbes that associate with and colonize the root system of corn were identified. This step corresponds to the “Measure the Microbiome Composition” depicted in FIG. 1A and FIG. 1B.


Root samples from corn plants grown in agronomically relevant soils were collected, and microbial populations extracted from the rhizosphere and endosphere. Genomic DNA from these samples was extracted, followed by 16S amplicon sequencing to profile the community composition.


A Kosakonia sacchari microbe (strain PBC6.1) was isolated and classified through 16S rRNA and whole genome sequencing. This is a particularly interesting nitrogen fixer capable of colonizing to nearly 21% abundance of the root-associated microbiota (FIG. 2). To assess strain sensitivity to exogenous nitrogen, nitrogen fixation rates in pure culture were measured with the classical acetylene reduction assay (ARA) and varying levels of glutamine supplementation. The species exhibited a high level of nitrogen fixing activity in nitrogen-free media, yet exogenous fixed nitrogen repressed nif gene expression and nitrogenase activity (Strain PBC6.1, FIG. 3C, FIG. 3D). Additionally, when released ammonia was measured in the supernatant of PBC6.1 grown in nitrogen-fixing conditions, very little release of fixed nitrogen could be detected (FIG. 3E).


We hypothesized that PBC6.1 could be a significant contributor of fixed nitrogen in fertilized fields if regulatory networks controlling nitrogen metabolism were remodeled to allow optimal nitrogenase expression and ammonia release in the presence of fixed nitrogen.


Sufficient genetic diversity should exist within the PBC6.1 genome to enable broad phenotypic remodeling (as a result of remodeling the underlying genetic architecture in a non-intergeneric manner) without the insertion of transgenes or synthetic regulatory elements. The isolated strain has a genome of at least 5.4 Mbp and a canonical nitrogen fixation gene cluster. Related nitrogen metabolism pathways in PBC6.1 are similar to those of the model organism for nitrogen fixation, Klebsiella oxytoca m5a1.


Several gene regulatory network nodes were identified which may augment nitrogen fixation and subsequent transfer to a host plant, particularly in high exogenous concentrations of fixed nitrogen (FIG. 3A). The nifLA operon directly regulates the rest of the nif cluster through transcriptional activation by NifA and nitrogen- and oxygen-dependent repression of NifA by NifL. Disruption of nifL can abolish inhibition of NifA and improve nif expression in the presence of both oxygen and exogenous fixed nitrogen. Furthermore, expressing nifA under the control of a nitrogen-independent promoter may decouple nitrogenase biosynthesis from regulation by the NtrB/NtrC nitrogen sensing complex.


The assimilation of fixed nitrogen by the microbe to glutamine by glutamine synthetase (GS) is reversibly regulated by the two-domain adenylyltransferase (ATase) enzyme GlnE through the adenylylation and deadenylation of GS to attenuate and restore activity, respectively. Truncation of the GlnE protein to delete its adenylyl-removing (AR) domain may lead to constitutively adenylated glutamine synthetase, limiting ammonia assimilation by the microbe and increasing intra- and extracellular ammonia.


Finally, reducing expression of AmtB, the transporter responsible for uptake of ammonia, could lead to greater extracellular ammonia.


To generate rationally designed microbial phenotypes without the use of transgenes, two approaches were employed to remodel the underlying genetic architecture of the microbe: (1) creating markerless deletions of genomic sequences encoding protein domains or whole genes, and (2) rewiring regulatory networks by intragenomic promoter rearrangement.


Through an iterative remodeling process, several non-transgenic derivative strains of PBC6.1 were generated (Table 25).









TABLE 25







List of isolated and derivative K. sacchari strains


used in this work. Prm, promoter sequence


derived from the PBC6.1 genome; ΔglnEAR1 and


ΔglnEAR2, different truncated versions


of glnE gene removing the adenylyl-removing domain


sequence.










Strain ID
Genotype







PBC6.1
WT



PBC6.14
ΔnifL::Prm1



PBC6.15
ΔnifL::Prm5



PBC6.22
ΔnifL::Prm3



PBC6.37
ΔnifL::Prm1 ΔglnEAR2



PBC6.38
ΔnifL::Prm1 ΔglnEAR1



PBC6.93
ΔnifL::Prm1 ΔglnEAR2 ΔamtB



PBC6.94
ΔnifL::Prm1 ΔglnEAR1 ΔamtB










Several in vitro assays were performed to characterize specific phenotypes of the derivative strains. The ARA was used to assess strain sensitivity to exogenous nitrogen, in which PBC6.1 exhibited repression of nitrogenase activity at high glutamine concentrations (FIG. 3D). In contrast, most derivative strains showed a derepressed phenotype with varying levels of acetylene reduction observed at high glutamine concentrations. Transcriptional rates of nifA in samples analyzed by qPCR correlated well with acetylene reduction rates (FIG. 4), supporting the hypothesis that ni/L disruption and insertion of a nitrogen-independent promoter to drive nifA can lead to nif cluster derepression.


Strains with altered GlnE or AmtB activity showed markedly increased ammonium excretion rates compared to wild type or derivative strains without these mutations (FIG. 3E), illustrating the effect of these genotypes on ammonia assimilation and reuptake.


Two experiments were performed to study the interaction of PBC6.1 derivatives (remodeled microbes) with corn plants and quantify incorporation of fixed nitrogen into plant tissues. First, rates of microbial nitrogen fixation were quantified in a greenhouse study using isotopic tracers. Briefly, plants are grown with 15N labeled fertilizer, and diluted concentrations of 15N in plant tissues indicate contributions of fixed nitrogen from microbes. Corn seedlings were inoculated with selected microbial strains, and plants were grown to the V6 growth stage. Plants were subsequently deconstructed to enable measurement of microbial colonization and gene expression as well as measurement of 15N/14N ratios in plant tissues by isotope ratio mass spectrometry (IRMS). Analysis of the aerial tissue showed a small, nonsignificant contribution by PBC6.38 to plant nitrogen levels, and a significant contribution by PBC6.94 (p=0.011). Approximately 20% of the nitrogen found in above-ground corn leaves was produced by PBC6.94, with the remainder coming from the seed, potting mix, or “background” fixation by other soilborne microbes (FIG. 5C). This illustrates that our microbial breeding and remodeling pipeline can generate remodeled strains capable of making significant nitrogen contributions to plants in the presence of nitrogen fertilizer. Microbial transcription within plant tissues was measured, and expression of the nif gene cluster was observed in derivative remodeled strains, but not the wild type strain (FIG. 5B), showing the importance of nif derepression for contribution of BNF to crops in fertilized conditions. Root colonization measured by qPCR demonstrated that colonization density is different for each of the strains tested (FIG. 5A). A 50 fold difference in colonization was observed between PBC6.38 and PBC6.94. This difference could be an indication that PBC6.94 has reduced fitness in the rhizosphere relative to PBC6.38 as a result of high levels of fixation and excretion.


Methods


Media


Minimal medium contains (per liter) 25 g Na2HPO4, 0.1 g CaCL2-2H2O, 3 g KH2PO4, 0.25 g MgSO4·7H2O, 1 g NaCl, 2.9 mg FeCl3, 0.25 mg Na2MoO4·2H2O, and 20 g sucrose. Growth medium is defined as minimal medium supplemented with 50 ml of 200 mM glutamine per liter.


Isolation of Diazotrophs


Corn seedlings were grown from seed (DKC 66-40, DeKalb, IL) for two weeks in a greenhouse environment controlled from 22° C. (night) to 26° C. (day) and exposed to 16 hour light cycles in soil collected from San Joaquin County, CA. Roots were harvested and washed with sterile deionized water to remove bulk soil. Root tissues were homogenized with 2 mm stainless steel beads in a tissue lyser (TissueLyser II, Qiagen P/N 85300) for three minutes at setting 30, and the samples were centrifuged for 1 minute at 13,000 rpm to separate tissue from root-associated bacteria. Supernatants were split into two fractions, and one was used to characterize the microbiome through 16S rRNA amplicon sequencing and the remaining fraction was diluted and plated on Nitrogen-free Broth (NfB) media supplemented with 1.5% agar. Plates were incubated at 30° C. for 5-7 days. Colonies that emerged were tested for the presence of the nifH gene by colony PCR with primers Ueda19f and Ueda406r. Genomic DNA from strains with a positive nifH colony PCR was isolated (QIAamp DNA Mini Kit, Cat No. 51306, QIAGEN, Germany) and sequenced (Illumina MiSeq v3, SeqMatic, Fremont, CA). Following sequence assembly and annotation, the isolates containing nitrogen fixation gene clusters were utilized in downstream research.


Microbiome Profiling of Isolation Seedlings


Genomic DNA was isolated from root-associated bacteria using the ZR-96 Genomic DNA I Kit (Zymo Research P/N D3011), and 16S rRNA amplicons were generated using nextera-barcoded primers targeting 799f and 1114r. The amplicon libraries were purified and sequenced with the Illumina MiSeq v3 platform (SeqMatic, Fremont, CA). Reads were taxonomically classified using Kraken using the minikraken database (FIG. 2).


Acetylene Reduction Assay (ARA)


A modified version of the Acetylene Reduction Assay was used to measure nitrogenase activity in pure culture conditions. Strains were propagated from single colony in SOB (RPI, P/N S25040-1000) at 30° C. with shaking at 200 RPM for 24 hours and then subcultured 1:25 into growth medium and grown aerobically for 24 hours (30° C., 200 RPM). 1 ml of the minimal media culture was then added to 4 ml of minimal media supplemented with 0 to 10 mM glutamine in air-tight Hungate tubes and grown anaerobically for 4 hours (30° C., 200 RPM). 10% headspace was removed then replaced by an equal volume of acetylene by injection, and incubation continued for 1 hr. Subsequently, 2 ml of headspace was removed via gas tight syringe for quantification of ethylene production using an Agilent 6850 gas chromatograph equipped with a flame ionization detector (FID).


Ammonium Excretion Assay


Excretion of fixed nitrogen in the form of ammonia was measured using batch fermentation in anaerobic bioreactors. Strains were propagated from single colony in 1 ml/well of SOB in a 96 well DeepWell plate. The plate was incubated at 30° C. with shaking at 200 RPM for 24 hours and then diluted 1:25 into a fresh plate containing 1 ml/well of growth medium. Cells were incubated for 24 hours (30° C., 200 RPM) and then diluted 1:10 into a fresh plate containing minimal medium. The plate was transferred to an anaerobic chamber with a gas mixture of >98.5% nitrogen, 1.2-1.5% hydrogen and <30 ppM oxygen and incubated at 1350 RPM, room temperature for 66-70 hrs. Initial culture biomass was compared to ending biomass by measuring optical density at 590 nm. Cells were then separated by centrifugation, and supernatant from the reactor broth was assayed for free ammonia using the Megazyme Ammonia Assay kit (P/N K-AMIAR) normalized to biomass at each timepoint.


Extraction of Root-Associated Microbiome


Roots were shaken gently to remove loose particles, and root systems were separated and soaked in a RNA stabilization solution (Thermo Fisher P/N AM7021) for 30 minutes. The roots were then briefly rinsed with sterile deionized water. Samples were homogenized using bead beating with ½-inch stainless steel ball bearings in a tissue lyser (TissueLyser II, Qiagen P/N 85300) in 2 ml of lysis buffer (Qiagen P/N 79216). Genomic DNA extraction was performed with ZR-96 Quick-gDNA kit (Zymo Research P/N D3010), and RNA extraction using the RNeasy kit (Qiagen P/N 74104).


Root Colonization Assay


Four days after planting, 1 ml of a bacterial overnight culture (approximately 109 cfu) was applied to the soil above the planted seed. Seedlings were fertilized three times weekly with 25 ml modified Hoagland's solution supplemented with 0.5 mM ammonium nitrate. Four weeks after planting, root samples were collected and the total genomic DNA (gDNA) was extracted. Root colonization was quantified using qPCR with primers designed to amplify unique regions of either the wild type or derivative strain genome. QPCR reaction efficiency was measured using a standard curve generated from a known quantity of gDNA from the target genome. Data was normalized to genome copies per g fresh weight using the tissue weight and extraction volume. For each experiment, the colonization numbers were compared to untreated control seedlings.


In Planta Transcriptomics


Transcriptional profiling of root-associated microbes was measured in seedlings grown and processed as described in the Root Colonization Assay. Purified RNA was sequenced using the Illumina NextSeq platform (SeqMatic, Fremont, CA). Reads were mapped to the genome of the inoculated strain using bowtie2 using ‘--very-sensitive-local’ parameters and a minimum alignment score of 30. Coverage across the genome was calculated using samtools. Differential coverage was normalized to housekeeping gene expression and visualized across the genome using Circos and across the nif gene cluster using DNAplotlib. Additionally, the in planta transcriptional profile was quantified via targeted Nanostring analysis. Purified RNA was processed on an nCounter Sprint (Core Diagnostics, Hayward, CA).


15N Dilution Greenhouse Study


A 15N fertilizer dilution experiment was performed to assess optimized strain activity in planta. A planting medium containing minimal background N was prepared using a mixture of vermiculite and washed sand (5 rinses in DI H2O). The sand mixture was autoclaved for 1 hour at 122° C. and approximately 600 g measured out into 40 cubic inch (656 mL) pots, which were saturated with sterile DI H2O and allowed to drain 24 hours before planting. Corn seeds (DKC 66-40) were surface sterilized in 0.625% sodium hypochlorite for 10 minutes, then rinsed five times in sterile distilled water and planted 1 cm deep. The plants were maintained under fluorescent lamps for four weeks with 16-hour day length at room temperatures averaging 22° C. (night) to 26° C. (day).


Five days after planting, seedlings were inoculated with a 1 ml suspension of cells drenched directly over the emerging coleoptile. Inoculum was prepared from 5 ml overnight cultures in SOB, which were spun down and resuspended twice in 5 ml PBS to remove residual SOB before final dilution to OD of 1.0 (approximately 109 CFU/ml). Control plants were treated with sterile PBS, and each treatment was applied to ten replicate plants.


Plants were fertilized with 25 ml fertilizer solution containing 2% 15N-enriched 2 mM KNO3 on 5, 9, 14, and 19 days after planting, and the same solution without KNO3 on 7, 12, 16, and 18 days after planting. The fertilizer solution contained (per liter) 3 mmol CaCl2), 0.5 mmol KH2PO4, 2 mmol MgSO4, 17.9 μmol FeSO4, 2.86 mg H3BO3, 1.81 mg MnCl2·4H2O, 0.22 mg ZnSO4·7H2O, 51 μg CuSO4·5H2O, 0.12 mg Na2MoO4·2H2O, and 0.14 nmol NiCl2. All pots were watered with sterile DI H2O as needed to maintain consistent soil moisture without runoff.


At four weeks, plants were harvested and separated at the lowest node into samples for root gDNA and RNA extraction and aerial tissue for IRMS. Aerial tissues were wiped as needed to remove sand, placed whole into paper bags and dried for at least 72 hours at 60° C. Once completely dry, total aerial tissue was homogenized by bead beating and 5-7 mg samples were analyzed by isotope ratio mass spectrometry (IRMS) for δ15N by the MBL Stable Isotope Laboratory (The Ecosystems Center, Woods Hole, MA). Percent NDFA was calculated using the following formula: % NDFA=(δ15N of UTC average— δ15N of sample)/(δ15N of UTC average)×100.


Example 3: Field Trials with Remodeled Microbes of the Disclosure—Summer 2016

In order to evaluate the efficacy of remodeled strains of the present disclosure on corn growth and productivity under varying nitrogen regimes, field trials were conducted.


Trials were conducted with (1) seven subplot treatments of six strains plus the control—four main plots comprised 0, 15, 85, and 100% of maximum return to nitrogen (MRTN) with local verification. The control (UTC only) was conducted with 10 100% MRTN plus, 5, 10, or 15 pounds. Treatments had four replications.


Plots of corn (minimum) were 4 rows of 30 feet in length, with 124 plots per location. All observations were taken from the center two rows of the plots, and all destructive sampling was taken from the outside rows. Seed samples were refrigerated until 1.5 to 2 hours prior to use.


Local Agricultural Practice: The seed was a commercial corn without conventional fungicide and insecticide treatment. All seed treatments were applied by a single seed treatment specialist to assure uniformity. Planting date, seeding rate, weed/insect management, etc. were left to local agricultural practices. With the exception of fungicide applications, standard management practices were followed.


Soil Characterization: Soil texture and soil fertility were evaluated. Soil samples were pre-planted for each replicate to insure residual nitrate levels lower than 501bs/Ac. Soil cores were taken from 0 cm to 30 cm. The soil was further characterized for pH, CEC, total K and P.


Assessments: The initial plant population was assessed 14 days after planting (DAP)/acre, and were further assessed for: (1) vigor (1 to 10 scale, w/10=excellent) 14 DAP & V10; (2) recordation of disease ratings any time symptoms are evident in the plots; (3) record any differences in lodging if lodging occurs in the plots; (4) yield (Bu/acre), adjusted to standard moisture pct; (5) test weight; and (6) grain moisture percentage.


Sampling Requirements: The soil was sampled at three timepoints (prior to trial initiation, V10-VT, 1 week post-harvest). All six locations and all plots were sampled at 10 grams per sample (124 plots×3 timepoints×6 locations).


Colonization Sampling: Colonization samples were collected at two timepoints (V10 and VT) for five locations and six timepoints (V4, V8, V10, VT, R5, and Post-Harvest). Samples were collected as follows: (1) from 0% and 100% MRTN, 60 plots per location; (2) 4 plants per plot randomly selected from the outside rows; (3) 5 grams of root, 8 inches of stalk, and top three leaves-bagged and IDed each separately—12/bags per plot; (4) five locations (60 plots×2 timepoints×12 bags/plot); and one location (60 plots×6 timepoints×12 bags/plot.


Normalized difference vegetation index (NDVI) determination was made using a Greenseeker instrument at two timepoints (V4-V6 and VT). Assessed each plot at all six locations (124 plots×2 timepoints×6 locations).


Root analysis was performed with Win Rhizo from one location that best illustrated treatment differentiation. Ten plants per plot were randomly sampled (5 adjacent from each outside row; V3-V4 stage plants were preferred) and gently washed to remove as much dirt as reasonable. Ten roots were placed in a plastic bag and labelled. Analyzed with WinRhizo Root Analysis.


Stalk Characteristics were measured at all six locations between R2 and R5. The stalk diameter of ten plants per plot at the 6″ height were recorded, as was the length of the first internode above the 6″ mark. Ten plants were monitored; five consecutive plants from the center of the two inside rows. Six locations were evaluated (124 plots×2 measures×6 locations).


The tissue nitrates were analyzed from all plots and all locations. An 8″ segment of stalk beginning 6″ above the soil when the corn is between one and three weeks after black layer formation; leaf sheaths were removed. All locations and plots were evaluated (6 locations×124 plots).


The following weather data was recorded for all locations from planting to harvest: daily maximum and minimum temperatures, soil temperature at seeding, daily rainfall plus irrigation (if applied), and any unusual weather events such as excessive rain, wind, cold, or heat.


Yield data across all six locations is presented in Table 26. Nitrogen rate had a significant impact on yield, but strains across nitrogen rates did not. However, at the lowest nitrogen rate, strains CI006, CM029, and CI019 numerically out-yielded the UTC by 4 to 6 bu/acre. Yield was also numerically increased 2 to 4 bu/acre by strains CM029, CI019, and CM081 at 15% MRTN.









TABLE 26





Yield data across all six locations
























Vigor_
Vigor_
Stalk Diameter
Internode
NDVI_
NDVI_


MRTN %
YLD (bu)
E
L
(mm)
Length (in)
Veg
Rep





 0
143.9
7.0
5.7
18.87
7.18
64.0
70.6


 15
165.9
7.2
6.3
19.27
7.28
65.8
72.5


 85
196.6
7.1
7.1
20.00
7.31
67.1
74.3


100
197.3
7.2
7.2
20.23
7.37
66.3
72.4







Vigor_
Vigor_
Stalk Diameter
Internode
NDVI_
NDVI_


Strain
YLD (bu)
E
L
(mm)
Length (in)
Veg
Rep





CI006 (1)
176.6
7.2
6.6
19.56
18.78 
66.1
72.3


CM029 (2)
176.5
7.1
6.5
19.54
18.61 
65.4
71.9


CM038 (3)
175.5
7.2
6.5
19.58
18.69 
65.7
72.8


CI019 (4)
176.0
7.1
6.6
19.51
18.69 
65.5
72.9


CM081 (5)
176.2
7.1
6.6
19.57
18.69 
65.8
73.1


CM029/CM081
174.3
7.1
6.6
19.83
18.79 
66.2
72.5


(6)









UTC (7)
176.4
7.1
6.6
19.54
18.71 
65.9
71.7







Vigor_
Vigor_
Stalk Diameter
Internode
NDVI_
NDVI_


MRTN / Strain
YLD (bu)
E
L
(mm)
Length (in)
Veg
Rep





 0 1
145.6
7.0
5.6
19.07
7.12
63.5
70.3


 0 2
147.0
7.0
5.5
18.74
7.16
64.4
70.4


 0 3
143.9
7.0
5.5
18.83
7.37
64.6
70.5


 0 4
146.0
6.9
5.7
18.86
7.15
63.4
70.7


 0 5
141.7
7.0
5.8
18.82
7.05
63.6
70.9


 0 6
142.2
7.2
5.8
19.12
7.09
64.7
69.9


 0 7
141.2
7.0
5.8
18.64
7.32
64,0
71.4


 15 1
164.2
7.3
6.1
19.09
7.21
66.1
71.5


 15 2
167.3
7.2
6.3
19.32
7.29
65.5
72.7


 15 3
165.6
7.3
6.3
19.36
7.23
64.8
72.5


 15 4
167.9
7.3
6.4
19.31
7.51
66.1
72.3


 15 5
169.3
7.2
6.2
19.05
7.32
66.0
72.8


 15 6
161.9
7.1
6.3
19.45
7.20
66.2
72.2


 15 7
165.1
7.3
6.4
19.30
7.18
66.0
73.3


 85 1
199.4
7.3
7.2
19.70
7.32
67,2
74.0


 85 2
195.1
7.1
7.2
19.99
7.09
66,5
74.4


 85 3
195.0
7.0
7.0
20.05
7.26
67.3
74.6


 85 4
195.6
7.2
7.1
20.04
7.29
66.4
74.4


 85 5
196.4
7.2
7.0
19.87
7.39
67.3
74.5


 85 6
195.1
7.0
6.9
20.35
7.34
67.4
74.4


 85 7
199.5
6.9
7.2
19.97
7.48
67.4
74.1


100 1
197.1
7.2
7.3
20.38
7.68
67.5
73.4


100 2
196.5
7.0
7.1
20.11
7.21
65.3
70.2


100 3
197.6
7.5
7.3
20.08
7.42
66.3
73.4


100 4
194.6
7.1
7.1
19.83
7.40
66.1
74.1


100 5
197.4
7.2
7.3
20.53
7.36
66.2
74.3


100 6
198.1
7.2
7.4
20.40
7.16
66.6
73.6


100 7
199.9
7.2
7.2
20.26
7.32
66.2
68.1









Another analysis approach is presented in Table 27. The table comprises the four locations where the response to nitrogen was the greatest which suggests that available residual nitrogen was lowest. This approach does not alter the assessment that the nitrogen rate significantly impacted yield, which strains did not when averaged across all nitrogen rates. However, the numerical yield advantage at the lowest N rate is more pronounced for all strains, particularly CI006, CM029, and CM029/CM081 where yields were increased from 8 to 10 bu/acre. At 1500 MRTN, strain CM081 outyielded UTC by 5 bu.









TABLE 27







Yield data across four locations


4 Location Average - SGS, AgIdea, Bennett, RFR















YLD (bu)
Vigor_E
Vigor_L
Stalk Diameter (mm)
Internode Length (in)















MRTN %







0
137.8
7.3
5.84
18.10
5.36


15
162.1
7.5
6.63
18.75
5.40


85
199.2
7.4
7.93
19.58
5.62


100
203.5
7.5
8.14
19.83
5.65


Strain







CI006 (1)
175.4
7.5
7.08
19.03
5.59


CM029 (2)
176.1
7.4
7.08
19.09
5.39


CM038 (3)
175.3
7.5
7.05
19.01
5.59


CM019 (4)
174.8
7.5
7.16
19.02
5.45


CM081 (5)
176.7
7.4
7.16
19.00
5.53


CM029/CM081 (6)
175.1
7.4
7.17
19.33
5.46


UTC (7)
176.0
7.3
7.27
18.98
5.55


MRTN/Strain


















0
1
140.0
7.3
5.69
18.32
5.28


0
2
140.7
7.4
5.69
18.19
5.23


0
3
135.5
7.3
5.63
17.95
5.50


0
4
138.8
7.3
5.81
17.99
5.36


0
5
136.3
7.3
6.06
18.05
5.34


0
6
141.4
7.5
6.00
18.43
5.30


0
7
131.9
7.3
6.00
17.75
5.48


15
1
158.0
7.6
6.44
18.53
5.34


15
2
164.1
7.5
6.56
19.13
5.42


15
3
164.3
7.6
6.63
18.68
5.51


15
4
163.5
7.6
6.81
18.84
5.34


15
5
166.8
7.5
6.63
18.60
5.39


15
6
156.6
7.4
6.56
18.86
5.41


15
7
161.3
7.5
6.81
18.62
5.42


85
1
199.4
7.6
8.00
19.15
5.63


85
2
199.0
7.4
8.09
19.49
5.46


85
3
198.2
7.4
7.75
19.88
5.69


85
4
196.8
7.4
8.00
19.65
5.60


85
5
199.5
7.4
7.75
19.26
5.70


85
6
198.7
7.3
7.81
19.99
5.61


85
7
202.8
7.2
8.13
19.66
5.65


100
1
204.3
7.4
8.19
20.11
6.10


100
2
200.6
7.3
8.00
19.53
5.46


100
3
203.3
7.7
8.19
19.55
5.67


100
4
200.2
7.6
8.00
19.59
5.49


100
5
203.9
7.4
8.19
20.08
5.68


100
6
203.8
7.5
8.31
20.05
5.52


100
7
208.1
7.4
8.13
19.90
5.63









The results from the field trial are also illustrated in FIGS. 9-15. The results indicate that the microbes of the disclosure are able to increase plant yield, which points to the ability of the taught microbes to increase nitrogen fixation in an important agricultural crop, i.e. corn.


The field based results further validate the disclosed methods of non-intergenerially modifying the genome of selected microbial strains, in order to bring about agriculturally relevant results in a field setting when applying said engineered strains to a crop.



FIG. 6 depicts the lineage of modified remodeled strains that were derived from strain CI006 (WT Kosakonia sacchari). The field data demonstrates that an engineered derivative of the CI006 WT strain, i.e. CM029, is able to bring about numerically relevant results in a field setting. For example, Table 26 illustrates that at 0% MRTN CM029 yielded 147.0 bu/acre compared to untreated control at 141.2 bu/acre (an increase of 5.8 bu/acre). Table 26 also illustrates that at 15% MRTN CM029 yielded 167.3 bu/acre compared to untreated control at 165.1 bu/acre (an increase of 2.2 bu/acre). Table 27 is supportive of these conclusions and illustrates that at 0% MRTN CM029 yielded 140.7 bu/acre compared to untreated control at 131.9 bu/acre (an increase of 8.8 bu/acre). Table 27 also illustrates that at 15% MRTN CM029 yielded 164.1 bu/acre compared to untreated control at 161.3 bu/acre (an increase of 2.8 bu/acre).



FIG. 7 depicts the lineage of modified remodeled strains that were derived from strain CI019 (WT Rahnella aquatilis). The field data demonstrates that an engineered derivative of the CI019 WT strain, i.e. CM081, is able to bring about numerically relevant results in a field setting. For example, Table 26 illustrates that at 15% MRTN CM081 yielded 169.3 bu/acre compared to untreated control at 165.1 bu/acre (an increase of 4.2 bu/acre). Table 27 is supportive of these conclusions and illustrates that at 0% MRTN CM081 yielded 136.3 bu/acre compared to untreated control at 131.9 bu/acre (an increase of 4.4 bu/acre). Table 27 also illustrates that at 15% MRTN CM081 yielded 166.8 bu/acre compared to untreated control at 161.3 bu/acre (an increase of 5.5 bu/acre).


Further, one can see in Table 27 that the combination of CM029/CM081 at 0% MRTN yielded 141.4 bu/acre compared to untreated control at 131.9 bu/acre (an increase of 9.5 bu/acre).


Example 4: Field Trials with Remodeled Microbes of the Disclosure—Summer 2017

In order to evaluate the efficacy of remodeled strains of the present disclosure on corn growth and productivity under varying nitrogen regimes, field trials were conducted. The below field data demonstrates that the non-intergeneric microbes of the disclosure are able to fix atmospheric nitrogen and deliver said nitrogen to a plant-resulting in increased yields—in both a nitrogen limiting environment, as well as a non-nitrogen limiting environment.


Trials were conducted at seven locations across the United states with six geographically diverse Midwestern locations. Five nitrogen regimes were used for fertilizer treatments: 100% of standard agricultural practice of the site/region, 100% minus 25 pounds, 100% minus 50 pounds, 100% minus 75 pounds, and 0%; all per acre. The pounds of nitrogen per acre for the 100% regime depended upon the standard agricultural practices of the site/region. The aforementioned nitrogen regimes ranged from about 153 pounds per acre to about 180 pounds per acre, with an average of about 164 pounds of nitrogen per acre.


Within each fertilizer regime there were 14 treatments. Each regime had six replications, and a split plot design was utilized. The 14 treatments included: 12 different microbes, 1 UTC with the same fertilizer rate as the main plot, and 1 UTC with 100% nitrogen. In the 100% nitrogen regime the 2nd UTC is 100 plus 25 pounds.


Plots of corn, at a minimum, were 4 rows of 30 feet in length (30 inches between rows) with 420 plots per location. All observations, unless otherwise noted, were taken from the center two rows of the plants, and all destructive sampling was taken from the outside rows. Seed samples were refrigerated until 1.5 to 2 hours prior to use.


Local Agricultural Practice: The seed was a commercial corn applied with a commercial seed treatment with no biological co-application. The seeding rate, planting date, weed/insect management, harvest times, and other standard management practices were left to the norms of local agricultural practices for the regions, with the exception of fungicide application (if required).


Microbe Application: The microbes were applied to the seed in a seed treatment over seeds that had already received a normal chemical treatment. The seed were coated with fermentation broth comprising the microbes.


Soil Characterization: Soil texture and soil fertility were evaluated. Standard soil sampling procedures were utilized, which included soil cores of depths from 0-30 cm and 30-60 cm. The standard soil sampling included a determination of nitrate nitrogen, ammonium nitrogen, total nitrogen, organic matter, and CEC. Standard soil sampling further included a determination of pH, total potassium, and total phosphorous. To determine the nitrogen fertilizer levels, preplant soil samples from each location were taken to ensure that the 0-12″ and potentially the 12″ to 24″ soil regions for nitrate nitrogen.


Prior to planting and fertilization, 2 ml soil samples were collected from 0 to 6-12″ from the UTC. One sample per replicate per nitrogen region was collected using the middle of the row. (5 fertilizer regimes×6 replicates=thirty soil samples).


Post-planting (V4-V6), 2 ml soil samples were collected from 0 to 6-12″ from the UTC. One sample per replicate per nitrogen region was collected using the middle of the row. (5 fertilizer regimes×6 replicates=thirty soil samples).


Post-harvest (V4-V6), 2 ml soil samples were collected from 0 to 6-12″ from the UTC. One sample per replicate per nitrogen region was collected using the middle of the row. Additional post-harvest soil sample collected at 0-12″ from the UTC and potentially 12-24″ from the UTC (5 fertilizer regimes×6 replicates=thirty soil samples).


A V6-V10 soil sample from each fertilizer regime (excluding the treatment of 100% and 100%+25 lbs [in the 100% block] for all fertilizer regimes at 0-12″ and 12-24″. (5 fertilizer regimes×2 depths=10 samples per location).


Post-harvest soil sample from each fertilizer regime (excluding the treatment of 100% and 100%+25 lbs [in the 100% block] for all fertilizer regimes at 0-12″ and 12-24″. (5 fertilizer regimes×2 depths=10 samples per location).


Assessments: The initial plant population was assessed at ˜50% UTC and the final plant population was assessed prior to harvest. Assessment included (1) potentially temperature (temperature probe); (2) vigor (1-10 scale with 10=excellent) at V4 and V8-V10; (3) plant height at V8-V10 and V14; (4) yield (bushels/acre) adjusted to standard moisture percentage; (5) test weight; (6) grain moisture percentage; (7) stalk nitrate tests at black layer (420 plots×7 locations); (8) colonization with 1 plant per plot in zip lock bag at 0% and 100% fertilizer at V4-V6 (1 plant×14 treatments×6 replicates×2 fertilizer regimes=168 plants); (9) transcriptomics with 1 plant per plot in zip lock bag at 0% and 100% fertilizer at V4-V6 (1 plant×14 treatments×6 replicates×2 fertilizer regimes=168 plants); (10) Normalized difference vegetative index (NDVI) or normalized difference red edge (NDRE) determination using a Greenseeker instrument at two time points (V4-V6 and VT) to assess each plot at all 7 locations (420 plots×2 time points×7 locations=5,880 data points); (11) stalk characteristics measured at all 7 locations between R2 and R5 by recording the stalk diameter of 10 plants/plot at 6″ height, record length of first internode above the 6″ mark, 10 plants monitored (5 consecutive plants from center of two inside rows) (420 plots×10 plants×7 locations=29,400 data points).


Monitoring Schedule: Practitioners visited all trials at V3-V4 stage to assess early-season response to treatments and during reproductive growth stage to monitor maturity. Local cooperator visited research trial on an on-going basis.


Weather Information: Weather data spanning from planting to harvest was collected and consisted of daily minimum and maximum temperatures, soil temperature at seeding, daily rainfall plus irrigation (if applied), and unusual weather events such as excessive wind, rain, cold, heat.


Data Reporting: Including the data indicated above, the field trials generated data points including soil textures; row spacing; plot sizes; irrigation; tillage; previous crop; seeding rate; plant population; seasonal fertilizer inputs including source, rate, timing, and placement; harvest area dimensions, method of harvest, such as by hand or machine and measurement tools used (scales, yield monitor, etc.)


Results: Select results from the aforementioned field trial are reported in FIG. 16 and FIG. 17.


In FIG. 16, it can be seen that a remodeled microbe of the disclosure (i.e. 6-403) resulted in a higher yield than the wild type strain (WT) and a higher yield than the untreated control (UTC). The “−25 lbs N” treatment utilizes 25 lbs less N per acre than standard agricultural practices of the region. The “100% N” UTC treatment is meant to depict standard agricultural practices of the region, in which 100% of the standard utilization of N is deployed by the farmer. The microbe “6-403” was deposited as NCMA 201708004 and can be found in Table 1. This is a mutant Kosakonia sacchari (also called CM037) and is a progeny mutant strain from CI006 WT.


In FIG. 17, the yield results obtained demonstrate that the remodeled microbes of the disclosure perform consistently across locations. Furthermore, the yield results demonstrate that the microbes of the disclosure perform well in both a nitrogen stressed environment (i.e. a nitrogen limiting environment), as well as an environment that has sufficient supplies of nitrogen (i.e. anon-nitrogen-limiting condition). The microbe “6-881” (also known as CM094, PBC6.94), and which is a progeny mutant Kosakonia sacchari strain from CI006 WT, was deposited as NCMA 201708002 and can be found in Table 1. The microbe “137-1034,” which is a progeny mutant Klebsiella variicola strain from CI137 WT, was deposited as NCMA 201712001 and can be found in Table 1. The microbe “137-1036,” which is a progeny mutant Klebsiella variicola strain from CI137 WT, was deposited as NCMA 201712002 and can be found in Table 1. The microbe “6-404” (also known as CM38, PBC6.38), and which is a progeny mutant Kosakonia sacchari strain from CI006 WT, was deposited as NCMA 201708003 and can be found in Table 1.


Example 5: Genus of Non-Intergeneric Remodeled Microbes Beneficial for Agricultural Systems

The remodeled microbes of the present disclosure were evaluated and compared against one another for the production of nitrogen produced in an acre across a season. See FIG. 8, FIG. 24, and FIG. 25.


It is hypothesized by the inventors that in order for a population of engineered non-intergeneric microbes to be beneficial in a modern row crop agricultural system, then the population of microbes needs to produce at least one pound or more of nitrogen per acre per season.


To that end, the inventors have surprisingly discovered a functional genus of microbes that are able to contribute, inter alia, to: increasing yields in non-leguminous crops; and/or lessening a farmer's dependence upon exogenous nitrogen application; and/or the ability to produce at least one pound of nitrogen per acre per season, even in non-nitrogen-limiting environments, said genus being defined by the product of colonization ability×mmol of N produced per microbe per hour (i.e. the line partitioning FIGS. 8, 24, and 25).


With respect to FIGS. 8, 24, and 25, certain data utilizing microbes of the disclosure was aggregated, in order to depict a heatmap of the pounds of nitrogen delivered per acre-season by microbes of the disclosure, which are recorded as a function of microbes per g-fresh weight by mmol of nitrogen/microbe-hr. Below the thin line that transects the larger images are the microbes that deliver less than one pound of nitrogen per acre-season, and above the line are the microbes that deliver greater than one pound of nitrogen per acre-season.


Field Data & Wild Type Colonization Heatmap: The microbes utilized in the FIG. 8 heatmap were assayed for N production in corn. For the WT strains CI006 and CI019, corn root colonization data was taken from a single field site. For the remaining strains, colonization was assumed to be the same as the WT field level. N-fixation activity was determined using an in vitro ARA assay at 5 mM glutamine. The table below the heatmap in FIG. 8 gives the precise value of mmol N produced per microbe per hour (mmol N/Microbe hr) along with the precise CFU per gram of fresh weight (CFU/g fw) for each microbe shown in the heatmap.


Field Data Heatmap: The data utilized in the FIG. 24 heatmap is derived from microbial strains assayed for N production in corn in field conditions. Each point represents lb N/acre produced by a microbe using corn root colonization data from a single field site. N-fixation activity was determined using in vitro ARA assay at 5 mM N in the form of glutamine or ammonium phosphate. The below Table 28 gives the precise value of mmol N produced per microbe per hour (mmol N/Microbe hr) along with the precise CFU per gram of fresh weight (CFU/g fw) for each microbe shown in the heatmap of FIG. 24.


Greenhouse & Laboratory Data Heatmap: The data utilized in the FIG. 25 heatmap is derived from microbial strains assayed for N production in corn in laboratory and greenhouse conditions. Each point represents lb N/acre produced by a single strain. White points represent strains in which corn root colonization data was gathered in greenhouse conditions. Black points represent mutant strains for which corn root colonization levels are derived from average field corn root colonization levels of the wild-type parent strain. Hatched points represent the wild type parent strains at their average field corn root colonization levels. In all cases, N-fixation activity was determined by in vitro ARA assay at 5 mM N in the form of glutamine or ammonium phosphate. The below Table 29 gives the precise value of mmol N produced per microbe per hour (mmol N/Microbe hr) along with the precise CFU per gram of fresh weight (CFU/g fw) for each microbe shown in the heatmap of FIG. 25.









TABLE 28







FIG. 24 - Field Data Heatmap












Activity (mmol
Peak Colonization




Strain Name
N/Microbe hr)
(CFU/g fw)
N Produced/acre season
Taxonomic Designation














CI006
3.88E−16
1.50E+07
0.24

Kosakonia sacchari



6-404
1.61E−13
3.50E+05
7.28

Kosakonia sacchari



6-848
1.80E−13
2.70E+05
1.97

Kosakonia sacchari



6-881
1.58E−13
5.00E+05
3.20

Kosakonia sacchari



6-412
4.80E−14
1.30E+06
2.53

Kosakonia sacchari



6-403
1.90E−13
1.30E+06
10.00

Kosakonia sacchari



CI019
5.33E−17
2.40E+06
0.01

Rahnella aquatilis



19-806
6.65E−14
2.90E+06
7.80

Rahnella aquatilis



19-750
8.90E−14
2.60E+05
0.94

Rahnella aquatilis



19-804
1.72E−14
4.10E+05
0.29

Rahnella aquatilis



CI137
3.24E−15
6.50E+06
0.85

Klebsiella variicola



137-1034
1.16E−14
6.30E+06
7.96

Klebsiella variicola



137-1036
3.47E−13
1.30E+07
182.56

Klebsiella variicola



137-1314
1.70E−13
1.99E+04
0.14

Klebsiella variicola



137-1329
1.65E−13
7.25E+04
0.48

Klebsiella variicola



63
3.60E−17
3.11E+05
0.00

Rahnella aquatilis



63-1146
1.90E−14
5.10E+05
0.39

Rahnella aquatilis



1021
1.77E−14
2.69E+07
19.25

Kosakonia pseudosacchari



728
5.56E−14
1445240.09
3.25

Klebsiella variicola

















TABLE 29







FIG. 25 - Greenhouse & Laboratory Data Heatmap












Activity (mmol N/
Peak Colonization




Strain Name
Microbe hr)
(CFU/g fw)
N Produced/acre season
Taxonomic Designation














CI006
 3.88E−16
1.50E+07
0.24

Kosakonia sacchari



6-400
 2.72E−13
1.79E+05
1.97

Kosakonia sacchari



6-397
 1.14E−14
1.79E+05
0.08

Kosakonia sacchari



CI137
 3.24E−15
6.50E+06
0.85

Klebsiella variicola



137-1586
 1.10E−13
1.82E+06
8.10

Klebsiella variicola



137-1382
 4.81E−12
1.82E+06
354.60

Klebsiella variicola



1021
 1.77E−14
2.69E+07
19.25

Kosakonia pseudosacchari



1021-1615
 1.20E−13
2.69E+07
130.75

Kosakonia pseudosacchari



1021-1619
 3.93E−14
2.69E+07
42.86

Kosakonia pseudosacchari



1021-1612
 1.20E−13
2.69E+07
130.75

Kosakonia pseudosacchari



1021-1623
 4.73E−17
2.69E+07
0.05

Kosakonia pseudosacchari



1293
 5.44E−17
8.70E+08
1.92

Azospirillum lipolerum



1116
 1.05E−14
1.37E+07
5.79

Enterobacter sp.



1113
 8.05E−15
4.13E+07
13.45

Enterobacter sp.



910
 1.19E−13
1.34E+06
6.46

Kluyvera iniermedia



910-1246
 2.16E−13
1.34E+06
11.69

Kluyvera intermedia



850
7.2301E−16
1.17E+06
0.03

Achromobacter spiritinus



852
 5.96E−16
1.07E+06
0.03

Achromobacter marplatensis



853
 6.42E−16
2.55E+06
0.07

Microbacterium morale










Conclusions: The data in FIGS. 8, 24, 25, and Tables 28 and 29, illustrates more than a dozen representative members of the described genus (i.e. microbes to the right of the line in the figures). Further, these numerous representative members come from a diverse array of taxonomic genera, which can be found in the above Tables 28 and 29. Further still, the inventors have discovered numerous genetic attributes that depict a structure/function relationship that is found in many of the microbes. These genetic relationships can be found in the numerous tables of the disclosure setting forth the genetic modifications introduced by the inventors, which include introducing at least one genetic variation into at least one gene, or non-coding polynucleotide, of the nitrogen fixation or assimilation genetic regulatory network.


Consequently, the newly discovered genus is supported by: (1) a robust dataset, (2) over a dozen representative members, (3) members from diverse taxonomic genera, and (4) classes of genetic modifications that define a structure/function relationship, in the underlying genetic architecture of the genus members.


Example 6: Growth Chamber Assays for Combined Clothianidin and Non-Intergenic Remodeled Microbes in Corn

An experiment will be conducted utilizing one or more of the deposited nine microbes described in Table 1 (6 non-intergenic remodeled microbes and 3 WT microbes), in combination with an insecticide, clothianidin. The microbe and clothianidin combination are used to treat corn seed in growth chamber experiments.


Growth chamber experiments are conducted in which corn seed is allowed to germinate under controlled standard growth conditions. The experiment includes: (a) untreated corn seed, (b) corn seed treated with clothianidin, (c) corn seed treated with one or more of the microbes described in Table 1, and (d) corn seed treated with a combination of clothianidin and one or more of the microbes described in Table 1. There will be approximately 100 seeds per treatment.


The corn seed treated with the combination of one or more of the 6 non-intergenic remodeled microbes from Table 1 and clothianidin is expected to exhibit (1) greater numbers of seed that germinate, (2) faster germination times, and (3) reaching the third leaf collar vegetative stage faster; as compared to all of the other treatment groups.


The corn seed treated with the combination of one or more of the 6 non-intergenic remodeled microbes from Table 1 and clothianidin is expected to reveal a synergistic effect as compared to the other treatment groups.


Example 7: Growth Chamber Assays for Combined Thiamethoxam and Non-Intergenic Remodeled Microbes in Corn

An experiment will be conducted utilizing one or more of the deposited nine microbes described in Table 1 (6 non-intergenic remodeled microbes and 3 WT microbes), in combination with an insecticide, thiamethoxam. The microbe and thiamethoxam combination are used to treat corn seed in growth chamber experiments.


Growth chamber experiments are conducted in which corn seed is allowed to germinate under controlled standard growth conditions. The experiment includes: (a) untreated corn seed, (b) corn seed treated with thiamethoxam, (c) corn seed treated with one or more of the microbes described in Table 1, and (d) corn seed treated with a combination of thiamethoxam and one or more of the microbes described in Table 1. There will be approximately 100 seeds per treatment.


The corn seed treated with the combination of one or more of the 6 non-intergenic remodeled microbes from Table 1 and thiamethoxam is expected to exhibit (1) greater numbers of seed that germinate, (2) faster germination times, and (3) reaching the third leaf collar vegetative stage faster; as compared to all of the other treatment groups.


The corn seed treated with the combination of one or more of the 6 non-intergenic remodeled microbes from Table 1 and thiamethoxam is expected to reveal a synergistic effect as compared to the other treatment groups.


Example 8: Growth Chamber Assays for Combined Chlorantraniliprole and Non-Intergenic Remodeled Microbes in Corn

An experiment will be conducted utilizing one or more of the deposited nine microbes described in Table 1 (6 non-intergenic remodeled microbes and 3 WT microbes), in combination with an insecticide, chlorantraniliprole. The microbe and chlorantraniliprole combination are used to treat corn seed in growth chamber experiments.


Growth chamber experiments are conducted in which corn seed is allowed to germinate under controlled standard growth conditions. The experiment includes: (a) untreated corn seed, (b) corn seed treated with chlorantraniliprole, (c) corn seed treated with one or more of the microbes described in Table 1, and (d) corn seed treated with a combination of chlorantraniliprole and one or more of the microbes described in Table 1. There will be approximately 100 seeds per treatment.


The corn seed treated with the combination of one or more of the 6 non-intergenic remodeled microbes from Table 1 and chlorantraniliprole is expected to exhibit (1) greater numbers of seed that germinate, (2) faster germination times, and (3) reaching the third leaf collar vegetative stage faster; as compared to all of the other treatment groups.


The corn seed treated with the combination of one or more of the 6 non-intergenic remodeled microbes from Table 1 and chlorantraniliprole is expected to reveal a synergistic effect as compared to the other treatment groups.


Example 9: Growth Chamber Assays for Combined Imidacloprid and Non-Intergenic Remodeled Microbes in Corn

An experiment will be conducted utilizing one or more of the deposited nine microbes described in Table 1 (6 non-intergenic remodeled microbes and 3 WT microbes), in combination with an insecticide, imidacloprid. The microbe and imidacloprid combination are used to treat corn seed in growth chamber experiments.


Growth chamber experiments are conducted in which corn seed is allowed to germinate under controlled standard growth conditions. The experiment includes: (a) untreated corn seed, (b) corn seed treated with imidacloprid, (c) corn seed treated with one or more of the microbes described in Table 1, and (d) corn seed treated with a combination of imidacloprid and one or more of the microbes described in Table 1. There will be approximately 100 seeds per treatment.


The corn seed treated with the combination of one or more of the 6 non-intergenic remodeled microbes from Table 1 and imidacloprid is expected to exhibit (1) greater numbers of seed that germinate, (2) faster germination times, and (3) reaching the third leaf collar vegetative stage faster; as compared to all of the other treatment groups.


The corn seed treated with the combination of one or more of the 6 non-intergenic remodeled microbes from Table 1 and imidacloprid is expected to reveal a synergistic effect as compared to the other treatment groups.


Example 10: Growth Chamber Assays for Combined MAXIM QUATTRO and Non-Intergenic Remodeled Microbes in Corn

An experiment will be conducted utilizing one or more of the deposited nine microbes described in Table 1 (6 non-intergenic remodeled microbes and 3 WT microbes), in combination with a fungicide, MAXIM QUATTRO (4 actives—fludioxonil+mefenoxam (also called metalaxyl)+azoxystrobin+thiabendazole). The microbe and MAXIM QUATTRO combination are used to treat corn seed in growth chamber experiments.


Growth chamber experiments are conducted in which corn seed is allowed to germinate under controlled standard growth conditions. The experiment includes: (a) untreated corn seed, (b) corn seed treated with MAXIM QUATTRO, (c) corn seed treated with one or more of the microbes described in Table 1, and (d) corn seed treated with a combination of MAXIM QUATTRO and one or more of the microbes described in Table 1. There will be approximately 100 seeds per treatment.


The corn seed treated with the combination of one or more of the 6 non-intergenic remodeled microbes from Table 1 and MAXIM QUATTRO is expected to exhibit (1) greater numbers of seed that germinate, (2) faster germination times, and (3) reaching the third leaf collar vegetative stage faster; as compared to all of the other treatment groups.


The corn seed treated with the combination of one or more of the 6 non-intergenic remodeled microbes from Table 1 and MAXIM QUATTRO is expected to reveal a synergistic effect as compared to the other treatment groups.


Example 11: Growth Chamber Assays for Combined Metalaxyl and Non-Intergenic Remodeled Microbes in Corn

An experiment will be conducted utilizing one or more of the deposited nine microbes described in Table 1 (6 non-intergenic remodeled microbes and 3 WT microbes), in combination with a fungicide, metalaxyl. The microbe and metalaxyl combination are used to treat corn seed in growth chamber experiments.


Growth chamber experiments are conducted in which corn seed is allowed to germinate under controlled standard growth conditions. The experiment includes: (a) untreated corn seed, (b) corn seed treated with metalaxyl, (c) corn seed treated with one or more of the microbes described in Table 1, and (d) corn seed treated with a combination of metalaxyl and one or more of the microbes described in Table 1. There will be approximately 100 seeds per treatment.


The corn seed treated with the combination of one or more of the 6 non-intergenic remodeled microbes from Table 1 and metalaxyl is expected to exhibit (1) greater numbers of seed that germinate, (2) faster germination times, and (3) reaching the third leaf collar vegetative stage faster; as compared to all of the other treatment groups.


The corn seed treated with the combination of one or more of the 6 non-intergenic remodeled microbes from Table 1 and metalaxyl is expected to reveal a synergistic effect as compared to the other treatment groups.


Example 12: Growth Chamber Assays for Combined Ipconazole and Non-Intergenic Remodeled Microbes in Corn

An experiment will be conducted utilizing one or more of the deposited nine microbes described in Table 1 (6 non-intergenic remodeled microbes and 3 WT microbes), in combination with a fungicide, ipconazole. The microbe and ipconazole combination are used to treat corn seed in growth chamber experiments.


Growth chamber experiments are conducted in which corn seed is allowed to germinate under controlled standard growth conditions. The experiment includes: (a) untreated corn seed, (b) corn seed treated with ipconazole, (c) corn seed treated with one or more of the microbes described in Table 1, and (d) corn seed treated with a combination of ipconazole and one or more of the microbes described in Table 1. There will be approximately 100 seeds per treatment.


The corn seed treated with the combination of one or more of the 6 non-intergenic remodeled microbes from Table 1 and ipconazole is expected to exhibit (1) greater numbers of seed that germinate, (2) faster germination times, and (3) reaching the third leaf collar vegetative stage faster; as compared to all of the other treatment groups.


The corn seed treated with the combination of one or more of the 6 non-intergenic remodeled microbes from Table 1 and ipconazole is expected to reveal a synergistic effect as compared to the other treatment groups.


Example 13: Growth Chamber Assays for Combined RAXIL PRO MD and Non-Intergenic Remodeled Microbes in Corn

An experiment will be conducted utilizing one or more of the deposited nine microbes described in Table 1 (6 non-intergenic remodeled microbes and 3 WT microbes), in combination with a fungicide, RAXIL PRO MD (3 actives—tebuconazole+prothioconazole+Metalaxyl, and ethoxylated tallow alkyl amines as surfactant). The microbe and RAXIL PRO MD combination are used to treat corn seed in growth chamber experiments.


Growth chamber experiments are conducted in which corn seed is allowed to germinate under controlled standard growth conditions. The experiment includes: (a) untreated corn seed, (b) corn seed treated with RAXIL PRO MD, (c) corn seed treated with one or more of the microbes described in Table 1, and (d) corn seed treated with a combination of RAXIL PRO MD and one or more of the microbes described in Table 1. There will be approximately 100 seeds per treatment.


The corn seed treated with the combination of one or more of the 6 non-intergenic remodeled microbes from Table 1 and RAXIL PRO MD is expected to exhibit (1) greater numbers of seed that germinate, (2) faster germination times, and (3) reaching the third leaf collar vegetative stage faster; as compared to all of the other treatment groups.


The corn seed treated with the combination of one or more of the 6 non-intergenic remodeled microbes from Table 1 and RAXIL PRO MD is expected to reveal a synergistic effect as compared to the other treatment groups.


Example 14: Methods and Assays for Detection of Non-Intergeneric Remodeled Microbes

The present disclosure teaches primers, probes, and assays that are useful for detecting the microbes utilized in the various aforementioned Examples. The assays are able to detect the non-natural nucleotide “junction” sequences in the derived/mutant non-intergeneric remodeled microbes. These non-naturally occurring nucleotide junctions can be used as a type of diagnostic that is indicative of the presence of a particular genetic alteration in a microbe.


The present techniques are able to detect these non-naturally occurring nucleotide junctions via the utilization of specialized quantitative PCR methods, including uniquely designed primers and probes. The probes can bind to the non-naturally occurring nucleotide junction sequences. That is, sequence-specific DNA probes consisting of oligonucleotides that are labelled with a fluorescent reporter, which permits detection only after hybridization of the probe with its complementary sequence can be used. The quantitative methods can ensure that only the non-naturally occurring nucleotide junction will be amplified via the taught primers, and consequently can be detected via either a non-specific dye, or via the utilization of a specific hybridization probe. Another aspect of the method is to choose primers such that the primers flank either side of a junction sequence, such that if an amplification reaction occurs, then said junction sequence is present.


Consequently, genomic DNA can be extracted from samples and used to quantify the presence of microbes of the disclosure by using qPCR. The primers utilized in the qPCR reaction can be primers designed by Primer Blast (https://www.ncbi.nlm.nih.gov/tools/primer-blast/) to amplify unique regions of the wild-type genome or unique regions of the engineered non-intergeneric mutant strains. The qPCR reaction can be carried out using the SYBR GreenER qPCR SuperMix Universal (Thermo Fisher P/N 11762100) kit, using only forward and reverse amplification primers; alternatively, the Kapa Probe Force kit (Kapa Biosystems P/N KK4301) can be used with amplification primers and a TaqMan probe containing a FAM dye label at the 5′ end, an internal ZEN quencher, and a minor groove binder and fluorescent quencher at the 3′ end (Integrated DNA Technologies).


Certain primer, probe, and non-native junction sequences—which can be used in the qPCR methods—are listed in the below Table 30. Specifically, the non-native junction sequences can be found in SEQ ID NOs: 372-405.









TABLE 30







Microbial Detection



























Junction







Junc-
up/down
SEQ
100 bp
SEQ
100 bp
SEQ
SEQ “/”

F
R



base
tion
stream
ID
upstream of
ID
downstream of
ID
indicating;
Junction
primer
primer
Probe


CI
Name
junction
NO
junction
NO
junction
NO
junction
des.
SEQ
SEQ
SEQ





1021
ds1131
up
304
TGGTGTCCGGGC
338
TTCTTGGTTCTCT
372
5′-
disrupted
N/A
N/A
N/A






GAACGTCGCCAG

GGAGCGCTTTATC

TGGTGTCCGGGC
nifL gene/









GTGGCACAAATT

GGCATCCTGACTG

GAACGTCGCCAG
PinfC









GTCAGAACTACG

AAGAATTIGCAGG

GTGGCACAAATT










ACACGACTAACC

CTTCTTCCCAACC

GTCAGAACTACG










GACCGCAGGAGP

TGGCTTGCACCCG

ACACGACTAACC










GTGCGATGACCC

TGCAGGTAGTTGT

GACCGCAGGAGT










TGAATATGATGA

GATGAACAT

GTGCGATGACCC










TGGA



TGAATATGATGA














TGGA/














TTCTTGGTTCTC














TGGAGCGCTTTA














TCGGCATCCTGA














CTGAAGAATTTG














CAGGCTTCTTCC














CAACCTGGCTTG














CACCCGTGCAGG














TAGTTGTGATGA














ACAT-3′









1021
ds1131
down
305
CGGAAAACGAGT
339
GCGATAGAACTCA
373
5′-
PinfC/
N/A
N/A
N/A






TCAAACGGCGCG

CTTCACGCCCCGA

CGGAAAACGAGT
disrupted









TCCCAATCGTAT

AGGGGGAAGCTGC

TCAAACGGCGCG
nifL gene









TAATGGCGAGAT

CTGACCCTACGAT

TCCCAATCGTAT










TCGCGCCACGGA

TCCCGCTATTTCA

TAATGGCGAGAT










AGTTCGCTTAAC

TTCACTGACCGGA

TCGCGCCACGGA










AGGTCTGGAAGG

GGTTCAAAATGAC

AGTTCGCTTAAC










CGAGCAGCTTGG

CCAGCGAAC

AGGTCTGGAAGG










TATT



CGAGCAGCTTGG














TATT/














GCGATAGAACTC














ACTTCACGCCCC














GAAGGGGGAAGC














TGCCTGACCCTA














CGATTCCCGCTA














TTTCATTCACTG














ACCGGAGGTTCA














AAATGACCCAGC














GAAC-3′









1021
ds1133
N/A
306
CGCCAGAGAGTT
340
TCCCTGTGCGCCG
374
5′-
5′ UTR
N/A
N/A
N/A






GAAATCGAACAT

CGTCGCCGATGGT

CGCCAGAGAGTT
and ATG/









TTCCGTAATACC

GGCCAGCCAACTG

GAAATCGAACAT
truncated









GCCATTACCCAG

GCGCGCTACCCGA

TTCCGTAATACC
glnE gene









GAGCCGTTCTGG

TCCTGCTCGATGA

GCCATTACCCAG










TTGCACAGCGGA

ACTGCTCGACCCG

GAGCCGTTCTGG










AAACGTTAACGA

AACACGCTCTATC

TTGCACAGCGGA










AAGGATATTTCG

AACCGACGG

AAACGTTAACGA










CATG



AAGGATATTTCG














CATG/














TCCCTGTGCGCC














GCGTCGCCGATG














GTGGCCAGCCAA














CTGGCGCGCTAC














CCGATCCTGCTC














GATGAACTGCTC














GACCCGAACACG














CTCTATCAACCG














ACGG-3′









1021
ds1145
up
307
CGGGCGAACGTC
341
CGTTCTGTAATAA
375
5′-
disrupted
N/A
N/A
N/A






GCCAGGTGGCAC

TAACCGGACAATT

CGGGCGAACGTC
nifL gene/









AAATTGTCAGAA

CGGACTGATTAAA

GCCAGGFGGCAC
Prml









CTACGACACGAC

AAAGCGCCCTCGC

AAATTGTCAGAA










TAACCGACCGCA

GGCGCTTTTTTTA

CTACGACACGAC










GGAGTGTGCGAT

TATTCTCGACTCC

TAACCGACCGCA










GACCCTGAATAT

ATTTAAAATAAAA

GGAGTGTGCGAT










GATGATGGATGC

AATCCAATC

GACCCTGAATAT










CAGC



GATGATGGATGC














CAGC/














CGTTCTGTAATA














ATAACCGGACAA














TTCGGACTGATT














AAAAAAGCGCCC














TCGCGGCGCTTT














TTTTATATTCTC














GACTCCATTTAA














AATAAAAAATCC














AATC-3′









1021
ds1145
down
308
TCAACCTAAAAA
342
AACTCACTTCACG
376
5′-
Prml/
N/A
N/A
N/A






AGTTTGTGTAAT

CCCCGAAGGGGGA

TCAACCTAAAAA
disrupted









ACTTGTAACGCT

AGCTGCCTGACCC

AGTTTGTGTAAT
nifL gene









ACATGGAGATTA

TACGATTCCCGCT

ACTTGTAACGCT










ACTCAATCTAGA

ATTTCATTCACTG

ACATGGAGATTA










GGGTATTAATAA

ACCGGAGGTTCAA

ACTCAATCTAGA










TGAATCGTACTA

AATGACCCAGCGA

GGGTATTAATAA










AACTGGTACTGG

ACCGAGTCG

TGAATCGTACTA










GCGC



AACTGGTACTGG














GCGC/














AACTCACTTCAC














GCCCCGAAGGGG














GAAGCTGCCTGA














CCCTACGATTCC














CGCTATTTCATT














CACTGACCGGAG














GTTCAAAATGAC














CCAGCGAACCGA














GTCG-3′









1021
ds1148
up
309
CGGGCGAACGTC
343
CGCGTCAGGTTGA
377
5′-
disrupted
N/A
N/A
N/A






GCCAGGTGGCAC

ACGTAAAAAAGTC

CGGGCGAACGTC
nifL gene/









AAATTGTCAGAA

GGTCTGCGCAAAG

GCCAGGTGGCAC
Prm7









CTACGACACGAC

CACGTCGTCGTCC

AAATTGTCAGAA










TAACCGACCGCA

GCAGTTCTCCAAA

CTACGACACGAC










GGAGTGTGCGAT

CGTTAATTGGTTT

TAACCGACCGCA










GACCCTGAATAT

CTGCTTCGGCAGA

GGAGTGTGCGAT










GATGATGGATGC

ACGATTGGC

GACCCTGAATAT










CAGC



GATGATGGATGC














CAGC/














CGCGTCAGGTTG














AACGTAAAAAAG














TCGGTCTGCGCA














AAGCACGTCGTC














GTCCGCAGTTCT














CCAAACGTTAAT














TGGTTTCTGCTT














CGGCAGAACGAT














TGGC-3′









1021
ds1148
down
310
AATTTTCTGCCC
344
AACTCACTTCACG
378
5′-
Prm4/
N/A
N/A
N/A






AAATGGCTGGGA

CCCCGAAGGGGGA

AATTTTCTGCCC
disrupted









TTGTTCATTTTT

AGCTGCCTGACCC

AAATGGCTGGGA
nifL gene









TGTTTGCCTTAC

TACGATTCCCGCT

TTGTTCATTTTT










AACGAGAGTGAC

ATTTCATTCACTG

TGTTTGCCTTAC










AGTACGCGCGGG

ACCGGAGGTTCAA

AACGAGAGTGAC










TAGTTAACTCAA

AATGACCCAGCGA

AGTACGCGCGGG










CATCTGACCGGT

ACCGAGTCG

TAGTTAACTCAA










CGAT



CATCTGACCGGT














CGAT/














AACTCACTTCAC














GCCCCGAAGGGG














GAAGCTGCCTGA














CCCTACGATTCC














CGCTATTTCATT














CACTGACCGGAG














GTTCAAAATGAC














CCAGCGAACCGA














GTCG-3′









CI006
ds126
N/A
311
GTAACCAATAAA
345
CCGATCCCCATC
379
5′-
5′ UTR up
N/A
N/A
N/A






GGCCACCACGCC

ACTGTGTGTCTT

GTAACCAATAAA
to ATG-









AGACCACACGAT

GTATTACAGTGC

GGCCACCACGCC
4 bp of









AGTGATGGCAAC

CGCTTCGTCGGC

AGACCACACGAT
amtB gene/









ACTTTCCAGCTG

TTCGCCGGTACG

AGTGATGGCAAC
disrupted









CACCAGCACCTG

AATACGAATGAC

ACTTTCCAGCTG
amtB gene









ATGGCCCATGGT

GCGTTGCAGCTC

CACCAGCACCTG










CACACCTTCAGC

AGCAACGAAAAT

ATGGCCCATGGT










GAAA

TTTG

CACACCTTCAGC














GAAA/














CCGATCCCCATC














ACTGTGTGTCTT














GTATTACAGTGC














CGCTTCGTCGGC














TTCGCCGGTACG














AATACGAATGAC














GCGTTGCAGCTC














AGCAACGAAAAT














TTTG-3′









CI019
ds172
down
312
TGGTATTGTCAG
346
CCGTCTCTGAAG
380
5′-
Prm1.2/
SEQ
SEQ
N/A






TCTGAATGAAGC

CTCTCGGTGAAC

TGGTATTGTCAG
disrupted 
ID
ID







TCTTGAAAAAGC

ATTGTTGCGAGG

TCTGAATGAAGC
nifLgene
NO:
NO:







TGAGGAAGCGGG

CAGGATGCGAGC

TCTTGAAAAAGC

406
407







CGTCGATTTAGT

TGGTTGTGTTTT

TGAGGAAGCGGG

CAAG
TGCC







AGAAATCAGTCC

GACATTACCGAT

CGTCGATTTAGT

AAGT
TCGC







GAATGCCGAGCC

AATGTGCCGCGT

AGAAATCAGTCC

TCGC
AACA







GCCAGTTTGTCG

GAACGGGTGCGT

GAATGCCGAGCC

CTCA
ATGT







AATC

TATG

GCCAGTTTGTCG

CAGG
TCAC











AATC/














CCGTCTCTGAAG














CTCTCGGTGAAC














ATTGTTGCGAGG














CAGGATGCGAGC














TGGTTGTGTTTT














GACATTACCGAT














AATGTGCCGCGT














GAACGGGTGCGT














TATG-3′









CI019
ds172
up
313
ACCGATCCGCAG
347
TGAACATCACTG
381
5′-
disrupted
N/A
N/A
N/A






GCGCGCATTTGT

ATGCACAAGCTA

ACCGATCCGCAG
nifL gene/









TATGCCAATCCG

CCTATGTCGAAG

GCGCGCATTTGT
PRM1.2









GCATTCTGCCGC

AATTAACTAAAA

TATGCCAATCCG










CAGACGGGTTTT

AACTGCAAGATG

GCATTCTGCCGC










GCACTTGAGACA

CAGGCATTCGCG

CAGACGGGTTTT










CTTTTGGGCGAG

TTAAAGCCGACT

GCACTTGAGACA










AACCACCGTCTG

TGAGAAATGAGA

CTTTTGGGCGAG










CTGG

AGAT

AACCACCGTCTG














CTGG/














TGAACATCACTG














ATGCACAAGCTA














CCTATGTCGAAG














AATTAACTAAAA














AACTGCAAGATG














CAGGCATTCGCG














TTAAAGCCGACT














TGAGAAATGAGA














AGAT-3′









CI019
ds175
down
314
CGGGAACCGGTG
348
CCGTCTCTGAAG
382
5′-
Prm3.1/
SEQ
SEQ
SEQ






TTATAATGCCGC

CTCTCGGTGAAC

CGGGAACCGGTG
disrupted
ID
ID
ID






GCCCTCATATTG

ATTGTTGCGAGG

TTATAATGCCGC
nifL gene
NO:
NO:
NO:






TGGGGATTTCTT

CAGGATGCGAGC

GCCCTCATATTG

408
409
410/






AATGACCTATCC

TGGTTGTGTTTT

TGGGGATTTCTT

CGCC
GGCA
56-






TGGGTCCTAAAG

GACATTACCGAT

AATGACCTATCC

CTCA
TAAC
FAM/






TTGTAGTTGACA

AATGTGCCGCGT

TGGGTCCTAAAG

TATT
GCAC
TAAC






TTAGCGGAGCAC

GAACGGGTGCGT

TTGTAGTTGACA

GTGG
CCGT
CCGT






TAAC

TATG

TTAGCGGAGCAC

GGAT
TCA
C/ZE










TAAC/



N/TC










CCGTCTCTGAAG



TGAA










CTCTCGGTGAAC



GCTC










ATTGTTGCGAGG



TCGG










CAGGATGCGAGC



T/3I










TGGTTGTGTTTT



ABkF










GACATTACCGAT



Q/










AATGTGCCGCGT














GAACGGGTGCGT














TATG-3′









CI019
ds175
up
315
ACCGATCCGCAG
349
TACAGTAGCGCC
383
5′-
disrupted
N/A
N/A
N/A






GCGCGCATTTGT

TCTCAAAAATAG

ACCGATCCGCAG
nifL gene/









TATGCCAATCCG

ATAAACGGCTCA

GCGCGCATTTGT
Prm3.1









GCATTCTGCCGC

TGTACGTGGGCC

TATGCCAATCCG










CAGACGGGTTTT

GTTTATTTTTTC

GCATTCTGCCGC










GCACTTGAGACA

TACCCATAATCG

CAGACGGGTTTT










CTTTTGGGCGAG

GGAACCGGTGTT

GCACTTGAGACA










AACCACCGTCTG

ATAATGCCGCGC

CTTTTGGGCGAG










CTGG

CCTC

AACCACCGTCTG














CTGG/














TACAGTAGCGCC














TCTCAAAAATAG














ATAAACGGCTCA














TGTACGTGGGCC














GTTTATTTTTTC














TACCCATAATCG














GGAACCGGTGTT














ATAATGCCGCGC














CCTC-3′









CI006
ds20
down
316
TCAACCTAAAAA
350
AACTCACTTCAC
384
5′-
PRM1/
SEQ
SEQ
SEQ






AGTTTGTGTAAT

ACCCCGAAGGGG

TCAACCTAAAAA
disrupted
ID
ID
ID






ACTTGTAACGCT

GAAGTTGCCTGA

AGTTTGTGTAAT
nifL gene
NO:
NO:
NO:






ACATGGAGATTA

CCCTACCATTCC

ACTTGTAACGCT

411
412
413/






ACTCAATCTAGA

CGCTATTTCATT

ACATGGAGATTA

TAAA
CAAA
56-






GGGTATTAATAA

CACTGACCGGAG

ACTCAATCTAGA

CTGG
TCGA
FAM/






TGAATCGFACTA

GTTCAAAATGAC

GGGTATTAATAA

TACT
AGCG
AAGT






AACTGGTACTGG

CCAGCGAACCGA

TGAATCGTACTA

GGGC
CCAG
TGCC






GCGC

GTCG

AACTGGTACTGG

GCAA
ACGG
T/ZE










GCGC/

CT
TAT
N/GA










AACTCACTTCAC



CCCT










ACCCCGAAGGGG



ACGA










GAAGTTGCCTGA



TTCC










CCCTACGATTCC



C/3I










CGCTATTTCATT



ABkF










CACTGACCGGAG



Q/










GTTCAAAATGAC














CCAGCGAACCGA














GTCG-3′









CI006
ds20
up
317
GGGCGACAAACG
351
CGTCCTGTAATA
385
5′-
disrupted
N/A
N/A
N/A






GCCTGGTGGCAC

ATAACCGGACAA

GGGCGACAAACG
nifL gene/









AAATTGTCAGAA

TTCGGACTGATT

GCCTGGTGGCAC
Prm1









CTACGACACGAC

AAAAAAGCGCCC

AAATTGTCAGAA










TAACTGACCGCA

TTGTGGCGCTTT

CTACGACACGAC










GGAGTGTGCGAT

TTTTATAYFCCC

TAACTGACCGCA










GACCCTGAATAT

GCCTCCATTTAA

GGAGTGTGCGAT










GATGTGGATGCC

AATAAAAAATCC

GACCCTGAATAT










GGC

AATC

GATGATGGATGC














CGGC/














CGTCCTGTAATA














ATAACCGGACAA














TTCGGACTGATT














AAAAAAGCGCCC














TTGTGGCGCTTT














TTTTATATTCCC














GCCTCCATTTAA














AATAAAAAATCC














AATC-3′









CI006
ds24
up
318
GGGCGACAAACG
352
GGACATCATCGC
386
5′-
disrupted
SEQ
SEQ
SEQ






GCCTGGTGGCAC

GACAAACAATAT

GGGCGACAAACG
nigL gene/
ID
ID
ID






AAATTGTCAGAA

TAATACCGGCAA

GCCTGGTGGCAC
Prm5
NO:
NO:
NO:






CTACGACACGAC

CCACACCGGCAA

AAATTGTCAGAA

414
415
416/






TAACTGACCGCA

TTTACGAGACTG

CTACGACACGAC

GGTG
GCGC
56-






GGAGTGTGCGAT

CGCAGGCATCCT

TAACTGACCGCA

CACT
AGTC
FAM/






GACCCTGAATAT

TTCTCCCGTCAA

GGAGTGTGCGAT

CTTT
TCGT
CAGG






GATGATGGATGC

TTTCTGTCAAAT

GACCCTGAATAT

GCAT
AAAT
AGTG






CGGC

AAAG

GATGATGGATGC

GGTT
TGCC
T/ZE










CGGC/



N/GC










GGACATCATCGC



GATG










GACAAACAATAT



ACCC










TAATACCGGCAA



TGAA










CCACACCGGCAA



T/3I










TTTACGAGACTG



ABkF










CGCAGGCATCCT



Q










TTCTCCCGTCAA














TTTCTGTCAAAT














AAAG-3′









CI006
ds24
down
319
TAAGAATFATCT
353
AACTCACTTCAC
387
5′-
Prm5/
N/A
N/A
N/A






GGATGAATGTGC

ACCCCGAAGGGG

TAAGAATTATCT
disrupted









CATTAAATGCGC

GAAGTTGCCTGA

GGATGAATGTGC
nifL gene









AGCATAATGGTG

CCCTACGATTCC

CATTAAATGCGC










CGTTGTGCGGGA

CGCTATTTCATT

AGCATAATGGTG










AAACTGCTTTTT

CACTGACCGGAG

CGTTGTGCGGGA










TTTGAAAGGGTT

GTTCAAAATGAC

AAACTGCTTTTT










GGTCAGTAGCGG

CCAGCGAACCGA

TTTGAAAGGGTT










AAAC

GTCG

GGTCAGTAGCGG














AAAC/














AACTCACTTCAC














ACCCCGAAGGGG














GAAGTTGCCTGA














CCCTACGATTCC














CGCTATTTCATT














CACTGACCGGAG














GTTCAAAATGAC














CCAGCGAACCGA














GTCG-3′









CI006
ds30
N/A
320
CGCCAGAGAGTC
354
TTTAACGATCTG
388
5′-
5′ UTR
N/A
N/A
N/A






GAAATCGAACAT

ATTGGCGATGAT

CGCCAGAGAGTC
and ATG/









TTCCGTAATACC

GAAACGGATTCG

GAAATCGAACAT
truncated









GCGATTACCCAG

CCGGAAGATGCG

TTCCGTAATACC
glnE gene









GAGCCGTTCTGG

CTTTCTGAGAGC

GCGATTACCCAG










TTGCACAGCGGA

TGGCGCGAATTG

GAGCCGTTCTGG










AAACGTTAACGA

TGGCAGGATGCG

TTGCACAGCGGA










AAGGATATTTCG

TTGCAGGAGGAG

AAACGTTAACGA










CATG

GATT

AAGGATATTTCG














CATG/














TTTAACGATCTG














ATTGGCGATGAT














GAAACGGATTCG














CCGGAAGATGCG














CTTTCTGAGAGC














TGGCGCGAATTG














TGGCAGGATGCG














TTGCAGGAGGAG














GATT-3′









CI006
ds31
N/A
321
CGCCAGAGAGTC
355
GCACTGAAACAC
389
5′-
5′ UTR
N/A
N/A
N/A






GAAATCGAACAT

CTCATTTCCCTG

CGCCAGAGAGTC
and ATG/









TTCCGTAATACC

TGTGCCGCGTCG

GAAATCGAACAT
truncated









GCGATTACCCAG

CCGATGGTTGCC

TTCCGTAATACC
glnE gene









GAGCCGTTCTGG

AGTCAGCTGGCG

GCGATTACCCAG










TTGCACAGCGGA

CGCTACCCGATC

GAGCCGTTCTGG










AAACGTTAACGA

CTGCTTGATGAA

TTGCACAGCGGA










AAGGATATTTCG

TTGCTCGACCCG

AAACGTTAACGA










CATG

AATA

AAGGATATTTCG














CATG/














GCACTGAAACAC














CTCATTTCCCTG














TGTGCCGCGTCG














CCGATGGTTGCC














AGTCAGCTGGCG














CGCTACCCGATC














CTGCTTGATGAA














TTGCTCGACCCG














AATA-3′









CI019
ds34
N/A
322
GATGATGGATGC
356
GCGCTCAAACAG
390
5′-
5′ UTR
N/A
N/A
N/A






TTTCTGGTTAAA

TTAATCCGTCTG

GATGATGGATGC
and ATG/









CGGGCAACCTCG

TGTGCCGCCTCG

TTTCTGGTTAAA
truncated









TTAACTGACTGA

CCGATGGTCGCG

CGGGCAACCTCG
glnE gene









CTAGCCTGGGCA

ACACAACTTGCA

TTAACTGACTGA










AACTGCCCGGGC

CGTCATCCTTTA

CTAGCCTGGGCA










TTTTTTTTGCAA

TTGCTCGATGAA

AACTGCCCGGGC










GGAATCTGATTT

CTGCTCGACCCG

TTTTTTTTGCAA










CATG

CGCA

GGAATCTGATTT














CATG/














GCGCTCAAACAG














TTAATCCGTCTG














TGTGCCGCCTCG














CCGATGGTCGCG














ACACAACTTGCA














CGTCATCCTTTA














TTGCTCGATGAA














CTGCTCGACCCG














CGCA-3′









CI019
ds70
up
323
ACCGATCCGCAG
357
AGTCTGAACTCA
391
5′-
disrupted
N/A
N/A
N/A






GCGCGCATTTGT

TCCTGCGGCAGT

ACCGATCCGCAG
nifL gene/









TATGCCAATCCG

CGGTGAGACGTA

GCGCGCATTTGT
Prm4









GCATTCTGCCGC

TTTTTGACCAAA

TATGCCAATCCG










CAGACGGGTTTT

GAGTGATCTACA

GCATTCTGCCGC










GCACTTGAGACA

TCACGGAATTTT

CAGACGGGTTTT










CTTTTGGGCGAG

GTGGTTGTTGCT

GCACTTGAGACA










AACCACCGTCTG

GCTTAAAAGGGC

CTTTTGGGCGAG










CTGG

AAAT

AACCACCGTCTG














CTGG/














AGTCTGAACTCA














TCCTGCGGCAGT














CGGTGAGACGTA














TTTTTGACCAAA














GAGTGATCTACA














TCACGGAATTTT














GTGGTTGTTGCT














GCTTAAAAGGGC














AAAT-3′









CI019
ds70
down
324
CATCGGACACCA
358
CCGTCTCTGAAG
392
5′-
Prm4/
N/A
N/A
N/A






CCAGCTTACAAA

CTCTCGGTGAAC

CATCGGACACCA
disrupted









TTGCCTGATTGC

ATTGTTGCGAGG

CCAGCTTACAAA
nifL gene









GGCCCCGATGGC

CAGGATGCGAGC

TTGCCTGATTGC










CGGTATCACTGA

TGGTTGTGTTTT

GGCCCCGATGGC










CCGACCATTTCG

GACATTACCGAT

CGGTATCACTGA










TGCCTTATGTCA

AATGTGCCGCGT

CCGACCATTTCG










TGCGATGGGGGC

GAACGGGTGCGT

TGCCTTATGTCA










TGGG

TATG

TGCGATGGGGGC














TGGG/














CCGTCTCTGAAG














CTCTCGGTGAAC














ATTGTTGCGAGG














CAGGATGCGAGC














TGGTTGTGTTTT














GACATTACCGAT














AATGTGCCGCGT














GAACGGGTGCGT














TATG-3′









 137
ds799
down
325
TCTTCAACAACT
359
GCCATTGAGCTG
393
5′-
PinfC/
SEQ.
SEQ
SEQ






GGAGGAATAAGG

GCTTCCCGACCG

TCTTCAACAACT
disrupted
ID
ID
ID






TATTAAAGGCGG

CAGGGCGGCACC

GGAGGAATAAGG
nifL gene
NO:
NO:
NO:






AAAACGAGTTCA

TGCCTGACCCTG

TATTAAAGGCGG

417
418
419/






AACGGCACGTCC

CGTTTCCCGCTG

AAAACGAGTTCA

CTCG
AGGG
56-






GAATCGTATCAA

TTTAACACCCTG

AACGGCACGTCC

GCAG
TGTT
FAM/






TGGCGAGATTCG

ACCGGAGGTGAA

GAATCGTATCAA

CATG
AAAC
AACG






CGCCCTGGAAGT

GCATGATCCCTG

TGGCGAGATTCG

GACG
AGCG
GCAC






TCGC

AATC

CGCCCTGGAAGT

TAA
GGAA
G/ZE










TCGC/


A
N/TC










GCCATTGAGCTG



CGAA










GCTTCCCGACCG



TCGT










CAGGGCGGCACC



ATCA










TGCCTGACCCTG



A/3I










CGTTTCCCGCTG



ABkF










TTTAACACCCTG



Q/










ACCGGAGGTGAA














GCATGATCCCTG














AATC-3′









 137
ds799
up
326
TCCGGGTTCGGC
360
AGCGTCAGGTAC
394
5′-
disrupted
N/A
N/A
N/A






TTACCCCGCCGC

CGGTCATGATTC

TCCGGGTTCGGC
nifL gene/









GTTTTGCGCACG

ACCGTGCGATTC

TTACCCCGCCGC
PinfC









GTGTCGGACAAT

TCGGTTCCCTGG

GTTTTGCGCACG










TTGTCATAACTG

AGCGCTTCATTG

GTGTCGGACAAT










CGACACAGGAGT

GCATCCTGACCG

TTGTCATAACTG










TTGCGATGACCC

AAGAGTTCGCTG

CGACACAGGAGT










TGAATATGATGC

GCTTCTTCCCAA

TTGCGATGACCC










TCGA

CCTG

TGAATATGATGC














TCGA/














AGCGTCAGGTAC














CGGTCATGATTC














ACCGTGCGATTC














TCGGTTCCCTGG














AGCGCTTCATTG














GCATCCTGACCG














AAGAGTTCGCTG














GCTTCTTCCCAA














CCTG-3′









 137
ds809
N/A
327
ATCGCAGCGTCT
361
GCGCTGAAGCAC
395
5′-
5′ UTR
SEQ
SEQ
SEQ






TTGAATATTTCC

CTGATCACGCTC

ATCGCAGCGTCT
and ATG/
ID
ID
ID






GTCGCCAGGCGC

TGCGCGGCGTCG

TTGAATATTTCC
truncated
NO:
NO:
NO:






TGGCTGCCGAGC

CCGATGGTCGCC

GTCGCCAGGCGC
glnE gene
420
421
422/






CGTTCTGGCTGC

AGCCAGCTGGCG

TGGCTGCCGAGC

GAGC
GCCG
56-






ATAGTGGAAAAC

CGCCACCCGCTG

CGTTCTGGCTGC

CGTT
TCGG
FAM/






GATAATTTCAGG

CTGCTGGATGAG

ATAGTGGAAAAC

CTGG
CTGA
TTAT






CCAGGGAGCCCT

CTGCTGGATCCC

GATAATTTCAGG

CTGC
TAGA
GGCG






TATG

AACA

CCAGGGAGCCCT

ATAG
GG
C/ZE










TATG/



N/TG










GCGCTGAAGCAC



AAGC










CTGATCACGCTC



ACCT










TGCGCGGCGTCG



GATC










CCGATGGTCGCC



A/3I










AGCCAGCTGGCG



ABkF










CGCCACCCGCTG



Q/










CTGCTGGATGAG














CTGCTGGATCCC














AACA-3′









 137
ds843
up
328
TCCGGGTTCGGC
362
GCCCGCTGACCG
396
5′-
disrupted
N/A
N/A
N/A






TTACCCCGCCGC

ACCAGAACTTCC

TCCGGGTTCGGC
nifL gene/









GTTTTGCGCACG

ACCTTGGACTCG

TTACCCCGCCGC
Prm1.2









GTGTCGGACAAT

GCTATACCCTTG

GTTTTGCGCACG










TTGTCATAACTG

GCGTGACGGCGC

GTGTCGGACAAT










CGACACAGGAGT

GCGATAACTGGG

TTGTCATAACTG










TTGCGATGACCC

ACTACATCCCCA

CGACACAGGAGT










TGAATATGATGC

TTCCGGTGATCT

TTGCGATGACCC










TCGA

TACC

TGAATATGATGC














TCGA/














GCCCGCTGACCG














ACCAGAACTTCC














ACCTTGGACTCG














GCTATACCCTTG














GCGTGACGGCGC














GCGATAACTGGG














ACTACATCCCCA














TTCCGGTGATCT














TACC-3′









 137
ds843
down
329
TCACTTTTTAGC
363
GCCATTGAGCTG
397
5′-
Prm1.2/
N/A
N/A
N/A






AAAGTTGCACTG

GCTTCCCGACCG

TCACTTTTTAGC
disrupted









GACAAAAGGTAC

CAGGGCGGCACC

AAAGTTGCACTG
nifL gene









CACAATTGGTGT

TGCCTGACCCTG

GACAAAAGGTAC










ACTGATACTCGA

CGTTTCCCGCTG

CACAATTGGTGT










CACAGCATTAGT

TTTAACACCCTG

ACTGATACTCGA










GTCGATTTTTCA

ACCGGAGGTGAA

CACAGCATTAGT










TATAAAGGTAAT

GCATGATCCCTG

GTCGATTTTTCA










TTTG

AATC

TATAAAGGTAAT














TTTG/














GCCATTGAGCTG














GcrrcCCGACCG














CAGGGCGGCACC














TGCCTGACCCTG














CGTTTCCCGCTG














TTTAACACCCTG














ACCGGAGGTGAA














GCATGATCCCTG














AATC-3′









 137
ds853
up
330
TCCGGGTTCGGC
364
GCTAAAGTTCTC
398
5′-
disrupted
N/A
N/A
N/A






TTACCCCGCCGC

GGCTAATCGCTG

TCCGGGTTCGGC
nifL gene/









GTTTTGCGCACG

ATAACATTTGAC

TTACCCCGCCGC
Prm6.2









GTGTCGGACAAT

GCAATGCGCAAT

GTTTTGCGCACG










TTGTCATAACTG

AAAAGGGCATCA

GTGTCGGACAAT










CGACACAGGAGT

TTTGATGCCCTT

TTGTCATAACTG










TTGCGATGACCC

TTTGCACGCTTT

CGACACAGGAGT










TGAATATGATGC

CATACCAGAACC

TTGCGATGACCC










TCGA

TGGC

TGAATATGATGC














TCGA/














GCTAAAGTTCTC














GGCTAATCGCTG














ATAACATTTGAC














GCAATGCGCAAT














AAAAGGGCATCA














TTTGATGCCCTT














TTTGCACGCTTT














CATACCAGAACC














TGGC-3′









 137
ds853
down
331
GTTCTCCTTTGC
365
GCCATTGAGCTG
399
5′-
Prm6.2/
N/A
N/A
N/A






AATAGCAGGGAA

GCTTCCCGACCG

GTTCTCCTTTGC
disrupted









GAGGCGCCAGAA

CAGGGCGGCACC

AATAGCAGGGAA
nifL gene









CCGCCAGCGTTG

TGCCTGACCCTG

GAGGCGCCAGAA










AAGCAGTTTGAA

CGTTTCCCGCTG

CCGCCAGCGTTG










CGCGTTCAGTGT

TTTAACACCCTG

AAGCAGTTTGAA










ATAATCCGAAAC

ACCGGAGGTGAA

CGCGTTCAGTGT










TTAATTTCGGTT

GCATGATCCCTG

ATAATCCGAAAC










TGGA

AATC

TTAATTTCGGTT














TGGA/














GCCATTGAGCTG














GCTTCCCGACCG














CAGGGCGGCACC














TGCCTGACCCTG














CGTTTCCCGCTG














TTTAACACCCTG














ACCGGAGGTGAA














GCATGATCCCTG














AATC-3′









 137
ds857
up
332
TCCGGGTTCGGC
366
CGCCGTCCTCGC
400
5′-
disrupted
N/A
N/A
N/A






TTACCCCGCCGC

AGTACCATTGCA

TCCGGGTTCGGC
nifL gene/









GTTTTGCGCACG

ACCGACTTTACA

TTACCCCGCCGC
Prm8.2









GTGTCGGACAAT

GCAAGAAGTGAT

GTTTTGCGCACG










TTGTCATAACTG

TCTGGCACGCAT

GTGTCGGACAAT










CGACACAGGAGT

GGAACAAATTCT

TTGTCATAACTG










TTGCGATGACCC

TGCCAGTCGGGC

CGACACAGGAGT










TGAATATGATGC

TTTATCCGATGA

TTGCGATGACCC










TCGA

CGAA

TGAATATGATGC














TCGA/














CGCCGTCCTCGC














AGTACCATTGCA














ACCGACTTTACA














GCAAGAAGTGAT














TCTGGCACGCAT














GGAACAAATTCT














TGCCAGTCGGGC














TTTATCCGATGA














CGAA-3′









137
ds857
down
333
GATATGCCTGAA
367
GCCATTGAGCTG
401
5′-
Prm8.2/
N/A
N/A
N/A






GTATTCAATTAC

GCTTCCCGACCG

GATATGCCTGAA
disrupted









TTAGGCATTTAC

CAGGGCGGCACC

GTATTCAATTAC
nifL gene









TTAACGCAGGCA

TGCCTGACCCTG

TTAGGCATTTAC










GGCAATTTTGAT

CGTTTCCCGCTG

TTAACGCAGGCA










GCTGCCTATGAA

TTTAACACCCTG

GGCAATTTTGAT










GCGTTTGATTCT

ACCGGAGGTGAA

GCTGCCTATGAA










GTACTTGAGCTT

GCATGATCCCTG

GCGTTTGATTCT










GATC

AATC

GTACTTGAGCTT














GATC/














GCCATTGAGCTG














GCTTCCCGACCG














CAGGGCGGCACC














TGCCTGACCCTG














CGTTTCCCGCTG














TTTAACACCCTG














ACCGGAGGTGAA














GCATGATCCCTG














AATC-3′









  63
ds908
down
334
TGGTATTGTCAG
368
TCTTTAGATCTC
402
5′-
PinfC/
SEQ
SEQ
N/A






TCTGAATGAAGC

TCGGTCCGCCCT

TGGTATTGTCAG
disrupted
ID
ID







TCTTGAAAAAGC

GATGGCGGCACC

TCTGAATGAAGC
nifL gene
NO:
NO:







TGAGGAAGCGGG

TTGCTGACGTTA

TCTTGAAAAAGC

423
424







CGTCGATTTAGT

CCGGTGCCGGTA

TGAGGAAGCGGG

GGAA
GGGC







AGAAATCAGTCC

CAGCAGGTTATC

CGTCGATTTAGT

AACG
GGAC







GAATGCCGAGCC

ACCGGAGGCTTA

AGAAATCAGTCC

AGTT
CGAG







GCCAGTTTGTCG

AAATGACCCAGT

GAATGCCGAGCC

CAAC
AGAT







AATC

TACC

GCCAGTTTGTCG

CGGC
CTAA











AATC/














TCTTTAGATCTC














TCGGTCCGCCCT














GATGGCGGCACC














TTGCTGACGTTA














CGCCTGCCGGTA














CAGCAGGTTATC














ACCGGAGGCTTA














AAATGACCCAGT














TACC-3′









  63
ds908
up
335
TGCAAATTGCAC
369
TGAATATCACTG
403
5′-
disrupted
N/A
N/A
N/A






GGTTATTCCGGG

ACTCACAAGCTA

TGCAAATTGCAC
nifL gene/









TGAGTATATGTG

CCTATGFCGAAG

GGTTATTCCGGG
PinfC









TGATTTGGGTTC

AATTAACTAAAA

TGAGTATATGTG










CGGCATTGCGCA

AACTGCAAGATG

TGATTTGGGTTC










ATAAAGGGGAGA

CAGGCATTCGCG

CGGCATTGCGCA










AAGACATGAGCA

TTAAAGCCGACT

ATAAAGGGGAGA










TCACGGCGTTAT

TGAGAAATGAGA

AAGACATGAGCA










CAGC

AGAT

TCACGGCGTTAT














CAGC/














TGAATATCACTG














ACTCACAAGCTA














CCTATGTCGAAG














AATTAACTAAAA














AACTGCAAGATG














CAGGCATTCGCG














TTAAAGCCGACT














TGAGAAATGAGA














AGAT-3′









 910
ds960
up
336
TCAGGGCTGCGG
370
CTGGGGTCACTG
404
5′-
disrupted
N/A
N/A
N/A






ATGTCGGGCGTT

GAGCGCTTTATC

TCAGGGCTGCGG
nifL gene/









TCACAACACAAA

GGCATCCTGACC

ATGTCGGGCGTT
PinfC









ATGTTGTAAATG

GAAGAATTTGCC

TCACAACACAAA










CGACACAGCCGG

GGTTTCTTCCCG

ATGTTGTAAATG










GCCTGAAACCAG

ACCTGGCTGGCC

CGACACAGCCGG










GAGCGTGTGATG

CCTGTTCAGGTT

GCCTGAAACCAG










ACCTTTAATATG

GTGGTGATGAAT

GAGCGTGTGATG










ATGC

ATCA

ACCTTTAATATG














ATGC/














CTGGGGTCACTG














GAGCGCTTTATC














GGCATCCTGACC














GAAGAATTTGCC














GGTTTCTTCCCG














ACCTGGCTGGCC














CCTGTTCAGGTT














GTGGTGATGAAT














ATCA-3′









 910
ds960
down
337
CGGAAAACGAGT
371
GCAATAGAACTA
405
5′-
PinfC/
N/A
N/A
N/A






TCAAACGGCACG

ACTACCCGCCCT

CGGAAAACGAGT
disrupted









TCCGAATCGTAT

GAAGGCGGTACC

TCAAACGGCACG
nifL gene









CAATGGCGAGAT

TGCCTGACCCTG

TCCGAATCGTAT










TCGCGCCCAGGA

CGATTCCCGTTA

CAATGGCGAGAT










AGTTCGCTTAAC

TTTCATTCACTG

TCGCGCCCAGGA










TGGTCTGGAAGG

ACCGGAGGCCCA

AGTTCGCTTAAC










TGAGCAGCTGGG

CGATGACCCAGC

TGGTCTGGAAGG










TATT

GACC

TGAGCAGCTGGG














TATT/














GCAATAGAACTA














ACTACCCGcccr














GAAGGCGGTACC














TGCCTGACCCTG














CGATTCCCGTTA














TTTCATTCACTG














ACCGGAGGCCCA














CGATGACCCAGC














GACC-3′
















TABLE 31







Remodeled Non-intergeneric Microbes











Strain Name
Genotype
SEQ ID NO







CI006
16S rDNA - contig 5
62



CI006
16S rDNA - contig 8
63



CI019
16S rDNA
64



CI006
nifH
65



CI006
nifD
66



CI006
nifK
67



CI006
niftL
68



CI006
nifA
69



CI019
nifH
70



CI019
nifD
71



CI019
nifK
72



CI019
nifL
73



CI019
nifA
74



CI006
Prm5 with 500 bp
75




flanking regions




CI006
nifLA operon - upstream
76




intergenic region plus





nifL and nifA CDSs




CI006
nifL (Amino Acid)
77



CI006
nifA (Amino Acid)
78



CI006
glnE
79



CI006
glnE_KO1
80



CI006
glnE (Amino Acid)
81



CI006
glnE_ KO1 (Amino Acid)
82



CI006
GlnE ATase domain
83




(Amino Acid)




CM029
Prm5 inserted into nifL
84




region

















TABLE 32







Remodeled Non-intergeneric Microbes

















Associated







Novel



Strain
SEQ ID


Junction If


Strain
ID
NO
Genotype
Description
Applicable





CI63;
63
SEQ ID
16S
N/A
N/A


CI063

NO 85





CI63;
63
SEQ ID
nifH
N/A
NIA


CI063

NO 86





CI63;
63
SEQ ID
nifD1
1 of 2 unique genes annotated as nifD
N/A


CI063

NO 87

in 63 genome



CI63;
63
SEQ ID
nifD2
2 of 2 unique genes annotated as nifD N/A



CI063

NO 88

in 63 genome



CI63;
63
SEQ ID
nifK1
1 of 2 unique genes annotated as nifK.
N/A


CI063

NO 89

in 63 genome



CI63;
63
SEQ ID
nifK2
2 of 2 unique genes annotated as nifK
N/A


CI063

NO 90

in 63 genome



CI63;
63
SEQ ID
nifL
N/A
N/A


CI063

NO 91





CI63;
63
SEQ ID
nifA
N/A
N/A


CI063

NO 92





CI63;
63
SEQ ID
glnE
N/A
NIA


CI063

NO 93





CI63;
63
SEQ ID
amtB
N/A
N/A


CI063

NO 94





CI63;
63
SEQ ID
PinfC
500 bp immediately upstrea of the ATG
N/A


CI063

NO 95

start codon of the infC gene



CI137
137
SEQ ID
16S
N/A
N/A




NO 96





CI137
137
SEQ ID
nifH1
1 of 2 unique genes annotated as nifH
N/A




NO 97

in 137 genome



CI137
137
SEQ ID
nifH2
2 of 2 unique genes annotated as nifH





NO 98

in 137 genome
N/A


CI137
137
SEQ ID
nifD1
1 of 2 unique genes annotated as nifD
N/A




NO 99

in 137 genome



CI137
137
SEQ ID
nifD2
2 of 2 unique genes annotated as nifH
N/A




NO 100

in 137 genome



CI137
137
SEQ ID
nifK1
1 of 2 unique genes annotated as nifK
N/A




NO 101

in 137 genome



CI137
137
SEQ ID
nifK2
2 of 2 unique genes annotated as nifK
N/A




NO 102

in 137 genome



CI137
137
SEQ ID
nifL
N/A
N/A




NO 103





CI137
137
SEQ ID
nifA
N/A
N/A




NO 104





CI137
137
SEQ ID
glnE
N/A
N/A




NO 105





CI137
137
SEQ ID
PinfC
500 bp immediately upstream of the
N/A




NO 106

TTG start codon of infC



CI137
137
SEQ ID
amtB
N/A
N/A




NO 107





CI137
137
SEQ ID
Prm8.2
internal promoter located in nlpI gene;
N/A




NO 108

299 bp starting at 81 bp after the A of







the ATG of the nlpI gene



CI137
137
SEQ ID
Prm.6.2
300 bp upstream of the secE gene
N/A




NO 109

starting at 57 bp upstream of the A of







the ATG of secE



CI137
137
SEQ ID
Prm1.2
400 bp immediately upstream of the
N/A




NO 110

ATG of cspE gene



none
728
SEQ ID
16S
N/A
N/A




NO 111





none
728
SEQ ID
nifH
N/A
N/A




NO 112





none
728
SEQ ID
nifD1
1 of 2 unique genes annotated as nifD
N/A




NO 113

in 728 genome



none
728
SEQ ID
nifD2
2 of 2 unique genes annotated as nifD
N/A




NO 114

in 728 genome



none
728
SEQ ID
nifK1
1 of 2 unique genes annotated as nifK
N/A




NO 115

in 728 genome



none
728
SEQ ID
nifK2
2 of 2 unique genes annotated as nifK
N/A




NO 116

in 728 genome



none
728
SEQ ID
nifL
N/A
N/A




NO 117





none
728
SEQ ID
nifA
N/A
N/A




NO 118





none
728
SEQ ID
glnE
N/A
N/A




NO 119





none
728
SEQ ID
amtB
N/A
N/A




NO 120





none
850
SEQ ID
16S
N/A
N/A




NO 121





none
852
SEQ ID
16S
N/A
N/A




NO 122





none
853
SEQ ID
16S
N/A
N/A




NO 123





none
910
SEQ ID
16S
N/A
N/A




NO 124





none
910
SEQ ID
nifH
N/A
N/A




NO 125





none
910
SEQ ID
Dinitrogenase iron-
N/A
N/A




NO 126
molybdenum







cofactor CDS




none
910
SEQ ID
nifD1
N/A
N/A




NO 127





none
910
SEQ ID
nifD2
N/A
N/A




NO 128





none
910
SEQ ID
nifK1
N/A
N/A




NO 129





none
910
SEQ ID
nifK2
N/A
N/A




NO 130





none
910
SEQ ID
nifL
N/A
N/A




NO 131





none
910
SEQ ID
nifA
N/A
N/A




NO 132





none
910
SEQ ID
glnE
N/A
N/A




NO 133





none
910
SEQ ID
amtB
N/A
N/A




NO 134





none
910
SEQ ID
PinfC
498 bp immediately upstream of the
N/A




NO 135

ATG of the infC gene



none
1021
SEQ ID
16S
N/A
NIA




NO 136





none
1021
SEQ ID
nifH
N/A
N/A




NO 137





none
1071
SEQ ID
nifD1
1 of 2 unique genes annotated as nifD
N/A




NO 138

in 910 genome



none
1021
SEQ ID
nifD2
2 of 2 unique genes annotated as nifD
N/A




NO 139

in 910 genome



none
1021
SEQ ID
nifK1
1 of 2 unique genes annotated as nifK
N/A




NO 140

in 910 genome



none
1021
SEQ ID
nifK2
2 of 2 unique genes annotated as nifK
N/A




NO 141

in 910 genome



none
1021
SEQ ID
nifL
N/A
N/A




NO 142





none
1021
SEQ ID
nifA
N/A
N/A




NO 143





none
1021
SEQ ID
glnE
N/A
N/A




NO 144





none
1021
SEQ ID
amtB
N/A
N/A




NO 145





none
1021
SEQ ID
PinfC
500 bp immediately upstream of the
N/A




NO 146

ATG start codon of the infC gene



none
1021
SEQ ID
PinfC
348 bp includes the 319 bp immediately
N/A




NO 147

upstream of the ATG start codon of the







lpp gene and the first 29 bp of the lpp







gene



none
1021
SEQ ID
Prm1
339 bp upstream of the sspA gene,
N/A




NO 148

ending at 46 bp upstream of the ATG of







the sspA gene



none
1113
SEQ ID
16S
N/A
N/A




NO 149





none
1113
SEQ ID
nifH
N/A
N/A




NO 150





none
1113
SEQ ID
nifD1
1 of 2 unique genes annotated as nifD
N/A




NO 151

in 1113 genome



none
1113
SEQ ID
nifD2
2 of 2 unique genes annotated as nifD
N/A




NO 152

in 1113 genome



none
1113
SEQ ID
nifK
N/A
N/A




NO 153





none
1113
SEQ ID
nifL
N/A
N/A




NO 154





none
1113
SEQ ID
nifA
due to a gap in the sequence assembly,
N/A




NO 155
partial gene
we can only identify a partial gene







from the 1113 genome



none
1111
SEQ ID
glnE
N/A
N/A




NO 156





none
1116
SEQ ID
16S

N/A




NO 157





none
1116
SEQ ID
nifH

N/A




NO 158





none
1116
SEQ ID
nifD1
1 of 2 unique genes annotated as nifD
N/A




NO 159

in 1116 genome



none
1116
SEQ ID
nifD2
2 of 2 unique genes annotated as nifD
N/A




NO 160

in 1116 genome



none
1116
SEQ ID
nifK1
1 of 2 unique genes annotated as nifK
N/A




NO 161

in 1116 genome



none
1116
SEQ ID
nifK2
2 of 2 unique genes annotated as nifK
N/A




NO 162

in 1116 genome



none
1116
SEQ ID
nifL
N/A
N/A




NO 163





none
1116
SEQ ID
nifA
N/A
N/A




NO 164





none
1116
SEQ ID
glnE
N/A
N/A




NO 165





none
1116
SEQ ID
amtB
N/A
N/A




NO 166





none
1293
SEQ ID
16S
N/A
N/A




NO 167





none
1293
SEQ ID
nifH
N/A
N/A




NO 168





none
1293
SEQ ID
nifD1
1 of 2 unique genes annotated as nifD
N/A




NO 169

in 1293 genome



none
1293
SEQ ID
nifD2
2 of 2 unique genes annotated as nifD
N/A




NO 170

in 1293 genome



none
1293
SEQ ID
nifK
1 of 2 unique genes annotated as nifK
N/A




NO 171

in 1293 genome



none
1293
SEQ ID
nifK1
2 of 2 unique genes annotated as nifK
N/A




NO 172

in 1293 genome



none
1293
SEQ ID
nifA
N/A
N/A




NO 173





none
1293
SEQ ID
glnE
N/A
N/A




NO 174





none
1293
SEQ ID
amtB1
1 of 2 unique genes annotated as amtB
N/A




NO 175

in 1293 genome



none
1293
SEQ ID
amtB1
2 of 2 unique genes annotated as amtB
N/A




NO 176

in 1293 genome



none
1021-
SEQ ID
ΔnifL::PinfC
starting at 24 bp after the A of the ATG
ds1131



1612
NO 177

start codon, 1375 bp of nifL have been







deleted and replaced with the 1021







PinfC promoter sequence



none
1021-
SEQ ID
ΔnifL::PinfC with
starting at 24 bp after the A of the ATG
ds1131



1612
NO 178
500 bp flank
start codon, 1375 bp of nifL have been







deleted and replaced with the 1021







PinfC promoter sequence; 500 bp







flanking the nifL gene upstream and







downstream are included



none
1021-
SEQ ID
glnEΔAR-2
glnE gene with 1673 bp immediately
ds1133



1612
NO 179

downstream of the ATG start codon







deleted, resulting in a truncated glnE







protein lacking the adenylyl-removing







(AR) domain



none
1021-
SEQ ID
glnEΔAR-2 with
glnE gene with 1673 bp immediately
ds1133



1612
NO 180
500 bp flank
downstream of the ATG start codon







deleted, resulting, in a truncated glnE







protein lacking the adenylyl-removing







(AR) domain; 500 bp flanking the glnE,







gene upstream and downstream are







included



none
1021-
SEQ ID
ΔnifL::Prm1
starting at 24 bp after the A of the ATG
ds1145



1615
NO 181

start codon, 1375 bp of nifL have been







deleted and replaced with the 1021







Prm1 promoter sequence



none
1021-
SEQ ID
ΔnifL::Prm1 with
starting at 24 bp after the A of the ATG
ds1145



1615
NO 182
500 bp flank
start codon, 1375 bp of nifL have been







deleted and replaced with the 1021 rm1







promoter sequence; 500 bp flanking the







nifL gene upstream and downstream







are included



none
1021-
SEQ ID
glnEΔAR-2
glnE gene with 1673 bp immediately
ds1133



1615
NO 183

downstream of the ATG start codon







deleted, resulting in a truncated glnE







protein lacking the adenylyl-removing







(AR) domain



none
1021-
SEQ ID
glnEΔAR-2 with
glnE gene with 1673 bp immediately
ds1133



1615
NO 184
500 bp flank
downstream of the ATG start codon







deleted, resulting in a truncated glnE







protein lacking the adenytyl-removing







(AR) domain; 500 bp flanking the glnE







gene upstream and downstream are







included



none
1021-
SEQ ID
ΔnifL::Prm1
starting at 24 bp after the A of the ATG
ds1145



1619
NO 185

start codon, 1375 bp of nifL have been







deleted and replaced with the 1021







Prm1 promoter sequence



none
1021-
SEQ ID
ΔnifL::Prm1 with
starting at 24 bp after the A of the ATG
ds1145



1619
NO 186
500 bp flank
start codon, 1375 bp of nifL have been







deleted and replaced with the 1021 rm1







promoter sequence; 500 bp flanking the







nifL gene upstream and downstream







are included



none
1021-
SEQ ID
glnEΔAR-2
glnE gene with 1673 bp immediately
ds1133



1623
NO 187

downstream of the ATG start codon







deleted, resulting in a truncated glnE







protein lacking the adenylyl-removing







(AR) domain



none
1021-
SEQ ID
glnEΔAR-2 with
glnE gene with 1673 bp immediately
ds1133



1623
NO 188
500 bp flank
downstream of the ATG start codon







deleted, resulting in a truncated glnE







protein lacking the aderlylyl-removing







(AR) domain; 500 bp flanking the glnE







gene upstream and downstream are







included



none
1021-
SEQ ID
ΔnifL::Prm7
starting at 24 bp after the A of the ATG
ds1148



1623
NO 189

start codon, 1375 bp of nifL have been







deleted and replaced with the 1021







Prm7 promoter sequence



none
1021-
SEQ ID
ΔnifL::Prm7 with
starting at 24 bp after the A of the ATG
ds1148



1623
NO 190
500 bp flank
start codon, 1375 bp of nifL have been







deleted and replaced with the 1021 rm7







promoter sequence; 500 bp flanking the







nifL gene upstream and downstream







are included



none
137-
SEQ ID
glnEΔAR-2
glnE gene with 1290 bp immediately
ds809



1034
NO 191

downstream of the ATG start codon







deleted, resulting in a truncated glnE







protein lacking the adenytyl-iremoving







(AR) domain



none
137-
SEQ ID
glnEΔAR-2 with
glnE gene with 1290 bp immediately
ds809



1034
NO 192
500 bp flank
downstream of the ATG start codon







deleted, resulting in a truncated glnE







protein lacking the adenylyl-removing







(AR) domain; 500 bp flanking the glnE







gene upstream and downstream are







included



none
137-
SEQ ID
ΔnifL::PinfC
starting at 24 bp after the A of the ATG
ds799



1036
NO 193

start codon, 1372 bp of nifL have been







deleted and replaced with the 137







PinfC promoter sequence



none
137-
SEQ ID
ΔnifL::PinfC with
starting at 24 bp after the A of the ATG
ds799



1036
NO 194
500 bp flank
start codon, 1372 bp of nifL have been







deleted and replaced with the 137







PinfC promoter sequence; 500 bp







flanking the nifL gene upstream and







downstream are included



none
137-
SEQ ID
glnEΔAR-2 36 bp
glnE gene with 1290 bp immediately
none



1314
NO 195
deletion
downstream of the ATG start codon







deleted AND 36 bp deleted beginning at







1472 bp downstream of the start codon,







resulting in a truncated glnE protein







lacking the adenylyl-removing (AR)







domain



none
137-
SEQ ID
glnEΔAR-2 36 bp
glnE gene with 1290 bp immediately
none



1314
NO 196
deletion
downstream of the ATG start codon







deleted AND 36 bp deleted beginning at







1472 bp downstream of the start codon,







resulting in a truncated glnE protein







lacking the adenylyl-removing (AR)







domain; 500 bp flanking the nifL gene







upstream and downstream are included



none
137-
SEQ ID
ΔnifL::Prm8.2
starting at 24 bp after the A of the ATG
ds857



1314
NO 197

start codon, 1372 bp of nifL have been







deleted and replaced with the 137







Prm8.2 promoter sequence



none
137-
SEQ ID
ΔnifL::Prm8.2 with
starting at 24 bp after the A of the ATG
ds857



1314
NO 198
500 bp flank
start codon, 1372 bp of nifL have been







deleted and replaced with the 137







Prm8.2 promoter sequence; 500 bp







flanking the nifL gene upstream and







downstream are included



none
137-
SEQ ID
glnEΔAR-2 36 bp
glnE gene with 11290 bp immediately
none



1329
NO 199
deletion
downstream of the ATG start codon







deleted AND 36 bp deleted beginning at







1472 bp downstream of the start codon,







resulting in a truncated glnE protein







lacking the adenylyl-removing (AR)







domain



none
137-
SEQ ID
glnEΔAR-2 36 bp
glnE gene with 11290 bp immediately
none



1329
NO 200
deletion
downstream of the ATG start codon







deleted AND 36 bp deleted beginning at







1472 bp downstream of the start codon







resulting in a truncated glnE protein







lacking the adenylyl-removing (AR)







domain; 500 bp flanking the nifL gene







upstream and downstream are included



none
137-
SEQ ID
ΔnifL::Prm6.2
starting at 24 bp after the A of the ATG
ds853



1329
NO 201

start codon, 1372 bp of nifL have been







deleted and replaced with the 137







Prm6.2 promoter sequence



none
137-
SEQ ID
ΔnifL::Prm6.2 with
starting at 24 bp after the A of the ATG
ds853



1329
NO 202
500 bp flank
start codon, 1372 bp of nifL have been







deleted and replaced with the 137







Prm6.2 promoter sequence; 500 bp







flanking the nifL gene upstream and







downstream are included



none
137-
SEQ ID
ΔnifL::Prm1.2
starting at 24 bp after the A of the ATG
ds843



1382
NO 203

start codon, 13-72 bp of nifL have been







deleted and replaced with the 137







Prm1.2 promoter sequence



none
137-
SEQ ID
ΔnifL::Prm1.2 with
starting at 24 bp after the A of the ATG
ds843



1382
NO 204
500 bp flank
start codon, 1372 bp of nifL have been







deleted and replaced with the 137







Prm1.2 promoter sequence; 500 bp







flanking the nifL gene upstream and







downstream are included



none
137-
SEQ ID
glnEΔAR-2 36 bp
glnE gene with 1290 bp immediately
none



1382
NO 205
deletion
downstream of the ATG start codon







deleted AND 36 bp deleted beginning at







1472 bp downstream of tile start codon,







resulting in a truncated glnE protein







lacking the adenylyl-removing (AR)







domain



none
137-
SEQ ID
glnEΔAR-2 36 bp
glnE gene with 1290 bp immediately
none



1382
NO 206
deletion
downstream of the ATG start codon







deleted AND 36 bp deleted beginning at







1472 bp downstream of the start codon,







resulting in a truncated glnE protein







lacking the adenylyl-removing (AR)







domain, 500 bp flanking the nifL gene







upstream and downstream are included



none
137-
SEQ ID
ΔnifL::PinfC
starting at 24 bp after the A of the ATG
ds799



1586
NO 207

start codon, 1372 bp of nifL have been







deleted and replaced with the 137







PinfC promoter sequence



none
137-
SEQ ID
ΔnifL::PinfC with
starting at 24 bp after the A of the ATG
ds799



1586
NO 208
500 bp flank
start codon, 1372 bp of nifL have been







deleted and replaced with the 137







PinfC promoter sequence; 500 bp







flanking the nifL gene upstream and







downstream are included



none
137-
SEQ ID
glnEΔAR-2
glnE gene with 1290 bp immediately
ds809



1586
NO 209

downstream of the ATG start codon







deleted, resulting in a truncated glnE







protein lacking the adenylyl-removing







(AR) domain



none
137-
SEQ ID
glnEΔAR-2 with
glnE gene with 1290 bp immediately
ds809



1586
NO 210
500 bp flank
downstream of the ATG start codon







deleted, resulting in a truncated glnE







protein lacking the adenylyl-removing







(AR) domain; 500 bp flanking the glnE







gene upstream and downstream are







included



none
19-594
SEQ ID
glnEΔAR-2
glnE gene with 1650 bp immediately
ds34




NO 211

downstream of the ATG start codon







deleted, resulting in a truncated glnE







protein lacking the adenylyl-removing







(AR) domain



none
19-594
SEQ ID
glnEΔAR-2 with
glnE gene with 1650 bp immediately
ds34




NO 212
500 bp flank
downstream of the ATG start codon







deleted, resulting in a truncated glnE







protein lacking the adenylyl-removing







(AR) domain; 500 bp flanking the glnE







gene upstream and downstream are







included



none
19-594
SEQ ID
ΔnifL::Prm6.1
starting at 221 bp after the A of the
ds180




NO 213

ATG start codon, 845 bp of nifL have







been deleted and replaced with the







CI019 Prm6.1 promoter sequence



none
19-594
SEQ ID
ΔnifL::Prm6.1 with
starting at 221 bp after the A of the
ds180




NO 214
500 bp flank
ATG start codon, 845 bp of nifL have







been deleted and replaced with the







CI019 Prm6.1promoter sequence;







500 bp flanking the nifL gene upstream







and downstream are included



none
19-714
SEQ ID
ΔnifL::Prm6.1
starting at 221 bp after the A of the
ds180




NO 215

ATG start codon, 845 bp of nifL have







been deleted and replaced with the







CI019 Prm6.1 promoter sequence



none
19-714
SEQ ID
ΔnifL::Prm6.1 with
starting at 221 bp after the A of the
ds180




NO 216
500 bp flank
ATG start codon, 845 bp of nifL have







been deleted and replaced with the







CI019 Prm6.1promoter sequence;







500 bp flanking the nifL gene upstream







and downstream are included



none
19-715
SEQ ID
ΔnifL::Prm7.1
starting at 221 bp after the A of the
ds181




NO 217

ATG start codon, 845 bp of nifL have







been deleted and replaced with the







CI019 Prm7.1 promoter sequence



none
19-715
SEQ ID
ΔnifL::Prm7.1 with
starting at 221 bp after the A of the
ds181




NO 218
500 bp flank
ATG start codon, 845 bp of nifL have







been deleted and replaced with the







CI019 Prm76.1 promoter sequence;







500 bp flanking the nifL gene upstream







and downstream are included



19-713
19-750
SEQ ID
ΔnifL::Prm1.2
starting at 221 bp after the A of the
ds172




NO 219

ATG start codon, 845 bp of nifL have







been deleted and replaced with the







CI019 Prm1.2 promoter sequence



19-713
19-750
SEQ ID
ΔnifL::Prm1.2 with
starting at 221 bp after the A of the
ds172




NO 220
500 bp flank
ATG start codon, 845 bp of nifL have







been deleted and replaced with the







CI019 Prm1.2 promoter sequence;







500 bp flanking the nifL gene upstream







and downstream are included



17-724
19-804
SEQ ID
ΔnifL::Prm1.2
starting at 221 bp after the A of the
ds172




NO 221

ATG start codon, 845 bp of nifL have







been deleted and replaced with the







CI019 Prm1.2 promoter sequence



17-724
19-804
SEQ ID
ΔnifL::Prm1.2 with
starting at 221 bp after the A of the
ds172




NO 222
500 bp flank
ATG start codon, 845 bp of nifL have







been deleted and replaced with the







CI019 Prm1.2 promoter sequence;







500 bp flanking the nifL gene upstream







and downstream are included



17-724
19-804
SEQ ID
glnEΔAR-2
glnE gene with 1650 bp immediately
ds34




NO 223

downstream of the ATG start codon







deleted, resulting in a truncated glnE







protein lacking the adenylyl-removing







(AR) domain



17-724
19-804
SEQ ID
glnEΔAR-2 with
glnE gene with 1650 bp immediately
ds34




NO 224
500 bp flank
downstream of the ATG start codon







deleted, resulting in a truncated glnE







protein lacking the adenylyl-removing







(AR) domain; 500 bp flanking the glnE







gene upstream and downstream are







included



19-590
19-806
SEQ ID
ΔnifL::Prm3.1
starting at 221 bp after the A of the
ds175




NO 225

ATG start codon, 845 bp of nifL have







been deleted and replaced with the







CI019 Prm3.1 promoter sequence



19-590
19-806
SEQ ID
ΔnifL::Prm3.1 with
starting at 221 bp after the A of the
ds175




NO 226
500 bp flank
ATG start codon, 845 bp of nifL have







been deleted and replaced with the







CI019 Prm3.1 promoter sequence;







500 bp flanking the nifL gene upstream







and downstream are included



19-590
19-806
SEQ ID
glnEΔAR-2
glnE gene with 1650 bp immediately
ds34




NO 227

downstream of the ATG start codon







deleted, resulting in a truncated glnE







protein lacking the adenylyl-removing







(AR) domain



19-590
19-806
SEQ ID
glnEΔAR-2 with
glnE gene with 1650 bp immediately
ds34




NO 228
500 bp flank
downstream of the ATG start codon







deleted, resulting in a truncated glnE







protein lacking the adenylyl-removing







(AR) domain; 500 bp flanking the glnE







gene upstream and downstream are







included



none
63-
SEQ ID
ΔnifL::PinfC
starting at 24 bp after the A of the ATG
ds908



1146
NO 229

start codon, 1375 bp of nifL have been







deleted and replaced with the 63 PinfC







promote sequence



none
63-
SEQ ID
ΔnifL::PinfC with
starting at 24 bp after the A of the ATG
ds908



1146
NO 230
500 bp flank
start codon, 1375 bp of nifL have been







deleted and replaced with the 63 PindC







flank promoter sequence; 5004 flanking the







nifL gene upstream and downstream







are included



CM015;
6-397
SEQ ID
ΔnifL::Prm5
starting at 31 bp after the A of the ATG
ds24


PBC6.15

NO 231

start codon, 1375 bp of nifL have been







deleted and replaced with the CI006







Prm5 promoter sequence



CM015;
6-397
SEQ ID
ΔnifL::Prm5 with
starting at 31 bp after the A of the ATG
ds24


PBC6.15

NO 232
500 bp flank
start codon, 1375 bp of nifL have been







deleted and replaced with the CI006







Prm5 promoter sequence; 500 bp







flanking the nifL gene upstream and







downstream are included



CM014
6-400
SEQ ID
ΔnifL::Prm1
starting at 31 bp after the A of the ATG
ds20




NO 233

start codon, 1375 bp of nifL have been







deleted and replaced with the CI006







Prm1 promoter sequence



CM014
6-400
SEQ ID
ΔnifL::Prm1 with
starting at 31 bp after the A of the ATG
ds20




NO 234
500 bp flank
start codon, 1375 bp of nifL have been







deleted and replaced with the CI006







Prm1 promoter sequence; 500 bp







flanking the nifL gene upstream and







downstream are included



CM037;
6-403
SEQ ID
ΔnifL::Prm1
starting at 31 bp after the A of the ATG
ds20


PBC6.37

NO 235

start codon, 1375 bp of nifL have been







deleted and replaced with the CI006







Prm1 promoter sequence



CM037;
6-403
SEQ ID
ΔnifL::Prm1 with
starting at 31 bp after the A of the ATG
ds20


PBC6.38

NO 236
500 bp flank
start codon, 1375 bp of nifL have been







deleted and replaced with the CI006







Prm1 promoter sequence; 500 bp







flanking the nifL gene upstream and







downstream are included



CM037;
6-403
SEQ ID
glnEΔAR-2
glnE gene with 1644 bp immediately
ds31


PBC6.39

NO 237

downstream of the ATG start codon







deleted, resulting in a truncated glnE







protein lacking the adenylyl-removing







(AR) domain



CM037;
6-403
SEQ ID
glnEΔAR-2 with
glnE gene with 1644 bp immediately
ds31


PBC6.40

NO 238
500 bp flank
downstream of the ATG start codon







deleted, resulting in a truncated glnE







protein lacking the adenylyl-removing







(AR) domain; 500 bp flanking the ginE







gene upstream and downstream are







included



CM038;
6-404
SEQ ID
glnEΔAR-1
glnE gene with 1287 bp immediately
ds30


PBC6.38

NO 239

downstream of the ATG start codon







deleted, resulting in a truncated glnE







protein lacking the adenylyl-removing







(AR) domain



CM038;
6-404
SEQ ID
ΔnifL::Prm1
starting at 31 bp after the A of the ATG
ds20


PBC6,38

NO 240

start codon 1375 bp of nifL have been







deleted and replaced with the CI006







Prm1 promoter sequence



CM038;
6-404
SEQ ID
ΔnifL::Prm1 with
starting at 31 bp after the A of the ATG
ds20


PBC6.38

NO 241
500 bp flank
start codon, 1375 bp of nifL have been







deleted and replaced with the CI006







Prm1 promoter sequence; 500 bp







flanking the nifl gene upstream and







downstream are included



CM029
6-404
SEQ ID
glnEΔAR-1 with
glnE gene with 1287 bp immediately
ds30


PBC6.38

NO 242
500 bp flank
downstream of the ATG start codon







deleted, resulting in a truncated glnE







protein lacking the adenylyl-removing







(AR) domain; 500 bp flanking the glnE







gene upstream and downstream are







included



CM029;
6-412
SEQ ID
glnEΔAR-1
glnE gene with 1287 bp immediately
ds30


PBC6.29

NO 243

downstream of the ATG start codon







deleted, resulting in a truncated glnE







protein lacking the adenylyl-removing







(AR) domain



CM029 ;
6-412
SEQ ID
glnEΔAR-1 with
glnE gene with 1287 bp immediately
ds30


PBC6.29

NO 244
500 bp flank
downstream of the ATG start codon







deleted, resulting in a truncated gliff,







protein lacking the adenylyl-removing







(AR) domain; 500 bp flanking the glriE







gene upstream and downstream are







included



CM029;
6-412
SEQ ID
ΔnifL::Prm5
starting at 31 bp after the A of the ATG
ds24


PBC6.29

NO 245

start codon, 1375 bp of nifL have been







deleted and replaced with the CI006







Prm5 promoter sequence



CM029;
6-412
SEQ ID
ΔnifL::Prm5 with
starting at 31 bp after the A of the ATG
ds24


PBC6.29

NO 246
500 bp flank
start codon, 1375 bp of nifL have been







deleted and replaced with the CI006







Prm5 promoter sequence; 500 bp







flanking the nifL gene upstream and







downstream are included



CM093-
6-848
SEQ ID
ΔnifL::Prm1
starting at 31 bp after the A of the ATG
ds20


PBC6.93

NO 247

start codon, 1375 bp of nifL have been







deleted and replaced with the CI006







Prm1 promoter sequence



CM093;
6-848
SEQ ID
ΔnifL::Prm1 with
starting at 31 bp after the A of the ATG
ds20


PBC6.93

NO 248
500 bp flank
start codon, 1375 bp of nifL have been







deleted and replaced with the CI006







Prm1 promoter sequence; 500 bp







flanking the nifL gene upstream and







downstream are included



CM093;
6-848
SEQ ID
glnEΔAR-2
glnE gene with 1644 bp immediately
ds31


PBC693

NO 249

downstream of the ATG start codon







deleted, resulting in a truncated glnE







protein lacking the adenylyl-removing







(AR) domain



CM093;
6-848
SEQ ID
glnEΔAR-2
glnE gene with 1644 bp immediately
ds31


PBC6.93

NO 250
500 bp flank
downstream of the ATG start codon







deleted, resulting in a truncated glnE







protein lacking the adenylyl-removing







(AR) domain; 500 bp flanking the glnE,







gene upstream and downstream are







included



CM093;
6-848
SEQ ID
ΔamtB
First 1088 bp of amtB gene and 4 bp
ds126


PBC6,93

NO 251

upstream of start codon deleted; 199 bp







of gene remaining lacks a start codon;







no amtB protein is translated



CM093;
6-848
SEQ ID
ΔamtB with 500 bp
First 1088 bp of amtB gene and 4 bp
ds126


PBC6.93

NO 252
flank
upstream of start codon deleted; 199 bp







of gene remaining lacks a staff codon;







no amtB protein is translated



CM094;
6-881
SEQ ID
glnEΔAR-1
glnE gene with 1287 bp immediately
ds30


PBC6.94

NO 253

downstream of the ATG start codon







deleted, resulting in a truncated glnE







protein lacking the adenylyl-removing







(AR) domain



CM094;
6-881
SEQ ID
glnEΔAR-1
glnE gene with 1287 bp immediately
ds30


PBC6.94

NO 254
500 bp flank
downstream of the ATG start codon







deleted, resulting in a truncated glnE







protein lacking the adenylyl-removing







(AR) domain; 500 bp flanking the glnE







gene upstream and downstream are







included



CM094;
6-881
SEQ ID
ΔnifL::Prm1
starting at 31 bp after the A of the ATG
ds20


PBC6.94

NO 255

start codon, 1375 bp of nifL have been







deleted and replaced with the CI006







Prm1 promoter sequence



CM094;
6-881
SEQ ID
ΔnifL::Prm1 with
starting at 31 bp after the A of the ATG
ds20


PBC6.94

NO 256
500 bp flank
start codon, 1375 bp of nifL have been







deleted and replaced with the CI006







Prm1 promoter sequence; 500 bp







flanking the nifL gene upstream and







downstream are included



CM094;
6-881
SEQ ID
ΔamtB
First 1088 bp of amtB gene and 4 bp
ds126


PBC6.94

NO 257

upstream of start codon deleted; 199 bp







of gene remaining lacks a start codon;







no amtB protein is translated



CM094;
6-881
SEQ ID
ΔamtB with 500 bp
First 1088 bp of amtB gene and 4 bp
ds126


PBC6.94
NO
258
flank
upstream of start codon deleted; 1.99 bp







of gene remaining lacks a start codon;







no amtB protein is translated



none
910-
SEQ ID
ΔnifL::PinfC
starting at 20 bp after the A of the ATG
ds960



1246
NO 259

start codon, 1379 bp of nifL have been







deleted and replaced with the 910







PinfC promoter sequence



none
910-
SEQ ID
ΔnifL::PinfC with
starting at 20 bp after the A of the ATG
ds960



1246
NO 260
500 bp flank
start codon, 1379 bp of nifL have been







deleted and replaced with the 910







PinfC promoter sequence; 500 bp







flanking the nifL gene upstream and







downstream are included



PBC6.1,
CI006
SEQ ID
16S-1
1 of 3 unique 16S rDNA genes in the
N/A


6, CI6

NO 261

CI006 genome



PBC6.1,
CI006
SEQ ID
16S-2
2 of 3 unique 16S rDNA genes in the
N./A


6, CI6

NO 262

CI006 genome



PBC6.1,
CI006
SEQ ID
nifH
N/A
N/A


6, CI6

NO 263





PBC6.1,
CI006
SEQ ID
nifD2
2 of 2 unique genes annotated as nifD
N/A


6, CI6

NO 264

in CI006 genome



PBC6.1,
C1006
SEQ ID
nifK2
2 of 2 unique genes annotated as nifK



6, CI6

NO 265

in CI006 genome



PBC6.1,
CI006
SEQ ID
nifL
N/A
NIA


6, CI6

NO 266





PBC6.1,
CI006
SEQ ID
nifA
N/A
N/A


6, CI6

NO 267





PB C6.1,
CI006
SEQ ID
glnE
N/A
N/A


6, CI6

NO 268





PBC6.1,
CI006
SEQ ID
16S-3
3 of 3 unique 16S rDNA genes in the
N/ A


6, CI6

NO 269

CI006 gettome



PBC6.1,
CI006
SEQ ID
nifD1
1 of 2 unique genes annotated as nifD
N/A


6, CI6

NO 270

in CI006 genome



PBC6.1 ,
CI006
SEQ ID
nifK1
1 of 2 unique genes annotated as nifK
N/A


6, CI6

NO 271

in CI006 genome



PBC6.1
CI006
SEQ ID
amtB
N/A
N/A


6, CI6

NO 272





PBC6.1,
CI006
SEQ ID
Prm1
348 bp includes the 319 bp immediately
N/A


6, CI6

NO 273

upstream of the ATG start codon of the







lpp gene and the first 29 bp of the lpp







gene



PBC6.1,
CI006
SEQ ID
Prm5
313 bp starting at 432 bp upstream of the
N/A


6, CI6

NO 274

ATG start codon of the ompX gene and







ending 119 bp upstream of the ATG







start codon of the orapX gene



19, CI1.9
CI019
SEQ ID
nifL
N/A
N/A




NO 275





19, CI19
CI019
SEQ ID
nifA
N/A
N/A




NO 276





19, CI19
CI019
SEQ ID
16S-1
1 of 7 unique 16S rDNA genes in the
N/A




NO 277

CI019 genome



19, C19
CI019
SEQ ID
16S-2
2 of 7 unique 16S rDNA genes in the
N/A




NO 278

CI019 genome



19, C119
CI019
SEQ ID
16S-3
3 of 7 unique 16S rDNA genes in the
N/A




NO 279

CI019 genome



19, C119
CI019
SEQ ID
16S-4
4 of 7 unique 16S rDNA genes in the
N/A




NO 280

CI019 genome



19, CI19
CI019
SEQ ID
16S-5
5 of 7 unique 165 rDNA genes in the
N/A




NO 281

CI019 genome



19, CI19
CI019
SEQ ID
16S-6
6 of 7 unique 165 rDNA genes in the
N/A




NO 282

CI019 genome



19, CI19
CI019
SEQ ID
16S-7
7 of 7 unique 16S rDNA genes in the
N/A




NO 283

CI019 genome



19, CI19
CI019
SEQ ID
nifH1
1 of 2 unique genes annotated as nifH
N/A




NO 284

in CI019 genome



19, CI19
CI019
SEQ ID
nifH2
2 of 2 unique genes annotated as nifH
N/A




NO 285

in CI019 genome



19, CI19
CI019
SEQ ID
nifD1
1 of 2 unique genes annotated as nifD
N/A




NO 286

in CI019 genome



19, CI19
CI019
SEQ ID
nifD2
2 of 2 unique genes annotated as nifD
N/A




NO 287

in CI019 genome



19, CI19
CI019
SEQ ID
nifK1
1 of 2 unique genes annotated as nifK
N/A




NO 288

in CI019 genome



19, CI19
CI019
SEQ ID
nifK2
2 of 2 unique genes annotated as nifK
N/A




NO 289

in CI019 genome



19, CI19
CI019
SEQ ID
glnE
N/A
N/A




NO 290





19, CI19
CI019
SEQ ID
Prm4
449 bp immediately upstream of the
N/A




NO 291

ATG of the dscC 2 gene



19, CI19
CI019
SEQ ID
Prm1.2
500 bp inurtedittely upstream of the
N/A




NO 292

TTG start codon of the infC gene



19, CI19
CI019
SEQ ID
Prm3.1
170 bp immediately upstream of the
N/A




NO 293

ATG start codon of the rplN gene



19, CI20
CI020
SEQ ID
Prm6.1
142 bp immediately upstream of the
N/A




NO 294

ATG of a highly-expressed







hypothetical protein (annotated as







PROKKA_00662 in CI019 assembly







82)



19, CI21
CI021
SEQ ID
Prm7.1
293 bp immediately upstream of the
N/A




NO 295

ATG of the lpp gene







glnE gene with 1650 bp immediately



19-375,
CM67
SEQ ID
glnEΔAR-2
downstream of the ATG start codon
ds34


19-417,

NO 296

deleted, resulting in a truncated glnE



CM067



protein lacking the adenylyl-removing







(AR) domain



19-375,
CM67
SEQ ID
glnEΔAR-1 with
glnE gene with 1650 bp immediately
ds34


19-417,

NO 297
500 bp flank
downstream of the ATG start codon



CM067



deleted, resulting in a truncated glnE







rotein lacking, the adenyltyl-removing







(AR) domain; 500 bp flanking the glnE







gene upstream and downstream are







included



19-375,
CM67
SEQ ID
ΔnifL::null-v1
starting at 221 bp after the A of the
none


19-417,

NO 298

ATG start codon, 845 bp of nifL have



CM067



been deleted and replaced with the







31 bp sequence







“GGAGTCTGAACTCATCCTGCGA







TGGGGGCTG”



19-375,
CM6
SEQ ID
ΔnifL::null-v1 with
starting at 221 bp after the A of the
none


19-417

NO 299
500 bp flank
ATG start codon, 845 bp of nifL have



CM067



been deleted and replaced with the







31 bp sequence







“GGAGTCTGAACTCATCCTGCGA







TGGGGGCTG”; 500 bp flanking the







nifL gene upstream and downstream







are included



19-377,
CM69
SEQ ID
ΔnifL::null-v2
starting at 221 bp after the A of the
none


CM069

NO 300

ATG start codon, 845 bp of nifL have







been deleted and replaced with the 5 bp







sequence “TTAAA”



19-377,
CM69
SEQ ID
ΔnifL::null-v2
starting at 221 bp after the A of the
none


CM069

NO 301
500 bp flank
ATG start codon, 845 bp of ilia, have







been deleted and replaced with the 5 bp







sequence “TTAAA”; 500 bp flanking







the nifL gene upstream and







downstream are included



19-389,
CM81
SEQ ID
ΔnifL::Prm4
starting at 221 bp after the A of the
ds70


19-418,

NO 302

ATG start codon, 845 bp of nifL have



CM081



been deleted and replaced with the







CI19 Prm4 sequence



19-389,
CM81
SEQ ID
ΔnifL::Prm4 with
starting at 221 bp after the A of the
ds70


19-418,

NO 303
500 bp flank
ATG start codon, 845 bp of nifL have



CM081



been deleted and replaced with the







CI19 Prm4 sequence; 500 bp flanking







the nifL gene upstream and







downstream are included









NUMBERED EMBODIMENTS OF THE DISCLOSURE

Notwithstanding the appended claims, the disclosure sets forth the following numbered embodiments:


1. A seed treatment composition, comprising:

    • a. a plurality of non-intergeneric remodeled bacteria that have an average colonization ability per unit of plant root tissue of at least about 1.0×104 bacterial cells per gram of fresh weight of plant root tissue and produce fixed N of at least about 1×1017 mmol N per bacterial cell per hour; and
    • b. at least one pesticide.


2. The seed treatment composition, according to embodiment 1, wherein said pesticide is a fungicide.


3. The seed treatment composition, according to embodiment 1 or 2, wherein said pesticide is a fungicide selected from the group consisting of: fludioxonil, metalaxyl, mefenoxam, azoxystrobin, thiabendazole, ipconazole, tebuconazole, prothioconazole, and combinations thereof.


4. The seed treatment composition, according to embodiment 1, wherein said pesticide is an insecticide.


5. The seed treatment composition, according to embodiment 1 or 4, wherein said pesticide is a neonicotinoid insecticide.


6. The seed treatment composition, according to embodiment 1 or 4, wherein said pesticide is an insecticide selected from the group consisting of: imidacloprid, clothianidin, thiamethoxam, chlorantraniliprole, and combinations thereof.


7. The seed treatment composition, according to any one of embodiments 1-6, wherein said at least one pesticide is a fungicide and an insecticide combination.


8. The seed treatment composition, according to embodiment 1, wherein said pesticide is a nematicide.


9. The seed treatment composition, according to embodiment 1, wherein said pesticide is an herbicide.


10. The seed treatment composition, according to any one of embodiments 1-9, wherein said pesticide is selected from those in Table 13.


11. The seed treatment composition, according to any one of embodiments 1-10, wherein the non-intergeneric remodeled bacteria and pesticide exhibit a synergistic effect.


12. The seed treatment composition, according to any one of embodiments 1-11, wherein said seed treatment is disposed onto a seed.


13. The seed treatment composition, according to any one of embodiments 1-12, wherein said seed treatment is disposed onto a seed from the family Poaceae.


14. The seed treatment composition, according to any one of embodiments 1-13, wherein said seed treatment is disposed onto a cereal seed.


15. The seed treatment composition, according to any one of embodiments 1-14, wherein said seed treatment is disposed onto a corn, rice, wheat, barley, Sorghum, millet, oat, rye, or triticale seed.


16. The seed treatment composition, according to any one of embodiments 1-15, wherein said seed treatment is disposed onto a corn seed.


17. The seed treatment composition, according to any one of embodiments 1-16, wherein said seed treatment is disposed onto a genetically modified corn seed.


18. The seed treatment composition, according to any one of embodiments 1-17, wherein said seed treatment is disposed onto a genetically modified corn seed, wherein said corn comprises an herbicide tolerant trait.


19. The seed treatment composition, according to any one of embodiments 1-18, wherein said seed treatment is disposed onto a genetically modified corn seed, wherein said corn comprises an insect resistance trait.


20. The seed treatment composition, according to any one of embodiments 1-19, wherein said seed treatment is disposed onto a genetically modified corn seed, wherein said corn comprises an herbicide tolerant trait and also an insect resistance trait.


21. The seed treatment composition, according to any one of embodiments 1-20, wherein said seed treatment is disposed onto a genetically modified corn seed, wherein said corn comprises a trait listed in Table 19.


22. The seed treatment composition, according to any one of embodiments 1-16, wherein said seed treatment is disposed onto a non-genetically modified corn seed.


23. The seed treatment composition, according to any one of embodiments 1-23, wherein said seed treatment is disposed onto sweet corn, flint corn, popcorn, dent corn, pod corn, or flour corn.


24. The seed treatment composition, according to any one of embodiments 1-23, wherein the plurality of non-intergeneric remodeled bacteria produce 1% or more of the fixed nitrogen in a plant exposed thereto.


25. The seed treatment composition, according to any one of embodiments 1-24, wherein the non-intergeneric remodeled bacteria are capable of fixing atmospheric nitrogen in the presence of exogenous nitrogen.


26. The seed treatment composition, according to any one of embodiments 1-25, wherein each member of the plurality of non-intergeneric remodeled bacteria comprises at least one genetic variation introduced into at least one gene, or non-coding polynucleotide, of the nitrogen fixation or assimilation genetic regulatory network.


27. The seed treatment composition, according to any one of embodiments 1-26, wherein each member of the plurality of non-intergeneric remodeled bacteria comprises an introduced control sequence operably linked to at least one gene of the nitrogen fixation or assimilation genetic regulatory network.


28. The seed treatment composition, according to any one of embodiments 1-27, wherein each member of the plurality of non-intergeneric remodeled bacteria comprises a heterologous promoter operably linked to at least one gene of the nitrogen fixation or assimilation genetic regulatory network.


29. The seed treatment composition, according to any one of embodiments 1-28, wherein each member of the plurality of non-intergeneric remodeled bacteria comprises at least one genetic variation introduced into a member selected from the group consisting of: nifA, nifL, ntrB, ntrC, polynucleotide encoding glutamine synthetase, glnA, glnB, glnK, drat, amtB, polynucleotide encoding glutaminase, glnD, glnE, nifJ, nifH, nifD, nifK, nifY, nifE, nifN nifU, nifS, nifV, nifW, nifZ, nifM, nifF, nifB, nifQ, a gene associated with biosynthesis of a nitrogenase enzyme, or combinations thereof.


30. The seed treatment composition, according to any one of embodiments 1-29, wherein each member of the plurality of non-intergeneric remodeled bacteria comprises at least one genetic variation introduced into at least one gene, or non-coding polynucleotide, of the nitrogen fixation or assimilation genetic regulatory network that results in one or more of: increased expression or activity of NifA or glutaminase; decreased expression or activity of NifL, NtrB, glutamine synthetase, GlnB, GlnK, DraT, AmtB; decreased adenylyl-removing activity of GlnE; or decreased uridylyl-removing activity of GlnD.


31. The seed treatment composition, according to any one of embodiments 1-30, wherein each member of the plurality of non-intergeneric remodeled bacteria comprises a mutated nifL gene that has been altered to comprise a heterologous promoter inserted into said nifL gene.


32. The seed treatment composition, according to any one of embodiments 1-31, wherein each member of the plurality of non-intergeneric remodeled bacteria comprises a mutated glnE gene that results in a truncated GlnE protein lacking an adenylyl-removing (AR) domain.


33. The seed treatment composition, according to any one of embodiments 1-32, wherein each member of the plurality of non-intergeneric remodeled bacteria comprises a mutated amtB gene that results in the lack of expression of said amtB gene.


34. The seed treatment composition, according to any one of embodiments 1-33, wherein each member of the plurality of non-intergeneric remodeled bacteria comprises at least one of: a mutated nifL gene that has been altered to comprise a heterologous promoter inserted into said nifL gene; a mutated glnE gene that results in a truncated GlnE protein lacking an adenylyl-removing (AR) domain; a mutated amtB gene that results in the lack of expression of said amtB gene; and combinations thereof.


35. The seed treatment composition, according to any one of embodiments 1-34, wherein each member of the plurality of non-intergeneric remodeled bacteria comprises a mutated nifL gene that has been altered to comprise a heterologous promoter inserted into said nifL gene and a mutated glnE gene that results in a truncated GlnE protein lacking an adenylyl-removing (AR) domain.


36. The seed treatment composition, according to any one of embodiments 1-35, wherein each member of the plurality of non-intergeneric remodeled bacteria comprises a mutated nifL gene that has been altered to comprise a heterologous promoter inserted into said nifL gene and a mutated glnE gene that results in a truncated GlnE protein lacking an adenylyl-removing (AR) domain and a mutated amtB gene that results in the lack of expression of said amtB gene.


37. The seed treatment composition, according to any one of embodiments 1-36, wherein the plurality of non-intergeneric remodeled bacteria are present at a concentration of about 1×105 to about 1×107 cfu per seed.


38. The seed treatment composition, according to any one of embodiments 1-37, wherein the plurality of non-intergeneric remodeled bacteria comprise at least two different species of bacteria.


39. The seed treatment composition, according to any one of embodiments 1-38, wherein the plurality of non-intergeneric remodeled bacteria comprise at least two different strains of the same species of bacteria.


40. The seed treatment composition, according to any one of embodiments 1-39, wherein the plurality of non-intergeneric remodeled bacteria comprise bacteria selected from: Rahnella aquatilis, Klebsiella variicola, Achromobacter spiritinus, Achromobacter marplatensis, Microbacterium murale, Kluyvera intermedia, Kosakonia pseudosacchari, Enterobacter sp., Azospirillum lipoferum, Kosakonia sacchari, and combinations thereof.


41. The seed treatment composition, according to any one of embodiments 1-40, wherein the plurality of non-intergeneric remodeled bacteria are endophytic, epiphytic, or rhizospheric.


42. The seed treatment composition, according to any one of embodiments 1-41, wherein the plurality of non-intergeneric remodeled bacteria comprise bacteria selected from: a bacteria deposited as NCMA 201701002, a bacteria deposited as NCMA 201708004, a bacteria deposited as NCMA 201708003, a bacteria deposited as NCMA 201708002, a bacteria deposited as NCMA 201712001, a bacteria deposited as NCMA 201712002, and combinations thereof.


43. The seed treatment composition, according to any one of embodiments 1-42, wherein the plurality of non-intergeneric remodeled bacteria comprise bacteria with a nucleic acid sequence that shares at least about 90% sequence identity to a nucleic acid sequence selected from SEQ ID NOs: 177-260, 296-303.


44. The seed treatment composition, according to any one of embodiments 1-43, wherein the plurality of non-intergeneric remodeled bacteria comprise bacteria with a nucleic acid sequence that shares at least about 95% sequence identity to a nucleic acid sequence selected from SEQ ID NOs: 177-260, 296-303.


45. The seed treatment composition, according to any one of embodiments 1-44, wherein the plurality of non-intergeneric remodeled bacteria comprise bacteria with a nucleic acid sequence that shares at least about 99% sequence identity to a nucleic acid sequence selected from SEQ ID NOs: 177-260, 296-303.


46. The seed treatment composition, according to any one of embodiments 1-45, wherein the plurality of non-intergeneric remodeled bacteria comprise bacteria with a nucleic acid sequence selected from SEQ ID NOs: 177-260, 296-303.


While preferred embodiments of the present disclosure have been shown and described herein, it will be obvious to those skilled in the art that such embodiments are provided by way of example only. Numerous variations, changes, and substitutions will now occur to those skilled in the art without departing from the disclosure. It should be understood that various alternatives to the embodiments of the disclosure described herein may be employed in practicing the disclosure. It is intended that the following Claims define the scope of the disclosure and that methods and structures within the scope of these claims and their equivalents be covered thereby.


INCORPORATION BY REFERENCE

All references, articles, publications, patents, patent publications, and patent applications cited herein are incorporated by reference in their entireties for all purposes. However, mention of any reference, article, publication, patent, patent publication, and patent application cited herein is not, and should not be taken as, an acknowledgment or any form of suggestion that they constitute valid prior art or form part of the common general knowledge in any country in the world. Further, U.S. Pat. No. 9,975,817, issued on May 22, 2018, and entitled: Methods and Compositions for Improving Plant Traits, is hereby incorporated by reference. Further, PCT/US2018/013671, filed Jan. 12, 2018, and entitled: Methods and Compositions for Improving Plant Traits, is hereby incorporated by reference.

Claims
  • 1. A seed treatment composition, comprising: a. a plurality of non-intergeneric remodeled bacteria that have an average colonization ability per unit of plant root tissue of at least about 1.0×107 bacterial cells per gram of fresh weight of plant root tissue and produce fixed N of at least about 1×10−17 mmol N per bacterial cell per hour; andb. at least one pesticide;wherein said pesticide is selected from the group consisting of fludioxonil, metalaxyl, mefenoxam, azoxystrobin, thiabendazole, ipconazole, tebuconazole, prothioconazole, imidacloprid, clothianidin, thiamethoxam, chlorantraniliprole, Bacillus pumilus, Bacillus subtilis, captan, hydrogen peroxide, mancozeb, pyraclostrobin, Streptomyces griseoviridis, Streptomyces lydicus, thiram, Trichoderma harzianum Rifai, trifloxystrobin, chlorpyrifos, abamectin, Bacillus firmus, harpin alpha beta protein, indole butyric acid, carboxin, permethrin, triticonazole, ethaboxam, carboxyanilide, broflanilide, and combinations thereof;wherein each member of the plurality of non-intergeneric remodeled bacteria comprises at least one genetic variation introduced into a member selected from the group consisting of: nifA, nifL, ntrB, ntrC, polynucleotide encoding glutamine synthetase, glnA, glnB, glnK, draT, amtB, polynucleotide encoding glutaminase, glnD, glnE, nifJ, nifH, nifD, nifK, nifY, nifE, nifN, nifU, nifS, nifV, nifW, nifZ, nifM, nifF, nifB, nifQ, a gene associated with biosynthesis of a nitrogenase enzyme, and combinations thereof; and wherein the plurality of non-intergeneric remodeled bacteria comprise non-intergeneric remodeled bacteria selected from the group consisting of: Rahnella aquatilis, Klebsiella variicola, Achromobacter spiritinus, Achromobacter marplatensis, Microbacterium murale, Kluyvera intermedia, Kosakonia pseudosacchari, Enterobacter sp., Azospirillum lipoferum, Kosakonia sacchari, and combinations thereof.
  • 2. The seed treatment composition according to claim 1, further comprising a seed.
  • 3. The seed treatment composition according to claim 2, wherein said seed is selected from the group consisting of a seed from the family Poaceae, a cereal seed, a rice seed, a wheat seed, a barley seed, a Sorghum seed, a millet seed, an oat seed, a rye seed, a triticale seed, a genetically modified corn seed, a non-genetically modified corn seed, sweet corn seed, flint corn seed, popcorn seed, dent corn seed, pod corn seed, and flour corn seed.
  • 4. The seed treatment composition according to claim 1, further comprising a genetically modified corn seed.
  • 5. The seed treatment composition according to claim 4, wherein said genetically modified corn seed comprises a trait selected from the group consisting of a herbicide tolerant trait, an insect resistance trait, and combinations thereof.
  • 6. The seed treatment composition according to claim 1, wherein the plurality of non-intergeneric remodeled bacteria produces 1% or more of the fixed nitrogen in a plant exposed thereto.
  • 7. The seed treatment composition according to claim 1, wherein the plurality of non-intergeneric remodeled bacteria is capable of fixing atmospheric nitrogen in the presence of exogenous nitrogen.
  • 8. The seed treatment composition according to claim 1, wherein the each member of the plurality of non-intergeneric remodeled bacteria comprises at least one genetic variation introduced into at least one gene or non-coding polynucleotide of a nitrogen fixation or assimilation genetic regulatory network.
  • 9. The seed treatment composition according to claim 1, wherein the each member of the plurality of non-intergeneric remodeled bacteria comprises an introduced control sequence operably linked to at least one gene of a nitrogen fixation or assimilation genetic regulatory network.
  • 10. The seed treatment composition according to claim 1, wherein the each member of the plurality of non-intergeneric remodeled bacteria comprises a heterologous promoter operably linked to at least one gene of a nitrogen fixation or assimilation genetic regulatory network.
  • 11. The seed treatment composition according to claim 1, wherein the each member of the plurality of non-intergeneric remodeled bacteria comprises at least one genetic variation introduced into at least one gene or non-coding polynucleotide of a nitrogen fixation or assimilation genetic regulatory network that results in one or more of: increased expression or activity of NifA or glutaminase; decreased expression or activity of NifL, NtrB, glutamine synthetase, GlnB, GlnK, DraT, AmtB; decreased adenylyl-removing activity of GlnE; or decreased uridylyl-removing activity of GlnD.
  • 12. The seed treatment composition according to claim 1, wherein the each member of the plurality of non-intergeneric remodeled bacteria comprises at least one of: a mutated nifL gene that has been altered to comprise a heterologous promoter inserted into said nifL gene; a mutated glnE gene that results in a truncated GlnE protein lacking an adenylyl-removing (AR) domain; a mutated amtB gene that results in the lack of expression of said amtB gene; and combinations thereof.
  • 13. The seed treatment composition according to claim 1, wherein the plurality of non-intergeneric remodeled bacteria is present at a concentration of about 1×105 to about 1×107 colony forming units (cfu) per seed.
  • 14. The seed treatment composition according to claim 1, wherein the plurality of non-intergeneric remodeled bacteria comprises at least two different species of non-intergeneric remodeled bacteria.
  • 15. The seed treatment composition according to claim 1, wherein the plurality of non-intergeneric remodeled bacteria comprises at least two different strains of the same species of non-intergeneric remodeled bacteria.
  • 16. The seed treatment composition according to claim 1, wherein the plurality of non-intergeneric remodeled bacteria is endophytic, epiphytic, or rhizospheric.
  • 17. The seed treatment composition according to claim 1, wherein the plurality of non-intergeneric remodeled bacteria comprises a non-intergeneric remodeled Klebsiella variicola bacteria deposited as NCMA 201712002.
  • 18. The seed treatment composition according to claim 1, wherein the plurality of non-intergeneric remodeled bacteria comprises a non-intergeneric remodeled bacteria comprising a nucleic acid sequence that shares at least about 90% sequence identity to a nucleic acid sequence selected from SEQ ID NOs: 177-260, 296-303.
  • 19. The seed treatment composition of claim 1, wherein the plurality of non-intergeneric remodeled bacteria produces greater than 1 pound of fixed N per acre per season.
  • 20. A corn seed treatment composition, comprising: a. a plurality of non-intergeneric remodeled bacteria that have an average colonization ability per unit of plant root tissue of at least about 1.0×107 bacterial cells per gram of fresh weight of plant root tissue and produce fixed N of at least about 1×10−17 mmol N per bacterial cell per hour; andb. at least one pesticide selected from the group consisting of: clothianidin, thiamethoxam, chlorantraniliprole, imidacloprid, fludioxonil, mefenoxam, azoxystrobin, thiabendazole, metalaxyl, ipconazole, tebuconazole, prothioconazole, and combinations thereof;wherein each member of the plurality of non-intergeneric remodeled bacteria comprises at least one genetic variation introduced into a member selected from the group consisting of: nifA, nifL, ntrB, ntrC, polynucleotide encoding glutamine synthetase, glnA, glnB, glnK, draT, amtB, polynucleotide encoding glutaminase, glnD, glnE, nifJ, nifH, nifD, nifK, nifY, nifE, nifN, nifU, nifS, nifV, nifW, nifZ, nifM, nifF, nifB, nifQ, a gene associated with biosynthesis of a nitrogenase enzyme, and combinations thereof.
  • 21. The corn seed treatment composition according to claim 20, further comprising a corn seed.
  • 22. The corn seed treatment composition according to claim 20, further comprising a genetically modified corn seed.
  • 23. The corn seed treatment composition according to claim 22, wherein said genetically modified corn seed comprises a trait selected from the group consisting of a herbicide tolerant trait, an insect resistance trait, and combinations thereof.
  • 24. The corn seed treatment composition according to claim 20, wherein the plurality of non-intergeneric remodeled bacteria produces 1% or more of the fixed nitrogen in a plant exposed thereto.
  • 25. The corn seed treatment composition according to claim 20, wherein the plurality of non-intergeneric remodeled bacteria is capable of fixing atmospheric nitrogen in the presence of exogenous nitrogen.
  • 26. The corn seed treatment composition according to claim 20, wherein the each member of the plurality of non-intergeneric remodeled bacteria comprises at least one genetic variation introduced into at least one gene or non-coding polynucleotide of a nitrogen fixation or assimilation genetic regulatory network.
  • 27. The corn seed treatment composition according to claim 20, wherein the each member of the plurality of non-intergeneric remodeled bacteria comprises an introduced control sequence operably linked to at least one gene of a nitrogen fixation or assimilation genetic regulatory network.
  • 28. The corn seed treatment composition according to claim 20, wherein the each member of the plurality of non-intergeneric remodeled bacteria comprises a heterologous promoter operably linked to at least one gene of a nitrogen fixation or assimilation genetic regulatory network.
  • 29. The corn seed treatment composition according to claim 20, wherein the each member of the plurality of non-intergeneric remodeled bacteria comprises at least one genetic variation introduced into at least one gene, or non-coding polynucleotide, of a nitrogen fixation or assimilation genetic regulatory network that results in one or more of: increased expression or activity of NifA or glutaminase; decreased expression or activity of NifL, NtrB, glutamine synthetase, GlnB, GlnK, DraT, AmtB; decreased adenylyl-removing activity of GlnE; or decreased uridylyl-removing activity of GlnD.
  • 30. The seed treatment composition according to claim 20, wherein the each member of the plurality of non-intergeneric remodeled bacteria comprises at least one of: a mutated nifL gene that has been altered to comprise a heterologous promoter inserted into said nifL gene; a mutated glnE gene that results in a truncated GlnE protein lacking an adenylyl-removing (AR) domain; a mutated amtB gene that results in the lack of expression of said amtB gene; and combinations thereof.
  • 31. The seed treatment composition according to claim 20, wherein the plurality of non-intergeneric remodeled bacteria is present at a concentration of about 1×105 to about 1×107 cfu per seed.
  • 32. The seed treatment composition according to claim 20, wherein the plurality of non-intergeneric remodeled bacteria comprises at least two different species of the non-intergeneric remodeled bacteria.
  • 33. The seed treatment composition according to claim 20, wherein the plurality of non-intergeneric remodeled bacteria comprises at least two different strains of the same species of the non-intergeneric remodeled bacteria.
  • 34. The seed treatment composition according to claim 20, wherein the plurality of non-intergeneric remodeled bacteria comprises non-intergeneric remodeled bacteria selected from: Rahnella aquatilis, Klebsiella variicola, Achromobacter spiritinus, Achromobacter marplatensis, Microbacterium murale, Kluyvera intermedia, Kosakonia pseudosacchari, Enterobacter sp., Azospirillum lipoferum, Kosakonia sacchari, and combinations thereof.
  • 35. The corn seed treatment composition according to claim 20, wherein the plurality of non-intergeneric remodeled bacteria is endophytic, epiphytic, or rhizospheric.
  • 36. The corn seed treatment composition according to claim 20, wherein the plurality of non-intergeneric remodeled bacteria comprises a non-intergeneric remodeled Klebsiella variicola bacteria deposited as NCMA 201712002.
  • 37. The corn seed treatment composition according to claim 20, wherein the plurality of non-intergeneric remodeled bacteria comprises a non-intergeneric remodeled bacteria comprising a nucleic acid sequence that shares at least about 90% sequence identity to a nucleic acid sequence selected from SEQ ID NOs: 177-260, 296-303.
  • 38. The corn seed treatment composition of claim 20, wherein the plurality of non-intergeneric remodeled bacteria produces greater than 1 pound of fixed N per acre per season.
  • 39. A corn seed treatment composition, comprising: a. a plurality of non-intergeneric remodeled bacteria that have an average colonization ability per unit of plant root tissue of at least about 1.0×107 bacterial cells per gram of fresh weight of plant root tissue and produce fixed N of at least about 1×10−17 mmol N per bacterial cell per hour; andb. at least one pesticide selected from the group consisting of: clothianidin, thiamethoxam, chlorantraniliprole, imidacloprid, fludioxonil, mefenoxam, azoxystrobin, thiabendazole, metalaxyl, ipconazole, tebuconazole, prothioconazole, and combinations thereof;wherein the plurality of non-intergeneric remodeled bacteria comprise non-intergeneric remodeled bacteria selected from the group consisting of: Rahnella aquatilis, Klebsiella variicola, Achromobacter spiritinus, Achromobacter marplatensis, Microbacterium murale, Kluyvera intermedia, Kosakonia pseudosacchari, Enterobacter sp., Azospirillum lipoferum, Kosakonia sacchari, and combinations thereof.
  • 40. The corn seed treatment composition according to claim 39, further comprising a corn seed.
  • 41. The corn seed treatment composition according to claim 39, further comprising a genetically modified corn seed.
  • 42. The corn seed treatment composition according to claim 41, wherein said genetically modified corn seed comprises a trait selected from the group consisting of a herbicide tolerant trait, an insect resistance trait, and combinations thereof.
  • 43. The corn seed treatment composition according to claim 39, wherein the plurality of non-intergeneric remodeled bacteria produces 1% or more of the fixed nitrogen in a plant exposed thereto.
  • 44. The corn seed treatment composition according to claim 39, wherein the plurality of non-intergeneric remodeled bacteria is capable of fixing atmospheric nitrogen in the presence of exogenous nitrogen.
  • 45. The corn seed treatment composition according to claim 39, wherein each member of the plurality of non-intergeneric remodeled bacteria comprises at least one genetic variation introduced into at least one gene or non-coding polynucleotide of a nitrogen fixation or assimilation genetic regulatory network.
  • 46. The corn seed treatment composition according to claim 39, wherein each member of the plurality of non-intergeneric remodeled bacteria comprises an introduced control sequence operably linked to at least one gene of a nitrogen fixation or assimilation genetic regulatory network.
  • 47. The corn seed treatment composition according to claim 39, wherein each member of the plurality of non-intergeneric remodeled bacteria comprises a heterologous promoter operably linked to at least one gene of a nitrogen fixation or assimilation genetic regulatory network.
  • 48. The corn seed treatment composition according to claim 39, wherein each member of the plurality of non-intergeneric remodeled bacteria comprises at least one genetic variation introduced into a member selected from the group consisting of: nifA, nifL, ntrB, ntrC, polynucleotide encoding glutamine synthetase, glnA, glnB, glnK, draT, amtB, polynucleotide encoding glutaminase, glnD, glnE, nifJ, nifH, nifD, nifK, nifY, nifE, nifN, nifU, nifS, nifV, nifW, nifZ, nifM, nifF, nifB, nifQ, a gene associated with biosynthesis of a nitrogenase enzyme, and combinations thereof.
  • 49. The corn seed treatment composition according to claim 39, wherein each member of the plurality of non-intergeneric remodeled bacteria comprises at least one genetic variation introduced into at least one gene or non-coding polynucleotide of a nitrogen fixation or assimilation genetic regulatory network that results in one or more of: increased expression or activity of NifA or glutaminase; decreased expression or activity of NifL, NtrB, glutamine synthetase, GlnB, GlnK, DraT, AmtB; decreased adenylyl-removing activity of GlnE; or decreased uridylyl-removing activity of GlnD.
  • 50. The corn seed treatment composition according to claim 39, wherein each member of the plurality of non-intergeneric remodeled bacteria comprises at least one of: a mutated nifL gene that has been altered to comprise a heterologous promoter inserted into said nifL gene; a mutated glnE gene that results in a truncated GlnE protein lacking an adenylyl-removing (AR) domain; a mutated amtB gene that results in the lack of expression of said amtB gene; and combinations thereof.
  • 51. The corn seed treatment composition according to claim 39, wherein the plurality of non-intergeneric remodeled bacteria is present at a concentration of about 1×105 to about 1×107 cfu per seed.
  • 52. The corn seed treatment composition according to claim 39, wherein the plurality of non-intergeneric remodeled bacteria comprises at least two different species of bacteria.
  • 53. The corn seed treatment composition according to claim 39, wherein the plurality of non-intergeneric remodeled bacteria comprises at least two different strains of the same species of bacteria.
  • 54. The corn seed treatment composition according to claim 39, wherein the plurality of non-intergeneric remodeled bacteria is endophytic, epiphytic, or rhizospheric.
  • 55. The corn seed treatment composition according to claim 39, wherein the plurality of non-intergeneric remodeled bacteria comprises a non-intergeneric remodeled Klebsiella variicola bacteria deposited as NCMA 201712002.
  • 56. The corn seed treatment composition according to claim 39, wherein the plurality of non-intergeneric remodeled bacteria comprises non-intergeneric remodeled bacteria comprising a nucleic acid sequence that shares at least about 90% sequence identity to a nucleic acid sequence selected from SEQ ID NOs: 177-260, 296-303.
  • 57. The corn seed treatment composition of claim 39, wherein the plurality of non-intergeneric remodeled bacteria produces greater than 1 pound of fixed N per acre per season.
CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims the benefit of International PCT Application No. PCT/US2019/039217, filed Jun. 26, 2019, which claims priority to U.S. Provisional Application No. 62/690,621, filed on Jun. 27, 2018 and U.S. Provisional Application No. 62/808,693, filed on Feb. 21, 2019. These applications are hereby incorporated by reference in their entirety for all purposes

PCT Information
Filing Document Filing Date Country Kind
PCT/US2019/039217 6/26/2019 WO
Publishing Document Publishing Date Country Kind
WO2020/006064 1/2/2020 WO A
US Referenced Citations (209)
Number Name Date Kind
1520545 Murphy Dec 1924 A
4782022 Puhler et al. Nov 1988 A
4832728 Allan et al. May 1989 A
5071743 Slilaty et al. Dec 1991 A
5188960 Payne et al. Feb 1993 A
5229291 Nielsen et al. Jul 1993 A
5354670 Nickoloff et al. Oct 1994 A
5427785 Ronson et al. Jun 1995 A
5610044 Lam Mar 1997 A
5780270 Lesley Jul 1998 A
5789166 Bauer et al. Aug 1998 A
5877012 Estruch et al. Mar 1999 A
5916029 Smith et al. Jun 1999 A
6033861 Schafer et al. Mar 2000 A
6033874 Baum et al. Mar 2000 A
6083499 Narva et al. Jul 2000 A
6107279 Estruch et al. Aug 2000 A
6114148 Seed et al. Sep 2000 A
6127180 Narva et al. Oct 2000 A
6137033 Estruch et al. Oct 2000 A
6218188 Cardineau et al. Apr 2001 B1
6248535 Danenberg et al. Jun 2001 B1
6326351 Donovan et al. Dec 2001 B1
6340593 Cardineau et al. Jan 2002 B1
6391548 Bauer et al. May 2002 B1
6399330 Donovan et al. Jun 2002 B1
6548289 Beynon et al. Apr 2003 B1
6548291 Narva et al. Apr 2003 B1
6596509 Bauer et al. Jul 2003 B1
6624145 Narva et al. Sep 2003 B1
6673610 Miyawaki et al. Jan 2004 B2
6713285 Bauer et al. Mar 2004 B2
6773900 Short et al. Aug 2004 B2
6841358 Locht et al. Jan 2005 B1
6949626 Donovan et al. Sep 2005 B2
6962705 Malvar et al. Nov 2005 B2
7064249 Corbin et al. Jun 2006 B2
7070982 Malvar et al. Jul 2006 B2
7084331 Isawa et al. Aug 2006 B2
7105332 Abad et al. Sep 2006 B2
7132265 Bauer et al. Nov 2006 B2
7244820 Miles et al. Jul 2007 B2
7329736 Abad et al. Feb 2008 B2
7378499 Abad et al. May 2008 B2
7385107 Donovan et al. Jun 2008 B2
7449552 Abad et al. Nov 2008 B2
7462760 Abad et al. Dec 2008 B2
7470427 Cocking Dec 2008 B2
7476781 Abad et al. Jan 2009 B2
7485451 VanderGheynst et al. Feb 2009 B2
7491698 Hey et al. Feb 2009 B2
7491869 Abad et al. Feb 2009 B2
7504229 Donovan et al. Mar 2009 B2
7615686 Miles et al. Nov 2009 B2
7803943 Mao et al. Sep 2010 B2
7858849 Cerf et al. Dec 2010 B2
8076142 Huang et al. Dec 2011 B2
8084416 Sampson et al. Dec 2011 B2
8084418 Hey et al. Dec 2011 B2
8137665 Cocking Mar 2012 B2
8236757 Carozzi et al. Aug 2012 B2
8237020 Miles et al. Aug 2012 B2
8268584 Harwood et al. Sep 2012 B1
8304604 Lira et al. Nov 2012 B2
8304605 Lira et al. Nov 2012 B2
8319019 Abad et al. Nov 2012 B2
8334366 Hughes et al. Dec 2012 B1
8334431 Sampson et al. Dec 2012 B2
8377671 Cournac et al. Feb 2013 B2
8481026 Woodruff et al. Jul 2013 B1
8513494 Wu et al. Aug 2013 B2
8530411 Cerf et al. Sep 2013 B2
8575433 Cerf et al. Nov 2013 B2
8686233 Cerf et al. Apr 2014 B2
8759619 Sampson et al. Jun 2014 B2
8795965 Zhang Aug 2014 B2
8802933 Abad et al. Aug 2014 B2
8802934 Abad et al. Aug 2014 B2
9150851 Wigley et al. Oct 2015 B2
9321697 Das et al. Apr 2016 B2
9487451 Doty et al. Nov 2016 B2
9512431 Mirsky et al. Dec 2016 B2
9657298 Soto, Sr. et al. May 2017 B2
9957509 Mirsky et al. May 2018 B2
9975817 Temme May 2018 B2
9994557 Davidson et al. Jun 2018 B2
10384983 Temme et al. Aug 2019 B2
10525318 Dougherty Jan 2020 B2
10556839 Temme et al. Feb 2020 B2
10577358 Davidson et al. Mar 2020 B2
10602744 Wigley Mar 2020 B2
10662432 Mirsky et al. May 2020 B2
10918106 Bayer et al. Feb 2021 B2
10919814 Temme Feb 2021 B2
10934226 Temme Mar 2021 B2
10968446 Zhao et al. Apr 2021 B2
11479516 Voigt et al. Oct 2022 B2
11565979 Temme et al. Jan 2023 B2
11739032 Temme et al. Aug 2023 B2
20040197916 Carozzi et al. Oct 2004 A1
20040197917 Carozzi et al. Oct 2004 A1
20040210964 Carozzi et al. Oct 2004 A1
20040210965 Carozzi et al. Oct 2004 A1
20040216186 Carozzi et al. Oct 2004 A1
20040235663 Cocking Nov 2004 A1
20040241847 Okuyama et al. Dec 2004 A1
20050081262 Cook et al. Apr 2005 A1
20050266541 Dillon Dec 2005 A1
20060033867 Krisko et al. Feb 2006 A1
20060127988 Wood et al. Jun 2006 A1
20060191034 Baum Aug 2006 A1
20060243011 Someus Nov 2006 A1
20080295207 Baum et al. Nov 2008 A1
20080311632 Figge et al. Dec 2008 A1
20090068159 Baum et al. Mar 2009 A1
20090105076 Stewart et al. Apr 2009 A1
20090137390 Triplett May 2009 A1
20090144852 Tomso et al. Jun 2009 A1
20090162477 Nadel et al. Jun 2009 A1
20090258404 Mikkelsen et al. Oct 2009 A1
20090308121 Reddy et al. Dec 2009 A1
20100005543 Sampson et al. Jan 2010 A1
20100017914 Hart et al. Jan 2010 A1
20100028870 Welch et al. Feb 2010 A1
20100267147 Qiao Oct 2010 A1
20100298211 Carozzi et al. Nov 2010 A1
20110023184 Desai et al. Jan 2011 A1
20110104690 Yu et al. May 2011 A1
20110183347 Hyde et al. Jul 2011 A1
20110263488 Carozzi et al. Oct 2011 A1
20120015806 Paikray et al. Jan 2012 A1
20120107889 Doty et al. May 2012 A1
20120192605 McSpadden Gardener et al. Aug 2012 A1
20120266332 Kuykendall Oct 2012 A1
20120278954 Bowen et al. Nov 2012 A1
20120284813 Olivier et al. Nov 2012 A1
20120311745 Meade et al. Dec 2012 A1
20120311746 Meade et al. Dec 2012 A1
20120317681 Meade et al. Dec 2012 A1
20120317682 Meade et al. Dec 2012 A1
20120324605 Meade et al. Dec 2012 A1
20120324606 Meade et al. Dec 2012 A1
20120331589 Meade et al. Dec 2012 A1
20120331590 Meade et al. Dec 2012 A1
20130116170 Graser et al. May 2013 A1
20130126428 Jones et al. May 2013 A1
20130167268 Narva et al. Jun 2013 A1
20130167269 Narva et al. Jun 2013 A1
20140011261 Wang et al. Jan 2014 A1
20140155283 Venkateswaran et al. Jun 2014 A1
20140182018 Lang et al. Jun 2014 A1
20140223598 Sampson et al. Aug 2014 A1
20140223599 Sampson et al. Aug 2014 A1
20140230504 Finlayson et al. Aug 2014 A1
20140273226 Wu Sep 2014 A1
20140301990 Gregory et al. Oct 2014 A1
20140329326 Mirsky et al. Nov 2014 A1
20140336050 Soto, Sr. et al. Nov 2014 A1
20150080261 Wigley et al. Mar 2015 A1
20150101373 Munusamy et al. Apr 2015 A1
20150128670 Das May 2015 A1
20150237807 Valiquette Aug 2015 A1
20150239789 Kang et al. Aug 2015 A1
20150274690 Davidson et al. Oct 2015 A1
20150315570 Zhao et al. Nov 2015 A1
20160174570 Vujanovic et al. Jun 2016 A1
20160264929 Barney et al. Sep 2016 A1
20160292355 Lou et al. Oct 2016 A1
20160295868 Jones Oct 2016 A1
20170152519 Mirsky et al. Jun 2017 A1
20170367349 Gruver Dec 2017 A1
20180002243 Temme Jan 2018 A1
20180020671 Wigley et al. Jan 2018 A1
20180065896 Van Iersel et al. Mar 2018 A1
20180073028 Mirsky et al. Mar 2018 A1
20180290942 Voigt et al. Oct 2018 A1
20180297906 Temme Oct 2018 A1
20190039964 Temme Feb 2019 A1
20190144352 Temme May 2019 A1
20190339964 Young et al. Nov 2019 A1
20200087221 Temme Mar 2020 A1
20200115715 Mirsky et al. Apr 2020 A1
20200299637 Voigt et al. Sep 2020 A1
20200308594 Tamsir et al. Oct 2020 A1
20200331820 Tamsir Oct 2020 A1
20210009483 Temme et al. Jan 2021 A1
20210163374 Bloch Jun 2021 A1
20210214282 Temme Jul 2021 A1
20210284995 Zhao et al. Sep 2021 A1
20210315212 Rezaei et al. Oct 2021 A1
20210329917 Bayer et al. Oct 2021 A1
20210345618 Bloch et al. Nov 2021 A1
20220017911 Temme et al. Jan 2022 A1
20220025389 Bayer et al. Jan 2022 A1
20220090095 Higgins et al. Mar 2022 A1
20220106238 Rezaei et al. Apr 2022 A1
20220127627 Bloch et al. Apr 2022 A1
20220132861 Reisinger et al. May 2022 A1
20220151241 Reisinger et al. May 2022 A1
20220162544 Voigt et al. May 2022 A1
20220211048 Temme et al. Jul 2022 A1
20220282340 Ryu et al. Sep 2022 A1
20220396530 Tamsir et al. Dec 2022 A1
20220411344 Voigt et al. Dec 2022 A1
20230019267 Hapes et al. Jan 2023 A1
20230033451 Reisinger et al. Feb 2023 A1
20230062568 Temme et al. Mar 2023 A1
20230276807 Reisinger et al. Sep 2023 A1
20230295559 Eskiyenenturk et al. Sep 2023 A1
Foreign Referenced Citations (77)
Number Date Country
2051071 Mar 1993 CA
1355294 Jun 2002 CN
WO 8704182 Jul 1987 WO
WO 9305154 Mar 1993 WO
WO 9801088 Jan 1998 WO
WO 9909834 Mar 1999 WO
WO 0057183 Sep 2000 WO
WO 0107567 Feb 2001 WO
WO 2004074462 Sep 2004 WO
WO 2005021585 Mar 2005 WO
WO 2005038032 Apr 2005 WO
WO 2006005100 Jan 2006 WO
WO 2006083891 Aug 2006 WO
WO 2006119457 Nov 2006 WO
WO-2007027776 Mar 2007 WO
WO 2009060012 May 2009 WO
WO 2009091557 Jul 2009 WO
WO 2011103247 Aug 2011 WO
WO 2011103248 Aug 2011 WO
WO 2011099019 Aug 2011 WO
WO 2011099024 Aug 2011 WO
WO 2011154960 Dec 2011 WO
WO 2012139004 Oct 2012 WO
WO 2012174271 Dec 2012 WO
WO 2013076687 May 2013 WO
WO 2013132518 Sep 2013 WO
WO 2014042517 Mar 2014 WO
WO 2014071182 May 2014 WO
WO 2014201044 Dec 2014 WO
WO 2015061764 Apr 2015 WO
WO 2016016629 Feb 2016 WO
WO 2016016630 Feb 2016 WO
WO 2016100727 Jun 2016 WO
WO-2016130586 Aug 2016 WO
WO 2016146955 Sep 2016 WO
WO-2016172655 Oct 2016 WO
WO 2016178580 Nov 2016 WO
WO 2016179046 Nov 2016 WO
WO 2016181228 Nov 2016 WO
WO 2016191828 Dec 2016 WO
WO 2017011602 Jan 2017 WO
WO 2017042833 Mar 2017 WO
WO 2017062412 Apr 2017 WO
WO 2017069717 Apr 2017 WO
WO 2018132774 Jul 2018 WO
WO 2019032926 Feb 2019 WO
WO 2019084059 May 2019 WO
WO 2019084342 May 2019 WO
WO 2019140125 Jul 2019 WO
WO 2020006064 Jan 2020 WO
WO 2020006246 Jan 2020 WO
WO 2020014498 Jan 2020 WO
WO 2020061363 Mar 2020 WO
WO 2020068946 Apr 2020 WO
WO 2020092940 May 2020 WO
WO 2020118111 Jun 2020 WO
WO 2020132632 Jun 2020 WO
WO 2020146372 Jul 2020 WO
WO 2020163251 Aug 2020 WO
WO 2020191072 Sep 2020 WO
WO 2020190363 Sep 2020 WO
WO 2020191201 Sep 2020 WO
WO 2020219893 Oct 2020 WO
WO 2020219932 Oct 2020 WO
WO 2021113352 Jun 2021 WO
WO 2021146209 Jul 2021 WO
WO 2021221689 Nov 2021 WO
WO 2021221690 Nov 2021 WO
WO 2021222567 Nov 2021 WO
WO 2021222643 Nov 2021 WO
WO 2021231449 Nov 2021 WO
WO 2021247915 Dec 2021 WO
WO 2022029661 Feb 2022 WO
WO 2022140656 Jun 2022 WO
WO-2022260676 Dec 2022 WO
WO-2022261433 Dec 2022 WO
WO-2023278804 Jan 2023 WO
Non-Patent Literature Citations (739)
Entry
U.S. Pat. No. 8,476,226, Nov. 9, 1999, Koenck (Withdrawn).
Baum, et al. “Control of coleopteran insect pests through RNA interference.” Nat Biotechnol. Nov. 2007;25(11):1322-6. doi: 10.1038/nbt1359.
Bosmans, et al. “Sea anemone venom as a source of insecticidal peptides acting on voltage-gated Na+ channels.” Toxicon. Mar. 15, 2007;49(4):550-60. doi: 10.1016/j.toxicon.2006.11.029.
Chakroun, et al. “Bacterial Vegetative Insecticidal Proteins (Vip) from Entomopathogenic Bacteria.” Microbiol Mol Biol Rev. Mar. 2, 2016;80(2):329-50. doi: 10.1128/MMBR.00060-15.
Chiang, et al. “Mutagenic oligonucleotide-directed PCR amplification (Mod-PCR): an efficient method for generating random base substitution mutations in a DNA sequence element.” PCR Methods Appl. Feb. 1993;2(3):210-7. doi: 10.1101/gr.2.3.210.
Crickmore, et al. “Revision of the nomenclature for the Bacillus thuringiensis pesticidal crystal proteins.” Microbiol Mol Biol Rev. Sep. 1998;62(3):807-13.
Crickmore, et al. “Bacillus thuringiensis toxin nomenclature.” 2016, available at: http://www.lifesci.sussex.ac.uk/home/Neil_Crickmore/Bt/.
Das, et al. “Microbial assay of N 2 fixation rate, a simple alternate for acetylene reduction assay.” MethodsX. Jan. 6, 2018;5:909-914. doi: 10.1016/j.mex.2017.11.010.
Eyraud, et al. “Expression and biological activity of the cystine knot bioinsecticide PA1b (Pea Albumin 1 Subunit b).” PLoS One. Dec. 11, 2013;8(12):e81619. doi: 10.1371/journal.pone.0081619.
Gaby, et al. “A comprehensive aligned nifH gene database: a multipurpose tool for studies of nitrogen-fixing bacteria.” Database (Oxford). Feb. 5, 2014;2014:bau001. doi: 10.1093/database/bau001.
GenBank: CP007215.3. Kosakonia sacchari SP1 chromosome, complete genome. Sep. 19, 2017.
GenBank: AAA22353.1. crystal protein [Bacillus thuringiensis]. Apr. 26, 1993.
GenBank: AAA22552.1. insecticidal crystal protein, partial (plasmid) [Bacillus thuringiensis]. Dec. 3, 2020.
GenBank: BAA00257.1. unnamed protein product [Bacillus thuringiensis serovar aizawai]. Dec. 20, 2002.
GenBank: CAA31886.1. unnamed protein product [Bacillus thuringiensis]. Apr. 18, 2005.
GenBank: BAA04468.1. insecticidal crystal protein [Bacillus thuringiensis]. Jun. 15, 2010.
GenBank: AAA86265.1. CryIA(a), partial [Bacillus thuringiensis serovar kurstaki]. Jan. 30, 1996.
GenBank: AAD46139.1. insecticidal crystal protein [Bacillus thuringiensis]. Aug. 1, 1999.
GenBank: BAA77213.1. BtT84A1 crystal protein [Bacillus thuringiensis]. Mar. 14, 2003.
GenBank: AAD55382.1. 135 kDa insecticidal protein [Bacillus thuringiensis serovar kurstaki]. Sep. 16, 1999.
GenBank: CAA70856.1. delta-endotoxin [Bacillus thuringiensis serovar kurstaki]. May 13, 2010.
GenBank: AAP80146.1. delta-endotoxin [Bacillus thuringiensis]. Jun. 30, 2003.
GenBank: AAM44305.1. crystal protein Cry1Aa13 [Bacillus thuringiensis serovar sotto]. Dec. 28, 2004.
GenBank: AAP40639.1. Cry1Aa [Bacillus thuringiensis]. Dec. 1, 2004.
GenBank: AAY66993.1. Cry1Aa, partial [Bacillus thuringiensis]. Jul. 26, 2016.
GenBank: HQ439776.1. Bacillus thuringiensis strain PS9-E2 Cry1A-like protein gene, partial cds. Jul. 25, 2016.
GenBank: AAE33526.1. Sequence 7 from patent U.S. Pat. No. 5,973,231. Aug. 31, 2000.
GenBank: ACU57499.1. pesticidal crystal protein [Bacillus thuringiensis]. Feb. 3, 2010.
GenBank: GQ483512.1. Bacillus thuringiensis strain G7-1 Cry56Aa-like protein gene, complete cds. Jun. 1, 2011.
GenBank: JX025567.1. Bacillus thuringiensis strain HS18-1 Cry56Aa1-like protein gene, complete cds. May 7, 2016.
GenBank: ANC87261.1. tRNA pseudouridine(55) synthase TruB [Sphingomonas sp. NIC1]. Feb. 28, 2017.
GenBank: ANC87260.1. 30S ribosomal protein S15 [Sphingomonas sp. NIC1]. Feb. 28, 2017.
GenBank: JN790647.1. Bacillus thuringiensis strain Bm59-2 cry4-like protein gene, complete cds. Sep. 26, 2015.
GenBank: ACR43758.2. cry4 delta-toxin-like protein (plasmid) [Bacillus thuringiensis]. Jul. 24, 2016.
GenBank: ACU24782.1. Cry [Bacillus thuringiensis serovar jegathesan]. May 10, 2013.
GenBank: EA057254.1. Sequence 628 from patent U.S. Pat. No. 7,166,424. Feb. 7, 2007.
GenBank: EEM99278.1. Pesticidial crystal protein cry 15Aa [Bacillus thuringiensis IBL 4222]. Apr. 30, 2009.
GenBank: GU810818.1. Bacillus thuringiensis serovar malayensis strain 4AV1 crystal protein gene, complete cds. Apr. 3, 2010.
GenBank: EA057253.1. Sequence 626 from patent U.S. Pat. No. 7,166,424. Feb. 7, 2007.
GenBank: EEM99279.1. Pesticidial crystal protein cry 15Aa [Bacillus thuringiensis IBL 4222]. Apr. 30, 2009.
GenBank: HM035087.1. Bacillus thuringiensis strain Sbt009 pesticidal crystal protein Cry0092 (cry0092) gene, complete cds. Dec. 31, 2013.
GenBank: HM132125.1. Bacillus thuringiensis strain HD868(E5) Cry7-like protein gene, partial cds. Jul. 25, 2016.
GenBank: EEM19308.1. hypothetical protein bthur0001_55730 [Bacillus thuringiensis serovar tochigiensis BGSC 4Y1]. Apr. 30, 2009.
GenBank: HM485581.1. Bacillus thuringiensis strain Sbt021 Cry0211 gene, complete cds. Dec. 31, 2019.
GenBank: HM461686.1. Saturnispora sp. DC198 isolate CB013 internal transcribed spacer 1, partial sequence; 5.8S ribosomal RNA gene and internal transcribed spacer 2, complete sequence; and 28S ribosomal RNA gene, partial sequence. Jul. 10, 2017.
GenBank: BAJ05397.1. crystal protein [Bacillus thuringiensis]. Jul. 10, 2014.
GenBank: HM485582.1. Bacillus thuringiensis strain Sbt009 Cry0094 gene, complete cds. Dec. 31, 2019.
GenBank: HQ113114.1. Bacillus thuringiensis Cry20-like protein (cry20) gene, complete cds. Dec. 30, 2012.
GenBank: HQ401006.1. Bacillus thuringiensis strain BtMC28 Cry-like protein (cry) gene, complete cds. Nov. 1, 2013.
GenBank: JQ821388.1. Bacillus thuringiensis strain BtMC28 Cry69Aa-like protein gene, complete cds. Jan. 1, 2016.
GenBank: JN209957.1. Bacillus thuringiensis strain hs18-1 insecticidal Cry-like protein (cry) gene, complete cds. Jul. 1, 2015.
GenBank: JN646781.1. Bacillus thuringiensis strain HS18-1 cry-like protein gene, complete cds. Sep. 26, 2015.
GenBank: EEL67276.1. 83-kDa crystal protein [Bacillus mycoides]. Jun. 25, 2018.
GenBank: JX025568.1. Bacillus thuringiensis strain HS18-1 Cry53Ab1-like protein gene, complete cds. May 7, 2016.
GenBank: JX025569.1. Bacillus thuringiensis strain HS18-1 mosquitocidal toxin-like protein gene, complete cds. May 7, 2016.
GenBank: X03182.1. Bacillus thuringiensis gene for crystal protein (Mr 28 000). Oct. 23, 2008.
GenBank: Z14147.1. B.thuringiensis gene for CytB toxin. Apr. 18, 2005.
GenBank: X98793.1. B.thuringiensis cyt1Ab1 gene. Apr. 18, 2005.
GenBank: U37196.1. Bacillus thuringiensis delta endotoxin gene, complete cds. Jul. 11, 1996.
GenBank: U52043.1. Bacillus thuringiensis plasmid pRX80 CytB toxin homolog (cytB) gene, complete cds. Jul. 26, 2016.
GenBank: HQ439788. Bacillus thuringiensis strain PS9-C12 Cry1A-like protein gene, partial cds Jul. 25, 2016.
Genbank: HQ439790. Bacillus thuringiensis strain PS9-D12 Cry1A-like protein gene, partial cds Nov. 4, 2016.
GenBank: HQ685121. Bacillus thuringiensis strain LS-R-21 Cry1Aa gene, partial cds Jul. 25, 2016.
GenBank: JF340156. Bacillus thuringiensis strain SK-798 plasmid Cry toxin (cry) gene, complete cds Jul. 25, 2016.
GenBank: JN651496. Bacillus thuringiensis strain LTS-209 Cry1Aa gene, partial cds Feb. 5, 2015.
GenBank: KC158223. Bacillus thuringiensis strain Lip plasmid insecticidal crystal protein (cry1Aa) gene, complete cds Jul. 26, 2016.
GenBank: AAA22330. entomocidal protoxin [Bacillus thuringiensis] Apr. 26, 1993.
GenBank: AAA22613. insecticidal endotoxin [Bacillus thuringiensis] Apr. 26, 1993.
GenBank: AAA22561. crystal protein precursor [Bacillus thuringiensis] Apr. 26, 1993.
GenBank: BAA00071. delta-endotoxin [Bacillus thuringiensis serovar kurstaki str. HD-1] Sep. 29, 2007.
GenBank: CAA31620. unnamed protein product [Bacillus thuringiensis] Apr. 18, 2005.
GenBank: AAA22551. insecticidal protein [ Bacillus thuringiensis] Apr. 26, 1993.
GenBank: CAA38701. unnamed protein product [Bacillus thuringiensis] Oct. 23, 2008.
GenBank: A29125, Replaced by Q93T21. Cry1Ab16 Nov. 28, 2006.
GenBank: AAV83251. Transcriptional regulator, Cro/CI family [Idiomarina loihiensis L2TR] Jan. 31, 2014.
GenBank: AAC64003. crystal protein (plasmid) [Bacillus thuringiensis serovar kurstaki] Jul. 25, 016.
GenBank: AAN76494. insecticidal protein P [Bacillus thuringiensis] Dec. 2, 2002.
GenBank: AAG16877. delta endotoxin [Bacillus thuringiensis] Oct. 1, 2000.
GenBank: AA013302. ze28d06.r1 Soares retina N2b4HR Homo sapiens cDNA clone Image:360299 5′, mRNA sequence Nov. 29, 1996.
GenBank: AAK55546. Cry1Ab16 [Bacillus thuringiensis] Oct. 16, 2002.
GenBank: AAT46415. parasporal crystal protein [Bacillus thuringiensis] Jun. 20, 2004.
GenBank: AAQ88259. cry1A toxin [Bacillus thuringiensis] Aug. 19, 2005.
GenBank: AAW31761. Cry1Ab [Bacillus thuringiensis] May 20, 2009.
GenBank: ABB72460. insecticidal crystal protein Cry1Ab [Bacillus thuringiensis] Dec. 1, 2008.
GenBank: ABS18384. delta-endotoxin Cry1Ab [Bacillus thuringiensis] Feb. 19, 2008.
GenBank: ABW87320. endotoxin [Bacillus thuringiensis] Nov. 6, 2007.
GenBank: HQ439778. Bacillus thuringiensis strain HD12 Cry1A-like protein gene, partial cds 25-Jul. 2016.
GenBank: HQ439777. Bacillus thuringiensis strain N32-2-2 Cry1A-like protein gene, partial cds Jul. 25, 2016.
GenBank: HQ685122. Bacillus thuringiensis strain LS-R-30 Cry1Ab gene, partial cds Jul. 25, 2016.
GenBank: HQ847729. Bacillus thuringiensis serovar kurstaki strain DOR BT-1 delta-endotoxin Cry1Ab (cry1Ab) gene, complete cds May 6, 2013.
GenBank: JN135249. Bacillus thuringiensis strain SSy 125-c clone HA1 delta-endotoxin (cry1A) gene, partial cds Sep. 15, 2012.
GenBank: JN135250. Bacillus thuringiensis strain SSy 125-c clone HA2 delta-endotoxin-like (cry1A) gene, complete sequence Sep. 15, 2012.
GenBank: JN135251. Bacillus thuringiensis strain SSy 141-c clone HA6 delta-endotoxin (cry1A) gene, complete cds Sep. 15, 2012.
GenBank: JN135252. Bacillus thuringiensis strain SSy 126-c clone HA7 delta-endotoxin (cry1A) gene, complete cds Sep. 15, 2012.
GenBank: JN135253. Bacillus thuringiensis strain SSy 126-c clone HA9 delta-endotoxin (cry1A) gene, complete cds Sep. 15, 2012.
GenBank: JN135254. Bacillus thuringiensis strain SSy111-c clone HA11 delta-endotoxin (cry1A) gene, complete cds Sep. 15, 2012.
GenBank: AAS93798. cry1A type crystal protein, partial [Bacillus thuringiensis serovar kenyae] Jul. 26, 2016.
GenBank: KC156668. Bacillus thuringiensis strain ARP102 pesticidal protein gene, complete cds Nov. 14, 2013.
GenBank: AAK14336. insecticidal crystal protein BTRX24 [Bacillus thuringiensis serovar kunthalaRX24] Mar. 1, 2001.
GenBank: AAK14337. insecticidal crystal protein BTRX28 [Bacillus thuringiensis serovar kunthalaRX28] -Mar. 1, 2001.
GenBank: AAK14338. insecticidal crystal protein BTRX27 [Bacillus thuringiensis serovar kunthalaRX27] Mar. 1, 2001.
GenBank: ABG88858. Cry1Ab-like BT toxin OL2, partial [Bacillus thuringiensis] Jul. 14, 2016.
GenBank: AAA22331. crystal protein [Bacillus thuringiensis] Apr. 26, 1993.
GenBank: AAA22338. delta-endotoxin [Bacillus thuringiensis] Apr. 26, 1993.
GenBank: CAA38098. unnamed protein product, partial [Bacillus thuringiensis] Jul. 26, 2016.
GenBank: AAA73077. insecticidal delta endotoxin [Bacillus thuringiensis serovar kurstaki] Apr. 26, 1993.
GenBank: AAA22339. cryIA(c)3 [Bacillus thuringiensis serovar kurstaki] Apr. 26, 1993.
GenBank: AAA86266. CryIA(c), partial [Bacillus thuringiensis serovar kurstaki] Jan. 30, 1996.
GenBank: AAB46989. insecticidal delta-endotoxin CryIA(c) [Bacillus thuringiensis serovar kurstaki] Feb. 11, 1997.
GenBank: AAC44841. crystal protein [Bacillus thuringiensis serovar kurstaki] Feb. 18, 1997.
GenBank: AAB49768. Cry1Ac delta-endotoxin [Bacillus thuringiensis] Mar. 17, 1997.
GenBank: CAA05505. insecticidal crystal protein [Bacillus thuringiensis serovar kurstaki str. YBT-1520] Apr. 15, 2005.
GenBank: CAA10270. crystal toxin protein [Bacillus thuringiensis] Apr. 15, 2005.
GenBank: AAD38701. insecticidal protein Cry1Ac, partial [Bacillus thuringiensis serovar kurstaki str. HD-1] Jul. 26, 2016.
GenBank: AAQ06607. Cry1Ac [Bacillus thuringiensis] Apr. 1, 2004.
GenBank: AAN07788. insecticidal crystal protein Cry1Ac [Bacillus thuringiensis] Sep. 29, 2002.
GenBank: AAU87037. Cry1Ac (plasmid) [Bacillus thuringiensis] Jul. 26, 2016.
GenBank: AAX18704. Cry1Ac [Bacillus thuringiensis serovar kenyae] Dec. 1, 2008.
GenBank: AAY88347. Cry [Bacillus thuringiensis] Jun. 1, 2007.
GenBank: ABD37053. insecticidal crystal protein [Bacillus thuringiensis serovar kurstaki] Feb. 20, 2006.
GenBank: ABB89046. delta-endocytoxin [Bacillus thuringiensis] May 1, 2006.
GenBank: AAY66992. Cry1Ac, partial [Bacillus thuringiensis] May 16, 2007.
GenBank: ABZ01836. Cry1Ac22 [Bacillus thuringiensis serovar kurstaki] Dec. 1, 2008.
GenBank: CAQ30431. pesticidal crystal protein cry1Ac, partial (plasmid) [Bacillus thuringiensis serovar kurstaki] Jul. 26, 2016.
GenBank: ABL01535. crystal protein [Bacillus thuringiensis] Dec. 6, 2006.
GenBank: FJ513324. Bacillus thuringiensis strain Tm37-6 pesticidal crystal protein (cry1Ac) gene, partial cds Jul. 24, 2016.
GenBank: FJ617446. Bacillus thuringiensis strain Tm41-4 Cry1Ac-like protein (cry1Ac) gene, partial cds Jul. 24, 2016.
GenBank: FJ617447. Bacillus thuringiensis strain Tm44-1B Cry1Ac-like protein (cry1Ac) gene, partial cds Jul. 24, 2016.
GenBank: ACM90319. Cry1Ac [Bacillus thuringiensis] Feb. 17, 2009.
GenBank: DQ438941. Bacillus thuringiensis strain INTA TA24-6 insecticidal crystal protein (Cry1Ac) gene, partial cds Apr. 1, 2006.
GenBank: GQ227507. Bacillus thuringiensis strain S1478-1 toxin crystal protein gene, complete cds Oct. 15, 2010.
GenBank: GU446674. Bacillus thuringiensis strain S3299-1 Cry1Ac gene, complete cds Feb. 1, 2011.
GenBank: HM061081. Bacillus thuringiensis strain ZQ-89 insecticidal crystal protein Cry1Ac gene, complete cds Jun. 1, 2010.
GenBank: GQ866913. Bacillus thuringiensis strain SK-711 plasmid crystal protein (cry1) gene, partial cds Jul. 25, 2016.
GenBank: HQ230364. Synthetic construct clone EC-783 cry1Ac protein (cry1Ac) gene, cry1Ac18 allele, complete cds Dec. 18, 2010.
GenBank: JF340157. Bacillus thuringiensis strain SK-784 plasmid Cry toxin (cry) gene, complete cds Jul. 25, 2016.
GenBank: JN387137. Bacillus thuringiensis isolate SK-958 truncated crystal protein PT-958 gene, complete cds Dec. 1, 2012.
GenBank: JQ317685. Bacillus thuringiensis strain SK-793 plasmid truncated crystal protein gene, complete cds Jul. 25, 2016.
GenBank: AAA22340. cryIA(d) [Bacillus thuringiensis serovar aizawai] Apr. 26, 1993.
GenBank: CAA01880. PS81RR1 endotoxin, partial [Bacillus thuringiensis] Sep. 25, 1995.
GenBank: AAA22410. delta-endotoxin [Bacillus thuringiensis serovar alesti] Apr. 26, 1993.
GenBank: AAB82749. insecticidal crystal protein, partial (plasmid) [Bacillus thuringiensis] Jul. 26, 2016.
GenBank: AAD46137. lepidoteran-specific toxin [Bacillus thuringiensis] Aug. 1, 1999.
GenBank: AAQ14326. delta-endotoxin Cry1A [Bacillus thuringiensis] Mar. 4, 2008.
GenBank: ABB76664. Cry1A-type pesticidal crystal protein, partial [Bacillus thuringiensis serovar alesti] Jul. 26, 2016.
GenBank: HQ439779. Bacillus thuringiensis strain S6 Cry1A-like protein gene, partial cds Jul. 25, 2016.
GenBank: AA039719. zf09e10.s1 Soares_fetal_heart_NbHH19W Homo sapiens cDNA clone Image:376458 3′ similar to GB:X02162 Apolipoprotein A-I Precursor (Human), mRNA sequence Aug. 30, 1996.
GenBank: HQ439780. Bacillus thuringiensis strain SC6H8 Cry1A-like protein gene, partial cds Jul. 25, 2016.
GenBank: AAK14339. insecticidal crystal protein BTRX3 [Bacillus thuringiensis serovar kunthalanags3] Mar. 1, 2001.
GenBank: CAA29898. unnamed protein product [Bacillus thuringiensis] Apr. 18, 2005.
GenBank: CAA65003. cry1Ba2 [Bacillus thuringiensis serovar entomocidus] Apr. 18, 2005.
GenBank: AAK63251. Cry1Ba [Bacillus thuringiensis] Sep. 30, 2003.
GenBank: AAK51084. delta-endotoxin Cry1Ba2 [Bacillus thuringiensis serovar entomocidus] Nov. 2, 2006.
GenBank: AB020894. Oryza sativa Indica Group RNA for retroposon p-SINE1-RC207 Dec. 20, 2010.
GenBank: ABL60921. Cry1B (plasmid) [Bacillus thuringiensis] Jul. 14, 2016.
GenBank: HQ439781. Bacillus thuringiensis strain N17-37 Cry1B-like protein gene, partial cds Jul. 25, 2016.
GenBank: AAA22344. crystal protein [Bacillus thuringiensis] Apr. 25, 1994.
GenBank: HQ439782. Bacillus thuringiensis strain WBT-2 Cry1B-like protein gene, partial cds Jul. 25, 2016.
GenBank: CAA86568. delta-endotoxin [Bacillus thuringiensis serovar morrisoni] Apr. 18, 2005.
GenBank: AAD10292. insecticidal crystal protein CryE1 [Bacillus thuringiensis serovar wuhanensis] Jan. 29, 1999.
GenBank: AAM93496. CryIBII [Bacillus thuringiensis] Aug. 13, 2002.
GenBank: AAC32850. Cry1Bel delta-endotoxin (plasmid) [Bacillus thuringiensis] Apr. 3, 2020.
GenBank: AAQ52387. Sequence 63 from patent U.S. Pat. No. 6,593,293 Aug. 17, 2003.
GenBank: ACV96720. Cry1Be2 [Bacillus thuringiensis] Aug. 10, 2010.
GenBank: HM070026. Bacillus thuringiensis strain SC6H8 Cry1Be-like protein gene, partial cds Jul. 25, 2016.
GenBank: CAC50778. unnamed protein product [Bacillus thuringiensis] Aug. 8, 2001.
GenBank: AAQ52380. Sequence 38 from patent U.S. Pat. No. 6,593,293 Aug. 17, 2003.
GenBank: AA039720. zf09e11.r1 Soares_fetal_heart_NbHH19W Homo sapiens cDNA clone Image:376460 5′ similar to GB:L19686_rnal Macrophage Migration Inhibitory Factor (Human), mRNA sequence Jan. 28, 2011.
GenBank: HQ589331. Bacillus thuringiensis strain PS46L insecticidal crystal protein DIG-3 gene, complete cds Dec. 20, 2010.
GenBank: KC156700. Bacillus thuringiensis strain ARP260 pesticidal protein gene, complete cds Nov. 14, 2013.
GenBank: CAA30396. unnamed protein product [Bacillus thuringiensis] Apr. 18, 2005.
GenBank: CAA31951. unnamed protein product, partial [Bacillus thuringiensis] Jul. 26, 2016.
GenBank: AAA22343. cryIC(b) [Bacillus thuringiensis serovar aizawai] Apr. 26, 1993.
GenBank: CAA01886. bt15 [Bacillus thuringiensis serovar entomocidus] Sep. 25, 1995.
GenBank: CAA65457. delta-endotoxin, partial [Bacillus thuringiensis serovar aizawai] Jul. 14, 2016.
GenBank: AAF37224. toxin Cry1Ca6 [Bacillus thuringiensis] Mar. 2, 2000.
GenBank: AAG50438. Cry1Ca (plasmid) [Bacillus thuringiensis] Jul. 26, 2016.
GenBank: AAM00264. insecticidal protein Cry1Ca [Bacillus thuringiensis] Apr. 2, 2002.
GenBank: AAL79362. delta-endotoxin [Bacillus thuringiensis] Jun. 22, 2007.
GenBank: AAN16462. insecticidal protein Cry1C, partial [Bacillus thuringiensis] Jul. 24, 2016.
GenBank: AAX53094. insecticidal crystal protein Cry1C [Bacillus thuringiensis] Mar. 28, 2005.
GenBank: HM070027. Bacillus thuringiensis strain mo3-E7 Cry1Ca-like protein gene, partial cds Jul. 25, 2016.
GenBank: HQ412621. Bacillus thuringiensis strain LB-R-78 Cr1C-like protein gene, partial cds Nov. 30, 2012.
GenBank: JN651493. Bacillus thuringiensis strain LTS-38 Cry1Ca gene, partial cds Feb. 5, 2015.
GenBank: M97880. Bacillus thuringiensis cryIC-related gene sequence Jun. 11, 1993.
GenBank: AAG35409. insecticidal crystal protein Cry1Cb (plasmid) [Bacillus thuringiensis] Jul. 26, 2016.
GenBank: ACD50894. Cry1C [Bacillus thuringiensis] May 20, 2008.
GenBank: AAX63901. crystal endotoxin, partial [Bacillus thuringiensis] Jul. 26, 2016.
GenBank: CAA38099. unnamed protein product [Bacillus thuringiensis] Oct. 23, 2008.
GenBank: 176415. replaced by X01939 Yeast tRNA-Cys (GCA) Jun. 11, 2003.
GenBank: HQ439784. Bacillus thuringiensis strain HD12 Cry 1D-like protein gene, partial cds Jul. 25, 2016.
GenBank: CAA80234. crystal protein [Bacillus thuringiensis] Apr. 18, 2005.
GenBank: AAK48937. insecticidal crystal protein [Bacillus thuringiensis] May 1, 2001.
GenBank: ABK35074. insecticidal delta endotoxin [Bacillus thuringiensis] Nov. 2, 2007.
GenBank: CAA37933. crystal protein [Bacillus thuringiensis] Apr. 18, 2005.
GenBank: CAA39609. crystal protein [Bacillus thuringiensis] Apr. 18, 2005.
GenBank: AAA22345. cryIE(a) [Bacillus thuringiensis serovar kenyae] Apr. 26, 1993.
GenBank: AAD04732. Cry1Ea4 [Bacillus thuringiensis] Jan. 14, 1999.
GenBank: A15535. bt18 gene Feb. 18, 1994.
GenBank: AAL50330. Cry032 [Bacillus thuringiensis] Dec. 27, 2001.
GenBank: AAW72936. insecticidal delta endotoxin CryIEa [Bacillus thuringiensis] Mar. 25, 2005.
GenBank: ABX11258. Cry1Ea [Bacillus thuringiensis] Nov. 19, 2007.
GenBank: HQ439785. Bacillus thuringiensis strain S6 Cry1E-like protein gene, partial cds Jul. 25, 2016.
GenBank: ADR00398. Cry1Ea10 [Bacillus thuringiensis] May 27, 2011.
GenBank: JQ652456. Bacillus thuringiensis strain BRC-XQ12 insecticidal crystal protein Cr1Ea11 (cry1Ea11) gene, partial cds Mar. 31, 2012.
GenBank: AAA22346. cryIE(b) [Bacillus thuringiensis serovar aizawai] Apr. 26, 1993.
GenBank: AAA22348. insecticidal crystal protein [Bacillus thuringiensis serovar aizawai] Apr. 26, 1993.
GenBank: AAA22347. cryIF [Bacillus thuringiensis serovar aizawai] Apr. 26, 1993.
GenBank: HM439638. Bacillus thuringiensis strain mo3-D10 Cry1F-like protein gene, partial cds Jul. 25, 2016.
GenBank: CAA80235. crystal protein [Bacillus thuringiensis] Apr. 18, 2005.
GenBank: BAA25298. CryINA67-1 [Bacillus thuringiensis serovar morrisoni] Feb. 5, 1999.
GenBank: AAF21767. crystal protein Cry1Fb [Bacillus thuringiensis serovar morrisoni] Jan. 1, 2000.
GenBank: AAC10641. Sequence 4 from patent U.S. Pat. No. 5,686,069 Apr. 3, 1998.
GenBank: AA013295. ze28c04.r1 Soares retina N2b4HR Homo sapiens cDNA clone Image:360294 5′, mRNA sequence Nov. 29, 1996.
GenBank: ACD50892. Cry1F [Bacillus thuringiensis] May 20, 2008.
GenBank: ACD50893. Cry1F [Bacillus thuringiensis] May 20, 2008.
GenBank: CAA80233. crystal protein [Bacillus thuringiensis] Apr. 18, 2005.
GenBank: CAA70506. delta-endotoxin [Bacillus thuringiensis serovar wuhanensis] Apr. 2, 1997.
GenBank: AAD10291. insecticidal crystal protein CryH2 [Bacillus thuringiensis serovar wuhanensis] Jan. 29, 1999.
GenBank: AA013756. mh27b10.r1 Soares mouse placenta 4NbMP13.5 14.5 Mus musculus cDNA clone Image:443707 5′ similar to GB:D17400 6-Pyruvoyl Tetrahydrobiopterin Synthase (Human), mRNA sequence Jan. 21, 1997.
GenBank: AAQ52381. Sequence 40 from patent U.S. Pat. No. 6,593,293 Aug. 17, 2003.
GenBank: CAA80236. crystal protein [Bacillus thuringiensis] Apr. 18, 2005.
GenBank: AAA79694. crystal toxin [Bacillus thuringiensis] Oct. 19, 1995.
GenBank: HQ439786. Bacillus thuringiensis strain WBT-2 Cry 1H-like protein gene, partial cds Jul. 25, 2016.
GenBank: AAF01213. endotoxin, partial [Bacillus thuringiensis] Jul. 26, 2016.
GenBank: CAA44633. delta-endotoxin [Bacillus thuringiensis serovar kurstaki] Apr. 18, 2005.
GenBank: AAA22354. insecticidal protein [Bacillus thuringiensis serovar kurstaki] Jul. 27, 1993.
GenBank: AAC36999. insecticidal protein [Bacillus thuringiensis serovar kurstaki] Jul. 31, 1995.
GenBank: AAB00958. CGCryV [Bacillus thuringiensis] May 28, 1996.
GenBank: CAA70124. Bt toxin [Bacillus thuringiensis] Apr. 18, 2005.
GenBank: AAC26910. insecticidal protein (plasmid) [Bacillus thuringiensis serovar kurstaki] Jul. 25, 2016.
GenBank: AAM73516. Cry [Bacillus thuringiensis] Jul. 5, 2006.
GenBank: AAK66742. Cry1Ia [Bacillus thuringiensis] Oct. 1, 2003.
GenBank: AAQ08616. Cry1Ia, partial [Bacillus thuringiensis] Jul. 25, 2016.
GenBank: AAP86782. Cr1I [Bacillus thuringiensis] Sep. 25, 2003.
GenBank: CAC85964. delta-endotoxin [Bacillus thuringiensis serovar kurstaki] Jul. 22, 2003.
GenBank: AAV53390. delta endotoxin [Bacillus thuringiensis] Nov. 30, 2007.
GenBank: ABF83202. Cry1Ia [Bacillus thuringiensis] Apr. 11, 2008.
GenBank: ACG63871. Cry1Ia [Bacillus thuringiensis] Aug. 17, 2008.
GenBank: FJ617445. Bacillus thuringiensis strain E-1B Cry1Ia-like protein (cry1Ia) gene, complete cds Jun. 1, 2011.
GenBank: FJ617448. Bacillus thuringiensis strain E-1A Cry1Ia-like protein (cry1Ia) gene, complete cds Jun. 1, 2011.
GenBank: GU989199. Bacillus thuringiensis strain BT-MX2 Cry 1I toxin crystal protein gene, complete cds Apr. 26, 2010.
GenBank: HQ439787. Bacillus thuringiensis strain SC6H8 Cry 1I-like protein gene, partial cds Jul. 25, 2016.
GenBank: ADK23801. Cry1I toxin Crystal protein [Bacillus thuringiensis] Jul. 18, 2010.
GenBank: JQ228426. Bacillus thuringiensis strain wulE-3 Cry1Ia (cry1Ia) gene, complete cds Nov. 30, 2015.
GenBank: JQ228424. Bacillus thuringiensis strain wulE-4 Cry1Ia (cry1la) gene, partial cds Nov. 30, 2015.
GenBank: JQ228427. Bacillus thuringiensis strain wu2B-6 Cry1Ia (cry1Ia) gene, complete cds Nov. 30, 2015.
GenBank: JQ228428. Bacillus thuringiensis strain wu2G-11 Cry1Ia (cry1Ia) gene, complete cds Nov. 30, 2015.
GenBank: JQ228429. Bacillus thuringiensis strain wu2G-12 Cry1Ia (cry1Ia) gene, complete cds Nov. 30, 2015.
GenBank: JQ228430. Bacillus thuringiensis strain you1D-9 Cry1Ia (cry1Ia) gene, complete cds Nov. 30, 2015.
GenBank: JQ228431. Bacillus thuringiensis strain you2D-3 Cry1Ia (cry1Ia) gene, complete cds Nov. 30, 2015.
GenBank: JQ228432. Bacillus thuringiensis strain you2E-3 Cry1Ia (cry1Ia) gene, complete cds Nov. 30, 2015.
GenBank: JQ228433. Bacillus thuringiensis strain you2F-3 Cry1Ia (cry1Ia) gene, complete cds Nov. 30, 2015.
GenBank: JQ228434. Bacillus thuringiensis strain wu1H-3 Cry1Ia (cry1Ia) gene, complete cds Nov. 30, 2015.
GenBank: JQ317686. Bacillus thuringiensis strain BGSC 4J4 plasmid crystal protein (cry1I) gene, complete cds Jul. 25, 2016.
GenBank: JX944038. Bacillus thuringiensis strain SC-7 Cry1Ia (cry1Ia) gene, complete cds Oct. 17, 2012.
GenBank: JX944039. Bacillus thuringiensis strain SC-13 Cry1Ia (cry1Ia) gene, partial cds Oct. 17, 2012.
GenBank: JX944040. Bacillus thuringiensis strain SC-51 Cry1Ia (cry1Ia) gene, complete cds Oct. 17, 2012.
GenBank: AAA82114. cry V465 [Bacillus thuringiensis] Nov. 29, 1995.
GenBank: ABW88019. Cry1Ib-type protein [Bacillus thuringiensis] Nov. 7, 2007.
GenBank: ACD75515. Cry1Ib-type protein [Bacillus thuringiensis] May 28, 2008.
GenBank: HM051227. Bacillus thuringiensis delta endotoxin gene, complete cds Jan. 1, 2011.
GenBank: HM070028. Bacillus thuringiensis strain mo3-D8 Cry1Fa-like protein gene, partial cds Jul. 25, 2016.
GenBank: ADK38579. insecticidal crystal protein Cry1Ib [Bacillus thuringiensis] Jul. 24, 2010.
GenBank: JN571740. Bacillus thuringiensis strain SK-935 plasmid Cry1I-like protein gene, partial cds Jul. 25, 2016.
GenBank: JN675714. Bacillus thuringiensis strain BT HMM-AND7B Cry1I crystal toxin protein (cry1I) gene, complete cds Oct. 2, 2011.
GenBank: JN675715. Bacillus thuringiensis strain BT HMM-BRT2A Cry1I crystal toxin protein (cry1I) gene, complete cds Oct. 2, 2011.
GenBank: JN675716. Bacillus thuringiensis strain BT HMM Cry1I crystal toxin protein (cry1I) gene, complete cds Oct. 2, 2011.
GenBank: JQ228423. Bacillus thuringiensis strain HD12 Cry1Ib (cry1lb) gene, partial cds Nov. 30, 2015.
GenBank: AAC62933. crystal protein toxin (plasmid) [Bacillus thuringiensis] Jul. 25, 2016.
GenBank: AAE71691. Sequence 2 from patent U.S. Pat. No. 6,232,439 Aug. 8, 2001.
GenBank: AAD44366. insecticidal crystal protein [Bacillus thuringiensis] Jul. 20, 1999.
GenBank: JQ228422. Bacillus thuringiensis strain HD12 Cry1Id (cry1Id) gene, complete cds Nov. 30, 2015.
GenBank: AAG43526. Cry1I (plasmid) [Bacillus thuringiensis] Jul. 14, 2016.
GenBank: HM439636. Bacillus thuringiensis strain T03B001 Cry1I-like protein gene, partial cds May 6, 2015.
GenBank: KC156647. Bacillus thuringiensis strain ARP058 pesticidal protein gene, complete cds Nov. 14, 2013.
GenBank: KC156681. Bacillus thuringiensis strain ARP058 pesticidal protein gene, complete cds Nov. 14, 2013.
GenBank: AAQ52382. Sequence 42 from patent U.S. Pat. No. 6,593,293 Aug. 17, 2003.
GenBank: KC156701. Bacillus thuringiensis strain ARP166 pesticidal protein gene, complete cds Nov. 14, 2013.
GenBank: AAC31094. Sequence 10 from patent U.S. Pat. No. 5,723,758 Aug. 10, 1998.
GenBank: ABG88859. Cry1I-like Bt toxin OL3, partial [Bacillus thuringiensis] Jul. 14, 2016.
GenBank: AAA22341. crystal protein [Bacillus thuringiensis] Apr. 26, 1994.
GenBank: HM070030. Bacillus thuringiensis strain WBT-2 Cry1Ja-like protein gene, partial cds Jul. 25, 2016.
GenBank: JQ228425. Bacillus thuringiensis strain FH21 Cry1Ja (cry1Ja) gene, partial cds Nov. 30, 2015.
GenBank: AAA98959. delta-endotoxin CryET1 [Bacillus thuringiensis] Nov. 26, 2012.
GenBank: AAC31092. Sequence 6 from patent U.S. Pat. No. 5,723,758 Aug. 10, 1998.
GenBank: AAQ52372. Sequence 22 from patent U.S. Pat. No. 6,593,293 Aug. 17, 2003.
GenBank: CAC50779. unnamed protein product [Bacillus thuringiensis] Aug. 8, 2001.
GenBank: AAB00376. Cry1K [Bacillus thuringiensis] May 20, 1996.
GenBank: HQ439783. Bacillus thuringiensis strain WBT-2 Cry1K-like protein gene, partial cds Jul. 25, 2016.
GenBank: AAS60191. crystal protein [Bacillus thuringiensis serovar kurstaki] Mar. 17, 2004.
GenBank: HM070031. Bacillus thuringiensis strain SC6H8 Cry1La-like protein gene, partial cds Jul. 25, 2016.
GenBank: FJ884067. Bacillus thuringiensis strain LBIT 1189 clone S1189-B Cry1-like delta-endotoxin gene, complete cds Jul. 24, 2016.
GenBank: KC156659. Bacillus thuringiensis strain ARP080 pesticidal protein gene, complete cds Nov. 14, 2013.
GenBank: KC156648. Bacillus thuringiensis strain ARP009 pesticidal protein gene, complete cds Nov. 14, 2013.
GenBank: KC156678. Bacillus thuringiensis strain ARP146 pesticidal protein gene, complete cds Nov. 14, 2013.
GenBank: AAC31091. Sequence 4 from patent U.S. Pat. No. 5,723,758 Aug. 10, 1998.
GenBank: AAA22335. P2 crystal protein [Bacillus thuringiensis] Apr. 26, 1993.
GenBank: AAA83516. insecticidal crystal protein [Bacillus thuringiensis serovar kurstaki] Dec. 15, 1995.
GenBank: D86064, Replaced by Q8XAT0. RecName: Full=rho operon leader peptide Feb. 10, 2021.
GenBank: AAC04867. insecticidal crystal protein [Bacillus thuringiensis] Mar. 24, 2010.
GenBank: CAA10671. Cry2Aa protein [Bacillus thuringiensis] Apr. 15, 2005.
GenBank: CAA10672. Cry2Aa protein [Bacillus thuringiensis] Apr. 15, 2005.
GenBank: CAA10670. Cry2A protein, partial [Bacillus thuringiensis] Jul. 26, 2016.
GenBank: AA013750.1. mh26g10.r1 Soares mouse placenta 4NbMP13.5 14.5 Mus musculus cDNA clone Image:443682 5′, mRNA sequence Jan. 21, 1997.
GenBank: AAQ04263.1. Cry2Aa [Bacillus thuringiensis] Nov. 1, 2004.
GenBank: AAQ52384.1. Sequence 46 from patent U.S. Pat. No. 6,593,293 Aug. 17, 2003.
GenBank: AB183671.1 Scrippsiella sp. MBIC11143 gene for 18S rRNA, partial sequence, strain: MBIC11143 Jul. 22, 2004.
GenBank: ABL01536.1 crystal protein [Bacillus thuringiensis] Dec. 6, 2006.
GenBank: ACF04939.1 Cry2Aa [Bacillus thuringiensis serovar kenyae] Jul. 23, 2009.
GenBank: JN426947.1 Bacillus thuringiensis strain SSy77 clone HA22 delta-endotoxin (cry2A) gene, complete cds Sep. 15, 2012.
GenBank: AAA22342.1 crystal protein B2 [Bacillus thuringiensis] Apr. 26, 1993.
GenBank: CAA39075.1 crystal protein CryIIB [Bacillus thuringiensis serovar kurstaki] Apr. 18, 2005.
GenBank: AAG36762.1 Cry2Ab [Bacillus thuringiensis] Dec. 2, 2000.
GenBank: AA013296.1 ze28c05.r1 Soares retina N2b4HR Homo sapiens cDNA clone Image:360296 5′ similar to GB:X58295 rnal Plasma Glutathione Peroxidase Precursor (Human), mRNA sequence Nov. 29, 1996.
GenBank: AAQ04609.1 Cry2Ab [Bacillus thuringiensis] Nov. 1, 2004.
GenBank: AAP59457.1 crystal delta-endotoxin Cry2ab-HB [Bacillus thuringiensis] Jun. 17, 2003.
GenBank: AAZ66347.1 delta endotoxin [Bacillus thuringiensis] Aug. 1, 2007.
GenBank: ABC95996.1 Cry2Ab [Bacillus thuringiensis] Feb. 8, 2006.
GenBank: ABC74968.1 crystal delta-endotoxin cry2Ab [Bacillus thuringiensis] Feb. 1, 2007.
GenBank: EF157306.1 Bacillus thuringiensis strain 1yD cry2A-type insecticidal crystal protein gene, complete cds Jan. 3, 2007.
GenBank: CAM84575.1 cry2Ab11 delta endotoxin [Bacillus thuringiensis] Jan. 10, 2009.
GenBank: ABM21764.1 cry2A-type insecticidal crystal protein [Bacillus thuringiensis] Jan. 3, 2007.
GenBank: ACG76120.1 pesticidal crystal protein [Bacillus thuringiensis] Aug. 20, 2008.
GenBank: ACG76121.1. pesticidal crystal protein [Bacillus thuringiensis] Aug. 20, 2008.
GenBank: HM037126.1. Bacillus thuringiensis strain YY1 insecticidal crystal protein gene, complete cds Mar. 1, 2011.
GenBank: GQ866914.1. Bacillus thuringiensis strain SK-793 plasmid crystal protein (cry2A) gene, complete cds Jul. 25, 2016.
GenBank: HQ439789.1. Bacillus thuringiensis strain PS9-C12 Cry2A-like protein gene, partial cds Jul. 25, 2016.
GenBank: JN135255.1. Bacillus thuringiensis strain SSy125-c clone HA13 delta-endotoxin (cry2A) gene, complete cds Sep. 15, 2012.
GenBank: JN135256.1. Bacillus thuringiensis strain SSy60-b clone HA14 delta-endotoxin (cry2A) gene, complete cds Sep. 15, 2012.
GenBank: JN135257.1. Bacillus thuringiensis strain SSy60-b clone HA16 delta-endotoxin (cry2A) gene, complete cds Sep. 15, 2012.
GenBank: JN135258.1. Bacillus thuringiensis strain SSy 141-c clone HA17 delta-endotoxin (cry2A) gene, complete cds Sep. 15, 2012.
GenBank: JN135259.1. Bacillus thuringiensis strain SSy 126-c clone HA18 delta-endotoxin (cry2A) gene, complete cds Sep. 15, 2012.
GenBank: JN135260.1. Bacillus thuringiensis strain SSy 126-c clone HA19 delta-endotoxin (cry2A) gene, partial cds Sep. 15, 2012.
GenBank: JN135261.1. Bacillus thuringiensis strain SSy 111-c clone HA20 delta-endotoxin (cry2A) gene, complete cds Sep. 15, 2012.
GenBank: JN415485.1. Bacillus thuringiensis serovar kurstaki strain MnD insecticidal crystal protein (cry2Ab) gene, complete cds Apr. 17, 2013.
GenBank: JN426946.1. Bacillus thuringiensis strain SSy77 clone HA21 delta-endotoxin (cry2A) gene, complete cds Sep. 15, 2012.
GenBank: JN415764.1. Bacillus thuringiensis strain SP41 Cry2Ab protein (cry2Ab) gene, complete cds Aug. 28, 2011.
GenBank: JN651494.1. Bacillus thuringiensis strain LTS-7 Cry2Aab gene, complete cds Feb. 5, 2015.
GenBank: CAA40536.1. CryIIC delta-endotoxin [Bacillus thuringiensis] Apr. 18, 2005.
GenBank: AAG35410.1. insecticidal crystal protein Cry2Ac [Bacillus thuringiensis] Jan. 3, 2006.
GenBank: AAQ52385.1. Sequence 48 from patent U.S. Pat. No. 6,593,293 Aug. 17, 2003.
GenBank: ABC95997.1. Cry2Ac [Bacillus thuringiensis] BCT Feb. 8, 2006.
GenBank: ABC74969.1. crystal delta-endotoxin cry2Ac [Bacillus thuringiensis] Mar. 12, 2007.
GenBank: ABC74793.1. insecticidal crystal protein Cry2Ac [Bacillus thuringiensis serovar wuhanensis] Jan. 29, 2006.
GenBank: CAL18690.1. cry2Ac7 protein [Bacillus thuringiensis] Dec. 17, 2008.
GenBank: CAM09325.1. insecticidal crystal protein Cry2Ac8 [Bacillus thuringiensis] Jan. 10, 2009.
GenBank: ABN15104.1. insecticidal crystal protein [Bacillus thuringiensis] Feb. 12, 2007.
GenBank: CAM83895.1. Cry2Ac11 protein [Bacillus thuringiensis] Jan. 10, 2009.
GenBank: CAM83896.1. Cry2Ac12 protein [Bacillus thuringiensis] Jan. 10, 2009.
GenBank: AAF09583.1. crystal protein [Bacillus thuringiensis] Nov. 22, 1999.
GenBank: ABC86927.1. crystal protein Cry2Ad [Bacillus thuringiensis] Feb. 4, 2006.
GenBank: CAK29504.1. crystal protein [Bacillus thuringiensis] Jan. 10, 2009.
GenBank: CAM32331.1. crystal protein cry2Ad4 [Bacillus thuringiensis] Jan. 10, 2009.
GenBank: CA078739.1. SCRLAM1010G05.g AM1 Saccharum hybrid cultivar SP80-3280 cDNA clone SCRLAM1010G05 5′, mRNA sequence Feb. 1, 2011.
GenBank: AAQ52362.1. Sequence 2 from patent U.S. Pat. No. 6,593,293 Aug. 17, 2003.
GenBank: AB030519.1. Crematogaster borneensis mitochondrial COI gene for cytochrome oxidase subunit I, partial cds, isolate:aL 1 Jul. 26, 2016.
GenBank: GQ866915.1. Bacillus thuringiensis strain SK-758 plasmid crystal protein (cry2A) gene, complete cds Jul. 25, 2016.
GenBank: ACH91610.1. Cry2Ag [Bacillus thuringiensis] Jul. 2, 2010.
GenBank: EU939453.1. Bacillus thuringiensis strain BTSC6H8 cry2-like protein gene, complete cds Dec. 30, 2011.
GenBank: ACL80665.1. Cry2Ah2 [Bacillus thuringiensis] Jan. 19, 2009.
GenBank: GU073380.1. Bacillus thuringiensis strain HYW-8 delta-endotoxin (cry2A) gene, complete cds Dec. 30, 2012.
GenBank: KC156702.1. Bacillus thuringiensis strain ARP193 pesticidal protein gene, complete cds Nov. 14, 2013.
GenBank: FJ788388.1. Bacillus thuringiensis strain T 20 Cry2A (cry2A) gene, complete cds Mar. 1, 2012.
GenBank: KC156660.1. Bacillus thuringiensis strain ARP067 pesticidal protein gene, complete cds Nov. 14, 2013.
GenBank: KC156658.1. Bacillus thuringiensis strain ARP026 pesticidal protein gene, complete cds Nov. 14, 2013.
GenBank: AAA22336.1. delta-endotoxin, partial [Bacillus thuringiensis] Apr. 26, 1993.
GenBank: AAA22541.1. insecticidal crystal protein [Bacillus thuringiensis] Apr. 26, 1993.
GenBank: CAA68482.1. unnamed protein product [Bacillus thuringiensis] Apr. 18, 2005.
GenBank: AAA22542.1. insect control protein [Bacillus thuringiensis] Apr. 26, 1993.
GenBank: AAA50255.1. crystal protein [Bacillus thuringiensis serovar morrisoni] Feb. 27, 2002.
GenBank: AAC43266.1. CryIIIA [Bacillus thuringiensis serovar tenebrionis] ec. 9, 1994.
GenBank: CAB41411.1. Cry3Aa protein [Bacillus thuringiensis] Apr. 15, 2005.
GenBank: AAS79487.1. insecticidal crystal protein [Bacillus thuringiensis] Apr. 6, 2004.
GenBank: AAW05659.1. Sequence 2 from patent U.S. Pat. No. 6,797,490 Dec. 15, 2004.
GenBank: AAU29411.1. Cry3Aa protein [Bacillus thuringiensis] Apr. 29, 2005.
GenBank: AAW82872.1. Cry3 delta endotoxin [Bacillus thuringiensis serovar tenebrionis] Feb. 14, 2005.
GenBank: ABY49136.1. Cry3A [Bacillus thuringiensis serovar tenebrionis] Jan. 1, 2008.
GenBank: CAA34983.1. unnamed protein product, partial [Bacillus thuringiensis] Jul. 26, 2016.
GenBank: CAA00645.1. toxin [Bacillus thuringiensis] Apr. 14, 2005.
GenBank: JQ397327.1. Bacillus thuringiensis strain ML090 delta-endotoxin Cry3Ba3 (cry3Ba3) gene, complete cds Jan. 10, 2016.
GenBank: AAA22334.1. cryIIIB2 [Bacillus thuringiensis] Apr. 26, 1993.
GenBank: AAA74198.1. Cry3Bb2 [Bacillus thuringiensis] Aug. 10, 1995.
GenBank: I15475.1. Sequence 2 from patent U.S. Pat. No. 5,466,597 Apr. 2, 1996.
GenBank: CAA42469.1. CryIIID [Bacillus thuringiensis serovar kurstaki] Apr. 18, 2005.
GenBank: CAA68485.1. unnamed protein product [Bacillus thuringiensis] Apr. 18, 2005.
GenBank: BAA00179.1. 130 kDa insecticidal protein (ISRH4) (plasmid) [Bacillus thuringiensis serovar israelensis] Jul. 26, 2016.
GenBank: CAD30148.1. pesticidial crystal protein cry4AA (plasmid) [Bacillus thuringiensis serovar israelensis] Oct. 23, 2008.
GenBank: AFB18317.1. putative cry4A [Bacillus thuringiensis serovar israelensis] Feb. 21, 2012.
GenBank: AAY96321.1. cry4A insecticidal protein, partial [Bacillus thuringiensis] Jul. 26, 2016.
GenBank: CAA30312.1. unnamed protein product [Bacillus thuringiensis] Oct. 23, 2008.
GenBank: CAA30114.1. unnamed protein product [Bacillus thuringiensis] Apr. 18, 2005.
GenBank: AAA22337.1. mosquitocidal protein [Bacillus thuringiensis] Apr. 26, 1993.
GenBank: BAA00178.1. 130 kDa insecticidal protein (ISRH3) (plasmid) [Bacillus thuringiensis serovar israelensis] Jul. 26, 2016.
GenBank: CAD30095.1. pesticidial crystal protein cry4BA (plasmid) [Bacillus thuringiensis serovar israelensis] Oct. 23, 2008.
GenBank: ABC47686.1. pesticidal crystal protein cry4B-like [Bacillus thuringiensis] Dec. 22, 2005.
GenBank: EU646202.1. Bacillus thuringiensis strain BTy41 Cry4-like protein gene, complete cds Dec. 20, 2011.
GenBank: FJ403208.1. Bacillus thuringiensis strain HS18-1 pesticidal crystal protein (cry4Da1) gene, complete cds Apr. 3, 2010.
GenBank: FJ597622.1. Bacillus thuringiensis strain Ywc2-8 pesticidal crystal protein (cry4C) gene, complete cds Jun. 1, 2011.
GenBank: FJ403207.1. Bacillus thuringiensis strain BtMC282 pesticidal crystal protein (cry4Db) gene, complete cds Jun. 1, 2011.
GenBank: AAA67694.1. delta-endotoxin, partial [Bacillus thuringiensis serovar darmstadiensis] May 30, 1995.
GenBank: AAA67693.1. delta-endotoxin, partial [Bacillus thuringiensis serovar darmstadiensis] May 30, 1995.
GenBank: 134543.1. Sequence 42 from patent U.S. Pat. No. 5, 596,071 Feb. 6, 1997.
GenBank: ABQ82087.1. CryAd [Bacillus thuringiensis] Jul. 23, 2016.
GenBank: AAA68598.1. delta endotoxin, partial [Bacillus thuringiensis] Jun. 21, 1995.
GenBank: ABW88931.1. Cry5B-like protein [Bacillus thuringiensis YBT-1518] Nov. 17, 2008.
GenBank: AFJ04417.1. Cry5B-like delta endotoxin [Bacillus thuringiensis] May 8, 2012.
GenBank: HM461869.1. Bacillus thuringiensis strain Sbt003 nematicidal crystal protein (cry0031) gene, complete cds Dec. 31, 2013.
GenBank: HM461870.1. Bacillus thuringiensis strain Sbt003 Cry0032 (cry0032) gene, complete cds Dec. 31, 2013.
GenBank: HM485580.1. Bacillus thuringiensis strain Sbt003 Cry0033 gene, complete cds Dec. 31, 2019.
GenBank: AAA22357.1. delta-endotoxin, partial [Bacillus thuringiensis] Apr. 26, 1993.
GenBank: AAM46849.1. nematocidal crystal protein R1 [Bacillus thuringiensis YBT-1518] Dec. 23, 2009.
GenBank: ABH03377.1. nematocidal crystal protein 6A [Bacillus thuringiensis] Aug. 5, 2006.
GenBank: AAA22358.1. delta-endotoxin, partial [Bacillus thuringiensis] Apr. 26, 1993.
GenBank: AAA22351.1. crystal protein [Bacillus thuringiensis serovar galleriae] Apr. 26, 1993.
GenBank: AAA21120.1. CryIII delta-endotoxin, partial [Bacillus thuringiensis serovar dakota] Aug. 27, 1994.
GenBank: AAA21121.1. CryIII delta-endotoxin, partial [Bacillus thuringiensis serovar kumamtoensis] Aug. 27, 1994.
GenBank: ABX24522.1. Cry7Ab3 delta-endotoxin [Bacillus thuringiensis] Sep. 11, 2008.
GenBank: EU380678.1. Bacillus thuringiensis strain HQ122 cry7-like protein gene, complete cds Feb. 1, 2011.
GenBank: ABX79555.2. crystal Cry7-like protein (plasmid) [Bacillus thuringiensis serovar monterrey] Jul. 26, 2016.
GenBank: ACJ44005.1. cytochrome oxidase subunit II, partial (mitochondrion) [Coenonympha tullia scotica].
GenBank: ADB89216.1. insecticidal crystal protein Cry7Ab [Bacillus thuringiensis] Jul. 1, 2010.
GenBank: GU145299.1. Bacillus thuringiensis strain GWS7 Cry7Ab8 protein (cry7Ab8) gene, complete cds Nov. 11, 2009.
GenBank: ADD92572.1. insecticidal crystal protein [Bacillus thuringiensis] Mar. 27, 2010.
GenBank: ABB70817.1. insecticidal crystal protein Cry7Bal [Bacillus thuringiensis serovar huazhongensis] Nov. 1, 2006.
GenBank: KC156653.1. Bacillus thuringiensis strain ARP013 pesticidal protein gene, complete cds Nov. 14, 2013.
GenBank: ABR67863.2. pesticidal crystal protein [Bacillus thuringiensis] Dec. 20, 2016.
GenBank: KC156698.1. Bacillus thuringiensis strain ARP269 pesticidal protein gene, complete cds Nov. 14, 2013.
GenBank: ACQ99547.1. pesticidal crystal protein, partial [Bacillus thuringiensis] Jul. 24, 2016.
GenBank: HM572236.1. Bacillus thuringiensis cry7-like protein 2 gene, partial cds Dec. 30, 2019.
GenBank: KC156679.1. Bacillus thuringiensis strain ARP140 pesticidal protein gene, complete cds Nov. 14, 2013.
GenBank: HM035086.1. Bacillus thuringiensis strain Sbt009 pesticidal crystal protein Cry0091 (cry0091) gene, complete cds Dec. 31, 2013.
GenBank: HM132124.1. Bacillus thuringiensis strain HD868(D8) Cry7-like protein gene, partial cds Jul. 25, 2016.
GenBank: EEM19403.1. hypothetical protein bthur0001_54740 [Bacillus thuringiensis serovar tochigiensis BGSC 4Y1] Apr. 30, 2009.
GenBank: HM035088.1. Bacillus thuringiensis strain Sbt009 pesticidal crystal protein Cry0093 (cry0093) gene, complete cds Dec. 31, 2013.
GenBank: EEM19090.1. hypothetical protein bthur0001_58040 [Bacillus thuringiensis serovar tochigiensis BGSC 4Y1] Apr. 30, 2009.
GenBank: HM572235.1. Bacillus thuringiensis cry7-like protein 1 gene, partial cds Dec. 30, 2019.
GenBank: KC156682.1. Bacillus thuringiensis strain ARP162 pesticidal protein gene, complete cds Nov. 14, 2013.
GenBank: HM572237.1. Bacillus thuringiensis cry7-like protein 3 gene, partial cds Dec. 30, 2019.
GenBank: KC156669.1. Bacillus thuringiensis strain ARP103 pesticidal protein gene, complete cds Nov. 14, 2013.
GenBank: KCL56650.1. DNA-3-methyladenine glycosylase I TagA [Mycobacterium tuberculosis TB_RSA170] Apr. 30, 2014.
GenBank: KC156654.1. Bacillus thuringiensis strain ARP012 pesticidal protein gene, complete cds Nov. 14, 2013.
GenBank: KC156697.1. Bacillus thuringiensis strain ARP271 pesticidal protein gene, complete cds Nov. 14, 2013.
GenBank: KC156651.1. Bacillus thuringiensis strain ARP021 pesticidal protein gene, complete cds Nov. 14, 2013.
GenBank: KC156665.1. Bacillus thuringiensis strain ARP112 pesticidal protein gene, complete cds Nov. 14, 2013.
GenBank: KC156671.1. Bacillus thuringiensis strain ARP114 pesticidal protein gene, complete cds Nov. 14, 2013.
GenBank: KC156680.1. Bacillus thuringiensis strain ARP171 pesticidal protein gene, complete cds Nov. 14, 2013.
GenBank: BAM99306.1. 155 kDa hypothetical protein delta-endotoxin [Bacillus thuringiensis serovar dakota] Mar. 14, 2013.
GenBank: BAM99307.1. 131 kDa hypothetical protein, delta-endotoxin [Bacillus thuringiensis serovar dakota] Mar. 14, 2013.
GenBank: AAA21117.1. CryIII delta-endotoxin, partial [Bacillus thuringiensis serovar kumamtoensis] Aug. 27, 1994.
GenBank: EU044830.1. Bacillus thuringiensis strain B-JJX insecticidal crystal protein gene, complete cds Dec. 31, 2009.
GenBank: KC156662.1. Bacillus thuringiensis strain ARP068 pesticidal protein gene, complete cds Nov. 14, 2013.
GenBank: KC156684.1. Brevibacillus laterosporus strain ARP215 pesticidal protein gene, complete cds Nov. 14, 2013.
GenBank: AAA21118.1. CryIII delta-endotoxin, partial [Bacillus thuringiensis serovar kumamtoensis] Aug. 27, 1994.
GenBank: CAD57542.1. unnamed protein product [Bacillus thuringiensis] Nov. 23, 2002.
GenBank: CAD57543.1. unnamed protein product [Bacillus thuringiensis] Nov. 23, 2002.
GenBank: AAA21119.1. CryIII delta-endotoxin [Bacillus thuringiensis serovar japonensis] Aug. 27, 1994.
GenBank: AAR98783.1. HBF-1 CryIII delta-endotoxin [Bacillus thuringiensis] Oct. 29, 2007.
GenBank: EU625349.1. Bacillus thuringiensis strain FB-32 delta-endotoxin gene, complete cds May 1, 2011.
GenBank: ADB54826.1. CryIII insecticidal crystal protein [Bacillus thuringiensis] Jan. 26, 2010.
GenBank: BAC07226.1. cry8 [Bacillus thuringiensis serovar galleriae] Dec. 20, 2003.
GenBank: BD133574.1. Protein having insecticidal activity, DNA encoding the protein, and controlling agent and controlling method of noxious organisms Sep. 18, 2002.
GenBank: BD133575.1. Protein having insecticidal activity, DNA encoding the protein, and controlling agent and controlling method of noxious organisms Sep. 18, 2002.
GenBank: BAF93483.1. Cry8Dlike [Bacillus thuringiensis] Dec. 6, 2008.
GenBank: AAQ73470.1. Cry8Ea1 [Bacillus thuringiensis] Mar. 18, 2009.
GenBank: EU047597.1. Bacillus thuringiensis strain B-DLL crystal protein gene, complete cds Dec. 30, 2009.
GenBank: KC855216.1. Bacillus thuringiensis strain GWL Cry8Ea3 gene, complete cds Jun. 30, 2015.
GenBank: AAT48690.1. Cry8X [Bacillus thuringiensis] Mar. 18, 2009.
GenBank: HQ174208.1. Bacillus thuringiensis strain B-DLL insecticidal crystal protein (cry8Fa2) gene, complete cds Oct. 2, 2010.
GenBank: AFH78109.1. Cry8x [Bacillus thuringiensis] Apr. 21, 2012.
GenBank: AAT46073.1. crystal protein [Bacillus thuringiensis] Sep. 22, 2009.
GenBank: ABC42043.1. toxin pbt145-1 [Bacillus thuringiensis] Dec. 30, 2008.
GenBank: FJ198072.1. Bacillus thuringiensis strain FCD114 spherical crystal protein (cry8Ga) gene, complete cds Aug. 10, 2010.
GenBank: AAW81032.2. Cry8 [Bacillus thuringiensis] Jan. 3, 2011.
GenBank: EU381044.1. Bacillus thuringiensis strain btsu4 cry8-like protein gene, complete cds Feb. 20, 2011.
GenBank: GU073381.1. Bacillus thuringiensis strain HW-11 delta-endotoxin (cry8) gene, complete cds Dec. 30, 2012.
GenBank: HM044664.1. Bacillus thuringiensis Cry0301 (cry0301) gene, complete cds Dec. 31, 2013.
GenBank: KC156674.1. Bacillus thuringiensis strain ARP124 pesticidal protein gene, complete cds Nov. 14, 2013.
GenBank: GU325772.1. Bacillus thuringiensis strain F4 Cry8L (cry8L) gene, complete cds Dec. 30, 2012.
GenBank: KC156677.1. Bacillus thuringiensis strain ARP135 pesticidal protein gene, complete cds Nov. 14, 2013.
GenBank: EU625348.1. Bacillus thuringiensis strain FPT-2 delta-endotoxin gene, complete cds May 1, 2011.
GenBank: FJ422558.1. Bacillus thuringiensis delta-endotoxin (cry8) gene, partial cds Dec. 26, 2011.
GenBank: ACN87262.1. Cry8Ka2 delta-endotoxin (plasmid) [Bacillus thuringiensis serovar kenyae] Jul. 24, 2016.
GenBank: HM123758.1. Bacillus thuringiensis strain ST8 Cry8Kb gene, complete cds Dec. 30, 2013.
GenBank: KC156675.1. Bacillus thuringiensis strain ARP158 pesticidal protein gene, complete cds Nov. 14, 2013.
GenBank: GU325771.1. Bacillus thuringiensis strain F4 Cry8M (cry8M) gene, complete cds Dec. 30, 2012.
GenBank: HM044665.1. Bacillus thuringiensis Cry0302 (cry0302) gene, complete cds Dec. 31, 2013.
GenBank: EEM86551.1. Pesticidal crystal protein cry8Ba [Bacillus thuringiensis serovar pulsiensis Bgsc 4CC1] Apr. 30, 2009.
GenBank: HM210574.1. Bacillus thuringiensis strain NARC Bt17(C6) Cry8-like protein gene, complete cds Dec. 31, 2013.
GenBank: HM640939.1. Bacillus thuringiensis strain Q52-7 Cry8Ga4 (cry8Ga4) gene, complete cds Jul. 30, 2011.
GenBank: HQ388415.1. Bacillus thuringiensis strain ST8 Cry8-like protein gene, complete cds Dec. 30, 2013.
GenBank: HQ441166.1. Bacillus thuringiensis strain ST8 Cry8-like protein gene, complete cds Dec. 30, 2013.
GenBank: KC152468.1. Bacillus thuringiensis strain INTA Fr7-4 Cry8 gene, complete cds Dec. 24, 2012.
GenBank: AFP87548.1. Cry8-like delta-endotoxin [Bacillus thuringiensis] Aug. 19, 2012.
GenBank: JQ740599.1. Bacillus thuringiensis strain 62 Cry8Sa1 gene, complete cds Feb. 14, 2016.
GenBank: KC156673.1. Bacillus thuringiensis strain ARP110 pesticidal protein gene, complete cds Nov. 14, 2013.
GenBank: FJ770571.1. Bacillus thuringiensis serovar canadensis plasmid cry8b delta-toxin-like gene, complete sequence Sep. 9, 2010.
GenBank: ABS53003.1. crystal protein [Bacillus thuringiensis] Jul. 30, 2007.
GenBank: CAA41122.1. delta-endotoxin CryIG protoxin [Bacillus thuringiensis serovar galleriae] Apr. 18, 2005.
GenBank: CAA41425.1. crystal protein, partial [Bacillus thuringiensis] Jul. 26, 2016.
GenBank: GQ249293.1. Bacillus thuringiensis strain SC5(D2) cry9Aa-like protein gene, complete cds Dec. 31, 2012.
GenBank: GQ249294.1. Bacillus thuringiensis strain T03C001 cry9Aa-like protein gene, complete cds Dec. 31, 2012.
GenBank: JX174110.1. Bacillus sp. enrichment culture clone BGSN1 Cry9Aa5 gene, complete cds Jul. 13, 2013.
GenBank: AAQ52376.1. Sequence 30 from patent U.S. Pat. No. 6,593,293 Aug. 17, 2003.
GenBank: CAA52927.1. delta-endotoxin, partial [Bacillus thuringiensis] Jul. 26, 2016.
GenBank: GU299522.1. Bacillus thuringiensis insecticidal crystal protein Cry9Ba2 gene, complete cds Dec. 1, 2010.
GenBank: AAV28716.1. Cry9Bb delta-endotoxin [Bacillus thuringiensis serovar japonensis] Jul. 7, 2008.
GenBank: CAA85764.1. unnamed protein product [Bacillus thuringiensis] Apr. 18, 2005.
GenBank: AAQ52375.1. Sequence 28 from patent U.S. Pat. No. 6,593,293 Aug. 17, 2003.
GenBank: BAA19948.1. cry9Dal1[Bacillus thuringiensis serovar japonensis] Feb. 6, 1999.
GenBank: AAB97923.1. delta-endotoxin, partial [Bacillus thuringiensis serovar japonensis] Mar. 3, 2017.
GenBank: GQ249297.1. Bacillus thuringiensis strain T03B001 cry9Eb-like protein gene, complete cds Oct. 31, 2013.
GenBank: AAX78439.1. crystal protein Cry9Db1 [Bacillus thuringiensis] Apr. 11, 2005.
GenBank: KC156683.1. Bacillus thuringiensis strain ARP168 pesticidal protein gene, complete cds Nov. 14, 2013.
GenBank: BAA34908.1. Cry9 like protein [Bacillus thuringiensis serovar aizawai] Feb. 5, 1999.
GenBank: AA012908.1. ze27b02.s1 Soares retina N2b4HR Homo sapiens cDNA clone Image:360171 3′ similar to GB:M11560 Fructose-Bisphosphate Aldolase a (Human), mRNA sequence Nov. 29, 1996.
GenBank: ABM21765.1. cry9Ea3 insecticidal crystal protein [Bacillus thuringiensis] Jan. 17, 2007.
GenBank: ACE88267.1. pesticidal crystal protein [Bacillus thuringiensis] un. 24, 2008.
GenBank: ACF04743.1. Cry9Ea protein (plasmid) [Bacillus thuringiensis] Jul. 26, 2016.
GenBank: ACG63872.1. Cry9Ea [Bacillus thuringiensis] Aug. 17, 2008.
GenBank: FJ380927.1. Bacillus thuringiensis strain Bt4 plasmid Cry9Ea (cry9Ea) gene, complete cds Jul. 24, 2016.
GenBank: GQ249292.1. Bacillus thuringiensis strain SC5(E8) cry9Ea-like protein gene, complete cds Dec. 31, 2012.
GenBank: JN651495.1. Bacillus thuringiensis strain LTS-7 Cry9Ea gene, complete cds Feb. 5, 2015.
GenBank: CAC50780.1. unnamed protein product [Bacillus thuringiensis] Aug. 8, 2001.
GenBank: GQ249298.1. Bacillus thuringiensis strain T23001 cry9Eb-like protein gene, complete cds Oct. 31, 2013.
GenBank: KC156646.1. Bacillus thuringiensis strain ARP057 pesticidal protein gene, complete cds Nov. 14, 2013.
GenBank: AAC63366.2. delta-endotoxin [Bacillus thuringiensis] Dec. 10, 2003.
GenBank: AAX78440.1. crystal protein Cry9Ed1 [Bacillus thuringiensis] Apr. 11, 2005.
GenBank: GQ249296.1. Bacillus thuringiensis strain T03B001 cry9Ea-like protein gene, complete cds Oct. 31, 2013.
GenBank: KC156664.1. Bacillus thuringiensis strain ARP095 pesticidal protein gene, complete cds Nov. 14, 2013.
GenBank: KC156692.1. Bacillus thuringiensis strain ARP212 pesticidal protein gene, complete cds Nov. 14, 2013.
GenBank: KC156699.1. Bacillus thuringiensis strain ARP188 pesticidal protein gene, complete cds Nov. 14, 2013.
GenBank: AAA22614.1. insecticidal endotoxin (put.); putative [Bacillus thuringiensis] Apr. 26, 1993.
GenBank: E00614.1. DNA encoding a polypeptide having insecticidal activity(BTI endotoxin) Nov. 4, 2005.
GenBank: CAD30098.1. pesticidial crystal protein cry 10AA (plasmid) [Bacillus thuringiensis serovar israelensis] Oct. 23, 2008.
GenBank: AFB18318.1. putative cry 10 [Bacillus thuringiensis serovar israelensis] Feb. 21, 2012.
GenBank: DQ167578.1. Bacillus thuringiensis strain LDC-9 pesticidal crystal protein c10Aa-like (cry 10Aa) gene, partial sequence Feb. 6, 2006.
GenBank: AAA22611.1. 67 kd mosquitocidal protein, partial [Bacillus thuringiensis] Apr. 26, 1993.
GenBank: CAD30081.1. pesticidial crystal protein cry11AA (plasmid) [Bacillus thuringiensis serovar israelensis] Oct. 23, 2008.
GenBank: AFB18319.1. putative cry 11A, partial [Bacillus thuringiensis serovar israelensis] Feb. 21, 2012.
GenBank: DQ166531.1. Bacillus thuringiensis strain LDC-9 mosquito toxic crystal protein-like (cry11Aa) gene, partial sequence Feb. 2, 2006.
GenBank: CAA60504.1. mosquitocidal toxin [Bacillus thuringiensis] Mar. 20, 1996.
GenBank: AAC97162.1. d-endotoxin [Bacillus thuringiensis serovar medellin] Dec. 18, 1998.
GenBank: HM068615.1. Bacillus thuringiensis strain K34 delta-endotoxin (cry11Bb2) gene, complete cds Jul. 17, 2012.
GenBank: AAA22355.1. delta-endotoxin, partial [Bacillus thuringiensis] Apr. 26, 1993.
GenBank: AAA22356.1. delta-endotoxin [Bacillus thuringiensis] Apr. 26, 1993.
GenBank: AAA21516.1. delta endotoxin [Bacillus thuringiensis serovar sotto]. Sep. 17, 1994.
GenBank: KC156652.1. Bacillus thuringiensis strain ARP001 pesticidal protein gene, complete cds. Nov. 14, 2013.
GenBank: AAA22333.1. crystal protein [Bacillus thuringiensis]. Apr. 26, 1993.
GenBank: CAA63860.1. cbm71 mosquitocidal toxin [[Clostridium] bifermentans]. Jun. 17, 1996.
GenBank: CAA67841.1. cbm72 mosquitocidal toxin [[Clostridium] bifermentans]. Jul. 17, 1998.
GenBank: CAA67506.1. parasporal crystal protein [Paenibacillus popilliae]. Apr. 18, 2005.
GenBank: AAF89667.1. parasporal crystal protein Cry 18Ba1 [Paenibacillus popilliae]. Aug. 1, 2000.
GenBank: AAF89668.1. parasporal crystal protein Cry 18Ca1 [Paenibacillus popilliae ATCC 14706]. Aug. 1, 2000.
GenBank: CAA68875.1. mosquitocidal toxin [Bacillus thuringiensis serovar jegathesan]. Apr. 18, 2005.
GenBank: BAA32397.1. insecticidal protein [Bacillus thuringiensis serovar higo]. Aug. 19, 1998.
GenBank: AFM37572.1. crystal protein [Bacillus thuringiensis serovar vazensis]. Jun. 19, 2012.
GenBank: AAB93476.1. mosquitocidal toxin [Bacillus thuringiensis]. Dec. 29, 1997.
GenBank: ACS93601.1. Cry20-like delta endotoxin (plasmid) [Bacillus thuringiensis]. Jul. 24, 2016.
GenBank: KC156694.1. Bacillus thuringiensis strain ARP192 pesticidal protein gene, complete cds. Nov. 14, 2013.
GenBank: GQ144333.1. Bacillus thuringiensis strain Y-5 insecticidal crystal protein (cry20) pseudogene, complete sequence. Jun. 17, 2009.
GenBank: I32932.1. Sequence 5 from patent U.S. Pat. No. 5,589,382. Feb. 6, 1997.
GenBank: I66477.1. Sequence 8 from patent U.S. Pat. No. 5,670,365. Dec. 28, 1997.
GenBank: BAC06484.1. Cry21Ba1 [Bacillus thuringiensis serovar roskildiensis]. Jan. 25, 2005.
GenBank: JF521577.1. Bacillus thuringiensis strain Sbt072 Cry21Ba1-like protein gene, complete cds. Feb. 21, 2013.
GenBank: KC156687.1. Bacillus thuringiensis strain ARP258 pesticidal protein gene, complete cds. Nov. 14, 2013.
GenBank: JF521578.1. Bacillus thuringiensis strain Sbt072 Cry21Ba2-like protein gene, complete cds. Feb. 21, 2013.
GenBank: I34547.1. Sequence 50 from patent U.S. Pat. No. 5,596,071. Feb. 6, 1997.
GenBank: CAD43579.1. unnamed protein product [Bacillus thuringiensis]. Aug. 9, 2002.
GenBank: ACD93211.1. delta-endotoxin [Bacillus thuringiensis]. Jun. 9, 2008.
GenBank: AAK50456.1. insecticidal crystal protein CryET70 [Bacillus thuringiensis]. Jun. 15, 2001.
GenBank: CAD43577.1. unnamed protein product, partial [Bacillus thuringiensis]. Aug. 9, 2002.
GenBank: CAD43578.1. unnamed protein product [Bacillus thuringiensis]. Aug. 9, 2002.
GenBank: KC156672.1. Bacillus thuringiensis strain ARP148 pesticidal protein gene, complete cds. Nov. 14, 2013.
GenBank: AAF76375.1. crystal protein [Bacillus thuringiensis]. Jun. 16, 2000.
GenBank: AAC61891.1. insecticidal protein Jeg72, partial [Bacillus thuringiensis serovar jegathesan]. Sep. 30, 1998.
GenBank: BAD32657.1. delta-endotoxin [Bacillus thuringiensis]. Aug. 17, 2005.
GenBank: CAJ43600.1. pesticidal crystal protein cry24-like [Bacillus thuringiensis]. Mar. 27, 2007.
GenBank: AAC61892.1. insecticidal protein Jeg74 [Bacillus thuringiensis serovar jegathesan]. Sep. 30, 1998.
GenBank: AAD25075.1. Cry26Aa1 protein [Bacillus thuringiensis serovar finitimus]. Jan. 14, 2000.
GenBank: BAA82796.1. 94kDa mosquitocidal toxin [Bacillus thuringiensis serovar higo]. Jul. 1, 2009.
GenBank: AAD24189.1. Cry28Aa1 delta-endotoxin [Bacillus thuringiensis serovar finitimus]. Jan. 14, 2000.
GenBank: AAG00235.1. parasporal inclusion protein Cry [Bacillus thuringiensis serovar finitimus]. Aug. 16, 2000.
GenBank: CAC80985.1. Cry29Aa protein [Bacillus thuringiensis serovar medellin]. Jan. 13, 2006.
GenBank: CAC80986.1. Cry30Aa protein [Bacillus thuringiensis serovar medellin]. Jan. 13, 2006.
GenBank: BAD00052.1. putative mosquitocidal toxin [Bacillus thuringiensis serovar entomocidus]. Oct. 5, 2006.
GenBank: BAD67157.1. Cry30-like [Bacillus thuringiensis]. Oct. 28, 2004.
GenBank: ACU24781.1. Cry30Ca [Bacillus thuringiensis serovar jegathesan]. May 10, 2013.
GenBank: EF095955.1. Bacillus thuringiensis strain y41 Cry30-like protein gene, complete cds. Dec. 29, 2009.
GenBank: BAE80088.1. delta-endotoxin [Bacillus thuringiensis serovar aizawai]. Mar. 1, 2006.
GenBank: ACC95445.1. Cry 30Ea1 [Bacillus thuringiensis]. Mar. 1, 2009.
GenBank: FJ499389.1. Bacillus thuringiensis strain Ywc2-8 pesticidal crystal protein (cry30) gene, complete cds. Jun. 1, 2011.
GenBank: ACI22625.1. Cry30-like protein [Bacillus thuringiensis]. Jun. 1, 2009.
GenBank: ACG60020.1. Cry30-like protein [Bacillus thuringiensis]. Aug. 13, 2008.
GenBank: HQ638217.1. Bacillus thuringiensis strain S2160-1 Cry30-like protein gene, complete cds. Jun. 25, 2012.
GenBank: BAB11757.1. 81-kDa leukemia toxin [Bacillus thuringiensis]. Aug. 12, 2000.
GenBank: AAL87458.1. 83-KDa crystal protein [Bacillus thuringiensis]. Jun. 21, 2007.
GenBank: BAE79808.1. Cry31-like 81-kDa protein [Bacillus thuringiensis]. Feb. 23, 2006.
GenBank: BAF32571.1. hypothetical protein [Bacillus thuringiensis]. Sep. 21, 2006.
GenBank: BAF32572.1. hypothetical protein [Bacillus thuringiensis]. Sep. 20, 2006.
GenBank: BAI44026.1. M019CP78A (plasmid) [Bacillus thuringiensis]. Jul. 24, 2016.
GenBank: BAE79809.1. Cry31-like 82-kDa protein [Bacillus thuringiensis]. Feb. 23, 2006.
GenBank: BAF32570.1. hypothetical protein [Bacillus thuringiensis]. Sep. 20, 2006.
GenBank: BAF34368.1. hypothetical protein [Bacillus thuringiensis]. Oct. 7, 2006.
GenBank: AB731600.1. Bacillus thuringiensis gene for parasporin 1 like protein, complete cds. May 24, 2013.
GenBank: BAI44022.1. M019CP78B (plasmid) [Bacillus thuringiensis]. Jul. 24, 2016.
GenBank: AAG36711.1. crystal protein [Bacillus thuringiensis serovar yunnanensis]. Jan. 4, 2002.
GenBank: GU063849.1. Bacillus thuringiensis strain FBG-1 delta-endotoxin (cry32A) gene, complete cds. Dec. 30, 2012.
GenBank: GU063850.1. Bacillus thuringiensis strain FZ-2 delta-endotoxin (cry32A) gene, complete cds. Dec. 30, 2012.
GenBank: BAB78601.1. crystal protein CryE6L [Bacillus thuringiensis]. Dec. 6, 2001.
GenBank: BAB78602.1. crystal protein CryE6Q [Bacillus thuringiensis]. Dec. 6, 2001.
GenBank: KC156708.1. Bacillus thuringiensis strain ARP227 pesticidal protein gene, complete cds. Nov. 14, 2013.
GenBank: BAB78603.1. crystal protein CryE6S [Bacillus thuringiensis]. Dec. 6, 2001.
GenBank: GU324274.1. Bacillus thuringiensis strain HYD-3 Cry32 gene, complete cds. Dec. 31, 2012.
GenBank: KC156686.1. Bacillus thuringiensis strain ARP239 pesticidal protein gene, complete cds. Nov. 14, 2013.
GenBank: KC156663.1. Bacillus thuringiensis strain ARP092 pesticidal protein gene, complete cds. Nov. 14, 2013.
GenBank: KC156656.1. Bacillus thuringiensis strain ARP055 pesticidal protein gene, complete cds. Nov. 14, 2013.
GenBank: KC156657.1. Bacillus thuringiensis strain ARP052 pesticidal protein gene, complete cds. Nov. 14, 2013.
GenBank: KC156661.1. Bacillus thuringiensis strain ARP076 pesticidal protein gene, complete cds. Nov. 14, 2013.
GenBank: KC156666.1. Bacillus thuringiensis strain ARP096 pesticidal protein gene, complete cds. Nov. 14, 2013.
GenBank: KC156667.1. Bacillus thuringiensis strain ARP104 pesticidal protein gene, complete cds. Nov. 14, 2013.
GenBank: KC156685.1. Bacillus thuringiensis strain ARP262 pesticidal protein gene, complete cds. Nov. 14, 2013.
GenBank: KC156688.1. Bacillus thuringiensis strain ARP259 pesticidal protein gene, complete cds. Nov. 14, 2013.
GenBank: KC156689.1. Bacillus thuringiensis strain ARP203 pesticidal protein gene, complete cds. Nov. 14, 2013.
GenBank: KC156690.1. Bacillus thuringiensis strain ARP256 pesticidal protein gene, complete cds. Nov. 14, 2013.
GenBank: KC156704.1. Bacillus thuringiensis strain ARP242 pesticidal protein gene, complete cds. Nov. 14, 2013.
GenBank: KC156691.1. Bacillus thuringiensis strain ARP179 pesticidal protein gene, complete cds. Nov. 14, 2013.
GenBank: KC156703.1. Bacillus thuringiensis strain ARP218 pesticidal protein gene, complete cds. Nov. 14, 2013.
GenBank: KC156705.1. Bacillus thuringiensis strain ARP277 pesticidal protein gene, complete cds. Nov. 14, 2013.
GenBank: KC156706.1. Bacillus thuringiensis strain ARP174 pesticidal protein gene, complete cds. Nov. 14, 2013.
GenBank: KC156707.1. Bacillus thuringiensis strain ARP229 pesticidal protein gene, complete cds. Nov. 14, 2013.
GenBank: KC156709.1. Bacillus thuringiensis strain ARP185 pesticidal protein gene, complete cds. Nov. 14, 2013.
GenBank: KC156710.1. Bacillus thuringiensis strain ARP220 pesticidal protein gene, complete cds. Nov. 14, 2013.
GenBank: KC156655.1. Bacillus thuringiensis strain ARP050 pesticidal protein gene, complete cds. Nov. 14, 2013.
GenBank: AAL26871.1. crystal protein NT40KD [Bacillus thuringiensis serovar dakota]. Apr. 10, 2003.
GenBank: AAG50341.1. 13.6 kDa insecticidal crystal protein [Bacillus thuringiensis]. Jan. 11, 2007.
GenBank: AAK64560.1. crystal protein ET74 [Bacillus thuringiensis]. Jan. 30, 2006.
GenBank: AAT29032.1. 14 kDa component of binary insecticidal crystal protein [Bacillus thuringiensis]. Apr. 6, 2005.
GenBank: AAT29030.1. 14 kDa component of binary insecticidal crystal protein [Bacillus thuringiensis]. Apr. 6, 2005.
GenBank: AAG41671.1. 13.6 kDa insecticidal crystal protein [Bacillus thuringiensis]. Sep. 26, 2002.
GenBank: AAG50118.113.6 kDa insecticidal crystal protein [Bacillus thuringiensis]. Mar. 4, 2002.
GenBank: AAK64562.1. crystal protein ET79 [Bacillus thuringiensis]. Jan. 30, 2006.
GenBank: AAT29029.114 kDa component of binary insecticidal crystal protein [Bacillus thuringiensis]. Apr. 6, 2005.
GenBank: AAK64565.1. crystal protein ET80 [Bacillus thuringiensis]. Jan. 30, 2006.
GenBank: AAT29033.114 kDa component of binary insecticidal crystal protein [Bacillus thuringiensis]. Apr. 6, 2005.
GenBank: AAT29031.114 kDa component of binary insecticidal crystal protein [Bacillus thuringiensis]. Apr. 6, 2005.
GenBank: AAG50342.1. 43.8 kDa insecticidal crystal protein [Bacillus thuringiensis]. Jan. 11, 2007.
GenBank: AAT29028.144 kDa component of binary insecticidal crystal protein [Bacillus thuringiensis]. Apr. 6, 2005.
GenBank: AAT29025.1. 44 kDa component of binary insecticidal crystal protein [Bacillus thuringiensis]. Apr. 6, 2005.
GenBank: AAG41672.1. 43.8 kDa insecticidal crystal protein [Bacillus thuringiensis]. Sep. 26, 2002.
GenBank: AAK64563.1. crystal protein ET71 [Bacillus thuringiensis]. Jan. 30, 2006.
GenBank: AY536891.1. Bacillus thuringiensis strain KR1369 44 kDa component of binary insecticidal crystal protein (cry35A) gene, complete cds. Apr. 6, 2005.
GenBank: AAG50117.1. 43.8 kDa insecticidal crystal protein [Bacillus thuringiensis]. Mar. 4, 2002.
GenBank: AAK64566.1. crystal protein ET76 [Bacillus thuringiensis]. Jan. 30, 2006.
GenBank: AAT29027.1. 44 kDa component of binary insecticidal crystal protein [Bacillus thuringiensis]. Apr. 6, 2005.
GenBank: AAT29026.1. 44 kDa component of binary insecticidal crystal protein [Bacillus thuringiensis]. Apr. 6, 2005.
GenBank: AAK64558.1. crystal protein ET69 [Bacillus thuringiensis]. Jun. 26, 2001.
GenBank: AAF76376.1. crystal protein [Bacillus thuringiensis]. Jun. 16, 2000.
GenBank: AAK64559.1. crystal protein ET75 [Bacillus thuringiensis]. Jan. 30, 2006.
GenBank: BAB72016.2. mosquitocidal toxin, partial [Bacillus thuringiensis serovar aizawai]. Jul. 26, 2016.
GenBank: BAB72018.1. putative mosquitocidal toxin, partial [Bacillus thuringiensis serovar aizawai]. Jul. 26, 2016.
GenBank: BAC77648.1. putative mosquitocidal toxin [Bacillus thuringiensis serovar aizawai]. Jun. 14, 2003.
GenBank: EU381045.1. Bacillus thuringiensis strain BTY41 cry40-like protein gene, complete cds. Dec. 1, 2011.
GenBank: ACF15199.1. toxin protein [Bacillus thuringiensis]. Apr. 1, 2009.
GenBank: BAD35163.1. cancer cell-killing Cry protein [Bacillus thuringiensis]. Jan. 20, 2006.
GenBank: HM461871.1. Bacillus thuringiensis strain Sbt021 parasporin 3-like protein (cry0212) gene, complete cds. Dec. 31, 2013.
GenBank: BAD35166.1. Cry protein [Bacillus thuringiensis]. Aug. 10, 2004.
GenBank: BAD15301.1. parasporal crystal protein [Paenibacillus lentimorbus]. Jul. 26, 2016.
GenBank: BAD95474.1. Cryhimel [Paenibacillus popilliae]. Apr. 9, 2005.
Genbank: BAD15303.1. parasporal crystal protein [Paenibacillus lentimorbus]. Jul. 26, 2016.
GenBank: KC156676.1. Brevibacillus laterosporus strain ARP132 pesticidal protein gene, complete cds. Nov. 14, 2013.
GenBank: KC156695.1. Brevibacillus laterosporus strain ARP252 pesticidal protein gene, complete cds. Nov. 14, 2013.
GenBank: KC156696.1. Brevibacillus laterosporus strain ARP191 pesticidal protein gene, complete cds. Nov. 14, 2013.
GenBank: BAD15305.1. parasporal crystal protein, partial [Paenibacillus lentimorbus]. Jul. 26, 2016.
GenBank: BAD08532.1. putative mosquitocidal toxin [Bacillus thuringiensis serovar entomocidus]. Feb. 22, 2008.
GenBank: BAD22577.1. parasporin 1470D [Bacillus thuringiensis serovar shandongiensis]. May 26, 2006.
GenBank: BAC79010. crystal protein [Bacillus thuringiensis serovar dakota]. Oct. 5, 2006.
GenBank: BAG68906.1. parasporin2 [Bacillus thuringiensis serovar shandongiensis]. Oct. 24, 2013.
GenBank: BAD35170.1. crystal protein (plasmid) [Bacillus thuringiensis]. Jul. 26, 2016.
GenBank: AAY24695.1. Cry [Bacillus thuringiensis]. Oct. 24, 2005.
GenBank: CAJ18351.1. Crystal toxin [Lysinibacillus sphaericus]. Jul. 26, 2016.
GenBank: CAJ86545.1. Cry48Aa protein [Lysinibacillus sphaericus]. Aug. 31, 2007.
GenBank: CAJ86546.1. Cry48Aa protein [Lysinibacillus sphaericus]. Aug. 31, 2007.
GenBank: CAJ86548.1. Cry48Aa protein, partial [Lysinibacillus sphaericus]. Jul. 26, 2016.
GenBank: CAJ86549.1. Cry48Aa protein, partial [Lysinibacillus sphaericus] Jul. 26, 2016.
GenBank: CAH56541.1. Crystal toxin [Lysinibacillus sphaericus]. Jul. 26, 2016.
GenBank: CAJ86541.1. Cry49Aa protein [Lysinibacillus sphaericus]. Aug. 31, 2007.
GenBank: CAJ86543.1. Cry49Aa protein [Lysinibacillus sphaericus]. Aug. 31, 2007.
GenBank: CAJ86544.1. Cry49Aa protein [Lysinibacillus sphaericus]. Aug. 31, 2007.
GenBank: CAJ86542.1. Cry49Aa protein [Lysinibacillus sphaericus]. Aug. 31, 2007.
GenBank: BAE86999.1. esticidal crystal protein [Bacillus thuringiensis serovar sotto]. Mar. 16, 2006.
GenBank: GU446675.1. Bacillus thuringiensis strain S2160-1 Cry50-like protein gene, complete cds. Jun. 25, 2012.
GenBank: AB114444.1. Marsupenaeus japonicus con mRNA for crustocalcin-a, complete cds. Dec. 4, 2004.
GenBank: GU570697.1. Bacillus thuringiensis strain EG2934 parasporal crystal protein gene, complete cds. May 16, 2012.
GenBank: EF613489.1. Bacillus thuringiensis Cry 19-like protein gene, complete cds. Dec. 31, 2010.
GenBank: FJ361760.1. Bacillus thuringiensis strain Bm59-2 pesticidal crystal protein (cry53Ba) gene, partial cds. Jul. 24, 2016.
GenBank: EF633476.1. Bacillus thuringiensis strain Y41 Cry toxin (cry) gene, complete cds. Dec. 30, 2010.
GenBank: FJ361759.1. Bacillus thuringiensis strain Btmc28 pesticidal crystal protein (cry52Ab) gene, partial cds. Jul. 24, 2016.
GenBank: ACA52194.1. insecticidal crystal protein Cry54Aa [Bacillus thuringiensis]. Jun. 1, 2009.
GenBank: GQ140349.1. Bacillus thuringiensis strain FBG25 Cry54 (cry54) gene, complete cds. Dec. 22, 2012.
GenBank: GU446677.1. Bacillus thuringiensis strain S2160-1 Cry54-like protein gene, complete cds. Jun. 25, 2012.
GenBank: ABW88932.1. Cry1518-45 [Bacillus thuringiensis YBT-1518]. Nov. 17, 2008.
GenBank: JQ916908.1. Bacillus thuringiensis strain BtMC28 cry-like protein gene, partial cds; and hypothetical protein gene, complete cds. Jan. 1, 2016.
Kant, et al. “Understanding plant response to nitrogen limitation for the improvement of crop nitrogen use efficiency.” J Exp Bot. Feb. 2011;62(4):1499-509. doi: 10.1093/jxb/erq297.
King, et al. “Spider-venom peptides: structure, pharmacology, and potential for control of insect pests.” Annu Rev Entomol. 2013;58:475-96. doi: 10.1146/annurev-ento-120811-153650.
Lin, et al. “PC, a Novel Oral Insecticidal Toxin from Bacillus bombysepticus Involved in Host Lethality via APN and BR-175.” Sci Rep. Jun. 9, 2015;5:11101. doi: 10.1038/srep11101.
Martinelli, et al. “Structure-function studies on jaburetox, a recombinant insecticidal peptide derived from jack bean (Canavalia ensiformis) urease.” Biochim Biophys Acta. Mar. 2014;1840(3):935-44. doi: 10.1016/j.bbagen.2013.11.010.
Naimov, et al. “Solubilization, activation, and insecticidal activity of Bacillus thuringiensis serovar thompsoni HD542 crystal proteins.” Appl Environ Microbiol. Dec. 2008;74(23):7145-51. doi: 10.1128/AEM.00752-08.
NCBI Reference Sequence: ZP_04123838.1. Cancer cell-killing Cry protein [Bacillus thuringiensis serovar pakistani str. T13001]. Nov. 27, 2012.
NCBI Reference Sequence: ZP_04099945.1. hypothetical protein bthur0009_56320 [Bacillus thuringiensis serovar andalousiensis BGSC 4AW1]. Nov. 27, 2012.
NCBI Reference Sequence: ZP_04099652.1 Cancer cell-killing Cry protein [Bacillus thuringiensis serovar andalousiensis BGSC 4AW1]. Nov. 27, 2012.
NCBI Reference Sequence: ZP_04148882.1. Pesticidal crystal protein cry4Aa [Bacillus thuringiensis serovar tochigiensis BGSC 4Y1]. Nov. 27, 2012.
NCBI Reference Sequence: ZP_04123426.1 Pesticidal crystal protein cry5Ba [Bacillus thuringiensis serovar pakistani str. T13001] Nov. 27, 2012.
NCBI Reference Sequence: ZP_04123980.1 Pesticidal crystal protein cry5Ba [Bacillus thuringiensis serovar pakistani str. T13001] Nov. 27, 2012.
NCBI Reference Sequence: ZP_04124038.1 Pesticidal crystal protein cry5Aa [Bacillus thuringiensis serovar pakistani str. T13001] Nov. 27, 2012.
Nestmann, et al. “Mutagenesis by nitrosoguanidine, ethyl methanesulfonate, and mutator gene mutH in continuous cultures of Escherichia coli.” Mutat Res. Jun. 1975;28(3):323-30. doi: 10.1016/0027-5107(75)90226-2.
Parker and Feil. “Pore-forming protein toxins: from structure to function.” Prog Biophys Mol Biol. May 2005;88(1):91-142. doi: 10.1016/j.pbiomolbio.2004.01.009.
Purcell, et al. “Cholesterol oxidase: a potent insecticidal protein active against boll weevil larvae.” Biochem Biophys Res Commun. Nov. 15, 1993;196(3):1406-13. doi: 10.1006/bbrc.1993.2409.
Qaim and Zilberman. “Yield effects of genetically modified crops in developing countries.” Science. Feb. 7, 2003;299(5608):900-2. doi: 10.1126/science.1080609.
Sanahuja, et al. “Bacillus thuringiensis: a century of research, development and commercial applications.” Plant Biotechnol J. Apr. 2011;9(3):283-300. doi: 10.1111/j.1467-7652.2011.00595.x.
Schuler, et al. “Potential side effects of insect-resistant transgenic plants on arthropod natural enemies.” Trends Biotechnol. May 1999;17(5):210-6. doi: 10.1016/S0167-7799(98)01298-0.
Shamseldin. “The Role of Different Genes Involoved in Symbiotic Nitrogen Fixation—Review.” Global Journal of Biotechnology & Biochemistry 8 (4): 84-94, 2013.
Stemmer. “DNA shuffling by random fragmentation and reassembly: in vitro recombination for molecular evolution.” Proc Natl Acad Sci U S A. Oct. 25, 1994;91(22):10747-51. doi: 10.1073/pnas.91.22.10747.
Stemple. “Tilling—a high-throughput harvest for functional genomics.” Nat Rev Genet. Feb. 2004;5(2):145-50. doi: 10.1038/nrg1273.
Tijssen. “Hybridization with Nucelic Acid Probes.” Laboratory Techniques In Biochemistry and Molecular Biology. Department for Physiological Chemistry, University of Utrecht, Utrecht, The Netherlands, vol. 24 (1993) 65 pages.
Yarza, et al. “Uniting the classification of cultured and uncultured bacteria and archaea using 16S rRNA gene sequences.” Nat Rev Microbiol. Sep. 2014;12(9):635-45. doi: 10.1038/nrmicro3330.
Bali, A. et al. “Excretion of ammonium by a nifL mutant of Azotobacter vinelandii fixing nitrogen”; Appl Environ Microbiol; 58(5):1711-8. May 1, 1992. doi: 10.1128/aem.58.5.1711-1718.1992.
Blanco, G. et al. “Sequence and molecular analysis of the nifL gene of Azotobacter vinelandii”; Mol Microbiol;9(4):869-79. Aug. 1, 1993. doi: 10.1111/j.1365-2958.1993.tb01745.x.
De-Yi, Q. et al. “Construction of Genetically Engineered Strains of Enterobacter Cloacae (nifL-Ac)”; Acta Phytophysiologica Sinica; 25(3): 269-273. Jan. 1, 1999. (English Translation).
Hidaka, M. et al. “Promotion of the Growth of Rice by Inoculation of Nitrogen-Fixing-Activity-Enhanced Bacteria to the Rhizosphere”; Nitrogen Fixation: From Molecules to Crop Productivity: 445-445. Jan. 1, 2002.
Kim, Y. et al. “Constitutive expression of nitrogenase system in Klebsiella oxytoca by gene targeting mutation to the chromosomal nifLA operon”; Journal of Biotechnology; 10(3-4):293-301. Jun. 1, 1989.
Marx, C. J. et al. “Broad-Host-Range cre-lox System for Antibiotic Marker Recycling in Gram-Negative Bacteria”; Biotechniques;33(5):1062-7. Nov. 1, 2002. doi: 10.2144/02335rr01.
International Search Report and Written Opinion dated Nov. 19, 2019 for International Application No. PCT/2019/39217, 13 pages.
Office Action dated Feb. 23, 2022 for Chinese Application No. 201980053954.X , with English translation, 16 pages.
Extended European Search Report dated May 27, 2022 for European Application No. 19825926.9, 13 pages.
Gomes, Y. C. B. et al., “Joint use of fungicides, insecticides and inoculants in the treatment of soybean seed,” Rev. Ceres, 64(3):258-265 (2017); https://doi.org/10.1590/0034-737X201764030006.
Kutcher, H. R. et al., “Rhizobium inoculant and seed-applied fungicide effects on field pea production,” Can. J. Plant Sci., 82:645-651 (2002).
Office Action and Search Report, dated Dec. 19, 2022, for Brazilian Application No. 112020026771-6 (6 total pages).
Office Action, dated May 30, 2023, for Chinese Patent Application No. 201980053954.X (4 total pages).
Related Publications (1)
Number Date Country
20220079163 A1 Mar 2022 US
Provisional Applications (2)
Number Date Country
62808693 Feb 2019 US
62690621 Jun 2018 US