COMPOSITIONS AND METHODS RELATING TO PLANT MESSENGER PACKS

Information

  • Patent Application
  • 20220192201
  • Publication Number
    20220192201
  • Date Filed
    April 24, 2020
    4 years ago
  • Date Published
    June 23, 2022
    a year ago
Abstract
Disclosed herein are methods for manufacturing plant messenger packs (PMPs), which can be formulated for use in a variety of agricultural and therapeutic methods.
Description
BACKGROUND

There is need in the art for methods of manufacturing plant messenger packs for use in a variety of agricultural, therapeutic, or commercial applications.


SUMMARY OF THE INVENTION

Described herein are methods for manufacturing of industrial and scaled preparations of PMPs, e.g., methods of manufacturing commercially acceptable and/or pharmaceutically acceptable preparations of PMPs.


In one aspect, the disclosure features a method for producing plant messenger packs (PMPs), the method comprising (a) providing a pectin-rich preparation from a plant comprising extracellular vesicles (EVs), the preparation having a turbidity of 0.8 AU or greater at an absorbance of 650 nm; (b) treating the preparation to reduce the turbidity of the preparation or a fraction thereof; and (c) separating PMPs from the preparation or fraction thereof, thereby producing PMPs.


In another aspect, the disclosure features a method for producing plant messenger packs (PMPs), the method comprising (a) providing a pectin-rich preparation having a viscosity of at least 1.4 cP at 20° C. from a plant comprising EVs; (b) treating the preparation to reduce the viscosity of the preparation or a fraction thereof; and (c) separating PMPs from the preparation or fraction thereof, thereby producing PMPs.


In another aspect, the disclosure features a method for producing plant messenger packs (PMPs), the method comprising (a) providing a pectin-rich preparation from a plant comprising EVs; (b) treating the preparation with an agent that reduces pectin gelation; (c) concentrating the preparation, wherein the viscosity of the concentrated preparation is reduced by at least 10% relative to a concentrated preparation that has not been treated with the agent that reduces pectin gelation; and (d) separating PMPs from the preparation or fraction thereof, thereby producing PMPs.


In another aspect, the disclosure features a method for producing plant messenger packs (PMPs), the method comprising (a) providing a pectin-rich preparation from a plant comprising EVs; (b) treating the preparation to reduce high molecular weight pectin in the preparation or a fraction thereof; and (c) separating PMPs from the preparation or fraction thereof, thereby producing PMPs.


In another aspect, the disclosure features a method for producing plant messenger packs (PMPs), the method comprising (a) providing a pectin-rich preparation from a plant comprising EVs; (b) contacting the preparation or a fraction thereof with a chelating agent; and (c) separating PMPs from the chelated preparation or fraction thereof, thereby producing PMPs.


In another aspect, the disclosure features a method for manufacturing PMPs, the method comprising (a) processing at least 500 g of a pectin-rich plant or plant part comprising EVs into a preparation; (b) contacting the preparation or a fraction thereof with a chelating agent; and (c) processing the chelated preparation or fraction thereof to separate PMPs, wherein the contacting is performed in an amount and for a time sufficient to reduce high molecular weight pectin in the chelated preparation or fraction thereof by at least 10%.


In some aspects, the processing of step (c) comprises separating the PMPs from the chelated preparation or fraction thereof. In some aspects, the chelating agent reduces gelation of pectin in the chelated preparation or fraction thereof. In some aspects, the chelating agent is EDTA or EGTA. In some aspects, the EDTA or EGTA is in a solution with MES, Tris, or PBS.


In some aspects, the method further comprises treating the preparation with a pectinase enzyme.


In another aspect, the disclosure features a method for producing PMPs, the method comprising (a) providing a pectin-rich preparation from a plant comprising EVs; (b) contacting the preparation or a fraction thereof with a pectinase enzyme; and (c) separating PMPs from the preparation or fraction thereof, thereby producing PMPs.


In some aspects, the method further comprises removal or inactivation of the pectinase enzyme.


In some aspects, the pectin concentration in the preparation is at least 0.1%.


In some aspects, the PMPs of step (c) are concentrated at least 10× relative to the preparation of step (a).


In some aspects, the separating or processing comprises centrifugation. In some aspects, the centrifugation is differential centrifugation.


In some aspects, the separating or processing comprises one or more filtration steps. In some aspects, the one or more filtration steps comprise tangential flow filtration. In some aspects, the tangential flow filtration comprises exchanging the volume of the preparation at least 10 times. In some aspects, the one or more filtration steps comprise size exclusion chromatography. In some aspects, the one or more filtration steps comprise tangential flow filtration and size exclusion chromatography. In some aspects, the separating or processing comprises one, two, or all three of centrifugation, tangential flow filtration, and size exclusion chromatography. In some aspects, the separating or processing comprises one or more of a wash step, dilution, pH modification, dialysis, and removal of contaminants.


In some aspects, pectin concentration in the PMPs of step (c) is reduced by at least 10% relative to PMPs produced from a preparation that has not been treated.


In some aspects, providing the preparation comprises processing a plant or a plant part to release EVs. In some aspects, the processing comprises blending a plant or a plant part. In some aspects, the plant part is a juice sac of a grapefruit or lemon.


In some aspects, the processing comprises mashing a plant or a plant part through a strainer. In some aspects, the processing comprises cold pressing a plant or a plant part.


In some aspects, the preparation is obtained from a pectin-rich plant or a pectin-rich plant part. In some aspects, the plant is a citrus plant. In some aspects, the citrus plant is a grapefruit or lemon. In some aspects, the plant is a flowering plant. In some aspects, the plant is a vegetable. In some aspects, the plant is a fruit.


In some aspects, the viscosity of the preparation is monitored, e.g., is monitored before, during, or after treatment, e.g., in an in-process control.


In some aspects, the viscosity of the preparation is reduced by at least 5% relative to a preparation that has not been treated.


In some aspects, the method comprises formulating the PMPs produced in step (c) with a carrier.


In some aspects, the carrier is an agriculturally acceptable carrier. In some aspects, the PMPs are formulated for delivery to a plant. In some aspects, the carrier is a pharmaceutically acceptable carrier.


In some aspects, the PMPs are formulated for administration to a human.


In some aspects, the PMPs are formulated with a liquid, a solid, an aerosol, a paste, a gel, or a gas composition.


In some aspects, the PMPs are stable for at least 24 hours, 48 hours, seven days, or 30 days.


In some aspects, the PMPs are stable at a temperature of at least 4° C. In some aspects, the PMPs are stable at a temperature of at least 20° C., 24° C., or 37° C.


In some aspects, the PMPs are at a concentration of at least 1, 10, 50, 100, or 250 μg PMP protein/ml.


In some aspects, the method comprises loading the PMPs with a heterologous functional agent.


In some aspects, the heterologous functional agent is a heterologous agricultural agent. In some aspects, the heterologous agricultural agent is a pesticidal agent. In some aspects, the heterologous agricultural agent is a fertilizing agent. In some aspects, the heterologous agricultural agent is an herbicidal agent. In some aspects, the heterologous agricultural agent is a plant-modifying agent.


In some aspects, the heterologous functional agent is a heterologous therapeutic agent. In some aspects, the heterologous functional agent comprises an antifungal agent, an antibacterial agent, a virucidal agent, an anti-viral agent, an insecticidal agent, a nematicidal agent, an antiparasitic agent, or an insect repellent.


In another aspect, the disclosure features a PMP composition comprising a plurality of PMPs, wherein the PMPs are produced by a process comprising the steps of (a) providing a pectin-rich preparation from a plant comprising extracellular vesicles (EVs), the preparation having a turbidity of 0.8 AU or greater at an absorbance of 650 nm; (b) treating the preparation to reduce the turbidity of the preparation or a fraction thereof; and (c) separating PMPs from the preparation or fraction thereof, thereby producing PMPs.


In another aspect, the disclosure features a PMP composition comprising a plurality of PMPs, wherein the PMPs are produced by a process comprising the steps of (a) providing a pectin-rich preparation having a viscosity of at least 1.4 cP at 20° C. from a plant comprising EVs; (b) treating the preparation to reduce the viscosity of the preparation or a fraction thereof; and (c) separating PMPs from the preparation or fraction thereof, thereby producing PMPs.


In another aspect, the disclosure features a PMP composition comprising a plurality of PMPs, wherein the PMPs are produced by a process comprising the steps of (a) providing a pectin-rich preparation from a plant comprising EVs; (b) treating the preparation with an agent that reduces pectin gelation; (c) concentrating the preparation, wherein the viscosity of the concentrated preparation is reduced by at least 10% relative to a concentrated preparation that has not been treated with the agent that reduces pectin gelation; and (d) separating PMPs from the preparation or fraction thereof, thereby producing PMPs.


In another aspect, the disclosure features a PMP composition comprising a plurality of PMPs, wherein the PMPs are produced by a process comprising the steps of (a) providing a pectin-rich preparation from a plant comprising EVs; (b) treating the preparation to reduce high molecular weight pectin in the preparation or a fraction thereof; and (c) separating PMPs from the preparation or fraction thereof, thereby producing PMPs.


In another aspect, the disclosure features a PMP composition comprising a plurality of PMPs, wherein the PMPs are produced by a process comprising the steps of (a) providing a pectin-rich preparation from a plant comprising EVs; (b) contacting the preparation or a fraction thereof with a chelating agent; and (c) separating PMPs from the chelated preparation or fraction thereof, thereby producing PMPs.


In another aspect, the disclosure features a PMP composition comprising a plurality of PMPs, wherein the PMPs are produced by a process comprising the steps of (a) processing at least 500 g of a pectin-rich plant or plant part comprising EVs into a preparation; (b) contacting the preparation or a fraction thereof with a chelating agent; and (c) processing the chelated preparation or fraction thereof to separate PMPs, wherein the contacting is performed in an amount and for a time sufficient to reduce high molecular weight pectin in the chelated preparation or fraction thereof by at least 10%.


In some aspects, the processing of step (c) comprises separating the PMPs from the chelated preparation or fraction thereof. In some aspects, the chelating agent reduces polymerization of pectin in the chelated preparation or fraction thereof. In some aspects, the chelating agent is EDTA or EGTA. In some aspects, the EDTA or EGTA is in a solution with MES, Tris, or PBS.


In some aspects, the PMP composition further comprises treating the preparation with a pectinase enzyme.


In another aspect, the disclosure features a PMP composition comprising a plurality of PMPs, wherein the PMPs are produced by a process comprising the steps of (a) providing a pectin-rich preparation from a plant comprising EVs; (b) contacting the preparation or a fraction thereof with a pectinase enzyme; and (c) separating PMPs from the preparation or fraction thereof, thereby producing PMPs.


In some aspects, the PMP composition further comprises removal or inactivation of the pectinase enzyme.


In some aspects, the PMP composition further comprises a carrier. In some aspects, the carrier is an agriculturally acceptable carrier. In some aspects, the carrier is a pharmaceutically acceptable carrier.


In some aspects, the composition is formulated as a liquid, a solid, an aerosol, a paste, a gel, or a gas composition.


In some aspects, the PMP composition is stable for at least 24 hours, 48 hours, seven days, or 30 days.


In some aspects, the PMP composition is stable at a temperature of at least 4° C. In some aspects, the PMP composition is stable at a temperature of at least 20° C., 24° C., or 37° C.


In some aspects, the PMPs in the composition are at a concentration of at least 1, 10, 50, 100, or 250 μg PMP protein/ml.


In another aspect, the disclosure features a method of increasing the fitness of a plant, the method comprising delivering to the plant an effective amount of the PMP composition of any one the above aspects, wherein the method increases the fitness of the plant relative to an untreated plant.


In another aspect, the disclosure features a method of decreasing the fitness of a plant pest, the method comprising delivering to the plant pest an effective amount of the PMP composition of any one of the above aspects, wherein the method decreases the fitness of the plant pest relative to an untreated plant pest.


In another aspect, the disclosure features a method of treating an infection in an animal in need thereof, the method comprising administering to the animal an effective amount of the PMP composition of any one of the above aspects.


In another aspect, the disclosure features a method of decreasing the fitness of a pathogen, the method comprising delivering to the pathogen an effective amount of the PMP composition of any one of the above aspects, wherein the method is effective to decrease the fitness of the pathogen relative to an untreated pathogen.


In another aspect, the disclosure features a method of decreasing the fitness of an animal pathogen vector, the method comprising delivering to the vector an effective amount of the PMP composition of any one of the above aspects, wherein the method decreases the fitness of the vector relative to an untreated vector.


In another aspect, the disclosure features a method for producing plant messenger packs (PMPs), the method comprising (a) providing a pectin-rich preparation from a plant comprising EVs; (b) (i) treating the preparation to reduce the turbidity of the preparation or a fraction thereof; (ii) treating the preparation to reduce the viscosity of the preparation or a fraction thereof; (iii) treating the preparation to reduce high molecular weight pectin in the preparation or a fraction thereof; (iv) contacting the preparation or a fraction thereof with a chelating agent; or (v) contacting the preparation or a fraction thereof with a pectinase enzyme; (c) intermittently or continuously measuring the viscosity of the preparation or fraction thereof during step (b); (d) ending step (b) when the viscosity of the preparation or fraction thereof is below a predetermined level that informs that the preparation or fraction thereof of step (e) will have reduced gelation relative to a preparation or fraction thereof that has not been treated; and (e) separating PMPs from the preparation or fraction thereof.


In some aspects, viscosity is measured in-process during step (b). In some aspects, viscosity is measured intermittently during step (b). In some aspects, viscosity is measured continuously during at least a portion of step (b). In some aspects, viscosity is measured continuously during step (b).


In some aspects, the predetermined level of viscosity is 1.4 cP when viscosity is measured at 20° C. In some aspects, the temperature of the composition during step (b) is 20° C.


In another aspect, the disclosure features a method for producing plant messenger packs (PMPs), the method comprising (a) providing a pectin-rich preparation from a plant comprising EVs; (b) (i) treating the preparation to reduce the turbidity of the preparation or a fraction thereof; (ii) treating the preparation to reduce the viscosity of the preparation or a fraction thereof; (iii) treating the preparation to reduce high molecular weight pectin in the preparation or a fraction thereof; (iv) contacting the preparation or a fraction thereof with a chelating agent; or (v) contacting the preparation or a fraction thereof with a pectinase enzyme; (c) intermittently or continuously measuring the turbidity of the preparation or fraction thereof during step (b); (d) ending step (b) when the turbidity of the preparation or fraction thereof is below a predetermined level that informs that the preparation or fraction thereof of step (e) will have reduced gelation relative to a preparation or fraction thereof that has not been treated; and (e) separating PMPs from the preparation or fraction thereof.


In some aspects, turbidity is measured in-process during step (b). In some aspects, turbidity is measured intermittently during step (b). In some aspects, turbidity is measured continuously during at least a portion of step (b). In some aspects, turbidity is measured continuously during step (b).


In some aspects, the predetermined level of turbidity is 0.8 AU at an absorbance of 650 nm.


Other features and advantages of the invention will be apparent from the following Detailed Description and the Claims.


Definitions

As used herein, the term “pectin”, or “pectic polysaccharide” refers to a polysaccharide, e.g., a polysaccharide occurring in a plant cell wall or a middle lamella, e.g., a galacturonic acid-rich polysaccharide. Exemplary pectins include homogalacturonans, rhamnogalacturonan I, and the substituted galacturonans rhamnogalacturonan II (RG-II) and xylogalacturonan (XGA). Pectins may be classified as low-methoxyl pectins or high-methoxyl pectins based on the degree of methyl esterification.


In some aspects, the degree of methyl esterification is at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or 100% methyl esterification.


In one example, the pectin is low-methoxyl pectin, e.g., a pectin having less than 50% methyl esterification. Gelation (e.g., increased viscosity of a preparation or solution comprising pectin) of low-methoxyl pectin results from ionic linkage between two carboxyl groups belonging to two different pectin chains via calcium bridges. Gelation of low-methoxyl pectin is increased in the presence of calcium, e.g., Ca2+ ions.


In another example, the pectin is high-methoxyl pectin, e.g., a pectin having 50% or more methyl esterification. Gelation of high-methoxyl pectin results from cross-linking of pectin molecules, involving a combination of hydrogen bonds and hydrophobic interactions between the pectin molecules.


In some aspects, the pectin is a high molecular weight pectin. Pectins having higher molecular weight have higher viscosity. Viscosity may be measured, for example, as described in Sayah et al., PLoS ONE, 11(9), e0161751, 2016.


The presence and amount of pectin in a substance (e.g., a plant preparation) may be detected using any known assay for pectins. For example, the assay may be performed using a Pectin Identification Assay Kit (Megazyme; K-PECID). The assay may involve treating the substance with an enzyme, e.g., a pectinase, and measuring the level of an enzymatic product of pectin, e.g., a sugar. The assay may be a colorimetric assay, e.g., a colorimeteric assay to detect galacturonic acid, a component of pectin, following contacting the substance with a pectinase and 3,5-dinitrosalicylic acid (DNS).


As used herein, the term “pectin-rich” refers to a substance, e.g., a plant preparation, comprising more than 0.01%, more than 0.05%, more than 0.1%, more than 0.5%, more than 1%, more than 5%, or more than 10% pectin. In other examples, a “pectin-rich” preparation may include between 0.1%-10% pectin, for example, between 0.5%-5% pectin.


As used herein, the term “pectinase” or “pectic enzyme” refers to an enzyme or a mixture of enzymes capable of degrading a pectin. Exemplary pectinases include pectolyase (pectin lyase) and polygalacturonase (pectin depolymerase).


As used herein, the term “plant preparation” refers to a product resulting from preparing or processing of a plant or a plant part. The plant preparation may be a liquid, a gel, or a gel-like solution. In one example, the viscosity of the plant preparation is 1.4 cP at 20° C. In other examples, the plant preparation is a blended plant or a blended plant part (e.g., a blended citrus fruit or a blended juice sac of a citrus fruit). In other aspects, the plant preparation is the product of a plant or a plant part (e.g., a citrus fruit or a juice sac of a citrus fruit) being mashed through a strainer. In other examples, the plant preparation is the product of cold pressing a plant or a plant part (e.g., a citrus fruit or a juice sac of a citrus fruit). A plant preparation may contain, without limitation, plant cell wall components; pectin; plant organelles (e.g., mitochondria; plastids such as chloroplasts, leucoplasts or amyloplasts; and nuclei); plant chromatin (e.g., a chromosome from the nucleus); or plant molecular aggregates (e.g., protein aggregates, protein-nucleic acid aggregates, lipoprotein aggregates, or lipido-proteic structures), or any other cellular or apoplastic component found in a plant or a plant part.


As used herein, the term “chelation” refers to the process of treating a preparation, solution, or system comprising a metal ion with a chelating agent (chelator). Typically, the chelating agent binds the metal ion to form a chelate (i.e., a compound having a metal ion covalently bound to two or more non-metallic ions in the compound), thus diminishing the chemical effect (e.g., reactivity) of the metal ion in the preparation, solution, or system. In some aspects, the metal ion is a calcium ion (e.g., Ca2+), a magnesium ion (e.g., Mg2+), an iron ion, a lead ion, or a copper ion. Chelating agents include, but are not limited to ethylenediaminetetraacetic acid (EDTA) and ethylene glycol-bis(β-aminoethyl ether)-N,N,N′,N′-tetraacetic acid (EGTA). The chelating agent may be formulated with sodium hydroxide (NaOH). In other examples, the chelating agent is formulated with 2-(N-morpholino)ethanesulfonic acid (MES), tris(hydroxymethyl)aminomethane (Tris), or phosphate buffered saline (PBS).


As used herein, the term “chelated preparation” or “chelated solution” refers to a preparation or solution treated with a chelating agent in an amount and for a time sufficient to diminish the reactivity of a metal ion in the solution by at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or 100%. In some aspects, the reactivity of the metal ion is quantified as esterification of pectins, e.g., in an assay for viscosity or turbidity of the solution.


As used herein, the term “juice sac” or “juice vesicle” refers to a juice-containing membrane-bound component of the endocarp (carpel) of a hesperidium, e.g., a citrus fruit. In some aspects, the juice sacs are separated from other portions of the fruit, e.g., the rind (exocarp or flavedo), the inner rind (mesocarp, albedo, or pith), the central column (placenta), the segment walls, or the seeds. In some aspects, the juice sacs are juice sacs of a grapefruit, a lemon, a lime, or an orange.


As used herein, the term “turbidity” or “turbid” refers to the relative opacity or cloudiness of a liquid, solution, or preparation (e.g., a PMP preparation), e.g., due to particulate matter suspended in the solution (e.g., pectin). Turbidity may be measured by, e.g., measuring the absorbance or optical density of a liquid, solution, or preparation at 650 nm (A650 nm or OD650). Other wavelengths (e.g., wavelengths greater than 650 nm) may also be appropriate for measuring turbidity.


As used herein, “decreasing the fitness of a plant pest” refers to any disruption to pest physiology, or any activity carried out by said pest, as a consequence of administration of a PMP composition described herein, including, but not limited to, any one or more of the following desired effects: (1) decreasing a population of a pest by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, 100% or more; (2) decreasing the reproductive ability or rate of a pest (e.g., insect) by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, 100% or more; (3) decreasing the mobility of a pest by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, 100% or more; (4) decreasing the body weight of a pest by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, 100% or more; (5) decreasing the metabolic rate or activity of a pest by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, 100% or more; or (6) decreasing plant infestation by a pest by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, 100% or more. A decrease in pest fitness can be determined in comparison to a pest to which the pest control (e.g., biopesticide or biorepellent) composition has not been administered.


As used herein “decreasing the fitness of a pathogen” refers to any disruption to pathogen physiology as a consequence of administration of a PMP composition described herein, including, but not limited to, any one or more of the following desired effects: (1) decreasing a population of a pathogen by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, 100% or more; (2) decreasing the reproductive ability or rate of a pathogen by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, 100% or more; (3) decreasing the mobility of a pathogen by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, 100% or more; (4) decreasing the body weight or mass of a pathogen by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, 100% or more; (5) decreasing the metabolic rate or activity of a pathogen by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, 100% or more; or (6) decreasing pathogen transmission (e.g., vertical or horizontal transmission of a pathogen from one insect to another) by a pathogen by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, 100% or more. A decrease in pathogen fitness can be determined, e.g., in comparison to an untreated pathogen.


As used herein “decreasing the fitness of a vector” refers to any disruption to vector physiology, or any activity carried out by said vector, as a consequence of administration of a PMP composition described herein, including, but not limited to, any one or more of the following desired effects: (1) decreasing a population of a vector by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, 100% or more; (2) decreasing the reproductive ability or rate of a vector (e.g., insect, e.g., mosquito, tick, mite, louse) by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, 100% or more; (3) decreasing the mobility of a vector (e.g., insect, e.g., mosquito, tick, mite, louse) by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, 100% or more; (4) decreasing the body weight of a vector (e.g., insect, e.g., mosquito, tick, mite, louse) by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, 100% or more; (5) increasing the metabolic rate or activity of a vector (e.g., insect, e.g., mosquito, tick, mite, louse) by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, 100% or more; (6) decreasing vector-vector pathogen transmission (e.g., vertical or horizontal transmission of a vector from one insect to another) by a vector (e.g., insect, e.g., mosquito, tick, mite, louse) by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, 100% or more; (7) decreasing vector-animal pathogen transmission by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, 100% or more; (8) decreasing vector (e.g., insect, e.g., mosquito, tick, mite, louse) lifespan by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, 100% or more; (9) increasing vector (e.g., insect, e.g., mosquito, tick, mite, louse) susceptibility to pesticides (e.g., insecticides) by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, 100% or more; or (10) decreasing vector competence by a vector (e.g., insect, e.g., mosquito, tick, mite, louse) by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, 100% or more. A decrease in vector fitness can be determined, e.g., in comparison to an untreated vector.


As used herein, the term “untreated” refers to an animal (e.g., a mammal), a plant, or a plant pest that has not been contacted with or delivered a PMP composition, including a separate animal, plant, or plant pest that has not been delivered the PMP composition, the same animal, plant, or plant pest undergoing treatment assessed at a time point prior to delivery of the PMP composition, or the same animal, plant, or plant pest undergoing treatment assessed at an untreated part of the animal, plant, or plant pest (that is, at an area of the animal, plant, or plant pest not contacted with the PMP composition).


As used herein, the term “effective amount,” “effective concentration,” or “concentration effective to” refers to an amount of a PMP, or a composition thereof, sufficient to effect the recited result or to reach a target level (e.g., a predetermined or threshold level) in or on a target organism.


As used herein, the term “heterologous” refers to an agent (e.g., a functional agent) that is either (1) exogenous to the plant (e.g., originating from a source that is not the plant or plant part from which the PMP is produced) (e.g., added the PMP using loading approaches described herein) or (2) endogenous to the plant cell or tissue from which the PMP is produced, but present in the PMP (e.g., added to the PMP using loading approaches described herein, genetic engineering, or in vitro or in vivo approaches) at a concentration that is higher than that found in nature (e.g., higher than a concentration found in a naturally-occurring plant extracellular vesicle). As used herein, the term “functional agent” refers to an agent (e.g., an agricultural agent (e.g., pesticidal agent, fertilizing agent, herbicidal agent, or a plant-modifying agent), a pathogen control agent (e.g., an antifungal agent, an antibacterial agent, a virucidal agent, an anti-viral agent, an insecticidal agent, a nematicidal agent, an antiparasitic agent, or an insect repellent), or a therapeutic agent) that is or can be associated with PMPs (e.g., loaded into or not PMPs, (e.g., encapsulated by, embedded in, or conjugated to PMPs)) using in vivo or in vitro methods and is capable of effecting the recited result (e.g., increasing or decreasing the fitness of an animal, plant, plant pest, plant symbiont, animal (e.g., human) pathogen, or animal pathogen vector) in accordance with the present compositions or methods.


As used herein, the term “agricultural agent” refers to an agent that can act on a plant, a plant pest, or a plant symbiont, such as a pesticidal agent, pest repellent, fertilizing agent, herbicidal agent, plant-modifying agent, or plant-symbiont modifying agent.


As used herein, the term “fertilizing agent” refers to an agent that is capable of increasing the fitness of a plant (e.g., a plant nutrient or a plant growth regulator) or a plant symbiont (e.g., a nucleic acid or a peptide).


As used herein, the term “pesticidal agent” refers to an agent, composition, or substance therein, that controls or decreases the fitness (e.g., kills or inhibits the growth, proliferation, division, reproduction, or spread) of an agricultural, environmental, or domestic/household pest, such as an insect, mollusk, nematode, fungus, bacterium, weed, or virus. Pesticides are understood to include naturally occurring or synthetic insecticides (larvicides or adulticides), insect growth regulators, acaricides (miticides), molluscicides, nematicides, ectoparasiticides, bactericides, fungicides, or herbicides. The term “pesticidal agent” may further encompass other bioactive molecules such as antibiotics, antivirals pesticides, antifungals, antihelminthics, nutrients, and/or agents that stun or slow insect movement.


As used herein, the term “plant-modifying agent” refers to an agent that can alter the genetic properties (e.g., increase gene expression, decrease gene expression, or otherwise alter the nucleotide sequence of DNA or RNA) or biochemical properties of a plant in a manner the results in an increase in plant fitness.


As used herein, the term “pathogen control agent” refers to an agent that can act on an animal (e.g., a human), an animal pathogen, or a pathogen vector, such as an antifungal agent, an antibacterial agent, a virucidal agent, an anti-viral agent, an insecticidal agent, a nematicidal agent, an antiparasitic agent, or an insect repellent.


As used herein, the term “therapeutic agent” refers to an agent that promotes, improves, or stabilizes the health of a mammal, such as a human or a non-human agricultural animal. Therapeutic agents include pathogen control agents (e.g., agents having antipathogen activity (e.g., antibacterial, antifungal, antinematicidal, antiparasitic, or antiviral activity) and agents used for the prevention or treatment of a condition or a disease. Exemplary therapeutic agents include, e.g., small molecules, nucleic acids (e.g., siRNA, miRNA, and mRNA), peptides, proteins, antibodies and antibody fragments, antigens, enzymes, gene editing proteins, and vaccines.


As used herein, “increase the fitness of a plant” refers to an increase in the fitness of the plant directly resulting from contact with a PMP composition described herein and includes, for example, an improved yield, improved vigor of the plant, or improved quality or amount of a harvested product from the plant, an improvement in pre- or post-harvest traits deemed desirable for agriculture or horticulture (e.g., taste, appearance, shelf life), or for an improvement of traits that otherwise benefit humans (e.g., decreased allergen production). An improved yield of a plant relates to an increase in the yield of a product (e.g., as measured by plant biomass, grain, seed or fruit yield, protein content, carbohydrate or oil content or leaf area) of the plant by a measurable amount over the yield of the same product of the plant produced under the same conditions, but without the application of the instant compositions or compared with application of conventional plant-modifying agents (e.g., plant-modifying agents delivered without a PMP). For example, yield can be increased by at least about 0.5%, about 1%, about 2%, about 3%, about 4%, about 5%, about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 100%, or more than 100%. Yield can be expressed in terms of an amount by weight or volume of the plant or a product of the plant on some basis. The basis can be expressed in terms of time, growing area, weight of plants produced, or amount of a raw material used. An increase in the fitness of plant can also be measured in other ways, such as by an increase or improvement of the vigor rating, increase in the stand (the number of plants per unit of area), increase in plant height, increase in stalk circumference, increase in plant canopy, improvement in appearance (such as greener leaf color as measured visually), improvement in root rating, increase in seedling emergence, protein content, increase in leaf size, increase in leaf number, fewer dead basal leaves, increase in tiller strength, decrease in nutrient or fertilizer requirements, increase in seed germination, increase in tiller productivity, increase in flowering, increase in seed or grain maturation or seed maturity, less plant lodging, increased shoot growth, or any combination of these factors, by a measurable or noticeable amount over the same factor of the plant produced under the same conditions, but without the administration of the instant compositions or with application of conventional agricultural agents.


As used herein, the term “pest” refers to organisms that cause damage to plants or other organisms, are present where they are not wanted, or otherwise are detrimental to humans, for example, by impacting human agricultural methods or products. Pests may include, for example, invertebrates (e.g., insects, nematodes, or mollusks), microorganisms (e.g., phytopathogens, endophytes, obligate parasites, facultative parasites, or facultative saprophytes), such as bacteria, fungi, or viruses; or weeds.


As used herein, the term “formulated for delivery to a plant” refers to a PMP composition that includes an agriculturally acceptable carrier. As used herein, an “agriculturally acceptable” carrier or excipient is one that is suitable for use in agriculture, e.g., for use on plants. In certain embodiments the agriculturally acceptable carrier or excipient does not have undue adverse side effects to the plants, the environment, or to humans or animals who consume the resulting agricultural products derived therefrom commensurate with a reasonable benefit/risk ratio.


As used herein, the term “formulated for delivery to an animal” refers to a PMP composition that includes a pharmaceutically acceptable carrier. As used herein, a “pharmaceutically acceptable” carrier or excipient is one that is suitable for administration to an animal (e.g., human), e.g., without undue adverse side effects to the animal (e.g., human or agricultural animal such as a cow, pig, steer, chicken, or turkey).


As used herein, the term “plant” refers to whole plants, plant organs, plant tissues, seeds, plant cells, seeds, and progeny of the same. Plant cells include, without limitation, cells from seeds, suspension cultures, embryos, meristematic regions, callus tissue, leaves, roots, shoots, gametophytes, sporophytes, pollen, and microspores. Plant parts include differentiated and undifferentiated tissues including, but not limited to the following: roots, stems, shoots, leaves, pollen, seeds, fruit, harvested produce, tumor tissue, sap (e.g., xylem sap and phloem sap), and various forms of cells and culture (e.g., single cells, protoplasts, embryos, and callus tissue). In some aspects, the plant or plant part is pectin-rich. In some examples, the plant is a citrus plant, e.g., a grapefruit or a lemon. In some examples, the plant part is a juice sac, e.g. a juice sac of a grapefruit or a juice sac of a lemon. In other examples, the plant is Arabidopsis.


As used herein, the term “plant culture” refers to a plant or a plurality of plants, plant parts, plant cells, or plant tissue that is propagated in or on a medium, e.g., a liquid, gaseous, gel, semi-solid, or solid medium. Plant culture includes, but is not limited to, culture of naturally occurring plants, plant parts, plant cells, or plant tissue or genetically modified plants, plant parts, plant cells, or plant tissues. Plant cultures can be classified, for example, as unorganized cultures (e.g., plant cell cultures such as callus, suspension, or protoplast cultures) or organized cultures (such as root, seedling, embryo, or entire plant cultures) depending on the tissue source and the level of differentiation of the cultured plant material. The plant culture may be a hydroponic culture. As used herein, the term “hydroponic” refers to a hydrated growth system for a plant or plant part (e.g., a plant root) that does not include a natural soil. Such hydroponic growth systems include, e.g., a plant growth system comprising a liquid or semi-liquid (e.g., aqueous), gel, semi-solid, or hydrated solid culture medium. Hydroponic cultures may include aquaponic, hydroculture, or aquaculture growth systems.


As used herein, the term “plant extracellular vesicle”, “plant EV”, or “EV” refers to an enclosed lipid-bilayer structure naturally occurring in a plant. Optionally, the plant EV includes one or more plant EV markers. As used herein, the term “plant EV marker” refers to a component that is naturally associated with a plant, such as a plant protein, a plant nucleic acid, a plant small molecule, a plant lipid, or a combination thereof, including but not limited to any of the plant EV markers listed in the Appendix. In some instances, the plant EV marker is an identifying marker of a plant EV but is not a pesticidal agent. In some instances, the plant EV marker is an identifying marker of a plant EV and also a pesticidal agent (e.g., either associated with or encapsulated by the plurality of PMPs, or not directly associated with or encapsulated by the plurality of PMPs).


As used herein, the term “plant messenger pack” or “PMP” refers to a lipid structure (e.g., a lipid bilayer, unilamellar, multilamellar structure; e.g., a vesicular lipid structure), that is about 5-2000 nm (e.g., at least 5-1000 nm, at least 5-500 nm, at least 400-500 nm, at least 25-250 nm, at least 50-150 nm, or at least 70-120 nm) in diameter that is derived from (e.g., enriched, isolated or purified from) a plant source or segment, portion, or extract thereof, including lipid or non-lipid components (e.g., peptides, nucleic acids, or small molecules) associated therewith and that has been enriched, isolated or purified from a plant, a plant part, or a plant cell, the enrichment or isolation removing one or more contaminants or undesired components from the source plant. PMPs may be highly purified preparations of naturally occurring EVs. Preferably, at least 1% of contaminants or undesired components from the source plant are removed (e.g., at least 2%, 5%, 10%, 15%, 20%, 25%, 30%, 40%, 45%, 50%, 55%, 60%, 70%, 80%, 90%, 95%, 96%, 98%, 99%, or 100%) of one or more contaminants or undesired components from the source plant, e.g., plant cell wall components; pectin; plant organelles (e.g., mitochondria; plastids such as chloroplasts, leucoplasts or amyloplasts; and nuclei); plant chromatin (e.g., a plant chromosome); or plant molecular aggregates (e.g., protein aggregates, protein-nucleic acid aggregates, lipoprotein aggregates, or lipido-proteic structures). Preferably, a PMP is at least 30% pure (e.g., at least 40% pure, at least 50% pure, at least 60% pure, at least 70% pure, at least 80% pure, at least 90% pure, at least 99% pure, or 100% pure) relative to the one or more contaminants or undesired components from the source plant as measured by weight (w/w), spectral imaging (% transmittance), or conductivity (S/m).


PMPs may optionally include additional agents, such as heterologous functional agents, e.g., pesticidal agents, fertilizing agents, plant-modifying agents, therapeutic agents, polynucleotides, polypeptides, or small molecules. The PMPs can carry or associate with additional agents (e.g., heterologous functional agents) in a variety of ways to enable delivery of the agent to a target plant, e.g., by encapsulating the agent, incorporation of the agent in the lipid bilayer structure, or association of the agent (e.g., by conjugation) with the surface of the lipid bilayer structure. Heterologous functional agents can be incorporated into the PMPs either in vivo (e.g., in planta) or in vitro (e.g., in tissue culture, in cell culture, or synthetically incorporated).


As used herein, the term “repellent” refers to an agent, composition, or substance therein, that deters pests from approaching or remaining on a plant or a pathogen vector (e.g., insects, e.g., mosquitos, ticks, mites, or lice) from approaching or remaining on an animal. A repellent may, for example, decrease the number of pests on or in the vicinity of a plant, but may not necessarily kill or decrease the fitness of the pest.


As used herein, the term “stable PMP composition” (e.g., a composition including loaded or non-loaded PMPs) refers to a PMP composition that over a period of time (e.g., at least 24 hours, at least 48 hours, at least 1 week, at least 2 weeks, at least 3 weeks, at least 4 weeks, at least 30 days, at least 60 days, or at least 90 days) retains at least 5% (e.g., at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100%) of the initial number of PMPs (e.g., PMPs per mL of solution) relative to the number of PMPs in the PMP composition (e.g., at the time of production or formulation) optionally at a defined temperature range (e.g., a temperature of at least 24° C. (e.g., at least 24° C., 25° C., 26° C., 27° C., 28° C., 29° C., or 30° C.), at least 20° C. (e.g., at least 20° C., 21° C., 22° C., or 23° C.), at least 4° C. (e.g., at least 5° C., 10° C., or 15° C.), at least −20° C. (e.g., at least −20° C., −15° C., −10° C., −5° C., or 0° C.), or −80° C. (e.g., at least −80° C., −70° C., −60° C., −50° C., −40° C., or −30° C.)); or retains at least 5% (e.g., at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100%) of its activity (e.g., fertilizing, pesticidal, and/or repellent activity) relative to the initial activity of the PMP (e.g., at the time of production or formulation) optionally at a defined temperature range (e.g., a temperature of at least 24° C. (e.g., at least 24° C., 25° C., 26° C., 27° C., 28° C., 29° C., or 30° C.), at least 20° C. (e.g., at least 20° C., 21° C., 22° C., or 23° C.), at least 4° C. (e.g., at least 5° C., 10° C., or 15° C.), at least −20° C. (e.g., at least −20° C., −15° C., −10° C., −5° C., or 0° C.), or −80° C. (e.g., at least −80° C., −70° C., −60° C., −50° C., −40° C., or −30° C.)).





BRIEF DESCRIPTION OF THE DRAWINGS

The application file contains at least one drawing executed in color. Copies of this patent or patent application with color drawings will be provided by the Office upon request and payment of the necessary fee.



FIG. 1A is an exemplary workflow for grapefruit PMP production using a blender, ultracentrifugation, and sucrose gradient purification, which resulted in gelling at all production steps.



FIG. 1B is an exemplary workflow for grapefruit PMP production using a milder juice extraction method by gently pressing isolated juice sacs through a mesh filter (strainer), followed by ultracentrifugation and sucrose gradient purification. This production process resulted in gelling at all steps of the production process.



FIG. 1C is an exemplary workflow for producing PMPs from the juice of one grapefruit using a juice press, followed by differential centrifugation to remove large debris, 20× concentration of the juice using TFF, and size exclusion chromatography to isolate the PMP containing fractions. The PMP fractions are analyzed for PMP concentration (NanoFCM), Particle size (NanoFCM) and protein concentration (bicinchoninic acid assay (BCA)).



FIG. 1D is a scatter plot showing PMP final concentration (PMPs/mL) in PMP-containing size exclusion chromatography (SEC) fractions. PMPs are eluted in fractions 4-6.



FIG. 1E is a size distribution plot of different SEC elution fractions and a table indicating the PMP size distribution per SEC fraction as measured by NanoFCM.



FIG. 2A is an exemplary workflow for PMP production from 1 liter of grapefruit juice (˜7 grapefruits) using a juice press, followed by differential centrifugation to remove large debris, 100× concentration of the juice using tangential flow filtration (TFF), and size exclusion chromatography to isolate the PMP-containing fractions. The PMP fractions are analyzed for PMP concentration (NanoFCM), particle size (NanoFCM) and protein concentration (BCA).



FIG. 2B is a set of graphs showing PMP production in 150 mL of grapefruit juice (1 grapefruit) and 1000 mL of grapefruit juice. The upper panels show the results of a BCA assay. The lower panels show PMP yield, as measured by NanoFCM.



FIG. 3A is an exemplary workflow of a PMP production process for enhanced removal of contaminants comprising incubation with 500 mM EDTA (pH 8.6) to a final concentration of 50 mM EDTA (pH 7.2-8); dialysis; TFF; and size exclusion chromatography.



FIG. 3B is a graph showing that incubation of the crude grapefruit PMP fraction with a final concentration of 50 mM EDTA (pH 7.2-8), followed by overnight dialysis using a 300 kDa membrane, successfully removes contaminants present in the late elution fractions after SEC, as shown by absorbance at 280 nm. Arrow indicates peak containing contaminants. Dialysis buffers used were PBS without calcium/magnesium pH 7.4, MES pH 6, and Tris pH 8.6.



FIG. 3C is a graph showing that incubation of the crude grapefruit PMP fraction with a final concentration of 50 mM EDTA, pH 7.2-8, followed by overnight dialysis using a 300 kDa membrane, successfully removes contaminants present in the late elution fractions after SEC, as shown by BCA protein analysis, which is sensitive to the presence of sugars and pectins. Arrow indicates peak containing contaminants. Dialysis buffers used were PBS without calcium/magnesium pH 7.4, MES pH 6, and Tris pH 8.6.



FIG. 4A is an exemplary workflow describing the crude production of PMPs from citrus fruit or plant cell culture. Briefly, juice or culture medium is collected and subsequently centrifuged at 1000×g for 10 minutes, 3000× g for 20 minutes, and 10,000×g for 40 minutes to remove large debris to produce the crude PMP fraction.



FIG. 4B is an exemplary workflow describing the production of pure PMPs and subsequent characterization methods. Briefly, PMPs are incubated in a final concentration of 50 mM EDTA (pH 7) for 30 minutes, and subsequently passaged through a 1 μm and a 0.45 μm filter. Filtered juice or medium is concentrated 5× by Tangential Flow Filtration (TFF) with PBS washing, and dialyzed overnight in PBS using a 300 kDa dialysis membrane to remove contaminants. Subsequently, the dialyzed juice is further concentrated by TFF to a final concentration of 20×. Size exclusion chromatography is then used to elute the PMP-containing fractions.



FIG. 5A is a photograph of a lemon juice preparation treated with 6 units (6 U) pectinase (+pectinase) or not treated with pectinase (-pectinase). Images were taken with an iPhone to show the difference in turbidity FIG. 5B is a photograph of grapefruit juice treated with 0.5 U pectinase (+pectinase) or not treated with pectinase (-pectinase). Images were taken with an iPhone to show the difference in turbidity.



FIG. 5C is a bar graph showing turbidity of pectinase-treated and untreated juice, as quantified as the volume of juice processed per filter.



FIG. 6 is a bar graph of grapefruit PMP concentration measured by nano-flow cytometry (NanoFCM) for PMP preparations produced from pectinase-treated and untreated juice.



FIG. 7A is an exemplary workflow of PMPs that were purified from 4 liters of pectinase and EDTA treated grapefruit juice as described above, and were concentrated 5× using a Spectrum 300 kDa TFF, washed by 6 volume exchanges with PBS, and concentrated to a final concentration of 20×. Next, size exclusion chromatography was used to elute the PMP-containing fractions.



FIG. 7B is a graph showing the absorbance at 280 nm (NanoDrop) of eluted SEC fractions produced by the method shown in FIG. 7A of 9 different columns (A-J), showing the efficient removal of the pectin, sugars, protein and other contaminants in the late SEC fractions, while PMPs are detected in early SEC fractions 3-7.



FIG. 7C is a graph showing the protein concentration (BCA) of eluted SEC fractions produced by the method shown in FIG. 7A of 9 different columns (A-J), indicating the efficient removal of the pectin, sugars, protein and other contaminants in the late SEC fractions, while PMPs are detected in early SEC fractions 3-7.



FIG. 8A is a graph showing the light transmittance spectrum of standard concentrations of pectin (0.1-1%) dissolved in ultrapure water. The transmittance spectrum was measured on a SpectraMax i3×.



FIG. 8B is a graph showing the light transmittance spectrum of grapefruit juice that was treated with pectinase compared to untreated juice.



FIG. 9A is a diagram showing an experimental overview of the treatment of alfalfa sprouts with DyLight800 nm-labeled PMPs that were produced with or without pectinase treatment.



FIG. 9B is an infrared heatmap showing that the removal of pectins during Lemon PMP production, does not affect uptake in alfalfa sprouts. PMPs are labeled with DyLight™800 (DL800). Infrared images are taken on an Odyssey scanner, and a heat map of PMP uptake is shown.



FIG. 10A is a schematic diagram showing a protocol for grapefruit PMP production using a destructive juicing step involving the use of a blender, followed by ultracentrifugation and sucrose gradient purification. Images are included of the grapefruit juice after centrifugation at 1000×g for 10 min and the sucrose gradient band pattern after ultracentrifugation at 150,000×g for 2 hours.



FIG. 10B is a plot of the PMP particle distribution measured by the Spectradyne NCS1.



FIG. 11 is a schematic diagram showing a protocol for grapefruit PMP production using a mild juicing step involving use of a mesh filter, followed by ultracentrifugation and sucrose gradient purification. Images are included of the grapefruit juice after centrifugation at 1000×g for 10 min and the sucrose gradient band pattern after ultracentrifugation at 150,000×g for 2 hours.



FIG. 12A is a schematic diagram showing a protocol for grapefruit PMP production using ultracentrifugation, followed by size exclusion chromatography (SEC) to isolate the PMP-containing fractions. The eluted SEC fractions are analyzed for particle concentration (NanoFCM), median particle size (NanoFCM), and protein concentration (BCA).



FIG. 12B is a graph showing particle concentration per mL in eluted size exclusion chromatography (SEC) fractions (NanoFCM). The fractions containing the majority of PMPs (“PMP fraction”) are indicated with an arrow. PMPs are eluted in fractions 2-4.



FIG. 12C is a set of graphs and a table showing particle size in nm for selected SEC fractions, as measured using NanoFCM. The graphs show PMP size distribution in fractions 1, 3, 5, and 8.



FIG. 12D is a graph showing protein concentration in μg/mL in SEC fractions, as measured using a BCA assay. The fraction containing the majority of PMPs (“PMP fraction”) is labeled, and an arrow indicates a fraction containing contaminants.



FIG. 13A is a schematic diagram showing a protocol for scaled PMP production from 1 liter of grapefruit juice (˜7 grapefruits) using a juice press, followed by differential centrifugation to remove large debris, 100× concentration of the juice using TFF, and size exclusion chromatography (SEC) to isolate the PMP containing fractions. The SEC elution fractions are analyzed for particle concentration (NanoFCM), median particle size (NanoFCM) and protein concentration (BCA).



FIG. 13B is a pair of graphs showing protein concentration (BCA assay, top panel) and particle concentration (NanoFCM, bottom panel) of SEC eluate volume (ml) from a scaled starting material of 1000 ml of grapefruit juice, showing a high amount of contaminants in the late SEC elution volumes.



FIG. 13C is a graph showing that incubation of the crude grapefruit PMP fraction with a final concentration of 50 mM EDTA, pH 7.15 followed by overnight dialysis using a 300 kDa membrane, successfully removed contaminants present in the late SEC elution fractions, as shown by absorbance at 280 nm. There was no difference in the dialysis buffers used (PBS without calcium/magnesium pH 7.4, MES pH 6, Tris pH 8.6).



FIG. 13D is a graph showing that incubation of the crude grapefruit PMP fraction with a final concentration of 50 mM EDTA, pH 7.15, followed by overnight dialysis using a 300 kDa membrane, successfully removed contaminants present in the late elution fractions after SEC, as shown by BCA protein analysis, which, besides detecting protein, is sensitive to the presence of sugars and pectins. There was no difference in the dialysis buffers used (PBS without calcium/magnesium pH 7.4, MES pH 6, Tris pH 8.6).



FIG. 14A is a graph showing particle concentration (particles/ml) in eluted BMS plant cell culture SEC fractions, as measured by nano-flow cytometry (NanoFCM). PMPs were eluted in SEC fractions 4-6.



FIG. 14B is a graph showing absorbance at 280 nm (A.U.) in eluted BMS SEC fractions, measured on a SpectraMax® spectrophotometer. PMPs were eluted in fractions 4-6; fractions 9-13 contained contaminants.



FIG. 14C is a graph showing protein concentration (μg/ml) in eluted BMS SEC fractions, as determined by BCA analysis. PMPs were eluted in fractions 4-6; fractions 9-13 contained contaminants.



FIG. 14D is a scatter plot showing particles in the combined BMS PMP-containing SEC fractions as measured by nano-flow cytometry (NanoFCM). PMP concentration (particles/ml) was determined using a bead standard according to NanoFCM's instructions.



FIG. 14E is a graph showing the size distribution of BMS PMPs (nm) for the gated particles (background subtracted) of FIG. 14D. Median PMP size (nm) was determined using Exo bead standards according to NanoFCM's instructions.



FIG. 15A is a scatter plot and a graph showing particle size in AF488-labeled lemon PMPs as measured by nanoflow cytometry (NanoFCM). The top panel is a scatter plot showing AF488-labeled lemon PMPs. Particles were gated on the FITC fluorescence signal, relative to unlabeled particles and background signal. The labeling efficiency was 89.4% as determined by the number of fluorescent particles relative to the total number of particles detected. The final AF488-PMP concentration (2.91×1012 PMPs/ml) was determined from the number of fluorescent particles and using a bead standard with a known concentration according to NanoFCM's instructions. The bottom panel is a size (nm) distribution graph of 488-labeled lemon PMPs. The median PMP size was determined using Exo bead standards according to NanoFCM's instructions. The median lemon AF488-PMPs size was 79.4 nm+/−14.7 nm (SD).



FIG. 15B is a set of photomicrographs showing uptake of lemon (LM) PMPs labeled with Alexa Fluor® 488 (AF488) by the plant cell lines Glycine max (soy bean), Tritium aestivum (wheat), and maize BMS cell culture. Brightfield panels show the position of cells; panels labeled “GFP” show fluorescence of AF488. Uptake of PMPs by a cell is indicated by the presence of the AF488 signal in the cell. Free AF488 (“Free dye”) is shown as a control.



FIG. 16 is a pair of diagrams and a set of photomicrographs showing uptake of lemon (LM) and grapefruit (GF) PMPs labeled with DL800 by Arabidopsis thaliana seedlings and alfalfa sprouts. Intensity of fluorescence of DL800 dye is displayed. Intensity of fluorescence was measured at 22 hpt (hours post-treatment) for Arabidopsis thaliana seedlings and at 24 hpt for alfalfa sprouts. Seedlings incubated with no dye (“negative control”) and with free DL800 dye (“DL800 dye only”) are shown as controls.





DETAILED DESCRIPTION OF THE INVENTION

Featured herein are methods for manufacturing of industrial and scaled preparations of PMPs, e.g., methods of manufacturing commercially acceptable and/or pharmaceutically acceptable preparations of PMPs, e.g., Good Manufacturing Practices (GMP) preparations of PMPs. Such methods may include one or more of chelation, enzymatic digestion, and differential separation (e.g., by centrifugation or tangential flow filtration), which will, e.g., clarify the solution, reduce its viscosity, reduce undesired components or contaminants, and/or enrich the preparations in PMPs so as to enable utilization at higher volume/mass scales. The PMPs manufactured using the methods herein are useful in a variety of agricultural and therapeutic compositions and methods.


I. PMP Production Methods

A plant messenger pack (PMP) is a lipid (e.g., lipid bilayer, unilamellar, or multilamellar structure) structure that includes a plant EV, or segment, portion, or extract (e.g., lipid extract) thereof. A plant EV is an enclosed lipid-bilayer structure that naturally occurs in a plant. Plant EVs may be about 5-2000 nm in diameter. Plant EVs can originate from a variety of plant biogenesis pathways. In nature, plant EVs can be found in the intracellular and extracellular compartments of plants, such as the plant apoplast, the compartment located outside the plasma membrane and formed by a continuum of cell walls and the extracellular space. Alternatively, PMPs can be enriched plant EVs found in cell culture media upon secretion from plant cells. Plant EVs can be separated from plants (e.g., from the apoplastic fluid), thereby providing PMPs, by a variety of methods further described herein. Further, the PMPs can optionally include a heterologous functional agent (e.g., a heterologous agricultural agent (e.g., pesticidal agent, fertilizing agent, herbicidal agent, plant-modifying agent) or a heterologous therapeutic agent (e.g., a pathogen control agent such as an antifungal agent, an antibacterial agent, a virucidal agent, an anti-viral agent, an insecticidal agent, a nematicidal agent, an antiparasitic agent, or an insect repellent)), which may be introduced (e.g., loaded into or onto the PMP) in vivo or in vitro.


As such, the PMPs can include a heterologous functional agent that is loaded into or onto the PMP by the plant from which the PMP is produced. For example, the pesticidal agent loaded in to the PMP in vivo may be a factor endogenous to the plant or a factor exogenous to the plant (e.g., as expressed by a heterologous genetic construct in a genetically engineered plant). Alternatively, the PMPs may be loaded with a heterologous functional agent in vitro (e.g., following production by a variety of methods further described herein).


PMPs can include plant EVs, or segments, portions, or extracts, thereof, in which the plant EVs are about 5-2000 nm in diameter. For example, the PMP can include a plant EV, or segment, portion, or extract thereof, that has a mean diameter of about 5-50 nm, about 50-100 nm, about 100-150 nm, about 150-200 nm, about 200-250 nm, about 250-300 nm, about 300-350 nm, about 350-400 nm, about 400-450 nm, about 450-500 nm, about 500-550 nm, about 550-600 nm, about 600-650 nm, about 650-700 nm, about 700-750 nm, about 750-800 nm, about 800-850 nm, about 850-900 nm, about 900-950 nm, about 950-1000 nm, about 1000-1250 nm, about 1250-1500 nm, about 1500-1750 nm, or about 1750-2000 nm. In some instances, the PMP includes a plant EV, or segment, portion, or extract thereof, that has a mean diameter of about 5-950 nm, about 5-900 nm, about 5-850 nm, about 5-800 nm, about 5-750 nm, about 5-700 nm, about 5-650 nm, about 5-600 nm, about 5-550 nm, about 5-500 nm, about 5-450 nm, about 5-400 nm, about 5-350 nm, about 5-300 nm, about 5-250 nm, about 5-200 nm, about 5-150 nm, about 5-100 nm, about 5-50 nm, or about 5-25 nm. In certain instances, the plant EV, or segment, portion, or extract thereof, has a mean diameter of about 50-200 nm. In certain instances, the plant EV, or segment, portion, or extract thereof, has a mean diameter of about 50-300 nm. In certain instances, the plant EV, or segment, portion, or extract thereof, has a mean diameter of about 200-500 nm. In certain instances, the plant EV, or segment, portion, or extract thereof, has a mean diameter of about 30-150 nm.


In some instances, the PMP may include a plant EV, or segment, portion, or extract thereof, that has a mean diameter of at least 5 nm, at least 50 nm, at least 100 nm, at least 150 nm, at least 200 nm, at least 250 nm, at least 300 nm, at least 350 nm, at least 400 nm, at least 450 nm, at least 500 nm, at least 550 nm, at least 600 nm, at least 650 nm, at least 700 nm, at least 750 nm, at least 800 nm, at least 850 nm, at least 900 nm, at least 950 nm, or at least 1000 nm. In some instances, the PMP includes a plant EV, or segment, portion, or extract thereof, that has a mean diameter less than 1000 nm, less than 950 nm, less than 900 nm, less than 850 nm, less than 800 nm, less than 750 nm, less than 700 nm, less than 650 nm, less than 600 nm, less than 550 nm, less than 500 nm, less than 450 nm, less than 400 nm, less than 350 nm, less than 300 nm, less than 250 nm, less than 200 nm, less than 150 nm, less than 100 nm, or less than 50 nm. A variety of methods (e.g., a dynamic light scattering method) standard in the art can be used to measure the particle diameter of the plant EV, or segment, portion, or extract thereof.


In some instances, the PMP may include a plant EV, or segment, portion, or extract thereof, that has a mean surface area of 77 nm2 to 3.2×106 nm2 (e.g., 77-100 nm2, 100-1000 nm2, 1000-1×104 nm2, 1×104-1×105 nm2, 1×105-1×106 nm2, or 1×106-3.2×106 nm2). In some instances, the PMP may include a plant EV, or segment, portion, or extract thereof, that has a mean volume of 65 nm3 to 5.3×108 nm3 (e.g., 65-100 nm3, 100-1000 nm3, 1000-1×104 nm3, 1×104-1×105 nm3, 1×105-1×106 nm3, 1×106-1×107 nm3, 1×107-1×108 nm3, 1×108-5.3×108 nm3). In some instances, the PMP may include a plant EV, or segment, portion, or extract thereof, that has a mean surface area of at least 77 nm2, (e.g., at least 77 nm2, at least 100 nm2, at least 1000 nm2, at least 1×104 nm2, at least 1×105 nm2, at least 1×106 nm2, or at least 2×106 nm2). In some instances, the PMP may include a plant EV, or segment, portion, or extract thereof, that has a mean volume of at least 65 nm3 (e.g., at least 65 nm3, at least 100 nm3, at least 1000 nm3, at least 1×104 nm3, at least 1×105 nm3, at least 1×106 nm3, at least 1×107 nm3, at least 1×108 nm3, at least 2×108 nm3, at least 3×108 nm3, at least 4×108 nm3, or at least 5×108 nm3.


In some instances, the PMP can have the same size as the plant EV or segment, extract, or portion thereof. Alternatively, the PMP may have a different size than the initial plant EV from which the PMP is produced. For example, the PMP may have a diameter of about 5-2000 nm in diameter. For example, the PMP can have a mean diameter of about 5-50 nm, about 50-100 nm, about 100-150 nm, about 150-200 nm, about 200-250 nm, about 250-300 nm, about 300-350 nm, about 350-400 nm, about 400-450 nm, about 450-500 nm, about 500-550 nm, about 550-600 nm, about 600-650 nm, about 650-700 nm, about 700-750 nm, about 750-800 nm, about 800-850 nm, about 850-900 nm, about 900-950 nm, about 950-1000 nm, about 1000-1200 nm, about 1200-1400 nm, about 1400-1600 nm, about 1600-1800 nm, or about 1800-2000 nm. In some instances, the PMP may have a mean diameter of at least 5 nm, at least 50 nm, at least 100 nm, at least 150 nm, at least 200 nm, at least 250 nm, at least 300 nm, at least 350 nm, at least 400 nm, at least 450 nm, at least 500 nm, at least 550 nm, at least 600 nm, at least 650 nm, at least 700 nm, at least 750 nm, at least 800 nm, at least 850 nm, at least 900 nm, at least 950 nm, at least 1000 nm, at least 1200 nm, at least 1400 nm, at least 1600 nm, at least 1800 nm, or about 2000 nm. A variety of methods (e.g., a dynamic light scattering method) standard in the art can be used to measure the particle diameter of the PMPs. In some instances, the size of the PMP is determined following loading of heterologous functional agents, or following other modifications to the PMPs.


In some instances, the PMP may have a mean surface area of 77 nm2 to 1.3×107 nm2 (e.g., 77-100 nm2, 100-1000 nm2, 1000-1×104 nm2, 1×104-1×105 nm2, 1×105-1×106 nm2, or 1×106-1.3×107 nm2).


In some instances, the PMP may have a mean volume of 65 nm3 to 4.2×109 nm3 (e.g., 65-100 nm3, 100-1000 nm3, 1000-1×104 nm3, 1×104-1×105 nm3, 1×105-1×106 nm3, 1×106-1×107 nm3, 1×107-1×108 nm3, 1×108-1×109 nm3, or 1×109-4.2×109 nm3). In some instances, the PMP has a mean surface area of at least 77 nm2, (e.g., at least 77 nm2, at least 100 nm2, at least 1000 nm2, at least 1×104 nm2, at least 1×105 nm2, at least 1×106 nm2, or at least 1×107 nm2). In some instances, the PMP has a mean volume of at least 65 nm3 (e.g., at least 65 nm3, at least 100 nm3, at least 1000 nm3, at least 1×104 nm3, at least 1×105 nm3, at least 1×106 nm3, at least 1×107 nm3, at least 1×108 nm3, at least 1×109 nm3, at least 2×109 nm3, at least 3×109 nm3, or at least 4×109 nm3).


In some instances, the PMP may include an intact plant EV. Alternatively, the PMP may include a segment, portion, or extract of the full surface area of the vesicle (e.g., a segment, portion, or extract including less than 100% (e.g., less than 90%, less than 80%, less than 70%, less than 60%, less than 50%, less than 40%, less than 30%, less than 20%, less than 10%, less than 10%, less than 5%, or less than 1%) of the full surface area of the vesicle) of a plant EV. The segment, portion, or extract may be any shape, such as a circumferential segment, spherical segment (e.g., hemisphere), curvilinear segment, linear segment, or flat segment. In instances where the segment is a spherical segment of the vesicle, the spherical segment may represent one that arises from the splitting of a spherical vesicle along a pair of parallel lines, or one that arises from the splitting of a spherical vesicle along a pair of non-parallel lines. Accordingly, the plurality of PMPs can include a plurality of intact plant EVs, a plurality of plant EV segments, portions, or extracts, or a mixture of intact and segments of plant EVs. One skilled in the art will appreciate that the ratio of intact to segmented plant EVs will depend on the particular isolation method used. For example, grinding or blending a plant, or part thereof, may produce PMPs that contain a higher percentage of plant EV segments, portions, or extracts than a non-destructive extraction method, such as vacuum-infiltration.


In instances where, the PMP includes a segment, portion, or extract of a plant EV, the EV segment, portion, or extract may have a mean surface area less than that of an intact vesicle, e.g., a mean surface area less than 77 nm2, 100 nm2, 1000 nm2, 1×104 nm2, 1×105 nm2, 1×106 nm2, or 3.2×106 nm2). In some instances, the EV segment, portion, or extract has a surface area of less than 70 nm2, 60 nm2, 50 nm2, 40 nm2, 30 nm2, 20 nm2, or 10 nm2). In some instances, the PMP may include a plant EV, or segment, portion, or extract thereof, that has a mean volume less than that of an intact vesicle, e.g., a mean volume of less than 65 nm3, 100 nm3, 1000 nm3, 1×104 nm3, 1×105 nm3, 1×106 nm3, 1×107 nm3, 1×108 nm3, or 5.3×108 nm3).


In instances where the PMP includes an extract of a plant EV, e.g., in instances where the PMP includes lipids extracted (e.g., with chloroform) from a plant EV, the PMP may include at least 1%, 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60% or more, of lipids extracted (e.g., with chloroform) from a plant EV. The PMPs in the plurality may include plant EV segments and/or plant EV-extracted lipids or a mixture thereof.


Further outlined herein are details regarding methods of producing PMPs, plant EV markers that can be associated with PMPs, and formulations for compositions including PMPs.


A. Production Methods


PMPs may be produced from plant EVs, or a segment, portion or extract (e.g., lipid extract) thereof, that occur naturally in plants, or parts thereof, including plant tissues or plant cells.


One exemplary method for producing PMPs includes (a) providing a pectin-rich preparation from a plant comprising extracellular vesicles (EVs), the preparation having a turbidity of 0.8 AU or greater at an absorbance of 650 nm; (b) treating the preparation to reduce the turbidity of the preparation or a fraction thereof; and (c) separating PMPs from the preparation or fraction thereof, thereby producing PMPs. In some examples, the turbidity of the preparation of step (a) is 0.5, 0.6, 0.7, 0.8, 0.81, 0.82, 0.83, 0.84, 0.85, 0.86, 0.87, 0.88, 0.89, 0.9, 1.0, 2.0, 3.0, or 4.0 AU or greater. In some examples, the turbidity of the preparation of step (a) is 0.86 or greater. In some examples, the preparation of step (a) has a percent light transmittance of 18% or lower, 17% or lower, 16% or lower, 15% or lower, 14% or lower, 13% or lower, 12% or lower, 11% or lower, 10% or lower, 9% or lower, 8% or lower, 7% or lower, 6% or lower, 5% or lower, 4% or lower, 3% or lower, 2% or lower, or 1% or lower at an absorbance of 650 nm. In some examples, the preparation of step (a) has a percent light transmittance of 14% or lower, e.g., 13.17% or lower. In some examples, the preparation of step (a) has a percent light transmittance of 16% or lower, e.g., 15.84% or lower.


A second exemplary method for producing PMPs includes (a) providing a pectin-rich preparation having a viscosity of at least 1.4 cP at 20° C. from a plant comprising EVs; (b) treating the preparation to reduce the viscosity of the preparation or a fraction thereof; and (c) separating PMPs from the preparation or fraction thereof, thereby producing PMPs.


A third exemplary method for producing PMPs includes (a) providing a pectin-rich preparation from a plant comprising EVs; (b) treating the preparation with an agent that reduces pectin gelation; (c) concentrating the preparation, wherein the viscosity of the concentrated preparation is reduced by at least 10% relative to a concentrated preparation that has not been treated with the agent that reduces pectin gelation; and (d) separating PMPs from the preparation or fraction thereof, thereby producing PMPs.


In some examples, the viscosity of the concentrated preparation is reduced by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, or 100% relative to a concentrated preparation that has not been treated with the agent that reduces pectin gelation. Viscosity of the concentrated preparation may be measured during the concentration or after the concentration, e.g., 5 minutes, 10 minutes, 20 minutes, 30 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, or more than 24 hours after the concentration.


The viscosity of the concentrated preparation that has not been treated with the agent that produces pectin gelation may be e.g., 1.4 cP when viscosity is measured at 20° C. In some examples, the viscosity of the concentrated preparation that has not been treated with the agent that produces pectin gelation is 1.01 cP, 1.1 cP, 1.2 cP, 1.3 cP, 1.4 cP, 1.5 cP, 1.6 cP, 1.7 cP, 1.8 cP, 1.9 cP, 2 cP, 3 cP, 4 cP, 5 cP, 6 cP, 7 cP, 8 cP, 9 cP, 10 cP, 20 cP, 50 cP, 100 cP, 250 cP, 500 cP, 750 cP, 1000 cP, 2000 cP, 5000 cP, 10,000 cP, 20,000 cP, 50,000 cP, 75,000 cP, or more than 75,000 cP at 20° C.


A fourth exemplary method for producing PMPs includes (a) providing a pectin-rich preparation from a plant comprising EVs; (b) treating the preparation to reduce high molecular weight pectin in the preparation or a fraction thereof; and (c) separating PMPs from the preparation or fraction thereof, thereby producing PMPs.


A fifth exemplary method for producing PMPs includes (a) providing a pectin-rich preparation from a plant comprising EVs; (b) contacting the preparation or a fraction thereof with a chelating agent; and (c) separating PMPs from the chelated preparation or fraction thereof, thereby producing PMPs.


A sixth exemplary method for producing PMPs includes (a) processing at least 500 g of a pectin-rich plant or plant part comprising EVs into a preparation; (b) contacting the preparation or a fraction thereof with a chelating agent; and (c) processing the chelated preparation or fraction thereof to separate PMPs, wherein the contacting is performed in an amount and for a time sufficient to reduce high molecular weight pectin in the chelated preparation or fraction thereof by at least 10%. The processing of step (c) may comprise separating the PMPs from the chelated preparation or fraction thereof.


In some examples of the fourth and fifth methods, the chelating agent reduces gelation of pectin in the chelated preparation or fraction thereof,


The chelating agent may be, e.g., ethylenediaminetetraacetic acid (EDTA) or ethylene glycol-bis(β-aminoethyl ether)-N,N,N′,N′-tetraacetic acid (EGTA). The chelating agent may act by chelating (e.g., binding to and diminishing the reactivity of) a metal ion, e.g., a calcium ion (e.g., Ca2+) in the plant preparation, and may diminish the reactivity of the metal ion in the solution by, e.g., at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or 100%. The reactivity of the metal ion in the solution may be quantified as esterification of pectins, e.g., in an assay for viscosity or turbidity of the solution. The chelating agent, e.g., EDTA or EGTA, may be formulated with a buffer, e.g., 2-(N-morpholino)ethanesulfonic acid (MES), tris(hydroxymethyl)aminomethane (Tris), or phosphate buffered saline (PBS). The chelating agent may be formulated with sodium hydroxide (NaOH).


The contacting of the preparation or fraction thereof with the chelating agent may be performed in an amount and for a time sufficient to reduce high molecular weight pectin in the chelated preparation or fraction thereof, e.g., reduce high molecular weight pectin by at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or 100%. The preparation may be contacted with the chelating agent for any suitable amount of time, e.g., 5 minutes, 10 minutes, 15 minutes, 20 minutes, 25 minutes, 30 minutes, 40 minutes, 50 minutes, 1 hour, 1.5 hours, 2 hours, 2.5 hours, 3 hours, 3.5 hours, 4 hours, 4.5 hours, 5 hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 11 hours, 12 hours, 24 hours, or more than 24 hours. The preparation or fraction thereof may be contacted with the chelating agent at any stage in the production process, e.g., contacted with the chelating agent at more than one stage in the production process.


Any one of the above-described methods may further comprise treating the plant preparation with a pectinase enzyme. The pectinase enzyme may be any enzyme or a mixture of enzymes capable of degrading a pectin (e.g., a high molecular weight pectin), e.g., a pectolyase (pectin lyase) enzyme or a polygalacturonase (pectin depolymerase) enzyme.


A seventh exemplary method for producing PMPs includes (a) providing a pectin-rich preparation from a plant comprising EVs; (b) contacting the preparation or a fraction thereof with a pectinase enzyme (e.g., an enzyme or a mixture of enzymes capable of degrading a pectin, e.g., a pectolyase enzyme or a polygalacturonase enzyme; and (c) separating PMPs from the preparation or fraction thereof, thereby producing PMPs.


The contacting of the preparation or fraction thereof with the pectinase enzyme may be performed in an amount and for a time sufficient to reduce high molecular weight pectin in the chelated preparation or fraction thereof, e.g., reduce high molecular weight pectin by at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or 100%. The preparation may be contacted with the pectinase enzyme for any suitable amount of time, e.g., 5 minutes, 10 minutes, 15 minutes, 20 minutes, 25 minutes, 30 minutes, 40 minutes, 50 minutes, 1 hour, 1.5 hours, 2 hours, 2.5 hours, 3 hours, 3.5 hours, 4 hours, 4.5 hours, 5 hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 11 hours, 12 hours, 24 hours, or more than 24 hours. The preparation or fraction thereof may be contacted with the pectinase enzyme at any stage in the production process, e.g., contacted with the pectinase enzyme at more than one stage in the production process.


The method may further involve removal or inactivation of the pectinase enzyme, e.g., inactivation of the pectinase enzyme by exposing the preparation to a temperature and for a time sufficient to deactivate the enzyme.


The PMPs provided herein can include a plant EV, or segment, portion, or extract thereof, isolated from a variety of plants or plant parts. The plant or plant part may be pectin-rich.


In some examples, the pectin-rich plant preparation derived from the plant part has a pectin concentration of at least 0.01%, e.g., has a pectin concentration of at least 0.02%, 0.04%, 0.06%, 0.08%, 0.1%, 0.2%, 0.4%, 0.6%, 0.8%, 1%, 2%, or more than 2%. In some examples, the pectin-rich plant preparation derived from the plant part has a pectin concentration of at least 0.1%. In some examples, the pectin concentration in the PMPs of step (c) is reduced by at least 1% relative to PMPs produced from a preparation that has not been treated, e.g., reduced by at least 1%, 2%, 3%, 4%, 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%, 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%. In some examples, the pectin concentration in the PMPs of step (c) is reduced by at least 10% relative to PMPs produced from a preparation that has not been treated.


In some examples, the pectin-rich plant preparation derived from the plant part has a viscosity of at least 1.01 cP at 20° C., e.g., has a viscosity of at least 1.01 cP, 1.02 cP, 1.03 cP, 1.04 cP, 1.05 cP, 1.1 cP, 1.2 cP, 1.3 cP, 1.4 cP, 1.5 cP, 2 cP, 3 cP, 4 cP, 5 cP, 6 cP, 7 cP, 8 cP, 9 cP, 10 cP, 20 cP, 50 cP, 100 cP, 250 cP, 500 cP, 750 cP, 1000 cP, 2000 cP, 5000 cP, 10,000 cP, 20,000 cP, 50,000 cP, 75,000 cP, or more than 75,000 cP at 20° C. In some examples, the pectin-rich plant preparation derived from the plant part has a viscosity of at least 1.4 cP.


In some examples, the viscosity of the preparation is reduced by at least 1% relative to a preparation that has not been treated (e.g., has not been treated to reduce viscosity, e.g., has not been treated with a chelating agent or a pectinase), e.g., is reduced by at least 1%, 2%, 3%, 4%, 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%, 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%. In some examples, the viscosity of the preparation is reduced by at least 5% relative to a preparation that has not been treated.


The viscosity of the preparation may be monitored. An exemplary method for producing PMPs includes (a) providing a pectin-rich preparation from a plant comprising EVs; (b) (i) treating the preparation to reduce the turbidity of the preparation or a fraction thereof; (ii) treating the preparation to reduce the viscosity of the preparation or a fraction thereof; (iii) treating the preparation to reduce high molecular weight pectin in the preparation or a fraction thereof; (iv) contacting the preparation or a fraction thereof with a chelating agent; or (v) contacting the preparation or a fraction thereof with a pectinase enzyme; (c) intermittently or continuously measuring the viscosity of the preparation or fraction thereof during step (b); (d) ending step (b) when the viscosity of the preparation or fraction thereof is below a predetermined level that informs that the preparation or fraction thereof of step (e) will have reduced gelation relative to a preparation or fraction thereof that has not been treated; and (e) separating PMPs from the preparation or fraction thereof.


Viscosity may be measured using any method known in the art, e.g., measured using a viscometer (e.g., a U-tube viscometer, a falling-sphere viscometer, a falling-piston viscometer, an oscillating-piston viscometer, a vibrational viscometer, a rotational viscometer, a bubble viscometer, or a rectangular slit viscometer) or a rheometer. Viscosity may be measured in-process (e.g., in-process during step (b) of the above-described method). Viscosity may be measured intermittently or continuously, e.g., continuously during all or a portion of step (b) of the above-described method.


The predetermined level of viscosity of step (d) may be, e.g., 1.4 cP when viscosity is measured at 20° C. In some examples, the predetermined level of viscosity of step (d) is 1.0 cP, 1.1 cP, 1.2 cP, 1.3 cP, 1.4 cP, 1.5 cP, 1.6 cP, 1.7 cP, 1.8 cP, 1.9 cP, 2 cP, 3 cP, 4 cP, 5 cP, 6 cP, 7 cP, 8 cP, 9 cP, 10 cP, 20 cP, 50 cP, 100 cP, 250 cP, 500 cP, 750 cP, 1000 cP, 2000 cP, 5000 cP, 10,000 cP, 20,000 cP, 50,000 cP, 75,000 cP, or more than 75,000 cP at 20° C.


The turbidity of the preparation may be monitored. An exemplary method for producing PMPs includes (a) providing a pectin-rich preparation from a plant comprising EVs; (b) (i) treating the preparation to reduce the turbidity of the preparation or a fraction thereof; (ii) treating the preparation to reduce the viscosity of the preparation or a fraction thereof; (iii) treating the preparation to reduce high molecular weight pectin in the preparation or a fraction thereof; (iv) contacting the preparation or a fraction thereof with a chelating agent; or (v) contacting the preparation or a fraction thereof with a pectinase enzyme; (c) intermittently or continuously measuring the turbidity of the preparation or fraction thereof during step (b); (d) ending step (b) when the turbidity of the preparation or fraction thereof is below a predetermined level that informs that the preparation or fraction thereof of step (e) will have reduced gelation relative to a preparation or fraction thereof that has not been treated; and (e) separating PMPs from the preparation or fraction thereof.


Turbidity may be measured using any method known in the art, e.g., measured based on absorbance of light (e.g., absorbance at 650 nm), light scattering (e.g., using a nephelometer), attenuation of a light beam (e.g., a Jackson Candle method), or visibility of a marker (e.g., a Secchi disk). Turbidity may be measured in-process (e.g., in-process during step (b) of the above-described method). Turbidity may be measured intermittently or continuously, e.g., continuously during all or a portion of step (b) of the above-described method. In some instances, turbidity is measured in a diluted sample.


The predetermined level of turbidity of step (d) may be, e.g., 0.8 arbitrary units (AU) absorbance as measured at 650 nm. In some examples, the predetermined level of turbidity of step (d) is 0, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1, 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3.0, 3.1, 3.2, 3.3, 3.4, 3.5, 3.6, 3.7, 3.8, 3.9, 4.0, 4.1, 4.2, 4.3, 4.4, 4.5, 4.6, 4.7, 4.8, 4.9, or 5.0 AU at an absorbance of 650 nm.


Upon isolating the plant EVs, thereby producing PMPs, the PMPs can be separated or collected into a crude PMP fraction. For instance, the separating step may involve separating the plurality of PMPs into a crude PMP fraction using centrifugation (e.g., differential centrifugation or ultracentrifugation) and/or filtration to separate the PMP-containing fraction from large contaminants, including plant tissue debris, plant cells, or plant cell organelles (e.g., nuclei or chloroplasts). As such, the crude PMP fraction will have a decreased number of large contaminants, including plant tissue debris, plant cells, or plant cell organelles (e.g., nuclei, mitochondria or chloroplasts), as compared to the initial sample from the source plant or plant part.


In some examples of the above methods, the separating or processing step involves centrifugation. The centrifugation may be differential centrifugation, e.g., differential centrifugation using a sucrose gradient. The centrifugation may be ultracentrifugation. The centrifugation step may separate the PMP-containing fraction from plant cells or cellular debris in the preparation or fraction thereof. In such instances, the PMP fraction will have a decreased number of plant cells or cellular debris, as compared to the initial preparation or fraction thereof.


In some examples of the above methods, the separating or processing step involves one or more filtration steps. The filtration may be tangential flow filtration. In some examples, the tangential flow filtration involves exchanging the volume of the preparation at least 2 times, e.g., exchanging the volume of the preparation at least 3 times, at least 4 times, at least 5 times, at least 6 times, at least 7 times, at least 8 times, at least 9 times, or at least 10 times. In some examples, the tangential flow filtration involves exchanging the volume of the preparation at least 10 times.


In some examples of the above methods, the separating or processing step involves size exclusion chromatography (SEC). In some examples, the SEC is performed using an SEC column that separates molecules having a size between 10 and 1000 nm, e.g., between 35 and 350 nm. In some examples, the SEC column has a resin pore size of at least 10 nm, 15 nm, 20 nm, 25 nm, 30 nm, 35 nm, 40 nm, 45 nm, 50 nm, 55 nm, 60 nm, 65 nm, 70 nm, 75 nm, or 80 nm, e.g., has a resin pore size of between 20 nm and 50 nm.


In some examples of the above methods, the separating or processing step involves one, two, or all three of centrifugation (e.g., differential centrifugation), tangential flow filtration, and size exclusion chromatography, e.g., involves centrifugation; involves tangential flow filtration; involves size exclusion chromatography; involves centrifugation and tangential flow filtration; involves centrifugation and size exclusion chromatography; involves tangential flow filtration and size exclusion chromatography; or involves centrifugation, tangential flow filtration, and size exclusion chromatography.


The separating or processing step of the method may comprise one or more of a wash step, dilution, pH modification, dialysis, and removal of contaminants.


The plant preparation or fraction thereof or the PMP fraction may be further purified by additional purification methods to produce a plurality of pure PMPs. For example, the crude PMP fraction can be separated from other plant components by ultracentrifugation, e.g., using a density gradient (iodixanol or sucrose) and/or use of other approaches to remove aggregated components (e.g., precipitation or size-exclusion chromatography). The resulting pure PMPs may have a decreased level of contaminants or undesired components from the source plant (e.g., one or more non-PMP components, such as protein aggregates, nucleic acid aggregates, protein-nucleic acid aggregates, free lipoproteins, lipido-proteic structures), nuclei, cell wall components, cell organelles, or a combination thereof) relative to one or more fractions generated during the earlier separation steps, or relative to a pre-established threshold level, e.g., a commercial release specification. For example, the pure PMPs may have a decreased level (e.g., by about 5%, 10%, 15%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or more than 100%; or by about 2× fold, 4× fold, 5× fold, 10× fold, 20× fold, 25× fold, 50× fold, 75× fold, 100× fold, or more than 100× fold) of plant organelles or cell wall components relative to the level in the initial sample. In some instances, the pure PMPs are substantially free (e.g., have undetectable levels) of one or more non-PMP components, such as protein aggregates, nucleic acid aggregates, protein-nucleic acid aggregates, free lipoproteins, lipido-proteic structures), nuclei, cell wall components, cell organelles, or a combination thereof. Further examples of the releasing and separation steps can be found in Example 1. The PMPs may be at a concentration of, e.g., 1×109, 5×109, 1×1010, 5×1010, 5×1010, 1×1011, 2×1011, 3×1011, 4×1011, 5×1011, 6×1011, 7×1011, 8×1011, 9×1011, 1×1012, 2×1012, 3×1012, 4×1012, 5×1012, 6×1012, 7×1012, 8×1012, 9×1012, 1×1013, or more than 1×1013 PMPs/mL.


For example, protein aggregates may be removed from isolated PMPs. For example, the isolated PMP solution can be taken through a range of pHs (e.g., as measured using a pH probe) to precipitate out protein aggregates in solution. The pH can be adjusted to, e.g., pH 3, pH 5, pH 7, pH 9, or pH 11 with the addition of, e.g., sodium hydroxide or hydrochloric acid. Once the solution is at the specified pH, it can be filtered to remove particulates. Alternatively, the isolated PMP solution can be flocculated using the addition of charged polymers, such as Polymin-P or Praestol 2640. Briefly, Polymin-P or Praestol 2640 is added to the solution and mixed with an impeller. The solution can then be filtered to remove particulates. Alternatively, aggregates can be solubilized by increasing salt concentration. For example NaCl can be added to the isolated PMP solution until it is at, e.g., 1 mol/L. The solution can then be filtered to isolate the PMPs. Alternatively, aggregates are solubilized by increasing the temperature. For example, the isolated PMPs can be heated under mixing until the solution has reached a uniform temperature of, e.g., 50° C. for 5 minutes. The PMP mixture can then be filtered to isolate the PMPs. Alternatively, soluble contaminants from PMP solutions can be separated by size-exclusion chromatography column according to standard procedures, where PMPs elute in the first fractions, whereas proteins and ribonucleoproteins and some lipoproteins are eluted later. The efficiency of protein aggregate removal can be determined by measuring and comparing the protein concentration before and after removal of protein aggregates via BCA/Bradford protein quantification.


Any of the production methods described herein can be supplemented with any quantitative or qualitative methods known in the art to characterize or identify the PMPs at any step of the production process. PMPs may be characterized by a variety of analysis methods to estimate PMP yield, PMP concentration, PMP purity, PMP composition, or PMP sizes. PMPs can be evaluated by a number of methods known in the art that enable visualization, quantitation, or qualitative characterization (e.g., identification of the composition) of the PMPs, such as microscopy (e.g., transmission electron microscopy), dynamic light scattering, nanoparticle tracking, spectroscopy (e.g., Fourier transform infrared analysis), or mass spectrometry (protein and lipid analysis). In certain instances, methods (e.g., mass spectroscopy) may be used to identify plant EV markers present on the PMP, such as markers disclosed in the Appendix. To aid in analysis and characterization, of the PMP fraction, the PMPs can additionally be labelled or stained. For example, the PMPs can be stained with 3,3′-dihexyloxacarbocyanine iodide (DIOC6), a fluorescent lipophilic dye, PKH67 (Sigma Aldrich); Alexa Fluor® 488 (Thermo Fisher Scientific), or DyLight™ 800 (Thermo Fisher). In the absence of sophisticated forms of nanoparticle tracking, this relatively simple approach quantifies the total membrane content and can be used to indirectly measure the concentration of PMPs (Rutter and Innes, Plant Physiol. 173(1): 728-741, 2017; Rutter et al, Bio. Protoc. 7(17): e2533, 2017). For more precise measurements, and to assess the size distributions of PMPs, nanoparticle tracking can be used.


In some examples of any of the above methods, the PMPs of step (c) may be concentrated at least 2× relative to the preparation of step (a) or relative to a control sample, e.g., are concentrated at least 3×, 4×, 5×, 6×, 7×, 8×, 9×, 10×, 11×, 12×, 13×, 14×, 15×, 16×, 17×, 18×, 19×, 20×, 25×, 50×, 75×, or more than 100×. In some examples of any of the above methods, the PMPs of step (c) are concentrated at least 10× relative to the preparation of step (a). The PMPs in the composition may be at a concentration of at least 1, 10, 50, 100, 250, 500, or 750 μg PMP protein/ml, 1, 1.5, 2, 2.5, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, 9, 9.5, or 10 mg PMP protein/ml.


The isolated PMPs may make up about 0.1% to about 100% of the composition, such as any one of about 0.01% to about 100%, about 1% to about 99.9%, about 0.1% to about 10%, about 1% to about 25%, about 10% to about 50%, about 50% to about 99%, or about 75% to about 100%. In some instances, the composition includes at least any of 0.1%, 0.5%, 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or more PMPs, e.g., as measured by wt/vol, percent PMP protein composition, and/or percent lipid composition (e.g., by measuring fluorescently labelled lipids); See, e.g., Example 3). In some instances, the concentrated agents are used as commercial products, e.g., the final user may use diluted agents, which have a substantially lower concentration of active ingredient. In some embodiments, the composition is formulated as a pest control concentrate formulation, e.g., an ultra-low-volume concentrate formulation.


Providing the pectin-rich plant preparation may comprise processing a plant or a plant part (e.g., a pectin-rich plant or pectin-rich plant part) to release EVs, thereby producing PMPs. For example, the processing may include or consist of blending a plant part, mashing a plant or plant part through a strainer, or cold pressing a plant or plant part.


PMPs can be produced from a plant, or part thereof, by a variety of methods. Any method that allows release of the EV-containing apoplastic fraction of a plant, or an otherwise extracellular fraction that contains PMPs comprising secreted EVs (e.g., cell culture media) is suitable in the present methods. EVs can be separated from the plant or plant part by either destructive (e.g., grinding or blending of a plant, or any plant part) or non-destructive (washing or vacuum infiltration of a plant or any plant part) methods. For instance, the plant, or part thereof, can be vacuum-infiltrated, ground, blended, or a combination thereof to isolate EVs from the plant or plant part, thereby producing PMPs. For instance, the isolating step may involve (b) isolating a crude PMP fraction from the initial sample (e.g., a plant, a plant part, or a sample derived from a plant or plant part), wherein the isolating step involves vacuum infiltrating the plant (e.g., with a vesicle isolation buffer) to release and collect the apoplastic fraction. Alternatively, the isolating step may involve grinding or blending the plant to release the EVs, thereby producing PMPs.


As illustrated by Example 1, PMPs can be produced from a variety of plants, or parts thereof (e.g., the leaf apoplast, seed apoplast, root, fruit, vegetable, pollen, phloem, or xylem sap). For example, PMPs can be isolated from the apoplastic fraction of a plant, such as the apoplast of a leaf (e.g., apoplast Arabidopsis thaliana leaves) or the apoplast of seeds (e.g., apoplast of sunflower seeds). Other exemplary PMPs are produced from roots (e.g., ginger roots), fruit juice (e.g., grapefruit juice), vegetables (e.g., broccoli), pollen (e.g., olive pollen), phloem sap (e.g., Arabidopsis phloem sap), xylem sap (e.g., tomato plant xylem sap), or cell culture supernatant (e.g. BY2 tobacco cell culture supernatant). In other examples, PMPs are produced from a citrus plant, e.g., a grapefruit or a lemon, or a juice sac of a citrus plant, e.g., a juice sac of a grapefruit or a lemon. In other examples, PMPs are produced from a flowering plant such as a pomegranate, a blueberry, duckweed (e.g., Wolffia globosa), broccoli, avocado, grape, tomato fruit, or onion.


PMPs may be isolated from any genera of plants (vascular or nonvascular), including but not limited to angiosperms (monocotyledonous and dicotyledonous plants), gymnosperms, ferns, selaginellas, horsetails, psilophytes, lycophytes, algae (e.g., unicellular or multicellular, e.g., archaeplastida), or bryophytes. In certain instances, PMPs can be produced from a vascular plant, for example monocotyledons or dicotyledons or gymnosperms. For example, PMPs can be produced from alfalfa, apple, Arabidopsis, banana, barley, canola, castor bean, chicory, chrysanthemum, clover, cocoa, coffee, cotton, cottonseed, corn, crambe, cranberry, cucumber, dendrobium, dioscorea, eucalyptus, fescue, flax, gladiolus, liliacea, linseed, millet, muskmelon, mustard, oat, oil palm, oilseed rape, papaya, peanut, pineapple, ornamental plants, Phaseolus, potato, rapeseed, rice, rye, ryegrass, safflower, sesame, sorghum, soybean, sugarbeet, sugarcane, sunflower, strawberry, tobacco, tomato, turfgrass, wheat or vegetable crops such as lettuce, celery, broccoli, cauliflower, cucurbits; fruit and nut trees, such as apple, pear, peach, orange, grapefruit, lemon, lime, almond, pecan, walnut, hazel; vines, such as grapes, kiwi, hops; fruit shrubs and brambles, such as raspberry, blackberry, gooseberry; forest trees, such as ash, pine, fir, maple, oak, chestnut, popular; with alfalfa, canola, castor bean, corn, cotton, crambe, flax, linseed, mustard, oil palm, oilseed rape, peanut, potato, rice, safflower, sesame, soybean, sugarbeet, sunflower, tobacco, tomato, or wheat.


PMPs may be produced from a whole plant (e.g., a whole rosettes or seedlings) or alternatively from one or more plant parts (e.g., a pectin-rich plant part, e.g., a leaf, seed, root, fruit, vegetable, pollen, phloem sap, or xylem sap). For example, PMPs can be produced from shoot vegetative organs/structures (e.g., leaves, stems, or tubers), roots, flowers and floral organs/structures (e.g., pollen, bracts, sepals, petals, stamens, carpels, anthers, or ovules), seed (including embryo, endosperm, or seed coat), fruit (the mature ovary), sap (e.g., phloem or xylem sap), plant tissue (e.g., vascular tissue, ground tissue, tumor tissue, or the like), and cells (e.g., single cells, protoplasts, embryos, callus tissue, guard cells, egg cells, or the like), or progeny of same. For instance, the isolation step may involve (a) providing a plant, or a part thereof. In some examples, the plant part is an Arabidopsis leaf. The plant may be at any stage of development. For example, the PMP can be produced from seedlings, e.g., 1 week, 2 week, 3 week, 4 week, 5 week, 6 week, 7 week, or 8 week old seedlings (e.g., Arabidopsis seedlings). Other exemplary PMPs can include PMPs produced from roots (e.g., ginger roots), fruit juice (e.g., grapefruit juice), vegetables (e.g., broccoli), pollen (e.g., olive pollen), phloem sap (e.g., Arabidopsis phloem sap), or xylem sap (e.g., tomato plant xylem sap).


PMPs may be produced from plant cultures, e.g., a plant cell culture or tissue culture or a culture comprising entire plants or plant parts (e.g., a hydroponic culture). As used herein, the term “plant culture” refers to a plurality of plant cells, plant parts, plants (e.g., entire plants), or plant tissue that is propagated in or on a liquid, gel, semi-solid, or solid medium. Plant cultures include, but are not limited to, cultures of naturally occurring plants, plant parts, plant cells, or plant tissue or genetically modified plants, plant parts, plant cells, or plant tissues.


As illustrated by Example 2, PMPs can be purified by a variety of methods, for example, by using a density gradient (iodixanol or sucrose) in conjunction with ultracentrifugation and/or methods to remove aggregated contaminants, e.g., precipitation or size-exclusion chromatography. For instance, Example 2 illustrates purification of PMPs that have been obtained via the separation steps outlined in Example 1. Further, PMPs can be characterized in accordance with the methods illustrated in Example 3.


In some instances, the PMPs of the present compositions and methods can be isolated from a plant, or part thereof, and used without further modification to the PMP. In other instances, the PMP can be modified prior to use, as outlined further herein.


B. Plant EV Markers


The PMPs of the present compositions and methods may have a range of markers that identify the PMP as being produced from a plant EV, and/or including a segment, portion, or extract thereof. As used herein, the term “plant EV marker” refers to a component that is naturally associated with a plant and incorporated into or onto the plant EV in planta, such as a plant protein, a plant nucleic acid, a plant small molecule, a plant lipid, or a combination thereof. Examples of plant EV markers can be found, for example, in Rutter and Innes, Plant Physiol. 173(1): 728-741, 2017; Raimondo et al., Oncotarget. 6(23): 19514, 2015; Ju et al., Mol. Therapy. 21(7):1345-1357, 2013; Wang et al., Molecular Therapy. 22(3): 522-534, 2014; and Regente et al, J of Exp. Biol. 68(20): 5485-5496, 2017; each of which is incorporated herein by reference. Additional examples of plant EV markers are listed in the Appendix, and are further outlined herein.


The plant EV marker can include a plant lipid. Examples of plant lipid markers that may be found in the PMP include phytosterol, campesterol, β-sitosterol, stigmasterol, avenasterol, glycosyl inositol phosphoryl ceramides (GIPCs), glycolipids (e.g., monogalactosyldiacylglycerol (MGDG) or digalactosyldiacylglycerol (DGDG)), or a combination thereof. For instance, the PMP may include GIPCs, which represent the main sphingolipid class in plants and are one of the most abundant membrane lipids in plants. Other plant EV markers may include lipids that accumulate in plants in response to abiotic or biotic stressors (e.g., bacterial or fungal infection), such as phosphatidic acid (PA) or phosphatidylinositol-4-phosphate (PI4P).


Alternatively, the plant EV marker may include a plant protein. In some instances, the protein plant EV marker may be an antimicrobial protein naturally produced by plants, including defense proteins that plants secrete in response to abiotic or biotic stressors (e.g., bacterial or fungal infection). Plant pathogen defense proteins include soluble N-ethylmalemide-sensitive factor association protein receptor protein (SNARE) proteins (e.g., Syntaxin-121 (SYP121; GenBank Accession No.: NP_187788.1 or NP_974288.1), Penetration1 (PEN1; GenBank Accession No: NP_567462.1)) or ABC transporter Penetration3 (PEN3; GenBank Accession No: NP_191283.2). Other examples of plant EV markers includes proteins that facilitate the long-distance transport of RNA in plants, including phloem proteins (e.g., Phloem protein2-A1 (PP2-A1), GenBank Accession No: NP_193719.1), calcium-dependent lipid-binding proteins, or lectins (e.g., Jacalin-related lectins, e.g., Helianthus annuus jacalin (Helja; GenBank: AHZ86978.1). For example, the RNA binding protein may be Glycine-Rich RNA Binding Protein-7 (GRP7; GenBank Accession Number: NP_179760.1). Additionally, proteins that regulate plasmodesmata function can in some instances be found in plant EVs, including proteins such as Synap-Totgamin A A (GenBank Accession No: NP_565495.1). In some instances, the plant EV marker can include a protein involved in lipid metabolism, such as phospholipase C or phospholipase D. In some instances, the plant protein EV marker is a cellular trafficking protein in plants. In certain instances where the plant EV marker is a protein, the protein marker may lack a signal peptide that is typically associated with secreted proteins. Unconventional secretory proteins seem to share several common features like (i) lack of a leader sequence, (ii) absence of PTMs specific for ER or Golgi apparatus, and/or (iii) secretion not affected by brefeldin A which blocks the classical ER/Golgi-dependent secretion pathway. One skilled in the art can use a variety of tools freely accessible to the public (e.g., SecretomeP Database; SUBA3 (SUBcellular localization database for Arabidopsis proteins)) to evaluate a protein for a signal sequence, or lack thereof.


In instances where the plant EV marker is a protein, the protein may have an amino acid sequence having at least 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, or 100% sequence identity to a plant EV marker, such as any of the plant EV markers listed in the Appendix. For example, the protein may have an amino acid sequence having at least 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, or 100% sequence identity to PEN1 from Arabidopsis thaliana (GenBank Accession Number: NP_567462.1).


In some instances, the plant EV marker includes a nucleic acid encoded in plants, e.g., a plant RNA, a plant DNA, or a plant PNA. For example, the PMP may include dsRNA, mRNA, a viral RNA, a microRNA (miRNA), or a small interfering RNA (siRNA) encoded by a plant. In some instances, the nucleic acid may be one that is associated with a protein that facilitates the long-distance transport of RNA in plants, as discussed herein. In some instances, the nucleic acid plant EV marker may be one involved in host-induced gene silencing (HIGS), which is the process by which plants silence foreign transcripts of plant pests (e.g., pathogens such as fungi). For example, the nucleic acid may be one that silences bacterial or fungal genes. In some instances, the nucleic acid may be a microRNA, such as miR159 or miR166, which target genes in a fungal pathogen (e.g., Verticillium dahliae). In some instances, the protein may be one involved in carrying plant defense compounds, such as proteins involved in glucosinolate (GSL) transport and metabolism, including Glucosinolate Transporter-1-1 (GTR1; GenBankAccesion No: NP_566896.2), Glucosinolate Transporter-2 (GTR2; NP_201074.1), or Epithiospecific Modifier 1 (ESM1; NP_188037.1).


In instances where the plant EV marker is a nucleic acid, the nucleic acid may have a nucleotide sequence having at least 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, or 100% sequence identity to a plant EV marker, e.g., such as those encoding the plant EV markers listed in the Appendix. For example, the nucleic acid may have a polynucleotide sequence having at least 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, or 100% sequence identity to miR159 or miR166.


In some instances, the plant EV marker includes a compound produced by plants. For example, the compound may be a defense compound produced in response to abiotic or biotic stressors, such as secondary metabolites. One such secondary metabolite that be found in PMPs are glucosinolates (GSLs), which are nitrogen and sulfur-containing secondary metabolites found mainly in Brassicaceae plants. Other secondary metabolites may include allelochemicals.


In some instances, the PMP may also be identified as being produced from a plant EV based on the lack of certain markers (e.g., lipids, polypeptides, or polynucleotides) that are not typically produced by plants, but are generally associated with other organisms (e.g., markers of animal EVs, bacterial EVs, or fungal EVs). For example, in some instances, the PMP lacks lipids typically found in animal EVs, bacterial EVs, or fungal EVs. In some instances, the PMP lacks lipids typical of animal EVs (e.g., sphingomyelin). In some instances, the PMP does not contain lipids typical of bacterial EVs or bacterial membranes (e.g., LPS). In some instances, the PMP lacks lipids typical of fungal membranes (e.g., ergosterol).


Plant EV markers can be identified using any approaches known in the art that enable identification of small molecules (e.g., mass spectroscopy, mass spectrometry), lipds (e.g., mass spectroscopy, mass spectrometry), proteins (e.g., mass spectroscopy, immunoblotting), or nucleic acids (e.g., PCR analysis). In some instances, a PMP composition described herein includes a detectable amount, e.g., a pre-determined threshold amount, of a plant EV marker described herein.


C. Loading of Agents


PMPs manufactured in accordance with the methods herein can be modified to include a heterologous functional agent (e.g., a heterologous agricultural agent (e.g., pesticidal agent, fertilizing agent, herbicidal agent, plant-modifying agent) or a heterologous therapeutic agent (e.g., a pathogen control agent such as an antifungal agent, an antibacterial agent, a virucidal agent, an anti-viral agent, an insecticidal agent, a nematicidal agent, an antiparasitic agent, or an insect repellent)), such as those described herein. The PMPs can carry or associate with such agents in a variety of ways to enable delivery of the agent to a target organism, e.g., by encapsulating the agent, incorporation of the component in the lipid bilayer structure, or association of the component (e.g., by conjugation) with the surface of the lipid bilayer structure of the PMP.


The heterologous functional agent can be incorporated or loaded into or onto the PMP by any methods known in the art that allow association, directly or indirectly, between the PMP and agent. Heterologous functional agent agents can be incorporated into the PMP by an in vivo method (e.g., in planta, e.g., through production of PMPs from a transgenic plant that comprises the heterologous agent), or in vitro (e.g., in tissue culture, or in cell culture), or both in vivo and in vitro methods.


In instances where the PMPs are loaded with a heterologous functional agent in vivo, the PMP may be produced from an EV, or segment, portion, or extract thereof, that has been loaded in planta, in tissue culture, or in cell culture. In planta methods include expression of the heterologous functional agent in a plant that has been genetically modified to express the heterologous functional agent. In some instances, the heterologous functional agent is exogenous to the plant. Alternatively, the heterologous functional agent may be naturally found in the plant, but expressed at an elevated level relative to that found in a non-genetically modified plant.


In some instances, the PMP can be loaded in vitro. The heterologous functional agent may be loaded onto or into (e.g., may be encapsulated by) the PMPs using, but not limited to, physical, chemical, and/or biological methods. For example, the heterologous functional agent may be introduced into PMP by one or more of electroporation, sonication, passive diffusion, stirring, lipid extraction, or extrusion. Loaded PMPs can be assessed to confirm the presence or level of the loaded agent using a variety methods, such as HPLC (e.g., to assess small molecules); immunoblotting (e.g., to assess proteins); and quantitative PCR (e.g., to assess nucleotides). However, it should be appreciated by those skilled in the art that the loading of a substance of interest into PMPs is not limited to the above-illustrated methods.


In some instances, the heterologous functional agent can be conjugated to the PMP, e.g., connected or joined, indirectly or directly, to the PMP. For instance, one or more pesticidal agents can be chemically linked to a PMP, such that the one or more pesticidal agents are joined (e.g., by covalent or ionic bonds) directly to the lipid bilayer of the PMP. In some instances, the conjugation of the heterologous functional agent to the PMPs can be achieved by first mixing the one or more heterologous functional agents with an appropriate cross-linking agent (e.g., N-ethylcarbo-diimide (“EDC”), which is generally utilized as a carboxyl activating agent for amide bonding with primary amines and also reacts with phosphate groups) in a suitable solvent. After a period of incubation sufficient to allow the heterologous functional agent to attach to the cross-linking agent, the cross-linking agent/heterologous functional agent mixture can then be combined with the PMPs and, after another period of incubation, subjected to a sucrose gradient (e.g., and 8, 30, 45, and 60% sucrose gradient) to separate the free heterologous functional agent and free PMPs from the heterologous functional agent conjugated to the PMPs. As part of combining the mixture with a sucrose gradient, and an accompanying centrifugation step, the PMPs conjugated to the heterologous functional agent are then seen as a band in the sucrose gradient, such that the conjugated PMPs can then be collected, washed, and dissolved in a suitable solution for use as described herein.


In some instances, the PMP is stably associated with the heterologous functional agent prior to and following delivery of the PMP, e.g., to a plant or to a pest. In other instances, the PMP is associated with the heterologous functional agent such that the heterologous functional agent becomes dissociated from the PMP following delivery of the PMP, e.g., to a plant or to a pest.


The PMP can be further modified with other components (e.g., lipids, e.g., sterols, e.g., cholesterol; or small molecules) to further alter the functional and structural characteristics of the PMP. For example, the PMPs can be further modified with stabilizing molecules that increase the stability of the PMP (e.g., for at least one day at room temperature, and/or stable for at least one week at 4° C.).


The PMPs can be loaded with various concentrations of the heterologous functional agent, depending on the particular agent or use. For example, in some instances, the PMPs are loaded such that the composition disclosed herein includes about 0.001, 0.01, 0.1, 1.0, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, 50, 60, 70, 80, 90, or 95 (or any range between about 0.001 and 95) or more wt % of a heterologous functional agent. In some instances, the PMPs are loaded such that the composition includes about 95, 90, 80, 70, 60, 50, 40, 30, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, 1.0, 0.1, 0.01, 0.001 (or any range between about 95 and 0.001) or less wt % of a heterologous functional agent. For example, the pest control (e.g., biopesticide or biorepellent) composition can include about 0.001 to about 0.01 wt %, about 0.01 to about 0.1 wt %, about 0.1 to about 1 wt %, about 1 to about 5 wt %, or about 5 to about 10 wt %, about 10 to about 20 wt % of the heterologous functional agent. In some instances, the PMP can be loaded with about 1, 5, 10, 50, 100, 200, or 500, 1,000, 2,000 (or any range between about 1 and 2,000) or more μg/ml of a heterologous functional agent. A liposome of the invention can be loaded with about 2,000, 1,000, 500, 200, 100, 50, 10, 5, 1 (or any range between about 2,000 and 1) or less μg/ml of a heterologous functional agent.


In some instances, the PMPs are loaded such that the composition disclosed herein includes at least 0.001 wt %, at least 0.01 wt %, at least 0.1 wt %, at least 1.0 wt %, at least 2 wt %, at least 3 wt %, at least 4 wt %, at least 5 wt %, at least 6 wt %, at least 7 wt %, at least 8 wt %, at least 9 wt %, at least 10 wt %, at least 15 wt %, at least 20 wt %, at least 30 wt %, at least 40 wt %, at least 50 wt %, at least 60 wt %, at least 70 wt %, at least 80 wt %, at least 90 wt %, or at least 95 wt % of a heterologous functional agent. In some instances, the PMP can be loaded with at least 1 μg/ml, at least 5 μg/ml, at least 10 μg/ml, at least 50 μg/ml, at least 100 μg/ml, at least 200 μg/ml, at least 500 μg/ml, at least 1,000 μg/ml, at least 2,000 μg/ml of a heterologous functional agent.


Examples of particular agents that can be loaded into the PMP are further outlined in the section entitled “Heterologous Functional Agents.”


D. Pharmaceutical Formulations


Included herein are compositions that can be formulated into pharmaceutical compositions, e.g., for administration to an animal, such as a human or a non-human agricultural animal (e.g., a cow, steer, pig, chicken, or turkey). The pharmaceutical composition may be administered to an animal with a pharmaceutically acceptable diluent, carrier, and/or excipient. Depending on the mode of administration and the dosage, the pharmaceutical composition of the methods described herein will be formulated into suitable pharmaceutical compositions to permit facile delivery. The single dose may be in a unit dose form as needed.


A pharmaceutical composition may be formulated for e.g., oral administration, intravenous administration (e.g., injection or infusion), or subcutaneous administration to an animal. For injectable formulations, various effective pharmaceutical carriers are known in the art (See, e.g., Remington: The Science and Practice of Pharmacy, 22nd ed., (2012) and ASHP Handbook on Injectable Drugs, 18th ed., (2014)).


Pharmaceutically acceptable carriers and excipients in the present compositions are nontoxic to recipients at the dosages and concentrations employed. Acceptable carriers and excipients may include buffers such as phosphate, citrate, HEPES, and TAE, antioxidants such as ascorbic acid and methionine, preservatives such as hexamethonium chloride, octadecyldimethylbenzyl ammonium chloride, resorcinol, and benzalkonium chloride, proteins such as human serum albumin, gelatin, dextran, and immunoglobulins, hydrophilic polymers such as polyvinylpyrrolidone, amino acids such as glycine, glutamine, histidine, and lysine, and carbohydrates such as glucose, mannose, sucrose, and sorbitol. The compositions may be formulated according to conventional pharmaceutical practice. The concentration of the compound in the formulation will vary depending upon a number of factors, including the dosage of the active agent (e.g., PMP) to be administered, and the route of administration.


For oral administration to an animal, the pharmaceutical composition can be prepared in the form of an oral formulation. Formulations for oral use can include tablets, caplets, capsules, syrups, or oral liquid dosage forms containing the active ingredient(s) in a mixture with non-toxic pharmaceutically acceptable excipients. These excipients may be, for example, inert diluents or fillers (e.g., sucrose, sorbitol, sugar, mannitol, microcrystalline cellulose, starches including potato starch, calcium carbonate, sodium chloride, lactose, calcium phosphate, calcium sulfate, or sodium phosphate); granulating and disintegrating agents (e.g., cellulose derivatives including microcrystalline cellulose, starches including potato starch, croscarmellose sodium, alginates, or alginic acid); binding agents (e.g., sucrose, glucose, sorbitol, acacia, alginic acid, sodium alginate, gelatin, starch, pregelatinized starch, microcrystalline cellulose, magnesium aluminum silicate, carboxymethylcellulose sodium, methylcellulose, hydroxypropyl methylcellulose, ethylcellulose, polyvinylpyrrolidone, or polyethylene glycol); and lubricating agents, glidants, and antiadhesives (e.g., magnesium stearate, zinc stearate, stearic acid, silicas, hydrogenated vegetable oils, or talc). Other pharmaceutically acceptable excipients can be colorants, flavoring agents, plasticizers, humectants, buffering agents, and the like. Formulations for oral use may also be provided in unit dosage form as chewable tablets, non-chewable tablets, caplets, capsules (e.g., as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium). The compositions disclosed herein may also further include an immediate-release, extended release or delayed-release formulation.


For parenteral administration to an animal, the pharmaceutical compositions may be formulated in the form of liquid solutions or suspensions and administered by a parenteral route (e.g., subcutaneous, intravenous, or intramuscular). The pharmaceutical composition can be formulated for injection or infusion. Pharmaceutical compositions for parenteral administration can be formulated using a sterile solution or any pharmaceutically acceptable liquid as a vehicle. Pharmaceutically acceptable vehicles include, but are not limited to, sterile water, physiological saline, or cell culture media (e.g., Dulbecco's Modified Eagle Medium (DMEM), α-Modified Eagles Medium (α-MEM), F-12 medium). Formulation methods are known in the art, see e.g., Gibson (ed.) Pharmaceutical Preformulation and Formulation (2nd ed.) Taylor & Francis Group, CRC Press (2009).


E. Agricultural Formulations


To allow ease of application, handling, transportation, storage, and activity, the active agent, here PMPs, can be formulated with other substances. PMPs can be formulated into, for example, baits, concentrated emulsions, dusts, emulsifiable concentrates, fumigants, gels, granules, microencapsulations, seed treatments, suspension concentrates, suspoemulsions, tablets, water soluble liquids, water dispersible granules or dry flowables, wettable powders, and ultra-low volume solutions. For further information on formulation types see “Catalogue of Pesticide Formulation Types and International Coding System” Technical Monograph n° 2, 5th Edition by CropLife International (2002).


Active agents (e.g., PMPs, additional pesticides) can be applied as aqueous suspensions or emulsions prepared from concentrated formulations of such agents. Such water-soluble, water-suspendable, or emulsifiable formulations are either solids, usually known as wettable powders, or water dispersible granules, or liquids usually known as emulsifiable concentrates, or aqueous suspensions. Wettable powders, which may be compacted to form water dispersible granules, comprise an intimate mixture of the pesticide, a carrier, and surfactants. The carrier is usually selected from among the attapulgite clays, the montmorillonite clays, the diatomaceous earths, or the purified silicates. Effective surfactants, including from about 0.5% to about 10% of the wettable powder, are found among sulfonated lignins, condensed naphthalenesulfonates, naphthalenesulfonates, alkylbenzenesulfonates, alkyl sulfates, and non-ionic surfactants such as ethylene oxide adducts of alkyl phenols.


Emulsifiable concentrates can comprise a suitable concentration of PMPs, such as from about 50 to about 500 grams per liter of liquid dissolved in a carrier that is either a water miscible solvent or a mixture of water-immiscible organic solvent and emulsifiers. Useful organic solvents include aromatics, especially xylenes and petroleum fractions, especially the high-boiling naphthalenic and olefinic portions of petroleum such as heavy aromatic naphtha. Other organic solvents may also be used, such as the terpenic solvents including rosin derivatives, aliphatic ketones such as cyclohexanone, and complex alcohols such as 2-ethoxyethanol. Suitable emulsifiers for emulsifiable concentrates are selected from conventional anionic and non-ionic surfactants.


Aqueous suspensions comprise suspensions of water-insoluble pesticides dispersed in an aqueous carrier at a concentration in the range from about 5% to about 50% by weight. Suspensions are prepared by finely grinding the pesticide and vigorously mixing it into a carrier comprised of water and surfactants. Ingredients, such as inorganic salts and synthetic or natural gums may also be added, to increase the density and viscosity of the aqueous carrier.


PMPs may also be applied as granular compositions that are particularly useful for applications to the soil. Granular compositions usually contain from about 0.5% to about 10% by weight of the pesticide, dispersed in a carrier that comprises clay or a similar substance. Such compositions are usually prepared by dissolving the formulation in a suitable solvent and applying it to a granular carrier which has been pre-formed to the appropriate particle size, in the range of from about 0.5 to about 3 mm. Such compositions may also be formulated by making a dough or paste of the carrier and compound and crushing and drying to obtain the desired granular particle size.


Dusts containing the present PMP formulation are prepared by intimately mixing PMPs in powdered form with a suitable dusty agricultural carrier, such as kaolin clay, ground volcanic rock, and the like. Dusts can suitably contain from about 1% to about 10% of the packets. They can be applied as a seed dressing or as a foliage application with a dust blower machine.


It is equally practical to apply the present formulation in the form of a solution in an appropriate organic solvent, usually petroleum oil, such as the spray oils, which are widely used in agricultural chemistry.


PMPs can also be applied in the form of an aerosol composition. In such compositions the packets are dissolved or dispersed in a carrier, which is a pressure-generating propellant mixture. The aerosol composition is packaged in a container from which the mixture is dispensed through an atomizing valve.


Another embodiment is an oil-in-water emulsion, wherein the emulsion comprises oily globules which are each provided with a lamellar liquid crystal coating and are dispersed in an aqueous phase, wherein each oily globule comprises at least one compound which is agriculturally active, and is individually coated with a monolamellar or oligolamellar layer including: (1) at least one non-ionic lipophilic surface-active agent, (2) at least one non-ionic hydrophilic surface-active agent and (3) at least one ionic surface-active agent, wherein the globules having a mean particle diameter of less than 800 nanometers. Further information on the embodiment is disclosed in U.S. patent publication 20070027034 published Feb. 1, 2007. For ease of use, this embodiment will be referred to as “OIWE.”


Additionally, generally, when the molecules disclosed above are used in a formulation, such formulation can also contain other components. These components include, but are not limited to, (this is a non-exhaustive and non-mutually exclusive list) wetters, spreaders, stickers, penetrants, buffers, sequestering agents, drift reduction agents, compatibility agents, anti-foam agents, cleaning agents, and emulsifiers. A few components are described forthwith.


A wetting agent is a substance that when added to a liquid increases the spreading or penetration power of the liquid by reducing the interfacial tension between the liquid and the surface on which it is spreading. Wetting agents are used for two main functions in agrochemical formulations: during processing and manufacture to increase the rate of wetting of powders in water to make concentrates for soluble liquids or suspension concentrates; and during mixing of a product with water in a spray tank to reduce the wetting time of wettable powders and to improve the penetration of water into water-dispersible granules. Examples of wetting agents used in wettable powder, suspension concentrate, and water-dispersible granule formulations are: sodium lauryl sulfate; sodium dioctyl sulfosuccinate; alkyl phenol ethoxylates; and aliphatic alcohol ethoxylates.


A dispersing agent is a substance which adsorbs onto the surface of particles and helps to preserve the state of dispersion of the particles and prevents them from reaggregating. Dispersing agents are added to agrochemical formulations to facilitate dispersion and suspension during manufacture, and to ensure the particles redisperse into water in a spray tank. They are widely used in wettable powders, suspension concentrates and water-dispersible granules. Surfactants that are used as dispersing agents have the ability to adsorb strongly onto a particle surface and provide a charged or steric barrier to reaggregation of particles. The most commonly used surfactants are anionic, non-ionic, or mixtures of the two types. For wettable powder formulations, the most common dispersing agents are sodium lignosulfonates. For suspension concentrates, very good adsorption and stabilization are obtained using polyelectrolytes, such as sodium naphthalene sulfonate formaldehyde condensates. Tristyrylphenol ethoxylate phosphate esters are also used. Non-ionics such as alkylarylethylene oxide condensates and EO-PO block copolymers are sometimes combined with anionics as dispersing agents for suspension concentrates. In recent years, new types of very high molecular weight polymeric surfactants have been developed as dispersing agents. These have very long hydrophobic ‘backbones’ and a large number of ethylene oxide chains forming the ‘teeth’ of a ‘comb’ surfactant. These high molecular weight polymers can give very good long-term stability to suspension concentrates because the hydrophobic backbones have many anchoring points onto the particle surfaces. Examples of dispersing agents used in agrochemical formulations are: sodium lignosulfonates; sodium naphthalene sulfonate formaldehyde condensates; tristyrylphenol ethoxylate phosphate esters; aliphatic alcohol ethoxylates; alkyl ethoxylates; EO-PO (ethylene oxide-propylene oxide) block copolymers; and graft copolymers.


An emulsifying agent is a substance which stabilizes a suspension of droplets of one liquid phase in another liquid phase. Without the emulsifying agent the two liquids would separate into two immiscible liquid phases. The most commonly used emulsifier blends contain alkylphenol or aliphatic alcohol with twelve or more ethylene oxide units and the oil-soluble calcium salt of dodecylbenzenesulfonic acid. A range of hydrophile-lipophile balance (“HLB”) values from 8 to 18 will normally provide good stable emulsions. Emulsion stability can sometimes be improved by the addition of a small amount of an EO-PO block copolymer surfactant.


A solubilizing agent is a surfactant which will form micelles in water at concentrations above the critical micelle concentration. The micelles are then able to dissolve or solubilize water-insoluble materials inside the hydrophobic part of the micelle. The types of surfactants usually used for solubilization are non-ionics, sorbitan monooleates, sorbitan monooleate ethoxylates, and methyl oleate esters.


Surfactants are sometimes used, either alone or with other additives such as mineral or vegetable oils as adjuvants to spray-tank mixes to improve the biological performance of the pesticide on the target. The types of surfactants used for bioenhancement depend generally on the nature and mode of action of the pesticide. However, they are often non-ionics such as: alkyl ethoxylates; linear aliphatic alcohol ethoxylates; aliphatic amine ethoxylates.


A carrier or diluent in an agricultural formulation is a material added to the pesticide to give a product of the required strength. Carriers are usually materials with high absorptive capacities, while diluents are usually materials with low absorptive capacities. Carriers and diluents are used in the formulation of dusts, wettable powders, granules, and water-dispersible granules.


Organic solvents are used mainly in the formulation of emulsifiable concentrates, oil-in-water emulsions, suspoemulsions, and ultra low volume formulations, and to a lesser extent, granular formulations. Sometimes mixtures of solvents are used. The first main groups of solvents are aliphatic paraffinic oils such as kerosene or refined paraffins. The second main group (and the most common) comprises the aromatic solvents such as xylene and higher molecular weight fractions of C9 and C10 aromatic solvents. Chlorinated hydrocarbons are useful as cosolvents to prevent crystallization of pesticides when the formulation is emulsified into water. Alcohols are sometimes used as cosolvents to increase solvent power. Other solvents may include vegetable oils, seed oils, and esters of vegetable and seed oils.


Thickeners or gelling agents are used mainly in the formulation of suspension concentrates, emulsions, and suspoemulsions to modify the rheology or flow properties of the liquid and to prevent separation and settling of the dispersed particles or droplets. Thickening, gelling, and anti-settling agents generally fall into two categories, namely water-insoluble particulates and water-soluble polymers. It is possible to produce suspension concentrate formulations using clays and silicas. Examples of these types of materials, include, but are not limited to, montmorillonite, bentonite, magnesium aluminum silicate, and attapulgite. Water-soluble polysaccharides have been used as thickening-gelling agents for many years. The types of polysaccharides most commonly used are natural extracts of seeds and seaweeds or are synthetic derivatives of cellulose. Examples of these types of materials include, but are not limited to, guar gum; locust bean gum; carrageenam; alginates; methyl cellulose; sodium carboxymethyl cellulose (SCMC); hydroxyethyl cellulose (HEC). Other types of anti-settling agents are based on modified starches, polyacrylates, polyvinyl alcohol, and polyethylene oxide. Another good anti-settling agent is xanthan gum.


Microorganisms can cause spoilage of formulated products. Therefore preservation agents are used to eliminate or reduce their effect. Examples of such agents include, but are not limited to: propionic acid and its sodium salt; sorbic acid and its sodium or potassium salts; benzoic acid and its sodium salt; p-hydroxybenzoic acid sodium salt; methyl p-hydroxybenzoate; and 1,2-benzisothiazolin-3-one (BIT).


The presence of surfactants often causes water-based formulations to foam during mixing operations in production and in application through a spray tank. In order to reduce the tendency to foam, anti-foam agents are often added either during the production stage or before filling into bottles. Generally, there are two types of anti-foam agents, namely silicones and non-silicones. Silicones are usually aqueous emulsions of dimethyl polysiloxane, while the non-silicone anti-foam agents are water-insoluble oils, such as octanol and nonanol, or silica. In both cases, the function of the anti-foam agent is to displace the surfactant from the air-water interface.


“Green” agents (e.g., adjuvants, surfactants, solvents) can reduce the overall environmental footprint of crop protection formulations. Green agents are biodegradable and generally derived from natural and/or sustainable sources, e.g., plant and animal sources. Specific examples are: vegetable oils, seed oils, and esters thereof, also alkoxylated alkyl polyglucosides.


In some instances, PMPs can be freeze-dried or lyophilized. See U.S. Pat. No. 4,311,712. The PMPs can later be reconstituted on contact with water or another liquid. Other components can be added to the lyophilized or reconstituted liposomes, for example, other pesticidal agents, agriculturally acceptable carriers, or other materials in accordance with the formulations described herein.


Other optional features of the composition include carriers or delivery vehicles that protect the pest control (e.g., biopesticide or biorepellent) composition against UV and/or acidic conditions. In some instances, the delivery vehicle contains a pH buffer. In some instances, the composition is formulated to have a pH in the range of about 4.5 to about 9.0, including for example pH ranges of about any one of 5.0 to about 8.0, about 6.5 to about 7.5, or about 6.5 to about 7.0.


The composition may additionally be formulated with an attractant (e.g., a chemoattractant) that attracts a pest to the vicinity of the composition. Attractants include pheromones, a chemical that is secreted by an animal, especially a pest, or chemoattractants which influences the behavior or development of others of the same species. Other attractants include sugar and protein hydrolysate syrups, yeasts, and rotting meat. Attractants also can be combined with an active ingredient and sprayed onto foliage or other items in the treatment area. Various attractants are known which influence a pest's behavior as a pest's search for food, oviposition, or mating sites, or mates. Attractants useful in the methods and compositions described herein include, for example, eugenol, phenethyl propionate, ethyl dimethylisobutyl-cyclopropane carboxylate, propyl benszodioxancarboxylate, cis-7,8-epoxy-2-methyloctadecane, trans-8,trans-0-dodecadienol, cis-9-tetradecenal (with cis-11-hexadecenal), trans-11-tetradecenal, cis-11-hexadecenal, (Z)-11,12-hexadecadienal, cis-7-dodecenyl acetate, cis-8-dodecenyul acetate, cis-9-dodecenyl acetate, cis-9-tetradecenyl acetate, cis-11-tetradecenyl acetate, trans-11-tetradecenyl acetate (with cis-11), cis-9,trans-11-tetradecadienyl acetate (with cis-9,trans-12), cis-9,trans-12-tetradecadienyl acetate, cis-7,cis-11-hexadecadienyl acetate (with cis-7,trans-11), cis-3,cis-13-octadecadienyl acetate, trans-3,cis-13-octadecadienyl acetate, anethole and isoamyl salicylate.


For further information on agricultural formulations, see “Chemistry and Technology of Agrochemical Formulations” edited by D. A. Knowles, copyright 1998 by Kluwer Academic Publishers. Also see “Insecticides in Agriculture and Environment-Retrospects and Prospects” by A. S. Perry, I. Yamamoto, I. Ishaaya, and R. Perry, copyright 1998 by Springer-Verlag.


II. Heterologous Functional Agents

The PMPs manufactured herein can further include a heterologous functional agent (e.g., a heterologous agricultural agent (e.g., pesticidal agent, fertilizing agent, herbicidal agent, plant-modifying agent) or a heterologous therapeutic agent (e.g., a pathogen control agent, such as an antifungal agent, an antibacterial agent, a virucidal agent, an anti-viral agent, an insecticidal agent, a nematicidal agent, an antiparasitic agent, or an insect repellent)). For example, the PMP may encapsulate the heterologous functional agent. Alternatively, the heterologous functional agent can be embedded on or conjugated to the surface of the PMP. In some instances, the PMPs include two or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, or more than 10) different heterologous functional agents. Heterologous functional agents may be added at any step during the manufacturing process effective to introduce the agent into the manufactured PMPs.


In certain instances, the heterologous functional agent (e.g., a heterologous agricultural agent (e.g., pesticidal agent, fertilizing agent, herbicidal agent, plant-modifying agent, a heterologous nucleic acid, a heterologous polypeptide, or a heterologous small molecule) or a heterologous therapeutic agent (e.g., a pathogen control agent such as an antifungal agent, an antibacterial agent, a virucidal agent, an anti-viral agent, a nematicidal agent, an antiparasitic agent, or an insect repellent)) can be modified. For example, the modification can be a chemical modification, e.g., conjugation to a marker, e.g., fluorescent marker or a radioactive marker. In other examples, the modification can include conjugation or operational linkage to a moiety that enhances the stability, delivery, targeting, bioavailability, or half-life of the agent, e.g., a lipid, a glycan, a polymer (e.g., PEG), a cation moiety.


Examples of heterologous functional agents that can be loaded into the PMPs manufactured herein are outlined below.


A. Heterologous Agricultural Agents


The PMPs manufactured herein can include a heterologous agricultural agent (e.g., an agent that effects a plant or an organism that associates with a plant and can be loaded into a PMP), such as a pesticidal agent, herbicidal agent, fertilizing agent, or a plant-modifying agent.


For example, in some instances, the PMPs may include a pesticidal agent. The pesticidal agent can be an antifungal agent, an antibacterial agent, an insecticidal agent, a molluscicidal agent, a nematicidal agent, a virucidal agent, or a combination thereof. The pesticidal agent can be a chemical agent, such as those well known in the art. Alternatively or additionally, the pesticidal agent can be a peptide, a polypeptide, a nucleic acid, a polynucleotide, or a small molecule. The pesticidal agent may be an agent that can decrease the fitness of a variety of plant pests or can be one that targets one or more specific target plant pests (e.g., a specific species or genus of plant pests).


In some instances, the PMPs may include one or more heterologous fertilizing agents. Examples of heterologous fertilizing agents include plant nutrients or plant growth regulators, such as those well known in the art. Alternatively, or additionally, the fertilizing agent can be a peptide, a polypeptide, a nucleic acid, or a polynucleotide that can increase the fitness of a plant symbiont. The fertilizing agent may be an agent that can increase the fitness of a variety of plants or plant symbionts or can be one that targets one or more specific target plants or plant symbionts (e.g., a specific species or genera of plants or plant symbionts).


In other instances, the PMPs may include one or more heterologous plant-modifying agents. In some instances, the plant-modifying agent can include a peptide or a nucleic acid.


i. Antibacterial Agents


The PMP compositions described herein can further include an antibacterial agent. In some instances, the PMP compositions include two or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, or more than 10) different antibacterial agents. For example, the antibacterial agent can decrease the fitness of (e.g., decrease growth or kill) a bacterial plant pest (e.g., a bacterial plant pathogen). A PMP composition including an antibiotic as described herein can be contacted with a target pest, or plant infested thereof, in an amount and for a time sufficient to: (a) reach a target level (e.g., a predetermined or threshold level) of antibiotic concentration inside or on the target pest; and (b) decrease fitness of the target pest. The antibacterials described herein may be formulated in a PMP composition for any of the methods described herein, and in certain instances, may be associated with the PMP thereof.


As used herein, the term “antibacterial agent” refers to a material that kills or inhibits the growth, proliferation, division, reproduction, or spread of bacteria, such as phytopathogenic bacteria, and includes bactericidal (e.g., disinfectant compounds, antiseptic compounds, or antibiotics) or bacteriostatic agents (e.g., compounds or antibiotics). Bactericidal antibiotics kill bacteria, while bacteriostatic antibiotics only slow their growth or reproduction.


Bactericides can include disinfectants, antiseptics, or antibiotics. The most used disinfectants can comprise: active chlorine (i.e., hypochlorites (e.g., sodium hypochlorite), chloramines, dichloroisocyanurate and trichloroisocyanurate, wet chlorine, chlorine dioxide etc.), active oxygen (peroxides, such as peracetic acid, potassium persulfate, sodium perborate, sodium percarbonate and urea perhydrate), iodine (iodpovidone (povidone-iodine, Betadine), Lugol's solution, iodine tincture, iodinated nonionic surfactants), concentrated alcohols (mainly ethanol, 1-propanol, called also n-propanol and 2-propanol, called isopropanol and mixtures thereof; further, 2-phenoxyethanol and 1- and 2-phenoxypropanols are used), phenolic substances (such as phenol (also called carbolic acid), cresols (called Lysole in combination with liquid potassium soaps), halogenated (chlorinated, brominated) phenols, such as hexachlorophene, triclosan, trichlorophenol, tribromophenol, pentachlorophenol, Dibromol and salts thereof), cationic surfactants, such as some quaternary ammonium cations (such as benzalkonium chloride, cetyl trimethylammonium bromide or chloride, didecyldimethylammonium chloride, cetylpyridinium chloride, benzethonium chloride) and others, non-quaternary compounds, such as chlorhexidine, glucoprotamine, octenidine dihydrochloride etc.), strong oxidizers, such as ozone and permanganate solutions; heavy metals and their salts, such as colloidal silver, silver nitrate, mercury chloride, phenylmercury salts, copper sulfate, copper oxide-chloride, copper hydroxide, copper octanoate, copper oxychloride sulfate, copper sulfate, copper sulfate pentahydrate, etc. Heavy metals and their salts are the most toxic, and environment-hazardous bactericides and therefore, their use is strongly oppressed or canceled; further, also properly concentrated strong acids (phosphoric, nitric, sulfuric, amidosulfuric, toluenesulfonic acids) and alkalis (sodium, potassium, calcium hydroxides).


As antiseptics (i.e., germicide agents that can be used on human or animal body, skin, mucoses, wounds and the like), few of the above mentioned disinfectants can be used, under proper conditions (mainly concentration, pH, temperature and toxicity toward man/animal). Among them, important are: properly diluted chlorine preparations (i.e., Daquin's solution, 0.5% sodium or potassium hypochlorite solution, pH-adjusted to pH 7-8, or 0.5-1% solution of sodium benzenesulfochloramide (chloramine B)), some iodine preparations, such as iodopovidone in various galenics (ointment, solutions, wound plasters), in the past also Lugol's solution, peroxides as urea perhydrate solutions and pH-buffered 0.1-0.25% peracetic acid solutions, alcohols with or without antiseptic additives, used mainly for skin antisepsis, weak organic acids such as sorbic acid, benzoic acid, lactic acid and salicylic acid some phenolic compounds, such as hexachlorophene, triclosan and Dibromol, and cation-active compounds, such as 0.05-0.5% benzalkonium, 0.5-4% chlorhexidine, 0.1-2% octenidine solutions.


The PMP composition described herein may include an antibiotic. Any antibiotic known in the art may be used. Antibiotics are commonly classified based on their mechanism of action, chemical structure, or spectrum of activity.


The antibiotic described herein may target any bacterial function or growth processes and may be either bacteriostatic (e.g., slow or prevent bacterial growth) or bactericidal (e.g., kill bacteria). In some instances, the antibiotic is a bactericidal antibiotic. In some instances, the bactericidal antibiotic is one that targets the bacterial cell wall (e.g., penicillins and cephalosporins); one that targets the cell membrane (e.g., polymyxins); or one that inhibits essential bacterial enzymes (e.g., rifamycins, lipiarmycins, quinolones, and sulfonamides). In some instances, the bactericidal antibiotic is an aminoglycoside (e.g., kasugamycin). In some instances, the antibiotic is a bacteriostatic antibiotic. In some instances the bacteriostatic antibiotic targets protein synthesis (e.g., macrolides, lincosamides, and tetracyclines). Additional classes of antibiotics that may be used herein include cyclic lipopeptides (such as daptomycin), glycylcyclines (such as tigecycline), oxazolidinones (such as linezolid), or lipiarmycins (such as fidaxomicin). Examples of antibiotics include rifampicin, ciprofloxacin, doxycycline, ampicillin, and polymyxin B. The antibiotic described herein may have any level of target specificity (e.g., narrow- or broad-spectrum). In some instances, the antibiotic is a narrow-spectrum antibiotic, and thus targets specific types of bacteria, such as gram-negative or gram-positive bacteria. Alternatively, the antibiotic may be a broad-spectrum antibiotic that targets a wide range of bacteria.


Other non-limiting examples of antibiotics are found in Table 1. One skilled in the art will appreciate that a suitable concentration of each antibiotic in the composition depends on factors such as efficacy, stability of the antibiotic, number of distinct antibiotics, the formulation, and methods of application of the composition.









TABLE 1







Examples of Antibiotics








Antibiotics
Action





Penicillins, cephalosporins, vancomycin
Cell wall synthesis


Polymixin, gramicidin
Membrane active agent,



disrupt cell membrane


Tetracyclines, macrolides,
Inhibit protein synthesis


chloramphenicol, clindamycin,


spectinomycin


Sulfonamides
Inhibit folate-dependent



pathways


Ciprofloxacin
Inhibit DNA-gyrase


Isoniazid, rifampicin, pyrazinamide,
Antimycobacterial agents


ethambutol, (myambutol)l, streptomycin









ii. Antifungal Agents


The PMP compositions described herein can further include an antifungal agent. In some instances, the PMP compositions include two or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, or more than 10) different antifungal agents. For example, the antifungal agent can decrease the fitness of (e.g., decrease growth or kill) a fungal plant pest. A PMP composition including an antifungal as described herein can be contacted with a target fungal pest, or plant infested therewith, in an amount and for a time sufficient to: (a) reach a target level (e.g., a predetermined or threshold level) of antibiotic concentration inside or on the target fungus; and (b) decrease fitness of the target fungus. The antifungals described herein may be formulated in a PMP composition for any of the methods described herein, and in certain instances, may be associated with the PMP thereof.


As used herein, the term “fungicide” or “antifungal agent” refers to a substance that kills or inhibits the growth, proliferation, division, reproduction, or spread of fungi, such as phytopathogenic fungi. Many different types of antifungal agent have been produced commercially. Non limiting examples of antifungal agents include: azoxystrobin, mancozeb, prothioconazole, folpet, tebuconazole, difenoconazole, captan, bupirimate, or fosetyl-Al. Further exemplary fungicides include, but are not limited to, strobilurins, azoxystrobin, dimoxystrobin, enestroburin, fluoxastrobin, kresoxim-methyl, metominostrobin, picoxystrobin, pyraclostrobin, trifloxystrobin, orysastrobin, carboxamides, carboxanilides, benalaxyl, benalaxyl-M, benodanil, carboxin, mebenil, mepronil, fenfuram, fenhexamid, flutolanil, furalaxyl, furcarbanil, furametpyr, metalaxyl, metalaxyl-M (mefenoxam), methfuroxam, metsulfovax, ofurace, oxadixyl, oxycarboxin, penthiopyrad, pyracarbolid, salicylanilide, tecloftalam, thifluzamide, tiadinil, N-biphenylamides, bixafen, boscalid, carboxylic acid morpholides, dimethomorph, flumorph, benzamides, flumetover, fluopicolid (picobenzamid), zoxamid, carboxamides, carpropamid, diclocymet, mandipropamid, silthiofam, azoles, triazoles, bitertanol, bromuconazole, cyproconazole, difenoconazole, diniconazole, enilconazole, epoxiconazole, fenbuconazole, flusilazol, fluquinconazole, flutriafol, hexaconazole, imibenconazole, ipconazole, metconazole, myclobutanil, penconazole, propiconazole, prothioconazole, simeconazole, tebuconazole, tetraconazole, triadimenol, triadimefon, triticonazole, Imidazoles, cyazofamid, imazalil, pefurazoate, prochloraz, triflumizole, benzimidazoles, benomyl, carbendazim, fuberidazole, thiabendazole, ethaboxam, etridiazole, hymexazol, nitrogen-containing heterocyclyl compounds, pyridines, fuazinam, pyrifenox, pyrimidines, bupirimate, cyprodinil, ferimzone, fenarimol, mepanipyrim, nuarimol, pyrimethanil, piperazines, triforine, pyrroles, fludioxonil, fenpiclonil, morpholines, aldimorph, dodemorph, fenpropimorph, tridemorph, dicarboximides, iprodione, procymidone, vinclozolin, acibenzolar-S-methyl, anilazine, captan, captafol, dazomet, diclomezin, fenoxanil, folpet, fenpropidin, famoxadon, fenamidon, octhilinone, probenazole, proquinazid, pyroquilon, quinoxyfen, tricyclazole, carbamates, dithiocarbamates, ferbam, mancozeb, maneb, metiram, metam, propineb, thiram, zineb, ziram, diethofencarb, flubenthiavalicarb, iprovalicarb, propamocarb, guanidines, dodine, iminoctadine, guazatine, kasugamycin, polyoxins, streptomycin, validamycin A, organometallic compounds, fentin salts, sulfur-containing heterocyclyl compounds, isoprothiolane, dithianone, organophosphorous compounds, edifenphos, fosetyl, fosetyl-aluminum, iprobenfos, pyrazophos, tolclofos-methyl, Organochlorine compounds, thiophanate-methyl, chlorothalonil, dichlofluanid, tolylfluanid, flusulfamide, phthalide, hexachlorobenzene, pencycuron, quintozene, nitrophenyl derivatives, binapacryl, dinocap, dinobuton, spiroxamine, cyflufenamid, cymoxanil, metrafenon, N-2-cyanophenyl-3,4-dichloroisothiazol-5-carboxamide (isotianil), N-(3′,4′,5′-trifluorobiphenyl-2-yl)-3-difluoromethyl-1-methylpyrazole-4-carboxamide, 3-[5-(4-chlorophenyl)-2,3-dimethylisoxazolidin-3-yl]-pyridine, N-(3′,4′-dichloro-4-fluorobiphenyl-2-yl)-3-difluoromethyl-1-methylpyrazol-e-4-carboxamide, 5-chloro-7-(4-methylpiperidin-1-yl)-6-(2,4,6-trifluorophenyl)-[1,2,4]tria-zolo[1,5-a]pyrimidine, 2-butoxy-6-iodo-3-propylchromen-4-one, N,N-dimethyl-3-(3-bromo-6-fluoro-2-methylindole-1-sulfonyl)-[1,2,4]triazo-le-1-sulfonamide, methyl-(2-chloro-5-[1-(3-methylbenzyloxyimino)-ethyl]benzyl)carbamate, methyl-(2-chloro-5-[1-(6-methylpyridin-2-ylmethoxy-imino)ethyl]benzyl)carbamate, methyl 3-(4-chlorophenyl)-3-(2-isopropoxycarbonylamino-3-methylbutyryl-amino)propionate, 4-fluorophenyl N-(1-(1-(4-cyanophenyl)ethanesulfonyl)but-2-yl)carbamate, N-(2-(4-[3-(4-chlorophenyl)prop-2-ynyloxy]-3-methoxyphenyl)ethyl)-2-metha-nesulfonylamino-3-methylbutyramide, N-(2-(4-[3-(4-chlorophenyl)prop-2-ynyloxy]-3-methoxyphenyl)ethyl)-2-ethan-esulfonylamino-3-methylbutyramide, N-(4′-bromobiphenyl-2-yl)-4-difluoromethyl-2-methylthiazol-5-carboxamide, N-(4′-trifluoromethylbiphenyl-2-yl)-4-difluoromethyl-2-methylthiazol-5-carboxamide, N-(4′-chloro-3′-fluorobiphenyl-2-yl)-4-difluoromethyl-2-methylt-hiazol-5-carboxamide, or methyl 2-(ortho-((2,5-dimethylphenyloxy-methylene)phenyl)-3-methoxyacrylate. One skilled in the art will appreciate that a suitable concentration of each antifungal in the composition depends on factors such as efficacy, stability of the antifungal, number of distinct antifungals, the formulation, and methods of application of the composition.


iii. Insecticides


The PMP compositions described herein can further include an insecticide. In some instances, the PMP compositions include two or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, or more than 10) different insecticide agents. For example, the insecticide can decrease the fitness of (e.g., decrease growth or kill) an insect plant pest. A PMP composition including an insecticide as described herein can be contacted with a target insect pest, or plant infested therewith, in an amount and for a time sufficient to: (a) reach a target level (e.g., a predetermined or threshold level) of insecticide concentration inside or on the target insect; and (b) decrease fitness of the target insect. The insecticides described herein may be formulated in a PMP composition for any of the methods described herein, and in certain instances, may be associated with the PMP thereof.


As used herein, the term “insecticide” or “insecticidal agent” refers to a substance that kills or inhibits the growth, proliferation, reproduction, or spread of insects, such as agricultural insect pests. Non limiting examples of insecticides are shown in Table 2. Additional non-limiting examples of suitable insecticides include biologics, hormones or pheromones such as azadirachtin, Bacillus species, Beauveria species, codlemone, Metarrhizium species, Paecilomyces species, Bacillus thuringiensis, and Verticillium species, and active compounds having unknown or non-specified mechanisms of action such as fumigants (such as aluminium phosphide, methyl bromide and sulphuryl fluoride) and selective feeding inhibitors (such as cryolite, flonicamid and pymetrozine). One skilled in the art will appreciate that a suitable concentration of each insecticide in the composition depends on factors such as efficacy, stability of the insecticide, number of distinct insecticides, the formulation, and methods of application of the composition.









TABLE 2







Examples of insecticides








Class
Compounds





chloronicotinyls/
acetamiprid, clothianidin, dinotefuran, imidacloprid, nitenpyram,


neonicotinoids
nithiazine, thiacloprid, thiamethoxam, imidaclothiz, (2E)-1-[(2-



chloro-1,3-thiazol-5-yl)methyl]-3,5-dimethyl-N-nitro-1,3,5-tri-azinan-



2-imine, acetylcholinesterase (AChE) inhibitors (such as



carbamates and organophosphates)


carbamates
alanycarb, aldicarb, aldoxycarb, allyxycarb, aminocarb, bendiocarb,



benfuracarb, bufencarb, butacarb, butocarboxim, butoxycarboxim,



carbaryl, carbofuran, carbosulfan, chloethocarb, dimetilan,



ethiofencarb, fenobucarb, fenothiocarb, formetanate, furathiocarb,



isoprocarb, metam-sodium, methiocarb, methomyl, metolcarb,



oxamyl, phosphocarb, pirimicarb, promecarb, propoxur, thiodicarb,



thiofanox, triazamate, trimethacarb, XMC, xylylcarb


organophosphates
acephate, azamethiphos, azinphos (-methyl, -ethyl), bromophos-



ethyl, bromfenvinfos (-methyl), butathiofos, cadusafos,



carbophenothion, chlorethoxyfos, chlorfenvinphos, chlormephos,



chlorpyrifos (-methyl/-ethyl), coumaphos, cyanofenphos,



cyanophos, demeton-S-methyl, demeton-S-methylsulphon, dialifos,



diazinon, dichlofenthion, dichlorvos/DDVP, dicrotophos,



dimethoate, dimethylvinphos, dioxabenzofos, disulfoton, EPN,



ethion, ethoprophos, etrimfos, famphur, fenamiphos, fenitrothion,



fensulfothion, fenthion, flupyrazofos, fonofos, formothion,



fosmethilan, fosthiazate, heptenophos, iodofenphos, iprobenfos,



isazofos, isofenphos, isopropyl O-salicylate, isoxathion, malathion,



mecarbam, methacrifos, methamidophos, methidathion,



mevinphos, monocrotophos, naled, omethoate, oxydemeton-



methyl, parathion (-methyl/-ethyl), phenthoate, phorate, phosalone,



phosmet, phosphamidon, phosphocarb, phoxim, pirimiphos



(-methyl/-ethyl), profenofos, propaphos, propetamphos, prothiofos,



prothoate, pyraclofos, pyridaphenthion, pyridathion, quinalphos,



sebufos, sulfotep, sulprofos, tebupirimfos, temephos, terbufos,



tetrachlorvinphos, thiometon, triazophos, triclorfon, vamidothion


pyrethroids
acrinathrin, allethrin (d-cis-trans, d-trans), cypermethrin (alpha-,



beta-, theta-, zeta-), permethrin (cis-, trans-), beta-cyfluthrin,



bifenthrin, bioallethrin, bioallethrin-S-cyclopentyl-isomer,



bioethanomethrin, biopermethrin, bioresmethrin, chlovaporthrin,



cis-cypermethrin, cis-resmethrin, cis-permethrin, clocythrin,



cycloprothrin, cyfluthrin, cyhalothrin, cyphenothrin, DDT,



deltamethrin, empenthrin (1R-isomer), esfenvalerate, etofenprox,



fenfluthrin, fenpropathrin, fenpyrithrin, fenvalerate, flubrocythrinate,



flucythrinate, flufenprox, flumethrin, fluvalinate, fubfenprox, gamma-



cyhalothrin, imiprothrin, kadethrin, lambda, cyhalothrin,



metofluthrin, phenothrin (1R-trans isomer), prallethrin, profluthrin,



protrifenbute, pyresmethrin, resmethrin, RU 15525, silafluofen, tau-



fluvalinate, tefluthrin, terallethrin, tetramethrin (1R-isomer),



tralocythrin, tralomethrin, transfluthrin, ZXI 8901, pyrethrins



(pyrethrum)


oxadiazines
indoxacarb, acetylcholine receptor modulators (such as spinosyns)


spinosyns
spinosad


cyclodiene
camphechlor, chlordane, endosulfan, gamma-HCH, HCH,



heptachlor,


organochlorines
lindane, methoxychlor


fiproles
acetoprole, ethiprole, vaniliprole, fipronil


mectins
abamectin, avermectin, emamectin, emamectin-benzoate,



fenoxycarb, hydroprene, kinoprene, methoprene, ivermectin,



lepimectin, epofenonane, pyriproxifen, milbemectin, milbemycin,



triprene


diacylhydrazines
chromafenozide, halofenozide, methoxyfenozide, tebufenozide


benzoylureas
bistrifluoron, chlorfluazuron, diflubenzuron, fluazuron, flucycloxuron,



flufenoxuron, hexaflumuron, lufenuron, novaluron, noviflumuron,



penfluoron, teflubenzuron, triflumuron


organotins
azocyclotin, cyhexatin, fenbutatin oxide


pyrroles
chlorfenapyr


dinitrophenols
binapacyrl, dinobuton, dinocap, DNOC


METIs
fenazaquin, fenpyroximate, pyrimidifen, pyridaben, tebufenpyrad,



tolfenpyrad, rotenone, acequinocyl, fluacrypyrim, microbial



disrupters of the intestinal membrane of insects (such as Bacillus




thuringiensis strains), inhibitors of lipid synthesis (such as tetronic




acids and tetramic acids)


tetronic acids
spirodiclofen, spiromesifen, spirotetramat


tetramic acids
cis-3-(2,5-dimethylphenyl)-8-methoxy-2-oxo-1-azaspiro[4.5]dec-3-



en-4-yl ethyl carbonate (alias: carbonic acid, 3-(2,5-



dimethylphenyl)-8-methoxy-2-oxo-1-azaspiro[4.5]dec-3-en-4-yl



ethyl ester; CAS Reg. No.: 382608-10-8), carboxamides (such as



flonicamid), octopaminergic agonists (such as amitraz), inhibitors of



the magnesium-stimulated ATPase (such as propargite), ryanodin



receptor agonists (such as phthalamides or rynaxapyr)


phthalamides
N2-[1,1-dimethyl-2-(methylsulphonyl)ethyl]-3-iodo-N1-[2-methyl--4-



[1,2,2,2-tetrafluoro-1-(trifluoromethyl)ethyl]phenyl]-1,2-benzenedi-



carboxamide (i.e., flubendiamide; CAS reg. No.: 272451-65-7)









iv. Nematicide


The PMP compositions described herein can further include a nematicide. In some instances, the PMP compositions include two or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, or more than 10) different nematicides. For example, the nematicide can decrease the fitness of (e.g., decrease growth or kill) a nematode plant pest. A PMP composition including a nematicide as described herein can be contacted with a target nematode pest, or plant infested therewith, in an amount and for a time sufficient to: (a) reach a target level (e.g., a predetermined or threshold level) of nematicide concentration inside or on the target nematode; and (b) decrease fitness of the target nematode. The nematicides described herein may be formulated in a PMP composition for any of the methods described herein, and in certain instances, may be associated with the PMP thereof.


As used herein, the term “nematicide” or “nematicidal agent” refers to a substance that kills or inhibits the growth, proliferation, reproduction, or spread of nematodes, such as agricultural nematode pests. Non limiting examples of nematicides are shown in Table 3. One skilled in the art will appreciate that a suitable concentration of each nematicide in the composition depends on factors such as efficacy, stability of the nematicide, number of distinct nematicides, the formulation, and methods of application of the composition.









TABLE 3





Examples of Nematicides
















FUMIGANTS
D-D, 1,3-Dichloropropene, Ethylene Dibromide, 1,2-Dibromo-3-



Chloropropane, Methyl Bromide, Chloropicrin, Metam Sodium, Dazomet,



Methyl Isothiocyanate (MITC), Sodium Tetrathiocarbonate, Chloropicrin,


CARBAMATES
Aldicarb, Aldoxycarb, Carbofuran, Oxamyl, Cleothocarb


ORGANOPHOSPHATES
Ethoprophos, Fenamiphos, Cadusafos, Fosthiazate, Fensulfothion,



Thionazin, Isazofos,


BIOCHEMICALS
DITERA ®, CLANDOSAN ®, SINCOCIN ®









v. Molluscicide


The PMP compositions described herein can further include a molluscicide. In some instances, the PMP compositions include two or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, or more than 10) different molluscicides. For example, the molluscicide can decrease the fitness of (e.g., decrease growth or kill) a mollusk plant pest. A PMP composition including a molluscicide as described herein can be contacted with a target mollusk pest, or plant infested therewith, in an amount and for a time sufficient to: (a) reach a target level (e.g., a predetermined or threshold level) of molluscicide concentration inside or on the target mollusk; and (b) decrease fitness of the target mollusk. The molluscicides described herein may be formulated in a PMP composition for any of the methods described herein, and in certain instances, may be associated with the PMP thereof.


As used herein, the term “molluscicide” or “molluscicidal agent” refers to a substance that kills or inhibits the growth, proliferation, reproduction, or spread of mollusks, such as agricultural mollusk pests. A number of chemicals can be employed as a molluscicide, including metal salts such as iron(III) phosphate, aluminium sulfate, and ferric sodium EDTA,[3][4], metaldehyde, methiocarb, or acetylcholinesterase inhibitors. One skilled in the art will appreciate that a suitable concentration of each molluscicide in the composition depends on factors such as efficacy, stability of the molluscicide, number of distinct molluscicides, the formulation, and methods of application of the composition.


vi. Virucides


The PMP compositions described herein can further include a virucide. In some instances, the PMP compositions include two or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, or more than 10) different virucides. For example, the virucide can decrease the fitness of (e.g., decrease or eliminate) a viral plant pathogen. A PMP composition including a virucide as described herein can be contacted with a target virus, or plant infested therewith, in an amount and for a time sufficient to: (a) reach a target level (e.g., a predetermined or threshold level) of virucide concentration; and (b) decrease or eliminate the target virus. The virucides described herein may be formulated in a PMP composition for any of the methods described herein, and in certain instances, may be associated with the PMP thereof.


As used herein, the term “virucide” or “antiviral” refers to a substance that kills or inhibits the growth, proliferation, reproduction, development, or spread of viruses, such as agricultural virus pathogens. A number of agents can be employed as a virucide, including chemicals or biological agents (e.g., nucleic acids, e.g., dsRNA). One skilled in the art will appreciate that a suitable concentration of each virucide in the composition depends on factors such as efficacy, stability of the virucide, number of distinct virucides, the formulation, and methods of application of the composition.


vii. Herbicides


The PMP compositions described herein can further include one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more than 10) herbicides. For example, the herbicide can decrease the fitness of (e.g., decrease or eliminate) a weed. A PMP composition including an herbicide as described herein can be contacted with a target weed in an amount and for a time sufficient to: (a) reach a target level (e.g., a predetermined or threshold level) of herbicide concentration on the plant and (b) decrease the fitness of the weed. The herbicides described herein may be formulated in a PMP composition for any of the methods described herein, and in certain instances, may be associated with the PMP thereof.


As used herein, the term “herbicide” refers to a substance that kills or inhibits the growth, proliferation, reproduction, or spread of weeds. A number of chemicals can be employed as a herbicides, including Glufosinate, Propaquizafop, Metamitron, Metazachlor, Pendimethalin, Flufenacet, Diflufenican, Clomazone, Nicosulfuron, Mesotrione, Pinoxaden, Sulcotrione, Prosulfocarb, Sulfentrazone, Bifenox, Quinmerac, Triallate, Terbuthylazine, Atrazine, Oxyfluorfen, Diuron, Trifluralin, or Chlorotoluron. Further examples of herbicides include, but are not limited to, benzoic acid herbicides, such as dicamba esters, phenoxyalkanoic acid herbicides, such as 2,4-D, MCPA and 2,4-DB esters, aryloxyphenoxypropionic acid herbicides, such as clodinafop, cyhalofop, fenoxaprop, fluazifop, haloxyfop, and quizalofop esters, pyridinecarboxylic acid herbicides, such as aminopyralid, picloram, and clopyralid esters, pyrimidinecarboxylic acid herbicides, such as aminocyclopyrachlor esters, pyridyloxyalkanoic acid herbicides, such as fluoroxypyr and triclopyr esters, and hydroxybenzonitrile herbicides, such as bromoxynil and ioxynil esters, esters of the arylpyridine carboxylic acids, and arylpyrimidine carboxylic acids of the generic structures disclosed in U.S. Pat. Nos. 7,314,849, 7,300,907, and 7,642,220, each of which is incorporated by reference herein in its entirety. In certain embodiments, the herbicide can be selected from the group consisting of 2,4-D, 2,4-DB, acetochlor, acifluorfen, alachlor, ametryn, amitrole, asulam, atrazine, azafenidin, benefin, bensulfuron, bensulide, bentazon, bromacil, bromoxynil, butylate, carfentrazone, chloramben, chlorimuron, chlorproham, chlorsulfuron, clethodim, clomazone, clopyralid, cloransulam, cyanazine, cycloate, DCPA, desmedipham, dichlobenil, diclofop, diclosulam, diethatyl, difenzoquat, diflufenzopyr, dimethenamid-p, diquat, diuron, DSMA, endothall, EPTC, ethalfluralin, ethametsulfuron, ethofumesate, fenoxaprop, fluazifop-P, flucarbazone, flufenacet, flumetsulam, flumiclorac, flumioxazin, fluometuron, fluroxypyr, fluthiacet, fomesafen, foramsulfuron, glufosinate, glyphosate, halosulfuron, haloxyfop, hexazinone, imazamethabenz, imazamox, imazapic, imazaquin, imazethapyr, isoxaben, isoxaflutole, lactofen, linuron, MCPA, MCPB, mesotrione, methazole, metolachlor-s, metribuzin, metsulfuron, molinate, MSMA, napropamide, naptalam, nicosulfuron, norflurazon, oryzalin, oxadiazon, oxasulfuron, oxyfluorfen, paraquat, pebulate, pelargonic acid, pendimethalin, phenmedipham, picloram, primisulfuron, prodiamine, prometryn, pronamide, propachlor, propanil, prosulfuron, pyrazon, pyridate, pyrithiobac, quinclorac, quizalofop, rimsulfuron, sethoxydim, siduron, simazine, sulfentrazone, sulfometuron, sulfosulfuron, tebuthiuron, terbacil, thiazopyr, thifensulfuron, thiobencarb, tralkoxydim, triallate, triasulfuron, tribenuron, triclopyr, trifluralin, triflusulfuron, vernolate. One skilled in the art will appreciate that a suitable concentration of each herbicide in the composition depends on factors such as efficacy, stability of the herbicide, number of distinct herbicides, the formulation, and methods of application of the composition.


viii. Repellents


The PMP compositions described herein can further include a repellent. In some instances, the PMP compositions include two or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, or more than 10) different repellents. For example, the repellent can repel any of the pests described herein (e.g., insects, nematodes, or mollusks); microorganisms (e.g., phytopathogens or endophytes, such as bacteria, fungi, or viruses); or weeds. A PMP composition including a repellent as described herein can be contacted with a target plant, or plant infested therewith, in an amount and for a time sufficient to: (a) reach a target level (e.g., a predetermined or threshold level) of repellent concentration; and (b) decrease the levels of the pest on the plant relative to an untreated plant. The repellent described herein may be formulated in a PMP composition for any of the methods described herein, and in certain instances, may be associated with the PMP thereof.


In some instances, the repellent is an insect repellent. Some examples of well-known insect repellents include: benzil; benzyl benzoate; 2,3,4,5-bis(butyl-2-ene)tetrahydrofurfural (MGK Repellent 11); butoxypolypropylene glycol; N-butylacetanilide; normal-butyl-6,6-dimethyl-5,6-dihydro-1,4-pyrone-2-carboxylate (Indalone); dibutyl adipate; dibutyl phthalate; di-normal-butyl succinate (Tabatrex); N,N-diethyl-meta-toluamide (DEET); dimethyl carbate (endo,endo)-dimethyl bicyclo[2.2.1] hept-5-ene-2,3-dicarboxylate); dimethyl phthalate; 2-ethyl-2-butyl-1,3-propanediol; 2-ethyl-1,3-hexanediol (Rutgers 612); di-normal-propyl isocinchomeronate (MGK Repellent 326); 2-phenylcyclohexanol; p-methane-3,8-diol, and normal-propyl N,N-diethylsuccinamate. Other repellents include citronella oil, dimethyl phthalate, normal-butylmesityl oxide oxalate and 2-ethyl hexanediol-1,3 (See, Kirk-Othmer Encyclopedia of Chemical Technology, 2nd Ed., Vol. 11: 724-728; and The Condensed Chemical Dictionary, 8th Ed., p 756).


An insect repellent may be a synthetic or nonsynthetic insect repellent. Examples of synthetic insect repellents include methyl anthranilate and other anthranilate-based insect repellents, benzaldehyde, DEET (N,N-diethyl-m-toluamide), dimethyl carbate, dimethyl phthalate, icaridin (i.e., picaridin, Bayrepel, and KBR 3023), indalone (e.g., as used in a “6-2-2” mixture (60% Dimethyl phthalate, 20% Indalone, 20% Ethylhexanediol), IR3535 (3-[N-Butyl-N-acetyl]-aminopropionic acid, ethyl ester), metofluthrin, permethrin, SS220, or tricyclodecenyl allyl ether. Examples of natural insect repellents include beautyberry (Callicarpa) leaves, birch tree bark, bog myrtle (Myrica Gale), catnip oil (e.g., nepetalactone), citronella oil, essential oil of the lemon eucalyptus (Corymbia citriodora; e.g., p-menthane-3,8-diol (PMD)), neem oil, lemongrass, tea tree oil from the leaves of Melaleuca alternifolia, tobacco, or extracts thereof.


ix. Fertilizing Agents


The PMP compositions described herein can further include a heterologous fertilizing agent. In some instances, the heterologous fertilizing agent is associated with the PMPs. For example, a PMP may encapsulate the heterologous fertilizing agent. Additionally, or alternatively, the heterologous fertilizing agent can be embedded on or conjugated to the surface of the PMP.


Examples of heterologous fertilizing agents include plant nutrients or plant growth regulators, such as those well known in the art. Alternatively, or additionally, the fertilizing agent can be a peptide, a polypeptide, a nucleic acid, or a polynucleotide that can increase the fitness of a plant symbiont. The fertilizing agent may be an agent that can increase the fitness of a variety of plants or plant symbionts or can be one that targets one or more specific target plants or plant symbionts (e.g., a specific species or genera of plants or plant symbionts).


In some instances, the heterologous fertilizing agent can be modified. For example, the modification can be a chemical modification, e.g., conjugation to a marker, e.g., fluorescent marker or a radioactive marker. In other examples, the modification can include conjugation or operational linkage to a moiety that enhances the stability, delivery, targeting, bioavailability, or half-life of the agent, e.g., a lipid, a glycan, a polymer (e.g., PEG), a cation moiety.


Examples of heterologous fertilizing agents that can be used in the presently disclosed PMP compositions and methods are outlined below.


In some instances, the heterologous fertilizing agent includes any material of natural or synthetic origin that is applied to soils or to plant tissues to supply one or more plant nutrients essential to the growth of plants. The plant nutrient may include a macronutrient, micronutrient, or a combination thereof. Plant macronutrients include nitrogen, phosphorus, potassium, calcium, magnesium, and/or sulfur. Plant micronutrients include copper, iron, manganese, molybdenum, zinc, boron, silicon, cobalt, and/or vanadium. Examples of plant nutrient fertilizers include 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, or methylene urea, phosphorous fertilizers such as diammonium phosphate, monoammonium phosphate, ammonium polyphosphate, concentrated superphosphate and triple superphosphate, or potassium fertilizers such as potassium chloride, potassium sulfate, potassium-magnesium sulfate, potassium nitrate. Such compositions can exist as free salts or ions within the composition. Fertilizers may be designated by the content of one or more of its components, such as nitrogen, phosphorous, or potassium. The content of these elements in a fertilizer may be indicated by the N—P—K value (where N=nitrogen content by weight percentage, P=phosphorous content by weight percentage, and K=potassium content by weight percentage).


Inorganic fertilizers, on the other hand, are manufactured from non-living materials and include, for example, ammonium nitrate, ammonium sulfate, urea, potassium chloride, potash, ammonium phosphate, anhydrous ammonia, and other phosphate salts. Inorganic fertilizers are readily commercially available and contain nutrients in soluble form that are immediately available to the plant. Inorganic fertilizers are generally inexpensive, having a low unit cost for the desired element. One skilled in the art will appreciate that the exact amount of a given element in a fertilizing agent may be calculated and administered to the plant or soil.


Fertilizers may be further classified as either organic fertilizers or inorganic fertilizers. Organic fertilizers include fertilizers having a molecular skeleton with a carbon backbone, such as in compositions derived from living matter. Organic fertilizers are made from materials derived from living things. Animal manures, compost, bonemeal, feather meal, and blood meal are examples of common organic fertilizers. Organic fertilizers, on the other hand, are typically not immediately available to plants and require soil microorganisms to break the fertilizer components down into simpler structures prior to use by the plants. In addition, organic fertilizers may not only elicit a plant growth response as observed with common inorganic fertilizers, but natural organic fertilizers may also stimulate soil microbial population growth and activities. Increased soil microbial population (e.g., plant symbionts) may have significant beneficial effects on the physical and chemical properties of the soil, as well as increasing disease and pest resistance.


In one aspect, a PMP composition including a plant nutrient as described herein can be contacted with the plant in an amount and for a time sufficient to: (a) reach a target level (e.g., a predetermined or threshold level) of plant nutrient concentration inside or on the plant, and (b) increase the fitness of the plant relative to an untreated plant.


In another aspect, a PMP composition including a plant nutrient as described herein can be contacted with the plant symbiont in an amount and for a time sufficient to: (a) reach a target level (e.g., a predetermined or threshold level) of plant nutrient concentration inside or on the plant symbiont (e.g., a bacterial or fungal endosymbiont), and (b) increase the fitness of the plant symbiont relative to an untreated plant symbiont.


The heterologous fertilizing agent may include a plant growth regulator. Exemplary plant growth regulators include auxins, cytokinins, gibberellins, and abscisic acid. In some instances, the plant growth regulator is abscisic cacid, 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 (prohexadione-calcium), prohydrojasmon, thidiazuron, triapenthenol, tributyl phosphorotrithioate, 2,3,5-tri-iodobenzoic acid, trinexapac-ethyl and uniconazole. Other plant growth regulators that can be incorporated seed coating compositions are described in US 2012/0108431, which is incorporated by reference in its entirety.


x. Plant-Modifying Agents


The PMP compositions described herein include one or more heterologous plant-modifying agents. For example, the PMPs may encapsulate the heterologous plant-modifying agent. Alternatively or additionally, the heterologous plant-modifying agent can be embedded on or conjugated to the surface of the PMP.


In some instances, the plant-modifying agent can include a peptide or a nucleic acid. The plant-modifying agent may be an agent that increases the fitness of a variety of plants or can be one that targets one or more specific plants (e.g., a specific species or genera of plants). Additionally, in some instances, the PMP compositions include two or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, or more than 10) different plant-modifying agents.


Further, in some instances, the heterologous plant-modifying agent (e.g., an agent including a nucleic acid molecule or peptide) can be modified. For example, the modification can be a chemical modification, e.g., conjugation to a marker, e.g., fluorescent marker or a radioactive marker. In other examples, the modification can include conjugation or operational linkage to a moiety that enhances the stability, delivery, targeting, bioavailability, or half-life of the agent, e.g., a lipid, a glycan, a polymer (e.g., PEG), a cation moiety.


Examples of heterologous plant-modifying agents (e.g., peptides or nucleic acids) that can be used in the presently disclosed PMP compositions and methods are outlined below.


B. Polypeptides


The PMP composition (e.g., PMPs) described herein may include a heterologous polypeptide. In some instances, the PMP composition described herein includes a polypeptide or functional fragments or derivative thereof that modifies an animal (e.g., a mammal) or a plant (e.g., increases the fitness of the animal or plant). For example, the polypeptide can increase the fitness of an animal or a plant. A PMP composition including a polypeptide as described herein can be contacted with an animal or a plant in an amount and for a time sufficient to: (a) reach a target level (e.g., a predetermined or threshold level) of polypeptide concentration; and (b) modify the animal or plant (e.g., increase the fitness of the animal or plant).


Examples of polypeptides that can be used herein can include an enzyme (e.g., a metabolic recombinase, a helicase, an integrase, a RNAse, a DNAse, or an ubiquitination protein), a pore-forming protein, a signaling ligand, a cell penetrating peptide, a transcription factor, a receptor, an antibody, a nanobody, a peptide or protein therapeutic, a gene editing protein (e.g., CRISPR-Cas system, TALEN, or zinc finger), riboprotein, a protein aptamer, or a chaperone.


Polypeptides included herein may include naturally occurring polypeptides or recombinantly produced variants. In some instances, the polypeptide may be a functional fragments or variants thereof (e.g., an enzymatically active fragment or variant thereof). For example, the polypeptide may be a functionally active variant of any of the polypeptides described herein with at least 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%, or 99% identity, e.g., over a specified region or over the entire sequence, to a sequence of a polypeptide described herein or a naturally occurring polypeptide. In some instances, the polypeptide may have at least 50% (e.g., at least 50%, 60%, 70%, 80%, 90%, 95%, 97%, 99%, or greater) identity to a protein of interest.


The polypeptides described herein may be formulated in a composition for any of the uses described herein. The compositions disclosed herein may include any number or type (e.g., classes) of polypeptides, such as at least about any one of 1 polypeptide, 2, 3, 4, 5, 10, 15, 20, or more polypeptides. A suitable concentration of each polypeptide in the composition depends on factors such as efficacy, stability of the polypeptide, number of distinct polypeptides in the composition, the formulation, and methods of application of the composition. In some instances, each polypeptide in a liquid composition is from about 0.1 ng/mL to about 100 mg/mL. In some instances, each polypeptide in a solid composition is from about 0.1 ng/g to about 100 mg/g.


Methods of making a polypeptide are routine in the art. See, in general, Smales & James (Eds.), Therapeutic Proteins: Methods and Protocols (Methods in Molecular Biology), Humana Press (2005); and Crommelin, Sindelar & Meibohm (Eds.), Pharmaceutical Biotechnology: Fundamentals and Applications, Springer (2013).


Methods for producing a polypeptide involve expression in plant cells, although recombinant proteins can also be produced using insect cells, yeast, bacteria, mammalian cells, or other cells under the control of appropriate promoters. Mammalian expression vectors may comprise nontranscribed elements such as an origin of replication, a suitable promoter and enhancer, and other 5′ or 3′ flanking nontranscribed sequences, and 5′ or 3′ nontranslated sequences such as necessary ribosome binding sites, a polyadenylation site, splice donor and acceptor sites, and termination sequences. DNA sequences derived from the SV40 viral genome, for example, SV40 origin, early promoter, enhancer, splice, and polyadenylation sites may be used to provide the other genetic elements required for expression of a heterologous DNA sequence. Appropriate cloning and expression vectors for use with bacterial, fungal, yeast, and mammalian cellular hosts are described in Green & Sambrook, Molecular Cloning: A Laboratory Manual (Fourth Edition), Cold Spring Harbor Laboratory Press (2012).


Various mammalian cell culture systems can be employed to express and manufacture a recombinant polypeptide agent. Examples of mammalian expression systems include CHO cells, COS cells, HeLA and BHK cell lines. Processes of host cell culture for production of protein therapeutics are described in, e.g., Zhou and Kantardjieff (Eds.), Mammalian Cell Cultures for Biologics Manufacturing (Advances in Biochemical Engineering/Biotechnology), Springer (2014). Purification of proteins is described in Franks, Protein Biotechnology: Isolation, Characterization, and Stabilization, Humana Press (2013); and in Cutler, Protein Purification Protocols (Methods in Molecular Biology), Humana Press (2010). Formulation of protein therapeutics is described in Meyer (Ed.), Therapeutic Protein Drug Products: Practical Approaches to formulation in the Laboratory, Manufacturing, and the Clinic, Woodhead Publishing Series (2012).


In some instances, the PMP composition includes an antibody or antigen binding fragment thereof. For example, an agent described herein may be an antibody that blocks or potentiates activity and/or function of a component of the plant. The antibody may act as an antagonist or agonist of a polypeptide (e.g., enzyme or cell receptor) in the plant. The making and use of antibodies against a target antigen is known in the art. See, for example, Zhiqiang An (Ed.), Therapeutic Monoclonal Antibodies: From Bench to Clinic, 1st Edition, Wiley, 2009 and also Greenfield (Ed.), Antibodies: A Laboratory Manual, 2nd Edition, Cold Spring Harbor Laboratory Press, 2013, for methods of making recombinant antibodies, including antibody engineering, use of degenerate oligonucleotides, 5′-RACE, phage display, and mutagenesis; antibody testing and characterization; antibody pharmacokinetics and pharmacodynamics; antibody purification and storage; and screening and labeling techniques.


C. Nucleic Acids


Numerous nucleic acids are useful in the PMP compositions and methods described herein. The PMP compositions disclosed herein may include any number or type (e.g., classes) of heterologous nucleic acids (e.g., DNA molecule or RNA molecule, e.g., mRNA, guide RNA (gRNA), or inhibitory RNA molecule (e.g., siRNA, shRNA, or miRNA), or a hybrid DNA-RNA molecule), such as at least about 1 class or variant of a nucleic acid, 2, 3, 4, 5, 10, 15, 20, or more classes or variants of nucleic acids. A suitable concentration of each nucleic acid in the composition depends on factors such as efficacy, stability of the nucleic acid, number of distinct nucleic acids, the formulation, and methods of application of the composition. Examples of nucleic acids useful herein include an antisense RNA, a short interfering RNA (siRNA), a short hairpin (shRNA), a microRNA (miRNA), an (asymmetric interfering RNA) aiRNA, a peptide nucleic acid (PNA), a morpholino, a locked nucleic acid (LNA), a piwi-interacting RNA (piRNA), a ribozyme, a deoxyribozymes (DNAzyme), an aptamer (DNA, RNA), a circular RNA (circRNA), a guide RNA (gRNA), or a DNA molecule


A PMP composition including a nucleic acid as described herein can be contacted with a plant in an amount and for a time sufficient to: (a) reach a target level (e.g., a predetermined or threshold level) of nucleic acid concentration; and (b) modify the plant (e.g., increase the fitness of the plant).


i. Nucleic Acid Encoding Peptides


In some instances, the PMP composition includes a heterologous nucleic acid encoding a polypeptide. Nucleic acids encoding a polypeptide may have a length from about 10 to about 50,000 nucleotides (nts), about 25 to about 100 nts, about 50 to about 150 nts, about 100 to about 200 nts, about 150 to about 250 nts, about 200 to about 300 nts, about 250 to about 350 nts, about 300 to about 500 nts, about 10 to about 1000 nts, about 50 to about 1000 nts, about 100 to about 1000 nts, about 1000 to about 2000 nts, about 2000 to about 3000 nts, about 3000 to about 4000 nts, about 4000 to about 5000 nts, about 5000 to about 6000 nts, about 6000 to about 7000 nts, about 7000 to about 8000 nts, about 8000 to about 9000 nts, about 9000 to about 10,000 nts, about 10,000 to about 15,000 nts, about 10,000 to about 20,000 nts, about 10,000 to about 25,000 nts, about 10,000 to about 30,000 nts, about 10,000 to about 40,000 nts, about 10,000 to about 45,000 nts, about 10,000 to about 50,000 nts, or any range therebetween.


The PMP composition may also include functionally active variants of a nucleic acid sequence of interest. In some instances, the variant of the nucleic acids has at least 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%, or 99% identity, e.g., over a specified region or over the entire sequence, to a sequence of a nucleic acid of interest. In some instances, the invention includes a functionally active polypeptide encoded by a nucleic acid variant as described herein. In some instances, the functionally active polypeptide encoded by the nucleic acid variant has at least 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%, or 99% identity, e.g., over a specified region or over the entire amino acid sequence, to a sequence of a polypeptide of interest or the naturally derived polypeptide sequence.


Certain methods for expressing a nucleic acid encoding a protein may involve expression in cells, including insect, yeast, plant, bacteria, or other cells under the control of appropriate promoters. Expression vectors may include nontranscribed elements, such as an origin of replication, a suitable promoter and enhancer, and other 5′ or 3′ flanking nontranscribed sequences, and 5′ or 3′ nontranslated sequences such as necessary ribosome binding sites, a polyadenylation site, splice donor and acceptor sites, and termination sequences. DNA sequences derived from the SV40 viral genome, for example, SV40 origin, early promoter, enhancer, splice, and polyadenylation sites may be used to provide the other genetic elements required for expression of a heterologous DNA sequence. Appropriate cloning and expression vectors for use with bacterial, fungal, yeast, and mammalian cellular hosts are described in Green et al., Molecular Cloning: A Laboratory Manual, Fourth Edition, Cold Spring Harbor Laboratory Press, 2012.


Genetic modification using recombinant methods is generally known in the art. A nucleic acid sequence coding for a desired gene can be obtained using recombinant methods known in the art, such as, for example by screening libraries from cells expressing the gene, by deriving the gene from a vector known to include the same, or by isolating directly from cells and tissues containing the same, using standard techniques. Alternatively, a gene of interest can be produced synthetically, rather than cloned.


Expression of natural or synthetic nucleic acids is typically achieved by operably linking a nucleic acid encoding the gene of interest to a promoter, and incorporating the construct into an expression vector. Expression vectors can be suitable for replication and expression in bacteria. Expression vectors can also be suitable for replication and integration in eukaryotes. Typical cloning vectors contain transcription and translation terminators, initiation sequences, and promoters useful for expression of the desired nucleic acid sequence.


Additional promoter elements, e.g., enhancers, regulate the frequency of transcriptional initiation. Typically, these are located in the region 30-110 basepairs (bp) upstream of the start site, although a number of promoters have recently been shown to contain functional elements downstream of the start site as well. The spacing between promoter elements frequently is flexible, so that promoter function is preserved when elements are inverted or moved relative to one another. In the thymidine kinase (tk) promoter, the spacing between promoter elements can be increased to 50 bp apart before activity begins to decline. Depending on the promoter, it appears that individual elements can function either cooperatively or independently to activate transcription.


One example of a suitable promoter is the immediate early cytomegalovirus (CMV) promoter sequence. This promoter sequence is a strong constitutive promoter sequence capable of driving high levels of expression of any polynucleotide sequence operatively linked thereto. Another example of a suitable promoter is Elongation Growth Factor-1α (EF-1α). However, other constitutive promoter sequences may also be used, including, but not limited to the simian virus 40 (SV40) early promoter, mouse mammary tumor virus (MMTV), human immunodeficiency virus (HIV) long terminal repeat (LTR) promoter, MoMuLV promoter, an avian leukemia virus promoter, an Epstein-Barr virus immediate early promoter, a Rous sarcoma virus promoter, as well as human gene promoters such as, but not limited to, the actin promoter, the myosin promoter, the hemoglobin promoter, and the creatine kinase promoter.


Alternatively, the promoter may be an inducible promoter. The use of an inducible promoter provides a molecular switch capable of turning on expression of the polynucleotide sequence which it is operatively linked when such expression is desired, or turning off the expression when expression is not desired. Examples of inducible promoters include, but are not limited to a metallothionine promoter, a glucocorticoid promoter, a progesterone promoter, and a tetracycline promoter.


The expression vector to be introduced can also contain either a selectable marker gene or a reporter gene or both to facilitate identification and selection of expressing cells from the population of cells sought to be transfected or infected through viral vectors. In other aspects, the selectable marker may be carried on a separate piece of DNA and used in a co-transfection procedure. Both selectable markers and reporter genes may be flanked with appropriate regulatory sequences to enable expression in the host cells. Useful selectable markers include, for example, antibiotic-resistance genes, such as neo and the like.


Reporter genes may be used for identifying potentially transformed cells and for evaluating the functionality of regulatory sequences. In general, a reporter gene is a gene that is not present in or expressed by the recipient source and that encodes a polypeptide whose expression is manifested by some easily detectable property, e.g., enzymatic activity. Expression of the reporter gene is assayed at a suitable time after the DNA has been introduced into the recipient cells. Suitable reporter genes may include genes encoding luciferase, beta-galactosidase, chloramphenicol acetyl transferase, secreted alkaline phosphatase, or the green fluorescent protein gene (e.g., Ui-Tei et al., FEBS Letters 479:79-82, 2000). Suitable expression systems are well known and may be prepared using known techniques or obtained commercially. In general, the construct with the minimal 5′ flanking region showing the highest level of expression of reporter gene is identified as the promoter. Such promoter regions may be linked to a reporter gene and used to evaluate agents for the ability to modulate promoter-driven transcription.


In some instances, an organism may be genetically modified to alter expression of one or more proteins. Expression of the one or more proteins may be modified for a specific time, e.g., development or differentiation state of the organism. In one instances, the invention includes a composition to alter expression of one or more proteins, e.g., proteins that affect activity, structure, or function. Expression of the one or more proteins may be restricted to a specific location(s) or widespread throughout the organism.


ii. Synthetic mRNA


The PMP composition may include a synthetic mRNA molecule, e.g., a synthetic mRNA molecule encoding a polypeptide. The synthetic mRNA molecule can be modified, e.g., chemically. The mRNA molecule can be chemically synthesized or transcribed in vitro. The mRNA molecule can be disposed on a plasmid, e.g., a viral vector, bacterial vector, or eukaryotic expression vector. In some examples, the mRNA molecule can be delivered to cells by transfection, electroporation, or transduction (e.g., adenoviral or lentiviral transduction).


In some instances, the modified RNA agent of interest described herein has modified nucleosides or nucleotides. Such modifications are known and are described, e.g., in WO 2012/019168. Additional modifications are described, e.g., in WO 2015/038892; WO 2015/038892; WO 2015/089511; WO 2015/196130; WO 2015/196118 and WO 2015/196128 A2.


In some instances, the modified RNA encoding a polypeptide of interest has one or more terminal modification, e.g., a 5′ cap structure and/or a poly-A tail (e.g., of between 100-200 nucleotides in length). The 5′ cap structure may be selected from the group consisting of CapO, Capl, ARCA, inosine, NI-methyl-guanosine, 2′fluoro-guanosine, 7-deaza-guanosine, 8-oxo-guanosine, 2-amino-guanosine, LNA-guanosine, and 2-azido-guanosine. In some cases, the modified RNAs also contain a 5′ UTR including at least one Kozak sequence, and a 3′ UTR. Such modifications are known and are described, e.g., in WO 2012/135805 and WO 2013/052523. Additional terminal modifications are described, e.g., in WO 2014/164253 and WO 2016/011306, WO 2012/045075, and WO 2014/093924. Chimeric enzymes for synthesizing capped RNA molecules (e.g., modified mRNA) which may include at least one chemical modification are described in WO 2014/028429.


In some instances, a modified mRNA may be cyclized, or concatemerized, to generate a translation competent molecule to assist interactions between poly-A binding proteins and 5′-end binding proteins. The mechanism of cyclization or concatemerization may occur through at least 3 different routes: 1) chemical, 2) enzymatic, and 3) ribozyme catalyzed. The newly formed 5′-/3′-linkage may be intramolecular or intermolecular. Such modifications are described, e.g., in WO 2013/151736.


Methods of making and purifying modified RNAs are known and disclosed in the art. For example, modified RNAs are made using only in vitro transcription (IVT) enzymatic synthesis. Methods of making IVT polynucleotides are known in the art and are described in WO 2013/151666, WO 2013/151668, WO 2013/151663, WO 2013/151669, WO 2013/151670, WO 2013/151664, WO 2013/151665, WO 2013/151671, WO 2013/151672, WO 2013/151667 and WO 2013/151736. Methods of purification include purifying an RNA transcript including a polyA tail by contacting the sample with a surface linked to a plurality of thymidines or derivatives thereof and/or a plurality of uracils or derivatives thereof (polyT/U) under conditions such that the RNA transcript binds to the surface and eluting the purified RNA transcript from the surface (WO 2014/152031); using ion (e.g., anion) exchange chromatography that allows for separation of longer RNAs up to 10,000 nucleotides in length via a scalable method (WO 2014/144767); and subjecting a modified mRNA sample to DNAse treatment (WO 2014/152030).


Formulations of modified RNAs are known and are described, e.g., in WO 2013/090648. For example, the formulation may be, but is not limited to, nanoparticles, poly(lactic-co-glycolic acid)(PLGA) microspheres, lipidoids, lipoplex, liposome, polymers, carbohydrates (including simple sugars), cationic lipids, fibrin gel, fibrin hydrogel, fibrin glue, fibrin sealant, fibrinogen, thrombin, rapidly eliminated lipid nanoparticles (reLNPs) and combinations thereof.


Modified RNAs encoding polypeptides in the fields of human disease, antibodies, viruses, and a variety of in vivo settings are known and are disclosed in for example, Table 6 of International Publication Nos. WO 2013/151666, WO 2013/151668, WO 2013/151663, WO 2013/151669, WO 2013/151670, WO 2013/151664, WO 2013/151665, WO 2013/151736; Tables 6 and 7 International Publication No. WO 2013/151672; Tables 6, 178 and 179 of International Publication No. WO 2013/151671; Tables 6, 185 and 186 of International Publication No WO 2013/151667. Any of the foregoing may be synthesized as an IVT polynucleotide, chimeric polynucleotide or a circular polynucleotide, and each may include one or more modified nucleotides or terminal modifications.


iii. Inhibitory RNA


In some instances, the PMP composition includes an inhibitory RNA molecule, e.g., that acts via the RNA interference (RNAi) pathway. In some instances, the inhibitory RNA molecule decreases the level of gene expression in a plant and/or decreases the level of a protein in the plant. In some instances, the inhibitory RNA molecule inhibits expression of a plant gene. For example, an inhibitory RNA molecule may include a short interfering RNA, short hairpin RNA, and/or a microRNA that targets a gene in the plant. Certain RNA molecules can inhibit gene expression through the biological process of RNA interference (RNAi). RNAi molecules include RNA or RNA-like structures typically containing 15-50 base pairs (such as about 18-25 base pairs) and having a nucleobase sequence identical (complementary) or nearly identical (substantially complementary) to a coding sequence in an expressed target gene within the cell. RNAi molecules include, but are not limited to: short interfering RNAs (siRNAs), double-strand RNAs (dsRNA), short hairpin RNAs (shRNA), meroduplexes, dicer substrates, and multivalent RNA interference (U.S. Pat. Nos. 8,084,599 8,349,809, 8,513,207 and 9,200,276). A shRNA is a RNA molecule including a hairpin turn that decreases expression of target genes via RNAi. shRNAs can be delivered to cells in the form of plasmids, e.g., viral or bacterial vectors, e.g., by transfection, electroporation, or transduction). A microRNA is a non-coding RNA molecule that typically has a length of about 22 nucleotides. MiRNAs bind to target sites on mRNA molecules and silence the mRNA, e.g., by causing cleavage of the mRNA, destabilization of the mRNA, or inhibition of translation of the mRNA. In some instances, the inhibitory RNA molecule decreases the level and/or activity of a negative regulator of function. In other instances, the inhibitor RNA molecule decreases the level and/or activity of an inhibitor of a positive regulator of function. The inhibitory RNA molecule can be chemically synthesized or transcribed in vitro.


In some instances, the nucleic acid is a DNA, a RNA, or a PNA. In some instances, the RNA is an inhibitory RNA. In some instances, the inhibitory RNA inhibits gene expression in a plant. In some instances, the nucleic acid is an mRNA, a modified mRNA, or a DNA molecule that, in the plant, increases expression of an enzyme (e.g., a metabolic recombinase, a helicase, an integrase, a RNAse, a DNAse, or an ubiquitination protein), a pore-forming protein, a signaling ligand, a cell penetrating peptide, a transcription factor, a receptor, an antibody, a nanobody, a gene editing protein (e.g., CRISPR-Cas system, TALEN, or zinc finger), riboprotein, a protein aptamer, or a chaperone. In some instances, the nucleic acid is an mRNA, a modified mRNA, or a DNA molecule that increases the expression of an enzyme (e.g., a metabolic enzyme, a recombinase enzyme, a helicase enzyme, an integrase enzyme, a RNAse enzyme, a DNAse enzyme, or an ubiquitination protein), a pore-forming protein, a signaling ligand, a cell penetrating peptide, a transcription factor, a receptor, an antibody, a nanobody, a gene editing protein (e.g., a CRISPR-Cas system, a TALEN, or a zinc finger), a riboprotein, a protein aptamer, or a chaperone. In some instances, the increase in expression in the plant is an increase in expression of about 5%, 10%, 15%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or more than 100% relative to a reference level (e.g., the expression in an untreated plant). In some instances, the increase in expression in the plant is an increase in expression of about 2× fold, about 4× fold, about 5× fold, about 10× fold, about 20× fold, about 25× fold, about 50× fold, about 75× fold, or about 100× fold or more, relative to a reference level (e.g., the expression in an untreated plant).


In some instances, the nucleic acid is an antisense RNA, a siRNA, a shRNA, a miRNA, an aiRNA, a PNA, a morpholino, a LNA, a piRNA, a ribozyme, a DNAzyme, an aptamer (DNA, RNA), a circRNA, a gRNA, or a DNA molecules (e.g., an antisense polynucleotide) to reduces, in the plant, expression of, e.g., an enzyme (a metabolic enzyme, a recombinase enzyme, a helicase enzyme, an integrase enzyme, a RNAse enzyme, a DNAse enzyme, a polymerase enzyme, a ubiquitination protein, a superoxide management enzyme, or an energy production enzyme), a transcription factor, a secretory protein, a structural factor (actin, kinesin, ortubulin), a riboprotein, a protein aptamer, a chaperone, a receptor, a signaling ligand, or a transporter. In some instances, the decrease in expression in the plant is a decrease in expression of about 5%, 10%, 15%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or more than 100% relative to a reference level (e.g., the expression in an untreated plant). In some instances, the decrease in expression in the plant is a decrease in expression of about 2× fold, about 4× fold, about 5× fold, about 10× fold, about 20× fold, about 25× fold, about 50× fold, about 75× fold, or about 100× fold or more, relative to a reference level (e.g., the expression in an untreated plant).


RNAi molecules include a sequence substantially complementary, or fully complementary, to all or a fragment of a target gene. RNAi molecules may complement sequences at the boundary between introns and exons to prevent the maturation of newly-generated nuclear RNA transcripts of specific genes into mRNA for transcription. RNAi molecules complementary to specific genes can hybridize with the mRNA for a target gene and prevent its translation. The antisense molecule can be DNA, RNA, or a derivative or hybrid thereof. Examples of such derivative molecules include, but are not limited to, peptide nucleic acid (PNA) and phosphorothioate-based molecules such as deoxyribonucleic guanidine (DNG) or ribonucleic guanidine (RNG).


RNAi molecules can be provided as ready-to-use RNA synthesized in vitro or as an antisense gene transfected into cells which will yield RNAi molecules upon transcription. Hybridization with mRNA results in degradation of the hybridized molecule by RNAse H and/or inhibition of the formation of translation complexes. Both result in a failure to produce the product of the original gene.


The length of the RNAi molecule that hybridizes to the transcript of interest may be around 10 nucleotides, between about 15 or 30 nucleotides, or about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30 or more nucleotides. The degree of identity of the antisense sequence to the targeted transcript may be at least 75%, at least 80%, at least 85%, at least 90%, or at least 95.


RNAi molecules may also include overhangs, i.e., typically unpaired, overhanging nucleotides which are not directly involved in the double helical structure normally formed by the core sequences of the herein defined pair of sense strand and antisense strand. RNAi molecules may contain 3′ and/or 5′ overhangs of about 1-5 bases independently on each of the sense strands and antisense strands. In some instances, both the sense strand and the antisense strand contain 3′ and 5′ overhangs. In some instances, one or more of the 3′ overhang nucleotides of one strand base pairs with one or more 5′ overhang nucleotides of the other strand. In other instances, the one or more of the 3′ overhang nucleotides of one strand base do not pair with the one or more 5′ overhang nucleotides of the other strand. The sense and antisense strands of an RNAi molecule may or may not contain the same number of nucleotide bases. The antisense and sense strands may form a duplex wherein the 5′ end only has a blunt end, the 3′ end only has a blunt end, both the 5′ and 3′ ends are blunt ended, or neither the 5′ end nor the 3′ end are blunt ended. In another instance, one or more of the nucleotides in the overhang contains a thiophosphate, phosphorothioate, deoxynucleotide inverted (3′ to 3′ linked) nucleotide or is a modified ribonucleotide or deoxynucleotide.


Small interfering RNA (siRNA) molecules include a nucleotide sequence that is identical to about 15 to about 25 contiguous nucleotides of the target mRNA. In some instances, the siRNA sequence commences with the dinucleotide AA, includes a GC-content of about 30-70% (about 30-60%, about 40-60%, or about 45%-55%), and does not have a high percentage identity to any nucleotide sequence other than the target in the genome in which it is to be introduced, for example as determined by standard BLAST search.


siRNAs and shRNAs resemble intermediates in the processing pathway of the endogenous microRNA (miRNA) genes (Bartel, Cell 116:281-297, 2004). In some instances, siRNAs can function as miRNAs and vice versa (Zeng et al., Mol. Cell 9:1327-1333, 2002; Doench et al., Genes Dev. 17:438-442, 2003). Exogenous siRNAs downregulate mRNAs with seed complementarity to the siRNA (Birmingham et al., Nat. Methods 3:199-204, 2006). Multiple target sites within a 3′ UTR give stronger downregulation (Doench et al., Genes Dev. 17:438-442, 2003).


Known effective siRNA sequences and cognate binding sites are also well represented in the relevant literature. RNAi molecules are readily designed and produced by technologies known in the art. In addition, there are computational tools that increase the chance of finding effective and specific sequence motifs (Pei et al., Nat. Methods 3(9):670-676, 2006; Reynolds et al., Nat. Biotechnol. 22(3):326-330, 2004; Khvorova et al., Nat. Struct. Biol. 10(9):708-712, 2003; Schwarz et al., Cell 115(2):199-208, 2003; Ui-Tei et al., Nucleic Acids Res. 32(3):936-948, 2004; Heale et al., Nucleic Acids Res. 33(3):e30, 2005; Chalk et al., Biochem. Biophys. Res. Commun. 319(1):264-274, 2004; and Amarzguioui et al., Biochem. Biophys. Res. Commun. 316(4):1050-1058, 2004).


The RNAi molecule modulates expression of RNA encoded by a gene. Because multiple genes can share some degree of sequence homology with each other, in some instances, the RNAi molecule can be designed to target a class of genes with sufficient sequence homology. In some instances, the RNAi molecule can contain a sequence that has complementarity to sequences that are shared amongst different gene targets or are unique for a specific gene target. In some instances, the RNAi molecule can be designed to target conserved regions of an RNA sequence having homology between several genes thereby targeting several genes in a gene family (e.g., different gene isoforms, splice variants, mutant genes, etc.). In some instances, the RNAi molecule can be designed to target a sequence that is unique to a specific RNA sequence of a single gene.


An inhibitory RNA molecule can be modified, e.g., to contain modified nucleotides, e.g., 2′-fluoro, 2′-o-methyl, 2′-deoxy, unlocked nucleic acid, 2′-hydroxy, phosphorothioate, 2′-thiouridine, 4′-thiouridine, 2′-deoxyuridine. Without being bound by theory, it is believed that such modifications can increase nuclease resistance and/or serum stability, or decrease immunogenicity.


In some instances, the RNAi molecule is linked to a delivery polymer via a physiologically labile bond or linker. The physiologically labile linker is selected such that it undergoes a chemical transformation (e.g., cleavage) when present in certain physiological conditions, (e.g., disulfide bond cleaved in the reducing environment of the cell cytoplasm). Release of the molecule from the polymer, by cleavage of the physiologically labile linkage, facilitates interaction of the molecule with the appropriate cellular components for activity.


The RNAi molecule-polymer conjugate may be formed by covalently linking the molecule to the polymer. The polymer is polymerized or modified such that it contains a reactive group A. The RNAi molecule is also polymerized or modified such that it contains a reactive group B. Reactive groups A and B are chosen such that they can be linked via a reversible covalent linkage using methods known in the art.


Conjugation of the RNAi molecule to the polymer can be performed in the presence of an excess of polymer. Because the RNAi molecule and the polymer may be of opposite charge during conjugation, the presence of excess polymer can reduce or eliminate aggregation of the conjugate. Alternatively, an excess of a carrier polymer, such as a polycation, can be used. The excess polymer can be removed from the conjugated polymer prior to administration of the conjugate. Alternatively, the excess polymer can be co-administered with the conjugate.


The making and use of inhibitory agents based on non-coding RNA such as ribozymes, RNAse P, siRNAs, and miRNAs are also known in the art, for example, as described in Sioud, RNA Therapeutics: Function, Design, and Delivery (Methods in Molecular Biology). Humana Press (2010).


iv. Gene Editing


The PMP compositions described herein may include a component of a gene editing system. For example, the agent may introduce an alteration (e.g., insertion, deletion (e.g., knockout), translocation, inversion, single point mutation, or other mutation) in a gene in the plant. Exemplary gene editing systems include the zinc finger nucleases (ZFNs), Transcription Activator-Like Effector-based Nucleases (TALEN), and the clustered regulatory interspaced short palindromic repeat (CRISPR) system. ZFNs, TALENs, and CRISPR-based methods are described, e.g., in Gaj et al., Trends Biotechnol. 31(7):397-405, 2013.


In a typical CRISPR/Cas system, an endonuclease is directed to a target nucleotide sequence (e.g., a site in the genome that is to be sequence-edited) by sequence-specific, non-coding guide RNAs that target single- or double-stranded DNA sequences. Three classes (I-III) of CRISPR systems have been identified. The class II CRISPR systems use a single Cas endonuclease (rather than multiple Cas proteins). One class II CRISPR system includes a type II Cas endonuclease such as Cas9, a CRISPR RNA (crRNA), and a trans-activating crRNA (tracrRNA). The crRNA contains a guide RNA, i.e., typically an about 20-nucleotide RNA sequence that corresponds to a target DNA sequence. The crRNA also contains a region that binds to the tracrRNA to form a partially double-stranded structure which is cleaved by RNase III, resulting in a crRNA/tracrRNA hybrid. The RNAs serve as guides to direct Cas proteins to silence specific DNA/RNA sequences, depending on the spacer sequence. See, e.g., Horvath et al., Science 327:167-170, 2010; Makarova et al., Biology Direct 1:7, 2006; Pennisi, Science 341:833-836, 2013. The target DNA sequence must generally be adjacent to a protospacer adjacent motif (PAM) that is specific for a given Cas endonuclease; however, PAM sequences appear throughout a given genome. CRISPR endonucleases identified from various prokaryotic species have unique PAM sequence requirements; examples of PAM sequences include 5′-NGG (SEQ ID NO: 78) (Streptococcus pyogenes), 5′-NNAGAA (SEQ ID NO: 79) (Streptococcus thermophilus CRISPR1), 5′-NGGNG (SEQ ID NO: 80) (Streptococcus thermophilus CRISPR3), and 5′-NNNGATT (SEQ ID NO: 81) (Neisseria meningiditis).


Some endonucleases, e.g., Cas9 endonucleases, are associated with G-rich PAM sites, e.g., 5′-NGG (SEQ ID NO: 78), and perform blunt-end cleaving of the target DNA at a location 3 nucleotides upstream from (5′ from) the PAM site. Another class II CRISPR system includes the type V endonuclease Cpf1, which is smaller than Cas9; examples include AsCpf1 (from Acidaminococcus sp.) and LbCpf1 (from Lachnospiraceae sp.). Cpf1-associated CRISPR arrays are processed into mature crRNAs without the requirement of a tracrRNA; in other words a Cpf1 system requires only the Cpf1 nuclease and a crRNA to cleave the target DNA sequence. Cpf1 endonucleases, are associated with T-rich PAM sites, e.g., 5′-TTN. Cpf1 can also recognize a 5′-CTA PAM motif. Cpf1 cleaves the target DNA by introducing an offset or staggered double-strand break with a 4- or 5-nucleotide 5′ overhang, for example, cleaving a target DNA with a 5-nucleotide offset or staggered cut located 18 nucleotides downstream from (3′ from) from the PAM site on the coding strand and 23 nucleotides downstream from the PAM site on the complimentary strand; the 5-nucleotide overhang that results from such offset cleavage allows more precise genome editing by DNA insertion by homologous recombination than by insertion at blunt-end cleaved DNA. See, e.g., Zetsche et al., Cell 163:759-771, 2015.


For the purposes of gene editing, CRISPR arrays can be designed to contain one or multiple guide RNA sequences corresponding to a desired target DNA sequence; see, for example, Cong et al., Science 339:819-823, 2013; Ran et al., Nature Protocols 8:2281-2308, 2013. At least about 16 or 17 nucleotides of gRNA sequence are required by Cas9 for DNA cleavage to occur; for Cpf1 at least about 16 nucleotides of gRNA sequence is needed to achieve detectable DNA cleavage. In practice, guide RNA sequences are generally designed to have a length of between 17-24 nucleotides (e.g., 19, 20, or 21 nucleotides) and complementarity to the targeted gene or nucleic acid sequence. Custom gRNA generators and algorithms are available commercially for use in the design of effective guide RNAs. Gene editing has also been achieved using a chimeric single guide RNA (sgRNA), an engineered (synthetic) single RNA molecule that mimics a naturally occurring crRNA-tracrRNA complex and contains both a tracrRNA (for binding the nuclease) and at least one crRNA (to guide the nuclease to the sequence targeted for editing). Chemically modified sgRNAs have also been demonstrated to be effective in genome editing; see, for example, Hendel et al., Nature Biotechnol. 985-991, 2015.


Whereas wild-type Cas9 generates double-strand breaks (DSBs) at specific DNA sequences targeted by a gRNA, a number of CRISPR endonucleases having modified functionalities are available, for example: a nickase version of Cas9 generates only a single-strand break; a catalytically inactive Cas9 (dCas9) does not cut the target DNA but interferes with transcription by steric hindrance. dCas9 can further be fused with an effector to repress (CRISPRi) or activate (CRISPRa) expression of a target gene. For example, Cas9 can be fused to a transcriptional repressor (e.g., a KRAB domain) or a transcriptional activator (e.g., a dCas9-VP64 fusion). A catalytically inactive Cas9 (dCas9) fused to Fokl nuclease (dCas9-Fokl) can be used to generate DSBs at target sequences homologous to two gRNAs. See, e.g., the numerous CRISPR/Cas9 plasmids disclosed in and publicly available from the Addgene repository (Addgene, 75 Sidney St., Suite 550A, Cambridge, Mass. 02139; addgene.org/crispr/). A double nickase Cas9 that introduces two separate double-strand breaks, each directed by a separate guide RNA, is described as achieving more accurate genome editing by Ran et al., Cell 154:1380-1389, 2013.


CRISPR technology for editing the genes of eukaryotes is disclosed in US Patent Application Publications US 2016/0138008 A1 and US 2015/0344912 A1, and in U.S. Pat. Nos. 8,697,359, 8,771,945, 8,945,839, 8,999,641, 8,993,233, 8,895,308, 8,865,406, 8,889,418, 8,871,445, 8,889,356, 8,932,814, 8,795,965, and 8,906,616. Cpf1 endonuclease and corresponding guide RNAs and PAM sites are disclosed in US Patent Application Publication 2016/0208243 A1.


In some instances, the desired genome modification involves homologous recombination, wherein one or more double-stranded DNA breaks in the target nucleotide sequence is generated by the RNA-guided nuclease and guide RNA(s), followed by repair of the break(s) using a homologous recombination mechanism (homology-directed repair). In such instances, a donor template that encodes the desired nucleotide sequence to be inserted or knocked-in at the double-stranded break is provided to the cell or subject; examples of suitable templates include single-stranded DNA templates and double-stranded DNA templates (e.g., linked to the polypeptide described herein). In general, a donor template encoding a nucleotide change over a region of less than about 50 nucleotides is provided in the form of single-stranded DNA; larger donor templates (e.g., more than 100 nucleotides) are often provided as double-stranded DNA plasmids. In some instances, the donor template is provided to the cell or subject in a quantity that is sufficient to achieve the desired homology-directed repair but that does not persist in the cell or subject after a given period of time (e.g., after one or more cell division cycles). In some instances, a donor template has a core nucleotide sequence that differs from the target nucleotide sequence (e.g., a homologous endogenous genomic region) by at least 1, at least 5, at least 10, at least 20, at least 30, at least 40, at least 50, or more nucleotides. This core sequence is flanked by homology arms or regions of high sequence identity with the targeted nucleotide sequence; in some instances, the regions of high identity include at least 10, at least 50, at least 100, at least 150, at least 200, at least 300, at least 400, at least 500, at least 600, at least 750, or at least 1000 nucleotides on each side of the core sequence. In some instances where the donor template is in the form of a single-stranded DNA, the core sequence is flanked by homology arms including at least 10, at least 20, at least 30, at least 40, at least 50, at least 60, at least 70, at least 80, or at least 100 nucleotides on each side of the core sequence. In instances, where the donor template is in the form of a double-stranded DNA, the core sequence is flanked by homology arms including at least 500, at least 600, at least 700, at least 800, at least 900, or at least 1000 nucleotides on each side of the core sequence. In one instance, two separate double-strand breaks are introduced into the cell or subject's target nucleotide sequence with a double nickase Cas9 (see Ran et al., Cell 154:1380-1389, 2013), followed by delivery of the donor template.


In some instances, the composition includes a gRNA and a targeted nuclease, e.g., a Cas9, e.g., a wild type Cas9, a nickase Cas9 (e.g., Cas9 D10A), a dead Cas9 (dCas9), eSpCas9, Cpf1, C2C1, or C2C3, or a nucleic acid encoding such a nuclease. The choice of nuclease and gRNA(s) is determined by whether the targeted mutation is a deletion, substitution, or addition of nucleotides, e.g., a deletion, substitution, or addition of nucleotides to a targeted sequence. Fusions of a catalytically inactive endonuclease e.g., a dead Cas9 (dCas9, e.g., D10A; H840A) tethered with all or a portion of (e.g., biologically active portion of) an (one or more) effector domain create chimeric proteins that can be linked to the polypeptide to guide the composition to specific DNA sites by one or more RNA sequences (sgRNA) to modulate activity and/or expression of one or more target nucleic acids sequences.


In instances, the agent includes a guide RNA (gRNA) for use in a CRISPR system for gene editing. In some instances, the agent includes a zinc finger nuclease (ZFN), or a mRNA encoding a ZFN, that targets (e.g., cleaves) a nucleic acid sequence (e.g., DNA sequence) of a gene in the plant. In some instances, the agent includes a TALEN, or an mRNA encoding a TALEN, that targets (e.g., cleaves) a nucleic acid sequence (e.g., DNA sequence) in a gene in the plant.


For example, the gRNA can be used in a CRISPR system to engineer an alteration in a gene in the plant. In other examples, the ZFN and/or TALEN can be used to engineer an alteration in a gene in the plant. Exemplary alterations include insertions, deletions (e.g., knockouts), translocations, inversions, single point mutations, or other mutations. The alteration can be introduced in the gene in a cell, e.g., in vitro, ex vivo, or in vivo. In some examples, the alteration increases the level and/or activity of a gene in the plant. In other examples, the alteration decreases the level and/or activity of (e.g., knocks down or knocks out) a gene in the plant. In yet another example, the alteration corrects a defect (e.g., a mutation causing a defect), in a gene in the plant.


In some instances, the CRISPR system is used to edit (e.g., to add or delete a base pair) a target gene in the plant. In other instances, the CRISPR system is used to introduce a premature stop codon, e.g., thereby decreasing the expression of a target gene. In yet other instances, the CRISPR system is used to turn off a target gene in a reversible manner, e.g., similarly to RNA interference. In some instances, the CRISPR system is used to direct Cas to a promoter of a gene, thereby blocking an RNA polymerase sterically.


In some instances, a CRISPR system can be generated to edit a gene in the plant, using technology described in, e.g., U.S. Publication No. 20140068797, Cong, Science 339: 819-823, 2013; Tsai, Nature Biotechnol. 32:6 569-576, 2014; U.S. Pat. Nos. 8,871,445; 8,865,406; 8,795,965; 8,771,945; and 8,697,359.


In some instances, the CRISPR interference (CRISPRi) technique can be used for transcriptional repression of specific genes in the plant. In CRISPRi, an engineered Cas9 protein (e.g., nuclease-null dCas9, or dCas9 fusion protein, e.g., dCas9-KRAB or dCas9-SID4X fusion) can pair with a sequence specific guide RNA (sgRNA). The Cas9-gRNA complex can block RNA polymerase, thereby interfering with transcription elongation. The complex can also block transcription initiation by interfering with transcription factor binding. The CRISPRi method is specific with minimal off-target effects and is multiplexable, e.g., can simultaneously repress more than one gene (e.g., using multiple gRNAs). Also, the CRISPRi method permits reversible gene repression.


In some instances, CRISPR-mediated gene activation (CRISPRa) can be used for transcriptional activation of a gene in the plant. In the CRISPRa technique, dCas9 fusion proteins recruit transcriptional activators. For example, dCas9 can be fused to polypeptides (e.g., activation domains) such as VP64 or the p65 activation domain (p65D) and used with sgRNA (e.g., a single sgRNA or multiple sgRNAs), to activate a gene or genes in the plant. Multiple activators can be recruited by using multiple sgRNAs—this can increase activation efficiency. A variety of activation domains and single or multiple activation domains can be used. In addition to engineering dCas9 to recruit activators, sgRNAs can also be engineered to recruit activators. For example, RNA aptamers can be incorporated into a sgRNA to recruit proteins (e.g., activation domains) such as VP64. In some examples, the synergistic activation mediator (SAM) system can be used for transcriptional activation. In SAM, MS2 aptamers are added to the sgRNA. MS2 recruits the MS2 coat protein (MCP) fused to p65AD and heat shock factor 1 (HSF1).


The CRISPRi and CRISPRa techniques are described in greater detail, e.g., in Dominguez et al., Nat. Rev. Mol. Cell Biol. 17:5-15, 2016, incorporated herein by reference. In addition, dCas9-mediated epigenetic modifications and simultaneous activation and repression using CRISPR systems, as described in Dominguez et al., can be used to modulate a gene in the plant.


D. Heterologous Therapeutic Agents


The PMPs manufactured herein can include a heterologous therapeutic agent (e.g., an agent that affects an animal (e.g., human), an animal pathogen, or a pathogen vector thereof, and can be loaded into a PMP), such as a pathogen control agent (e.g., antifungal agent, an antibacterial agent, a virucidal agent, an anti-viral agent, an insecticidal agent, a nematicidal agent, an antiparasitic agent, or an insect repellent). PMPs loaded with such agents can be formulated with a pharmaceutically acceptable carrier for delivery to an animal, an animal pathogen, or a pathogen vector thereof.


i. Antibacterial Agents


The PMP compositions described herein can further include an antibacterial agent. For example, a PMP composition including an antibiotic as described herein can be administered to an animal in an amount and for a time sufficient to: reach a target level (e.g., a predetermined or threshold level) of antibiotic concentration inside or on the animal; and/or treat or prevent a bacterial infection in the animal. The antibacterials described herein may be formulated in a PMP composition for any of the methods described herein, and in certain instances, may be associated with the PMP thereof. In some instances, the PMP compositions includes two or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, or more than 10) different antibacterial agents.


As used herein, the term “antibacterial agent” refers to a material that kills or inhibits the growth, proliferation, division, reproduction, or spread of bacteria, such as phytopathogenic bacteria, and includes bactericidal (e.g., disinfectant compounds, antiseptic compounds, or antibiotics) or bacteriostatic agents (e.g., compounds or antibiotics). Bactericidal antibiotics kill bacteria, while bacteriostatic antibiotics only slow their growth or reproduction.


Bactericides can include disinfectants, antiseptics, or antibiotics. The most used disinfectants can comprise: active chlorine (i.e., hypochlorites (e.g., sodium hypochlorite), chloramines, dichloroisocyanurate and trichloroisocyanurate, wet chlorine, chlorine dioxide etc.), active oxygen (peroxides, such as peracetic acid, potassium persulfate, sodium perborate, sodium percarbonate and urea perhydrate), iodine (iodpovidone (povidone-iodine, Betadine), Lugol's solution, iodine tincture, iodinated nonionic surfactants), concentrated alcohols (mainly ethanol, 1-propanol, called also n-propanol and 2-propanol, called isopropanol and mixtures thereof; further, 2-phenoxyethanol and 1- and 2-phenoxypropanols are used), phenolic substances (such as phenol (also called carbolic acid), cresols (called Lysole in combination with liquid potassium soaps), halogenated (chlorinated, brominated) phenols, such as hexachlorophene, triclosan, trichlorophenol, tribromophenol, pentachlorophenol, Dibromol and salts thereof), cationic surfactants, such as some quaternary ammonium cations (such as benzalkonium chloride, cetyl trimethylammonium bromide or chloride, didecyldimethylammonium chloride, cetylpyridinium chloride, benzethonium chloride) and others, non-quaternary compounds, such as chlorhexidine, glucoprotamine, octenidine dihydrochloride etc.), strong oxidizers, such as ozone and permanganate solutions; heavy metals and their salts, such as colloidal silver, silver nitrate, mercury chloride, phenylmercury salts, copper sulfate, copper oxide-chloride, copper hydroxide, copper octanoate, copper oxychloride sulfate, copper sulfate, copper sulfate pentahydrate, etc. Heavy metals and their salts are the most toxic, and environment-hazardous bactericides and therefore, their use is strongly oppressed or canceled; further, also properly concentrated strong acids (phosphoric, nitric, sulfuric, amidosulfuric, toluenesulfonic acids) and alkalis (sodium, potassium, calcium hydroxides). As antiseptics (i.e., germicide agents that can be used on human or animal body, skin, mucoses, wounds and the like), few of the above mentioned disinfectants can be used, under proper conditions (mainly concentration, pH, temperature and toxicity toward man/animal). Among them, important are: properly diluted chlorine preparations (i.e., Daquin's solution, 0.5% sodium or potassium hypochlorite solution, pH-adjusted to pH 7-8, or 0.5-1% solution of sodium benzenesulfochloramide (chloramine B)), some iodine preparations, such as iodopovidone in various galenics (ointment, solutions, wound plasters), in the past also Lugol's solution, peroxides as urea perhydrate solutions and pH-buffered 0.1-0.25% peracetic acid solutions, alcohols with or without antiseptic additives, used mainly for skin antisepsis, weak organic acids such as sorbic acid, benzoic acid, lactic acid and salicylic acid some phenolic compounds, such as hexachlorophene, triclosan and Dibromol, and cation-active compounds, such as 0.05-0.5% benzalkonium, 0.5-4% chlorhexidine, 0.1-2% octenidine solutions.


The PMP composition described herein may include an antibiotic. Any antibiotic known in the art may be used. Antibiotics are commonly classified based on their mechanism of action, chemical structure, or spectrum of activity.


The antibiotic described herein may target any bacterial function or growth processes and may be either bacteriostatic (e.g., slow or prevent bacterial growth) or bactericidal (e.g., kill bacteria). In some instances, the antibiotic is a bactericidal antibiotic. In some instances, the bactericidal antibiotic is one that targets the bacterial cell wall (e.g., penicillins and cephalosporins); one that targets the cell membrane (e.g., polymyxins); or one that inhibits essential bacterial enzymes (e.g., rifamycins, lipiarmycins, quinolones, and sulfonamides). In some instances, the bactericidal antibiotic is an aminoglycoside (e.g., kasugamycin). In some instances, the antibiotic is a bacteriostatic antibiotic. In some instances the bacteriostatic antibiotic targets protein synthesis (e.g., macrolides, lincosamides, and tetracyclines). Additional classes of antibiotics that may be used herein include cyclic lipopeptides (such as daptomycin), glycylcyclines (such as tigecycline), oxazolidinones (such as linezolid), or lipiarmycins (such as fidaxomicin). Examples of antibiotics include rifampicin, ciprofloxacin, doxycycline, ampicillin, and polymyxin B. The antibiotic described herein may have any level of target specificity (e.g., narrow- or broad-spectrum). In some instances, the antibiotic is a narrow-spectrum antibiotic, and thus targets specific types of bacteria, such as gram-negative or gram-positive bacteria. Alternatively, the antibiotic may be a broad-spectrum antibiotic that targets a wide range of bacteria.


Examples of antibacterial agents suitable for the treatment of animals include Penicillins (Amoxicillin, Ampicillin, Bacampicillin, Carbenicillin, Cloxacillin, Dicloxacillin, Flucloxacillin, Mezlocillin, Nafcillin, Oxacillin, Penicillin G, Crysticillin 300 A.S., Pentids, Permapen, Pfizerpen, Pfizerpen-AS, Wycillin, Penicillin V, Piperacillin, Pivampicillin, Pivmecillinam, Ticarcillin), Cephalosporins (Cefacetrile (cephacetrile), Cefadroxil (cefadroxyl), Cefalexin (cephalexin), Cefaloglycin (cephaloglycin), Cefalonium (cephalonium), Cefaloridine (cephaloradine), Cefalotin (cephalothin), Cefapirin (cephapirin), Cefatrizine, Cefazaflur, Cefazedone, Cefazolin (cephazolin), Cefradine (cephradine), Cefroxadine, Ceftezole, Cefaclor, Cefamandole, Cefmetazole, Cefonicid, Cefotetan, Cefoxitin, Cefprozil (cefproxil), Cefuroxime, Cefuzonam, Cefcapene, Cefdaloxime, Cefdinir, Cefditoren, Cefetamet, Cefixime, Cefmenoxime, Cefodizime, Cefotaxime, Cefpimizole, Cefpodoxime, Cefteram, Ceftibuten, Ceftiofur, Ceftiolene, Ceftizoxime, Ceftriaxone, Cefoperazone, Ceftazidime, Cefclidine, Cefepime, Cefluprenam, Cefoselis, Cefozopran, Cefpirome, Cefquinome, Ceftobiprole, Ceftaroline, Cefaclomezine, Cefaloram, Cefaparole, Cefcanel, Cefedrolor, Cefempidone, Cefetrizole, Cefivitril, Cefmatilen, Cefmepidium, Cefovecin, Cefoxazole, Cefrotil, Cefsumide, Cefuracetime, Ceftioxide, Combinations, Ceftazidime/Avibactam, Ceftolozane/Tazobactam), Monobactams (Aztreonam), Carbapenems (Imipenem, Imipenem/cilastatin, Doripenem, Ertapenem, Meropenem, Meropenem/vaborbactam), Macrolide (Azithromycin, Erythromycin, Clarithromycin, Dirithromycin, Roxithromycin, Telithromycin), Lincosamides (Clindamycin, Lincomycin), Streptogramins (Pristinamycin, Quinupristin/dalfopristin), Aminoglycoside (Amikacin, Gentamicin, Kanamycin, Neomycin, Netilmicin, Paromomycin, Streptomycin, Tobramycin), Quinolone (Flumequine, Nalidixic acid, Oxolinic acid, Piromidic acid, Pipemidic acid, Rosoxacin, Second Generation, Ciprofloxacin, Enoxacin, Lomefloxacin, Nadifloxacin, Norfloxacin, Ofloxacin, Pefloxacin, Rufloxacin, Balofloxacin, Gatifloxacin, Grepafloxacin, Levofloxacin, Moxifloxacin, Pazufloxacin, Sparfloxacin, Temafloxacin, Tosufloxacin, Besifloxacin, Delafloxacin, Clinafloxacin, Gemifloxacin, Prulifloxacin, Sitafloxacin, Trovafloxacin), Sulfonamides (Sulfamethizole, Sulfamethoxazole, Sulfisoxazole, Trimethoprim-Sulfamethoxazole), Tetracycline (Demeclocycline, Doxycycline, Minocycline, Oxytetracycline, Tetracycline, Tigecycline), Other (Lipopeptides, Fluoroquinolone, Lipoglycopeptides, Cephalosporin, Macrocyclics, Chloramphenicol, Metronidazole, Tinidazole, Nitrofurantoin, Glycopeptides, Vancomycin, Teicoplanin, Lipoglycopeptides, Telavancin, Oxazolidinones, Linezolid, Cycloserine 2, Rifamycins, Rifampin, Rifabutin, Rifapentine, Rifalazil, Polypeptides, Bacitracin, Polymyxin B, Tuberactinomycins, Viomycin, Capreomycin).


One skilled in the art will appreciate that a suitable concentration of each antibiotic in the composition depends on factors such as efficacy, stability of the antibiotic, number of distinct antibiotics, the formulation, and methods of application of the composition.


ii. Antifungal Agents


The PMP compositions described herein can further include an antifungal agent. For example, a PMP composition including an antifungal as described herein can be administered to an animal in an amount and for a time sufficient to reach a target level (e.g., a predetermined or threshold level) of antifungal concentration inside or on the animal; and/or treat or prevent a fungal infection in the animal. The antifungals described herein may be formulated in a PMP composition for any of the methods described herein, and in certain instances, may be associated with the PMP thereof. In some instances, the PMP compositions includes two or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, or more than 10) different antifungal agents.


As used herein, the term “fungicide” or “antifungal agent” refers to a substance that kills or inhibits the growth, proliferation, division, reproduction, or spread of fungi, such as fungi that are pathogenic to animals. Many different types of antifungal agent have been produced commercially. Non limiting examples of antifungal agents include: Allylamines (Amorolfin, Butenafine, Naftifine, Terbinafine), Imidazoles ((Bifonazole, Butoconazole, Clotrimazole, Econazole, Fenticonazole, Ketoconazole, Isoconazole, Luliconazole, Miconazole, Omoconazole, Oxiconazole, Sertaconazole, Sulconazole, Tioconazole, Terconazole); Triazoles (Albaconazole, Efinaconazole, Fluconazole, Isavuconazole, Itraconazole, Posaconazole, Ravuconazole, Terconazole, Voriconazole), Thiazoles (Abafungin), Polyenes (Amphotericin B, Nystatin, Natamycin, Trichomycin), Echinocandins (Anidulafungin, Caspofungin, Micafungin), Other (Tolnaftate, Flucytosine, Butenafine, Griseofulvin, Ciclopirox, Selenium sulfide, Tavaborole). One skilled in the art will appreciate that a suitable concentration of each antifungal in the composition depends on factors such as efficacy, stability of the antifungal, number of distinct antifungals, the formulation, and methods of application of the composition.


iii. Insecticides


The PMP compositions described herein can further include an insecticide. For example, the insecticide can decrease the fitness of (e.g., decrease growth or kill) an insect vector of an animal pathogen. A PMP composition including an insecticide as described herein can be contacted with an insect, in an amount and for a time sufficient to: (a) reach a target level (e.g., a predetermined or threshold level) of insecticide concentration inside or on the insect; and (b) decrease fitness of the insect. In some instances, the insecticide can decrease the fitness of (e.g., decrease growth or kill) a parasitic insect. A PMP composition including an insecticide as described herein can be contacted with a parasitic insect, or an animal infected therewith, in an amount and for a time sufficient to: (a) reach a target level (e.g., a predetermined or threshold level) of insecticide concentration inside or on the parasitic insect; and (b) decrease the fitness of the parasitic insect. The insecticides described herein may be formulated in a PMP composition for any of the methods described herein, and in certain instances, may be associated with the PMP thereof. In some instances, the PMP compositions include two or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, or more than 10) different insecticide agents.


As used herein, the term “insecticide” or “insecticidal agent” refers to a substance that kills or inhibits the growth, proliferation, reproduction, or spread of insects, such as insect vectors of animal pathogens or parasitic insects. Non limiting examples of insecticides are shown in Table 4. Additional non-limiting examples of suitable insecticides include biologics, hormones or pheromones such as azadirachtin, Bacillus species, Beauveria species, codlemone, Metarrhizium species, Paecilomyces species, thuringiensis, and Verticillium species, and active compounds having unknown or non-specified mechanisms of action such as fumigants (such as aluminium phosphide, methyl bromide and sulphuryl fluoride) and selective feeding inhibitors (such as cryolite, flonicamid and pymetrozine). One skilled in the art will appreciate that a suitable concentration of each insecticide in the composition depends on factors such as efficacy, stability of the insecticide, number of distinct insecticides, the formulation, and methods of application of the composition.









TABLE 4







Examples of insecticides








Class
Compounds





chloronicotinyls/
acetamiprid, clothianidin, dinotefuran, imidacloprid, nitenpyram,


neonicotinoids
nithiazine, thiacloprid, thiamethoxam, imidaclothiz, (2E)-1-[(2-



chloro-1,3-thiazol-5-yl)methyl]-3,5-dimethyl-N-nitro-1,3,5-tri-azinan-



2-imine, acetylcholinesterase (AChE) inhibitors (such as



carbamates and organophosphates)


carbamates
alanycarb, aldicarb, aldoxycarb, allyxycarb, aminocarb, bendiocarb,



benfuracarb, bufencarb, butacarb, butocarboxim, butoxycarboxim,



carbaryl, carbofuran, carbosulfan, chloethocarb, dimetilan,



ethiofencarb, fenobucarb, fenothiocarb, formetanate, furathiocarb,



isoprocarb, metam-sodium, methiocarb, methomyl, metolcarb,



oxamyl, phosphocarb, pirimicarb, promecarb, propoxur, thiodicarb,



thiofanox, triazamate, trimethacarb, XMC, xylylcarb


organophosphates
acephate, azamethiphos, azinphos (-methyl, -ethyl), bromophos-



ethyl, bromfenvinfos (-methyl), butathiofos, cadusafos,



carbophenothion, chlorethoxyfos, chlorfenvinphos, chlormephos,



chlorpyrifos (-methyl/-ethyl), coumaphos, cyanofenphos,



cyanophos, demeton-S-methyl, demeton-S-methylsulphon, dialifos,



diazinon, dichlofenthion, dichlorvos/DDVP, dicrotophos,



dimethoate, dimethylvinphos, dioxabenzofos, disulfoton, EPN,



ethion, ethoprophos, etrimfos, famphur, fenamiphos, fenitrothion,



fensulfothion, fenthion, flupyrazofos, fonofos, formothion,



fosmethilan, fosthiazate, heptenophos, iodofenphos, iprobenfos,



isazofos, isofenphos, isopropyl O-salicylate, isoxathion, malathion,



mecarbam, methacrifos, methamidophos, methidathion,



mevinphos, monocrotophos, naled, omethoate, oxydemeton-



methyl, parathion (-methyl/-ethyl), phenthoate, phorate, phosalone,



phosmet, phosphamidon, phosphocarb, phoxim, pirimiphos



(-methyl/-ethyl), profenofos, propaphos, propetamphos, prothiofos,



prothoate, pyraclofos, pyridaphenthion, pyridathion, quinalphos,



sebufos, sulfotep, sulprofos, tebupirimfos, temephos, terbufos,



tetrachlorvinphos, thiometon, triazophos, triclorfon, vamidothion


pyrethroids
acrinathrin, allethrin (d-cis-trans, d-trans), cypermethrin (alpha-,



beta-, theta-, zeta-), permethrin (cis-, trans-), beta-cyfluthrin,



bifenthrin, bioallethrin, bioallethrin-S-cyclopentyl-isomer,



bioethanomethrin, biopermethrin, bioresmethrin, chlovaporthrin,



cis-cypermethrin, cis-resmethrin, cis-permethrin, clocythrin,



cycloprothrin, cyfluthrin, cyhalothrin, cyphenothrin, DDT,



deltamethrin, empenthrin (1R-isomer), esfenvalerate, etofenprox,



fenfluthrin, fenpropathrin, fenpyrithrin, fenvalerate, flubrocythrinate,



flucythrinate, flufenprox, flumethrin, fluvalinate, fubfenprox,



gamma-cyhalothrin, imiprothrin, kadethrin, lambda, cyhalothrin,



metofluthrin, phenothrin (1R-trans isomer), prallethrin, profluthrin,



protrifenbute, pyresmethrin, resmethrin, RU 15525, silafluofen, tau-



fluvalinate, tefluthrin, terallethrin, tetramethrin (1R-isomer),



tralocythrin, tralomethrin, transfluthrin, ZXI 8901, pyrethrins



(pyrethrum)


oxadiazines
indoxacarb, acetylcholine receptor modulators (such as spinosyns)


spinosyns
spinosad


cyclodiene
camphechlor, chlordane, endosulfan, gamma-HCH, HCH,



heptachlor,


organochlorines
lindane, methoxychlor


fiproles
acetoprole, ethiprole, vaniliprole, fipronil


mectins
abamectin, avermectin, emamectin, emamectin-benzoate,



fenoxycarb, hydroprene, kinoprene, methoprene, ivermectin,



lepimectin, epofenonane, pyriproxifen, milbemectin, milbemycin,



triprene


diacylhydrazines
chromafenozide, halofenozide, methoxyfenozide, tebufenozide


benzoylureas
bistrifluoron, chlorfluazuron, diflubenzuron, fluazuron, flucycloxuron,



flufenoxuron, hexaflumuron, lufenuron, novaluron, noviflumuron,



penfluoron, teflubenzuron, triflumuron


organotins
azocyclotin, cyhexatin, fenbutatin oxide


pyrroles
chlorfenapyr


dinitrophenols
binapacyrl, dinobuton, dinocap, DNOC


METIs
fenazaquin, fenpyroximate, pyrimidifen, pyridaben, tebufenpyrad,



tolfenpyrad, rotenone, acequinocyl, fluacrypyrim, microbial



disrupters of the intestinal membrane of insects (such as Bacillus




thuringiensis strains), inhibitors of lipid synthesis (such as tetronic




acids and tetramic acids)


tetronic acids
spirodiclofen, spiromesifen, spirotetramat


tetramic acids
cis-3-(2,5-dimethylphenyl)-8-methoxy-2-oxo-1-azaspiro[4.5]dec-3-



en-4-yl ethyl carbonate (alias: carbonic acid, 3-(2,5-



dimethylphenyl)-8-methoxy-2-oxo-1-azaspiro[4.5]dec-3-en-4-yl



ethyl ester; CAS Reg. No.: 382608-10-8), carboxamides (such as



flonicamid), octopaminergic agonists (such as amitraz), inhibitors of



the magnesium-stimulated ATPase (such as propargite), ryanodin



receptor agonists (such as phthalamides or rynaxapyr)


phthalamides
N2-[1,1-dimethyl-2-(methylsulphonyl)ethyl]-3-iodo-N1-[2-methyl--4-



[1,2,2,2-tetrafluoro-1-(trifluoromethyl)ethyl]phenyl]-1,2-benzenedi-



carboxamide (i.e., flubendiamide; CAS reg. No.: 272451-65-7)









iv. Nematicides


The PMP compositions described herein can further include a nematicide. In some instances, the PMP composition includes two or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, or more than 10) different nematicides. For example, the nematicide can decrease the fitness of (e.g., decrease growth or kill) a parasitic nematode. A PMP composition including a nematicide as described herein can be contacted with a parasitic nematode, or an animal infected therewith, in an amount and for a time sufficient to: (a) reach a target level (e.g., a predetermined or threshold level) of nematicide concentration inside or on the target nematode; and (b) decrease fitness of the parasitic nematode. The nematicides described herein may be formulated in a PMP composition for any of the methods described herein, and in certain instances, may be associated with the PMP thereof.


As used herein, the term “nematicide” or “nematicidal agent” refers to a substance that kills or inhibits the growth, proliferation, reproduction, or spread of nematodes, such as a parasitic nematode. Non limiting examples of nematicides are shown in Table 5. One skilled in the art will appreciate that a suitable concentration of each nematicide in the composition depends on factors such as efficacy, stability of the nematicide, number of distinct nematicides, the formulation, and methods of application of the composition.









TABLE 5





Examples of Nematicides
















FUMIGANTS
D-D, 1,3-Dichloropropene, Ethylene Dibromide, 1,2-Dibromo-3-



Chloropropane, Methyl Bromide, Chloropicrin, Metam Sodium, Dazomet,



Methyl Isothiocyanate (MITC), Sodium Tetrathiocarbonate, Chloropicrin,


CARBAMATES
Aldicarb, Aldoxycarb, Carbofuran, Oxamyl, Cleothocarb


ORGANOPHOSPHATES
Ethoprophos, Fenamiphos, Cadusafos, Fosthiazate, Fensulfothion,



Thionazin, Isazofos,


BIOCHEMICALS
DITERA ®, CLANDOSAN ®, SINCOCIN ®









v. Antiparasitic Agent


The PMP compositions described herein can further include an antiparasitic agent. For example, the antiparasitic can decrease the fitness of (e.g., decrease growth or kill) a parasitic protozoan. A PMP composition including an antiparasitic as described herein can be contacted with a protozoan in an amount and for a time sufficient to: (a) reach a target level (e.g., a predetermined or threshold level) of antiparasitic concentration inside or on the protozoan, or animal infected therewith; and (b) decrease fitness of the protozoan. This can be useful in the treatment or prevention of parasites in animals. For example, a PMP composition including an antiparasitic agent as described herein can be administered to an animal in an amount and for a time sufficient to: reach a target level (e.g., a predetermined or threshold level) of antiparasitic concentration inside or on the animal; and/or treat or prevent a parasite (e.g., parasitic nematode, parasitic insect, or protozoan) infection in the animal. The antiparasitic described herein may be formulated in a PMP composition for any of the methods described herein, and in certain instances, may be associated with the PMP thereof. In some instances, the PMP composition includes two or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, or more than 10) different antiparasitic agents.


As used herein, the term “antiparasitic” or “antiparasitic agent” refers to a substance that kills or inhibits the growth, proliferation, reproduction, or spread of parasites, such as parasitic protozoa, parasitic nematodes, or parasitic insects. Examples of antiparasitic agents include Antihelmintics (Bephenium, Diethylcarbamazine, Ivermectin, Niclosamide, Piperazine, Praziquantel, Pyrantel, Pyrvinium, Benzimidazoles, Albendazole, Flubendazole, Mebendazole, Thiabendazole, Levamisole, Nitazoxanide, Monopantel, Emodepside, Spiroindoles), Scabicides (Benzyl benzoate, Benzyl benzoate/disulfiram, Lindane, Malathion, Permethrin), Pediculicides (Piperonyl butoxide/pyrethrins, Spinosad, Moxidectin), Scabicides (Crotamiton), Anticestodes (Niclosamide, Pranziquantel, Albendazole), Antiamoebics (Rifampin, Apmphotericin B); or Antiprotozoals (Melarsoprol, Eflornithine, Metronidazole, Tinidazole, Miltefosine, Artemisinin). In certain instances, the antiparasitic agent may be use for treating or prevening infections in livestock animals, e.g., Levamisole, Fenbendazole, Oxfendazole, Albendazole, Moxidectin, Eprinomectin, Doramectin, Ivermectin, or Clorsulon. One skilled in the art will appreciate that a suitable concentration of each antiparasitic in the composition depends on factors such as efficacy, stability of the antiparasitic, number of distinct antiparasitics, the formulation, and methods of application of the composition.


vi. Antiviral Agent


The PMP compositions described herein can further include an antiviral agent. A PMP composition including an antivirual agent as described herein can be administered to an animal in an amount and for a time sufficient to reach a target level (e.g., a predetermined or threshold level) of antiviral concentration inside or on the animal; and/or to treat or prevent a viral infection in the animal. The antivirals described herein may be formulated in a PMP composition for any of the methods described herein, and in certain instances, may be associated with the PMP thereof. In some instances, the PMP composition includes two or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, or more than 10) different antivirals.


As used herein, the term “antiviral” or “virucide” refers to a substance that kills or inhibits the growth, proliferation, reproduction, development, or spread of viruses, such as viral pathogens that infect animals. A number of agents can be employed as an antiviral, including chemicals or biological agents (e.g., nucleic acids, e.g., dsRNA). Examples of antiviral agents useful herein include Abacavir, Acyclovir (Aciclovir), Adefovir, Amantadine, Amprenavir (Agenerase), Ampligen, Arbidol, Atazanavir, Atripla, Balavir, Cidofovir, Combivir, Dolutegravir, Darunavir, Delavirdine, Didanosine, Docosanol, Edoxudine, Efavirenz, Emtricitabine, Enfuvirtide, Entecavir, Ecoliever, Famciclovir, Fomivirsen, Fosamprenavir, Foscarnet, Fosfonet, Fusion inhibitor, Ganciclovir, Ibacitabine, Imunovir, Idoxuridine, Imiquimod, Indinavir, Inosine, Integrase inhibitor, Interferon type III, Interferon type II, Interferon type I, Interferon, Lamivudine, Lopinavir, Loviride, Maraviroc, Moroxydine, Methisazone, Nelfinavir, Nevirapine, Nexavir, Nitazoxanide, Nucleoside analogues, Norvir, Oseltamivir (Tamiflu), Peginterferon alfa-2a, Penciclovir, Peramivir, Pleconaril, Podophyllotoxin, Raltegravir, Ribavirin, Rimantadine, Ritonavir, Pyramidine, Saquinavir, Sofosbuvir, Stavudine, Synergistic enhancer (antiretroviral), Telaprevir, Tenofovir, Tenofovir disoproxil, Tipranavir, Trifluridine, Trizivir, Tromantadine, Truvada, Valaciclovir (Valtrex), Valganciclovir, Vicriviroc, Vidarabine, Viramidine, Zalcitabine, Zanamivir (Relenza), or Zidovudine. One skilled in the art will appreciate that a suitable concentration of each antiviral in the composition depends on factors such as efficacy, stability of the antivirals, number of distinct antivirals, the formulation, and methods of application of the composition.


vii. Repellents


The PMP compositions described herein can further include a repellent. For example, the repellent can repel a vector of animal pathogens, such as insects. The repellent described herein may be formulated in a PMP composition for any of the methods described herein, and in certain instances, may be associated with the PMP thereof. In some instances, the PMP composition includes two or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, or more than 10) different repellents.


For example, a PMP composition including a repellent as described herein can be contacted with an insect vector or a habitat of the vector in an amount and for a time sufficient to: (a) reach a target level (e.g., a predetermined or threshold level) of repellent concentration; and/or (b) decrease the levels of the insect near or on nearby animals relative to a control. Alternatively, a PMP composition including a repellent as described herein can be contacted with an animal in an amount and for a time sufficient to: (a) reach a target level (e.g., a predetermined or threshold level) of repellent concentration; and/or (b) decrease the levels of the insect near or on the animal relative to an untreated animal.


Some examples of well-known insect repellents include: benzil; benzyl benzoate; 2,3,4,5-bis(butyl-2-ene)tetrahydrofurfural (MGK Repellent 11); butoxypolypropylene glycol; N-butylacetanilide; normal-butyl-6,6-dimethyl-5,6-dihydro-1,4-pyrone-2-carboxylate (Indalone); dibutyl adipate; dibutyl phthalate; di-normal-butyl succinate (Tabatrex); N,N-diethyl-meta-toluamide (DEET); dimethyl carbate (endo,endo)-dimethyl bicyclo[2.2.1] hept-5-ene-2,3-dicarboxylate); dimethyl phthalate; 2-ethyl-2-butyl-1,3-propanediol; 2-ethyl-1,3-hexanediol (Rutgers 612); di-normal-propyl isocinchomeronate (MGK Repellent 326); 2-phenylcyclohexanol; p-methane-3,8-diol, and normal-propyl N,N-diethylsuccinamate. Other repellents include citronella oil, dimethyl phthalate, normal-butylmesityl oxide oxalate and 2-ethyl hexanediol-1,3 (See, Kirk-Othmer Encyclopedia of Chemical Technology, 2nd Ed., Vol. 11: 724-728; and The Condensed Chemical Dictionary, 8th Ed., p 756).


In some instances, the repellent is an insect repellent, including synthetic or nonsynthetic insect repellents. Examples of synthetic insect repellents include methyl anthranilate and other anthranilate-based insect repellents, benzaldehyde, DEET (N,N-diethyl-m-toluamide), dimethyl carbate, dimethyl phthalate, icaridin (i.e., picaridin, Bayrepel, and KBR 3023), indalone (e.g., as used in a “6-2-2” mixture (60% Dimethyl phthalate, 20% Indalone, 20% Ethylhexanediol), IR3535 (3-[N-Butyl-N-acetyl]-aminopropionic acid, ethyl ester), metofluthrin, permethrin, SS220, or tricyclodecenyl allyl ether. Examples of natural insect repellents include beautyberry (Callicarpa) leaves, birch tree bark, bog myrtle (Myrica Gale), catnip oil (e.g., nepetalactone), citronella oil, essential oil of the lemon eucalyptus (Corymbia citriodora; e.g., p-menthane-3,8-diol (PMD)), neem oil, lemongrass, tea tree oil from the leaves of Melaleuca alternifolia, tobacco, or extracts thereof.


viii. Other Therapeutic Agents


In some examples, the therapeutic agent is an agent used for the prevention or treatment of a mammalian (for example, human) condition or a disease. The disease may be, e.g., a cancer, an autoimmune condition, or a metabolic disorder.


In some examples, the therapeutic agent is a small molecule or a nucleic acid (e.g., a siRNA, a miRNA, or an mRNA).


In some examples, the therapeutic agent is a protein or peptide therapeutic with enzymatic activity, regulatory activity, or targeting activity, e.g., a protein or peptide with activity that affects one or more of endocrine and growth regulation, metabolic enzyme deficiencies, hematopoiesis, hemostasis and thrombosis; gastrointestinal-tract disorders; pulmonary disorders; immunodeficiencies and/or immunoregulation; fertility; aging (e.g., anti-aging activity); autophagy regulation; epigenetic regulation; oncology; or infectious diseases (e.g., anti-microbial peptides, anti-fungals, or anti-virals).


In some examples, the therapeutic agent is an antibody (e.g., a monoclonal antibody, e.g., a monospecific, bispecific, or multispecific monoclonal antibody) or an antigen-binding fragment thereof (e.g., an scFv, (scFv)2, Fab, Fab′, and F(ab′)2, F(ab1)2, Fv, dAb, and Fd fragment, or a diabody), a nanobody, a conjugated antibody, or an antibody-related polypeptide.


In some examples, the therapeutic agent is an antimicrobial, antibacterial, antifungal, antinematicidal, antiparasitic, or antiviral polypeptide.


In some examples, the therapeutic agent is an allergenic, an allergen, or an antigen.


In some examples, the therapeutic agent is a vaccine (e.g., a conjugate vaccine, an inactivated vaccine, or a live attenuated vaccine), In some examples, the therapeutic agent is an enzyme, e.g., a metabolic recombinase, a helicase, an integrase, a RNAse, a DNAse, an ubiquitination protein. In some examples, the enzyme is a recombinant enzyme.


In some examples, the therapeutic agent is a gene editing protein, e.g., a component of a CRISPR-Cas system, TALEN, or zinc finger.


In some examples, the therapeutic agent is any one of a cytokine, a hormone, a signaling ligand, a transcription factor, a receptor, a receptor antagonist, a receptor agonist, a blocking or neutralizing polypeptide, a riboprotein, or a chaperone.


In some examples, the therapeutic agent is a pore-forming protein, a cell-penetrating peptide, a cell-penetrating peptide inhibitor, or a proteolysis targeting chimera (PROTAC).


In some examples, the therapeutic agent is any one of an aptamer, a blood derivative, a cell therapy, or an immunotherapy (e.g., a cellular immunotherapy.


In some aspects, the therapeutic agent is a protein vaccine, e.g., a vaccine for use in protecting against a deleterious foreign agent, treating an autoimmune disease, or treating cancer.


III. Methods of Use

The PMPs manufactured herein are useful in a variety of agricultural or therapeutic methods. Examples of methods of using PMPs are described further below.


A. Delivery to a Plant


Provided herein are methods of delivering a PMP composition (e.g., manufactured in accordance with the methods or bioreactors herein) to a plant, e.g., by contacting the plant, or part thereof, with the PMP composition. In some instances, plants may be treated with unloaded PMPs. In other instances, the PMPs include a heterologous functional agent, e.g., pesticidal agents (e.g., antibacterial agents, antifungal agents, nematicides, molluscicides, virucides, herbicides), pest control agents (e.g., repellents), fertilizing agents, or plant-modifying agents. PMPs intended for delivery to a plant may be formulated with an agriculturally acceptable carrier, e.g., formulated for delivery to a plant.


In one aspect, provided herein is a method of increasing the fitness of a plant, the method including delivering to the plant the PMP composition described herein (e.g., in an effective amount and duration) to increase the fitness of the plant relative to an untreated plant (e.g., a plant that has not been delivered the PMP composition).


An increase in the fitness of the plant as a consequence of delivery of a PMP composition can manifest in a number of ways, e.g., thereby resulting in a better production of the plant, for example, an improved yield, improved vigor of the plant or quality of the harvested product from the plant. An improved yield of a plant relates to an increase in the yield of a product (e.g., as measured by plant biomass, grain, seed or fruit yield, protein content, carbohydrate or oil content or leaf area) of the plant by a measurable amount over the yield of the same product of the plant produced under the same conditions, but without the application of the instant compositions or compared with application of conventional agricultural agents. For example, yield can be increased by at least about 0.5%, about 1%, about 2%, about 3%, about 4%, about 5%, about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 100%, or more than 100%. Yield can be expressed in terms of an amount by weight or volume of the plant or a product of the plant on some basis. The basis can be expressed in terms of time, growing area, weight of plants produced, or amount of a raw material used. For example, such methods may increase the yield of plant tissues including, but not limited to: seeds, fruits, kernels, bolls, tubers, roots, and leaves.


An increase in the fitness of a plant as a consequence of delivery of a PMP composition can also be measured by other methods, such as an increase or improvement of the vigor rating, the stand (the number of plants per unit of area), plant height, stalk circumference, stalk length, leaf number, leaf size, plant canopy, visual appearance (such as greener leaf color), root rating, emergence, protein content, increased tillering, bigger leafs, more leaves, less dead basal leaves, stronger tillers, less fertilizer needed, less seeds needed, more productive tillers, earlier flowering, early grain or seed maturity, less plant verse (lodging), increased shoot growth, earlier germination, or any combination of these factors, by a measurable or noticeable amount over the same factor of the plant produced under the same conditions, but without the administration of the instant compositions or with application of conventional agricultural agents.


Provided herein is a method of modifying or increasing the fitness of a plant, the method including delivering to the plant an effective amount of a PMP composition provided herein, wherein the method modifies the plant and thereby introduces or increases a beneficial trait in the plant (e.g., by about 1%, 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or more than 100%) relative to an untreated plant. In particular, the method may increase the fitness of the plant (e.g., by about 1%, 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or more than 100%) relative to an untreated plant.


In some instances, the increase in plant fitness is an increase (e.g., by about 1%, 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or more than 100%) in disease resistance, drought tolerance, heat tolerance, cold tolerance, salt tolerance, metal tolerance, herbicide tolerance, chemical tolerance, water use efficiency, nitrogen utilization, resistance to nitrogen stress, nitrogen fixation, pest resistance, herbivore resistance, pathogen resistance, yield, yield underwater-limited conditions, vigor, growth, photosynthetic capability, nutrition, protein content, carbohydrate content, oil content, biomass, shoot length, root length, root architecture, seed weight, or amount of harvestable produce.


In some instances, the increase in fitness is an increase (e.g., by about 1%, 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or more than 100%) in development, growth, yield, resistance to abiotic stressors, or resistance to biotic stressors. An abiotic stress refers to an environmental stress condition that a plant or a plant part is subjected to that includes, e.g., drought stress, salt stress, heat stress, cold stress, and low nutrient stress. A biotic stress refers to an environmental stress condition that a plant or plant part is subjected to that includes, e.g. nematode stress, insect herbivory stress, fungal pathogen stress, bacterial pathogen stress, or viral pathogen stress. The stress may be temporary, e.g. several hours, several days, several months, or permanent, e.g. for the life of the plant.


In some instances, the increase in plant fitness is an increase (e.g., by about 1%, 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or more than 100%) in quality of products harvested from the plant. For example, the increase in plant fitness may be an improvement in commercially favorable features (e.g., taste or appearance) of a product harvested from the plant. In other instances, the increase in plant fitness is an increase in shelf-life of a product harvested from the plant (e.g., by about 1%, 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or more than 100%).


Alternatively, the increase in fitness may be an alteration of a trait that is beneficial to human or animal health, such as a reduction in allergen production. For example, the increase in fitness may be a decrease (e.g., by about 1%, 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or more than 100%) in production of an allergen (e.g., pollen) that stimulates an immune response in an animal (e.g., human).


The modification of the plant (e.g., increase in fitness) may arise from modification of one or more plant parts. For example, the plant can be modified by contacting leaf, seed, pollen, root, fruit, shoot, flower, cells, protoplasts, or tissue (e.g., meristematic tissue) of the plant. As such, in another aspect, provided herein is a method of increasing the fitness of a plant, the method including contacting pollen of the plant with an effective amount of a PMP composition herein, wherein the method increases the fitness of the plant (e.g., by about 1%, 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or more than 100%) relative to an untreated plant.


In yet another aspect, provided herein is a method of increasing the fitness of a plant, the method including contacting a seed of the plant with an effective amount of a PMP composition disclosed herein, wherein the method increases the fitness of the plant (e.g., by about 1%, 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or more than 100%) relative to an untreated plant.


In another aspect, provided herein is a method including contacting a protoplast of the plant with an effective amount of a PMP composition herein, wherein the method increases the fitness of the plant (e.g., by about 1%, 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or more than 100%) relative to an untreated plant.


In a further aspect, provided herein is a method of increasing the fitness of a plant, the method including contacting a plant cell of the plant with an effective amount of a PMP composition herein, wherein the method increases the fitness of the plant (e.g., by about 1%, 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or more than 100%) relative to an untreated plant.


In another aspect, provided herein is a method of increasing the fitness of a plant, the method including contacting meristematic tissue of the plant with an effective amount of a PMP composition herein, wherein the method increases the fitness of the plant (e.g., by about 1%, 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or more than 100%) relative to an untreated plant.


In another aspect, provided herein is a method of increasing the fitness of a plant, the method including contacting an embryo of the plant with an effective amount of a PMP composition herein, wherein the method increases the fitness of the plant (e.g., by about 1%, 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or more than 100%) relative to an untreated plant.


In cases where an herbicide is included in the PMP, or compositions thereof, the methods may be further used to decrease the fitness of or kill weeds. In such instances, the method may be effective to decrease the fitness of the weed by about 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or more in comparison to an untreated weed (e.g., a weed to which the PMP composition has not been administered). For example, the method may be effective to kill the weed, thereby decreasing a population of the weed by about 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or more in comparison to an untreated weed. In some instances, the method substantially eliminates the weed. Examples of weeds that can be treated in accordance with the present methods are further described herein.


i. Plants


A variety of plants can be delivered or treated with a PMP composition described herein. Plants that can be delivered a PMP composition (i.e., “treated”) in accordance with the present methods include whole plants and parts thereof, including, but not limited to, shoot vegetative organs/structures (e.g., leaves, stems and tubers), roots, flowers and floral organs/structures (e.g., bracts, sepals, petals, stamens, carpels, anthers and ovules), seed (including embryo, endosperm, cotyledons, and seed coat) and fruit (the mature ovary), plant tissue (e.g., vascular tissue, ground tissue, and the like) and cells (e.g., guard cells, egg cells, and the like), and progeny of same. Plant parts can further refer parts of the plant such as the shoot, root, stem, seeds, stipules, leaves, petals, flowers, ovules, bracts, branches, petioles, internodes, bark, pubescence, tillers, rhizomes, fronds, blades, pollen, stamen, and the like.


The class of plants that can be treated in a method disclosed herein includes the class of higher and lower plants, including angiosperms (monocotyledonous and dicotyledonous plants), gymnosperms, ferns, horsetails, psilophytes, lycophytes, bryophytes, and algae (e.g., multicellular or unicellular algae). Plants that can be treated in accordance with the present methods further include any vascular plant, for example monocotyledons or dicotyledons or gymnosperms, including, but not limited to alfalfa, apple, Arabidopsis, banana, barley, canola, castor bean, chrysanthemum, clover, cocoa, coffee, cotton, cottonseed, corn, crambe, cranberry, cucumber, dendrobium, dioscorea, eucalyptus, fescue, flax, gladiolus, liliacea, linseed, millet, muskmelon, mustard, oat, oil palm, oilseed rape, papaya, peanut, pineapple, ornamental plants, Phaseolus, potato, rapeseed, rice, rye, ryegrass, safflower, sesame, sorghum, soybean, sugarbeet, sugarcane, sunflower, strawberry, tobacco, tomato, turfgrass, wheat and vegetable crops such as lettuce, celery, broccoli, cauliflower, cucurbits; fruit and nut trees, such as apple, pear, peach, orange, grapefruit, lemon, lime, almond, pecan, walnut, hazel; vines, such as grapes (e.g., a vineyard), kiwi, hops; fruit shrubs and brambles, such as raspberry, blackberry, gooseberry; forest trees, such as ash, pine, fir, maple, oak, chestnut, popular; with alfalfa, canola, castor bean, corn, cotton, crambe, flax, linseed, mustard, oil palm, oilseed rape, peanut, potato, rice, safflower, sesame, soybean, sugarbeet, sunflower, tobacco, tomato, and wheat. Plants that can be treated in accordance with the methods of the present invention include any crop plant, for example, forage crop, oilseed crop, grain crop, fruit crop, vegetable crop, fiber crop, spice crop, nut crop, turf crop, sugar crop, beverage crop, and forest crop. In certain instances, the crop plant that is treated in the method is a soybean plant. In other certain instances, the crop plant is wheat. In certain instances, the crop plant is corn. In certain instances, the crop plant is cotton. In certain instances, the crop plant is alfalfa. In certain instances, the crop plant is sugarbeet. In certain instances, the crop plant is rice. In certain instances, the crop plant is potato. In certain instances, the crop plant is tomato.


In certain instances, the plant is a crop. Examples of such crop plants include, but are not limited to, monocotyledonous and dicotyledonous plants including, but not limited to, fodder or forage legumes, ornamental plants, food crops, trees, or shrubs selected from Acer spp., Allium spp., Amaranthus spp., Ananas comosus, Apium graveolens, Arachis spp, Asparagus officinalis, Beta vulgaris, Brassica spp. (e.g., Brassica napus, Brassica rapa ssp. (canola, oilseed rape, turnip rape), Camellia sinensis, Canna indica, Cannabis saliva, Capsicum spp., Castanea spp., Cichorium endivia, Citrullus lanatus, Citrus spp., Cocos spp., Coffea spp., Coriandrum sativum, Corylus spp., Crataegus spp., Cucurbita spp., Cucumis spp., Daucus carota, Fagus spp., Ficus carica, Fragaria spp., Ginkgo biloba, Glycine spp. (e.g., Glycine max, Soja hispida or Soja max), Gossypium hirsutum, Helianthus spp. (e.g., Helianthus annuus), Hibiscus spp., Hordeum spp. (e.g., Hordeum vulgare), Ipomoea batatas, Juglans spp., Lactuca sativa, Linum usitatissimum, Litchi chinensis, Lotus spp., Luffa acutangula, Lupinus spp., Lycopersicon spp. (e.g., Lycopersicon esculenturn, Lycopersicon lycopersicum, Lycopersicon pyriforme), Malus spp., Medicago sativa, Mentha spp., Miscanthus sinensis, Morus nigra, Musa spp., Nicotiana spp., Olea spp., Oryza spp. (e.g., Oryza sativa, Oryza latifolia), Panicum miliaceum, Panicum virgatum, Passiflora edulis, Petroselinum crispum, Phaseolus spp., Pinus spp., Pistacia vera, Pisum spp., Poa spp., Populus spp., Prunus spp., Pyrus communis, Quercus spp., Raphanus sativus, Rheum rhabarbarum, Ribes spp., Ricinus communis, Rubus spp., Saccharum spp., Salix sp., Sambucus spp., Secale cereale, Sesamum spp., Sinapis spp., Solanum spp. (e.g., Solanum tuberosum, Solanum integrifolium or Solanum lycopersicum), Sorghum bicolor, Sorghum halepense, Spinacia spp., Tamarindus indica, Theobroma cacao, Trifolium spp., Triticosecale rimpaui, Triticum spp. (e.g., Triticum aestivum, Triticum durum, Triticum turgidum, Triticum hybernum, Triticum macha, Triticum sativum or Triticum vulgare), Vaccinium spp., Vicia spp., Vigna spp., Viola odorata, Vitis spp., and Zea mays. In certain embodiments, the crop plant is rice, oilseed rape, canola, soybean, corn (maize), cotton, sugarcane, alfalfa, sorghum, or wheat.


In certain instance, the compositions and methods can be used to treat post-harvest plants or plant parts, food, or feed products. In some instances, the food or feed product is a non-plant food or feed product (e.g., a product edible for humans, veterinary animals, or livestock (e.g., mushrooms)).


The plant or plant part for use in the present invention include plants of any stage of plant development. In certain instances, the delivery can occur during the stages of germination, seedling growth, vegetative growth, and reproductive growth. In certain instances, delivery to the plant occurs during vegetative and reproductive growth stages. Alternatively, the delivery can occur to a seed. The stages of vegetative and reproductive growth are also referred to herein as “adult” or “mature” plants.


ii. Weeds


In cases where an herbicide is included in the PMP, or compositions thereof, the methods may be further used to decrease the fitness of or kill weeds. In such instances, the method may be effective to decrease the fitness of the weed by about 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or more in comparison to an untreated weed (e.g., a weed to which the PMP composition has not been administered). For example, the method may be effective to kill the weed, thereby decreasing a population of the weed by about 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or more in comparison to an untreated weed. In some instances, the method substantially eliminates the weed. Examples of weeds that can be treated in accordance with the present methods are further described herein.


As used herein, the term weed refers to a plant that grows where it is not wanted. Such plants are typically invasive and, at times, harmful, or have the risk of becoming so. Weeds may be treated with the present PMP compositions to reduce or eliminate the presence, viability, or reproduction of the plant. For example, and without being limited thereto, the methods can be used to target weeds known to damage plants. For example, and without being limited thereto, the weeds can be any member of the following group of families: Gramineae, Umbelliferae, Papilionaceae, Cruciferae, Malvaceae, Eufhorbiaceae, Compositae, Chenopodiaceae, Fumariaceae, Charyophyllaceae, Primulaceae, Geraniaceae, Polygonaceae, Juncaceae, Cyperaceae, Aizoaceae, Asteraceae, Convolvulaceae, Cucurbitaceae, Euphorbiaceae, Polygonaceae, Portulaceae, Solanaceae, Rosaceae, Simaroubaceae, Lardizabalaceae, Liliaceae, Amaranthaceae, Vitaceae, Fabaceae, Primulaceae, Apocynaceae, Araliaceae, Caryophyllaceae, Asclepiadaceae, Celastraceae, Papaveraceae, Onagraceae, Ranunculaceae, Lamiaceae, Commelinaceae, Scrophulariaceae, Dipsacaceae, Boraginaceae, Equisetaceae, Geraniaceae, Rubiaceae, Cannabaceae, Hyperiacaceae, Balsaminaceae, Lobeliaceae, Caprifoliaceae, Nyctaginaceae, Oxalidaceae, Vitaceae, Urticaceae, Polypodiaceae, Anacardiaceae, Smilacaceae, Araceae, Campanulaceae, Typhaceae, Valerianaceae, Verbenaceae, Violaceae. For example, and without being limited thereto, the weeds can be any member of the group consisting of Lolium Rigidum, Amaramthus palmeri, Abutilon theopratsi, Sorghum halepense, Conyza Canadensis, Setaria verticillata, Capsella pastoris, and Cyperus rotundas. Additional weeds include, for example, Mimosapigra, salvinia, hyptis, senna, noogoora, burr, Jatropha gossypifolia, Parkinsonia aculeate, Chromolaena odorata, Cryptoslegia grandiflora, or Andropogon gayanus. Weeds can include monocotyledonous plants (e.g., Agrostis, Alopecurus, Avena, Bromus, Cyperus, Digitaria, Echinochloa, Lolium, Monochoria, Rottboellia, Sagittaria, Scirpus, Setaria, Sida or Sorghum) or dicotyledonous plants (Abutilon, Amaranthus, Chenopodium, Chrysanthemum, Conyza, Galium, Ipomoea, Nasturtium, Sinapis, Solanum, Stellaria, Veronica, Viola or Xanthium).


The compositions and related methods can be used to prevent infestation by or reduce the numbers of pathogens or pathogen vectors in any habitats in which they reside (e.g., outside of animals, e.g., on plants, plant parts (e.g., roots, fruits and seeds), in or on soil, water, or on another pathogen or pathogen vector habitat. Accordingly, the compositions and methods can reduce the damaging effect of pathogen vectors by for example, killing, injuring, or slowing the activity of the vector, and can thereby control the spread of the pathogen to animals. Compositions disclosed herein can be used to control, kill, injure, paralyze, or reduce the activity of one or more of any pathogens or pathogen vectors in any developmental stage, e.g., their egg, nymph, instar, larvae, adult, juvenile, or desiccated forms. The details of each of these methods are described further below.


B. Delivery to a Plant Pest


Provided herein are methods of delivering a PMP composition (e.g., manufactured in accordance with the methods or bioreactors herein) to a plant pest, e.g., by contacting the plant pest with the PMP composition. In some instances, plant pest may be treated with unloaded PMPs. In other instances, the PMPs include a heterologous functional agent, e.g., pesticidal agents (e.g., antibacterial agents, antifungal agents, nematicides, molluscicides, virucides, or herbicides) or pest control agents (e.g., repellents). For example, the methods can be useful for decreasing the fitness of a pest, e.g., to prevent or treat a pest infestation as a consequence of delivery of a PMP composition.


In one aspect, provided herein is a method of decreasing the fitness of a pest, the method including delivering to the pest the PMP composition described herein (e.g., in an effective amount and for an effective duration) to decrease the fitness of the pest relative to an untreated pest (e.g., a pest that has not been delivered the PMP composition).


In one aspect, provided herein is a method of decreasing a fungal infection in (e.g., treating) a plant having a fungal infection, wherein the method includes delivering to the plant pest a PMP composition including a plurality of PMPs (e.g., a PMP composition described herein).


In another aspect, provided herein is a method of decreasing a fungal infection in (e.g., treating) a plant having a fungal infection, wherein the method includes delivering to the plant pest a PMP composition including a plurality of PMPs (e.g., a PMP composition described herein), and wherein the plurality of PMPs include an antifungal agent. In some instances, the antifungal agent is a nucleic acid that inhibits expression of a gene (e.g., dc/1 and dc/2 (i.e., dc/1/2) in a fungus that causes the fungal infection. In some instances, the fungal infection is caused be a fungus belonging to a Sclerotinia spp. (e.g., Sclerotinia sclerotiorum), a Botrytis spp. (e.g., Botrytis cinerea), an Aspergillus spp., a Fusarium spp., or a Penicillium spp. In some instances, the composition includes a PMP produced from an Arabidopsis apoplast EV. In some instances, the method decreases or substantially eliminates the fungal infection.


In another aspect, provided herein is a method of decreasing a bacterial infection in (e.g., treating) a plant having a bacterial infection, wherein the method includes delivering to the plant pest a PMP composition including a plurality of PMPs (e.g., a PMP composition described herein).


In another aspect, provided herein is a method of decreasing a bacterial infection in (e.g., treating) a plant having a bacterial infection, wherein the method includes delivering to the plant pest a PMP composition including a plurality of PMPs, and wherein the plurality of PMPs include an antibacterial agent. In some instances, the antibacterial agent is streptomycin. In some instances, the bacterial infection is caused by a bacterium belonging to a Pseudomonas spp (e.g., Pseudomonas syringae). In some instances, the composition includes a PMP produced from an Arabidopsis apoplast EV. In some instances, the method decreases or substantially eliminates the bacterial infection.


In another aspect, provided herein is a method of decreasing the fitness of an insect plant pest, wherein the method includes delivering to the insect plant pest a PMP composition including a plurality of PMPs (e.g., a PMP composition described herein).


In another aspect, provided herein is a method of decreasing the fitness of an insect plant pest, wherein the method includes delivering to the insect plant pest a PMP composition including a plurality of PMPs (e.g., a PMP composition described herein), and wherein the plurality of PMPs includes an insecticidal agent. In some instances, the insecticidal agent is a peptide nucleic acid. In some instances, the insect plant pest is an aphid. In some instances, the insect plant pest is a lepidopteran (e.g., Spodoptera frugiperda). In some instances, the method decreases the fitness of the insect plant pest relative to an untreated insect plant pest


In another aspect, provided herein is a method of decreasing the fitness of a nematode plant pest, wherein the method includes delivering to the nematode plant pest a PMP composition including a plurality of PMPs (e.g., a PMP composition described herein).


In another aspect, provided herein is a method of decreasing the fitness of a nematode plant pest, wherein the method includes delivering to the nematode plant pest a PMP composition including a plurality of PMPs (e.g., a PMP composition described herein), and wherein the plurality of PMPs include a nematicidal agent. In some instances, the nematicidal agent is a neuropeptide (e.g., Mi-NLP-15b). In some instances, the nematode plant pest is a corn root-knot nematode. In some instances, the method decreases the fitness of the nematode plant pest relative to an untreated nematode plant pest.


In another aspect, provided herein is a method of decreasing the fitness of a weed, wherein the method includes delivering to the weed a PMP composition including a plurality of PMPs (e.g., a PMP composition described herein).


In another aspect, provided herein is a method of decreasing the fitness of a weed, wherein the method includes delivering to the weed a PMP composition including a plurality of PMPs (e.g., a PMP composition described herein), and wherein the plurality of PMPs include an herbicidal agent (e.g. Glufosinate). In some instances, the weed is an Indian goosegrass (Eleusine indica). In some instances, the method decreases the fitness of the weed relative to an untreated weed.


A decrease in the fitness of the pest as a consequence of delivery of a PMP composition can manifest in a number of ways. In some instances, the decrease in fitness of the pest may manifest as a deterioration or decline in the physiology of the pest (e.g., reduced health or survival) as a consequence of delivery of the PMP composition. In some instances, the fitness of an organism may be measured by one or more parameters, including, but not limited to, reproductive rate, fertility, lifespan, viability, mobility, fecundity, pest development, body weight, metabolic rate or activity, or survival in comparison to a pest to which the PMP composition has not been administered. For example, the methods or compositions provided herein may be effective to decrease the overall health of the pest or to decrease the overall survival of the pest. In some instances, the decreased survival of the pest is about 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or greater than 100% greater relative to a reference level (e.g., a level found in a pest that does not receive a PMP composition). In some instances, the methods and compositions are effective to decrease pest reproduction (e.g., reproductive rate, fertility) in comparison to a pest to which the PMP composition has not been administered. In some instances, the methods and compositions are effective to decrease other physiological parameters, such as mobility, body weight, life span, fecundity, or metabolic rate, by about 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or greater than 100% relative to a reference level (e.g., a level found in a pest that does not receive a PMP composition).


In some instances, the decrease in pest fitness may manifest as a decrease in the production of one or more nutrients in the pest (e.g., vitamins, carbohydrates, amino acids, or polypeptides) in comparison to a pest to which the PMP composition has not been administered. In some instances, the methods or compositions provided herein may be effective to decrease the production of nutrients in the pest (e.g., vitamins, carbohydrates, amino acids, or polypeptides) by about 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or greater than 100% relative to a reference level (e.g., a level found in a pest that does not receive a PMP composition).


In some instances, the decrease in pest fitness may manifest as an increase in the pest's sensitivity to a pesticidal agent and/or a decrease in the pest's resistance to a pesticidal agent in comparison to a pest to which the PMP composition has not been administered. In some instances, the methods or compositions provided herein may be effective to increase the pest's sensitivity to a pesticidal agent by about 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or greater than 100% relative to a reference level (e.g., a level found in a pest that does not receive a PMP composition). The pesticidal agent may be any pesticidal agent known in the art, including insecticidal agents. In some instances, the methods or compositions provided herein may increase the pest's sensitivity to a pesticidal agent by decreasing the pest's ability to metabolize or degrade the pesticidal agent into usable substrates in comparison to a pest to which the PMP composition has not been administered.


In some instances, the decrease in pest fitness may manifest as an increase in the pest's sensitivity to an allelochemical agent and/or a decrease in the pest's resistance to an allelochemical agent in comparison to a pest to which the PMP composition has not been administered. In some instances, the methods or compositions provided herein may be effective to decrease the pest's resistance to an allelochemical agent by about 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or greater than 100% relative to a reference level (e.g., a level found in a pest that does not receive a PMP composition). In some instances, the allelochemical agent is caffeine, soyacystatin, fenitrothion, monoterpenes, diterpene acids, or phenolic compounds (e.g., tannins, flavonoids). In some instances, the methods or compositions provided herein may increase the pest's sensitivity to an allelochemical agent by decreasing the pest's ability to metabolize or degrade the allelochemical agent into usable substrates in comparison to a pest to which the PMP composition has not been administered.


In some instances, the methods or compositions provided herein may be effective to decease the pest's resistance to parasites or pathogens (e.g., fungal, bacterial, or viral pathogens or parasites) in comparison to a pest to which the PMP composition has not been administered. In some instances, the methods or compositions provided herein may be effective to decrease the pest's resistance to a pathogen or parasite (e.g., fungal, bacterial, or viral pathogens; or parasitic mites) by about 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or greater than 100% relative to a reference level (e.g., a level found in a pest that does not receive a PMP composition).


In some instances, the methods or compositions provided herein may be effective to decrease the pest's ability to carry or transmit a plant pathogen (e.g., plant virus (e.g., TYLCV) or a plant bacterium (e.g., Agrobacterium spp)) in comparison to a pest to which the PMP composition has not been administered. For example, the methods or compositions provided herein may be effective to decrease the pest's ability to carry or transmit a plant pathogen (e.g., a plant virus (e.g., TYLCV) or plant bacterium (e.g., Agrobacterium spp)) by about 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or greater than 100% relative to a reference level (e.g., a level found in a pest that does not receive a PMP composition).


Additionally or alternatively, in cases where an herbicide is included in the PMP, or compositions thereof, the methods may be further used to decrease the fitness of or kill weeds. In such instances, the method may be effective to decrease the fitness of the weed by about 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or more in comparison to an untreated weed (e.g., a weed to which the PMP composition has not been administered). For example, the method may be effective to kill the weed, thereby decreasing a population of the weed by about 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or more in comparison to an untreated weed. In some instances, the method substantially eliminates the weed. Examples of weeds that can be treated in accordance with the present methods are further described herein.


In some instances, the decrease in pest fitness may manifest as other fitness disadvantages, such as a decreased tolerance to certain environmental factors (e.g., a high or low temperature tolerance), a decreased ability to survive in certain habitats, or a decreased ability to sustain a certain diet in comparison to a pest to which the PMP composition has not been administered. In some instances, the methods or compositions provided herein may be effective to decrease pest fitness in any plurality of ways described herein. Further, the PMP composition may decrease pest fitness in any number of pest classes, orders, families, genera, or species (e.g., 1 pest species, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, 200, 250, 500, or more pest species). In some instances, the PMP composition acts on a single pest class, order, family, genus, or species.


Pest fitness may be evaluated using any standard methods in the art. In some instances, pest fitness may be evaluated by assessing an individual pest. Alternatively, pest fitness may be evaluated by assessing a pest population. For example, a decrease in pest fitness may manifest as a decrease in successful competition against other insects, thereby leading to a decrease in the size of the pest population.


i. Fungi


The PMP compositions and related methods can be useful for decreasing the fitness of a fungus, e.g., to prevent or treat a fungal infection in a plant. Included are methods for delivering a PMP composition to a fungus by contacting the fungus with the PMP composition. Additionally or alternatively, the methods include delivering the PMP composition to a plant at risk of or having a fungal infection, by contacting the plant with the PMP composition.


The PMP compositions and related methods are suitable for delivery to fungi that cause fungal diseases in plants, including diseases caused by powdery mildew pathogens, for example Blumeria species, for example Blumeria graminis; Podosphaera species, for example Podosphaera leucotricha; Sphaerotheca species, for example Sphaerotheca fuliginea; Uncinula species, for example Uncinula necator; diseases caused by rust disease pathogens, for example Gymnosporangium species, for example Gymnosporangium sabinae; Hemileia species, for example Hemileia vastatrix; Phakopsora species, for example Phakopsora pachyrhizi and Phakopsora meibomiae; Puccinia species, for example Puccinia recondite, P. triticina, P. graminis or P. striiformis or P. hordei; Uromyces species, for example Uromyces appendiculatus; diseases caused by pathogens from the group of the Oomycetes, for example Albugo species, for example Algubo candida; Bremia species, for example Bremia lactucae; Peronospora species, for example Peronospora pisi, P. parasitica or P. brassicae; Phytophthora species, for example Phytophthora infestans; Plasmopara species, for example Plasmopara viticola; Pseudoperonospora species, for example Pseudoperonospora humuli or Pseudoperonospora cubensis; Pythium species, for example Pythium ultimum; leaf blotch diseases and leaf wilt diseases caused, for example, by Alternaria species, for example Alternaria solani; Cercospora species, for example Cercospora beticola; Cladiosporium species, for example Cladiosporium cucumerinum; Cochliobolus species, for example Cochliobolus sativus (conidia form: Drechslera, Syn: Helminthosporium), Cochliobolus miyabeanus; Colletotrichum species, for example Colletotrichum lindemuthanium; Cycloconium species, for example Cycloconium oleaginum; Diaporthe species, for example Diaporthe citri; Elsinoe species, for example Elsinoe fawcettii; Gloeosporium species, for example Gloeosporium laeticolor; Glomerella species, for example Glomerella cingulata; Guignardia species, for example Guignardia bidwelli; Leptosphaeria species, for example Leptosphaeria maculans, Leptosphaeria nodorum; Magnaporthe species, for example Magnaporthe grisea; Microdochium species, for example Microdochium nivale; Mycosphaerella species, for example Mycosphaerella graminicola, M. arachidicola and M. fifiensis; Phaeosphaeria species, for example Phaeosphaeria nodorum; Pyrenophora species, for example Pyrenophora teres, Pyrenophora tritici repentis; Ramularia species, for example Ramularia collo-cygni, Ramularia areola; Rhynchosporium species, for example Rhynchosporium secalis; Septoria species, for example Septoria apii, Septoria lycopersii; Typhula species, for example Typhula incarnata; Venturia species, for example Venturia inaequalis; root and stem diseases caused, for example, by Corticium species, for example Corticium graminearum; Fusarium species, for example Fusarium oxysporum; Gaeumannomyces species, for example Gaeumannomyces graminis; Rhizoctonia species, such as, for example Rhizoctonia solani; Sarocladium diseases caused for example by Sarocladium oryzae; Sclerotium diseases caused for example by Sclerotium oryzae; Tapesia species, for example Tapesia acuformis; Thielaviopsis species, for example Thielaviopsis basicola; ear and panicle diseases (including corn cobs) caused, for example, by Alternaria species, for example Alternaria spp.; Aspergillus species, for example Aspergillus flavus; Cladosporium species, for example Cladosporium cladosporioides; Claviceps species, for example Claviceps purpurea; Fusarium species, for example Fusarium culmorum; Gibberella species, for example Gibberella zeae; Monographella species, for example Monographella nivalis; Septoria species, for example Septoria nodorum; diseases caused by smut fungi, for example Sphacelotheca species, for example Sphacelotheca reiliana; Tilletia species, for example Tilletia caries, T. controversa; Urocystis species, for example Urocystis occulta; Ustilago species, for example Ustilago nuda, U. nuda tritici; fruit rot caused, for example, by Aspergillus species, for example Aspergillus flavus; Botrytis species, for example Botrytis cinerea; Penicillium species, for example Penicillium expansum and P. purpurogenum; Sclerotinia species, for example Sclerotinia sclerotiorum; Verticilium species, for example Verticilium alboatrum; seed and soilborne decay, mould, wilt, rot and damping-off diseases caused, for example, by Alternaria species, caused for example by Alternaria brassicicola; Aphanomyces species, caused for example by Aphanomyces euteiches; Ascochyta species, caused for example by Ascochyta lentis; Aspergillus species, caused for example by Aspergillus flavus; Cladosporium species, caused for example by Cladosporium herbarum; Cochliobolus species, caused for example by Cochliobolus sativus; (Conidiaform: Drechslera, Bipolaris Syn: Helminthosporium); Colletotrichum species, caused for example by Colletotrichum coccodes; Fusarium species, caused for example by Fusarium culmorum; Gibberella species, caused for example by Gibberella zeae; Macrophomina species, caused for example by Macrophomina phaseolina; Monographella species, caused for example by Monographella nivalis; Penicillium species, caused for example by Penicillium expansum; Phoma species, caused for example by Phoma lingam; Phomopsis species, caused for example by Phomopsis sojae; Phytophthora species, caused for example by Phytophthora cactorum; Pyrenophora species, caused for example by Pyrenophora graminea; Pyricularia species, caused for example by Pyricularia oryzae; Pythium species, caused for example by Pythium ultimum; Rhizoctonia species, caused for example by Rhizoctonia solani; Rhizopus species, caused for example by Rhizopus oryzae; Sclerotium species, caused for example by Sclerotium rolfsii; Septoria species, caused for example by Septoria nodorum; Typhula species, caused for example by Typhula incarnata; Verticillium species, caused for example by Verticillium dahliae; cancers, galls and witches' broom caused, for example, by Nectria species, for example Nectria galligena; wilt diseases caused, for example, by Monilinia species, for example Monilinia laxa; leaf blister or leaf curl diseases caused, for example, by Exobasidium species, for example Exobasidium vexans; Taphrina species, for example Taphrina deformans; decline diseases of wooden plants caused, for example, by Esca disease, caused for example by Phaemoniella clamydospora, Phaeoacremonium aleophilum and Fomitiporia mediterranea; Eutypa dyeback, caused for example by Eutypa lata; Ganoderma diseases caused for example by Ganoderma boninense; Rigidoporus diseases caused for example by Rigidoporus lignosus; diseases of flowers and seeds caused, for example, by Botrytis species, for example Botrytis cinerea; diseases of plant tubers caused, for example, by Rhizoctonia species, for example Rhizoctonia solani; Helminthosporium species, for example Helminthosporium solani; Club root caused, for example, by Plasmodiophora species, for example Plamodiophora brassicae; diseases caused by bacterial pathogens, for example Xanthomonas species, for example Xanthomonas campestris pv. oryzae; Pseudomonas species, for example Pseudomonas syringae pv. lachrymans; Erwinia species, for example Erwinia amylovora.


Fungal diseases on leaves, stems, pods and seeds caused, for example, by Alternaria leaf spot (Alternaria spec. atrans tenuissima), Anthracnose (Colletotrichum gloeosporoides dematium var. truncatum), brown spot (Septoria glycines), cercospora leaf spot and blight (Cercospora kikuchii), choanephora leaf blight (Choanephora infundibulifera trispora (Syn.)), dactuliophora leaf spot (Dactuliophora glycines), downy mildew (Peronospora manshurica), drechslera blight (Drechslera glycini), frogeye leaf spot (Cercospora sojina), leptosphaerulina leaf spot (Leptosphaerulina trifolii), phyllostica leaf spot (Phyllosticta sojaecola), pod and stem blight (Phomopsis sojae), powdery mildew (Microsphaera diffusa), pyrenochaeta leaf spot (Pyrenochaeta glycines), rhizoctonia aerial, foliage, and web blight (Rhizoctonia solani), rust (Phakopsora pachyrhizi, Phakopsora meibomiae), scab (Sphaceloma glycines), stemphylium leaf blight (Stemphylium botryosum), target spot (Corynespora cassiicola).


Fungal diseases on roots and the stem base caused, for example, by black root rot (Calonectria crotalariae), charcoal rot (Macrophomina phaseolina), fusarium blight or wilt, root rot, and pod and collar rot (Fusarium oxysporum, Fusarium orthoceras, Fusarium semitectum, Fusarium equiseti), mycoleptodiscus root rot (Mycoleptodiscus terrestris), neocosmospora (Neocosmospora vasinfecta), pod and stem blight (Diaporthe phaseolorum), stem canker (Diaporthe phaseolorum var. caulivora), phytophthora rot (Phytophthora megasperma), brown stem rot (Phialophora gregata), pythium rot (Pythium aphanidermatum, Pythium irregulare, Pythium debaryanum, Pythium myriotylum, Pythium ultimum), rhizoctonia root rot, stem decay, and damping-off (Rhizoctonia solani), sclerotinia stem decay (Sclerotinia sclerotiorum), sclerotinia southern blight (Sclerotinia rolfsih), thielaviopsis root rot (Thielaviopsis basicola).


In certain instances, the fungus is a Sclerotinia spp (Scelrotinia sclerotiorum). In certain instances, the fungus is a Botrytis spp (e.g., Botrytis cinerea). In certain instances, the fungus is an Aspergillus spp. In certain instances, the fungus is a Fusarium spp. In certain instances, the fungus is a Penicillium spp.


Compositions of the present invention are useful in various fungal control applications. The above-described compositions may be used to control fungal phytopathogens prior to harvest or post-harvest fungal pathogens. In one embodiment, any of the above-described compositions are used to control target pathogens such as Fusarium species, Botrytis species, Verticillium species, Rhizoctonia species, Trichoderma species, or Pythium species by applying the composition to plants, the area surrounding plants, or edible cultivated mushrooms, mushroom spawn, or mushroom compost. In another embodiment, compositions of the present invention are used to control post-harvest pathogens such as Penicillium, Geotrichum, Aspergillus niger, or Colletotrichum species.


Table 6 provides further examples of fungi, and plant diseases associated therewith, that can be treated or prevented using the PMP composition and related methods described herein.









TABLE 6







Fungal pests








Disease
Causative Agent






Alternaria leaf blight of wheat


Alternaria triticina




Alternaria leaf spot of cole crops


Alternaria japonica



American soybean rust

Phakopsora meibomiae



Ampelopsis rust

Phakopsora ampelopsidis



Anemone

Ochropsora ariae



Angular leaf spot of Citrus

Pseudocercospora angolensis



Arctic Rubus rust

Phragmidium arcticum



Ascochyta blight of broad beans

Didymella fabae



Ash dieback

Chalara fraxinea



Asia mountain Rosa rust

Phragmidium butleri



Asian filbert rust

Pucciniastrum coryli



Asian Kuehneola rose rust

Kuehneola japonica



Asian Mountain Rubus rust

Phragmidium assamense



Asian Phragmidium Rubus rust

Phragmidium arisanense



Asian pistacio rust

Pileolaria pistaciae



Asian rose rust

Gerwasia rosae



Asian Rubus rust

Hamaspora hashiokai



Asian soybean rust

Phakopsora pachyrhizi



Asian sugarcane smut

Sporisorium sacchari



Asian Wart bark, blister canker,

Botryosphaeria berengeriana f. sp. pyricola



ring rot, Physalospora canker of


pear and apple


Asian/European brown rot of

Monilinia fructigena



rosaceae


Asiatic brown fruit rot

Monilia polystroma



Barclay's Asian Rubus rust

Phragmidium barclayi



Black leaf blight of soybean

Arkoola nigra



Blister blight of tea

Exobasidium vexans



Blue stain of Mongolian oak

Ophiostoma longicollum



Box Rust or Boxwood Rust

Puccinia buxi



Brown rust of sugarcane

Puccinia melanocephala



Cherry leaf scorch

Apiognomonia erythrostoma



Chocolate spot of Ya Li pears

Alternaria yaliinficiens



Chrysanthemum White Rust

Puccinia horiana



Coffee Leaf Rust

Hemileia vastatrix



Common Asian Rubus Rust

Hamaspora acutissima



Common larch

Melampsora capraearum



Common potato and tomato rust

Puccinia pittieriana




Crumenulopsis pine dieback


Crumenulopsis sororia



Daylily Rust

Puccinia hemerocallidis



Digitalis Downy Mildew

Peronospora digitalis



Downy mildew (Plasmopara) of

Plasmopara obducens



Impatiens


Eggplant

Puccinia substriata var. substriata



Ergot of pearl millet

Claviceps fusiformis



European Larch canker

Lachnellula willkommii



Few-loculed Asian Rubus rust

Phragmidium pauciloculare



Flag smut of wheat

Urocystis agropyri



Gladiolus Rust

Uromyces transversalis




Goplana dioscoreae


Goplana dioscoreae



Grape leaf rust

Phakopsora euvitis



Gray Rubus rust

Phragmidium griseum



Himalayan rhododendron

Chrysomyxa himalensis



spruce rust


Hiratsuka Rubus rust

Phragmidium hiratsukanum



Horse's tooth or ergot of maize

Claviceps gigantea



Japanese apple rust

Gymnosporangium yamadae



Japanese Chamaecyparis

Gymnosporangium miyabei



Japanese ergot of sorghum

Claviceps sorghicola



Kamtschatka rose rust

Phragmidium kamtschatkae



Late wilt of maize

Harpophora maydis



Long-Spored Asian Rubus rust

Hamaspora longissima



Mai secco disease of Citrus

Phoma tracheiphila



Miscanthus

Puccinia miscanthi



Mulberry rust

Aecidium mori



Nambu Rubus rust

Phragmidium nambuanum



Neck rot of onion

Ciborinia allii



New Zealand Rubus Rust

Hamaspora australis



Northern blue stain of pine

Leptographium wingfieldii



Northern spruce

Chrysomyxa rhododendri



Oak Wilt

Ceratocystis fagacearum



Orange rust of sugarcane

Puccinia kuehnii




Peronospora radii


Peronospora radii



Pistachio Rust

Pileolaria terebinthi



Poinsettia scab

Sphaceloma poinsettiae



Potato smut

Thecaphora solani




Puccinia gladioli on Gladiolus


Puccinia gladioli




Puccinia glyceriae (anam.


Puccinia glyceriae




Aecidium hydrangea




Puccinia mccleanii on Gladiolus


Puccinia mccleanii




Puccinia psidii


Puccinia psidii




Pucciniastrum actinidiae on


Pucciniastrum actinidiae




Actinidia spp.



Red Miscanthus rust

Puccinia erythropus



Rust of European blackberry

Phragmidium bulbosum



Rust of Rubus saxitilis

Phragmidium acuminatum



Rust on Asian Rubus

Gerwasia rubi



Rust on South American Rubus

Gerwasia imperialis



Scots stem pine rust

Cronartium flaccidum



Shoot blight of boxwood

Calonectria pseudonaviculata



Sirex wasp fungus

Amylostereum areolatum



Solanum

Puccinia agrophila



South American Rubus rust

Gerwasia mayorii




Sporisorium smut of wild


Sporisorium pulverulentum



Saccharum


Spruce needle rust

Chrysomyxa abietis



Stackburn, seedling blight, leaf

Alternaria padwickii



spot of rice


Sudden needle drop of Spruce

Setomelanomma holmii



(SNEED)


Sugary disease or Asian ergot

Claviceps sorghi



of sorghum


Sweet potato rust

Endophyllum kaernbachii



Taiwan Rubus rust

Phragmidium formosanum



Tar spot of corn

Phyllachora maydis



Teak Rust

Olivea tectonae




Thekopsora areolate


Thekopsora areolata



Tip over disease of egglant

Diaporthe vexans



Tropical American Kuehneola

Kuehneola loeseneriana



rust of Rubus


Tropical American Mainsia

Mainsia rubi



Rubus rust


Tropical Soybean Rust

Aecidium glycines




Uromyces gladioli on Gladiolus


Uromyces gladioli




Uromyces nyikensis on


Uromyces nyikensis



Gladiolus



Uromycladium tepperianum on


Uromycladium tepperianum




Acacia spp.



Variable Rubus

Gerwasia variabilis



Wineberry Rubus rust

Hamaspora sinica var. sinica



Yamada Rubusrust

Phragmidium yamadanum



Anthracnose leaf blight and stalk

Colletotrichum graminicola anthracnose (teleomorph: Glomerella



rot

graminicola), Glomerella tucumanensis (anamorph: Glomerella





falcatum)




Aspergillus ear and kernel rot


Aspergillus flavus



Banded leaf and sheath spot

Rhizoctonia solani = Rhizoctonia microsclerotia (teleomorph:





Thanatephorus cucumeris)



Bean rust

Uromyces appendiculatus



Black bundle disease

Acremonium strictum = Cephalosporium acremonium



Black kernel rot

Lasiodiplodia theobromae = Botryodiplodia theobromae



Borde bianco

Marasmiellus sp.



Brown spot (black spot, stalk rot)

Physoderma maydis



Brown stripe downy mildew

Sclerophthora rayssiae var. zeae




Cephalosporium kernel rot


Acremonium strictum = Cephalosporium acremonium



Charcoal rot

Macrophomina phaseolina



Corn common rust

Puccinia sorghi



Corn southern rust

Puccinia polysora



Corn tropical rust

Physopella pallescens, P. zeae = Angiospora zeae




Corticium ear rot


Thanatephorus cucumeris = Corticium sasakii



Cotton rust

Puccinia schedonnardi



Cotton southwestern rust

Puccinia cacabata



Cotton tropical rust

Phakopsora gossypii



Crazy top downy mildew

Sclerophthora macrospora = S. macrospora




Curvularia leaf spot


Curvularia clavata, C. eragrostidis, = C. maculans (teleomorph:





Cochliobolus eragrostidis), Curvularia inaequalis, C. intermedia




(teleomorph: Cochliobolus intermedius), Curvularia lunata



(teleomorph: Cochliobolus lunatus), Curvularia pallescens



(teleomorph: Cochliobolus pallescens), Curvularia senegalensis, C.




tuberculata (teleomorph: Cochliobolus tuberculatus)




Didymella leaf spot


Didymella exitialis




Diplodia ear rot and stalk rot


Diplodia frumenti (teleomorph: Botryosphaeria festucae)




Diplodia ear rot, stalk rot, seed


Diplodia maydis = Stenocarpella maydis



rot and seedling blight



Diplodia leaf spot or leaf streak


Stenocarpella macrospora = Diplodia macrospore



Grape leaf Downey mildew

Plasmopara viticola



Dry ear rot (cob, kernel and stalk

Nigrospora oryzae (teleomorph: Khuskia oryzae)



rot)


Ear rots, minor

Aspergillus glaucus, A. niger, Aspergillus spp., Cunninghamella




sp., Curvularia pallescens, Doratomyces stemonitis =




Cephalotrichum stemonitis, Fusarium culmorum, Gonatobotrys





simplex, Pithomyces maydicus, Rhizopus microsporus, R.





stolonifer = R. nigricans, Scopulariopsis brumptii



epitea

Melampsora larici



Ergot (horse's tooth, diente del

Claviceps gigantea (anamorph: Sphacelia sp.)



caballo)


Eyespot

Aureobasidium zeae = Kabatiella zeae




Fusarium ear and stalk rot


Fusarium subglutinans = F. moniliforme var. subglutinans




Fusarium kernel, root and stalk


Fusarium moniliforme (teleomorph: Gibberella fujikuroi)



rot, seed rot and seedling blight



Fusarium stalk rot, seedling root


Fusarium avenaceum (teleomorph: Gibberella avenacea)



rot



Gibberella ear and stalk rot


Gibberella zeae (anamorph: Fusarium graminearum)



Gray ear rot

Botryosphaeria zeae = Physalospora zeae (anamorph:





Macrophoma zeae)



Gray leaf spot (Cercospora

Cercospora sorghi = C. sorghi var. maydis, C. zeae-maydis leaf




spot)


Green ear downy mildew

Sclerospora graminicola




Helminthosporium ear rot (race


Bipolaris zeicola = Helminthosporium carbonum



1)



Helminthosporium root rot


Exserohilum pedicellatum = Helminthosporium pedicellatum




(teleomorph: Setosphaeria)



Hormodendrum ear rot


Cladosporium cladosporioides = Hormodendrum cladosporioides,



(Cladosporium rot)

C. herbarum (teleomorph: Mycosphaerella tassiana)




Hyalothyridium leaf spot


Hyalothyridium maydis



Java downy mildew

Peronosclerospora maydis = Sclerospora maydis



Late wilt

Cephalosporium maydis



Leaf (brown) rust

Puccinia recondita (anamorph: Aecidium clematitis)



Leaf spots, minor

Alternaria alternata, Ascochyta maydis, A. tritici, A. zeicola,





Bipolaris victoriae = Helminthosporium victoriae (teleomorph:





Cochliobolus victoriae), C. sativus (anamorph: Bipolaris





sorokiniana = H. Exserohilum maydis, Leptothyrium





zeae, Ophiosphaerella herpotricha, Setosphaeria prolata)





Graphium penicillioides, Leptosphaeria prolatum = Drechslera





prolata (teleomorph: sorokinianum = H. sativum), Epicoccum





nigrum, (anamorph: Scolecosporiella sp.), Paraphaeosphaeria





michotii, Phoma sp., Septoria zeae, S. zeicola, S. zeina



Rust fungi
Puccinia veronicae-longifoliae


Musk rose rust
Phragmidium rosae-moschatae


Multiflora rose rust
Phragmidium rosae-multiflorae


Northern corn leaf blight

Exaerohilum turcicum = Helminthosporium turcicum, Setosphaeria





turcica



Northern corn leaf spot

Cochliobolus carbonum



Oat crown rust

Puccinia coronate



Oat stem Rust

Puccinia graminis



Peanut rust

Puccinia arachidis




Penicillium ear rot (blue eye,


Penicillium spp., P. chrysogenum, P. expansum, P. oxalicum



blue mold)


Bay willow-larch rust
Melampsora larici-pentandrae



Phaeocytostroma stalk rot and


Phaeocytostroma ambiguum, Phaeocytosporella zeae



root rot



Phaeosphaeria leaf spot


Phaeosphaeria maydis, Sphaerulina maydis



Philippine downy mildew

Peronosclerospora philippinensis = Sclerospora philippinensis




Physalospora ear rot


Botryosphaeria Botryosphaeria festucae = Physalospora zeicola,




(anamorph: Diplodia frumenti)


Potato common rust

Puccinia pittierianap



Potato deforming rust

Aecidium cantensis



Cereals and grasses

Erysiphe graminis



Powdery mildew


Rose Powdery mildew

Sphaerotheca pannosa



Wheat Powdery mildew

Blumeria graminis f. sp. tritici,



Barley Powdery mildew

Blumeria graminis f. sp. hordei



Grape Powdery mildew

Microsphaera diffusa



Legume Powdery mildew

Erysiphe necator (or Uncinula necator)



Grape Powdery mildew

Leveillula taurica, or Oidiopsis taurica



Onion Powdery mildew

Podosphaera leucotricha



Apple Powdery mildew

Podosphaera xanthii, Erysiphe cichoracearum, Podosphaera fusca,





Leveillula taurica



Cucurbits Powdery mildew

Microsphaera syringae



Lilacs Powdery mildew

Podosphaera aphanis, Geum rivale



Strawberry Powdery mildew

Erysiphe berberidis



Hawthorn Powdery mildew

Podosphaera oxyacanthae



Gooseberry Powdery mildew

Sphaerotheca mors-uvae



Purple leaf sheath

Hemiparasitic bacteria and fungi




Pyrenochaeta stalk rot and root


Phoma terrestris, Pyrenochaeta terrestris



rot



Pythium root rot


Pythium spp., P. arrhenomanes, P. graminicola




Pythium stalk rot


Pythium aphanidermatum = P. butleri L.



Red kernel disease (ear mold,

Epicoccum nigrum



leaf and seed rot)



Rhizoctonia ear rot


Rhizoctonia zeae (teleomorph: Waitea circinata)




Rhizoctonia root rot and stalk rot


Rhizoctonia solani, Rhizoctonia zeae



Root rots, minor

Alternaria alternata, Cercospora sorghi, Dictochaeta fertilis,





Fusarium acuminatum (teleomorph: Gibberella acuminate), F.





equiseti (teleomorph: G. intricans), F. oxysporum, F.





pallidoroseum, F. poae, F. roseum, F. cyanogena, (anamorph: F.





sulphureum), Microdochium bolleyi, Mucor sp., Periconia





circinata, Phytophthora cactorum, P. drechsleri, P. nicotianae var.





parasitica, Rhizopus arrhizus




Rostratum leaf spot (leaf


Setosphaeria rostrata, Helminthosporium (anamorph: Exserohilum



disease, ear and, stalk rot)

rostratum = Helminthosporium rostratum)



rugosae

Phragmidium rosae



Rust, common corn

Puccinia sorghi



Rust, southern corn

Puccinia polysora



Rust, tropical corn

Physopella pallescens, P. zeae = Angiospora zeae



sativae
Balansia oryzae



Sclerotium ear rot


Sclerotium rolfsii (teleomorph: Athelia rolfsii)



(southern blight)


Seed rot-seedling blight

Bipolaris sorokiniana, B. zeicola = Helminthosporium carbonum,





Diplodia maydis, Exserohilum pedicellatum, Exserohilum





turcicum = Helminthosporium turcicum, Fusarium avenaceum, F.





culmorum, F. moniliforme, Gibberella zeae (anamorph: F.





graminearum), Macrophomina





phaseolina, Penicillium spp., Phomopsis sp.,





Pythium spp., Rhizoctonia solani, R. zeae, Sclerotium rolfsii,





Spicaria sp.




Selenophoma leaf spot


Selenophoma sp.



Sheath rot

Gaeumannomyces graminis



Shuck rot

Myrothecium gramineum



sieboldii

Hamaspora rubi



Silage mold

Monascus purpureus, M. rubber



Smut, common

Ustilago zeae = U. maydis



Smut, false

Ustilaginoidea virens



Smut, head

Sphacelotheca reiliana = Sporisorium holci-sorghi




Sorghum downy mildew


Peronosclerospora sorghi = Sclerospora sorghi



Southern corn leaf blight and

Cochliobolus heterostrophus (anamorph: Bipolaris maydis =



stalk rot

Helminthosporium maydis)



Southern leaf spot

Stenocarpella macrospora = Diplodia macrospora



Soybean rust

Phakopsora pachyrhizi




Spontaneum downy mildew


Peronosclerospora spontanea = Sclerospora spontanea



Stalk rots, minor

Cercospora sorghi, Fusarium episphaeria, F. merismoides, F.





oxysportum, F. poae, F. roseum, F. solani (teleomorph: Nectria





haematococca), F. tricinctum, Mariannaea elegans, Mucor sp.,





Rhopographus zeae, Spicaria sp.



Stem rust

Puccinia graminis = P. graminis f. sp. secalis



Storage rots

Aspergillus spp., Penicillium spp. and other fungi



Sugarcane common rust

Puccinia melanocephala = P. eriantha



Sugarcane downy mildew

Peronosclerospora sacchari = Sclerospora sacchari



Tar spot

Phyllachora maydis



thunbergii

Phragmidium rubi




Trichoderma ear rot and root rot


Trichoderma viride = T. lignorum (teleomorph: Hypocrea sp.)



Wheat leaf (brown) rust

Puccinia triticina = P. Recondita f. Sp. tritici = P. tritici-duri



Wheat stem (black) rust

Puccinia graminis = P. graminis f. sp. tritici



Wheat stripe (yellow) rust

Puccinia striiformis (anamorph: P. uredoglumarum)



White ear rot, root and stalk rot

Stenocarpella maydis = Diplodia zeae



Yellow leaf blight

Ascochyta ischaemi, Phyllosticta maydis (teleomorph:





Mycosphaerella zeae-maydis)



Zonate leaf spot

Gloeocercospora sorghi










ii. Bacteria


The PMP compositions and related methods can be useful for decreasing the fitness of a bacterium, e.g., to prevent or treat a bacterial infection in a plant. Included are methods for delivering a PMP composition to a bacterium by contacting the bacteria with the PMP composition. Additionally or alternatively, the methods include delivering the biopesticide to a plant at risk of or having a bacterial infection, by contacting the plant with the PMP composition.


The PMP compositions and related methods are suitable for delivery to bacteria, or a plant infected therewith, including any bacteria described further below. For example, the bacteria may be one belonging to Actinobacteria or Proteobacteria, such as bacteria in the families of the Burkholderiaceae, Xanthomonadaceae, Pseudomonadaceae, Enterobacteriaceae, Microbacteriaceae, and Rhizobiaceae.


In some instances, the bacteria is an Acidovorax avenae subsp., including e.g., Acidovorax avenae subsp. avenae (=Pseudomonas avenae subsp. avenae), Acidovorax avenae subsp. cattleyae (=Pseudomonas cattleyae), or Acidovorax avenae subsp. citrulli (=Pseudomonas pseudoalcaligenes subsp. citrulli, Pseudomonas avenae subsp. citrulli)).


In some instances, the bacteria is a Burkholderia spp., including e.g., Burkholderia andropogonis (=Pseudomonas andropogonis, Pseudomonas woodsii), Burkholderia caryophylli (=Pseudomonas caryophylli), Burkholderia cepacia (=Pseudomonas cepacia), Burkholderia gladioli (=Pseudomonas gladioli), Burkholderia gladioli pv. agaricicola (=Pseudomnas gladioli pv. agaricicola), Burkholderia gladioli pv. alliicola (i.e., Pseudomonas gladioli pv. alliicola), Burkholderia gladioli pv. gladioli (i.e., Pseudomonas gladioli, Pseudomonas gladioli pv. gladioli), Burkholderia glumae (i.e., Pseudomonas glumae), Burkholderia plantarii (i.e., Pseudomonas plantari), Burkholderia solanacearum (i.e., Ralstonia solanacearum), or Ralstonia spp.


In some instances, the bacteria is a Liberibacter spp., including Candidatus Liberibacter spec., including e.g., Candidatus Liberibacter asiaticus, Liberibacter africanus (Laf), Liberibacter americanus (Lam), Liberibacter asiaticus (Las), Liberibacter europaeus (Leu), Liberibacter psyllaurous, or Liberibacter solanacearum (Lso).


In some instances, the bacteria is a Corynebacterium spp. including e.g., Corynebacterium fascians, Corynebacterium flaccumfaciens pv. flaccumfaciens, Corynebacterium michiganensis, Corynebacterium michiganense pv. tritici, Corynebacterium michiganense pv. nebraskense, or Corynebacterium sepedonicum.


In some instances, the bacteria is a Erwinia spp. including e.g., Erwinia amylovora, Erwinia ananas, Erwinia carotovora (i.e., Pectobacterium carotovorum), Erwinia carotovora subsp. atroseptica, Erwinia carotovora subsp. carotovora, Erwinia chrysanthemi, Erwinia chrysanthemi pv. zeae, Erwinia dissolvens, Erwinia herbicola, Erwinia rhapontic, Erwinia stewartiii, Erwinia tracheiphila, or Erwinia uredovora.


In some instances, the bacteria is a Pseudomonas syringae subsp., including e.g., Pseudomonas syringae pv. actinidiae (Psa), Pseudomonas syringae pv. atrofaciens, Pseudomonas syringae pv. coronafaciens, Pseudomonas syringae pv. glycinea, Pseudomonas syringae pv. lachrymans, Pseudomonas syringae pv. maculicola Pseudomonas syringae pv. papulans, Pseudomonas syringae pv. striafaciens, Pseudomonas syringae pv. syringae, Pseudomonas syringae pv. tomato, or Pseudomonas syringae pv. tabaci.


In some instances, the bacteria is a Streptomyces spp., including e.g., Streptomyces acidiscabies, Streptomyces albidoflavus, Streptomyces candidus (i.e., Actinomyces candidus), Streptomyces caviscabies, Streptomyces collinus, Streptomyces europaeiscabiei, Streptomyces intermedius, Streptomyces ipomoeae, Streptomyces luridiscabiei, Streptomyces niveiscabiei, Streptomyces puniciscabiei, Streptomyces retuculiscabiei, Streptomyces scabiei, Streptomyces scabies, Streptomyces setonii, Streptomyces steliiscabiei, Streptomyces turgidiscabies, or Streptomyces wedmorensis.


In some instances, the bacteria is a Xanthomonas axonopodis subsp., including e.g., Xanthomonas axonopodis pv. alfalfae (=Xanthomonas alfalfae), Xanthomonas axonopodis pv. aurantifolii (=Xanthomonas fuscans subsp. aurantifolii), Xanthomonas axonopodis pv. allii (=Xanthomonas campestris pv. allii), Xanthomonas axonopodis pv. axonopodis, Xanthomonas axonopodis pv. bauhiniae (=Xanthomonas campestris pv. bauhiniae), Xanthomonas axonopodis pv. begoniae (=Xanthomonas campestris pv. begoniae), Xanthomonas axonopodis pv. betlicola (=Xanthomonas campestris pv. betlicola), Xanthomonas axonopodis pv. biophyti (=Xanthomonas campestris pv. biophyti), Xanthomonas axonopodis pv. cajani (=Xanthomonas campestris pv. cajani), Xanthomonas axonopodis pv. cassavae (=Xanthomonas cassavae, Xanthomonas campestris pv. cassavae), Xanthomonas axonopodis pv. cassiae (=Xanthomonas campestris pv. cassiae), Xanthomonas axonopodis pv. citri (=Xanthomonas citri), Xanthomonas axonopodis pv. citrumelo (=Xanthomonas alfalfae subsp. citrumelonis), Xanthomonas axonopodis pv. clitoriae (=Xanthomonas campestris pv. clitoriae), Xanthomonas axonopodis pv. coracanae (=Xanthomonas campestris pv. coracanae), Xanthomonas axonopodis pv. cyamopsidis (=Xanthomonas campestris pv. cyamopsidis), Xanthomonas axonopodis pv. desmodii (=Xanthomonas campestris pv. desmodii), Xanthomonas axonopodis pv. desmodiigangetici (=Xanthomonas campestris pv. desmodiigangetici), Xanthomonas axonopodis pv. desmodiilaxiflori (=Xanthomonas campestris pv. desmodiilaxiflori), Xanthomonas axonopodis pv. desmodiirotundifolii (=Xanthomonas campestris pv. desmodiirotundifolii), Xanthomonas axonopodis pv. dieffenbachiae (=Xanthomonas campestris pv. dieffenbachiae), Xanthomonas axonopodis pv. erythrinae (=Xanthomonas campestris pv. erythrinae), Xanthomonas axonopodis pv. fascicularis (=Xanthomonas campestris pv. fasciculari), Xanthomonas axonopodis pv. glycines (=Xanthomonas campestris pv. glycines), Xanthomonas axonopodis pv. khayae (=Xanthomonas campestris pv. khayae), Xanthomonas axonopodis pv. lespedezae (=Xanthomonas campestris pv. lespedezae), Xanthomonas axonopodis pv. maculifoliigardeniae (=Xanthomonas campestris pv. maculifoliigardeniae), Xanthomonas axonopodis pv. malvacearum (=Xanthomonas citri subsp. malvacearum), Xanthomonas axonopodis pv. manihotis (=Xanthomonas campestris pv. manihotis), Xanthomonas axonopodis pv. martyniicola (=Xanthomonas campestris pv. martyniicola), Xanthomonas axonopodis pv. melhusii (=Xanthomonas campestris pv. melhusii), Xanthomonas axonopodis pv. nakataecorchori (=Xanthomonas campestris pv. nakataecorchori), Xanthomonas axonopodis pv. passiflorae (=Xanthomonas campestris pv. passiflorae), Xanthomonas axonopodis pv. patelii (=Xanthomonas campestris pv. patelii), Xanthomonas axonopodis pv. pedalii (=Xanthomonas campestris pv. pedalii), Xanthomonas axonopodis pv. phaseoli (=Xanthomonas campestris pv. phaseoli, Xanthomonas phaseoli), Xanthomonas axonopodis pv. phaseoli var. fuscans (=Xanthomonas fuscans), Xanthomonas axonopodis pv. phyllanthi (=Xanthomonas campestris pv. phyllanthi), Xanthomonas axonopodis pv. physalidicola (=Xanthomonas campestris pv. physalidicola), Xanthomonas axonopodis pv. poinsettiicola (=Xanthomonas campestris pv. poinsettiicola), Xanthomonas axonopodis pv. punicae (=Xanthomonas campestris pv. punicae), Xanthomonas axonopodis pv. rhynchosiae (=Xanthomonas campestris pv. rhynchosiae), Xanthomonas axonopodis pv. ricini (=Xanthomonas campestris pv. ricini), Xanthomonas axonopodis pv. sesbaniae (=Xanthomonas campestris pv. sesbaniae), Xanthomonas axonopodis pv. tamarindi (=Xanthomonas campestris pv. tamarindi), Xanthomonas axonopodis pv. vasculorum (=Xanthomonas campestris pv. vasculorum), Xanthomonas axonopodis pv. vesicatoria (=Xanthomonas campestris pv. vesicatoria, Xanthomonas vesicatoria), Xanthomonas axonopodis pv. vignaeradiatae (=Xanthomonas campestris pv. vignaeradiatae), Xanthomonas axonopodis pv. vignicola (=Xanthomonas campestris pv. vignicola), or Xanthomonas axonopodis pv. vitians (=Xanthomonas campestris pv. vitians).


In some instances, the bacteria is Xanthomonas campestris pv. musacearum, Xanthomonas campestris pv. pruni (=Xanthomonas arboricola pv. pruni), or Xanthomonas fragariae.


In some instances, the bacteria is a Xanthomonas translucens supsp. (=Xanthomonas campestris pv. hordei) including e.g., Xanthomonas translucens pv. arrhenatheri (=Xanthomonas campestris pv. arrhenatheri), Xanthomonas translucens pv. cerealis (=Xanthomonas campestris pv. cerealis), Xanthomonas translucens pv. graminis (=Xanthomonas campestris pv. graminis), Xanthomonas translucens pv. phlei (=Xanthomonas campestris pv. phlei), Xanthomonas translucens pv. phleipratensis (=Xanthomonas campestris pv. phleipratensis), Xanthomonas translucens pv. poae (=Xanthomonas campestris pv. poae), Xanthomonas translucens pv. secalis (=Xanthomonas campestris pv. secalis), Xanthomonas translucens pv. translucens (=Xanthomonas campestris pv. translucens), or Xanthomonas translucens pv. undulosa (=Xanthomonas campestris pv. undulosa).


In some instances, the bacteria is a Xanthomonas oryzae supsp., Xanthomonas oryzae pv. oryzae (=Xanthomonas campestris pv. oryzae), or Xanthomonas oryzae pv. oryzicola (=Xanthomonas campestris pv. oryzicola).


In some instances, the bacteria is a Xylella fastidiosa from the family of Xanthomonadaceae.


Table 7 shows further examples of bacteria, and diseases associated therewith, that can be treated or prevented using the PMP composition and related methods described herein.









TABLE 7







Bacterial pests








Disease
Causative Agent





Bacterial leaf blight and stalk rot

Pseudomonas avenae subsp. avenae



Bacterial leaf spot

Xanthomonas campestris pv. holcicola



Bacterial stalk rot

Enterobacter dissolvens = Erwinia dissolvens



Bacterial stalk and top rot

Erwinia carotovora subsp. carotovora, Erwinia





chrysanthemi pv. Zeae



Bacterial stripe

Pseudomonas andropogonis



Chocolate spot

Pseudomonas syringae pv. Coronafaciens



Goss's bacterial wilt blight (leaf

Clavibacter michiganensis subsp.



freckles and wilt)

nebraskensis = Cornebacterium michiganense




pv. Nebraskense



Holcus spot


Pseudomonas syringae pv. Syringae



Purple leaf sheath
Hemiparasitic bacteria


Seed rot-seedling blight

Bacillus subtilis



Stewart's disease (bacterial wilt)

Pantoea stewartii = Erwinia stewartii



Corn stunt (Mesa Central or Rio
Achapparramiento, stunt, Spiroplasma kunkelii


Grande stunt)


Soft rot

Dickeya dianthicola



Soft rot

Dickeya solani



Fire blight

Erwinia amylovora



Soft rot

P. atrosepticum



Soft rot

Pectobacterium carotovorum ssp. carotovorum



Soft rot

Pectobacterium wasabiae



Bacterial blight

Pseudomonas syringae pv. Porri and pv. Tomato



Brown blotch Disease

Pseudomonas tolaasii



Bacterial wilt

Ralstonia solanacearum



Bacteria wilt

Ralstonia solanacearum



Common scab

Streptomyces scabies



Common scab

Streptomyces scabies




Xanthomonasleaf blight of onion


Xanthomonas axonopodis pv. allii



Asiatic citrus canker

Xanthomonas axonopodis pv. citri



Citrus bacterial spot

Xanthomonas axonopodis pv. citrumelo



Bacterial spot

Xanthomonas campestris pv. vesicatoria



Pierce's Disease

Xylella fastidiosa










iii. Insects


The PMP compositions and related methods can be useful for decreasing the fitness of an insect, e.g., to prevent or treat an insect infestation in a plant. The term “insect” includes any organism belonging to the phylum Arthropoda and to the class Insecta or the class Arachnida, in any stage of development, i.e., immature and adult insects. Included are methods for delivering a PMP composition to an insect by contacting the insect with the PMP composition. Additionally or alternatively, the methods include delivering the biopesticide to a plant at risk of or having an insect infestation, by contacting the plant with the PMP composition.


The PMP compositions and related methods are suitable for preventing or treating infestation by an insect, or a plant infested therewith, including insects belonging to the following orders: Acari, Araneae, Anoplura, Coleoptera, Collembola, Dermaptera, Dictyoptera, Diplura, Diptera (e.g., spotted-wing Drosophila), Embioptera, Ephemeroptera, Grylloblatodea, Hemiptera (e.g., aphids, Greenhous whitefly), Homoptera, Hymenoptera, Isoptera, Lepidoptera, Mallophaga, Mecoptera, Neuroptera, Odonata, Orthoptera, Phasmida, Plecoptera, Protura, Psocoptera, Siphonaptera, Siphunculata, Thysanura, Strepsiptera, Thysanoptera, Trichoptera, or Zoraptera.


In some instances, the insect is from the class Arachnida, for example, Acarus spp., Aceria sheldoni, Aculops spp., Aculus spp., Amblyomma spp., Amphitetranychus viennensis, Argas spp., Boophilus spp., Brevipalpus spp., Bryobia graminum, Bryobia praetiosa, Centruroides spp., Chorioptes spp., Dermanyssus gallinae, Dermatophagoides pteronyssinus, Dermatophagoides farinae, Dermacentor spp., Eotetranychus spp., Epitrimerus pyri, Eutetranychus spp., Eriophyes spp., Glycyphagus domesticus, Halotydeus destructor, Hemitarsonemus spp., Hyalomma spp., Ixodes spp., Latrodectus spp., Loxosceles spp., Metatetranychus spp., Neutrombicula autumnalis, Nuphersa spp., Oligonychus spp., Ornithodorus spp., Ornithonyssus spp., Panonychus spp., Phyllocoptruta oleivora, Polyphagotarsonemus latus, Psoroptes spp., Rhipicephalus spp., Rhizoglyphus spp., Sarcoptes spp., Scorpio maurus, Steneotarsonemus spp., Steneotarsonemus spinki, Tarsonemus spp., Tetranychus spp., Trombicula alfreddugesi, Vaejovis spp., or Vasates lycopersici.


In some instances, the insect is from the class Chilopoda, for example, Geophilus spp. or Scutigera spp.


In some instances, the insect is from the order Collembola, for example, Onychiurus armatus.


In some instances, the insect is from the class Diplopoda, for example, Blaniulus guttulatus; from the class Insecta, e.g. from the order Blattodea, for example, Blattella asahinai, Blattella germanica, Blatta orientalis, Leucophaea maderae, Panchlora spp., Parcoblatta spp., Periplaneta spp., or Supella longipalpa.


In some instances, the insect is from the order Coleoptera, for example, Acalymma vittatum, Acanthoscelides obtectus, Adoretus spp., Agelastica alni, Agriotes spp., Alphitobius diaperinus, Amphimallon solstitialis, Anobium punctatum, Anoplophora spp., Anthonomus spp., Anthrenus spp., Apion spp., Apogonia spp., Atomaria spp., Attagenus spp., Bruchidius obtectus, Bruchus spp., Cassida spp., Cerotoma trifurcata, Ceutorrhynchus spp., Chaetocnema spp., Cleonus mendicus, Conoderus spp., Cosmopolites spp., Costelytra zealandica, Ctenicera spp., Curculio spp., Cryptolestes ferrugineus, Cryptorhynchus lapathi, Cylindrocopturus spp., Dermestes spp., Diabrotica spp. (e.g., corn rootworm), Dichocrocis spp., Dicladispa armigera, Diloboderus spp., Epilachna spp., Epitrix spp., Faustinus spp., Gibbium psylloides, Gnathocerus cornutus, Hellula undalis, Heteronychus arator, Heteronyx spp., Hylamorpha elegans, Hylotrupes bajulus, Hypera postica, Hypomeces squamosus, Hypothenemus spp., Lachnosterna consanguinea, Lasioderma serricorne, Latheticus oryzae, Lathridius spp., Lema spp., Leptinotarsa decemlineata, Leucoptera spp., Lissorhoptrus oryzophilus, Lixus spp., Luperodes spp., Lyctus spp., Megascelis spp., Melanotus spp., Meligethes aeneus, Melolontha spp., Migdolus spp., Monochamus spp., Naupactus xanthographus, Necrobia spp., Niptus hololeucus, Oryctes rhinoceros, Oryzaephilus surinamensis, Oryzaphagus oryzae, Otiorrhynchus spp., Oxycetonia jucunda, Phaedon cochleariae, Phyllophaga spp., Phyllophaga helleri, Phyllotreta spp., Popillia japonica, Premnotrypes spp., Prostephanus truncatus, Psylliodes spp., Ptinus spp., Rhizobius ventralis, Rhizopertha dominica, Sitophilus spp., Sitophilus oryzae, Sphenophorus spp., Stegobium paniceum, Sternechus spp., Symphyletes spp., Tanymecus spp., Tenebrio molitor, Tenebrioides mauretanicus, Tribolium spp., Trogoderma spp., Tychius spp., Xylotrechus spp., or Zabrus spp.


In some instances, the insect is from the order Diptera, for example, Aedes spp., Agromyza spp., Anastrepha spp., Anopheles spp., Asphondylia spp., Bactrocera spp., Bibio hortulanus, Calliphora erythrocephala, Calliphora vicina, Ceratitis capitata, Chironomus spp., Chrysomyia spp., Chrysops spp., Chrysozona pluvialis, Cochliomyia spp., Contarinia spp., Cordylobia anthropophaga, Cricotopus sylvestris, Culex spp., Culicoides spp., Culiseta spp., Cuterebra spp., Dacus oleae, Dasyneura spp., Delia spp., Dermatobia hominis, Drosophila spp., Echinocnemus spp., Fannia spp., Gasterophilus spp., Glossina spp., Haematopota spp., Hydrellia spp., Hydrellia griseola, Hylemya spp., Hippobosca spp., Hypoderma spp., Liriomyza spp., Lucilia spp., Lutzomyia spp., Mansonia spp., Musca spp. (e.g., Musca domestica), Oestrus spp., Oscinella frit, Paratanytarsus spp., Paralauterborniella subcincta, Pegomyia spp., Phlebotomus spp., Phorbia spp., Phormia spp., Piophila casei, Prodiplosis spp., Psila rosae, Rhagoletis spp., Sarcophaga spp., Simulium spp., Stomoxys spp., Tabanus spp., Tetanops spp., or Tipula spp.


In some instances, the insect is from the order Heteroptera, for example, Anasa tristis, Antestiopsis spp., Boisea spp., Blissus spp., Calocoris spp., Campylomma livida, Cavelerius spp., Cimex spp., Collaria spp., Creontiades dilutus, Dasynus piperis, Dichelops furcatus, Diconocoris hewetti, Dysdercus spp., Euschistus spp., Eurygaster spp., Heliopeltis spp., Horcias nobilellus, Leptocorisa spp., Leptocorisa varicornis, Leptoglossus phyllopus, Lygus spp., Macropes excavatus, Miridae, Monalonion atratum, Nezara spp., Oebalus spp., Pentomidae, Piesma quadrata, Piezodorus spp., Psallus spp., Pseudacysta persea, Rhodnius spp., Sahlbergella singularis, Scaptocoris castanea, Scotinophora spp., Stephanitis nashi, Tibraca spp., or Triatoma spp.


In some instances, the insect is from the order Homiptera, for example, Acizzia acaciaebaileyanae, Acizzia dodonaeae, Acizzia uncatoides, Acrida turrita, Acyrthosipon spp., Acrogonia spp., Aeneolamia spp., Agonoscena spp., Aleyrodes proletella, Aleurolobus barodensis, Aleurothrixus floccosus, Allocaridara malayensis, Amrasca spp., Anuraphis cardui, Aonidiella spp., Aphanostigma pini, Aphis spp. (e.g., Apis gossypii), Arboridia apicalis, Arytainilla spp., Aspidiella spp., Aspidiotus spp., Atanus spp., Aulacorthum solani, Bemisia tabaci, Blastopsylla occidentalis, Boreioglycaspis melaleucae, Brachycaudus helichrysi, Brachycolus spp., Brevicoryne brassicae, Cacopsylla spp., Calligypona marginata, Carneocephala fulgida, Ceratovacuna lanigera, Cercopidae, Ceroplastes spp., Chaetosiphon fragaefolii, Chionaspis tegalensis, Chlorita onukii, Chondracris rosea, Chromaphis juglandicola, Chrysomphalus ficus, Cicadulina mbila, Coccomytilus halli, Coccus spp., Cryptomyzus ribis, Cryptoneossa spp., Ctenarytaina spp., Dalbulus spp., Dialeurodes citri, Diaphorina citri, Diaspis spp., Drosicha spp., Dysaphis spp., Dysmicoccus spp., Empoasca spp., Eriosoma spp., Erythroneura spp., Eucalyptolyma spp., Euphyllura spp., Euscelis bilobatus, Ferrisia spp., Geococcus coffeae, Glycaspis spp., Heteropsylla cubana, Heteropsylla spinulosa, Homalodisca coagulata, Homalodisca vitripennis, Hyalopterus arundinis, Icerya spp., Idiocerus spp., Idioscopus spp., Laodelphax striatellus, Lecanium spp., Lepidosaphes spp., Lipaphis erysimi, Macrosiphum spp., Macrosteles facifrons, Mahanarva spp., Melanaphis sacchari, Metcalfiella spp., Metopolophium dirhodum, Monellia costalis, Monelliopsis pecanis, Myzus spp., Nasonovia ribisnigri, Nephotettix spp., Nettigonicla spectra, Nilaparvata lugens, Oncometopia spp., Orthezia praelonga, Oxya chinensis, Pachypsylla spp., Parabemisia myricae, Paratrioza spp., Parlatoria spp., Pemphigus spp., Pentatomidae spp. (e.g., Halyomorpha halys), Peregrinus maidis, Phenacoccus spp., Phloeomyzus passerinii, Phorodon humuli, Phylloxera spp., Pinnaspis aspidistrae, Planococcus spp., Prosopidopsylla flava, Protopulvinaria pyriformis, Pseudaulacaspis pentagona, Pseudococcus spp., Psyllopsis spp., Psylla spp., Pteromalus spp., Pyrilla spp., Quadraspidiotus spp., Quesada gigas, Rastrococcus spp., Rhopalosiphum spp., Saissetia spp., Scaphoideus titanus, Schizaphis graminum, Selenaspidus articulatus, Sogata spp., Sogatella furcifera, Sogatodes spp., Stictocephala festina, Siphoninus phillyreae, Tenalaphara malayensis, Tetragonocephela spp., Tinocallis caryaefoliae, Tomaspis spp., Toxoptera spp., Trialeurodes vaporariorum, Trioza spp., Typhlocyba spp., Unaspis spp., Viteus vitifolii, Zygina spp.; from the order Hymenoptera, for example, Acromyrmex spp., Athalia spp., Atta spp., Diprion spp., Hoplocampa spp., Lasius spp., Monomorium pharaonis, Sirex spp., Solenopsis invicta, Tapinoma spp., Urocerus spp., Vespa spp., or Xeris spp.


In some instances, the insect is from the order Isopoda, for example, Armadillidium vulgare, Oniscus asellus, or Porcellio scaber.


In some instances, the insect is from the order Isoptera, for example, Coptotermes spp., Cornitermes cumulans, Cryptotermes spp., Incisitermes spp., Microtermes obesi, Odontotermes spp., or Reticulitermes spp.


In some instances, the insect is from the order Lepidoptera, for example, Achroia grisella, Acronicta major, Adoxophyes spp., Aedia leucomelas, Agrotis spp., Alabama spp., Amyelois transitella, Anarsia spp., Anticarsia spp., Argyroploce spp., Barathra brassicae, Borbo cinnara, Bucculatrix thurberiella, Bupalus piniarius, Busseola spp., Cacoecia spp., Caloptilia theivora, Capua reticulana, Carpocapsa pomonella, Carposina niponensis, Cheimatobia brumata, Chilo spp., Choristoneura spp., Clysia ambiguella, Cnaphalocerus spp., Cnaphalocrocis medinalis, Cnephasia spp., Conopomorpha spp., Conotrachelus spp., Copitarsia spp., Cydia spp., Dalaca noctuides, Diaphania spp., Diatraea saccharalis, Earias spp., Ecdytolopha aurantium, Elasmopalpus lignosellus, Eldana saccharina, Ephestia spp., Epinotia spp., Epiphyas postvittana, Etiella spp., Eulia spp., Eupoecilia ambiguella, Euproctis spp., Euxoa spp., Feltia spp., Galleria mellonella, Gracillaria spp., Grapholitha spp., Hedylepta spp., Helicoverpa spp., Heliothis spp., Hofmannophila pseudospretella, Homoeosoma spp., Homona spp., Hyponomeuta padella, Kakivoria flavofasciata, Laphygma spp., Laspeyresia molesta, Leucinodes orbonalis, Leucoptera spp., Lithocolletis spp., Lithophane antennata, Lobesia spp., Loxagrotis albicosta, Lymantria spp., Lyonetia spp., Malacosoma neustria, Maruca testulalis, Mamstra brassicae, Melanitis leda, Mocis spp., Monopis obviella, Mythimna separata, Nemapogon cloacellus, Nymphula spp., Oiketicus spp., Oria spp., Orthaga spp., Ostrinia spp., Oulema oryzae, Panolis flammea, Parnara spp., Pectinophora spp., Perileucoptera spp., Phthorimaea spp., Phyllocnistis citrella, Phyllonorycter spp., Pieris spp., Platynota stultana, Plodia interpunctella, Plusia spp., Plutella xylostella, Prays spp., Prodenia spp., Protoparce spp., Pseudaletia spp., Pseudaletia unipuncta, Pseudoplusia includens, Pyrausta nubilalis, Rachiplusia nu, Schoenobius spp., Scirpophaga spp., Scirpophaga innotata, Scotia segetum, Sesamia spp., Sesamia inferens, Sparganothis spp., Spodoptera spp., Spodoptera praefica, Stathmopoda spp., Stomopteryx subsecivella, Synanthedon spp., Tecia solanivora, Thermesia gemmatalis, Tinea cloacella, Tinea pellionella, Tineola bisselliella, Tortrix spp., Trichophaga tapetzella, Trichoplusia spp., Tryporyza incertulas, Tuta absoluta, or Virachola spp.


In some instances, the insect is from the order Orthoptera or Saltatoria, for example, Acheta domesticus, Dichroplus spp., Gryllotalpa spp., Hieroglyphus spp., Locusta spp., Melanoplus spp., or Schistocerca gregaria.


In some instances, the insect is from the order Phthiraptera, for example, Damalinia spp., Haematopinus spp., Linognathus spp., Pediculus spp., Ptirus pubis, Trichodectes spp.


In some instances, the insect is from the order Psocoptera for example Lepinatus spp., or Liposcelis spp.


In some instances, the insect is from the order Siphonaptera, for example, Ceratophyllus spp., Ctenocephalides spp., Pulex irritans, Tunga penetrans, or Xenopsylla cheopsis.


In some instances, the insect is from the order Thysanoptera, for example, Anaphothrips obscurus, Baliothrips biformis, Drepanothrips reuteri, Enneothrips flavens, Frankliniella spp., Heliothrips spp., Hercinothrips femoralis, Rhipiphorothrips cruentatus, Scirtothrips spp., Taeniothrips cardamomi, or Thrips spp.


In some instances, the insect is from the order Zygentoma (=Thysanura), for example, Ctenolepisma spp., Lepisma saccharina, Lepismodes inquilinus, or Thermobia domestica.


In some instances, the insect is from the class Symphyla, for example, Scutigerella spp.


In some instances, the insect is a mite, including but not limited to, Tarsonemid mites, such as Phytonemus pallidus, Polyphagotarsonemus latus, Tarsonemus bilobatus, or the like; Eupodid mites, such as Penthaleus erythrocephalus, Penthaleus major, or the like; Spider mites, such as Oligonychus shinkajii, Panonychus citri, Panonychus mori, Panonychus ulmi, Tetranychus kanzawai, Tetranychus urticae, or the like; Eriophyid mites, such as Acaphylla theavagrans, Aceria tulipae, Aculops lycopersici, Aculops pelekassi, Aculus schlechtendali, Eriophyes chibaensis, Phyllocoptruta oleivora, or the like; Acarid mites, such as Rhizoglyphus robini, Tyrophagus putrescentiae, Tyrophagus similis, or the like; Bee brood mites, such as Varroa jacobsoni, Varroa destructor or the like; Ixodides, such as Boophilus microplus, Rhipicephalus sanguineus, Haemaphysalis longicornis, Haemophysalis flava, Haemophysalis campanulata, Ixodes ovatus, Ixodes persulcatus, Amblyomma spp., Dermacentor spp., or the like; Cheyletidae, such as Cheyletiella yasguri, Cheyletiella blakei, or the like; Demodicidae, such as Demodex canis, Demodex cati, or the like; Psoroptidae, such as Psoroptes ovis, or the like; Scarcoptidae, such as Sarcoptes scabiei, Notoedres cati, Knemidocoptes spp., or the like.


Table 8 shows further examples of insects that cause infestations that can be treated or prevented using the PMP compositions and related methods described herein.









TABLE 8







Insect pests








Common Name
Latin name





European corn borer

Ostrinia nubilalis



Corn earworm

Helicoverpa zea



Beet armyworm

Spodoptera exigua



Fall armyworm

Spodoptera frugiperda



Southwestern corn borer

Diatraea grandiosella



Lesser cornstalk borer

Elasmopalpus lignosellus



Stalk borer

Papaipema nebris



Common armyworm

Pseudaletia unipuncta



Black cutworm

Agrotis ipsilon



Western bean cutworm

Striacosta albicosta



Yellowstriped armyworm

Spodoptera ornithogalli



Western yellowstriped

Spodoptera praefica



armyworm


Southern armyworm

Spodoptera eridania



Southern armyworm

Spodoptera eridania



Variegated cutworm

Peridroma saucia



Stalk borer

Papaipema nebris



Cabbage looper

Trichoplusia ni



Tomato pinworm

Keiferia lycopersicella



Tobacco hornworm

Manduca sexta



Tomato hornworm

Manduca quinquemaculata



Imported cabbageworm

Artogeia rapae



Cabbage butterfly

Pieris brassicae



Cabbage looper

Trichoplusia ni



Diamondback moth

Plutella xylostella



Beet armyworm

Spodoptera exigua



Common cutworm

Agrotis segetum



Potato tuberworm

Phthorimaea operculella



Diamondback moth

Plutella xylostella



Sugarcane borer

Diatraea saccharalis



Glassy cutworm

Crymodes devastator



Dingy cutworm

Feltia ducens



Claybacked cutworm

Agrotis gladiaria



Green cloverworm

Plathypena scabra



Soybean looper

Pseudoplusia includes



Velvetbean caterpillar

Anticarsia gemmatalis



Northern corn rootworm

Coleoptera Diabrotica barberi



Southern corn rootworm

Diabrotica undecimpunctata



Western corn rootworm

Diabrotica virgifera



Maize weevil

Sitophilus zeamais



Colorado potato beetle

Leptinotarsa decemlineata



Tobacco flea beetle

Epitrix hirtipennis



Crucifer flea beetle

Phyllotreta cruciferae



Western black flea beetle

Phyllotreta pusilia



Pepper weevil

Anthonomus eugenii



Colorado potato beetle

Leptinotarsa decemlineata



Potato flea beetle

Epitrix cucumeris



Wireworms Melanpotus spp.

Hemicrepidus memnonius



Wireworms

Ceutorhychus assimilis



Cabbage seedpod weevil

Phyllotreta Cruciferae



Crucifer flea beetle

Melanolus spp.



Wireworm

Aeolus mellillus



Wheat wireworm

Aeolus mancus



Sand wireworm

Horistonotus uhlerii



Maize billbug

Sphenophorus maidis



Timothy bilibug

Sphenophorus zeae



Bluegrass billbug

Sphenophorus parvulus



Southern corn billbug

Sphenophorus callosus



White grubs

Phyllophaga spp.



Corn flea beetle

Chaetocnema pulicaria



Japanese beetle

Popillia japonica



Mexican bean beetle

Epilachna varivestis



Bean leaf beetle

Cerotoma trifurcate



Blister beetles

Epicauta pestifera Epicauta lemniscata



Corn leaf aphid

Homoptera Rhopalosiphum maidis



Corn root aphid

Anuraphis maidiradicis



Green peach aphid

Myzus persicae



Potato aphid

Macrosiphum euphorbiae



Greenhouse whitefly

Trileurodes vaporariorum



Sweetpotato whitefly

Bemisia tabaci



Silverleaf whitefly

Bemisia argentifolii



Cabbage aphid

Brevicoryne brassicae



Green peach aphid

Myzus persicae



Potato leafhopper

Empoasca fabae



Potato psyllid

Paratrioza cockerelli



Silverleaf whitefly

Bemisia argentifolii



Sweetpotato whitefly

Bemisia tabaci



Carrot aphid

Cavariella aegopodii



Cabbage aphid

Brevicoryne brassicae



West Indian canefly

Saccharosydne saccharivora



Yellow sugarcane aphid

Sipha flava



Threecornered alfalfa hopper

Spissistilus festinus




Lygus Hesperus


Hemiptera Lygus lineolaris




Lygus bug


Lygus rugulipennis



Green stink bug

Acrosternum hilare



Brown stick bug

Euschistus servus



Chinch bug

Blissus leucopterus leucopterus



Leafminer

Diptera Liriomyza trifolii



Vegetable leafminer

Liriomyza sativae



Tomato leafminer

Scrobipalpula absoluta



Seedcorn maggot

Delia platura



Cabbage maggot

Delia brassicae



Cabbage root fly

Delia radicum



Carrot rust fly

Psilia rosae



Sugarbeet root maggot

Tetanops myopaeformis



Differential grasshopper

Orthoptera Melanoplus differentialis



Redlegged grasshopper

Melanoplus femurrubrum



Twostriped grasshopper

Melanoplus bivittatus










iv. Mollusks


The PMP compositions and related methods can be useful for decreasing the fitness of a mollusk, e.g., to prevent or treat a mollusk infestation in a plant. The term “mollusk” includes any organism belonging to the phylum Mollusca. Included are methods for delivering a PMP composition to a mollusk by contacting the mollusk with the PMP composition. Additionally or alternatively, the methods include delivering the biopesticide to a plant at risk of or having a mollusk infestation, by contacting the plant with the PMP composition.


The PMP compositions and related methods are suitable for preventing or treating infestation by terrestrial Gastropods (e.g., slugs and snails) in agriculture and horticulture. They include all terrestrial slugs and snails which mostly occur as polyphagous pests on agricultural and horticultural crops. For example, the mollusk may belong to the family Achatinidae, Agriolimacidae, Ampullariidae, Arionidae, Bradybaenidae, Helicidae, Hydromiidae, Lymnaeidae, Milacidae, Urocyclidae, or Veronicellidae.


For example, in some instances, the mollusk is Achatina spp., Archachatina spp. (e.g., Archachatina marginata), Agriolimax spp., Anon spp. (e.g., A. ater, A. circumscriptus, A. distinctus, A. fasciatus, A. hortensis, A. intermedius, A. rufus, A. subfuscus, A. silvaticus, A. lusitanicus), Arliomax spp. (e.g., Ariolimax columbianus), Biomphalaria spp., Bradybaena spp. (e.g., B. fruticum), Bulinus spp., Cantareus spp. (e.g., C. asperses), Cepaea spp. (e.g., C. hortensis, C. nemoralis, C. hortensis), Cernuella spp., Cochlicella spp., Cochlodina spp. (e.g., C. laminata), Deroceras spp. (e.g., D. agrestis, D. empiricorum, D. laeve, D. panornimatum, D. reticulatum), Discus spp. (e.g., D. rotundatus), Euomphalia spp., Galba spp. (e.g., G. trunculata), Helicella spp. (e.g., H. itala, H. obvia), Helicigona spp. (e.g., H. arbustorum), Helicodiscus spp., Helix spp. (e.g., H. aperta, H. aspersa, H. pomatia), Limax spp. (e.g., L. cinereoniger, L. flavus, L. marginatus, L. maximus, L. tenellus), Limicolaria spp. (e.g., Limicolaria aurora), Lymnaea spp. (e.g., L. stagnalis), Mesodon spp. (e.g., Meson thyroidus), Monadenia spp. (e.g., Monadenia fidelis), Milax spp. (e.g., M. gagates, M. marginatus, M. sowerbyi, M. budapestensis), Oncomelania spp., Neohelix spp. (e.g., Neohelix albolabris), Opeas spp., Otala spp. (e.g., Otala lacteal), Oxyloma spp. (e.g., O. pfeiffen), Pomacea spp. (e.g., P. canaliculata), Succinea spp., Tandonia spp. (e.g., T. budapestensis, T. sowerbyi), Theba spp., Vallonia spp., or Zonitoides spp. (e.g., Z. nitidus).


v. Nematodes


The PMP compositions and related methods can be useful for decreasing the fitness of a nematode, e.g., to prevent or treat a nematode infestation in a plant. The term “nematode” includes any organism belonging to the phylum Nematoda. Included are methods for delivering a PMP composition to a nematode by contacting the nematode with the PMP composition. Additionally or alternatively, the methods include delivering the biopesticide to a plant at risk of or having a nematode infestation, by contacting the plant with the PMP composition.


The PMP compositions and related methods are suitable for preventing or treating infestation by nematodes that cause damage plants including, for example, Meloidogyne spp. (root-knot), Heterodera spp., Globodera spp., Pratylenchus spp., Helicotylenchus spp., Radopholus similis, Ditylenchus dipsaci, Rotylenchulus reniformis, Xiphinema spp., Aphelenchoides spp. and Belonolaimus longicaudatus. In some instances, the nematode is a plant parasitic nematodes or a nematode living in the soil. Plant parasitic nematodes include, but are not limited to, ectoparasites such as Xiphinema spp., Longidorus spp., and Trichodorus spp.; semiparasites such as Tylenchulus spp.; migratory endoparasites such as Pratylenchus spp., Radopholus spp., and Scutellonema spp.; sedentary parasites such as Heterodera spp., Globodera spp., and Meloidogyne spp., and stem and leaf endoparasites such as Ditylenchus spp., Aphelenchoides spp., and Hirshmaniella spp. Especially harmful root parasitic soil nematodes are such as cystforming nematodes of the genera Heterodera or Globodera, and/or root knot nematodes of the genus Meloidogyne. Harmful species of these genera are for example Meloidogyne incognita, Heterodera glycines (soybean cyst nematode), Globodera pallida and Globodera rostochiensis (potato cyst nematode), which species are effectively controlled with the PMP compositions described herein. However, the use of the PMP compositions described herein is in no way restricted to these genera or species, but also extends in the same manner to other nematodes.


Other examples of nematodes that can be targeted by the methods and compositions described herein include but are not limited to e.g. Aglenchus agricola, Anguina tritici, Aphelenchoides arachidis, Aphelenchoides fragaria and the stem and leaf endoparasites Aphelenchoides spp. in general, Belonolaimus gracilis, Belonolaimus longicaudatus, Belonolaimus nortoni, Bursaphelenchus cocophilus, Bursaphelenchus eremus, Bursaphelenchus xylophilus, Bursaphelenchus mucronatus, and Bursaphelenchus spp. in general, Cacopaurus pestis, Criconemella curvata, Criconemella onoensis, Criconemella ornata, Criconemella rusium, Criconemella xenoplax (=Mesocriconema xenoplax) and Criconemella spp. in general, Criconemoides femiae, Criconemoides onoense, Criconemoides ornatum and Criconemoides spp. in general, Ditylenchus destructor, Ditylenchus dipsaci, Ditylenchus myceliophagus and the stem and leaf endoparasites Ditylenchus spp. in general, Dolichodorus heterocephalus, Globodera pallida (=Heterodera pallida), Globodera rostochiensis (potato cyst nematode), Globodera solanacearum, Globodera tabacum, Globodera virginia and the sedentary, cyst forming parasites Globodera spp. in general, Helicotylenchus digonicus, Helicotylenchus dihystera, Helicotylenchus erythrine, Helicotylenchus multicinctus, Helicotylenchus nannus, Helicotylenchus pseudorobustus and Helicotylenchus spp. in general, Hemicriconemoides, Hemicycliophora arenaria, Hemicycliophora nudata, Hemicycliophora parvana, Heterodera avenae, Heterodera cruciferae, Heterodera glycines (soybean cyst nematode), Heterodera oryzae, Heterodera schachtii, Heterodera zeae and the sedentary, cyst forming parasites Heterodera spp. in general, Hirschmaniella gracilis, Hirschmaniella oryzae Hirschmaniella spinicaudata and the stem and leaf endoparasites Hirschmaniella spp. in general, Hoplolaimus aegyptii, Hoplolaimus califomicus, Hoplolaimus columbus, Hoplolaimus galeatus, Hoplolaimus indicus, Hoplolaimus magnistylus, Hoplolaimus pararobustus, Longidorus africanus, Longidorus breviannulatus, Longidorus elongatus, Longidorus laevicapitatus, Longidorus vineacola and the ectoparasites Longidorus spp. in general, Meloidogyne acronea, Meloidogyne africana, Meloidogyne arenaria, Meloidogyne arenaria thamesi, Meloidogyne artiella, Meloidogyne chitwoodi, Meloidogyne coffeicola, Meloidogyne ethiopica, Meloidogyne exigua, Meloidogyne fallax, Meloidogyne graminicola, Meloidogyne graminis, Meloidogyne hapla, Meloidogyne incognita, Meloidogyne incognita acrita, Meloidogyne javanica, Meloidogyne kikuyensis, Meloidogyne minor, Meloidogyne naasi, Meloidogyne paranaensis, Meloidogyne thamesi and the sedentary parasites Meloidogyne spp. in general, Meloinema spp., Nacobbus aberrans, Neotylenchus vigissi, Paraphelenchus pseudoparietinus, Paratrichodorus allius, Paratrichodorus lobatus, Paratrichodorus minor, Paratrichodorus nanus, Paratrichodorus porosus, Paratrichodorus teres and Paratrichodorus spp. in general, Paratylenchus hamatus, Paratylenchus minutus, Paratylenchus projectus and Paratylenchus spp. in general, Pratylenchus agilis, Pratylenchus alleni, Pratylenchus andinus, Pratylenchus brachyurus, Pratylenchus cerealis, Pratylenchus coffeae, Pratylenchus crenatus, Pratylenchus delattrei, Pratylenchus giibbicaudatus, Pratylenchus goodeyi, Pratylenchus hamatus, Pratylenchus hexincisus, Pratylenchus loosi, Pratylenchus neglectus, Pratylenchus penetrans, Pratylenchus pratensis, Pratylenchus scribneri, Pratylenchus teres, Pratylenchus thornei, Pratylenchus vulnus, Pratylenchus zeae and the migratory endoparasites Pratylenchus spp. in general, Pseudohalenchus minutus, Psilenchus magnidens, Psilenchus tumidus, Punctodera chalcoensis, Quinisulcius acutus, Radopholus citrophilus, Radopholus similis, the migratory endoparasites Radopholus spp. in general, Rotylenchulus borealis, Rotylenchulus parvus, Rotylenchulus reniformis and Rotylenchulus spp. in general, Rotylenchus laurentinus, Rotylenchus macrodoratus, Rotylenchus robustus, Rotylenchus uniformis and Rotylenchus spp. in general, Scutellonema brachyurum, Scutellonema bradys, Scutellonema clathricaudatum and the migratory endoparasites Scutellonema spp. in general, Subanguina radiciola, Tetylenchus nicotianae, Trichodorus cylindricus, Trichodorus minor, Trichodorus primitivus, Trichodorus proximus, Trichodorus similis, Trichodorus sparsus and the ectoparasites Trichodorus spp. in general, Tylenchorhynchus agri, Tylenchorhynchus brassicae, Tylenchorhynchus clarus, Tylenchorhynchus claytoni, Tylenchorhynchus digitatus, Tylenchorhynchus ebriensis, Tylenchorhynchus maximus, Tylenchorhynchus nudus, Tylenchorhynchus vulgaris and Tylenchorhynchus spp. in general, Tylenchulus semipenetrans and the semiparasites Tylenchulus spp. in general, Xiphinema americanum, Xiphinema brevicolle, Xiphinema dimorphicaudatum, Xiphinema index and the ectoparasites Xiphinema spp. in general.


Other examples of nematode pests include species belonging to the family Criconematidae, Belonolaimidae, Hoploaimidae, Heteroderidae, Longidoridae, Pratylenchidae, Trichodoridae, or Anguinidae.


Table 9 shows further examples of nematodes, and diseases associated therewith, that can be treated or prevented using the PMP compositions and related methods described herein.









TABLE 9







Nematode Pests








Disease
Causative Agent





Awl

Dolichoderus spp., D. heterocephalus



Bulb and stem (Europe)

Ditylenchus dipsaci



Burrowing

Radopholus similes R. similis



Cyst

Heterodera avenae, H. zeae, H.





schachti; Globodera





rostochiensis, G. pallida, and G.





tabacum; Heterodera trifolii, H.





medicaginis, H. ciceri, H.





mediterranea, H. cyperi, H.





salixophila, H. zeae, H. goettingiana, H.





riparia, H. humuli, H. latipons, H.





sorghi, H. fici, H. litoralis, and H.





turcomanica; Punctodera chalcoensis



Dagger

Xiphinema spp., X. americanum, X.





Mediterraneum



False root-knot

Nacobbus dorsalis



Lance

Hoplolaimus spp., H. galeatus



Lance, Columbia

Hoplolaimus Columbus



Lesion

Pratylenchus spp., P. brachyurus, P.





coffeae P. crenatus, P. hexincisus, P.





neglectus, P. penetrans, P. scribneri, P.





magnica, P. neglectus, P. thornei, P.





vulnus, P. zeae



Needle

Longidorus spp., L. breviannulatus



Others

Hirschmanniella species, Pratylenchoid





magnicauda



Ring

Criconemella spp., C. ornata



Root-knot

Meloidogyne spp., M. arenaria, M. chitwoodi,





M. artiellia, M. fallax, M. hapla,





M. javanica, M. incognita, M. microtyla,





M. partityla, M. panyuensis,





M, paranaensis



Spiral

Helicotylenchus spp.



Sting

Belonolaimus spp., B. longicaudatus



Stubby-root

Paratrichodorus spp., P. christiei,





P. minor, Quinisulcius acutus,





Trichodorus spp.



Stunt

Tylenchorhynchus dubius










vi. Viruses


The PMP compositions and related methods can be useful for decreasing the fitness of a virus, e.g., to prevent or treat a viral infection in a plant. Included are methods for delivering a PMP composition to a virus by contacting the virus with the PMP composition. Additionally or alternatively, the methods include delivering the PMP composition to a plant at risk of or having a viral infection, by contacting the plant with the PMP composition.


The PMP compositions and related methods are suitable for delivery to a virus that causes viral diseases in plants, including the viruses and diseases listed in Table 10.









TABLE 10







Viral Plant Pathogens








Disease
Causative Agent





Alfamoviruses:
Alfalfa mosaic alfamovirus


Bromoviridae


Alphacryptoviruses:
Alfalfa 1 alphacryptovirus, Beet 1 alphacryptovirus, Beet 2


Partitiviridae
alphacryptovirus, Beet 3 alphacryptovirus, Carnation 1



alphacryptovirus, Carrot temperate 1 alphacryptovirus, Carrot



temperate 3 alphacryptovirus, Carrot temperate 4 alphacryptovirus,



Cocksfoot alphacryptovirus, Hop trefoil 1 alphacryptovirus, Hop



trefoil 3 alphacryptovirus, Radish yellow edge alphacryptovirus,



Ryegrass alphacryptovirus, Spinach temperate alphacryptovirus,



Vicia alphacryptovirus, White clover 1 alphacryptovirus, White



clover 3 alphacryptovirus


Badnaviruses
Banana streak badnavirus, Cacao swollen shoot badnavirus, Canna



yellow mottle badnavirus, Commelina yellow mottle badnavirus,




Dioscorea bacilliform badnavirus, Kalanchoe top-spotting




badnavirus, Rice tungro bacilliform badnavirus, Schefflera ringspot



badnavirus, Sugarcane bacilliform badnavirus


Betacryptoviruses:
Carrot temperate 2 betacryptovirus, Hop trefoil 2 betacryptovirus,


Partitiviridae
Red clover 2 betacryptovirus, White clover 2 betacryptovirus


Bigeminiviruses:
Abutilon mosaic bigeminivirus, Ageratum yellow vein


Geminiviridae
bigeminivirus, Bean calico mosaic bigeminivirus, Bean golden



mosaic bigeminivirus, Bhendi yellow vein mosaic bigeminivirus,



Cassava African mosaic bigeminivirus, Cassava Indian mosaic



bigeminivirus, Chino del tomate bigeminivirus, Cotton leaf crumple



bigeminivirus, Cotton leaf curl bigeminivirus, Croton yellow vein



mosaic bigeminivirus, Dolichos yellow mosaic bigeminivirus,



Euphorbia mosaic bigeminivirus, Horsegram yellow mosaic



bigeminivirus, Jatropha mosaic bigeminivirus, Lima bean golden



mosaic bigeminivirus, Melon leaf curl bigeminivirus, Mung bean



yellow mosaic bigeminivirus, Okra leaf-curl bigeminivirus, Pepper



hausteco bigeminivirus, Pepper Texas bigeminivirus, Potato yellow



mosaic bigeminivirus, Rhynchosia mosaic bigeminivirus, Serrano



golden mosaic bigeminivirus, Squash leaf curl bigeminivirus,



Tobacco leaf curl bigeminivirus, Tomato Australian leafcurl



bigeminivirus, Tomato golden mosaic bigeminivirus, Tomato



Indian leafcurl bigeminivirus, Tomato leaf crumple bigeminivirus,



Tomato mottle bigeminivirus, Tomato yellow leaf curl



bigeminivirus, Tomato yellow mosaic bigeminivirus, Watermelon



chlorotic stunt bigeminivirus, Watermelon curly mottle



bigeminivirus


Bromoviruses:
Broad bean mottle bromovirus, Brome mosaic bromovirus, Cassia


Bromoviridae
yellow blotch bromovirus, Cowpea chlorotic mottle bromovirus,



Melandrium yellow fleck bromovirus, Spring beauty latent



bromovirus


Bymoviruses:
Barley mild mosaic bymovirus, Barley yellow mosaic bymovirus,


Potyviridae
Oat mosaic bymovirus, Rice necrosis mosaic bymovirus, Wheat



spindle streak mosaic bymovirus, Wheat yellow mosaic bymovirus


Capilloviruses
Apple stem grooving capillovirus, Cherry A capillovirus, Citrus



tatter leaf capillovirus, Lilac chlorotic leafspot capillovirus


Carlaviruses
Blueberry scorch carlavirus, Cactus 2 carlavirus, Caper latent



carlavirus, Carnation latent carlavirus, Chrysanthemum B



carlavirus, Dandelion latent carlavirus, Elderberry carlavirus, Fig S



carlavirus, Helenium S carlavirus, Honeysuckle latent carlavirus,



Hop American latent carlavirus, Hop latent carlavirus, Hop mosaic



carlavirus, Kalanchoe latent carlavirus, Lilac mottle carlavirus, Lily



symptomless carlavirus, Mulberry latent carlavirus, Muskmelon



vein necrosis carlavirus, Nerine latent carlavirus, Passiflora latent



carlavirus, Pea streak carlavirus, Poplar mosaic carlavirus, Potato M



carlavirus, Potato S carlavirus, Red clover vein mosaic carlavirus,



Shallot latent carlavirus, Strawberry pseudo mild yellow edge



carlavirus


Carmoviruses:
Bean mild mosaic carmovirus, Cardamine chlorotic fleck


Tombusviridae
carmovirus, Carnation mottle carmovirus, Cucumber leaf spot



carmovirus, Cucumber soil-borne carmovirus, Galinsoga mosaic



carmovirus, Hibiscus chlorotic ringspot carmovirus, Melon necrotic



spot carmovirus, Pelargonium flower break carmovirus, Turnip



crinkle carmovirus


Caulimoviruses
Blueberry red ringspot caulimovirus, Carnation etched ring



caulimovirus, Cauliflower mosaic caulimovirus, Dahlia mosaic



caulimovirus, Figwort mosaic caulimovirus, Horseradish latent



caulimovirus, Mirabilis mosaic caulimovirus, Peanut chlorotic



streak caulimovirus, Soybean chlorotic mottle caulimovirus, Sweet



potato caulimovirus, Thistle mottle caulimovirus


Closteroviruses
Beet yellow stunt closterovirus, Beet yellows closterovirus, Broad



bean severe chlorosis closterovirus, Burdock yellows closterovirus,



Carnation necrotic fleck closterovirus, Citrus tristeza closterovirus,



Clover yellows closterovirus, Grapevine stem pitting associated



closterovirus, Wheat yellow leaf closterovirus


Comoviruses:
Bean pod mottle comovirus, Bean rugose mosaic comovirus, Broad


Comoviridae
bean stain comovirus, Broad bean true mosaic comovirus, Cowpea



mosaic comovirus, Cowpea severe mosaic comovirus, Glycine



mosaic comovirus, Pea mild mosaic comovirus, Potato Andean



mottle comovirus, Quail pea mosaic comovirus, Radish mosaic



comovirus, Red clover mottle comovirus, Squash mosaic



comovirus, Ullucus C comovirus


Cucumoviruses:
Cucumber mosaic cucuamovirus, Peanut stunt cucumovirus, Tomato


Bromoviridae
aspermy cucumovirus


Cytorhabdoviruses:
Barley yellow striate mosaic cytorhabdovirus, Broad bean yellow


Rhabdoviridae
vein cytorhabdovirus, Broccoli necrotic yellows cytorhabdovirus,



Cereal northern mosaic cytorhabdovirus, Festuca leaf streak



cytorhabdovirus, Lettuce necrotic yellows cytorhabdovirus,



Sonchus cytorhabdovirus, Strawberry crinkle cytorhabdovirus


Dianthoviruses
Carnation ringspot dianthovirus, Red clover necrotic mosaic



dianthovirus, Sweet clover necrotic mosaic dianthovirus


Enamoviruses
Pea enation mosaic enamovirus


Fijiviruses:
Maize rough dwarf fijivirus, Oat sterile dwarf fijivirus, Pangola


Reoviridae
stunt fijivirus, Rice black-streaked dwarf fijivirus, Sugarcane Fiji



disease fijivirus


Furoviruses
Beet necrotic yellow vein furovirus, Beet soil-borne furovirus,



Broad bean necrosis furovirus, Oat golden stripe furovirus, Peanut



clump furovirus, Potato mop-top furovirus, Sorghum chlorotic spot



furovirus, Wheat soil-borne mosaic furovirus


Hordeiviruses

Anthoxanthum latent blanching hordeivirus, Barley stripe mosaic




hordeivirus, Lychnis ringspot hordeivirus, Poa semilatent



Hordeivirus


Hybrigeminiviruses:
Beet curly top hybrigeminivirus, Tomato pseudo curly top


Geminiviridae
hybrigeminivirus


Idaeoviruses
Raspberry bushy dwarf idaeovirus


Ilarviruses:
Apple mosaic ilarvirus, Asparagus 2 ilarvirus, Blueberry necrotic


Bromoviridae
shock ilarvirus, Citrus leaf rugose ilarvirus, Citrus variegation



ilarvirus, Elm mottle ilarvirus, Humulus japonicus ilarvirus,



Hydrangea mosaic ilarvirus, Lilac ring mottle ilarvirus, Parietaria



mottle ilarvirus, Plum American line pattern ilarvirus, Prune dwarf



ilarvirus, Prunus necrotic ringspot ilarvirus, Spinach latent ilarvirus,



Tobacco streak ilarvirus, Tulare apple mosaic ilarvirus


Ipomoviruses:
Sweet potato mild mottle ipomovirus, Sweet potato yellow dwarf


Potyviridae
ipomovirus


Luteoviruses
Barley yellow dwarf luteovirus, Bean leaf roll luteovirus, Beet mild



yellowing luteovirus, Beet western yellows luteovirus, Carrot red



leaf luteovirus, Groundnut rosette assistor luteovirus, Potato leafroll



luteovirus, Solanum yellows luteovirus, Soybean dwarf luteovirus,



Soybean Indonesian dwarf luteovirus, Strawberry mild yellow edge



luteovirus, Subterranean clover red leaf luteovirus, Tobacco



necrotic dwarf luteovirus


Machlomoviruses
Maize chlorotic mottle machlomovirus


Macluraviruses
Maclura mosaic macluravirus, Narcissus latent macluravirus


Marafiviruses
Bermuda grass etched-line marafivirus, Maize rayado fino



marafivirus, Oat blue dwarf marafivirus


Monogeminiviruses:
Chloris striate mosaic monogeminivirus, Digitaria striate mosaic


Geminiviridae
monogeminivirus, Digitaria streak monogeminivirus, Maize streak



monogeminivirus, Miscanthus streak monogeminivirus, Panicum



streak monogeminivirus, Paspalum striate mosaic



monogeminivirus, Sugarcane streak monogeminivirus, Tobacco



yellow dwarf monogeminivirus, Wheat dwarf monogeminivirus


Nanaviruses
Banana bunchy top nanavirus, Coconut foliar decay nanavirus,



Faba bean necrotic yellows nanavirus, Milk vetch dwarf nanavirus,



Subterranean clover stunt nanavirus


Necroviruses
Tobacco necrosis necrovirus, Carnation yellow stripe necrovirus,




Lisianthus necrosis necrovirus



Nepoviruses:
Arabis mosaic nepovirus, Arracacha A nepovirus, Artichoke Italian


Comoviridae
latent nepovirus, Artichoke yellow ringspot nepovirus, Blueberry



leaf mottle nepovirus, Cacao necrosis nepovirus, Cassava green



mottle nepovirus, Cherry leaf roll nepovirus, Cherry rasp leaf



nepovirus, Chicory yellow mottle nepovirus, Crimson clover latent



nepovirus, Cycas necrotic stunt nepovirus, Grapevine Bulgarian



latent nepovirus, Grapevine chrome mosaic nepovirus, Grapevine



fanleaf nepovirus, Hibiscus latent ringspot nepovirus, Lucerne



Australian latent nepovirus, Mulberry ringspot nepovirus,



Myrobalan latent ringspot nepovirus, Olive latent ringspot



nepovirus, Peach rosette mosaic nepovirus, Potato black ringspot



nepovirus, Potato U nepovirus, Raspberry ringspot nepovirus,



Tobacco ringspot nepovirus, Tomato black ring nepovirus, Tomato



ringspot nepovirus


Nucleorhabdoviruses:
Carrot latent nucleorhabdovirus, Coriander feathery red vein


Rhabdoviridae
nucleorhabdovirus, Cow parsnip mosaic nucleorhabdovirus,



Cynodon chlorotic streak nucleorhabdovirus, Datura yellow vein



nucleorhabdovirus, Eggplant mottled dwarf nucleorhabdovirus,



Maize mosaic nucleorhabdovirus, Pittosporum vein yellowing



nucleorhabdovirus, Potato yellow dwarf nucleorhabdovirus,



Sonchus yellow net nucleorhabdovirus, Sowthistle yellow vein



nucleorhabdovirus, Tomato vein clearing nucleorhabdovirus, Wheat



American striate mosaic nucleorhabdovirus


Oryzaviruses:
Echinochloa ragged stunt oryzavirus, Rice ragged stunt oryzavirus


Reoviridae


Ourmiaviruses
Cassava Ivorian bacilliform ourmiavirus, Epirus cherry



ourmiavirus, Melon Ourmia ourmiavirus, Pelargonium zonate spot



ourmiavirus


Phytoreoviruses:
Clover wound tumor phytoreovirus, Rice dwarf phytoreovirus, Rice


Reoviridae
gall dwarf phytoreovirus, Rice bunchy stunt phytoreovirus, Sweet



potato phytoreovirus


Potexviruses
Asparagus 3 potexvirus, Cactus × potexvirus, Cassava ×



potexvirus, Chicory × potexvirus, Clover yellow mosaic



potexvirus, Commelina × potexvirus, Cymbidium mosaic



potexvirus, Daphne × potexvirus, Foxtail mosaic potexvirus,



Hydrangea ringspot potexvirus, Lily × potexvirus, Narcissus



mosaic potexvirus, Nerine × potexvirus, Papaya mosaic potexvirus,



Pepino mosaic potexvirus, Plantago asiatica mosaic potexvirus,



Plantain × potexvirus, Potato aucuba mosaic potexvirus, Potato ×



potexvirus, Tulip × potexvirus, Viola mottle potexvirus, White



clover mosaic potexvirus


Potyviruses:
Alstroemeria mosaic potyvirus, Amaranthus leaf mottle potyvirus,


Potyviridae
Araujia mosaic potyvirus, Arracacha Y potyvirus, Artichoke latent



potyvirus, Asparagus 1 potyvirus, Banana bract mosaic potyvirus,



Bean common mosaic necrosis potyvirus, Bean common mosaic



potyvirus, Bean yellow mosaic potyvirus, Beet mosaic potyvirus,



Bidens mosaic potyvirus, Bidens mottle potyvirus, Cardamom



mosaic potyvirus, Carnation vein mottle potyvirus, Carrot thin leaf



potyyirus, Cassava brown streak potyvirus, Cassia yellow spot



potyvirus, Celery mosaic potyvirus, Chickpea bushy dwarf



potyvirus, Chickpea distortion mosaic potyvirus, Clover yellow



vein potyvirus, Commelina diffusa potyvirus, Commelina mosaic



potyvirus, Cowpea green vein-banding potyvirus, Cowpea



Moroccan aphid-borne mosaic potyvirus, Cowpea rugose mosaic



potyvirus, Crinum mosaic potyvirus, Daphne Y potyvirus, Dasheen



mosaic potyvirus, Datura Colombian potyvirus, Datura distortion



mosaic potyvirus, Datura necrosis potyvirus, Datura shoestring



potyvirus, Dendrobium mosaic potyvirus, Desmodium mosaic



potyvirus, Dioscorea alata potyvirus, Dioscorea green banding



mosaic potyvirus, Eggplant green mosaic potyvirus, Euphorbia



ringspot potyvirus, Freesia mosaic potyvirus, Groundnut eyespot



potyvirus, Guar symptomless potyvirus, Guinea grass mosaic



potyvirus, Helenium Y potyvirus, Henbane mosaic potyvirus,



Hippeastrum mosaic potyvirus, Hyacinth mosaic potyvirus, Iris



fulva mosaic potyvirus, Iris mild mosaic potyvirus, Iris severe



mosaic potyvirus, Johnsongrass mosaic potyvirus, Kennedya Y



potyvirus, Leek yellow stripe potyvirus, Lettuce mosaic potyvirus,



Lily mottle potyvirus, Maize dwarf mosaic potyvirus, Malva vein



clearing potyvirus, Marigold mottle potyvirus, Narcissus yellow



stripe potyvirus, Nerine potyvirus, Onion yellow dwarf potyvirus,



Ornithogalum mosaic potyvirus, Papaya ringspot potyvirus, Parsnip



mosaic potyvirus, Passiflora ringspot potyvirus, Passiflora South



African potyvirus, Passionfruit woodiness potyvirus, Patchouli



mosaic potyvirus, Pea mosaic potyvirus, Pea seed-borne mosaic



potyvirus, Peanut green mosaic potyvirus, Peanut mottle potyvirus,



Pepper Indian mottle potyvirus, Pepper mottle potyvirus, Pepper



severe mosaic potyvirus, Pepper veinal mottle potyvirus, Plum pox



potyvirus, Pokeweed mosaic potyvirus, Potato A potyvirus, Potato



V potyvirus, Potato Y potyvirus, Primula mosaic potyvirus,



Ranunculus mottle potyvirus, Sorghum mosaic potyvirus, Soybean



mosaic potyvirus, Statice Y potyvirus, Sugarcane mosaic potyvirus,



Sweet potato feathery mottle potyvirus, Sweet potato G potyvirus,



Swordbean distortion mosaic potyvirus, Tamarillo mosaic



potyvirus, Telfairia mosaic potyvirus, Tobacco etch potyvirus,



Tobacco vein-banding mosaic potyvirus, Tobacco vein mottling



potyvirus, Tobacco wilt potyvirus, Tomato Peru potyvirus,



Tradescantia-Zebrina potyvirus, Tropaeolum 1 potyvirus,



Tropaeolum 2 potyvirus, Tuberose potyvirus, Tulip band-breaking



potyvirus, Tulip breaking potyvirus, Tulip chlorotic blotch



potyvirus, Turnip mosaic potyvirus, Ullucus mosaic potyvirus,



Vallota mosaic potyvirus, Vanilla mosaic potyvirus, Vanilla



necrosis potyvirus, Voandzeia distortion mosaic potyvirus,



Watermelon mosaic 1 potyvirus, Watermelon mosaic 2 potyvirus,



Wild potato mosaic potyvirus, Wisteria vein mosaic potyvirus, Yam



mosaic potyvirus, Zucchini yellow fleck potyvirus, Zucchini yellow



mosaic potyvirus


Rymoviruses:
Hordeum mosaic rymovirus, Oat necrotic mottle


Potyviridae


Agropyron mosaic


rymovirus
rymovirus, Ryegrass mosaic rymovirus, Wheat streak mosaic



rymovirus


Satellite RNAs
Arabis mosaic satellite RNA, Chicory yellow mottle satellite RNA,



Cucumber mosaic satellite RNA, Grapevine fanleaf satellite RNA,



Strawberry latent ringspot satellite RNA, Tobacco ringspot satellite



RNA, Tomato black ring satellite RNA, Velvet tobacco mottle



satellite RNA


Satelliviruses
Maize white line mosaic satellivirus, Panicum mosaic satellivirus,



Tobacco mosaic satellivirus, Tobacco necrosis satellivirus


Sequiviruses:
Dandelion yellow mosaic sequivirus, Parsnip yellow fleck


Sequiviridae
sequivirus


Sobemoviruses
Bean southern mosaic sobemovirus, Blueberry shoestring



sobemovirus, Cocksfoot mottle sobemovirus, Lucerne transient



streak sobemovirus, Rice yellow mottle sobemovirus, Rottboellia



yellow mottle sobemovirus, Solanum nodiflorum mottle



sobemovirus, Sowbane mosaic sobemovirus, Subterranean clover



mottle sobemovirus, Turnip rosette sobemovirus, Velvet tobacco



mottle, sobemovirus


Tenuiviruses
Maize stripe tenuivirus, Rice grassy stunt tenuivirus, Rice hoja



blanca tenuivirus, Rice stripe tenuivirus


Tobamoviruses
Cucumber green mottle mosaic tobamovirus, Frangipani mosaic



tobamovirus, Kyuri green mottle mosaic tobamovirus,



Odontoglossum ringspot tobamovirus, Paprika mild mottle



tobamovirus, Pepper mild mottle tobamovirus, Ribgrass mosaic



tobamovirus, Opuntia Sammons' tobamovirus, Sunn-hemp mosaic



tobamovirus, Tobacco mild green mosaic tobamovirus, Tobacco



mosaic tobamovirus, Tomato mosaic tobamovirus, Ullucus mild



mottle tobamovirus


Tobraviruses
Pea early browning tobravirus, Pepper ringspot tobravirus, Tobacco



rattle tobravirus


Tombusviruses:
Artichoke mottled crinkle tombusvirus, Carnation Italian ringspot


Tombusviridae
tombusvirus, Cucumber necrosis tombusvirus, Cymbidium ringspot



tombusvirus, Eggplant mottled crinkle tombusvirus, Grapevine



Algerian latent tombusvirus, Lato River tombusvirus, Neckar River



tombusvirus, Pelargonium leaf curl tombusvirus, Pepper Moroccan



tombusvirus, Petunia asteroid mosaic tombusvirus, Tomato bushy



stunt tombusvirus


Tospoviruses:
Impatiens necrotic spot tospovirus, Peanut yellow spot tospovirus,


Bunyaviridae
Tomato spotted wilt tospovirus


Trichoviruses
Apple chlorotic leaf spot trichovirus, Heracleum latent trichovirus,



Potato T trichovirus


Tymoviruses
Abelia latent tymovirus, Belladonna mottle tymovirus, Cacao



yellow mosaic tymovirus, Clitoria yellow vein tymovirus,



Desmodium yellow mottle tymovirus, Dulcamara mottle tymovirus,



Eggplant mosaic tymovirus, Erysimum latent tymovirus, Kennedya



yellow mosaic tymovirus, Melon rugose mosaic tymovirus, Okra



mosaic tymovirus, Ononis yellow mosaic tymovirus, Passionfruit



yellow mosaic tymovirus, Physalis mosaic tymovirus, Plantago



mottle tymovirus, Potato Andean latent tymovirus, Scrophularia



mottle tymovirus, Turnip yellow mosaic, tymovirus, Voandzeia



necrotic mosaic tymovirus, Wild cucumber mosaic tymovirus


Umbraviruses
Bean yellow vein banding umbravirus, Carrot mottle mimic



umbravirus, Carrot mottle umbravirus, Carrot mottle mimic



umbravirus, Groundnut rosette umbravirus, Lettuce speckles mottle



umbravirus, Tobacco mottle umbravirus


Varicosaviruses
Freesia leaf necrosis varicosavirus, Lettuce big-vein varicosavirus,



Tobacco stunt varicosavirus


Waikaviruses:
Anthriscus yellows waikavirus, Maize chlorotic dwarf waikavirus,


Sequiviridae
Rice tungro spherical waikavirus


Putative
Alsike clover vein mosaic virus, Alstroemeria streak potyvirus,


Ungrouped
Amaranthus mosaic potyvirus, Amazon lily mosaic potyvirus,


Viruses
Anthoxanthum mosaic potyvirus, Apple stem pitting virus,



Aquilegia potyvirus, Asclepias rhabdovirus, Atropa belladonna



rhabdovirus, Barley mosaic virus, Barley yellow streak mosaic



virus, Beet distortion mosaic virus, Beet leaf curl rhabdovirus, Beet



western yellows ST9-associated RNA virus, Black raspberry



necrosis virus, Bramble yellow mosaic potyvirus, Brinjal mild



mosaic potyvirus, Broad bean B virus, Broad bean V potyvirus,



Broad bean yellow ringspot virus, Bryonia mottle potyvirus,



Burdock mosaic virus, Burdock mottle virus, Callistephus chinensis



chlorosis rhabdovirus, Canary reed mosaic potyvirus, Canavalia



maritima mosaic potyvirus, Carnation rhabdovirus, Carrot mosaic



potyvirus, Cassava symptomless rhabdovirus, Cassia mosaic virus,



Cassia ringspot virus, Celery yellow mosaic potyvirus, Celery



yellow net virus, Cereal flame chlorosis virus, Chickpea filiform



potyvirus, Chilli veinal mottle potyvirus, Chrysanthemum spot



potyvirus, Chrysanthemum vein chlorosis rhabdovirus, Citrus



leprosis rhabdovirus, Citrus ringspot virus, Clover mild mosaic



virus, Cocksfoot streak potyvirus, Colocasia bobone disease



rhabdovirus, Cucumber toad-skin rhabdovirus, Cucumber vein



yellowing virus, Cypripedium calceolus potyvirus, Datura innoxia



Hungarian mosaic potyvirus, Dioscorea trifida potyvirus, Dock



mottling mosaic potyvirus, Dodonaea yellows-associated virus,



Eggplant severe mottle potyvirus, Euonymus fasciation



rhabdovirus, Euonymus rhabdovirus, Fern potyvirus, Fig potyvirus,



Gerbera symptomless rhabdovirus, Grapevine fleck virus,



Grapevine stunt virus, Guar top necrosis virus, Habenaria mosaic



potyvirus, Holcus lanatus yellowing rhabdovirus, Holcus streak



potyvirus, Iris germanica leaf stripe rhabdovirus, Iris Japanese



necrotic ring virus, Isachne mosaic potyvirus, Kalanchoe isometric



virus, Kenaf vein-clearing rhabdovirus, Launaea mosaic potyvirus,



Lupin yellow vein rhabdovirus, Maize eyespot virus, Maize line



virus, Maize mottle/chlorotic stunt virus, Maize white line mosaic



virus, Malvastrum mottle virus, Melilotus mosaic potyvirus, Melon



vein-banding mosaic potyvirus, Melothria mottle potyvirus,



Mimosa mosaic virus, Mung bean mottle potyvirus, Narcissus



degeneration potyvirus, Narcissus late season yellows potyvirus,



Nerine Y potyvirus, Nothoscordum mosaic potyvirus, Oak ringspot



virus, Orchid fleck rhabdovirus, Palm mosaic potyvirus, Parsley



green mottle potyvirus, Parsley rhabdovirus, Parsnip leafcurl virus,



Passionfruit Sri Lankan mottle potyvirus, Passionfruit vein-clearing



rhabdovirus, Patchouli mottle rhabdovirus, Pea stem necrosis virus,



Peanut top paralysis potyvirus, Peanut veinal chlorosis rhabdovirus,



Pecteilis mosaic potyvirus, Pepper mild mosaic potyvirus, Perilla



mottle potyvirus, Pigeonpea proliferation rhabdovirus, Pigeonpea



sterility mosaic virus, Plantain 7 potyvirus, Plantain mottle



rhabdovirus, Pleioblastus chino potyvirus, Poplar decline potyvirus,



Primula mottle potyvirus, Purple granadilla mosaic virus,



Ranunculus repens symptomless rhabdovirus, Rice yellow stunt



virus, Saintpaulia leaf necrosis rhabdovirus, Sambucus vein



clearing rhabdovirus, Sarracenia purpurea rhabdovirus, Shamrock



chlorotic ringspot potyvirus, Soybean mild mosaic virus, Soybean



rhabdovirus, Soybean spherical virus, Soybean yellow vein virus,



Soybean Z potyvirus, Strawberry latent C rhabdovirus, Strawberry



mottle virus, Strawberry pallidosis virus, Sunflower mosaic



potyvirus, Sweet potato latent potyvirus, Teasel mosaic potyvirus,



Thimbleberry ringspot virus, Tomato mild mottle potyvirus,



Trichosanthes mottle potyvirus, Tulip halo necrosis virus, Tulip



mosaic virus, Turnip vein-clearing virus, Urd bean leaf crinkle



virus, Vigna sinensis mosaic rhabdovirus, Watercress yellow spot



virus, Watermelon Moroccan mosaic potyvirus, Wheat chlorotic



spot rhabdovirus, White bryony potyvirus, Wineberry latent virus,



Zinnia mild mottle potyvirus, Zoysia mosaic potyvirus









C. Delivery to a Plant Symbiont


Provided herein are methods of delivering to a plant symbiont a PMP composition disclosed herein. Included are methods for delivering a PMP composition to a symbiont (e.g., a bacterial endosymbiont, a fungal endosymbiont, or an insect) by contacting the symbiont with a PMP composition. The methods can be useful for increasing the fitness of plant symbiont, e.g., a symbiont that is beneficial to the fitness of a plant. In some instances, plant symbiont may be treated with unloaded PMPs. In other instances, the PMPs include a heterologous functional agent, e.g., fertilizing agents.


As such, the methods can be used to increase the fitness of a plant symbiont. In one aspect, provided herein is a method of increasing the fitness of a symbiont, the method including delivering to the symbiont the PMP composition described herein (e.g., in an effective amount and for an effective duration) to increase the fitness of the symbiont relative to an untreated symbiont (e.g., a symbiont that has not been delivered the PMP composition).


In one aspect, provided herein is a method of increasing the fitness of a fungus (e.g., a fungal endosymbiont of a plant), wherein the method includes delivering to the endosymbiont a PMP composition including a plurality of PMPs (e.g., a PMP composition described herein). For example, the plant symbiont may be an endosymbiotic fungus, such as a fungus of the genus Aspergillaceae, Ceratobasidiaceae, Coniochaetaceae, Cordycipitaceae, Corticiaceae, Cystofilobasidiaceae, Davidiellaceae, Debaryomycetaceae, Dothioraceae, Erysiphaceae, Filobasidiaceae, Glomerellaceae, Hydnaceae, Hypocreaceae, Leptosphaeriaceae, Montagnulaceae, Mortierellaceae, Mycosphaerellaceae, Nectriaceae, Orbiliaceae, Phaeosphaeriaceae, Pleosporaceae, Pseudeurotiaceae, Rhizopodaceae, Sclerotiniaceae, Stereaceae, or Trichocomacea.


In another aspect, provided herein is a method of increasing the fitness of a bacterium (e.g., a bacterial endosymbiont of a plant), wherein the method includes delivering to the bacteria a PMP composition including a plurality of PMPs (e.g., a PMP composition described herein). For example, the plant symbiont may be an endosymbiotic bacteria, such as a bacterium of the genus Acetobacteraceae, Acidobacteriaceae, Acidothermaceae, Aerococcaceae, Alcaligenaceae, Alicyclobacillaceae, Alteromonadaceae, Anaerolineaceae, Aurantimonadaceae, Bacillaceae, Bacteriovoracaceae, Bdellovibrionaceae, Bradyrhizobiaceae, Brevibacteriaceae, Brucellaceae, Burkholderiaceae, Carboxydocellaceae, Caulobacteraceae, Cellulomonadaceae, Chitinophagaceae, Chromatiaceae, Chthoniobacteraceae, Chthonomonadaceae, Clostridiaceae, Comamonadaceae, Corynebacteriaceae, Coxiellaceae, Cryomorphaceae, Cyclobacteriaceae, Cytophagaceae, Deinococcaceae, Dermabacteraceae, Dermacoccaceae, Enterobacteriaceae, Enterococcaceae, Erythrobacteraceae, Fibrobacteraceae, Flammeovirgaceae, Flavobacteriaceae, Frankiaceae, Fusobacteriaceae, Gaiellaceae, Gemmatimonadaceae, Geodermatophilaceae, Gly corny cetaceae, Haliangiaceae, Halomonadaceae, Holosporaceae, Hyphomicrobiaceae, lamiaceae, Intrasporangiaceae, Kineosporiaceae, Koribacteraceae, Lachnospiraceae, Lactobacillaceae, Legionellaceae, Leptospiraceae, Leuconostocaceae, Methylobacteriaceae, Methylocystaceae, Methylophilaceae, Microbacteriaceae, Micrococcaceae, Micromonosporaceae, Moraxellaceae, Mycobacteriaceae, Mycoplasmataceae, Myxococcaceae, Nakamurellaceae, Neisseriaceae, Nitrosomonadaceae, Nocardiaceae, Nocardioidaceae, Oceanospirillaceae, Opitutaceae, Oxalobacteraceae, Paenibacillaceae, Parachlamydiaceae, Pasteurellaceae, Patulibacteraceae, Peptostreptococcaceae, Phyllobacteriaceae, Piscirickettsiaceae, Planctomycetaceae, Planococcaceae, Polyangiaceae, Porphyromonadaceae, Prevotellaceae, Promicromonosporaceae, Pseudomonadaceae, Pseudonocardiaceae, Rhizobiaceae, Rhodobacteraceae, Rhodospirillaceae, Roseiflexaceae, Rubrobacteriaceae, Sandaracinaceae, Sanguibacteraceae, Saprospiraceae, Segniliparaceae, Shewanellaceae, Sinobacteraceae, Solibacteraceae, Solimonadaceae, Solirubrobacteraceae, Sphingobacteriaceae, Sphingomonadaceae, Spiroplasmataceae, Sporichthyaceae, Sporolactobacillaceae, Staphylococcaceae, Streptococcaceae, Streptomycetaceae, Syntrophobacteraceae, Veillonellaceae, Verrucomicrobiaceae, Weeksellaceae, Xanthobacteraceae, or Xanthomonadaceae.


In yet another aspect, provided herein is a method of increasing the fitness of an insect (e.g., an insect symbiont of a plant), wherein the method includes delivering to the insect a PMP composition including a plurality of PMPs (e.g., a PMP composition described herein). In some instances, the insect is a plant pollinator. For example, the insect may be of the genus Hymenoptera or Diptera. In some instances, the insect of the genus Hymenoptera is a bee. In other instances, the insect of the genus Diptera is a fly.


In some instances, the increase in symbiont fitness may manifest as an improvement in the physiology of the symbiont (e.g., improved health or survival) as a consequence of administration of the PMP composition. In some instances, the fitness of an organism may be measured by one or more parameters, including, but not limited to, reproductive rate, lifespan, mobility, fecundity, body weight, metabolic rate or activity, or survival in comparison to a symbiont to which the PMP composition has not been delivered. For example, the methods or compositions provided herein may be effective to improve the overall health of the symbiont or to improve the overall survival of the symbiont in comparison to a symbiont organism to which the PMP composition has not been administered. In some instances, the improved survival of the symbiont is about 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or greater than 100% greater relative to a reference level (e.g., a level found in a symbiont that does not receive a PMP composition). In some instances, the methods and compositions are effective to increase symbiont reproduction (e.g., reproductive rate) in comparison to a symbiont organism to which the PMP composition has not been administered. In some instances, the methods and compositions are effective to increase other physiological parameters, such as mobility, body weight, life span, fecundity, or metabolic rate, by about 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or greater than 100% relative to a reference level (e.g., a level found in a symbiont that does not receive a PMP composition).


In some instances, the increase in symbiont fitness may manifest as an increase in the frequency or efficacy of a desired activity carried out by the symbiont (e.g., pollination, predation on pests, seed spreading, or breakdown of waste or organic material) in comparison to a symbiont organism to which the PMP composition has not been administered. In some instances, the methods or compositions provided herein may be effective to increase the frequency or efficacy of a desired activity carried out by the symbiont (e.g., pollination, predation on pests, seed spreading, or breakdown of waste or organic material) by about 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or greater than 100% relative to a reference level (e.g., a level found in a symbiont that does not receive a PMP composition).


In some instances, the increase in symbiont fitness may manifest as an increase in the production of one or more nutrients in the symbiont (e.g., vitamins, carbohydrates, amino acids, or polypeptides) in comparison to a symbiont organism to which the PMP composition has not been administered. In some instances, the methods or compositions provided herein may be effective to increase the production of nutrients in the symbiont (e.g., vitamins, carbohydrates, amino acids, or polypeptides) by about 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or greater than 100% relative to a reference level (e.g., a level found in a symbiont that does not receive a PMP composition). In some instances, the methods or compositions provided herein may increase nutrients in an associated plant by increasing the production or metabolism of nutrients by one or more microorganisms (e.g., endosymbiont) in the symbiont.


In some instances, the increase in symbiont fitness may manifest as a decrease in the symbiont's sensitivity to a pesticidal agent and/or an increase in the symbiont's resistance to a pesticidal agent in comparison to a symbiont organism to which the PMP composition has not been administered. In some instances, the methods or compositions provided herein may be effective to decrease the symbiont's sensitivity to a pesticidal agent by about 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or greater than 100% relative to a reference level (e.g., a level found in a symbiont that does not receive a PMP composition).


In some instances, the increase in symbiont fitness may manifest as a decrease in the symbiont's sensitivity to an allelochemical agent and/or an increase in the symbiont's resistance to an allelochemical agent in comparison to a symbiont organism to which the PMP composition has not been administered. In some instances, the methods or compositions provided herein may be effective to increase the symbiont's resistance to an allelochemical agent by about 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or greater than 100% relative to a reference level (e.g., a level found in a symbiont that does not receive a PMP composition). In some instances, the allelochemical agent is caffeine, soyacystatin N, monoterpenes, diterpene acids, or phenolic compounds. In some instances, the methods or compositions provided herein may decrease the symbiont's sensitivity to an allelochemical agent by increasing the symbiont's ability to metabolize or degrade the allelochemical agent into usable substrates.


In some instances, the methods or compositions provided herein may be effective to increase the symbiont's resistance to parasites or pathogens (e.g., fungal, bacterial, or viral pathogens; or parasitic mites (e.g., Varroa destructor mite in honeybees)) in comparison to a symbiont organism to which the PMP composition has not been administered. In some instances, the methods or compositions provided herein may be effective to increase the symbiont's resistance to a pathogen or parasite (e.g., fungal, bacterial, or viral pathogens; or parasitic mites (e.g., Varroa destructor mite in honeybees)) by about 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or greater than 100% relative to a reference level (e.g., a level found in a symbiont that does not receive a PMP composition).


In some instances, the increase in symbiont fitness may manifest as other fitness advantages, such as improved tolerance to certain environmental factors (e.g., a high or low temperature tolerance), improved ability to survive in certain habitats, or an improved ability to sustain a certain diet (e.g., an improved ability to metabolize soy vs corn) in comparison to a symbiont organism to which the PMP composition has not been administered. In some instances, the methods or compositions provided herein may be effective to increase symbiont fitness in any plurality of ways described herein. Further, the PMP composition may increase symbiont fitness in any number of symbiont classes, orders, families, genera, or species (e.g., 1 symbiont species, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, 200, 250, 500, or more symbiont species). In some instances, the PMP composition acts on a single symbiont class, order, family, genus, or species.


Symbiont fitness may be evaluated using any standard methods in the art. In some instances, symbiont fitness may be evaluated by assessing an individual symbiont. Alternatively, symbiont fitness may be evaluated by assessing a symbiont population. For example, an increase in symbiont fitness may manifest as an increase in successful competition against other insects, thereby leading to an increase in the size of the symbiont population.


Examples of plant symbionts that can be treated with the present compositions or related methods are further described herein.


i. Fungi


The PMP compositions and related methods can be useful for increasing the fitness of a fungus, e.g., a fungus that is an endosymbiont of a plant (e.g., mycorrhizal fungus).


In some instances, the fungus is of the family Aspergillaceae, Ceratobasidiaceae, Coniochaetaceae, Cordycipitaceae, Corticiaceae, Cystofilobasidiaceae, Davidiellaceae, Debaryomycetaceae, Dothioraceae, Erysiphaceae, Filobasidiaceae, Glomerellaceae, Hydnaceae, Hypocreaceae, Leptosphaeriaceae, Montagnulaceae, Mortierellaceae, Mycosphaerellaceae, Nectriaceae, Orbiliaceae, Phaeosphaeriaceae, Pleosporaceae, Pseudeurotiaceae, Rhizopodaceae, Sclerotiniaceae, Stereaceae, or Trichocomacea.


In some instances, the fungus is a fungus having a mychorrhizal (e.g., ectomycorrhizal or endomycorrhizal) association with the roots of a plant, including fungi belonging to Glomeromycota, Basidiomycota, Ascomycota, or Zygomycota.


ii. Bacteria


The PMP compositions and related methods can be useful for increasing the fitness of a bacterium, e.g., a bacterium that is an endosymbiont of a plant (e.g., nitrogen-fixing bacteria).


For example, the bacterium may be of the genus Acidovorax, Agrobacterium, Bacillus, Burkholderia, Chryseobacterium, Curtobacterium, Enterobacter, Escherichia, Methylobacterium, Paenibacillus, Pantoea, Pseudomonas, Ralstonia, Rhizobium, Saccharibacillus, Sphingomonas, or Stenotrophomonas.


In some instances, the bacteria is of the family: Acetobacteraceae, Acidobacteriaceae, Acidothermaceae, Aerococcaceae, Alcaligenaceae, Alicyclobacillaceae, Alteromonadaceae, Anaerolineaceae, Aurantimonadaceae, Bacillaceae, Bacteriovoracaceae, Bdellovibrionaceae, Bradyrhizobiaceae, Brevibacteriaceae, Brucellaceae, Burkholderiaceae, Carboxydocellaceae, Caulobacteraceae, Cellulomonadaceae, Chitinophagaceae, Chromatiaceae, Chthoniobacteraceae, Chthonomonadaceae, Clostridiaceae, Comamonadaceae, Corynebacteriaceae, Coxiellaceae, Cryomorphaceae, Cyclobacteriaceae, Cytophagaceae, Deinococcaceae, Dermabacteraceae, Dermacoccaceae, Enterobacteriaceae, Enterococcaceae, Erythrobacteraceae, Fibrobacteraceae, Flammeovirgaceae, Flavobacteriaceae, Frankiaceae, Fusobacteriaceae, Gaiellaceae, Gemmatimonadaceae, Geodermatophilaceae, Gly corny cetaceae, Haliangiaceae, Halomonadaceae, Holosporaceae, Hyphomicrobiaceae, lamiaceae, Intrasporangiaceae, Kineosporiaceae, Koribacteraceae, Lachnospiraceae, Lactobacillaceae, Legionellaceae, Leptospiraceae, Leuconostocaceae, Methylobacteriaceae, Methylocystaceae, Methylophilaceae, Microbacteriaceae, Micrococcaceae, Micromonosporaceae, Moraxellaceae, Mycobacteriaceae, Mycoplasmataceae, Myxococcaceae, Nakamurellaceae, Neisseriaceae, Nitrosomonadaceae, Nocardiaceae, Nocardioidaceae, Oceanospirillaceae, Opitutaceae, Oxalobacteraceae, Paenibacillaceae, Parachlamydiaceae, Pasteurellaceae, Patulibacteraceae, Peptostreptococcaceae, Phyllobacteriaceae, Piscirickettsiaceae, Planctomycetaceae, Planococcaceae, Polyangiaceae, Porphyromonadaceae, Prevotellaceae, Promicromonosporaceae, Pseudomonadaceae, Pseudonocardiaceae, Rhizobiaceae, Rhodobacteraceae, Rhodospirillaceae, Roseiflexaceae, Rubrobacteriaceae, Sandaracinaceae, Sanguibacteraceae, Saprospiraceae, Segniliparaceae, Shewanellaceae, Sinobacteraceae, Solibacteraceae, Solimonadaceae, Solirubrobacteraceae, Sphingobacteriaceae, Sphingomonadaceae, Spiroplasmataceae, Sporichthyaceae, Sporolactobacillaceae, Staphylococcaceae, Streptococcaceae, Streptomycetaceae, Syntrophobacteraceae, Veillonellaceae, Verrucomicrobiaceae, Weeksellaceae, Xanthobacteraceae, or Xanthomonadaceae.


In some instances, the endosymbiotic bacterium is of a family selected from the group consisting of: Bacillaceae, Burkholderiaceae, Comamonadaceae, Enterobacteriaceae, Flavobacteriaceae, Methylobacteriaceae, Microbacteriaceae, Paenibacillileae, Pseudomonnaceae, Rhizobiaceae, Sphingomonadaceae, and Xanthomonadaceae.


In some instances, the endosymbiotic bacterium is of a genus selected from the group consisting of: Acidovorax, Agrobacterium, Bacillus, Burkholderia, Chryseobacterium, Curtobacterium, Enterobacter, Escherichia, Methylobacterium, Paenibacillus, Pantoea, Pseudomonas, Ralstonia, Saccharibacillus, Sphingomonas, and Stenotrophomonas.


iii. Insects


The PMP compositions and related methods can be useful for increasing the fitness of an insect, e.g., an insect that is beneficial to plant. The term insect includes any organism belonging to the phylum Arthropoda and to the class Insecta or the class Arachnida, in any stage of development, i.e., immature and adult insects. For example, the host may include insects that are used in agricultural applications, including insects that aid in the pollination of crops, spreading seeds, or pest control.


In some instances, the host aids in pollination of a plant (e.g., bees, beetles, wasps, flies, butterflies, or moths). In some instances, the host aiding in pollination of a plant is a bee. In some instances, the bee is in the family Andrenidae, Apidae, Colletidae, Halictidae, or Megachilidae. In some examples, the host aiding in pollination of a plant is beetle. In particular instances, the PMP composition may be used to increase the fitness of a honeybee.


In some instances, the host aiding in pollination of a plant is a beetle, e.g., a species in the family Buprestidae, Cantharidae, Cerambycidae, Chrysomelidae, Cleridae, Coccinellidae, Elateridae, Melandryidae, Meloidae, Melyridae, Mordellidae, Nitidulidae, Oedemeridae, Scarabaeidae, or Staphyllinidae.


In some instances, the host aiding in pollination of a plant is a butterfly or moth (e.g., Lepidoptera). In some instances, the butterfly or moth is a species in the family Geometridae, Hesperiidae, Lycaenidae, Noctuidae, Nymphalidae, Papilionidae, Pieridae, or Sphingidae.


In some instances, the host aiding in pollination of a plant is a fly (e.g., Diptera). In some instances, the fly is in the family Anthomyiidae, Bibionidae, Bombyliidae, Calliphoridae, Cecidomiidae, Certopogonidae, Chrionomidae, Conopidae, Culicidae, Dolichopodidae, Empididae, Ephydridae, Lonchopteridae, Muscidae, Mycetophilidae, Phoridae, Simuliidae, Stratiomyidae, or Syrphidae.


In some instances, the host aiding in pollination is an ant (e.g., Formicidae), sawfly (e.g., Tenthredinidae), or wasp (e.g., Sphecidae or Vespidae).


D. Delivery to an Animal Pathogen


Provided herein are methods of delivering a PMP composition (e.g., manufactured in accordance with the methods or bioreactors herein) to an animal (e.g., human) pathogen, such as one disclosed herein, by contacting the pathogen with a PMP composition. As used herein the term “pathogen” refers to an organism, such as a microorganism or an invertebrate, which causes disease or disease symptoms in an animal by, e.g., (i) directly infecting the animal, (ii) by producing agents that causes disease or disease symptoms in an animal (e.g., bacteria that produce pathogenic toxins and the like), and/or (iii) that elicit an immune (e.g., inflammatory response) in animals (e.g., biting insects, e.g., bedbugs). As used herein, pathogens include, but are not limited to bacteria, protozoa, parasites, fungi, nematodes, insects, viroids and viruses, or any combination thereof, wherein each pathogen is capable, either by itself or in concert with another pathogen, of eliciting disease or symptoms in animals, such as humans.


In some instances, animal (e.g., human) pathogen may be treated with unloaded PMPs. In other instances, the PMPs include a heterologous functional agent, e.g., a heterologous therapeutic agent (e.g., antibacterial agent, antifungal agent, insecticide, nematicide, antiparasitic agent, antiviral agent, or a repellent). The methods can be useful for decreasing the fitness of an animal pathogen, e.g., to prevent or treat a pathogen infection or control the spread of a pathogen as a consequence of delivery of the PMP composition.


Examples of pathogens that can be targeted in accordance with the methods described herein include bacteria (e.g., Streptococcus spp., Pneumococcus spp., Pseudomonas spp., Shigella spp, Salmonella spp., Campylobacter spp., or an Escherichia spp), fungi (Saccharomyces spp. or a Candida spp), parasitic insects (e.g., Cimex spp), parasitic nematodes (e.g., Heligmosomoides spp), or parasitic protozoa (e.g., Trichomoniasis spp).


For example, provided herein is a method of decreasing the fitness of a pathogen, the method including delivering to the pathogen a PMP composition described herein, wherein the method decreases the fitness of the pathogen relative to an untreated pathogen. In some embodiments, the method includes delivering the composition to at least one habitat where the pathogen grows, lives, reproduces, feeds, or infests. In some instances of the methods described herein, the composition is delivered as a pathogen comestible composition for ingestion by the pathogen. In some instances of the methods described herein, the composition is delivered (e.g., to a pathogen) as a liquid, a solid, an aerosol, a paste, a gel, or a gas.


Also provided herein is a method of decreasing the fitness of a parasitic insect, wherein the method includes delivering to the parasitic insect a PMP composition including a plurality of PMPs. In some instances, the method includes delivering to the parasitic insect a PMP composition including a plurality of PMPs, wherein the plurality of PMPs includes an insecticidal agent. For example, the parasitic insect may be a bedbug. Other non-limiting examples of parasitic insects are provided herein. In some instances, the method decreases the fitness of the parasitic insect relative to an untreated parasitic insect.


Additionally provided herein is a method of decreasing the fitness of a parasitic nematode, wherein the method includes delivering to the parasitic nematode a PMP composition including a plurality of PMPs. In some instances, the method includes delivering to the parasitic nematode a PMP composition including a plurality of PMPs, wherein the plurality of PMPs includes a nematicidal agent. For example, the parasitic nematode is Heligmosomoides polygyrus. Other non-limiting examples of parasitic nematodes are provided herein. In some instances, the method decreases the fitness of the parasitic nematode relative to an untreated parasitic nematode.


Further provided herein is a method of decreasing the fitness of a parasitic protozoan, wherein the method includes delivering to the parasitic protozoan a PMP composition including a plurality of PMPs. In some instances, the method includes delivering to the parasitic protozoan a PMP composition including a plurality of PMPs, wherein the plurality of PMPs includes an antiparasitic agent. For example, the parasitic protozoan may be T. vaginalis. Other non-limiting examples of parasitic protozoans are provided herein. In some instances, the method decreases the fitness of the parasitic protozoan relative to an untreated parasitic protozoan.


A decrease in the fitness of the pathogen as a consequence of delivery of a PMP composition can manifest in a number of ways. In some instances, the decrease in fitness of the pathogen may manifest as a deterioration or decline in the physiology of the pathogen (e.g., reduced health or survival) as a consequence of delivery of the PMP composition. In some instances, the fitness of an organism may be measured by one or more parameters, including, but not limited to, reproductive rate, fertility, lifespan, viability, mobility, fecundity, pathogen development, body weight, metabolic rate or activity, or survival in comparison to a pathogen to which the PMP composition has not been administered. For example, the methods or compositions provided herein may be effective to decrease the overall health of the pathogen or to decrease the overall survival of the pathogen. In some instances, the decreased survival of the pathogen is about 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or greater than 100% greater relative to a reference level (e.g., a level found in a pathogen that does not receive a PMP composition. In some instances, the methods and compositions are effective to decrease pathogen reproduction (e.g., reproductive rate, fertility) in comparison to a pathogen to which the PMP composition has not been administered. In some instances, the methods and compositions are effective to decrease other physiological parameters, such as mobility, body weight, life span, fecundity, or metabolic rate, by about 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or greater than 100% relative to a reference level (e.g., a level found in a pathogen that does not receive a PMP composition).


In some instances, the decrease in pest fitness may manifest as an increase in the pathogen's sensitivity to an antipathogen agent and/or a decrease in the pathogen's resistance to an antipathogen agent in comparison to a pathogen to which the PMP composition has not been delivered. In some instances, the methods or compositions provided herein may be effective to increase the pathogen's sensitivity to a pesticidal agent by about 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or greater than 100% relative to a reference level (e.g., a level found in a pest that does not receive a PMP composition).


In some instances, the decrease in pathogen fitness may manifest as other fitness disadvantages, such as a decreased tolerance to certain environmental factors (e.g., a high or low temperature tolerance), a decreased ability to survive in certain habitats, or a decreased ability to sustain a certain diet in comparison to a pathogen to which the PMP composition has not been delivered. In some instances, the methods or compositions provided herein may be effective to decrease pathogen fitness in any plurality of ways described herein. Further, the PMP composition may decrease pathogen fitness in any number of pathogen classes, orders, families, genera, or species (e.g., 1 pathogen species, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, 200, 250, 500, or more pathogen species). In some instances, the PMP composition acts on a single pest class, order, family, genus, or species.


Pathogen fitness may be evaluated using any standard methods in the art. In some instances, pest fitness may be evaluated by assessing an individual pathogen. Alternatively, pest fitness may be evaluated by assessing a pathogen population. For example, a decrease in pathogen fitness may manifest as a decrease in successful competition against other pathogens, thereby leading to a decrease in the size of the pathogen population.


The PMP compositions and related methods described herein are useful to decrease the fitness of an animal pathogen and thereby treat or prevent infections in animals. Examples of animal pathogens, or vectors thereof, that can be treated with the present compositions or related methods are further described herein.


i. Fungi


The PMP compositions and related methods can be useful for decreasing the fitness of a fungus, e.g., to prevent or treat a fungal infection in an animal. Included are methods for delivering a PMP composition to a fungus by contacting the fungus with the PMP composition. Additionally or alternatively, the methods include preventing or treating a fungal infection (e.g., caused by a fungus described herein) in an animal at risk of or in need thereof, by administering to the animal a PMP composition.


The PMP compositions and related methods are suitable for treatment or preventing of fungal infections in animals, including infections caused by fungi belonging to Ascomycota (Fusarium oxysporum, Pneumocystis jirovecii, Aspergillus spp., Coccidioides immitis/posadasii, Candida albicans), Basidiomycota (Filobasidiella neoformans, Trichosporon), Microsporidia (Encephalitozoon cuniculi, Enterocytozoon bieneusi), Mucoromycotina (Mucor circinelloides, Rhizopus oryzae, Lichtheimia corymbifera).


In some instances, the fungal infection is one caused by a belonging to the phylum Ascomycota, Basidomycota, Chytridiomycota, Microsporidia, or Zygomycota. The fungal infection or overgrowth can include one or more fungal species, e.g., Candida albicans, C. tropicalis, C. parapsilosis, C. glabrata, C. auris, C. krusei, Saccharomyces cerevisiae, Malassezia globose, M. restricta, or Debaryomyces hansenii, Gibberella moniliformis, Alternaria brassicicola, Cryptococcus neoformans, Pneumocystis carinii, P. jirovecii, P. murina, P. oryctolagi, P. wakefieldiae, and Aspergillus clavatus. The fungal species may be considered a pathogen or an opportunistic pathogen.


In some instances, the fungal infection is caused by a fungus in the genus Candida (i.e., a Candida infection). For example, a Candida infection can be caused by a fungus in the genus Candida that is selected from the group consisting of C. albicans, C. glabrata, C. dubliniensis, C. krusei, C. auris, C. parapsilosis, C. tropicalis, C. orthopsilosis, C. guilliermondii, C. rugose, and C. lusitaniae. Candida infections that can be treated by the methods disclosed herein include, but are not limited to candidemia, oropharyngeal candidiasis, esophageal candidiasis, mucosal candidiasis, genital candidiasis, vulvovaginal candidiasis, rectal candidiasis, hepatic candidiasis, renal candidiasis, pulmonary candidiasis, splenic candidiasis, otomycosis, osteomyelitis, septic arthritis, cardiovascular candidiasis (e.g., endocarditis), and invasive candidiasis.


ii. Bacteria


The PMP compositions and related methods can be useful for decreasing the fitness of a bacterium, e.g., to prevent or treat a bacterial infection in an animal. Included are methods for administering a PMP composition to a bacterium by contacting the bacteria with the PMP composition. Additionally or alternatively, the methods include preventing or treating a bacterial infection (e.g., caused by a bacterium described herein) in an animal at risk of or in need thereof, by administering to the animal a PMP composition.


The PMP compositions and related methods are suitable for preventing or treating a bacterial infection in animals caused by any bacteria described further below. For example, the bacteria may be one belonging to Bacillales (B. anthracis, B. cereus, S. aureus, L. monocytogenes), Lactobacillales (S. pneumoniae, S. pyogenes), Clostridiales (C. botulinum, C. difficile, C. perfringens, C. tetani), Spirochaetales (Borrelia burgdorferi, Treponema pallidum), Chlamydiales (Chlamydia trachomatis, Chlamydophila psittaci), Actinomycetales (C. diphtheriae, Mycobacterium tuberculosis, M. avium), Rickettsiales (R. prowazekii, R. rickettsii, R. typhi, A. phagocytophilum, E. chaffeensis), Rhizobiales (Brucella melitensis), Burkholderiales (Bordetella pertussis, Burkholderia mallei, B. pseudomallei), Neisseriales (Neisseria gonorrhoeae, N. meningitidis), Campylobacterales (Campylobacter jejuni, Helicobacter pylon), Legionellales (Legionella pneumophila), Pseudomonadales (A. baumannii, Moraxella catarrhalis, P. aeruginosa), Aeromonadales (Aeromonas sp.), Vibrionales (Vibrio cholerae, V. parahaemolyticus), Thiotrichales, Pasteurellales (Haemophilus influenzae), Enterobacteriales (Klebsiella pneumoniae, Proteus mirabilis, Yersinia pestis, Y. enterocolitica, Shigella flexneri, Salmonella enterica, E. coli).


iii. Parasitic Insects


The PMP compositions and related methods can be useful for decreasing the fitness of a parasitic insect, e.g., to prevent or treat a parasitic insect infection in an animal. The term “insect” includes any organism belonging to the phylum Arthropoda and to the class Insecta or the class Arachnida, in any stage of development, i.e., immature and adult insects. Included are methods for delivering a PMP composition to an insect by contacting the insect with the PMP composition. Additionally or alternatively, the methods include preventing or treating a parasitic insect infection (e.g., caused by a parasitic insect described herein) in an animal at risk of or in need thereof, by administering to the animal a PMP composition.


The PMP compositions and related methods are suitable for preventing or treating infection in animals by a parasitic insect, including infections by insects belonging to Phthiraptera: Anoplura (Sucking lice), Ischnocera (Chewing lice), Amblycera (Chewing lice). Siphonaptera: Pulicidae (Cat fleas), Ceratophyllidae (Chicken-fleas). Diptera: Culicidae (Mosquitoes), Ceratopogonidae (Midges), Psychodidae (Sandflies), Simuliidae (Blackflies), Tabanidae (Horse-flies), Muscidae (House-flies, etc.), Calliphoridae (Blowflies), Glossinidae (Tsetse-flies), Oestridae (Bot-flies), Hippoboscidae (Louse-flies). Hemiptera: Reduviidae (Assassin-bugs), Cimicidae (Bed-bugs). Arachnida: Sarcoptidae (Sarcoptic mites), Psoroptidae (Psoroptic mites), Cytoditidae (Air-sac mites), Laminosioptes (Cyst-mites), Analgidae (Feather-mites), Acaridae (Grain-mites), Demodicidae (Hair-follicle mites), Cheyletiellidae (Fur-mites), Trombiculidae (Trombiculids), Dermanyssidae (Bird mites), Macronyssidae (Bird mites), Argasidae (Soft-ticks), Ixodidae (Hard-ticks).


iv. Protozoa


The PMP compositions and related methods can be useful for decreasing the fitness of a parasitic protozoa, e.g., to prevent or treat a parasitic protozoa infection in an animal. The term “protozoa” includes any organism belonging to the phylum Protozoa. Included are methods for delivering a PMP composition to a parasitic protozoa by contacting the parasitic protozoa with the PMP composition. Additionally or alternatively, the methods include preventing or treating a protozoal infection (e.g., caused by a protozoan described herein) in an animal at risk of or in need thereof, by administering to the animal a PMP composition.


The PMP compositions and related methods are suitable for preventing or treating infection by parasitic protozoa in animals, including protozoa belonging to Euglenozoa (Trypanosoma cruzi, Trypanosoma brucei, Leishmania spp.), Heterolobosea (Naegleria fowleri), Diplomonadida (Giardia intestinalis), Amoebozoa (Acanthamoeba castellanii, Balamuthia mandrillaris, Entamoeba histolytica), Blastocystis (Blastocystis hominis), Apicomplexa (Babesia microti, Cryptosporidium parvum, Cyclospora cayetanensis, Plasmodium spp., Toxoplasma gondii).


v. Nematodes


The PMP compositions and related methods can be useful for decreasing the fitness of a parasitic nematode, e.g., to prevent or treat a parasitic nematode infection in an animal. Included are methods for delivering a PMP composition to a parasitic nematode by contacting the parasitic nematode with the PMP composition. Additionally or alternatively, the methods include preventing or treating a parasitic nematode infection (e.g., caused by a parasitic nematode described herein) in an animal at risk of or in need thereof, by administering to the animal a PMP composition.


The PMP compositions and related methods are suitable for preventing or treating infection by parasitic nematodes in animals, including nematodes belonging to Nematoda (roundworms): Angiostrongylus cantonensis (rat lungworm), Ascaris lumbricoides (human roundworm), Baylisascaris procyonis (raccoon roundworm), Trichuris trichiura (human whipworm), Trichinella spiralis, Strongyloides stercoralis, Wuchereria bancrofti, Brugia malayi, Ancylostoma duodenale and Necator americanus (human hookworms), Cestoda (tapeworms): Echinococcus granulosus, Echinococcus multilocularis, Taenia solium (pork tapeworm).


vi. Viruses


The PMP compositions and related methods can be useful for decreasing the fitness of a virus, e.g., to prevent or treat a viral infection in an animal. Included are methods for delivering a PMP composition to a virus by contacting the virus with the PMP composition. Additionally or alternatively, the methods include preventing or treating a viral infection (e.g., caused by a virus described herein) in an animal at risk of or in need thereof, by administering to the animal a PMP composition.


The PMP compositions and related methods are suitable for preventing or treating a viral infection in animals, including infections by viruses belonging to DNA viruses: Parvoviridae, Papillomaviridae, Polyomaviridae, Poxviridae, Herpesviridae; Single-stranded negative strand RNA viruses: Arenaviridae, Paramyxoviridae (Rubulavirus, Respirovirus, Pneumovirus, Moribillivirus), Filoviridae (Marburgvirus, Ebolavirus), Bornaoviridae, Rhabdoviridae, Orthomyxoviridae, Bunyaviridae, Nairovirus, Hantaviruses, Orthobunyavirus, Phlebovirus. Single-stranded positive strand RNA viruses: Astroviridae, Coronaviridae, Caliciviridae, Togaviridae (Rubivirus, Alphavirus), Flaviviridae (Hepacivirus, Flavivirus), Picornaviridae (Hepatovirus, Rhinovirus, Enterovirus); or dsRNA and Retro-transcribed Viruses: Reoviridae (Rotavirus, Coltivirus, Seadornavirus), Retroviridae (Deltaretrovirus, Lentivirus), Hepadnaviridae (Orthohepadnavirus).


E. Delivery to a Pathogen Vector


Provided herein are methods of delivering a PMP composition (e.g., manufactured in accordance with the methods or bioreactors herein) to pathogen vector, such as one disclosed herein, by contacting the pathogen vector with a PMP composition. As used herein, the term “vector” refers to an insect that can carry or transmit an animal pathogen from a reservoir to an animal. Exemplary vectors include insects, such as those with piercing-sucking mouthparts, as found in Hemiptera and some Hymenoptera and Diptera such as mosquitoes, bees, wasps, midges, lice, tsetse fly, fleas and ants, as well as members of the Arachnidae such as ticks and mites.


In some instances, the vector of the animal (e.g., human) pathogen may be treated with unloaded PMPs. In other instances, the PMPs include a heterologous functional agent, e.g., a heterologous therapeutic agent (e.g., antibacterial agent, antifungal agent, insecticide, nematicide, antiparasitic agent, antiviral agent, or a repellent). The methods can be useful for decreasing the fitness of a pathogen vector, e.g., to control the spread of a pathogen as a consequence of delivery of the PMP composition. Examples of pathogen vectors that can be targeted in accordance with the present methods include insects, such as those described herein.


For example, provided herein is a method of decreasing the fitness of an animal pathogen vector, the method including delivering to the vector an effective amount of the PMP compositions described herein, wherein the method decreases the fitness of the vector relative to an untreated vector. In some instances, the method includes delivering the composition to at least one habitat where the vector grows, lives, reproduces, feeds, or infests. In some instances, the composition is delivered as a comestible composition for ingestion by the vector. In some instances, the vector is an insect. In some instances, the insect is a mosquito, a tick, a mite, or a louse. In some instances, the composition is delivered (e.g., to the pathogen vector) as a liquid, a solid, an aerosol, a paste, a gel, or a gas.


For example, provided herein is a method of decreasing the fitness of an insect vector of an animal pathogen, wherein the method includes delivering to the vector a PMP composition including a plurality of PMPs. In some instances, the method includes delivering to the vector a PMP composition including a plurality of PMPs, wherein the plurality of PMPs includes an insecticidal agent. For example, the insect vector may be a mosquito, tick, mite, or louse. Other non-limiting examples of pathogen vectors are provided herein. In some instances, the method decreases the fitness of the vector relative to an untreated vector.


In some instances, the decrease in vector fitness may manifest as a deterioration or decline in the physiology of the vector (e.g., reduced health or survival) as a consequence of administration of a composition. In some instances, the fitness of an organism may be measured by one or more parameters, including, but not limited to, reproductive rate, lifespan, mobility, fecundity, body weight, metabolic rate or activity, or survival in comparison to a vector organism to which the composition has not been delivered. For example, the methods or compositions provided herein may be effective to decrease the overall health of the vector or to decrease the overall survival of the vector. In some instances, the decreased survival of the vector is about 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or greater than 100% greater relative to a reference level (e.g., a level found in a vector that does not receive a composition). In some instances, the methods and compositions are effective to decrease vector reproduction (e.g., reproductive rate) in comparison to a vector organism to which the composition has not been delivered. In some instances, the methods and compositions are effective to decrease other physiological parameters, such as mobility, body weight, life span, fecundity, or metabolic rate, by about 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or greater than 100% relative to a reference level (e.g., a level found in a vector that is not delivered the composition).


In some instances, the decrease in vector fitness may manifest as an increase in the vector's sensitivity to a pesticidal agent and/or a decrease in the vector's resistance to a pesticidal agent in comparison to a vector organism to which the composition has not been delivered. In some instances, the methods or compositions provided herein may be effective to increase the vector's sensitivity to a pesticidal agent by about 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or greater than 100% relative to a reference level (e.g., a level found in a vector that does not receive a composition). The pesticidal agent may be any pesticidal agent known in the art, including insecticidal agents. In some instances, the methods or compositions provided herein may increase the vector's sensitivity to a pesticidal agent by decreasing the vector's ability to metabolize or degrade the pesticidal agent into usable substrates in comparison to a vector to which the composition has not been delivered.


In some instances, the decrease in vector fitness may manifest as other fitness disadvantages, such as decreased tolerance to certain environmental factors (e.g., a high or low temperature tolerance), decreased ability to survive in certain habitats, or a decreased ability to sustain a certain diet in comparison to a vector organism to which the composition has not been delivered. In some instances, the methods or compositions provided herein may be effective to decrease vector fitness in any plurality of ways described herein. Further, the composition may decrease vector fitness in any number of vector classes, orders, families, genera, or species (e.g., 1 vector species, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, 200, 250, 500, or more vector species). In some instances, the composition acts on a single vector class, order, family, genus, or species.


Vector fitness may be evaluated using any standard methods in the art. In some instances, vector fitness may be evaluated by assessing an individual vector. Alternatively, vector fitness may be evaluated by assessing a vector population. For example, a decrease in vector fitness may manifest as a decrease in successful competition against other vectors, thereby leading to a decrease in the size of the vector population.


By decreasing the fitness of vectors that carry animal pathogens, the compositions provided herein are effective to reduce the spread of vector-borne diseases. The composition may be delivered to the insects using any of the formulations and delivery methods described herein, in an amount and for a duration effective to reduce transmission of the disease, e.g., reduce vertical or horizontal transmission between vectors and/or reduce transmission to animals. For example, the composition described herein may reduce vertical or horizontal transmission of a vector-borne pathogen by about 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or more in comparison to a vector organism to which the composition has not been delivered. As another example, the composition described herein may reduce vectorial competence of an insect vector by about 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or more in comparison to a vector organism to which the composition has not been delivered.


Non-limiting examples of diseases that may be controlled by the compositions and methods provided herein include diseases caused by Togaviridae viruses (e.g., Chikungunya, Ross River fever, Mayaro, Onyon-nyong fever, Sindbis fever, Eastern equine enchephalomyeltis, Wesetern equine encephalomyelitis, Venezualan equine encephalomyelitis, or Barmah forest); diseases caused by Flavivirdae viruses (e.g., Dengue fever, Yellow fever, Kyasanur Forest disease, Omsk haemorrhagic fever, Japaenese encephalitis, Murray Valley encephalitis, Rocio, St. Louis encephalitis, West Nile encephalitis, or Tick-borne encephalitis); diseases caused by Bunyaviridae viruses (e.g., Sandly fever, Rift Valley fever, La Crosse encephalitis, California encephalitis, Crimean-Congo haemorrhagic fever, or Oropouche fever); disease caused by Rhabdoviridae viruses (e.g., Vesicular stomatitis); disease caused by Orbiviridae (e.g., Bluetongue); diseases caused by bacteria (e.g., Plague, Tularaemia, Q fever, Rocky Mountain spotted fever, Murine typhus, Boutonneuse fever, Queensland tick typhus, Siberian tick typhus, Scrub typhus, Relapsing fever, or Lyme disease); or diseases caused by protozoa (e.g., Malaria, African trypanosomiasis, Nagana, Chagas disease, Leishmaniasis, Piroplasmosis, Bancroftian filariasis, or Brugian filariasis).


vii. Pathogen Vectors


The methods and compositions provided herein may be useful for decreasing the fitness of a vector for an animal pathogen. In some instances, the vector may be an insect. For example, the insect vector may include, but is not limited to those with piercing-sucking mouthparts, as found in Hemiptera and some Hymenoptera and Diptera such as mosquitoes, bees, wasps, midges, lice, tsetse fly, fleas and ants, as well as members of the Arachnidae such as ticks and mites; order, class or family of Acarina (ticks and mites) e.g. representatives of the families Argasidae, Dermanyssidae, Ixodidae, Psoroptidae or Sarcoptidae and representatives of the species Amblyomma spp., Anocenton spp., Argas spp., Boophilus spp., Cheyletiella spp., Chorioptes spp., Demodex spp., Dermacentor spp., Denmanyssus spp., Haemophysalis spp., Hyalomma spp., Ixodes spp., Lynxacarus spp., Mesostigmata spp., Notoednes spp., Ornithodoros spp., Ornithonyssus spp., Otobius spp., Otodectes spp., Pneumonyssus spp., Psoroptes spp., Rhipicephalus spp., Sancoptes spp., or Trombicula spp.; Anoplura (sucking and biting lice) e.g. representatives of the species Bovicola spp., Haematopinus spp., Linognathus spp., Menopon spp., Pediculus spp., Pemphigus spp., Phylloxera spp., or Solenopotes spp.; Diptera (flies) e.g. representatives of the species Aedes spp., Anopheles spp., Calliphora spp., Chrysomyia spp., Chrysops spp., Cochliomyia spp., Cw/ex spp., Culicoides spp., Cuterebra spp., Dermatobia spp., Gastrophilus spp., Glossina spp., Haematobia spp., Haematopota spp., Hippobosca spp., Hypoderma spp., Lucilia spp., Lyperosia spp., Melophagus spp., Oestrus spp., Phaenicia spp., Phlebotomus spp., Phormia spp., Acari (sarcoptic mange) e.g., Sarcoptidae spp., Sarcophaga spp., Simulium spp., Stomoxys spp., Tabanus spp., Tannia spp. or Zzpu/alpha spp.; Mallophaga (biting lice) e.g. representatives of the species Damalina spp., Felicola spp., Heterodoxus spp. or Trichodectes spp.; or Siphonaptera (wingless insects) e.g. representatives of the species Ceratophyllus spp., Xenopsylla spp; Cimicidae (true bugs) e.g. representatives of the species Cimex spp., Tritominae spp., Rhodinius spp., or Triatoma spp.


In some instances, the insect is a blood-sucking insect from the order Diptera (e.g., suborder Nematocera, e.g., family Colicidae). In some instances, the insect is from the subfamilies Culicinae, Corethrinae, Ceratopogonidae, or Simuliidae. In some instances, the insect is of a Culex spp., Theobaldia spp., Aedes spp., Anopheles spp., Aedes spp., Forciponiyia spp., Culicoides spp., or Helea spp.


In certain instances, the insect is a mosquito. In certain instances, the insect is a tick. In certain instances, the insect is a mite. In certain instances, the insect is a biting louse.


F. Application Methods


A plant described herein can be exposed to a PMP composition described herein in any suitable manner that permits delivering or administering the composition to the plant. The PMP composition may be delivered either alone or in combination with other active (e.g., fertilizing agents) or inactive substances and may be applied by, for example, spraying, injection (e.g., microinjection), through plants, pouring, dipping, in the form of concentrated liquids, gels, solutions, suspensions, sprays, powders, pellets, briquettes, bricks and the like, formulated to deliver an effective concentration of the PMP composition. Amounts and locations for application of the compositions described herein are generally determined by the habitat of the plant, the lifecycle stage at which the plant can be targeted by the PMP composition, the site where the application is to be made, and the physical and functional characteristics of the PMP composition.


In some instances, the composition is sprayed directly onto a plant e.g., crops, by e.g., backpack spraying, aerial spraying, crop spraying/dusting etc. In instances where the PMP composition is delivered to a plant, the plant receiving the PMP composition may be at any stage of plant growth. For example, formulated PMP compositions can be applied as a seed-coating or root treatment in early stages of plant growth or as a total plant treatment at later stages of the crop cycle. In some instances, the PMP composition may be applied as a topical agent to a plant.


Further, the PMP composition may be applied (e.g., in the soil in which a plant grows, or in the water that is used to water the plant) as a systemic agent that is absorbed and distributed through the tissues of a plant. In some instances, plants or food organisms may be genetically transformed to express the PMP composition.


Delayed or continuous release can also be accomplished by coating the PMP composition or a composition with the PMP composition(s) with a dissolvable or bioerodable coating layer, such as gelatin, which coating dissolves or erodes in the environment of use, to then make the PMP composition available, or by dispersing the agent in a dissolvable or erodable matrix. Such continuous release and/or dispensing devices may be advantageously employed to consistently maintain an effective concentration of one or more of the PMP compositions described herein.


In some instances, the PMP composition is delivered to a part of the plant, e.g., a leaf, seed, pollen, root, fruit, shoot, or flower, or a tissue, cell, or protoplast thereof. In some instances, the PMP composition is delivered to a cell of the plant. In some instances, the PMP composition is delivered to a protoplast of the plant. In some instances, the PMP composition is delivered to a tissue of the plant. For example, the composition may be delivered to meristematic tissue of the plant (e.g., apical meristem, lateral meristem, or intercalary meristem). In some instances, the composition is delivered to permanent tissue of the plant (e.g., simple tissues (e.g., parenchyma, collenchyma, or sclerenchyma) or complex permanent tissue (e.g., xylem or phloem)). In some instances, the composition is delivered to a plant embryo.


In some instances, the PMP composition may be recommended for field application as an amount of PMPs per hectare (g/ha or kg/ha) or the amount of active ingredient (e.g., PMP with or without a heterologous functional agent) or acid equivalent per hectare (kg a.i./ha or g a.i./ha). In some instances, a lower amount of heterologous functional agent in the present compositions may be required to be applied to soil, plant media, seeds plant tissue, or plants to achieve the same results as where the heterologous functional agent is applied in a composition lacking PMPs. For example, the amount of heterologous functional agent may be applied at levels about 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 50, or 100-fold (or any range between about 2 and about 100-fold, for example about 2- to 10-fold; about 5- to 15-fold, about 10- to 20-fold; about 10- to 50-fold) less than the same heterologous functional agent applied in a non-PMP composition, e.g., direct application of the same heterologous functional agent without PMPs. PMP compositions of the invention can be applied at a variety of amounts per hectare, for example at about 0.0001, 0.001, 0.005, 0.01, 0.1, 1, 2, 10, 100, 1,000, 2,000, 5,000 (or any range between about 0.0001 and 5,000) kg/ha. For example, about 0.0001 to about 0.01, about 0.01 to about 10, about 10 to about 1,000, about 1,000 to about 5,000 kg/ha.


G. Therapeutic Methods


The PMP compositions described herein are useful in a variety of therapeutic methods. For example, the methods and composition may be used for the prevention or treatment of pathogen infections in animals (e.g., humans); to treat or prevent a human disease or disorder; or to treat or prevent a disorder in agricultural animals (e.g., cows, steer, pigs, horses, or chickens) or in other veterinary species such as horses, dogs, or cats. As used herein, the term “treatment” refers to administering a pharmaceutical composition to an animal for prophylactic and/or therapeutic purposes. To “prevent” refers to prophylactic treatment of an animal who is not yet ill, but who is susceptible to, or otherwise at risk of, a particular disease. To “treat” refers to administering treatment to an animal already suffering from a disease to improve or stabilize the animal's condition. The present methods involve delivering the PMP compositions described herein to an animal, such as a human.


For example, provided herein is a method of treating an animal having a fungal infection, wherein the method includes administering to the animal an effective amount of a PMP composition including a plurality of PMPs. In some instances, the method includes administering to the animal an effective amount of a PMP composition including a plurality of PMPs, wherein the plurality of PMPs includes an antifungal agent. In some instances, the antifungal agent is a nucleic acid that inhibits expression of a gene in a fungus that causes the fungal infection (e.g., Enhanced Filamentous Growth Protein (EFG1)). In some instances, the fungal infection is caused by Candida albicans. In some instances, composition includes a PMP produced from an Arabidopsis apoplast EV. In some instances, the method decreases or substantially eliminates the fungal infection.


In another aspect, provided herein is a method of treating an animal having a bacterial infection, wherein the method includes administering to the animal an effective amount of a PMP composition including a plurality of PMPs. In some instances, the method includes administering to the animal an effective amount of a PMP composition including a plurality of PMPs, and wherein the plurality of PMPs includes an antibacterial agent (e.g., Amphotericin B). In some instances, the bacterium is a Streptococcus spp., Pneumococcus spp., Pseudamonas spp., Shigella spp, Salmonella spp., Campylobacter spp., or an Escherichia spp. In some instances, the composition includes a PMP produced from an Arabidopsis apoplast EV. In some instances, the method decreases or substantially eliminates the bacterial infection. In some instances, the animal is a human, a veterinary animal, or a livestock animal.


The present methods are useful to treat an infection (e.g., as caused by an animal pathogen) in an animal, which refers to administering treatment to an animal already suffering from a disease to improve or stabilize the animal's condition. This may involve reducing colonization of a pathogen in, on, or around an animal by one or more pathogens (e.g., by about 1%, 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100%) relative to a starting amount and/or allow benefit to the individual (e.g., reducing colonization in an amount sufficient to resolve symptoms). In such instances, a treated infection may manifest as a decrease in symptoms (e.g., by about 1%, 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100%). In some instances, a treated infection is effective to increase the likelihood of survival of an individual (e.g., an increase in likelihood of survival by about 1%, 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100%) or increase the overall survival of a population (e.g., an increase in likelihood of survival by about 1%, 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100%). For example, the compositions and methods may be effective to “substantially eliminate” an infection, which refers to a decrease in the infection in an amount sufficient to sustainably resolve symptoms (e.g., for at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 months) in the animal.


The present methods are useful to prevent an infection (e.g., as caused by an animal pathogen), which refers to preventing an increase in colonization in, on, or around an animal by one or more pathogens (e.g., by about 1%, 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or more than 100% relative to an untreated animal) in an amount sufficient to maintain an initial pathogen population (e.g., approximately the amount found in a healthy individual), prevent the onset of an infection, and/or prevent symptoms or conditions associated with infection. For example, individuals may receive prophylaxis treatment to prevent a fungal infection while being prepared for an invasive medical procedure (e.g., preparing for surgery, such as receiving a transplant, stem cell therapy, a graft, a prosthesis, receiving long-term or frequent intravenous catheterization, or receiving treatment in an intensive care unit), in immunocompromised individuals (e.g., individuals with cancer, with HIV/AIDS, or taking immunosuppressive agents), or in individuals undergoing long term antibiotic therapy.


The PMP composition can be formulated for administration or administered by any suitable method, including, for example, intravenously, intramuscularly, subcutaneously, intradermally, percutaneously, intraarterially, intraperitoneally, intralesionally, intracranially, intraarticularly, intraprostatically, intrapleurally, intratracheally, intrathecally, intranasally, intravaginally, intrarectally, topically, intratumorally, peritoneally, subconjunctivally, intravesicularly, mucosally, intrapericardially, intraumbilically, intraocularly, intraorbitally, orally, topically, transdermally, intravitreally (e.g., by intravitreal injection), by eye drop, by inhalation, by injection, by implantation, by infusion, by continuous infusion, by localized perfusion bathing target cells directly, by catheter, by lavage, in cremes, or in lipid compositions. Oral administration includes delivery of the compositions in food or animal feed, accordingly, the invention includes food and feed compositions comprising the PMP compositions described herein. The compositions utilized in the methods described herein can also be administered systemically or locally. The method of administration can vary depending on various factors (e.g., the compound or composition being administered and the severity of the condition, disease, or disorder being treated). In some instances, PMP composition is administered intravenously, intramuscularly, subcutaneously, topically, orally, transdermally, intraperitoneally, intraorbitally, by implantation, by inhalation, intrathecally, intraventricularly, or intranasally. Dosing can be by any suitable route, e.g., by injections, such as intravenous or subcutaneous injections, depending in part on whether the administration is brief or chronic. Various dosing schedules including but not limited to single or multiple administrations over various time-points, bolus administration, and pulse infusion are contemplated herein.


For the prevention or treatment of an infection or disease or disorder described herein, use of a PMP composition (when used alone or in combination with one or more other additional therapeutic agents) will depend on the type of disease to be treated, the severity and course of the disease, whether the administration for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the PMP composition. The PMP composition can be, e.g., administered to the patient at one time or over a series of treatments. For repeated administrations over several days or longer, depending on the condition, the treatment would generally be sustained until a desired suppression of disease symptoms occurs or the infection is no longer detectable. Such doses may be administered intermittently, e.g., every week or every two weeks (e.g., such that the patient receives, for example, from about two to about twenty, doses of the PMP composition. An initial higher loading dose, followed by one or more lower doses may be administered. However, other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques and assays.


In some instances, the amount of the PMP composition administered to individual (e.g., human) may be in the range of about 0.01 mg/kg to about 5 g/kg (e.g., about 0.01 mg/kg-0.1 mg/kg, about 0.1 mg/kg-1 mg/kg, about 1 mg/kg-10 mg/kg, about 10 mg/kg-100 mg/kg, about 100 mg/kg-1 g/kg, or about 1 g/kg-5 g/kg), of the individual's body weight. In some instances, the amount of the PMP composition administered to individual (e.g., human) is at least 0.01 mg/kg (e.g., at least 0.01 mg/kg, at least 0.1 mg/kg, at least 1 mg/kg, at least 10 mg/kg, at least 100 mg/kg, at least 1 g/kg, or at least 5 g/kg), of the individual's body weight. The dose may be administered as a single dose or as multiple doses (e.g., 2, 3, 4, 5, 6, 7, or more than 7 doses). In some instances, the PMP composition administered to the animal may be administered alone or in combination with an additional therapeutic agent. The dose of the antibody administered in a combination treatment may be reduced as compared to a single treatment. The progress of this therapy is easily monitored by conventional techniques.


IV. Kits

The present invention also provides a kit including a container having a PMP composition described herein. The kit may further include instructional material for applying or delivering the PMP composition to a plant in accordance with a method of the present invention. The skilled artisan will appreciate that the instructions for applying the PMP composition in the methods of the present invention can be any form of instruction. Such instructions include, but are not limited to, written instruction material (such as, a label, a booklet, a pamphlet), oral instructional material (such as on an audio cassette or CD) or video instructions (such as on a video tape or DVD).


EXAMPLES

The following are examples of the methods of the invention. It is understood that various other embodiments may be practiced, given the general description provided above.


Example 1: Isolation of Plant Messenger Packs from Plants

This example describes the isolation of crude plant messenger packs (PMPs) from various plant sources, including the leaf apoplast, seed apoplast, root, fruit, vegetable, pollen, phloem, xylem sap and plant cell culture medium.


Experimental Design:

a) PMP Isolation from the Apoplast of Arabidopsis thaliana Leaves



Arabidopsis (Arabidopsis thaliana Col-0) seeds are surface sterilized with 50% bleach and plated on 0.53 Murashige and Skoog medium containing 0.8% agar. The seeds are vernalized for 2 d at 4° C. before being moved to short-day conditions (9-h days, 22° C., 150 μEm−2). After 1 week, the seedlings are transferred to Pro-Mix PGX. Plants are grown for 4-6 weeks before harvest.


PMPs are isolated from the apoplastic wash of 4-6-week old Arabidopsis rosettes, as described by Rutter and Innes, Plant Physiol. 173(1): 728-741, 2017. Briefly, whole rosettes are harvested at the root and vacuum infiltrated with vesicle isolation buffer (20 mM MES, 2 mM CaCl2), and 0.1 M NaCl, pH6). Infiltrated plants are carefully blotted to remove excess fluid, placed inside 30-mL syringes, and centrifuged in 50 mL conical tubes at 700 g for 20 min at 2° C. to collect the apoplast extracellular fluid containing EVs. Next, the apoplast extracellular fluid is filtered through a 0.85 μm filter to remove large particles, and PMPs are purified as described in Example 2.


b) PMP Isolation from the Apoplast of Sunflower Seeds


Intact sunflower seeds (H. annuus L.), and are imbibed in water for 2 hours, peeled to remove the pericarp, and the apoplastic extracellular fluid is extracted by a modified vacuum infiltration-centrifugation procedure, adapted from Regente et al, FEBS Letters. 583: 3363-3366, 2009. Briefly, seeds are immersed in vesicle isolation buffer (20 mM MES, 2 mM CaCl2), and 0.1 M NaCl, pH6) and subjected to three vacuum pulses of 10 s, separated by 30 s intervals at a pressure of 45 kPa. The infiltrated seeds are recovered, dried on filter paper, placed in fritted glass filters and centrifuged for 20 min at 400 g at 4° C. The apoplast extracellular fluid is recovered, filtered through a 0.85 μm filter to remove large particles, and PMPs are purified as described in Example 2.


c) PMP Isolation from Ginger Roots


Fresh ginger (Zingiber officinale) rhizome roots are purchased from a local supplier and washed 3× with PBS. A total of 200 grams of washed roots is ground in a mixer (Osterizer 12-speed blender) at the highest speed for 10 min (pause 1 min for every 1 min of blending), and PMPs are isolated as described in Zhuang et al., J Extracellular Vesicles. 4(1):28713, 2015. Briefly, ginger juice is sequentially centrifuged at 1,000 g for 10 min, 3,000 g for 20 min and 10,000 g for 40 min to remove large particles from the PMP-containing supernatant. PMPs are purified as described in Example 2.


d) PMP Isolation from Grapefruit Juice


Fresh grapefruits (Citrus x paradisi) are purchased from a local supplier, their skins are removed, and the fruit is manually pressed, or ground in a mixer (Osterizer 12-speed blender) at the highest speed for 10 min (pause 1 min for every minute of blending) to collect the juice, as described by Wang et al., Molecular Therapy. 22(3): 522-534, 2014 with minor modifications. Briefly, juice/juice pulp is sequentially centrifuged at 1,000 g for 10 min, 3,000 g for 20 min, and 10,000 g for 40 min to remove large particles from the PMP-containing supernatant. PMPs are purified as described in Example 2.


e) PMP Isolation from Broccoli Heads


Broccoli (Brassica oleracea var. italica) PMPs are isolated as previously described (Deng et al., Molecular Therapy, 25(7): 1641-1654, 2017). Briefly, fresh broccoli is purchased from a local supplier, washed three times with PBS, and ground in a mixer (Osterizer 12-speed blender) at the highest speed for 10 min (pause 1 min for every minute of blending). Broccoli juice is then sequentially centrifuged at 1,000 g for 10 min, 3,000 g for 20 min, and 10,000 g for 40 min to remove large particles from the PMP-containing supernatant. PMPs are purified as described in Example 2.


f) PMP Isolation from Olive Pollen


Olive (Olea europaea) pollen PMPs are isolated as previously described in Prado et al., Molecular Plant. 7(3):573-577, 2014. Briefly, olive pollen (0.1 g) is hydrated in a humid chamber at room temperature for 30 min before transferring to petri dishes (15 cm in diameter) containing 20 ml germination medium: 10% sucrose, 0.03% Ca(NO3)2, 0.01% KNO3, 0.02% MgSO4, and 0.03% H3BO3. Pollen is germinated at 30° C. in the dark for 16 h. Pollen grains are considered germinated only when the tube is longer than the diameter of the pollen grain. Cultured medium containing PMPs is collected and cleared of pollen debris by two successive filtrations on 0.85 um filters by centrifugation. PMPs are purified as described in Example 2.


g) PMP Isolation from Arabidopsis Phloem Sap



Arabidopsis (Arabidopsis thaliana Col-0) seeds are surface sterilized with 50% bleach and plated on 0.53 Murashige and Skoog medium containing 0.8% agar. The seeds are vernalized for 2 d at 4° C. before being moved to short-day conditions (9-h days, 22° C., 150 μEm−2). After 1 week, the seedlings are transferred to Pro-Mix PGX. Plants are grown for 4-6 weeks before harvest.


Phloem sap from 4-6-week old Arabidopsis rosette leaves is collected as described by Tetyuk et al., JoVE. 80, 2013. Briefly, leaves are cut at the base of the petiole, stacked, and placed in a reaction tube containing 20 mM K2-EDTA for one hour in the dark to prevent sealing of the wound. Leaves are gently removed from the container, washed thoroughly with distilled water to remove all EDTA, put in a clean tube, and phloem sap is collected for 5-8 hours in the dark. Leaves are discarded, phloem sap is filtered through a 0.85 μm filter to remove large particles, and PMPs are purified as described in Example 2.


h) PMP Isolation from Tomato Plant Xylem Sap


Tomato (Solanum lycopersicum) seeds are planted in a single pot in an organic-rich soil, such as Sunshine Mix (Sun Gro Horticulture, Agawam, Mass.) and maintained in a greenhouse between 22° C. and 28° C. About two weeks after germination, at the two true-leaf stage, the seedlings are transplanted individually into pots (10 cm diameter and 17 cm deep) filled with sterile sandy soil containing 90% sand and 10% organic mix. Plants are maintained in a greenhouse at 22-28° C. for four weeks.


Xylem sap from 4-week old tomato plants is collected as described by Kohlen et al., Plant Physiology. 155(2):721-734, 2011. Briefly, tomato plants are decapitated above the hypocotyl, and a plastic ring is placed around the stem. The accumulating xylem sap is collected for 90 min after decapitation. Xylem sap is filtered through a 0.85 μm filter to remove large particles, and PMPs are purified as described in Example 2.


i) PMP Isolation from Tobacco BY-2 Cell Culture Medium


Tobacco BY-2 (Nicotiana tabacum L cv. Bright Yellow 2) cells are cultured in the dark at 26° C., on a shaker at 180 rpm in MS (Murashige and Skoog, 1962) BY-2 cultivation medium (pH 5.8) comprised MS salts (Duchefa, Haarlem, Netherlands, at #M0221) supplemented with 30 g/L sucrose, 2.0 mg/L potassium dihydrogen phosphate, 0.1 g/L myo-inositol, 0.2 mg/L 2,4-dichlorophenoxyacetic acid, and 1 mg/L thiamine HCl. The BY-2 cells are subcultured weekly by transferring 5% (v/v) of a 7-day-old cell culture into 100 mL fresh liquid medium. After 72-96 hours, BY-2 cultured medium is collected and centrifuged at 300 g at 4° C. for 10 minutes to remove cells. The supernatant containing PMPs is collected and cleared of debris by filtration on 0.85 um filter. PMPs are purified as described in Example 2.


Example 2: Production of Purified Plant Messenger Packs (PMPs)

In this example, purified PMPs are produced from crude PMP fractions as described in Example 1, using ultrafiltration combined with size-exclusion chromatography, a density gradient (iodixanol or sucrose), and the removal of aggregates by precipitation or size-exclusion chromatography.


Experimental Design:

a) Production of Purified Grapefruit PMPs Using Ultrafiltration Combined with Size-Exclusion Chromatography


The crude grapefruit PMP fraction from Example 1a is concentrated using 100-kDA molecular weight cut-off (MWCO) Amicon spin filter (Merck Millipore). Subsequently, the concentrated crude PMP solution is loaded onto a PURE-EV size exclusion chromatography column (HansaBioMed Life Sciences Ltd) and isolated according to the manufacturer's instructions. The purified PMP-containing fractions are pooled after elution. Optionally, PMPs can be further concentrated using a 100-kDa MWCO Amicon spin filter, or by Tangential Flow Filtration (TFF). The purified PMPs are analyzed as described in Example 3.


b) Production of Purified Arabidopsis Apoplast PMPs Using an Iodixanol Gradient


Crude Arabidopsis leaf apoplast PMPs are isolated as described in Example 1a, and purified PMPs are produced by using an iodixanol gradient as described in Rutter and Innes, Plant Physiol. 173(1): 728-741, 2017. To prepare discontinuous iodixanol gradients (OptiPrep; Sigma-Aldrich), solutions of 40% (v/v), 20% (v/v), 10% (v/v), and 5% (v/v) iodixanol are created by diluting an aqueous 60% OptiPrep stock solution in vesicle isolation buffer (VIB; 20 mM MES, 2 mM CaCl2, and 0.1 M NaCl, pH6). The gradient is formed by layering 3 ml of 40% solution, 3 mL of 20% solution, 3 mL of 10% solution, and 2 mL of 5% solution. The crude apoplast PMP solution from Example 1a is centrifuged at 40,000 g for 60 min at 4° C. The pellet is resuspended in 0.5 ml of VIB and layered on top of the gradient. Centrifugation is performed at 100,000 g for 17 h at 4° C. The first 4.5 ml at the top of the gradient is discarded, and subsequently 3 volumes of 0.7 ml that contain the apoplast PMPs are collected, brought up to 3.5 mL with VIB and centrifuged at 100,000 g for 60 min at 4° C. The pellets are washed with 3.5 ml of VIB and repelleted using the same centrifugation conditions. The purified PMP pellets are combined for subsequent analysis, as described in Example 3.


c) Production of Purified Grapefruit PMPs Using a Sucrose Gradient


Crude grapefruit juice PMPs are isolated as described in Example 1d, centrifuged at 150,000 g for 90 min, and the PMP-containing pellet is resuspended in 1 ml PBS as described (Mu et al., Molecular Nutrition & Food Research. 58(7):1561-1573, 2014). The resuspended pellet is transferred to a sucrose step gradient (8%/15%/30%/45%/60%) and centrifuged at 150,000 g for 120 min to produce purified PMPs. Purified grapefruit PMPs are harvested from the 30%/45% interface, and subsequently analyzed, as described in Example 3.


d) Removal of Aggregates from Grapefruit PMPs


In order to remove protein aggregates from produced grapefruit PMPs as described in Example 1d or purified PMPs from Example 2a-c, an additional purification step can be included. The produced PMP solution is taken through a range of pHs to precipitate protein aggregates in solution. The pH is adjusted to 3, 5, 7, 9, or 11 with the addition of sodium hydroxide or hydrochloric acid. pH is measured using a calibrated pH probe. Once the solution is at the specified pH, it is filtered to remove particulates. Alternatively, the isolated PMP solution can be flocculated using the addition of charged polymers, such as Polymin-P or Praestol 2640. Briefly, 2-5 g per L of Polymin-P or Praestol 2640 is added to the solution and mixed with an impeller. The solution is then filtered to remove particulates. Alternatively, aggregates are solubilized by increasing salt concentration. NaCl is added to the PMP solution until it is at 1 mol/L. The solution is then filtered to purify the PMPs. Alternatively, aggregates are solubilized by increasing the temperature. The isolated PMP mixture is heated under mixing until it has reached a uniform temperature of 50° C. for 5 minutes. The PMP mixture is then filtered to isolate the PMPs. Alternatively, soluble contaminants from PMP solutions are separated by size-exclusion chromatography column according to standard procedures, where PMPs elute in the first fractions, whereas proteins and ribonucleoproteins and some lipoproteins are eluted later. The efficiency of protein aggregate removal is determined by measuring and comparing the protein concentration before and after removal of protein aggregates via BCA/Bradford protein quantification. The produced PMPs are analyzed as described in Example 3.


Example 3: Plant Messenger Pack Characterization

This example describes the characterization of PMPs produced as described in Example 1 or Example 2.


Experimental Design:

a) Determining PMP Concentration


PMP particle concentration is determined by Nanoparticle Tracking Analysis (NTA) using a Malvern NanoSight, or by Tunable Resistive Pulse Sensing (TRPS) using an iZon qNano, following the manufacturer's instructions. The protein concentration of purified PMPs is determined by using the DC Protein assay (Bio-Rad). The lipid concentration of purified PMPs is determined using a fluorescent lipophilic dye, such as DiOC6 (ICN Biomedicals) as described by Rutter and Innes, Plant Physiol. 173(1): 728-741, 2017. Briefly, purified PMP pellets from Example 2 are resuspended in 100 ml of 10 mM DiOC6 (ICN Biomedicals) diluted with MES buffer (20 mM MES, pH 6) plus 1% plant protease inhibitor cocktail (Sigma-Aldrich) and 2 mM 2,29-dipyridyl disulfide. The resuspended PMPs are incubated at 37° C. for 10 min, washed with 3 mL of MES buffer, repelleted (40,000 g, 60 min, at 4° C.), and resuspended in fresh MES buffer. DiOC6 fluorescence intensity is measured at 485 nm excitation and 535 nm emission.


b) Biophysical and Molecular Characterization of PMPs


PMPs are characterized by electron and cryo-electron microscopy on a JEOL 1010 transmission electron microscope, following the protocol from Wu et al., Analyst. 140(2):386-406, 2015. The size and zeta potential of the PMPs are also measured using a Malvern Zetasizer or iZon qNano, following the manufacturer's instructions. Lipids are isolated from PMPs using chloroform extraction and characterized with LC-MS/MS as demonstrated in Xiao et al. Plant Cell. 22(10): 3193-3205, 2010. Glycosyl inositol phosphorylceramides (GIPCs) lipids are extracted and purified as described by Cacas et al., Plant Physiology. 170: 367-384, 2016, and analyzed by LC-MS/MS as described above. Total RNA, DNA, and protein are characterized using Quant-It kits from Thermo Fisher according to instructions. Proteins on the PMPs are characterized by LC-MS/MS following the protocol in Rutter and Innes, Plant Physiol. 173(1): 728-741, 2017. RNA and DNA are extracted using Trizol, prepared into libraries with the TruSeq Total RNA with Ribo-Zero Plant kit and the Nextera Mate Pair Library Prep Kit from Illumina, and sequenced on an Illumina MiSeq following manufacturer's instructions.


Example 4: Characterization of Plant Messenger Pack Stability

This example describes measuring the stability of PMPs under a wide variety of storage and physiological conditions.


Experimental Design:

PMPs produced as described in Examples 1 and 2 are subjected to various conditions. PMPs are suspended in water, 5% sucrose, or PBS and left for 1, 7, 30, and 180 days at −20° C., 4° C., 20° C., and 37° C. PMPs are also suspended in water and dried using a rotary evaporator system and left for 1, 7, and 30, and 180 days at 4° C., 20° C., and 37° C. PMPs are also suspended in water or 5% sucrose solution, flash-frozen in liquid nitrogen and lyophilized. After 1, 7, 30, and 180 days, dried and lyophilized PMPs are then resuspended in water. The previous three experiments with conditions at temperatures above 0° C. are also exposed to an artificial sunlight simulator in order to determine content stability in simulated outdoor UV conditions. PMPs are also subjected to temperatures of 37° C., 40° C., 45° C., 50° C., and 55° C. for 1, 6, and 24 hours in buffered solutions with a pH of 1, 3, 5, 7, and 9 with or without the addition of 1 unit of trypsin or in other simulated gastric fluids.


After each of these treatments, PMPs are bought back to 20° C., neutralized to pH 7.4, and characterized using some or all of the methods described in Example 3.


Example 5. Uptake of Pectinase-Treated PMPs by Alfalfa Sprouts

This example demonstrates that the removal of pectins during the PMP production process does not impact their in planta uptake and systemic transport. In this example, lemon PMPs were used as model PMPs, and Alfalfa sprouts were used as model plant.


a) Production of Lemon PMPs with or without the Addition of Pectinase


Lemons were obtained from a local market. Lemon juice (1260 ml) was collected using a juice press, and split into two fractions. 630 ml was untreated, and 630 ml was pH adjusted to pH4 with NaOH and incubated with 6 U/ml pectinase (Sigma, 17389) for 1.45 hrs at room temperature. Pectinase treated and untreated juice was subsequently centrifuged at 3000 g for 20 minutes, followed by 10,000 g for 40 minutes to remove large debris. Next, the processed juice was incubated with 500 mM EDTA pH8.6, to a final concentration of 50 mM EDTA, pH 7.19-7.25, for 30 minutes at room temperature to chelate calcium and prevent the formation of pectin macromolecules. Subsequently, the EDTA-treated juice was passaged through an 11 um, 1 um and 0.45 um filter to remove large particles. Filtered juice was washed (260 ml PBS during TFF procedure) and concentrated ˜1.6× to a total volume of 400 ml by Tangential Flow Filtration (TFF), and dialyzed overnight in PBS, pH 7.4 using a 300 kDa dialysis membrane. Subsequently, the dialyzed juice was further concentrated by TFF to a final concentration of 30 ml (˜21×). Next, we used size exclusion chromatography to elute the PMP-containing fractions, and analyzed the 280 nm absorbance (SpectraMax) to determine the PMP-containing fractions from late elution fractions containing contaminants. SEC fractions 4-6 (no pectinase treatment) and SEC fractions 4-7 (with pectinase treatment) containing purified PMPs were pooled together in the individual treatment groups. Pooled SEC fractions were dialyzed o/n in PBS, pH 7.4 using a 300 kDa dialysis membrane. Samples were sterilized by sequential filtration using 0.85 um, 0.4 um and 0.22 um syringe filters, and concentrated further by pelleting PMPs for 1.5 hrs at 40,000× g and finally the pellet is resuspended in Ultrapure water. The final PMP concentration for untreated lemon PMPs was 1.24×1012 PMPs/ml and median PMP size was 129 nm+/−12 nm SD; for pectinase-treated lemon PMPs the final concentration was 2.261012 PMPs/ml and median PMP size was 130 nm+/−11 nm (SD), as determined by nano-flow cytometry (NanoFCM) using concentration and size standards provided by the manufacturer.


b) Labeling of Lemon PMPs with DyLight 800 NHS Ester


Pectinase treated and untreated lemon PMPs were labeled with the DyLight 800 NHS Ester (Life Technologies, #46421) covalent membrane dye (DyL800). Briefly, DyL800 was dissolved in DMSO to a final concentration of 10 mg/ml, 200 ul of PMPs were mixed with 5 ul dye, incubated for 1 h at room temperature on a shaker, and labeled PMPs were washed 2-3 times by ultracentrifugation at 100,000×g for 1 hr at 4° C. and pellets were resuspended with 1.5 ml UltraPure water. To control for the presence of potential dye aggregates, a dye-only control sample was prepared according to the same procedure, adding 200 ul of UltraPure water instead of PMPs. The final DyL800-labeled PMP pellet and DyL800 dye-only control were resuspended in a minimal amount of UltraPure water and characterized by NanoFCM. The final concentration of non-pectinase treated Dyl800-labeled lemon PMPs was 3.2×1012 PMPs/ml, and of pectinase treated DyL800-labeled was 5.57×1012 PMPs/ml. The labeling efficiency could not be determined using the nanoFCM, as it cannot detect infrared.


c) Treatment of Alfalfa Sprouts with Pectinase Treated and Untreated DyL800-PMPs


To assess whether the removal of pectin during PMP production impacts PMP uptake, Alfalfa sprouts were obtained from a local supermarket, were treated with pectinase-treated and untreated DyLight800-Lemon PMPs, water (negative control), DyLight800 nm dye only (dye aggregate control) in half-strength Murashige and Skoog (MS), supplemented with 0.5% sucrose and 2.5 mM MES, pH 5.6 for 21 hours at 23° C. (FIG. 9A). Seedlings where then washed 3 times in MS medium, and imaged using an Odyssey infrared imager. There was no difference in uptake and transport of PMPs produced with or without pectinase treatment (FIG. 9B).


Example 6: PMP Production from Blended Fruit Juice Using Ultracentrifugation and Sucrose Gradient Purification

In this example, PMPs were produced from fruit by blending the fruit and using a combination of sequential centrifugation to remove debris, ultracentrifugation to pellet crude PMPs, and using a sucrose density gradient to purify PMPs. Grapefruit was used as a model fruit.


a) Production of Grapefruit PMPs by Ultracentrifugation and Sucrose Density Gradient Purification


An exemplary workflow for grapefruit PMP production using a blender, ultracentrifugation and sucrose gradient purification is shown in FIG. 10A. One red grapefruit was purchased from a local market, and the albedo, flavedo, and segment membranes were removed to collect juice sacs, which were homogenized using a blender at maximum speed for 10 minutes. One hundred mL juice was diluted 5× with PBS, followed by subsequent centrifugation at 1000×g for 10 minutes, 3000× g for 20 minutes, and 10,000× g for 40 minutes to remove large debris. 28 mL of cleared juice was ultracentrifuged on a Sorvall™ MX 120 Plus Micro-Ultracentrifuge at 150,000× g for 90 minutes at 4° C. using a S50-ST (4×7 mL) swing bucket rotor to obtain a crude PMP pellet which was resuspended in PBS pH 7.4. Next, a sucrose gradient was prepared in Tris-HCL pH7.2, crude PMPs were layered on top of the sucrose gradient (from top to bottom: 8, 15, 30, 45 and 60% sucrose), and spun down by ultracentrifugation at 150,000×g for 120 minutes at 4° C. using a S50-ST (4×7 mL) swing bucket rotor. One mL fractions were collected and PMPs were isolated at the 30-45% interface. The fractions were washed with PBS by ultracentrifugation at 150,000×g for 120 minutes at 4° C. and pellets were dissolved in a minimal amount of PBS.


PMP concentration (1×109 PMPs/mL) and median PMP size (121.8 nm) were determined using a Spectradyne nCS1™ particle analyzer, using a TS-400 cartridge (FIG. 10B). The zeta potential was determined using a Malvern Zetasizer Ultra and was −11.5+/−0.357 mV.


In this example, grapefruit PMPs were isolated using ultracentrifugation combined with sucrose gradient purification methods. However, this method induced gelling of the samples at all PMP production steps and in the final PMP solution.


Example 7: PMP Production from Mesh-Pressed Fruit Juice Using Ultracentrifugation and Sucrose Gradient Purification

In this example, cell wall and cell membrane contaminants were reduced during the PMP production process using a milder juicing process (mesh strainer). Grapefruit was used as a model fruit. a) Mild juicing reduces gelling during PMP production from grapefruit PMPs Juice sacs were isolated from a red grapefruit as described in Example 2. To reduce gelling during PMP production, instead of using a destructive blending method, juice sacs were gently pressed against a tea strainer mesh to collect the juice and to reduce cell wall and cell membrane contaminants. After differential centrifugation, the juice was clearer than after using a blender, and one clean PMP-containing sucrose band at the 30-45% intersection was observed after sucrose density gradient centrifugation (FIG. 11). There was overall less gelling during and after PMP production.


Our data shows that use of a mild juicing step reduces gelling caused by contaminants during PMP production when compared to a method comprising blending.


Example 8: PMP Production Using Ultracentrifugation and Size Exclusion Chromatography

This example describes the production of PMPs from fruits by using Ultracentrifugation (UC) and Size Exclusion Chromatography (SEC). In this example, grapefruit is used as a model fruit.


a) Production of Grapefruit PMPs Using UC and SEC


Juice sacs were isolated from a red grapefruit, as described in Example 6a, and were gently pressed against a tea strainer mesh to collect 28 ml juice. The workflow for grapefruit PMP production using UC and SEC is depicted in FIG. 12A. Briefly, juice was subjected to differential centrifugation at 1000×g for 10 minutes, 3000× g for 20 minutes, and 10,000× g for 40 minutes to remove large debris. 28 ml of cleared juice was ultracentrifuged on a Sorvall™ MX 120 Plus Micro-Ultracentrifuge at 100,000× g for 60 minutes at 4° C. using a S50-ST (4×7 mL) swing bucket rotor to obtain a crude PMP pellet which was resuspended in MES buffer (20 mM MES, NaCl, pH 6). After washing the pellets twice with MES buffer, the final pellet was resuspended in 1 ml PBS, pH 7.4. Next, we used size exclusion chromatography to elute the PMP-containing fractions. SEC elution fractions were analyzed by nano-flow cytometry using a NanoFCM to determine PMP size and concentration using concentration and size standards provided by the manufacturer. In addition, absorbance at 280 nm (SpectraMax®) and protein concentration (Pierce™ BCA assay, ThermoFisher) were determined on SEC fractions to identify in which fractions PMPs are eluted (FIGS. 12B-12D). SEC fractions 2-4 were identified as the PMP-containing fractions. Analysis of earlier- and later-eluting fractions indicated that SEC fraction 3 is the main PMP-containing fraction, with a concentration of 2.83×1011 PMPs/mL (57.2% of all particles in the 50-120 nm size range), with a median size of 83.6 nm+/−14.2 nm (SD). While the late elution fractions 8-13 had a very low concentration of particles as shown by NanoFCM, protein contaminants were detected in these fractions by BCA analysis.


Our data shows that TFF and SEC can be used to isolate purified PMPs from late-eluting contaminants, and that a combination of the analysis methods used here can identify PMP fractions from late-eluting contaminants.


Example 9: Scaled PMP Production Using Tangential Flow Filtration and Size Exclusion Chromatography Combined with EDTA/Dialysis to Reduce Contaminants

This example describes the scaled production of PMPs from fruits by using Tangential Flow Filtration (TFF) and Size Exclusion Chromatography (SEC), combined with an EDTA incubation to reduce the formation of pectin macromolecules, and overnight dialysis to reduce contaminants. In this example, grapefruit is used as a model fruit.


a) Production of Grapefruit PMPs Using TFF and SEC


Red grapefruits were obtained from a local market, and 1000 ml juice was isolated using a juice press. The workflow for grapefruit PMP production using TFF and SEC is depicted in FIG. 13A. Juice was subjected to differential centrifugation at 1000×g for 10 minutes, 3000× g for 20 minutes, and 10,000× g for 40 minutes to remove large debris. Cleared grapefruit juice was concentrated and washed once using a TFF (TFF-easy, HansaBioMed Life Sciences) to 2 mL (100×). Next, we used size exclusion chromatography to elute the PMP-containing fractions. SEC elution fractions were analyzed by nano-flow cytometry using a NanoFCM to determine PMP concentration using concentration and size standards provided by the manufacturer. In addition, protein concentration (Pierce™ BCA assay, ThermoFisher) was determined for SEC fractions to identify the fractions in which PMPs are eluted. The scaled production from 1 liter of juice (100× concentrated) also concentrated a high amount of contaminants in the late SEC fractions as can be detected by BCA assay (FIG. 13B, top panel). The overall total PMP yield (FIG. 13B, bottom panel) was lower in the scaled production when compared to single grapefruit isolations, which may indicate loss of PMPs.


b) Reducing Contaminants by EDTA Incubation and Dialysis


Red grapefruits were obtained from a local market, and 800 ml juice was isolated using a juice press. Juice was subjected to differential centrifugation at 1000×g for 10 minutes, 3000× g for 20 minutes, and 10,000× g for 40 minutes to remove large debris, and filtered through a 1 μm and 0.45 μm filter to remove large particles. Cleared grapefruit juice was split into 4 different treatment groups containing 125 ml juice each. Treatment Group 1 was processed as described in Example 9a, concentrated and washed (PBS) to a final concentration of 63×, and subjected to SEC. Prior to TFF, 475 ml juice was incubated with a final concentration of 50 mM EDTA, pH 7.15 for 1.5 hrs at RT to chelate iron and reduce the formation of pectin macromolecules. Afterwards, juice was split in three treatment groups that underwent TFF concentration with either a PBS (without calcium/magnesium) pH 7.4, MES pH 6, or Tris pH 8.6 wash to a final juice concentration of 63×. Next, samples were dialyzed in the same wash buffer overnight at 4° C. using a 300 kDa membrane and subjected to SEC. Compared to the high contaminant peak in the late elution fractions of the TFF only control, EDTA incubation followed by overnight dialysis strongly reduced contaminants, as shown by absorbance at 280 nm (FIG. 13C) and BCA protein analysis (FIG. 13D), which is sensitive to the presence of sugars and pectins. There was no difference in the dialysis buffers used (PBS without calcium/magnesium pH 7.4, MES pH 6, Tris pH 8.6).


Our data indicates that incubation with EDTA followed by dialysis reduces the amount of co-purified contaminants, facilitating scaled PMP production.


Example 10: PMP Production from Plant Cell Culture Medium

In this example, PMPs were produced from plant cell culture. The Zea mays Black Mexican Sweet (BMS) cell line is used as a model plant cell line.


a) Production of Zea mays BMS Cell Line PMPs


The Zea mays Black Mexican sweet (BMS) cell line was purchased from the ABRC and was grown in Murashige and Skoog basal medium pH 5.8, containing 4.3 g/L Murashige and Skoog Basal Salt Mixture (Sigma M5524), 2% sucrose (S0389, Millipore Sigma), 1× MS vitamin solution (M3900, Millipore Sigma), 2 mg/L 2,4-dichlorophenoxyacetic acid (D7299, Millipore Sigma) and 250 ug/L thiamine HCL (V-014, Millipore Sigma), at 24° C. with agitation (110 rpm), and was passaged 20% volume/volume every 7 days.


Three days after passaging, 160 ml BMS cells was collected and spun down at 500× g for 5 min to remove cells, and 10,000×g for 40 min to remove large debris. Medium was passed through a 0.45 μm filter to remove large particles, and filtered medium was concentrated and washed (100 ml MES buffer, 20 mM MES, 100 mM NaCL, pH 6) by TFF (5 nm pore size) to 4 mL (40×). Next, we used size exclusion chromatography to elute the PMP-containing fractions, which were analyzed by NanoFCM for PMP concentration, by absorbance at 280 nm (SpectraMax®), and by a protein concentration assay (Pierce™ BCA assay, ThermoFisher) to verify the PMP-containing fractions and late fractions containing contaminants (FIGS. 14A-14C). SEC fractions 4-6 contained purified PMPs (fractions 9-13 contained contaminants), and were pooled together. The final PMP concentration (2.84×1010 PMPs/ml) and median PMP size (63.2 nm+/−12.3 nm SD) in the combined PMP containing fractions were determined by NanoFCM, using concentration and size standards provided by the manufacturer (FIGS. 14D-14E).


These data show that PMPs were isolated, purified, and concentrated from plant liquid culture media.


Example 11: Uptake of PMPs by Plant Cells

This example describes the association with and uptake of PMPs by plant cells. In this example, lemon PMPs are used as a model PMP, and soy, wheat and corn cell lines are used as model plant cells.


a) Production of Grapefruit PMPs Using TFF Combined with SEC


Red organic grapefruits (Florida) were obtained from a local market. One liter of grapefruit juice was collected using a juice press, and was subsequently centrifuged at 3000×g for 20 minutes, followed by 10,000× g for 40 minutes to remove large debris. Next, 500 mM EDTA pH 8.6 was added to a final concentration of 50 mM EDTA, pH 7, and the solution was incubated for 30 minutes to chelate calcium and prevent the formation of pectin macromolecules. Subsequently the juice was passaged through 11 μm, 1 μm and 0.45 μm filters to remove large particles. Filtered juice was concentrated and washed (500 ml PBS) by Tangential Flow Filtration (TFF) (pore size 5 nm) to 400 ml (2.5×) and dialyzed overnight in PBS pH 7.4 (with one medium exchange) using a 300 kDa dialysis membrane to remove contaminants. Subsequently, the dialyzed juice was further concentrated by TFF to a final concentration of 50 ml (20×). Next, we used size exclusion chromatography to elute the PMP-containing fractions, which were analyzed by absorbance at 280 nm (SpectraMax®) and a protein concentration assay (Pierce™ BCA assay, ThermoFisher) to verify the PMP-containing fractions and late fractions containing contaminants. SEC fractions 4-6 contained purified PMPs (fractions 8-14 contained contaminants), were pooled together, and were filter sterilized by sequential filtration using 0.8 m, 0.45 m and 0.22 m syringe filters. The final PMP concentration (1.32×1011 PMPs/mL) and median PMP size (71.9 nm+/−14.5 nm) in the combined sterilized PMP-containing fractions were determined by NanoFCM using concentration and size standards provided by the manufacturer.


b) Production of Lemon PMPs Using TFF Combined with SEC


Lemons were obtained from a local market. One liter of lemon juice was collected using a juice press, and was subsequently centrifuged at 3000 g for 20 minutes, followed by 10,000 g for 40 minutes to remove large debris. Next, 500 mM EDTA pH 8.6 was added to a final concentration of 50 mM EDTA, pH 7, and the solution was incubated for 30 minutes to chelate calcium and prevent the formation of pectin macromolecules. Subsequently the juice was passaged through a coffee filter, 1 μm and 0.45 μm filters to remove large particles. Filtered juice was concentrated by Tangential Flow Filtration (TFF) (5 nm pore size) to 400 ml (2.5× concentrated) and dialyzed overnight in PBS pH 7.4 using a 300 kDa dialysis membrane to remove contaminants. Subsequently, the dialyzed juice was further concentrated by TFF to a final concentration of 50 ml (20×). Next, we used size exclusion chromatography to elute the PMP-containing fractions, which were analyzed by absorbance at 280 nm (SpectraMax®) and a protein concentration assay (Pierce™ BCA assay, ThermoFisher) to verify the PMP-containing fractions and late fractions containing contaminants. SEC fractions 4-6 contained purified PMPs (fractions 8-14 contained contaminants), were pooled together, and were filter sterilized by sequential filtration using 0.8 m, 0.45 m and 0.22 m syringe filters. The final PMP concentration (2.7×1011 PMPs/mL) and median PMP size (70.7 nm+/−15.8 nm) in the combined sterilized PMP-containing fractions were determined by NanoFCM, using concentration and size standards provided by the manufacturer.


c) Labeling of Lemon PMPs with Alexa Fluor 488 NHS Ester


Lemon PMPs were produced as described in Example 11b. PMPs were labeled with the Alexa Fluor 488® NHS Ester (Life Technologies, covalent membrane dye (AF488)). Briefly, AF488 was dissolved in DMSO to a final concentration of 10 mg/ml, 200 ul of PMPs (1.53E+13 PMPs/ml) were mixed with 5 ul dye, incubated for 1 h at room temperature on a shaker, and labeled PMPs were washed 2-3 times by ultracentrifuge at 100,000×g for 1 hr at 4° C. Pellets were resuspended with 1.5 ml UltraPure water. To control for the presence of potential dye aggregates, a dye-only control sample was prepared according to the same procedure, adding 200 ul of UltraPure water instead of PMPs. The final AF488-labeled PMP pellet and AF488 dye-only control were resuspended in a minimal amount of UltraPure water and characterized by NanoFCM. The final concentration of lemon 488-labeled PMPs was 2.91×1012 PMPs/ml with a median AF488-PMP size of 79.4 nm+/−14.7 nm and a labeling efficiency of 89.4% (FIG. 15A).


d) Uptake of AF488-Labeled Lemon PMPs by Plant Cells


Plant cell lines were purchased from the Deutsche Sammlung von Mikroorganismen und Zellkulturen (DSMZ) (Glycine max, #PC-1026; Triticum aestivum, #PC-998) and ABRC (Zea mays, Black Mexican sweet (BMS), and were grown in baffled vented 250 mL flasks in the dark, at 24° C. with agitation (110 rpm). Glycine max and Triticum aestivum were grown in 3.2 g/L Gamborg's B-5 Basal Medium with Minimal Organics supplemented (G5893, Millipore Sigma) pH 5.5, supplemented with 2% sucrose, and 2 mg/L 2,4-dichlorophenoxyacetic acid (2,4D) (D7299, Millipore Sigma) according to the supplier's instructions. BMS cells were grown in Murashige and Skoog basal medium pH 5.8, containing 4.3 g/L Murashige and Skoog Basal Salt Mixture (Sigma M5524), 2% sucrose (S0389, Millipore Sigma), 1× MS vitamin solution (M3900, Millipore Sigma), 2 mg/L 2,4-dichlorophenoxyacetic acid (D7299, Millipore Sigma) and 250 ug/L thiamine HCL (V-014, Millipore Sigma).


For treatment with AF488-PMPs, 5 mL of the cell suspensions was taken to determine the percent Pack Cell Volume (PCV). The PCV is defined as the volume of cells divided by the total volume of the cell culture aliquot, and expressed as a percentage. Cells were centrifuged for 5 min at 3900 rpm, and the volume of the cell pellet was determined. The % PCV for BMS, Glycine max, and Triticum aestivum were 20%, 15%, and 18%, respectively. For the uptake experiment, the % PCV of the cultures was adjusted to 2%, by diluting cells in their appropriate medium. Next, 125 μl of the plant cell suspensions was added to a 24 well plate, and duplicate samples were treated with 125 μl MES buffer (200 mM MES+10 mM NaCl, pH6) alone (negative control), AF488 dye only (dye only control) or a final concentration of 1×1012 AF488-PMPs/mL diluted in MES buffer to 125 μl. Cells were incubated for 2 hours at 24° C. in the dark, washed three times with 1 mL MES buffer to remove AF488-PMPs or free dye that had not been taken up, and resuspended in 300 μL of MES buffer for imaging on an epifluorescence microscope (EVOS FL Auto 2, Invitrogen). Compared to the AF488 dye only control which had no detectable fluorescence, a variable fluorescent signal could be detected in all plant cell lines, indicating PMP uptake (FIG. 15B). Triticum aestivum cells displayed the strongest fluorescence signal, indicating that out of the three plant cell lines tested, they had the highest uptake of AF488-labeled lemon PMPs.


Our data shows that PMPs can be taken up by plant cells in vitro.


Example 12: Uptake of PMPs in Plants

In this example, PMPs were taken up and systemically transported in planta. Grapefruit, lemon and Arabidopsis thaliana seedling PMPs are used as model PMPs, and Arabidopsis seedlings and alfalfa sprouts are used as model plants.


a) Labeling of Lemon and Grapefruit PMPs with DyLight 800 NHS Ester


Grapefruit and lemon PMPs were produced as described in Examples 11a and 11b. PMPs were labeled with the DyLight 800 NHS Ester (Life Technologies, #46421) covalent membrane dye (DyL800). Briefly, Dyl800 was dissolved in DMSO to a final concentration of 10 mg/ml, 200 μl of PMPs were mixed with 5 μl dye, incubated for 1 h at room temperature on a shaker, and labeled PMPs were washed 2-3 times by ultracentrifugation at 100,000×g for 1 hr at 4° C. and pellets were resuspended with 1.5 ml UltraPure water. To control for the presence of potential dye aggregates, a dye-only control sample was prepared according to the same procedure, adding 200 μl of UltraPure water instead of PMPs. The final DyL800-labeled PMP pellet and DyL800 dye-only control were resuspended in a minimal amount of UltraPure water and characterized by NanoFCM. The final concentration of grapefruit DyL800-labeled PMPs was 4.44×1012 PMPs/ml, and of lemon DyL800-labeled PMPs was 5.18×1012 PMPs/ml. The labeling efficiency could not be determined using the NanoFCM, as it cannot detect infrared.


b) Germination and Growth of Arabidopsis thaliana Seedlings


Wild type Arabidopsis thaliana Col-0 seeds were obtained from the ABRC and were surface sterilized with 70% ethanol, incubation with 50% bleach/0.1% triton X-100 for 10 minutes, and 4 sterile ddH2O washes to remove the bleach solution. Seeds were stratified for 1 d at 4° C. in the dark. Approximately 250 seeds were germinated per 100 cm2 plate (pre-coated with 0.5% fetal calf serum in water), containing 20 mL 0.5× MS medium (2.15 g/L Murashige and Skoog salts, 1% sucrose, pH 5.8), sealed with 3M surgical tape and grown in an incubator with a photoperiod of 16 h light at 23° C./8 h dark at 21° C.


c) Uptake of DyL800-Labeled Grapefruit, Lemon and Ats PMPs by Arabidopsis thaliana and Alfalfa


To assess whether PMPs can be taken up and transported systemically in planta, Arabidopsis seedlings were germinated in liquid culture as described in Example 12b on top of a mesh filter, to allow the roots to grow through the mesh, and to allow partial exposure of At seedlings to a PMP solution. Alfalfa sprouts were obtained from a local supermarket. 9 day-old Arabidopsis seedlings and Alfalfa sprouts were treated with a 0.5 ml solution of water (negative control), DyL800 dye only (dye control) DyL800-labeled grapefruit PMPs (1.6×1010 PMPs/ml), or lemon (5.1×1010 PMPs/ml) PMPs in 0.5×MS medium by partial root exposure (At seedlings in a mesh floating in a PMP solution, or in Alfalfa sprouts by partial root exposure in a 1.5 ml Eppendorf tube) for 22 or 24 hours, respectively, at 23° C. Plants where then washed 3 times in MS medium and imaged using an Odyssey® CLx infrared imager (Li-Cor).


Compared to the negative (some autofluorescence in Alfalfa sprout leafs) and dye only control, all PMP sources showed a fluorescence signal (white is high fluorescent signal, black is no signal) in both Arabidopsis seedlings and Alfalfa sprouts, indicating that PMPs are taken up by both plants (FIG. 16). The presence of fluorescence signal in Arabidopsis leafs or Alfalfa stem areas that were not exposed to the PMP solution indicates active transport of the PMPs in planta.


Our data indicate that PMPs derived from various plant sources can be taken up and transported in planta.


Example 13. PMP Preparations Resulting in Gelling

a) Production of PMPs Using a Blender


PMPs were produced from grapefruits using an exemplary workflow including blending, ultracentrifugation, and sucrose gradient purification, as shown in FIG. 1A. Briefly, grapefruit PMPs were produced by blending fruit in a blender at maximum speed for 10 minutes, followed by subsequent centrifugation at 1000×g for 10 minutes, 3000× g for 20 minutes, and 10,000×g for 40 minutes to remove large debris. Crude PMPs were pelleted by ultracentrifugation at 150,000×g for 90 minutes at 4° C. using a swing bucket rotor. PMPs were purified by sucrose density gradient using ultracentrifugation at 150,000×g for 120 minutes at 4° C. PMPs were isolated at the 30-35% interface, and the fraction was washed with PBS by ultracentrifugation. This production process resulted in gelling of the product at all steps of the production process.


b) Production of PMPs Using a Mesh Strainer Juicing Method


PMPs were produced from grapefruits using an exemplary workflow including a milder juice extraction method using a mesh strainer, ultracentrifugation, and sucrose gradient purification, as shown in FIG. 1B. Briefly, grapefruit PMPs were produced by isolating grapefruit juice sacs and gently pressing them through a tea strainer. The juice was collected and subsequently centrifuged at 1000×g for 10 minutes, 3000× g for 20 minutes, and 10,000×g for 40 minutes to remove large debris. Crude PMPs were pelleted by ultracentrifugation at 150,000×g for 90 minutes at 4° C. using a swing bucket rotor. PMPs were purified by sucrose density gradient using ultracentrifugation at 150,000×g for 120 minutes at 4 C. PMPs were isolated at the 30-35% interface, and the fraction was washed with PBS by ultracentrifugation. Compared to the sucrose gradient purification process in FIG. 1A, the gentle juicing process resulted in a whiter and cleaner PMP-containing sucrose band. However, this production process also resulted in gelling at all steps of the production process.


c) Production of PMPs Using a Juice Press


PMPs were produced from grapefruits using an exemplary workflow including a juice press, differential centrifugation to remove large debris, 20× concentration of the juice using tangential flow filtration (TFF), and size exclusion chromatography to isolate the PMP containing fractions, as shown in FIG. 1C. The PMP fractions were analyzed for PMP concentration and particle size using nano flow cytometry (NanoFCM) and for protein concentration using a bicinchoninic acid assay (BCA). PMP concentration in particles/mL is shown in FIG. 1D. PMPs are eluted in SEC fractions 4-6. The majority of PMPs are in SEC fraction 3 (Fr 3), as shown in FIG. 1E and Table 11. PMP production using a juice press resulted in less gelling compared to the methods described in Examples 13a and 13b.









TABLE 11







PMP size distribution in SEC fractions











SEC
Size

% gated



fraction
(nm)
SD
(50-120 nm)
Notes














Fr 1
60.5
11
4.9



Fr 3
83.6
14.2
57.2
Main PMP fraction


Fr 5
71.3
15.7
18.1


Fr 8
69
16.8
13









Example 14. Scaled PMP Preparations

a) Production of PMPs from a Large Volume of Grapefruit Juice


PMPs were produced from a large volume of grapefruit juice (1 liter, the juice of about 7 grapefruits) using an exemplary workflow including a juice press, differential centrifugation to remove large debris, 100× concentration of the juice using tangential flow filtration (TFF), and size exclusion chromatography to isolate the PMP-containing fractions, as shown in FIG. 2A. The PMP fractions were analyzed for PMP concentration and particle size using nano flow cytometry (NanoFCM) and for protein concentration using a bicinchoninic acid assay (BCA). In comparison to the PMP product from 150 mL of grapefruit juice (1 grapefruit), the PMP product from 1 L of grapefruit juice had a high amount of contaminants concentrated in the late SEC fractions, as detected using a BCA assay (FIG. 2B). Additionally, the overall PMP yield in particles/mL is lower than expected in the 1000 mL preparation, which may indicate loss of PMPs during the production process (FIG. 2B) as a result of pectin aggregation and gelling.


Example 15. Production of PMPs with Enhanced Removal of Contaminants

a) PMP Production Process Comprising Chelation and Dialysis


The PMP production process was modified to enhance the removal of contaminants. An exemplary workflow is provided in FIG. 3A. In short, a crude PMP preparation was produced from pressed juice, followed by subsequent centrifugation at 3000×g for 20 minutes, followed by centrifugation at 10,000×g for 40 minutes to remove large debris. To purify PMPs, the crude PMP preparation was incubated with 500 mM EDTA (pH 8.6) to a final concentration of 50 mM EDTA (pH 7.2-8) for 30 minutes to chelate calcium and prevent the formation of pectin macromolecules. Subsequently, the EDTA-treated crude PMP fraction was passaged through a 1 μm and a 0.45 μm filter. Filtered juice was washed with PBS and concentrated 5× by Tangential Flow Filtration (TFF). Concentrated juice was dialyzed in PBS overnight at 4° C. using a 300 kDa dialysis membrane to remove contaminants. Subsequently, the dialyzed juice was further concentrated by TFF to a final concentration of 20×. Next, size exclusion chromatography was used to elute the PMP-containing fractions. Combined PMP-containing fractions were further characterized as described below.


Incubation of the crude grapefruit PMP fraction with a final concentration of 50 mM EDTA (pH 7.2-8), followed by overnight dialysis using a 300 kDa membrane, successfully removed contaminants present in the late elution fractions after SEC, as shown by absorbance at 280 nm (FIG. 3B). The peak containing contaminants is indicated by an arrow. There was no difference in effectiveness among the dialysis buffers used (PBS without calcium/magnesium pH 7.4; MES pH 6; and Tris pH 8.6).


The PMP production process also successfully removed contaminants present in the late elution fractions after SEC as shown by BCA protein analysis, which is also sensitive to the presence of sugars and pectins (FIG. 3C). The peak containing contaminants is indicated by an arrow. There was no difference in effectiveness among the dialysis buffers used (PBS without calcium/magnesium pH 7.4; MES pH 6; and Tris pH 8.6).


b) PMP Production Process Comprising Chelation and Dialysis for Citrus Fruit or Plant Cell Culture


Citrus juice or plant cell culture medium is subjected to a PMP production process with enhanced removal of contaminants. An exemplary workflow is provided in FIGS. 4A and 4B. Briefly, juice or culture medium is collected and subsequently centrifuged at 1000×g for 10 minutes, 3000× g for 20 minutes, and 10,000×g for 40 minutes to remove large debris to produce the crude PMP fraction. The crude PMP fraction is incubated in a final concentration of 50 mM EDTA (pH 7) for 30 minutes, and subsequently passaged through a 1 μm and a 0.45 μm filter. Filtered juice or medium is concentrated 5× by Tangential Flow Filtration (TFF) with PBS washing, and dialyzed overnight in PBS using a 300 kDa dialysis membrane to remove contaminants. Subsequently, the dialyzed juice is further concentrated by TFF to a final concentration of 20×. Size exclusion chromatography is then used to elute the PMP-containing fractions, and the PMP-containing fractions are characterized by, e.g., analysis of PMP concentration and particle size using nano flow cytometry (NanoFCM) and of protein concentration using a bicinchoninic acid assay (BCA).


c) PMP Production Process Comprising Pectinase Treatment


A lemon juice preparation and a grapefruit juice preparation were subjected to a PMP production process including treatment with a pectinase.


Lemons were obtained from a local market. 1260 mL of lemon juice was collected using a juice press and was split into two fractions. 860 mL was untreated, and 835 mL was pH adjusted to pH 4 with NaOH and incubated with 6 U/mL pectinase (Sigma, 17389) for 1.45 hours at room temperature. Pectinase-treated and untreated juice was subsequently centrifuged at 3000 g for 20 minutes, followed by centrifugation at 10,000 g for 40 minutes to remove large debris. Turbidity was reduced in the pectinase-treated sample relative to the untreated sample (FIG. 5A).


Red organic grapefruits were obtained from a local market. 1695 mL of grapefruit juice was collected using a juice press and was split into two fractions. 860 mL was untreated, and 835 mL was pH adjusted to pH 4 with NaOH and incubated with 0.5 U/mL pectinase (Sigma, 17389) throughout subsequent processing steps. Pectinase-treated and untreated juice was subsequently centrifuged at 3000 g for 20 minutes, followed by centrifugation at 10,000 g for 40 minutes to remove large debris. Turbidity was reduced in the pectinase-treated sample relative to the untreated sample (FIG. 5B).


Turbidity of the grapefruit juice preparations was quantified as the volume of juice that could be processed per filter. The addition of pectinase during grapefruit PMP production reduced juice turbidity and facilitated sequential filtration of the differentially centrifuged juice prior to TFF, improving the production process. About four times more of the juice preparation can be processed per 1 um or 0.45 um filter when the juice preparation has been treated with a pectinase (FIG. 5C).


PMP concentration in the pectinase-treated grapefruit juice preparation was measured to be about 2.5 times lower than PMP concentration in the grapefruit juice preparation that was not treated with a pectinase (FIG. 6), which likely indicates the removal of pectinase particles with similar size properties as PMPs.


d) PMP Production Process Comprising Pectinase Treatment and Chelation


A grapefruit juice preparation was subjected to a PMP production process including treatment with a pectinase and chelation. An exemplary workflow is provided in FIG. 7A. Four liters of grapefruit juice were isolated using a juice press. The juice preparation was treated with 0.5 U/mL pectinase as described in Example 15c. The pectinase-treated juice preparation was then centrifuged at 1000×g for 10 minutes, 3000× g for 20 minutes, and 10,000×g for 40 minutes to remove large debris. The juice preparation was then treated with EDTA as described in Example 15a. The preparation was then concentrated 5× using a Spectrum® 300 kDa TFF filter module, washed by 6 volume exchanges with PBS, and concentrated to a final concentration of 20×. Next, size exclusion chromatography (maxiPURE-EVs columns, HansaBioMed Life Sciences) was used to elute the PMP-containing fractions.


For each of the nine columns (A-J), the PMP production process efficiently removed pectin, sugars, protein and other contaminants in the late SEC fractions, as measured by absorbance at 280 nm and by using a BCA protein concentration assay, while PMPs were detected in the early SEC fractions 3-7 (FIGS. 7B and 7C).


Example 16. Light Transmittance Spectra of Pectinase-Treated Juice

A light transmittance spectrum was collected for standard concentrations of pectin (0.1-1%), dissolved in ultrapure water. Increased pectin concentration reduced the % transmittance, i.e., increased the turbidity of the solution (FIG. 8A). The transmittance spectrum was measured on a SpectraMax® i3×.


The light transmittance spectra of a grapefruit juice preparation that was treated with a pectinase and a grapefruit juice preparation that was not treated with a pectinase were collected. In brief, a red grapefruit was juiced using a juice press and split into two fractions. One fraction was incubated with 1 U/mL pectinase (Sigma, 17389); as the pH of the juice was 3.5-4, the pH did not have to be adjusted for pectinase treatment. Pectinase-treated and untreated juice was subsequently centrifuged at 3000×g for 20 minutes, followed by centrifugation at 10,000×g for 40 minutes to remove large debris. Clarified supernatant was transferred to fresh tubes and brought to pH 7.5 with NaOH. The crude PMP juice fraction was subjected to transmittance spectrum analysis using a SpectraMax® i3×. Pectinase treatment increased the % transmittance, i.e., reduced the turbidity of the juice preparation (FIG. 8B).


Example 17. Delivery of a Pectinase-Treated PMP Preparation to a Plant

In this example, pectinase-treated PMPs were taken up and systemically transported in planta. Lemon PMPs are used as model PMPs, and alfalfa sprouts are used as model plants.


a) Labeling of Lemon PMPs with DyLight 800 NHS Ester


Pectinase-treated and untreated lemon PMPs were produced as described in Example 15c, using 0.5 U pectinase. PMPs were labeled with the DyLight 800 infrared membrane dye (Invitrogen; DyL800). Briefly, Dyl800 was dissolved in DMSO to a final concentration of 10 mg/ml, 200 μl of PMPs were mixed with 5 μl dye, incubated for 1 h at room temperature on a shaker, and labeled PMPs were washed 2-3 times by ultracentrifugation at 100,000×g for 1 hr at 4° C. and pellets were resuspended with 1.5 ml UltraPure water. To control for the presence of potential dye aggregates, a dye-only control sample was prepared according to the same procedure, adding 200 μl of UltraPure water instead of PMPs. The final DyL800-labeled PMP pellet and DyL800 dye-only control were resuspended in a minimal amount of UltraPure water and characterized by NanoFCM.


b) Uptake of DyL800-Labeled Lemon PMPs by Alfalfa Sprouts


To assess whether pectinase treatment affects uptake and/or systemic transport of PMPs, alfalfa sprouts were obtained from a local supermarket. Alfalfa sprouts were treated with a 0.5 mL solution of water (negative control), DyL800 dye only (dye control), pectinase-treated lemon PMPs, or untreated lemon PMPs in half-strength Murashige and Skoog (MS) medium supplemented with 0.5% sucrose and 2.5 mM MES, pH 5.6 by partial root exposure in a 1.5 ml Eppendorf tube for 21 hours at 23° C. Plants were then washed 3 times in MS medium and imaged using an Odyssey® CLx infrared imager (Li-Cor) (FIG. 9A). There was no difference in uptake and transport of PMPs produced with or without pectinase treatment, as can be observed by the similar infrared signal intensity and height of the signal in the stem of the treated alfalfa sprouts (FIG. 9B).


Example 18: PMP Production from 18 L of Citrus Fruit Juice

This example describes production of PMPs at 18 L scale. During the production process, citrus juice was treated with pectinase enzyme and incubated with EDTA to prevent pectin aggregation. In this example, grapefruit is used as a model fruit.


a) Production of Grapefruit PMPs from 18 Liters of Juice


Red grapefruits were obtained from a local grocery market. Fruits were washed with 1% Liquinox® detergent (Alconox®) and rinsed under warm water. Next, 18 L of juice was isolated using a commercial citrus juicer (Zumex®, Model No. 08826) and processed by progressive clarification. Large pulp fragments were removed by the juicer itself, and a subsequent filtration through a 600 μm nylon screen filter was performed. The juice was brought to pH 4 with 10N sodium hydroxide solution (VWR Chemicals BDH®), before the addition of pectinase enzyme at a final concentration of 0.5 U/mL (Pectinase from Aspergillus niger, TCI P0026-10). The enzymatic digestion of pectin was performed at room temperature (23° C.-25° C.) for at least 2 hours. Then, a series of differential centrifugations at 3000× g for 20 minutes and 10,000× g for 40 minutes was performed to remove large debris. Subsequent juice clarification was performed by vacuum filtration through 11 μm disk filters (Whatman®) set on a funnel-flask system. Further clarification was performed using a 1.2 μm glass fiber depth filter (Glass fiber, Sartopure® GF+1.2 μm, Sartorius) set on a peristaltic pump system (250 liter/m2/hour (LMH)), followed by a 0.8/0.45 μm PES depth filter (PES, Sartopore® 2, 0.8/0.45 um, 250 LMH). EDTA was added to the juice at a final concentration of 50 mM, and pH was brought to pH 7.5. The clarified juice was stored at 4° C. overnight and was then processed using a TFF system (Repligen, KrosFlo® KR2i TFF System) with a TFF mPES hollow fiber filter (mPES, 300 kDa pore size. Repligen), sequentially concentrated 12.5× (1.6 L), washed by diafiltration with 7 diavolumes of filter sterilized PBS pH7.4, and finally concentrated to 60× based on the initial juice volume (300 mL).


Next, we used size exclusion chromatography to elute the PMP-containing fractions (maxiPURE-EVs size exclusion chromatography columns, HansaBioMed Life Sciences). To identify the PMP-containing fractions, SEC elution fractions were analyzed by absorbance measurement at 280 nm (SpectraMax® spectrophotometer), protein quantification was performed by BCA assay (Pierce™ BCA Protein Assay Kit, Thermo Scientific™), and PMP concentration was determined by nano-flow cytometry (nanoFCM) using concentration standards provided by the manufacturer. SEC fractions 4-8, which contained PMPs, were then combined under aseptic conditions in a tissue culture hood and were syringe filter-sterilized through a 1 μm filter (Glass fiber, Acrodisc®, Pall Laboratory), 0.8/0.45 μm set-pore size filter (PES, Sartopore® 2, 0.8/0.45 μm PES) and finally a 0.2 μm set-pore size filter (Sartopore® 2, 0.2 μm PES). After sterilization, PMPs were concentrated in sterile tubes by ultracentrifugation at 40,000×g for 1.5 h at 4° C., and the resulting pellets were resuspended in sterile PBS pH 7.4 (GIBCO) to a final volume of 15 mL, which represents a 1200× concentration from the input juice volume. The PMP suspension was then analyzed by nanoFCM to determine the final PMP concentration (1.98×1013 PMPs/mL) and size (78.1 nm±19.6 nm) using concentration and size standards provided by the manufacturer, and protein concentration (8.39 mg/mL) was determined by BCA assay (Pierce™ BCA Protein Assay Kit, Thermo Scientific™).


Example 19: PMP Production from Homogenized Plant Sources

This example describes PMP production from homogenized plant sources, including large fruits, berries, whole plants, and vegetables. In this example, Wolffia globosa is used as a model plant, pomegranate and blueberries as model fruits, and broccoli as a model vegetable.


a) Production of Pomegranate PMPs


Pomegranates were obtained from a local grocery market. Fruits were washed with 1% detergent (Alconox®) and rinsed under warm water. Next, 8 L of juice was isolated using a juicer/mincer machine (Slowstar). Juice went through a progressive clarification process. The juice was brought to pH 4 with 10N sodium hydroxide solution (VWR Chemicals BDH®) before the addition of pectinase enzyme at a final concentration of 0.5 U/mL (Pectinase from Aspergillus niger, Sigma-Aldrich 17389). The enzymatic digestion of pectin was performed at room temperature for at least 2 hours (25° C.). Then, a series of differential centrifugations at 3000× g for 20 minutes and 10,000× g for 40 minutes was performed to remove large debris. Subsequent juice clarification was performed using Miracloth mesh (20-25 μm, Sigma-Aldrich) to remove additional debris. EDTA was added to the juice at a final concentration of 50 mM, and pH was brought to pH 7.5. Further clarification of the juice was performed by vacuum filtration through 11 μm disk filters (Whatman®) set on a funnel-flask system, followed by 1 μm glass fiber depth filter (Glass fiber, Acrodisc®, Pall Laboratory) set on a peristaltic pump system (250 LMH), followed by a 0.8/0.45 μm PES depth filter (PES, Sartopore® 2, 0.8/0.45 um, 250 LMH). The clarified juice was stored at 4° C. overnight. Next, the juice was processed using a TFF system (Repligen, KrosFlo® KR2i TFF System) with a TFF mPES hollow fiber filter (mPES, 300 kDa pore size) sequentially concentrated 10× (1.6 L), washed by diafiltration with 10 diavolumes of filter-sterilized PBS (pH 7.4), and finally concentrated to 70× based on the initial juice volume.


Next, we used size exclusion chromatography to elute the PMP-containing fractions (maxiPURE-EVs size exclusion chromatography columns, HansaBioMed Life Sciences). To identify the PMP-containing fractions, SEC elution fractions were analyzed by absorbance measurement at 280 nm (SpectraMax® spectrophotometer), protein quantification was performed by BCA assay, and PMP concentration was determined by nanoFCM using concentration standards provided by the manufacturer. SEC fractions 4-7, which contained PMPs, were then combined under aseptic conditions in a TC hood and were syringe filter-sterilized through a 1 μm filter (glass fiber 1 μm 37 mm, VWR), 0.45 um filter (PES, Whatman® PURADISC™) and finally through a 0.45/0.2 μm set pore-size filter (Sartopore® 2, 0.2 um PES). After sterilization, PMPs were concentrated by ultracentrifugation at 40,000×g for 1.5 hours at 4° C. The pellet was resuspended in sterile PBS pH7.4 (GIBCO) and final particle concentration (4.33×1013 PMPs/mL) and median size (79.3 nm±17.2 nm) were determined by NanoFCM. Protein concentration was determined using a BCA assay (15.8 mg/mL). PMP ultrastructural characterization was performed by cryo-electron microscopy.


b) Production of Blueberry PMPs


Blueberries (2.5 kg) were obtained from a local grocery market. Fruits were washed with 1% detergent (Alconox®) and rinsed under warm water. Next, the blueberry juice was isolated using a juicer/mincer machine (Slowstar). 2.6 L of PBS pH 7.4 buffer was added during the juicing process in order to retrieve the extracted material while reducing juice viscosity to a final volume of 5.2 L. The collected juice went through a progressive clarification process. First, the juice was brought to pH 4 with 10N sodium hydroxide solution (VWR Chemicals BDH®), before the addition of pectinase enzyme at a final concentration of 1 U/mL (Pectinase from Aspergillus niger, Sigma-Aldrich 17389). The enzymatic digestion of pectin was performed at room temperature (23° C.-25° C.) for at least 2 hours. Then, a series of differential centrifugations at 3000× g for 20 minutes and 10,000× g for 40 minutes was carried out to remove large debris. Subsequent juice clarification was performed using a Miracloth mesh (20-25 μm, Sigma-Aldrich) to remove additional debris. EDTA was added to the 4.5 L of juice at a final concentration of 50 mM, and pH was brought to pH 7.5. Further clarification of the juice was performed by vacuum-filtration through 11 μm disk filters (Whatman®) set on a funnel-flask system, followed by 1 μm filtration (Glass fiber, Acrodisc®, Pall Laboratory) set on a peristaltic pump system (250 LMH, liter/m2/hour) and a 0.45 μm filter (PES, CELLTREAT® Scientific Products) set on a vacuum system. The clarified juice was stored at 4° C. overnight. Next, the juice was processed using a TFF system (Repligen, KrosFlo® KR2i TFF System) with a TFF mPES hollow fiber filter (mPES, 300 kDa pore size), sequentially concentrated 10× (1.6 L), washed by diafiltration with 10 diavolumes of filter sterilized PBS pH7.4, and finally concentrated to 70× based on the initial juice volume.


Next, we used size exclusion chromatography to elute the PMP-containing fractions (maxiPURE-EVs size exclusion chromatography columns, HansaBioMed Life Sciences). To identify the PMP-containing fractions, SEC elution fractions were analyzed by absorbance measurement at 280 nm (SpectraMax® spectrophotometer), protein quantification was performed by BCA assay, and PMP concentration was determined by nanoFCM using concentration standards provided by the manufacturer. Based on nanoFCM particle counts, blueberry PMPs were eluted in early SEC fractions 6-8, and a high amount of contaminants are present in late SEC fractions (9-14). SEC fractions 6-8, which contained PMPs, were then combined under aseptic conditions in a TC hood and were syringe filter-sterilized through a 1 μm filter (Glass fiber, Acrodisc®, Pall Laboratory), and finally through a 0.45 μm filter (PES, Whatman® Puradisc). After sterilization, PMPs were concentrated by ultracentrifugation at 40,000×g for 1.5 h at 4° C. The pellet was resuspended in sterile PBS pH 7.4 (GIBCO), and final particle concentration (4.04×1011 PMPs/mL) and median size (77.2 nm+/−17.9 nm) were determined by NanoFCM. Protein concentration was determined by BCA assay (2 μg/mL). PMP ultrastructural characterization was performed by cryo-electron microscopy.


c) Production of Wolffia globosa (Duckweed) PMPs



Wolffia globosa strain 9349-31 was obtained from Rutgers Duckweed Stock Cooperative and cultured in house in frond medium at pH 5.8 (3.2 g/L Schenk & Hildebrandt basal salt mixture, 20 g/L sucrose, pH 5.8 adjusted with KOH) at 25° C. under continuous light and 100 rpm agitation conditions (Percival incubator). Wolffia globosa whole plants were harvested at 5% (weight/volume) density from 400 mL of culture through filtration using a Miracloth mesh (20-25 um, Sigma-Aldrich). Next, approximately 18 g of plants were blended in a food processor (Oster, 2 minutes at maximum speed) with the addition of 135 mL of PBS pH 7.4. The blended plants were processed through a progressive clarification process. First, the blended plants were brought to pH 4 with 10N sodium hydroxide solution (VWR Chemicals BDH®), before the addition of pectinase enzyme at a final concentration of 0.5 U/mL (Pectinase from Aspergillus niger, Sigma-Aldrich 17389). The enzymatic digestion of pectin was performed at room temperature for at least 2 hours (25° C.). Then, a series of differential centrifugations at 3000× g for 20 minutes was carried out to remove large debris. Subsequent clarification was performed using Miracloth mesh (20-25 μm, Sigma-Aldrich) to remove additional debris. EDTA was added to the blended extract at a final concentration of 50 mM, and pH was brought to pH 7.5. Further clarification was done by vacuum-filtration through 11 μm disk filters (Whatman®) set on a funnel-flask system, followed by 1 μm syringe filtration (Glass fiber, Acrodisc®, Pall Laboratory) and a 0.45 μm filter (PES, CELLTREAT® Scientific Products) set on a vacuum system. The clarified blended-plant solution was stored at 4° C. overnight. Samples were then processed successively using a TFF system and size exclusion chromatography. The TFF system (Repligen, KrosFlo® KR2i TFF System) used was set with a TFF mPES hollow fiber filter (mPES, 300 kDa pore size). The samples were sequentially concentrated 12.5×, washed by diafiltration with 7 diavolumes of filter sterilized PBS pH 7.4, and finally concentrated to 50× based on the initial juice volume.


Next, we used size exclusion chromatography to elute the PMP-containing fractions (maxiPURE-EVs size exclusion chromatography columns, HansaBioMed Life Sciences). To identify the PMP-containing fractions, SEC elution fractions were analyzed by absorbance measurement at 280 nm (SpectraMax® spectrophotometer), protein quantification was performed by BCA assay, and PMP concentration was determined by nanoFCM using concentration standards provided by the manufacturer. SEC fractions 4-7, which contained PMPs, were then combined under aseptic conditions in a TC hood and were syringe filter-sterilized through a 1 μm filter (Glass fiber, Acrodisc®, Pall Laboratory), and finally through an 0.45 μm filter (PES, Whatman® Puradisc). After sterilization, PMPs were concentrated by ultracentrifugation at 40,000×g, for 1.5 h at 4° C. The pellet was resuspended in sterile PBS pH7.4 (GIBCO) and final particle concentration (8.79×1012 PMPs/mL), and size (91.8 nm±23.4 nm) were determined by nanoFCM. Protein concentration was determined by BCA assay (2.99 mg/mL).


d) Production of Broccoli PMPs


Broccoli crowns were obtained from a local grocery market. Broccoli crowns were hand-washed with 1% detergent (Alconox®) and rinsed under warm water. Next, approximately 1.3 kg of broccoli was isolated using a juicer/mincer machine (Slowstar) with the addition of PBS pH 7.4. The resulting juice (800 mL) was processed through a 1 mm mesh metal strainer and Miracloth mesh (20-25 μm) to remove large debris. The broccoli juice was next processed through a progressive clarification process. First, the juice was brought to pH 4 with 10N sodium hydroxide solution (VWR Chemicals BDH®), before the addition of pectinase enzyme at a final concentration of 0.5 U/mL (Pectinase from Aspergillus niger, Sigma-Aldrich 17389). The enzymatic digestion of pectin was performed at room temperature for at least 2 hours (25° C.). Then, a series of differential centrifugations at 3000× g for 20 minutes and 10,000× g for 40 minutes to remove large debris were carried out. EDTA was added to the juice at a final concentration of 50 mM, and pH was brought to pH 7.5.


Further clarification of the juice was performed by vacuum filtration through 11 μm disk filters (Whatman®) set on a funnel-flask system, followed by 1 μm glass fiber syringe filtration (Glass fiber, Acrodisc®, Pall Laboratory) and 0.45 μm filtration (PES, CELLTREAT® Scientific Products). Next, the juice was processed using a TFF system (Repligen, KrosFlo® KR2i TFF System) with a TFF mPES hollow fiber filter (mPES, 300 kDa pore size) sequentially concentrated 10× (1.6 L), washed by diafiltration with 10 diavolumes of filter sterilized PBS (pH 7.4), and finally concentrated to 50× based on the initial juice volume.


Next, we used size exclusion chromatography to elute the PMP-containing fractions (maxiPURE-EVs, HansaBioMed Life Sciences). To identify the PMP-containing fractions, SEC elution fractions were analyzed by absorbance measurement at 280 nm (SpectraMax® spectrophotometer), protein quantification was performed by BCA assay, and PMP concentration was determined by nanoFCM using concentration standards provided by the manufacturer. SEC fractions 3-7, which contained PMPs, were then combined under aseptic conditions in a TC hood and were syringe filter-sterilized through a 1 μm filter (Glass fiber, Acrodisc®, Pall Laboratory), 0.45 μm filter (PES, Whatman® Puradisc) and finally through a 0.2 μm filter (PES, Whatman® Puradisc). After sterilization, PMPs were concentrated by ultracentrifugation at 40,000×g, for 1.5 h at 4° C. The resulting pellet was resuspended in sterile PBS pH7.4 (GIBCO), and final particle concentration (1.69×1012 PMPs/mL), and size (80.3 nm±20.9 nm) were determined by NanoFCM. Protein concentration was determined by BCA assay (1.25 mg/mL). PMP ultrastructural characterization was performed by cryo-electron microscopy.


e) Production of PMPs from Other Plant Sources


Using methods described above (e.g., in Examples 18 and 19), PMPs were also produced from avocado, grape, tomato fruits, and onion.


In some examples, PMPs are produced from plant culture, e.g., produced from plant cells (e.g., cell culture), plant tissue, plant parts, or whole plants grown in a culture media and processed using the methods described above (e.g., in Examples 18 and 19).


Other Embodiments

Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, the descriptions and examples should not be construed as limiting the scope of the invention. The disclosures of all patent and scientific literature cited herein are expressly incorporated in their entirety by reference.


Other embodiments are within the claims.









APPENDIX





Table 1: Plant EV-Markers

















Example Species
Accession No.
Protein Name






Arabidopsis thaliana

C0LGG8
Probable LRR receptor-like serine/threonine-protein kinase




At1g53430 (EC 2.7.11.1)



Arabidopsis thaliana

F4HQT8
Uncharacterized protein



Arabidopsis thaliana

F4HWU0
Protein kinase superfamily protein



Arabidopsis thaliana

F4I082
Bifunctional inhibitor/lipid-transfer protein/seed storage 2S




albumin superfamily protein



Arabidopsis thaliana

F4I3M3
Kinase with tetratricopeptide repeat domain-containing




protein



Arabidopsis thaliana

F4IB62
Leucine-rich repeat protein kinase family protein



Arabidopsis thaliana

O03042
Ribulose bisphosphate carboxylase large chain (RuBisCO




large subunit) (EC 4.1.1.39)



Arabidopsis thaliana

O03986
Heat shock protein 90-4 (AtHSP90.4) (AtHsp90-4) (Heat




shock protein 81-4) (Hsp81-4)



Arabidopsis thaliana

O04023
Protein SRC2 homolog (AtSRC2)



Arabidopsis thaliana

O04309
Jacalin-related lectin 35 (JA-responsive protein 1)




(Myrosinase-binding protein-like At3g16470)



Arabidopsis thaliana

O04314
PYK10-binding protein 1 (Jacalin-related lectin 30) (Jasmonic




acid-induced protein)



Arabidopsis thaliana

O04922
Probable glutathione peroxidase 2 (EC 1.11.1.9)



Arabidopsis thaliana

O22126
Fasciclin-like arabinogalactan protein 8 (AtAGP8)



Arabidopsis thaliana

O23179
Patatin-like protein 1 (AtPLP1 (EC 3.1.1.—) (Patatin-related




phospholipase A IIgamma) (pPLAIIg) (Phospholipase A IVA)




(AtPLAIVA)



Arabidopsis thaliana

O23207
Probable NAD(P)H dehydrogenase (quinone) FQR1-like 2




(EC 1.6.5.2)



Arabidopsis thaliana

O23255
Adenosylhomocysteinase 1 (AdoHcyase 1) (EC 3.3.1.1)




(Protein EMBRYO DEFECTIVE 1395) (Protein




HOMOLOGY-DEPENDENT GENE SILENCING 1)




(S-adenosyl-L-homocysteine hydrolase 1)




(SAH hydrolase 1)



Arabidopsis thaliana

O23482
Oligopeptide transporter 3 (AtOPT3)



Arabidopsis thaliana

O23654
V-type proton ATPase catalytic subunit A (V-ATPase subunit




A) (EC 3.6.3.14) (V-ATPase 69 kDa subunit) (Vacuolar H(+)-




ATPase subunit A) (Vacuolar proton pump subunit alpha)



Arabidopsis thaliana

O48788
Probable inactive receptor kinase At2g26730



Arabidopsis thaliana

O48963
Phototropin-1 (EC 2.7.11.1) (Non-phototropic hypocotyl




protein 1) (Root phototropism protein 1)



Arabidopsis thaliana

O49195
Vegetative storage protein 1



Arabidopsis thaliana

O50008
5-methyltetrahydropteroyltriglutamate--homocysteine




methyltransferase 1 (EC 2.1.1.14) (Cobalamin-independent




methionine synthase 1) (AtMS1) (Vitamin-B12-independent




methionine synthase 1)



Arabidopsis thaliana

O64696
Putative uncharacterized protein At2g34510



Arabidopsis thaliana

O65572
Carotenoid 9,10(9′,10′)-cleavage dioxygenase 1 (EC




1.14.99.n4) (AtCCD1) (Neoxanthin cleavage enzyme NC1)




(AtNCED1)



Arabidopsis thaliana

O65660
PLAT domain-containing protein 1 (AtPLAT1) (PLAT domain




protein 1)



Arabidopsis thaliana

O65719
Heat shock 70 kDa protein 3 (Heat shock cognate 70 kDa




protein 3) (Heat shock cognate protein 70-3) (AtHsc70-3)




(Heat shock protein 70-3) (AtHsp70-3)



Arabidopsis thaliana

O80517
Uclacyanin-2 (Blue copper-binding protein II) (BCB II)




(Phytocyanin 2) (Uclacyanin-II)



Arabidopsis thaliana

O80576
At2g44060 (Late embryogenesis abundant protein, group 2)




(Similar to late embryogenesis abundant proteins)



Arabidopsis thaliana

O80725
ABC transporter B family member 4 (ABC transporter




ABCB.4) (AtABCB4) (Multidrug resistance protein 4)




(P-glycoprotein 4)



Arabidopsis thaliana

O80837
Remorin (DNA-binding protein)



Arabidopsis thaliana

O80852
Glutathione S-transferase F9 (AtGSTF9) (EC 2.5.1.18)




(AtGSTF7) (GST class-phi member 9)



Arabidopsis thaliana

O80858
Expressed protein (Putative uncharacterized protein




At2g30930) (Putative uncharacterized protein At2g30930;




F7F1.14)



Arabidopsis thaliana

O80939
L-type lectin-domain containing receptor kinase IV.1




(Arabidopsis thaliana lectin-receptor kinase e) (AthlecRK-e)




(LecRK-IV.1) (EC 2.7.11.1) (Lectin Receptor Kinase 1)



Arabidopsis thaliana

O80948
Jacalin-related lectin 23 (Myrosinase-binding protein-like




At2g39330)



Arabidopsis thaliana

O82628
V-type proton ATPase subunit G1 (V-ATPase subunit G1)




(Vacuolar H(+)-ATPase subunit G isoform 1) (Vacuolar




proton pump subunit G1)



Arabidopsis thaliana

P10795
Ribulose bisphosphate carboxylase small chain 1A,




chloroplastic (RuBisCO small subunit 1A) (EC 4.1.1.39)



Arabidopsis thaliana

P10896
Ribulose bisphosphate carboxylase/oxygenase activase,




chloroplastic (RA) (RuBisCO activase)



Arabidopsis thaliana

P17094
60S ribosomal protein L3-1 (Protein EMBRYO DEFECTIVE




2207)



Arabidopsis thaliana

P19456
ATPase 2, plasma membrane-type (EC 3.6.3.6) (Proton




pump 2)



Arabidopsis thaliana

P20649
ATPase 1, plasma membrane-type (EC 3.6.3.6) (Proton




pump 1)



Arabidopsis thaliana

P22953
Probable mediator of RNA polymerase II transcription subunit




37e (Heat shock 70 kDa protein 1) (Heat shock cognate 70




kDa protein 1) (Heat shock cognate protein 70-1) (AtHsc70-1)




(Heat shock protein 70-1) (AtHsp70-1) (Protein




EARLY-RESPONSIVE TO DEHYDRATION 2)



Arabidopsis thaliana

P23586
Sugar transport protein 1 (Glucose transporter) (Hexose




transporter 1)



Arabidopsis thaliana

P24636
Tubulin beta-4 chain (Beta-4-tubulin)



Arabidopsis thaliana

P25696
Bifunctional enolase 2/transcriptional activator (EC 4.2.1.11)




(2-phospho-D-glycerate hydro-lyase 2) (2-phosphoglycerate




dehydratase 2) (LOW EXPRESSION OF OSMOTICALLY




RESPONSIVE GENES 1)



Arabidopsis thaliana

P25856
Glyceraldehyde-3-phosphate dehydrogenase GAPA1,




chloroplastic (EC 1.2.1.13) (NADP-dependent




glyceraldehydephosphate dehydrogenase A subunit 1)



Arabidopsis thaliana

P28186
Ras-related protein RABE1c (AtRABE1c) (Ras-related




protein Ara-3) (Ras-related protein Rab8A) (AtRab8A)



Arabidopsis thaliana

P30302
Aquaporin PIP2-3 (Plasma membrane intrinsic protein 2-3)




(AtPIP2; 3) (Plasma membrane intrinsic protein 2c) (PIP2c)




(RD28-PIP) (TMP2C) (Water stress-induced tonoplast




intrinsic protein) (WSI-TIP) [Cleaved into: Aquaporin PIP2-3,




N-terminally processed]



Arabidopsis thaliana

P31414
Pyrophosphate-energized vacuolar membrane proton pump




1 (EC 3.6.1.1) (Pyrophosphate-energized inorganic




pyrophosphatase 1) (H(+)-PPase 1) (Vacuolar proton




pyrophosphatase 1) (Vacuolar proton pyrophosphatase 3)



Arabidopsis thaliana

P32961
Nitrilase 1 (EC 3.5.5.1)



Arabidopsis thaliana

P38666
60S ribosomal protein L24-2 (Protein SHORT VALVE 1)



Arabidopsis thaliana

P39207
Nucleoside diphosphate kinase 1 (EC 2.7.4.6) (Nucleoside




diphosphate kinase I) (NDK I) (NDP kinase I) (NDPK I)



Arabidopsis thaliana

P42643
14-3-3-like protein GF14 chi (General regulatory factor 1)



Arabidopsis thaliana

P42737
Beta carbonic anhydrase 2, chloroplastic (AtbCA2)




(AtbetaCA2) (EC 4.2.1.1) (Beta carbonate dehydratase 2)



Arabidopsis thaliana

P42759
Dehydrin ERD10 (Low-temperature-induced protein LTI45)



Arabidopsis thaliana

P42761
Glutathione S-transferase F10 (AtGSTF10) (EC 2.5.1.18)




(AtGSTF4) (GST class-phi member 10) (Protein EARLY




RESPONSE TO DEHYDRATION 13)



Arabidopsis thaliana

P42763
Dehydrin ERD14



Arabidopsis thaliana

P42791
60S ribosomal protein L18-2



Arabidopsis thaliana

P43286
Aquaporin PIP2-1 (Plasma membrane intrinsic protein 2-1)




(AtPIP2; 1) (Plasma membrane intrinsic protein 2a) (PIP2a)




[Cleaved into: Aquaporin PIP2-1, N-terminally processed]



Arabidopsis thaliana

P46286
60S ribosomal protein L8-1 (60S ribosomal protein L2)




(Protein EMBRYO DEFECTIVE 2296)



Arabidopsis thaliana

P46422
Glutathione S-transferase F2 (AtGSTF2) (EC 2.5.1.18) (24




kDa auxin-binding protein) (AtPM24) (GST class-phi member




2)



Arabidopsis thaliana

P47998
Cysteine synthase 1 (EC 2.5.1.47) (At.OAS.5-8) (Beta-substituted




Ala synthase 1; 1) (ARAth-Bsas1; 1) (CSase A)




(AtCS-A) (Cys-3A) (O-acetylserine (thiol)-lyase 1) (OAS-TL




A) (O-acetylserine sulfhydrylase) (Protein ONSET OF LEAF




DEATH 3)



Arabidopsis thaliana

P48347
14-3-3-like protein GF14 epsilon (General regulatory factor




10)



Arabidopsis thaliana

P48491
Triosephosphate isomerase, cytosolic (TIM) (Triose-phosphate




isomerase) (EC 5.3.1.1)



Arabidopsis thaliana

P50318
Phosphoglycerate kinase 2, chloroplastic (EC 2.7.2.3)



Arabidopsis thaliana

P53492
Actin-7 (Actin-2)



Arabidopsis thaliana

P54144
Ammonium transporter 1 member 1 (AtAMT1; 1)



Arabidopsis thaliana

P92963
Ras-related protein RABB1c (AtRABB1c) (Ras-related




protein Rab2A) (AtRab2A)



Arabidopsis thaliana

P93004
Aquaporin PIP2-7 (Plasma membrane intrinsic protein 2-7)




(AtPIP2; 7) (Plasma membrane intrinsic protein 3) (Salt




stress-induced major intrinsic protein) [Cleaved into:




Aquaporin PIP2-7, N-terminally processed]



Arabidopsis thaliana

P93025
Phototropin-2 (EC 2.7.11.1) (Defective in chloroplast




avoidance protein 1) (Non-phototropic hypocotyl 1-like




protein 1) (AtKin7) (NPH1-like protein 1)



Arabidopsis thaliana

P93819
Malate dehydrogenase 1, cytoplasmic (EC 1.1.1.37)




(Cytosolic NAD-dependent malate dehydrogenase 1)




(cNAD-MDH1) (Cytosolic malate dehydrogenase 1)




(Cytosolic MDH1)



Arabidopsis thaliana

Q03250
Glycine-rich RNA-binding protein 7 (AtGR-RBP7) (AtRBG7)




(Glycine-rich protein 7) (AtGRP7) (Protein COLD,




CIRCADIAN RHYTHM, AND RNA BINDING 2) (Protein




CCR2)



Arabidopsis thaliana

Q05431
L-ascorbate peroxidase 1, cytosolic (AP) (AtAPx01) (EC




1.11.1.11)



Arabidopsis thaliana

Q06611
Aquaporin PIP1-2 (AtPIP1; 2) (Plasma membrane intrinsic




protein 1b) (PIP1b) (Transmembrane protein A) (AthH2)




(TMP-A)



Arabidopsis thaliana

Q07488
Blue copper protein (Blue copper-binding protein) (AtBCB)




(Phytocyanin 1) (Stellacyanin)



Arabidopsis thaliana

Q0WLB5
Clathrin heavy chain 2



Arabidopsis thaliana

Q0WNJ6
Clathrin heavy chain 1



Arabidopsis thaliana

Q1ECE0
Vesicle-associated protein 4-1 (Plant VAP homolog 4-1)




(AtPVA41) (Protein MEMBRANE-ASSOCIATED MANNITOL-INDUCED)




(AtMAMI) (VAMP-associated protein 4-1)



Arabidopsis thaliana

Q38882
Phospholipase D alpha 1 (AtPLDalpha1) (PLD alpha 1) (EC




3.1.4.4) (Choline phosphatase 1) (PLDalpha)




(Phosphatidylcholine-hydrolyzing phospholipase D 1)



Arabidopsis thaliana

Q38900
Peptidyl-prolyl cis-trans isomerase CYP19-1 (PPIase CYP19-1)




(EC 5.2.1.8) (Cyclophilin of 19 kDa 1) (Rotamase




cyclophilin-3)



Arabidopsis thaliana

Q39033
Phosphoinositide phospholipase C 2 (EC 3.1.4.11)




(Phosphoinositide phospholipase PLC2) (AtPLC2) (PI-PLC2)



Arabidopsis thaliana

Q39085
Delta(24)-sterol reductase (EC 1.3.1.72) (Cell elongation




protein DIMINUTO) (Cell elongation protein Dwarf1) (Protein




CABBAGE1) (Protein ENHANCED VERY-LOW-FLUENCE




RESPONSE 1)



Arabidopsis thaliana

Q39228
Sugar transport protein 4 (Hexose transporter 4)



Arabidopsis thaliana

Q39241
Thioredoxin H5 (AtTrxh5) (Protein LOCUS OF




INSENSITIVITY TO VICTORIN 1) (Thioredoxin 5) (AtTRX5)



Arabidopsis thaliana

Q39258
V-type proton ATPase subunit E1 (V-ATPase subunit E1)




(Protein EMBRYO DEFECTIVE 2448) (Vacuolar




H(+)-ATPase subunit E isoform 1) (Vacuolar proton pump subunit




E1)



Arabidopsis thaliana

Q42112
60S acidic ribosomal protein P0-2



Arabidopsis thaliana

Q42403
Thioredoxin H3 (AtTrxh3) (Thioredoxin 3) (AtTRX3)



Arabidopsis thaliana

Q42479
Calcium-dependent protein kinase 3 (EC 2.7.11.1)




(Calcium-dependent protein kinase isoform CDPK6) (AtCDPK6)



Arabidopsis thaliana

Q42547
Catalase-3 (EC 1.11.1.6)



Arabidopsis thaliana

Q56WH1
Tubulin alpha-3 chain



Arabidopsis thaliana

Q56WK6
Patellin-1



Arabidopsis thaliana

Q56X75
CASP-like protein 4D2 (AtCASPL4D2)



Arabidopsis thaliana

Q56ZI2
Patellin-2



Arabidopsis thaliana

Q7Y208
Glycerophosphodiester phosphodiesterase GDPDL1 (EC




3.1.4.46) (Glycerophosphodiester phosphodiesterase-like 1)




(ATGDPDL1) (Glycerophosphodiesterase-like 3) (Protein




SHV3-LIKE 2)



Arabidopsis thaliana

Q84VZ5
Uncharacterized GPI-anchored protein At5g19240



Arabidopsis thaliana

Q84WU7
Eukaryotic aspartyl protease family protein (Putative




uncharacterized protein At3g51330)



Arabidopsis thaliana

Q8GUL8
Uncharacterized GPI-anchored protein At5g19230



Arabidopsis thaliana

Q8GYA4
Cysteine-rich receptor-like protein kinase 10 (Cysteine-rich




RLK10) (EC 2.7.11.—) (Receptor-like protein kinase 4)



Arabidopsis thaliana

Q8GYN5
RPM1-interacting protein 4



Arabidopsis thaliana

Q8GZ99
At5g49760 (Leucine-rich repeat protein kinase family protein)




(Leucine-rich repeat receptor-like protein kinase) (Putative




receptor protein kinase)



Arabidopsis thaliana

Q8L636
Sodium/calcium exchanger NCL (Na(+)/Ca(2+)-exchange




protein NCL) (Protein NCX-like) (AtNCL)



Arabidopsis thaliana

Q8L7S1
At1g45200 (At1g45200/At1g45200) (Triacylglycerol




lipase-like 1)



Arabidopsis thaliana

Q8LAA6
Probable aquaporin PIP1-5 (AtPIP1; 5) (Plasma membrane




intrinsic protein 1d) (PIP1d)



Arabidopsis thaliana

Q8LCP6
Endoglucanase 10 (EC 3.2.1.4) (Endo-1,4-beta glucanase




10)



Arabidopsis thaliana

Q8RWV0
Transketolase-1, chloroplastic (TK) (EC 2.2.1.1)



Arabidopsis thaliana

Q8S8Q6
Tetraspanin-8



Arabidopsis thaliana

Q8VZG8
MDIS1-interacting receptor like kinase 2 (AtMIK2) (Probable




LRR receptor-like serine/threonine-protein kinase




At4g08850) (EC 2.7.11.1)



Arabidopsis thaliana

Q8VZU2
Syntaxin-132 (AtSYP132)



Arabidopsis thaliana

Q8W4E2
V-type proton ATPase subunit B3 (V-ATPase subunit B3)




(Vacuolar H(+)-ATPase subunit B isoform 3) (Vacuolar




proton pump subunit B3)



Arabidopsis thaliana

Q8W4S4
V-type proton ATPase subunit a3 (V-ATPase subunit a3)




(V-type proton ATPase 95 kDa subunit a isoform 3) (V-ATPase




95 kDa isoform a3) (Vacuolar H(+)-ATPase subunit a isoform




3) (Vacuolar proton pump subunit a3) (Vacuolar proton




translocating ATPase 95 kDa subunit a isoform 3)



Arabidopsis thaliana

Q93VG5
40S ribosomal protein S8-1



Arabidopsis thaliana

Q93XY5
Tetraspanin-18 (TOM2A homologous protein 2)



Arabidopsis thaliana

Q93YS4
ABC transporter G family member 22 (ABC transporter




ABCG.22) (AtABCG22) (White-brown complex homolog




protein 23) (AtWBC23)



Arabidopsis thaliana

Q93Z08
Glucan endo-1,3-beta-glucosidase 6 (EC 3.2.1.39)




((1 −> 3)-beta-glucan endohydrolase 6)




((1 −> 3)-beta-glucanase 6) (Beta-1,3-endoglucanase




6) (Beta-1,3-glucanase 6)



Arabidopsis thaliana

Q940M8
3-oxo-5-alpha-steroid 4-dehydrogenase (DUF1295)




(At1g73650/F25P22_7)



Arabidopsis thaliana

Q944A7
Probable serine/threonine-protein kinase At4g35230 (EC




2.7.11.1)



Arabidopsis thaliana

Q944G5
Protein NRT1/PTR FAMILY 2.10 (AtNPF2.10) (Protein




GLUCOSINOLATE TRANSPORTER-1)



Arabidopsis thaliana

Q94AZ2
Sugar transport protein 13 (Hexose transporter 13)




(Multicopy suppressor of snf4 deficiency protein 1)



Arabidopsis thaliana

Q94BT2
Auxin-induced in root cultures protein 12



Arabidopsis thaliana

Q94CE4
Beta carbonic anhydrase 4 (AtbCA4) (AtbetaCA4) (EC




4.2.1.1) (Beta carbonate dehydratase 4)



Arabidopsis thaliana

Q94KI8
Two pore calcium channel protein 1 (Calcium channel protein




1) (AtCCH1) (Fatty acid oxygenation up-regulated protein 2)




(Voltage-dependent calcium channel protein TPC1) (AtTPC1)



Arabidopsis thaliana

Q96262
Plasma membrane-associated cation-binding protein 1




(AtPCAP1) (Microtubule-destabilizing protein 25)



Arabidopsis thaliana

Q9C5Y0
Phospholipase D delta (AtPLDdelta) (PLD delta) (EC 3.1.4.4)



Arabidopsis thaliana

Q9C7F7
Non-specific lipid transfer protein GPI-anchored 1




(AtLTPG-1) (Protein LTP-GPI-ANCHORED 1)



Arabidopsis thaliana

Q9C821
Proline-rich receptor-like protein kinase PERK15 (EC




2.7.11.1) (Proline-rich extensin-like receptor kinase 15)




(AtPERK15)



Arabidopsis thaliana

Q9C8G5
CSC1-like protein ERD4 (Protein EARLY-RESPONSIVE TO




DEHYDRATION STRESS 4)



Arabidopsis thaliana

Q9C9C5
60S ribosomal protein L6-3



Arabidopsis thaliana

Q9CAR7
Hypersensitive-induced response protein 2 (AtHIR2)



Arabidopsis thaliana

Q9FFH6
Fasciclin-like arabinogalactan protein 13



Arabidopsis thaliana

Q9FGT8
Temperature-induced lipocalin-1 (AtTIL1)



Arabidopsis thaliana

Q9FJ62
Glycerophosphodiester phosphodiesterase GDPDL4 (EC




3.1.4.46) (Glycerophosphodiester phosphodiesterase-like 4)




(ATGDPDL4) (Glycerophosphodiesterase-like 1) (Protein




SHV3-LIKE 1)



Arabidopsis thaliana

Q9FK68
Ras-related protein RABA1c (AtRABA1c)



Arabidopsis thaliana

Q9FKS8
Lysine histidine transporter 1



Arabidopsis thaliana

Q9FM65
Fasciclin-like arabinogalactan protein 1



Arabidopsis thaliana

Q9FNH6
NDR1/HIN1-like protein 3



Arabidopsis thaliana

Q9FRL3
Sugar transporter ERD6-like 6



Arabidopsis thaliana

Q9FWR4
Glutathione S-transferase DHAR1, mitochondrial (EC




2.5.1.18) (Chloride intracellular channel homolog 1) (CLIC




homolog 1) (Glutathione-dependent dehydroascorbate




reductase 1) (AtDHAR1) (GSH-dependent dehydroascorbate




reductase 1) (mtDHAR)



Arabidopsis thaliana

Q9FX54
Glyceraldehyde-3-phosphate dehydrogenase GAPC2,




cytosolic (EC 1.2.1.12) (NAD-dependent




glyceraldehydephosphate dehydrogenase C subunit 2)



Arabidopsis thaliana

Q9LE22
Probable calcium-binding protein CML27 (Calmodulin-like




protein 27)



Arabidopsis thaliana

Q9LEX1
At3g61050 (CaLB protein) (Calcium-dependent lipid-binding




(CaLB domain) family protein)



Arabidopsis thaliana

Q9LF79
Calcium-transporting ATPase 8, plasma membrane-type (EC




3.6.3.8) (Ca(2+)-ATPase isoform 8)



Arabidopsis thaliana

Q9LJG3
GDSL esterase/lipase ESM1 (EC 3.1.1.—) (Extracellular lipase




ESM1) (Protein EPITHIOSPECIFIER MODIFIER 1)




(AtESM1)



Arabidopsis thaliana

Q9LJI5
V-type proton ATPase subunit d1 (V-ATPase subunit d1)




(Vacuolar H(+)-ATPase subunit d isoform 1) (Vacuolar proton




pump subunit d1)



Arabidopsis thaliana

Q9LME4
Probable protein phosphatase 2C 9 (AtPP2C09) (EC




3.1.3.16) (Phytochrome-associated protein phosphatase 2C)




(PAPP2C)



Arabidopsis thaliana

Q9LNP3
At1g17620/F11A6_23 (F1L3.32) (Late embryogenesis




abundant (LEA) hydroxyproline-rich glycoprotein family)




(Putative uncharacterized protein At1g17620)



Arabidopsis thaliana

Q9LNW1
Ras-related protein RABA2b (AtRABA2b)



Arabidopsis thaliana

Q9LQU2
Protein PLANT CADMIUM RESISTANCE 1 (AtPCR1)



Arabidopsis thaliana

Q9LQU4
Protein PLANT CADMIUM RESISTANCE 2 (AtPCR2)



Arabidopsis thaliana

Q9LR30
Glutamate--glyoxylate aminotransferase 1 (AtGGT2) (EC




2.6.1.4) (Alanine aminotransferase GGT1) (EC 2.6.1.2)




(Alanine--glyoxylate aminotransferase GGT1) (EC 2.6.1.44)




(Alanine-2-oxoglutarate aminotransferase 1) (EC 2.6.1.—)



Arabidopsis thaliana

Q9LSI9
Inactive LRR receptor-like serine/threonine-protein kinase




BIR2 (Protein BAK1-INTERACTING RECEPTOR-LIKE




KINASE 2)



Arabidopsis thaliana

Q9LSQ5
NAD(P)H dehydrogenase (quinone) FQR1 (EC 1.6.5.2)




(Flavodoxin-like quinone reductase 1)



Arabidopsis thaliana

Q9LUT0
Protein kinase superfamily protein (Putative uncharacterized




protein At3g17410) (Serine/threonine protein kinase-like




protein)



Arabidopsis thaliana

Q9LV48
Proline-rich receptor-like protein kinase PERK1 (EC 2.7.11.1)




(Proline-rich extensin-like receptor kinase 1) (AtPERK1)



Arabidopsis thaliana

Q9LX65
V-type proton ATPase subunit H (V-ATPase subunit H)




(Vacuolar H(+)-ATPase subunit H) (Vacuolar proton pump




subunit H)



Arabidopsis thaliana

Q9LYG3
NADP-dependent malic enzyme 2 (AtNADP-ME2)




(NADP-malic enzyme 2) (EC 1.1.1.40)



Arabidopsis thaliana

Q9M088
Glucan endo-1,3-beta-glucosidase 5 (EC 3.2.1.39)




((1 −> 3)-beta-glucan endohydrolase 5)




((1 −> 3)-beta-glucanase 5) (Beta-1,3-endoglucanase




5) (Beta-1,3-glucanase 5)



Arabidopsis thaliana

Q9M2D8
Uncharacterized protein At3g61260



Arabidopsis thaliana

Q9M386
Late embryogenesis abundant (LEA) hydroxyproline-rich




glycoprotein family (Putative uncharacterized protein




At3g54200) (Putative uncharacterized protein F24B22.160)



Arabidopsis thaliana

Q9M390
Protein NRT1/PTR FAMILY 8.1 (AtNPF8.1) (Peptide




transporter PTR1)



Arabidopsis thaliana

Q9M5P2
Secretory carrier-associated membrane protein 3 (AtSC3)




(Secretory carrier membrane protein 3)



Arabidopsis thaliana

Q9M8T0
Probable inactive receptor kinase At3g02880



Arabidopsis thaliana

Q9SDS7
V-type proton ATPase subunit C (V-ATPase subunit C)




(Vacuolar H(+)-ATPase subunit C) (Vacuolar proton pump




subunit C)



Arabidopsis thaliana

Q9SEL6
Vesicle transport v-SNARE 11 (AtVTI11) (Protein SHOOT




GRAVITROPISM 4) (Vesicle soluble NSF attachment protein




receptor VTI1a) (AtVTI1a) (Vesicle transport v-SNARE




protein VTI1a)



Arabidopsis thaliana

Q9SF29
Syntaxin-71 (AtSYP71)



Arabidopsis thaliana

Q9SF85
Adenosine kinase 1 (AK 1) (EC 2.7.1.20) (Adenosine




5′-phosphotransferase 1)



Arabidopsis thaliana

Q9SIE7
PLAT domain-containing protein 2 (AtPLAT2) (PLAT domain




protein 2)



Arabidopsis thaliana

Q9SIM4
60S ribosomal protein L14-1



Arabidopsis thaliana

Q9SIU8
Probable protein phosphatase 2C 20 (AtPP2C20) (EC




3.1.3.16) (AtPPC3; 1.2)



Arabidopsis thaliana

Q9SJ81
Fasciclin-like arabinogalactan protein 7



Arabidopsis thaliana

Q9SKB2
Leucine-rich repeat receptor-like serine/threonine/tyrosine-protein




kinase SOBIR1 (EC 2.7.10.1) (EC 2.7.11.1) (Protein




EVERSHED) (Protein SUPPRESSOR OF BIR1-1)



Arabidopsis thaliana

Q9SKR2
Synaptotagmin-1 (NTMC2T1.1) (Synaptotagmin A)



Arabidopsis thaliana

Q9SLF7
60S acidic ribosomal protein P2-2



Arabidopsis thaliana

Q9SPE6
Alpha-soluble NSF attachment protein 2 (Alpha-SNAP2)




(N-ethylmaleimide-sensitive factor attachment protein alpha 2)



Arabidopsis thaliana

Q9SRH6
Hypersensitive-induced response protein 3 (AtHIR3)



Arabidopsis thaliana

Q9SRY5
Glutathione S-transferase F7 (EC 2.5.1.18) (AtGSTF8) (GST




class-phi member 7) (Glutathione S-transferase 11)



Arabidopsis thaliana

Q9SRZ6
Cytosolic isocitrate dehydrogenase [NADP] (EC 1.1.1.42)



Arabidopsis thaliana

Q9SSK5
MLP-like protein 43



Arabidopsis thaliana

Q9SU13
Fasciclin-like arabinogalactan protein 2



Arabidopsis thaliana

Q9SU40
Monocopper oxidase-like protein SKU5 (Skewed roots)



Arabidopsis thaliana

Q9SUR6
Cystine lyase CORI3 (EC 4.4.1.35) (Protein CORONATINE




INDUCED 3) (Protein JASMONIC ACID RESPONSIVE 2)




(Tyrosine aminotransferase CORI3)



Arabidopsis thaliana

Q9SVC2
Syntaxin-122 (AtSYP122) (Synt4)



Arabidopsis thaliana

Q9SVF0
Putative uncharacterized protein AT4g38350 (Putative




uncharacterized protein F22I13.120)



Arabidopsis thaliana

Q9SW40
Major facilitator superfamily protein (Putative uncharacterized




protein AT4g34950) (Putative uncharacterized protein




T11I11.190)



Arabidopsis thaliana

Q9SYT0
Annexin D1 (AnnAt1) (Annexin A1)



Arabidopsis thaliana

Q9SZ11
Glycerophosphodiester phosphodiesterase GDPDL3 (EC




3.1.4.46) (Glycerophosphodiester phosphodiesterase-like 3)




(ATGDPDL3) (Glycerophosphodiesterase-like 2) (Protein




MUTANT ROOT HAIR 5) (Protein SHAVEN 3)



Arabidopsis thaliana

Q9SZN1
V-type proton ATPase subunit B2 (V-ATPase subunit B2)




(Vacuolar H(+)-ATPase subunit B isoform 2) (Vacuolar




proton pump subunit B2)



Arabidopsis thaliana

Q9SZP6
AT4g38690/F20M13_250 (PLC-like phosphodiesterases




superfamily protein) (Putative uncharacterized protein




AT4g38690) (Putative uncharacterized protein F20M13.250)



Arabidopsis thaliana

Q9SZR1
Calcium-transporting ATPase 10, plasma membrane-type




(EC 3.6.3.8) (Ca(2+)-ATPase isoform 10)



Arabidopsis thaliana

Q9T053
Phospholipase D gamma 1 (AtPLDgamma1) (PLD gamma 1)




(EC 3.1.4.4) (Choline phosphatase) (Lecithinase D)




(Lipophosphodiesterase II)



Arabidopsis thaliana

Q9T076
Early nodulin-like protein 2 (Phytocyanin-like protein)



Arabidopsis thaliana

Q9T0A0
Long chain acyl-CoA synthetase 4 (EC 6.2.1.3)



Arabidopsis thaliana

Q9T0G4
Putative uncharacterized protein AT4g10060 (Putative




uncharacterized protein T5L19.190)



Arabidopsis thaliana

Q9XEE2
Annexin D2 (AnnAt2)



Arabidopsis thaliana

Q9XGM1
V-type proton ATPase subunit D (V-ATPase subunit D)




(Vacuolar H(+)-ATPase subunit D) (Vacuolar proton pump




subunit D)



Arabidopsis thaliana

Q9XI93
At1g13930/F16A14.27 (F16A14.14) (F7A19.2 protein)




(Oleosin-B3-like protein)



Arabidopsis thaliana

Q9XIE2
ABC transporter G family member 36 (ABC transporter




ABCG.36) (AtABCG36) (Pleiotropic drug resistance protein




8) (Protein PENETRATION 3)



Arabidopsis thaliana

Q9ZPZ4
Putative uncharacterized protein (Putative uncharacterized




protein At1g09310) (T31J12.3 protein)



Arabidopsis thaliana

Q9ZQX4
V-type proton ATPase subunit F (V-ATPase subunit F)




(V-ATPase 14 kDa subunit) (Vacuolar H(+)-ATPase subunit F)




(Vacuolar proton pump subunit F)



Arabidopsis thaliana

Q9ZSA2
Calcium-dependent protein kinase 21 (EC 2.7.11.1)



Arabidopsis thaliana

Q9ZSD4
Syntaxin-121 (AtSYP121) (Syntaxin-related protein At-Syr1)



Arabidopsis thaliana

Q9ZV07
Probable aquaporin PIP2-6 (Plasma membrane intrinsic




protein 2-6) (AtPIP2; 6) (Plasma membrane intrinsic protein




2e) (PIP2e) [Cleaved into: Probable aquaporin PIP2-6,




N-terminally processed]



Arabidopsis thaliana

Q9ZVF3
MLP-like protein 328



Arabidopsis thaliana

Q9ZWA8
Fasciclin-like arabinogalactan protein 9



Arabidopsis thaliana

Q9ZSD4
SYR1, Syntaxin Related Protein 1, also known as SYP121,




PENETRATION1/PEN1 (Protein PENETRATION 1)



Citrus lemon

A1ECK0
Putative glutaredoxin



Citrus lemon

A9YVC9
Pyrophosphate--fructose 6-phosphate 1-phosphotransferase




subunit beta (PFP) (EC 2.7.1.90) (6-phosphofructokinase,




pyrophosphate dependent) (PPi-PFK)




(Pyrophosphate-dependent 6-phosphofructose-1-kinase)



Citrus lemon

B2YGY1
Glycosyltransferase (EC 2.4.1.—)



Citrus lemon

B6DZD3
Glutathione S-transferase Tau2 (Glutathione transferase




Tau2)



Citrus lemon

C3VIC2
Translation elongation factor



Citrus lemon

C8CPS0
Importin subunit alpha



Citrus lemon

D3JWB5
Flavanone 3-hydroxylase



Citrus lemon

E0ADY2
Putative caffeic acid O-methyltransferase



Citrus lemon

E5DK62
ATP synthase subunit alpha (Fragment)



Citrus lemon

E9M5S3
Putative L-galactose-1-phosphate phosphatase



Citrus lemon

F1CGQ9
Heat shock protein 90



Citrus lemon

F8WL79
Aminopeptidase (EC 3.4.11.—)



Citrus lemon

F8WL86
Heat shock protein



Citrus lemon

K9JG59
Abscisic acid stress ripening-related protein



Citrus lemon

Q000W4
Fe(III)-chelate reductase



Citrus lemon

Q39538
Heat shock protein (Fragment)



Citrus lemon

Q5UEN6
Putative signal recognition particle protein



Citrus lemon

Q8GV08
Dehydrin



Citrus lemon

Q8L893
Cytosolic phosphoglucomutase (Fragment)



Citrus lemon

Q8S990
Polygalacturonase-inhibiting protein



Citrus lemon

Q8W3U6
Polygalacturonase-inhibitor protein



Citrus lemon

Q93XL8
Dehydrin COR15



Citrus lemon

Q941Q1
Non-symbiotic hemoglobin class 1



Citrus lemon

Q9MBF3
Glycine-rich RNA-binding protein



Citrus lemon

Q9SP55
V-type proton ATPase subunit G (V-ATPase subunit G)




(Vacuolar proton pump subunit G)



Citrus lemon

Q9THJ8
Ribulose bisphosphate carboxylase large chain (EC 4.1.1.39)




(Fragment)



Citrus lemon

Q9ZST2
Pyrophosphate--fructose 6-phosphate 1-phosphotransferase




subunit alpha (PFP) (6-phosphofructokinase, pyrophosphate




dependent) (PPi-PFK) (Pyrophosphate-dependent




6-phosphofructose-1-kinase)



Citrus lemon

Q9ZWH6
Polygalacturonase inhibitor



Citrus lemon

S5DXI9
Nucleocapsid protein



Citrus lemon

S5NFC6
GTP cyclohydrolase



Citrus lemon

V4RG42
Uncharacterized protein



Citrus lemon

V4RGP4
Uncharacterized protein



Citrus lemon

V4RHN8
Uncharacterized protein



Citrus lemon

V4RJ07
Uncharacterized protein



Citrus lemon

V4RJK9
Adenosylhomocysteinase (EC 3.3.1.1)



Citrus lemon

V4RJM1
Uncharacterized protein



Citrus lemon

V4RJX1
40S ribosomal protein S6



Citrus lemon

V4RLB2
Uncharacterized protein



Citrus lemon

V4RMX8
Uncharacterized protein



Citrus lemon

V4RNA5
Uncharacterized protein



Citrus lemon

V4RP81
Glycosyltransferase (EC 2.4.1.—)



Citrus lemon

V4RPZ5
Adenylyl cyclase-associated protein



Citrus lemon

V4RTN9
Histone H4



Citrus lemon

V4RUZ4
Phosphoserine aminotransferase (EC 2.6.1.52)



Citrus lemon

V4RVF6
Uncharacterized protein



Citrus lemon

V4RXD4
Uncharacterized protein



Citrus lemon

V4RXG2
Uncharacterized protein



Citrus lemon

V4RYA0
Uncharacterized protein



Citrus lemon

V4RYE3
Uncharacterized protein



Citrus lemon

V4RYH3
Uncharacterized protein



Citrus lemon

V4RYX8
Uncharacterized protein



Citrus lemon

V4RZ12
Coatomer subunit beta'



Citrus lemon

V4RZ89
Uncharacterized protein



Citrus lemon

V4RZE3
Uncharacterized protein



Citrus lemon

V4RZF3
1,2-dihydroxy-3-keto-5-methylthiopentene dioxygenase (EC




1.13.11.54) (Acireductone dioxygenase (Fe(2+)-requiring))




(ARD) (Fe-ARD)



Citrus lemon

V4RZM7
Uncharacterized protein



Citrus lemon

V4RZX6
Uncharacterized protein



Citrus lemon

V4S1V0
Uncharacterized protein



Citrus lemon

V4S2B6
Uncharacterized protein



Citrus lemon

V4S2N1
Uncharacterized protein



Citrus lemon

V4S2S5
Uncharacterized protein (Fragment)



Citrus lemon

V4S346
Uncharacterized protein



Citrus lemon

V4S3T8
Uncharacterized protein



Citrus lemon

V4S409
Cyanate hydratase (Cyanase) (EC 4.2.1.104) (Cyanate




hydrolase) (Cyanate lyase)



Citrus lemon

V4S4E4
Histone H2B



Citrus lemon

V4S4F6
Flavin-containing monooxygenase (EC 1.—.—.—)



Citrus lemon

V4S4J1
Uncharacterized protein



Citrus lemon

V4S4K9
Uncharacterized protein



Citrus lemon

V4S535
Proteasome subunit alpha type (EC 3.4.25.1)



Citrus lemon

V4S5A8
Isocitrate dehydrogenase [NADP] (EC 1.1.1.42)



Citrus lemon

V4S5G8
Uncharacterized protein



Citrus lemon

V4S5I6
Uncharacterized protein



Citrus lemon

V4S5N4
Uncharacterized protein (Fragment)



Citrus lemon

V4S5Q3
Uncharacterized protein



Citrus lemon

V4S5X8
Uncharacterized protein



Citrus lemon

V4S5Y1
Uncharacterized protein



Citrus lemon

V4S6P4
Calcium-transporting ATPase (EC 3.6.3.8)



Citrus lemon

V4S6W0
Uncharacterized protein



Citrus lemon

V4S6W7
Uncharacterized protein (Fragment)



Citrus lemon

V4S6Y4
Uncharacterized protein



Citrus lemon

V4S773
Ribosomal protein L19



Citrus lemon

V4S7U0
Uncharacterized protein



Citrus lemon

V4S7U5
Uncharacterized protein



Citrus lemon

V4S7W4
Pyruvate kinase (EC 2.7.1.40)



Citrus lemon

V4S885
Uncharacterized protein



Citrus lemon

V4S8T3
Peptidyl-prolyl cis-trans isomerase (PPIase) (EC 5.2.1.8)



Citrus lemon

V4S920
Uncharacterized protein



Citrus lemon

V4S999
Uncharacterized protein



Citrus lemon

V4S9G5
Phosphoglycerate kinase (EC 2.7.2.3)



Citrus lemon

V4S9Q6
Beta-amylase (EC 3.2.1.2)



Citrus lemon

V4SA44
Serine/threonine-protein phosphatase (EC 3.1.3.16)



Citrus lemon

V4SAE0
Alpha-1,4 glucan phosphorylase (EC 2.4.1.1)



Citrus lemon

V4SAF6
Uncharacterized protein



Citrus lemon

V4SAI9
Eukaryotic translation initiation factor 3 subunit M (eIF3m)



Citrus lemon

V4SAJ5
Ribosomal protein



Citrus lemon

V4SAR3
Uncharacterized protein



Citrus lemon

V4SB37
Uncharacterized protein



Citrus lemon

V4SBI0
Elongation factor 1-alpha



Citrus lemon

V4SBI8
D-3-phosphoglycerate dehydrogenase (EC 1.1.1.95)



Citrus lemon

V4SBL9
Polyadenylate-binding protein (PABP)



Citrus lemon

V4SBR1
S-formylglutathione hydrolase (EC 3.1.2.12)



Citrus lemon

V4SBR6
Uncharacterized protein



Citrus lemon

V4SCG7
Uncharacterized protein



Citrus lemon

V4SCJ2
Uncharacterized protein



Citrus lemon

V4SCQ6
Peptidyl-prolyl cis-trans isomerase (PPIase) (EC 5.2.1.8)



Citrus lemon

V4SDJ8
Uncharacterized protein



Citrus lemon

V4SE41
Protein DETOXIFICATION (Multidrug and toxic compound




extrusion protein)



Citrus lemon

V4SE90
Uncharacterized protein



Citrus lemon

V4SED1
Succinate dehydrogenase [ubiquinone] flavoprotein subunit,




mitochondrial (EC 1.3.5.1)



Citrus lemon

V4SEI1
Uncharacterized protein



Citrus lemon

V4SEN9
Uncharacterized protein



Citrus lemon

V4SEX8
Uncharacterized protein



Citrus lemon

V4SF31
Uncharacterized protein



Citrus lemon

V4SF69
40S ribosomal protein S24



Citrus lemon

V4SF76
Cysteine synthase (EC 2.5.1.47)



Citrus lemon

V4SFK3
Uncharacterized protein



Citrus lemon

V4SFL4
Uncharacterized protein



Citrus lemon

V4SFW2
Uncharacterized protein



Citrus lemon

V4SGC9
Uncharacterized protein



Citrus lemon

V4SGJ4
Uncharacterized protein



Citrus lemon

V4SGN4
Uncharacterized protein



Citrus lemon

V4SGV6
Uncharacterized protein



Citrus lemon

V4SGV7
Uncharacterized protein



Citrus lemon

V4SHH1
Plasma membrane ATPase (EC 3.6.3.6) (Fragment)



Citrus lemon

V4SHI2
Uncharacterized protein



Citrus lemon

V4SHJ3
Uncharacterized protein



Citrus lemon

V4SI86
Uncharacterized protein



Citrus lemon

V4SI88
Uncharacterized protein



Citrus lemon

V4SIA2
Uncharacterized protein



Citrus lemon

V4SIC1
Phospholipase D (EC 3.1.4.4)



Citrus lemon

V4SJ14
Uncharacterized protein



Citrus lemon

V4SJ48
Uncharacterized protein



Citrus lemon

V4SJ69
Uncharacterized protein



Citrus lemon

V4SJD9
Uncharacterized protein



Citrus lemon

V4SJS7
Uncharacterized protein



Citrus lemon

V4SJT5
Uncharacterized protein



Citrus lemon

V4SKA2
Uncharacterized protein



Citrus lemon

V4SKG4
Glucose-6-phosphate isomerase (EC 5.3.1.9)



Citrus lemon

V4SKJ1
Uncharacterized protein



Citrus lemon

V4SL90
Uncharacterized protein



Citrus lemon

V4SLC6
Proteasome subunit beta type (EC 3.4.25.1)



Citrus lemon

V4SLI7
Uncharacterized protein



Citrus lemon

V4SLQ6
Uncharacterized protein



Citrus lemon

V4SMD8
Uncharacterized protein



Citrus lemon

V4SMN7
Uncharacterized protein



Citrus lemon

V4SMV5
Uncharacterized protein



Citrus lemon

V4SN00
Uncharacterized protein



Citrus lemon

V4SNA9
Uncharacterized protein



Citrus lemon

V4SNC1
Uncharacterized protein



Citrus lemon

V4SNC4
Aconitate hydratase (Aconitase) (EC 4.2.1.3)



Citrus lemon

V4SNZ3
Uncharacterized protein



Citrus lemon

V4SP86
Uncharacterized protein



Citrus lemon

V4SPM1
40S ribosomal protein S12



Citrus lemon

V4SPW4
40S ribosomal protein S4



Citrus lemon

V4SQ71
Uncharacterized protein



Citrus lemon

V4SQ89
Uncharacterized protein



Citrus lemon

V4SQ92
Uncharacterized protein



Citrus lemon

V4SQC7
Peroxidase (EC 1.11.1.7)



Citrus lemon

V4SQG3
Uncharacterized protein



Citrus lemon

V4SR15
Uncharacterized protein



Citrus lemon

V4SRN3
Transmembrane 9 superfamily member



Citrus lemon

V4SS09
Uncharacterized protein



Citrus lemon

V4SS11
Uncharacterized protein



Citrus lemon

V4SS50
Uncharacterized protein



Citrus lemon

V4SSB6
Uncharacterized protein



Citrus lemon

V4SSB8
Proteasome subunit alpha type (EC 3.4.25.1)



Citrus lemon

V4SSL7
Uncharacterized protein



Citrus lemon

V4SSQ1
Uncharacterized protein



Citrus lemon

V4SST6
Uncharacterized protein



Citrus lemon

V4SSW9
Uncharacterized protein



Citrus lemon

V4SSX5
Uncharacterized protein



Citrus lemon

V4SU82
Uncharacterized protein



Citrus lemon

V4SUD3
Uncharacterized protein



Citrus lemon

V4SUL7
Uncharacterized protein



Citrus lemon

V4SUP3
Uncharacterized protein



Citrus lemon

V4SUT4
UDP-glucose 6-dehydrogenase (EC 1.1.1.22)



Citrus lemon

V4SUY5
Uncharacterized protein



Citrus lemon

V4SV60
Serine/threonine-protein phosphatase (EC 3.1.3.16)



Citrus lemon

V4SV61
Uncharacterized protein



Citrus lemon

V4SVI5
Proteasome subunit alpha type (EC 3.4.25.1)



Citrus lemon

V4SVI6
Uncharacterized protein



Citrus lemon

V4SW04
Uncharacterized protein (Fragment)



Citrus lemon

V4SWD9
Uncharacterized protein



Citrus lemon

V4SWJ0
40S ribosomal protein S3a



Citrus lemon

V4SWQ9
Uncharacterized protein



Citrus lemon

V4SWR9
Uncharacterized protein



Citrus lemon

V4SWU9
Fructose-bisphosphate aldolase (EC 4.1.2.13)



Citrus lemon

V4SX11
Uncharacterized protein



Citrus lemon

V4SX99
Uncharacterized protein



Citrus lemon

V4SXC7
Proteasome subunit alpha type (EC 3.4.25.1)



Citrus lemon

V4SXQ5
Uncharacterized protein



Citrus lemon

V4SXW1
Beta-adaptin-like protein



Citrus lemon

V4SXY9
Uncharacterized protein



Citrus lemon

V4SY74
Uncharacterized protein



Citrus lemon

V4SY90
Uncharacterized protein



Citrus lemon

V4SY93
Uncharacterized protein



Citrus lemon

V4SYH9
Uncharacterized protein



Citrus lemon

V4SYK6
Uncharacterized protein



Citrus lemon

V4SZ03
Uncharacterized protein



Citrus lemon

V4SZ73
Uncharacterized protein



Citrus lemon

V4SZI9
Uncharacterized protein



Citrus lemon

V4SZX7
Uncharacterized protein



Citrus lemon

V4T057
Ribosomal protein L15



Citrus lemon

V4T0V5
Eukaryotic translation initiation factor 3 subunit A (eIF3a)




(Eukaryotic translation initiation factor 3 subunit 10)



Citrus lemon

V4T0Y1
Uncharacterized protein



Citrus lemon

V4T1Q6
Uncharacterized protein



Citrus lemon

V4T1U7
Uncharacterized protein



Citrus lemon

V4T2D9
Uncharacterized protein



Citrus lemon

V4T2M6
Tubulin beta chain



Citrus lemon

V4T3G2
Uncharacterized protein



Citrus lemon

V4T3P3
6-phosphogluconate dehydrogenase, decarboxylating (EC




1.1.1.44)



Citrus lemon

V4T3V9
Uncharacterized protein



Citrus lemon

V4T3Y6
Uncharacterized protein



Citrus lemon

V4T4H3
Uncharacterized protein



Citrus lemon

V4T4I7
Uncharacterized protein



Citrus lemon

V4T4M7
Superoxide dismutase [Cu—Zn] (EC 1.15.1.1)



Citrus lemon

V4T539
Uncharacterized protein



Citrus lemon

V4T541
Uncharacterized protein



Citrus lemon

V4T576
Uncharacterized protein



Citrus lemon

V4T5E1
Uncharacterized protein



Citrus lemon

V4T5I3
Uncharacterized protein



Citrus lemon

V4T5W7
Uncharacterized protein



Citrus lemon

V4T6T5
60S acidic ribosomal protein P0



Citrus lemon

V4T722
Uncharacterized protein



Citrus lemon

V4T785
Uncharacterized protein



Citrus lemon

V4T7E2
Uncharacterized protein



Citrus lemon

V4T7I7
Uncharacterized protein



Citrus lemon

V4T7N0
Proteasome subunit beta type (EC 3.4.25.1)



Citrus lemon

V4T7N4
Uncharacterized protein



Citrus lemon

V4T7T2
Uncharacterized protein



Citrus lemon

V4T7W5
Uncharacterized protein



Citrus lemon

V4T825
Uncharacterized protein



Citrus lemon

V4T846
Uncharacterized protein



Citrus lemon

V4T8E9
S-acyltransferase (EC 2.3.1.225) (Palmitoyltransferase)



Citrus lemon

V4T8G2
Uncharacterized protein



Citrus lemon

V4T8G9
Chorismate synthase (EC 4.2.3.5)



Citrus lemon

V4T8Y6
Uncharacterized protein



Citrus lemon

V4T8Y8
Uncharacterized protein



Citrus lemon

V4T939
Carboxypeptidase (EC 3.4.16.—)



Citrus lemon

V4T957
Uncharacterized protein



Citrus lemon

V4T998
Uncharacterized protein



Citrus lemon

V4T9B9
Uncharacterized protein



Citrus lemon

V4T9Y7
Uncharacterized protein



Citrus lemon

V4TA70
Uncharacterized protein



Citrus lemon

V4TAF6
Uncharacterized protein



Citrus lemon

V4TB09
Uncharacterized protein



Citrus lemon

V4TB32
Uncharacterized protein



Citrus lemon

V4TB89
Uncharacterized protein



Citrus lemon

V4TBN7
Phosphoinositide phospholipase C (EC 3.1.4.11)



Citrus lemon

V4TBQ3
Uncharacterized protein



Citrus lemon

V4TBS4
Uncharacterized protein



Citrus lemon

V4TBU3
Uncharacterized protein



Citrus lemon

V4TCA6
Uncharacterized protein



Citrus lemon

V4TCL3
Uncharacterized protein



Citrus lemon

V4TCS5
Pectate lyase (EC 4.2.2.2)



Citrus lemon

V4TD99
Uncharacterized protein



Citrus lemon

V4TDB5
Uncharacterized protein



Citrus lemon

V4TDI2
Uncharacterized protein



Citrus lemon

V4TDY3
Serine/threonine-protein kinase (EC 2.7.11.1)



Citrus lemon

V4TE72
Uncharacterized protein



Citrus lemon

V4TE95
Uncharacterized protein



Citrus lemon

V4TEC0
Uncharacterized protein



Citrus lemon

V4TED8
Uncharacterized protein



Citrus lemon

V4TES4
Uncharacterized protein



Citrus lemon

V4TEY9
Uncharacterized protein



Citrus lemon

V4TF24
Proteasome subunit alpha type (EC 3.4.25.1)



Citrus lemon

V4TF52
Uricase (EC 1.7.3.3) (Urate oxidase)



Citrus lemon

V4TFV8
Catalase (EC 1.11.1.6)



Citrus lemon

V4TGU1
Uncharacterized protein



Citrus lemon

V4TH28
Uncharacterized protein



Citrus lemon

V4TH78
Reticulon-like protein



Citrus lemon

V4THM9
Uncharacterized protein



Citrus lemon

V4TIU2
Ribulose-phosphate 3-epimerase (EC 5.1.3.1)



Citrus lemon

V4TIW6
Uncharacterized protein



Citrus lemon

V4TIY6
Uncharacterized protein



Citrus lemon

V4TIZ5
Uncharacterized protein



Citrus lemon

V4TJ75
Uncharacterized protein



Citrus lemon

V4TJC3
Uncharacterized protein



Citrus lemon

V4TJQ9
Uncharacterized protein



Citrus lemon

V4TK29
NEDD8-activating enzyme E1 regulatory subunit



Citrus lemon

V4TL04
Uncharacterized protein



Citrus lemon

V4TLL5
Uncharacterized protein



Citrus lemon

V4TLP6
Uncharacterized protein



Citrus lemon

V4TM00
Uncharacterized protein



Citrus lemon

V4TM19
Uncharacterized protein



Citrus lemon

V4TMB7
Uncharacterized protein (Fragment)



Citrus lemon

V4TMD1
Uncharacterized protein



Citrus lemon

V4TMD6
Uncharacterized protein



Citrus lemon

V4TMV4
Uncharacterized protein



Citrus lemon

V4TN30
Uncharacterized protein



Citrus lemon

V4TN38
Uncharacterized protein



Citrus lemon

V4TNY8
Uncharacterized protein



Citrus lemon

V4TP87
Carbonic anhydrase (EC 4.2.1.1) (Carbonate dehydratase)



Citrus lemon

V4TPM1
Homoserine dehydrogenase (HDH) (EC 1.1.1.3)



Citrus lemon

V4TQB6
Uncharacterized protein



Citrus lemon

V4TQM7
Uncharacterized protein



Citrus lemon

V4TQR2
Uncharacterized protein



Citrus lemon

V4TQV9
Uncharacterized protein



Citrus lemon

V4TS21
Proteasome subunit beta type (EC 3.4.25.1)



Citrus lemon

V4TS28
Annexin



Citrus lemon

V4TSD8
Uncharacterized protein (Fragment)



Citrus lemon

V4TSF8
Uncharacterized protein



Citrus lemon

V4TSI9
Uncharacterized protein



Citrus lemon

V4TT89
Uncharacterized protein



Citrus lemon

V4TTA0
Uncharacterized protein



Citrus lemon

V4TTR8
Uncharacterized protein



Citrus lemon

V4TTV4
Uncharacterized protein



Citrus lemon

V4TTZ7
Uncharacterized protein



Citrus lemon

V4TU54
Uncharacterized protein



Citrus lemon

V4TVB6
Uncharacterized protein



Citrus lemon

V4TVG1
Eukaryotic translation initiation factor 5A (eIF-5A)



Citrus lemon

V4TVJ4
Profilin



Citrus lemon

V4TVM6
Uncharacterized protein



Citrus lemon

V4TVM9
Uncharacterized protein



Citrus lemon

V4TVP7
Uncharacterized protein



Citrus lemon

V4TVT8
Uncharacterized protein



Citrus lemon

V4TW14
Uncharacterized protein



Citrus lemon

V4TWG9
T-complex protein 1 subunit delta



Citrus lemon

V4TWU1
Probable bifunctional methylthioribulose-1-phosphate




dehydratase/enolase-phosphatase E1 [Includes: Enolase-phosphatase




E1 (EC 3.1.3.77) (2,3-diketo-5-methylthio-1-phosphopentane




phosphatase); Methylthioribulose-1-phosphate dehydratase




(MTRu-1-P dehydratase) (EC 4.2.1.109)]



Citrus lemon

V4TWX8
Uncharacterized protein



Citrus lemon

V4TXH0
Glutamate decarboxylase (EC 4.1.1.15)



Citrus lemon

V4TXK9
Uncharacterized protein



Citrus lemon

V4TXU9
Thiamine thiazole synthase, chloroplastic (Thiazole




biosynthetic enzyme)



Citrus lemon

V4TY40
Uncharacterized protein



Citrus lemon

V4TYJ6
Uncharacterized protein



Citrus lemon

V4TYP5
60S ribosomal protein L13



Citrus lemon

V4TYP6
Uncharacterized protein



Citrus lemon

V4TYR6
Uncharacterized protein



Citrus lemon

V4TYZ8
Tubulin alpha chain



Citrus lemon

V4TZ91
Guanosine nucleotide diphosphate dissociation inhibitor



Citrus lemon

V4TZA8
Uncharacterized protein



Citrus lemon

V4TZJ1
Uncharacterized protein



Citrus lemon

V4TZK5
Uncharacterized protein



Citrus lemon

V4TZP2
Uncharacterized protein



Citrus lemon

V4TZT8
Uncharacterized protein



Citrus lemon

V4TZU3
Mitogen-activated protein kinase (EC 2.7.11.24)



Citrus lemon

V4TZU5
Dihydrolipoyl dehydrogenase (EC 1.8.1.4)



Citrus lemon

V4TZZ0
Uncharacterized protein



Citrus lemon

V4U003
Eukaryotic translation initiation factor 3 subunit K (eIF3k)




(eIF-3 p25)



Citrus lemon

V4U068
Uncharacterized protein



Citrus lemon

V4U088
Uncharacterized protein



Citrus lemon

V4U0J7
Uncharacterized protein



Citrus lemon

V4U133
Uncharacterized protein



Citrus lemon

V4U1A8
Uncharacterized protein



Citrus lemon

V4U1K1
Xylose isomerase (EC 5.3.1.5)



Citrus lemon

V4U1M1
Uncharacterized protein



Citrus lemon

V4U1V0
Uncharacterized protein



Citrus lemon

V4U1X7
Uncharacterized protein



Citrus lemon

V4U1X9
Proteasome subunit beta type (EC 3.4.25.1)



Citrus lemon

V4U251
Uncharacterized protein



Citrus lemon

V4U283
Uncharacterized protein



Citrus lemon

V4U2E4
Uncharacterized protein



Citrus lemon

V4U2F7
Uncharacterized protein



Citrus lemon

V4U2H8
Uncharacterized protein



Citrus lemon

V4U2L0
Malate dehydrogenase (EC 1.1.1.37)



Citrus lemon

V4U2L2
Uncharacterized protein



Citrus lemon

V4U2W4
V-type proton ATPase subunit C



Citrus lemon

V4U3L2
Uncharacterized protein



Citrus lemon

V4U3W8
Uncharacterized protein



Citrus lemon

V4U412
Uncharacterized protein



Citrus lemon

V4U4K2
Uncharacterized protein



Citrus lemon

V4U4M4
Uncharacterized protein



Citrus lemon

V4U4N5
Eukaryotic translation initiation factor 6 (eIF-6)



Citrus lemon

V4U4S9
Uncharacterized protein



Citrus lemon

V4U4X3
Serine hydroxymethyltransferase (EC 2.1.2.1)



Citrus lemon

V4U4Z9
Uncharacterized protein



Citrus lemon

V4U500
Uncharacterized protein



Citrus lemon

V4U5B0
Eukaryotic translation initiation factor 3 subunit E (eIF3e)




(Eukaryotic translation initiation factor 3 subunit 6)



Citrus lemon

V4U5B8
Glutathione peroxidase



Citrus lemon

V4U5R5
Citrate synthase



Citrus lemon

V4U5Y8
Uncharacterized protein



Citrus lemon

V4U6I5
ATP synthase subunit beta (EC 3.6.3.14)



Citrus lemon

V4U6Q8
Uncharacterized protein



Citrus lemon

V4U706
Uncharacterized protein



Citrus lemon

V4U717
Uncharacterized protein



Citrus lemon

V4U726
Uncharacterized protein



Citrus lemon

V4U729
Uncharacterized protein



Citrus lemon

V4U734
Serine/threonine-protein phosphatase (EC 3.1.3.16)



Citrus lemon

V4U7G7
Uncharacterized protein



Citrus lemon

V4U7H5
Uncharacterized protein



Citrus lemon

V4U7R1
Potassium transporter



Citrus lemon

V4U7R7
Mitogen-activated protein kinase (EC 2.7.11.24)



Citrus lemon

V4U833
Malic enzyme



Citrus lemon

V4U840
Uncharacterized protein



Citrus lemon

V4U8C3
Uncharacterized protein



Citrus lemon

V4U8J1
3-phosphoshikimate 1-carboxyvinyltransferase (EC 2.5.1.19)



Citrus lemon

V4U8J8
T-complex protein 1 subunit gamma



Citrus lemon

V4U995
Uncharacterized protein



Citrus lemon

V4U999
Uncharacterized protein



Citrus lemon

V4U9C7
Eukaryotic translation initiation factor 3 subunit D (eIF3d)




(Eukaryotic translation initiation factor 3 subunit 7) (eIF-3-zeta)



Citrus lemon

V4U9G8
Proline iminopeptidase (EC 3.4.11.5)



Citrus lemon

V4U9L1
Uncharacterized protein



Citrus lemon

V4UA63
Phytochrome



Citrus lemon

V4UAC8
Uncharacterized protein



Citrus lemon

V4UAR4
Uncharacterized protein



Citrus lemon

V4UB30
Uncharacterized protein



Citrus lemon

V4UBK8
V-type proton ATPase subunit a



Citrus lemon

V4UBL3
Coatomer subunit alpha



Citrus lemon

V4UBL5
Uncharacterized protein (Fragment)



Citrus lemon

V4UBM0
Uncharacterized protein



Citrus lemon

V4UBZ8
Aspartate aminotransferase (EC 2.6.1.1)



Citrus lemon

V4UC72
Uncharacterized protein



Citrus lemon

V4UC97
Beta-glucosidase (EC 3.2.1.21)



Citrus lemon

V4UCE2
Uncharacterized protein



Citrus lemon

V4UCT9
Acetyl-coenzyme A synthetase (EC 6.2.1.1)



Citrus lemon

V4UCZ1
Uncharacterized protein



Citrus lemon

V4UE34
Uncharacterized protein



Citrus lemon

V4UE78
Uncharacterized protein



Citrus lemon

V4UER3
Uncharacterized protein



Citrus lemon

V4UET6
Uncharacterized protein



Citrus lemon

V4UEZ6
Uncharacterized protein



Citrus lemon

V4UFD0
Uncharacterized protein



Citrus lemon

V4UFG8
Uncharacterized protein



Citrus lemon

V4UFK1
Uncharacterized protein



Citrus lemon

V4UG68
Eukaryotic translation initiation factor 3 subunit I (eIF3i)



Citrus lemon

V4UGB0
Uncharacterized protein



Citrus lemon

V4UGH4
Uncharacterized protein



Citrus lemon

V4UGL9
Uncharacterized protein



Citrus lemon

V4UGQ0
Ubiquitinyl hydrolase 1 (EC 3.4.19.12)



Citrus lemon

V4UH00
Uncharacterized protein



Citrus lemon

V4UH48
Uncharacterized protein



Citrus lemon

V4UH77
Proteasome subunit alpha type (EC 3.4.25.1)



Citrus lemon

V4UHD8
Uncharacterized protein



Citrus lemon

V4UHD9
Uncharacterized protein



Citrus lemon

V4UHF1
Uncharacterized protein



Citrus lemon

V4UHZ5
Uncharacterized protein



Citrus lemon

V4UI07
40S ribosomal protein S8



Citrus lemon

V4UI34
Eukaryotic translation initiation factor 3 subunit L (eIF3l)



Citrus lemon

V4UIF1
Uncharacterized protein



Citrus lemon

V4UIN5
Uncharacterized protein



Citrus lemon

V4UIX8
Uncharacterized protein



Citrus lemon

V4UJ12
Uncharacterized protein



Citrus lemon

V4UJ42
Uncharacterized protein



Citrus lemon

V4UJ63
Uncharacterized protein



Citrus lemon

V4UJB7
Uncharacterized protein (Fragment)



Citrus lemon

V4UJC4
Uncharacterized protein



Citrus lemon

V4UJX0
Phosphotransferase (EC 2.7.1.—)



Citrus lemon

V4UJY5
Uncharacterized protein



Citrus lemon

V4UK18
Uncharacterized protein



Citrus lemon

V4UK52
Uncharacterized protein



Citrus lemon

V4UKM9
Uncharacterized protein



Citrus lemon

V4UKS4
Uncharacterized protein



Citrus lemon

V4UKV6
40S ribosomal protein SA



Citrus lemon

V4UL30
Pyrophosphate-fructose 6-phosphate 1-phosphotransferase




subunit beta (PFP) (EC 2.7.1.90) (6-phosphofructokinase,




pyrophosphate dependent) (PPi-PFK)




(Pyrophosphate-dependent 6-phosphofructose-1-kinase)



Citrus lemon

V4UL39
Uncharacterized protein



Citrus lemon

V4ULH9
Uncharacterized protein



Citrus lemon

V4ULL2
Uncharacterized protein



Citrus lemon

V4ULS0
Uncharacterized protein



Citrus lemon

V4UMU7
Uncharacterized protein



Citrus lemon

V4UN36
Uncharacterized protein



Citrus lemon

V4UNT5
Uncharacterized protein



Citrus lemon

V4UNW1
Uncharacterized protein



Citrus lemon

V4UP89
Uncharacterized protein



Citrus lemon

V4UPE4
Uncharacterized protein



Citrus lemon

V4UPF7
Uncharacterized protein



Citrus lemon

V4UPK0
Uncharacterized protein



Citrus lemon

V4UPX5
Uncharacterized protein



Citrus lemon

V4UQ58
Uncharacterized protein



Citrus lemon

V4UQF6
Uncharacterized protein



Citrus lemon

V4UR21
Uncharacterized protein



Citrus lemon

V4UR80
Uncharacterized protein



Citrus lemon

V4URK3
Uncharacterized protein



Citrus lemon

V4URT3
Uncharacterized protein



Citrus lemon

V4US96
Uncharacterized protein



Citrus lemon

V4USQ8
Uncharacterized protein



Citrus lemon

V4UT16
Uncharacterized protein



Citrus lemon

V4UTC6
Uncharacterized protein



Citrus lemon

V4UTC8
Uncharacterized protein



Citrus lemon

V4UTP6
Uncharacterized protein



Citrus lemon

V4UTY0
Proteasome subunit alpha type (EC 3.4.25.1)



Citrus lemon

V4UU96
Uncharacterized protein



Citrus lemon

V4UUB6
Uncharacterized protein



Citrus lemon

V4UUJ9
Aminopeptidase (EC 3.4.11.—)



Citrus lemon

V4UUK6
Uncharacterized protein



Citrus lemon

V4UV09
Uncharacterized protein



Citrus lemon

V4UV83
Lysine--tRNA ligase (EC 6.1.1.6) (Lysyl-tRNA synthetase)



Citrus lemon

V4UVJ5
Diacylglycerol kinase (DAG kinase) (EC 2.7.1.107)



Citrus lemon

V4UW03
Uncharacterized protein



Citrus lemon

V4UW04
Uncharacterized protein



Citrus lemon

V4UWR1
Uncharacterized protein



Citrus lemon

V4UWV8
Uncharacterized protein



Citrus lemon

V4UX36
Uncharacterized protein



Citrus lemon

V4V003
Uncharacterized protein



Citrus lemon

V4V0J0
40S ribosomal protein S26



Citrus lemon

V4V1P8
Uncharacterized protein



Citrus lemon

V4V4V0
Uncharacterized protein



Citrus lemon

V4V5T8
Ubiquitin-fold modifier 1



Citrus lemon

V4V600
Uncharacterized protein



Citrus lemon

V4V622
Aldehyde dehydrogenase



Citrus lemon

V4V6W1
Uncharacterized protein



Citrus lemon

V4V6Z2
Uncharacterized protein



Citrus lemon

V4V738
Uncharacterized protein



Citrus lemon

V4V8H5
Vacuolar protein sorting-associated protein 35



Citrus lemon

V4V9P6
Eukaryotic translation initiation factor 3 subunit F (eIF3f)




(eIF-3-epsilon)



Citrus lemon

V4V9V7
Clathrin heavy chain



Citrus lemon

V4V9X3
Uncharacterized protein



Citrus lemon

V4VAA3
Superoxide dismutase (EC 1.15.1.1)



Citrus lemon

V4VAF3
Uncharacterized protein



Citrus lemon

V4VBQ0
Uncharacterized protein (Fragment)



Citrus lemon

V4VCL1
Proteasome subunit beta type (EC 3.4.25.1)



Citrus lemon

V4VCZ9
Uncharacterized protein



Citrus lemon

V4VDK1
Peptidylprolyl isomerase (EC 5.2.1.8)



Citrus lemon

V4VEA1
Uncharacterized protein



Citrus lemon

V4VEB3
Alanine--tRNA ligase (EC 6.1.1.7) (Alanyl-tRNA synthetase)




(AlaRS)



Citrus lemon

V4VEE3
Glutamine synthetase (EC 6.3.1.2)



Citrus lemon

V4VFM3
Uncharacterized protein



Citrus lemon

V4VFN5
Proteasome subunit beta type (EC 3.4.25.1)



Citrus lemon

V4VGD6
Uncharacterized protein



Citrus lemon

V4VGL9
Uncharacterized protein



Citrus lemon

V4VHI6
Uncharacterized protein



Citrus lemon

V4VIP4
Uncharacterized protein



Citrus lemon

V4VJT4
Uncharacterized protein



Citrus lemon

V4VK14
Uncharacterized protein



Citrus lemon

V4VKI5
Protein-L-isoaspartate O-methyltransferase (EC 2.1.1.77)



Citrus lemon

V4VKP2
Glyceraldehyde-3-phosphate dehydrogenase (EC 1.2.1.—)



Citrus lemon

V4VL73
Acyl-coenzyme A oxidase



Citrus lemon

V4VLL7
Uncharacterized protein



Citrus lemon

V4VN43
Uncharacterized protein (Fragment)



Citrus lemon

V4VQH3
Methylenetetrahydrofolate reductase (EC 1.5.1.20)



Citrus lemon

V4VTC9
Uncharacterized protein (Fragment)



Citrus lemon

V4VTT4
Uncharacterized protein



Citrus lemon

V4VTY7
Uncharacterized protein



Citrus lemon

V4VU14
Uncharacterized protein



Citrus lemon

V4VU32
Uncharacterized protein



Citrus lemon

V4VUK6
S-(hydroxymethyl)glutathione dehydrogenase (EC 1.1.1.284)



Citrus lemon

V4VVR8
Uncharacterized protein



Citrus lemon

V4VXE2
Uncharacterized protein



Citrus lemon

V4VY37
Phosphomannomutase (EC 5.4.2.8)



Citrus lemon

V4VYC0
Uncharacterized protein



Citrus lemon

V4VYV1
Uncharacterized protein



Citrus lemon

V4VZ80
Uncharacterized protein



Citrus lemon

V4VZJ7
Uncharacterized protein



Citrus lemon

V4W2P2
Alpha-mannosidase (EC 3.2.1.—)



Citrus lemon

V4W2Z9
Chloride channel protein



Citrus lemon

V4W378
Uncharacterized protein



Citrus lemon

V4W4G3
Uncharacterized protein



Citrus lemon

V4W5F1
Uncharacterized protein



Citrus lemon

V4W5N8
Uncharacterized protein



Citrus lemon

V4W5U2
Uncharacterized protein



Citrus lemon

V4W6G1
Uncharacterized protein



Citrus lemon

V4W730
Uncharacterized protein



Citrus lemon

V4W7J4
Obg-like ATPase 1



Citrus lemon

V4W7L5
Uncharacterized protein



Citrus lemon

V4W8C5
Uncharacterized protein



Citrus lemon

V4W8C9
Uncharacterized protein



Citrus lemon

V4W8D3
Uncharacterized protein



Citrus lemon

V4W951
Uncharacterized protein



Citrus lemon

V4W9F6
60S ribosomal protein L18a



Citrus lemon

V4W9G2
Uncharacterized protein (Fragment)



Citrus lemon

V4W9L3
Uncharacterized protein



Citrus lemon

V4W9Y8
Uncharacterized protein



Citrus lemon

V4WAP9
Coatomer subunit beta (Beta-coat protein)



Citrus lemon

V4WBK6
Cytochrome b-c1 complex subunit 7



Citrus lemon

V4WC15
Malic enzyme



Citrus lemon

V4WC19
Uncharacterized protein



Citrus lemon

V4WC74
Uncharacterized protein



Citrus lemon

V4WC86
Serine/threonine-protein phosphatase 2A 55 kDa regulatory




subunit B



Citrus lemon

V4WCS4
GTP-binding nuclear protein



Citrus lemon

V4WD80
Aspartate aminotransferase (EC 2.6.1.1)



Citrus lemon

V4WDK0
Uncharacterized protein



Citrus lemon

V4WDK3
ATP-dependent 6-phosphofructokinase (ATP-PFK)




(Phosphofructokinase) (EC 2.7.1.11) (Phosphohexokinase)



Citrus lemon

V4WE00
Uncharacterized protein



Citrus lemon

V4WEE3
Uncharacterized protein



Citrus lemon

V4WEN2
Uncharacterized protein



Citrus lemon

V4WG97
Autophagy-related protein



Citrus lemon

V4WGV2
Uncharacterized protein



Citrus lemon

V4WGW5
Uridine kinase (EC 2.7.1.48)



Citrus lemon

V4WHD4
Uncharacterized protein



Citrus lemon

V4WHF8
Sucrose synthase (EC 2.4.1.13)



Citrus lemon

V4WHK2
Pectinesterase (EC 3.1.1.11)



Citrus lemon

V4WHQ4
Uncharacterized protein



Citrus lemon

V4WHT6
Uncharacterized protein



Citrus lemon

V4WJ93
Uncharacterized protein



Citrus lemon

V4WJA9
Uncharacterized protein



Citrus lemon

V4WJB1
Uncharacterized protein



Citrus lemon

V9HXG3
Protein disulfide-isomerase (EC 5.3.4.1)



Citrus lemon

W8Q8K1
Putative inorganic pyrophosphatase



Citrus lemon

W8QJL0
Putative isopentenyl pyrophosphate isomerase












Grape
Accession Number
Identified Proteins





Grape
A5C5K3 (+2)
Adenosylhomocysteinase


Grape
Q9M6B5
Alcohol dehydrogenase 6


Grape
A3FA65 (+1)
Aquaporin PIP1; 3


Grape
Q0MX13 (+2)
Aquaporin PIP2; 2


Grape
A3FA69 (+4)
Aquaporin PIP2; 4


Grape
A5AFS1 (+2)
Elongation factor 1-alpha


Grape
UPI0001985702
elongation factor 2


Grape
D7T227
Enolase


Grape
D7TJ12
Enolase


Grape
A5B118 (+1)
Fructose-bisphosphate aldolase


Grape
E0CQ39
Glucose-6-phosphate isomerase


Grape
D7TW04
Glutathione peroxidase


Grape
A1YW90 (+3)
Glutathione S-transferase


Grape
A5BEW0
Histone H4


Grape
UPI00015C9A6A
HSC70-1 (heat shock cognate 70 kDa protein 1); ATP




binding isoform 1


Grape
D7FBC0 (+1)
Malate dehydrogenase


Grape
D7TBH4
Malic enzyme


Grape
A5ATB7 (+1)
Methylenetetrahydrofolate reductase


Grape
A5JPK7 (+1)
Monodehydroascorbate reductase


Grape
A5AKD8
Peptidyl-prolyl cis-trans isomerase


Grape
A5BQN6
Peptidyl-prolyl cis-trans isomerase


Grape
A5CAF6
Phosphoglycerate kinase


Grape
Q09VU3 (+1)
Phospholipase D


Grape
D7SK33
Phosphorylase


Grape
A5AQ89
Profilin


Grape
C5DB50 (+2)
Putative 2,3-bisphosphoglycerate-independent




phosphoglycerate mutase


Grape
D7TIZ5
Pyruvate kinase


Grape
A5BV65
Triosephosphate isomerase


Grapefruit
G8Z362 (+1)
(E)-beta-farnesene synthase


Grapefruit
Q5CD81
(E)-beta-ocimene synthase


Grapefruit
D0UZK1 (+2)
1,2 rhamnosyltransferase


Grapefruit
A7ISD3
1,6-rhamnosyltransferase


Grapefruit
Q80H98
280 kDa protein


Grapefruit
Q15GA4 (+2)
286 kDa polyprotein


Grapefruit
D7NHW9
2-phospho-D-glycerate hydrolase


Grapefruit
D0EAL9
349 kDa polyprotein


Grapefruit
Q9DTG5
349-kDa polyprotein


Grapefruit
O22297
Acidic cellulase


Grapefruit
Q8H986
Acidic class I chitinase


Grapefruit
D3GQL0
Aconitate hydratase 1


Grapefruit
K7N8A0
Actin


Grapefruit
A8W8Y0
Alcohol acyl transferase


Grapefruit
Q84V85
Allene oxide synthase


Grapefruit
F8WL79
Aminopeptidase


Grapefruit
Q09MG5
Apocytochrome f


Grapefruit
J7EIR8
Ascorbate peroxidase


Grapefruit
B9VRH6
Ascorbate peroxidase


Grapefruit
G9I820
Auxin-response factor


Grapefruit
J7ICW8
Beta-amylase


Grapefruit
Q8L5Q9
Beta-galactosidase


Grapefruit
A7BG60
Beta-pinene synthase


Grapefruit
C0KLD1
Beta-tubulin


Grapefruit
Q91QZ1
Capsid protein


Grapefruit
Q3SAK9
Capsid protein


Grapefruit
D2U833
Cation chloride cotransporter


Grapefruit
C3VPJ0 (+3)
Chalcone synthase


Grapefruit
D5LM39
Chloride channel protein


Grapefruit
Q9M4U0
Cinnamate 4-hydroxylase CYP73


Grapefruit
Q39627
Citrin


Grapefruit
G2XKD3
Coat protein


Grapefruit
Q3L2I6
Coat protein


Grapefruit
D5FV16
CRT/DRE binding factor


Grapefruit
Q8H6S5
CTV.2


Grapefruit
Q8H6Q8
CTV.20


Grapefruit
Q8H6Q7
CTV.22


Grapefruit
Q1I1D7
Cytochrome P450


Grapefruit
Q7Y045
Dehydrin


Grapefruit
F8WLD2
DNA excision repair protein


Grapefruit
Q09MI8
DNA-directed RNA polymerase subunit beta″


Grapefruit
D2WKC9
Ethylene response 1


Grapefruit
D2WKD2
Ethylene response sensor 1


Grapefruit
D7PVG7
Ethylene-insensitive 3-like 1 protein


Grapefruit
G3CHK8
Eukaryotic translation initiation factor 3 subunit E


Grapefruit
A9NJG4 (+3)
Fatty acid hydroperoxide lyase


Grapefruit
B8Y9B5
F-box family protein


Grapefruit
Q000W4
Fe(III)-chelate reductase


Grapefruit
Q6Q3H4
Fructokinase


Grapefruit
F8WL95
Gag-pol polyprotein


Grapefruit
Q8L5K4
Gamma-terpinene synthase, chloroplastic


Grapefruit
Q9SP43
Glucose-1-phosphate adenylyltransferase


Grapefruit
Q3HM93
Glutathione S-transferase


Grapefruit
D0VEW6
GRAS family transcription factor


Grapefruit
F8WL87
Heat shock protein


Grapefruit
H9NHK0
Hsp90


Grapefruit
Q8H6R4
Jp18


Grapefruit
G3CHK6
Leucine-rich repeat family protein


Grapefruit
B2YGX9 (+1)
Limonoid UDP-glucosyltransferase


Grapefruit
Q05KK0
MADS-box protein


Grapefruit
F8WLB4
Mechanosensitive ion channel domain-containing protein


Grapefruit
Q5CD82
Monoterpene synthase


Grapefruit
F8WLC4
MYB transcription factor


Grapefruit
A5YWA9
NAC domain protein


Grapefruit
Q09MC9
NAD(P)H-quinone oxidoreductase subunit 5, chloroplastic


Grapefruit
Q8H6R9
NBS-LRR type disease resistance protein


Grapefruit
Q8H6S0
NBS-LRR type disease resistance protein


Grapefruit
Q8H6R6
NBS-LRR type disease resistance protein


Grapefruit
J9WR93
p1a


Grapefruit
Q1X8V8
P23


Grapefruit
E7DSS0 (+4)
P23


Grapefruit
G0Z9I6
p27


Grapefruit
I3XHN0
p33


Grapefruit
B8YDL3
p33 protein


Grapefruit
B9VB22
p33 protein


Grapefruit
P87587
P346


Grapefruit
B9VB56
p349 protein


Grapefruit
I3RWW7
p349 protein


Grapefruit
B9VB20
p349 protein


Grapefruit
Q9WID7
p349 protein


Grapefruit
Q2XP16
P353


Grapefruit
O04886 (+1)
Pectinesterase 1


Grapefruit
F8WL74
Peptidyl-prolyl cis-trans isomerase


Grapefruit
Q0ZA67
Peroxidase


Grapefruit
F1CT41
Phosphoenolpyruvate carboxylase


Grapefruit
B1PBV7 (+2)
Phytoene synthase


Grapefruit
Q9ZWQ8
Plastid-lipid-associated protein, chloroplastic


Grapefruit
Q94FM1
Pol polyprotein


Grapefruit
Q94FM0
Pol polyprotein


Grapefruit
G9I825
Poly C-binding protein


Grapefruit
O64460 (+7)
Polygalacturonase inhibitor


Grapefruit
I3XHM8
Polyprotein


Grapefruit
C0STR9
Polyprotein


Grapefruit
H6U1F0
Polyprotein


Grapefruit
B8QHP8
Polyprotein


Grapefruit
I3V6C0
Polyprotein


Grapefruit
C0STS0
Polyprotein


Grapefruit
K0FGH5
Polyprotein


Grapefruit
Q3HWZ1
Polyprotein


Grapefruit
F8WLA5
PPR containing protein


Grapefruit
Q06652 (+1)
Probable phospholipid hydroperoxide glutathione




peroxidase


Grapefruit
P84177
Profilin


Grapefruit
Q09MB4
Protein ycf2


Grapefruit
A8C183
PSI reaction center subunit II


Grapefruit
A5JVP6
Putative 2b protein


Grapefruit
D0EFM2
Putative eukaryotic translation initiation factor 1


Grapefruit
Q18L98
Putative gag-pol polyprotein


Grapefruit
B5AMI9
Putative movement protein


Grapefruit
A1ECK5
Putative multiple stress-responsive zinc-finger protein


Grapefruit
B5AMJ0
Putative replicase polyprotein


Grapefruit
I7CYN5
Putative RNA-dependent RNA polymerase


Grapefruit
Q8RVR2
Putative terpene synthase


Grapefruit
B5TE89
Putative uncharacterized protein


Grapefruit
Q8JVF3
Putative uncharacterized protein


Grapefruit
F8WLB0
Putative uncharacterized protein ORF43


Grapefruit
A5JVP4
Putative viral replicase


Grapefruit
M1JAW3
Replicase


Grapefruit
H6VXK8
Replicase polyprotein


Grapefruit
J9UF50 (+1)
Replicase protein 1a


Grapefruit
J9RV45
Replicase protein 2a


Grapefruit
Q5EGG5
Replicase-associated polyprotein


Grapefruit
G9I823
RNA recognition motif protein 1


Grapefruit
J7EPC0
RNA-dependent RNA polymerase


Grapefruit
Q6DN67
RNA-directed RNA polymerase L


Grapefruit
A9CQM4
SEPALLATA1 homolog


Grapefruit
Q9SLS2
Sucrose synthase


Grapefruit
Q9SLV8 (+1)
Sucrose synthase


Grapefruit
Q38JC1
Temperature-induced lipocalin


Grapefruit
D0ELH6
Tetratricopeptide domain-containing thioredoxin


Grapefruit
D2KU75
Thaumatin-like protein


Grapefruit
C3VIC2
Translation elongation factor


Grapefruit
D5LY07
Ubiquitin/ribosomal fusion protein


Grapefruit
C6KI43
UDP-glucosyltransferase family 1 protein


Grapefruit
A0FKR1
Vacuolar citrate/H+ symporter


Grapefruit
Q944C8
Vacuolar invertase


Grapefruit
Q9MB46
V-type proton ATPase subunit E


Grapefruit
F8WL82
WD-40 repeat family protein



Helianthuus annuus

HanXRQChr03g0080391
Hsp90



Helianthuus annuus

HanXRQChr13g0408351
Hsp90



Helianthuus annuus

HanXRQChr13g0408441
Hsp90



Helianthuus annuus

HanXRQChr14g0462551
Hsp90



Helianthuus annuus

HanXRQChr02g0044471
Hsp70



Helianthuus annuus

HanXRQChr02g0044481
Hsp70



Helianthuus annuus

HanXRQChr05g0132631
Hsp70



Helianthuus annuus

HanXRQChr05g0134631
Hsp70



Helianthuus annuus

HanXRQChr05g0134801
Hsp70



Helianthuus annuus

HanXRQChr10g0299441
glutathione S-transferase



Helianthuus annuus

HanXRQChr16g0516291
glutathione S-transferase



Helianthuus annuus

HanXRQChr03g0091431
lactate/malate dehydrogenase



Helianthuus annuus

HanXRQChr13g0421951
lactate/malate dehydrogenase



Helianthuus annuus

HanXRQChr10g0304821
lactate/malate dehydrogenase



Helianthuus annuus

HanXRQChr12g0373491
lactate/malate dehydrogenase



Helianthuus annuus

HanXRQChr01g0031071
small GTPase superfamily, Rab type



Helianthuus annuus

HanXRQChr01g0031091
small GTPase superfamily, Rab type



Helianthuus annuus

HanXRQChr02g0050791
small GTPase superfamily, Rab type



Helianthuus annuus

HanXRQChr11g0353711
small GTPase superfamily, Rab type



Helianthuus annuus

HanXRQChr13g0402771
small GTPase superfamily, Rab type



Helianthuus annuus

HanXRQChr07g0190171
isocitrate/isopropylmalate dehydrogenase



Helianthuus annuus

HanXRQChr16g0532251
isocitrate/isopropylmalate dehydrogenase



Helianthuus annuus

HanXRQChr03g0079131
phosphoenolpyruvate carboxylase



Helianthuus annuus

HanXRQChr15g0495261
phosphoenolpyruvate carboxylase



Helianthuus annuus

HanXRQChr13g0388931
phosphoenolpyruvate carboxylase



Helianthuus annuus

HanXRQChr14g0442731
phosphoenolpyruvate carboxylase



Helianthuus annuus

HanXRQChr15g0482381
UTP--glucose-1-phosphate uridylyltransferase



Helianthuus annuus

HanXRQChr16g0532261
UTP--glucose-1-phosphate uridylyltransferase



Helianthuus annuus

HanXRQChr05g0135591
tubulin



Helianthuus annuus

HanXRQChr06g0178921
tubulin



Helianthuus annuus

HanXRQChr08g0237071
tubulin



Helianthuus annuus

HanXRQChr11g0337991
tubulin



Helianthuus annuus

HanXRQChr13g0407921
tubulin



Helianthuus annuus

HanXRQChr05g0145191
tubulin



Helianthuus annuus

HanXRQChr07g0187021
tubulin



Helianthuus annuus

HanXRQChr07g0189811
tubulin



Helianthuus annuus

HanXRQChr09g0253681
tubulin



Helianthuus annuus

HanXRQChr10g0288911
tubulin



Helianthuus annuus

HanXRQChr11g0322631
tubulin



Helianthuus annuus

HanXRQChr12g0367231
tubulin



Helianthuus annuus

HanXRQChr13g0386681
tubulin



Helianthuus annuus

HanXRQChr13g0393261
tubulin



Helianthuus annuus

HanXRQChr12g0371591
ubiquitin



Helianthuus annuus

HanXRQChr12g0383641
ubiquitin



Helianthuus annuus

HanXRQChr17g0569881
ubiquitin



Helianthuus annuus

HanXRQChr06g0171511
photosystem II HCF136, stability/assembly factor



Helianthuus annuus

HanXRQChr17g0544921
photosystem II HCF136, stability/assembly factor



Helianthuus annuus

HanXRQChr16g0526461
proteasome B-type subunit



Helianthuus annuus

HanXRQChr17g0565551
proteasome B-type subunit



Helianthuus annuus

HanXRQChr05g0149801
proteasome B-type subunit



Helianthuus annuus

HanXRQChr09g0241421
proteasome B-type subunit



Helianthuus annuus

HanXRQChr11g0353161
proteasome B-type subunit



Helianthuus annuus

HanXRQChr16g0506311
proteinase inhibitor family I3 (Kunitz)



Helianthuus annuus

HanXRQChr16g0506331
proteinase inhibitor family I3 (Kunitz)



Helianthuus annuus

HanXRQChr09g0265401
metallopeptidase (M10 family)



Helianthuus annuus

HanXRQChr09g0265411
metallopeptidase (M10 family)



Helianthuus annuus

HanXRQChr05g0154561
ATPase, AAA-type



Helianthuus annuus

HanXRQChr08g0235061
ATPase, AAA-type



Helianthuus annuus

HanXRQChr09g0273921
ATPase, AAA-type



Helianthuus annuus

HanXRQChr16g0498881
ATPase, AAA-type



Helianthuus annuus

HanXRQChr02g0058711
oxoacid dehydrogenase acyltransferase



Helianthuus annuus

HanXRQChr08g0214191
oxoacid dehydrogenase acyltransferase



Helianthuus annuus

HanXRQChr08g0208631
small GTPase superfamily, SAR1-type



Helianthuus annuus

HanXRQChr11g0331441
small GTPase superfamily, SAR1-type



Helianthuus annuus

HanXRQChr12g0371571
small GTPase superfamily, SAR1-type



Helianthuus annuus

HanXRQChr12g0383571
small GTPase superfamily, SAR1-type



Helianthuus annuus

HanXRQChr14g0446771
small GTPase superfamily, SAR1-type



Helianthuus annuus

HanXRQChr17g0539461
small GTPase superfamily, SAR1-type



Helianthuus annuus

HanXRQChr17g0548271
small GTPase superfamily, SAR1-type



Helianthuus annuus

HanXRQChr17g0569871
small GTPase superfamily, SAR1-type



Helianthuus annuus

HanXRQChr10g0311201
ATPase, V1 complex, subunit A



Helianthuus annuus

HanXRQChr12g0359711
ATPase, V1 complex, subunit A



Helianthuus annuus

HanXRQChr04g0124671
fructose-1,6-bisphosphatase



Helianthuus annuus

HanXRQChr06g0176631
fructose-1,6-bisphosphatase



Helianthuus annuus

HanXRQCPg0579861
photosystem II PsbD/D2, reaction centre



Helianthuus annuus

HanXRQChr00c0439g0574731
photosystem II PsbD/D2, reaction centre



Helianthuus annuus

HanXRQChr04g0099321
photosystem II PsbD/D2, reaction centre



Helianthuus annuus

HanXRQChr08g0210231
photosystem II PsbD/D2, reaction centre



Helianthuus annuus

HanXRQChr11g0326671
photosystem II PsbD/D2, reaction centre



Helianthuus annuus

HanXRQChr17g0549121
photosystem II PsbD/D2, reaction centre



Helianthuus annuus

HanXRQCPg0579731
photosystem II protein D1



Helianthuus annuus

HanXRQChr00c0126g0571821
photosystem II protein D1



Helianthuus annuus

HanXRQChr00c0165g0572191
photosystem II protein D1



Helianthuus annuus

HanXRQChr00c0368g0574171
photosystem II protein D1



Helianthuus annuus

HanXRQChr00c0454g0574931
photosystem II protein D1



Helianthuus annuus

HanXRQChr00c0524g0575441
photosystem II protein D1



Helianthuus annuus

HanXRQChr00c0572g0575941
photosystem II protein D1



Helianthuus annuus

HanXRQChr09g0257281
photosystem II protein D1



Helianthuus annuus

HanXRQChr11g0326571
photosystem II protein D1



Helianthuus annuus

HanXRQChr11g0327051
photosystem II protein D1



Helianthuus annuus

HanXRQChr16g0503941
photosystem II protein D1



Helianthuus annuus

HanXRQCPg0580061
photosystem II cytochrome b559



Helianthuus annuus

HanXRQChr01g0020331
photosystem II cytochrome b559



Helianthuus annuus

HanXRQChr10g0283581
photosystem II cytochrome b559



Helianthuus annuus

HanXRQChr10g0284271
photosystem II cytochrome b559



Helianthuus annuus

HanXRQChr10g0289291
photosystem II cytochrome b559



Helianthuus annuus

HanXRQChr10g0318171
photosystem II cytochrome b559



Helianthuus annuus

HanXRQChr11g0326851
photosystem II cytochrome b559



Helianthuus annuus

HanXRQChr16g0529011
photosystem II cytochrome b559



Helianthuus annuus

HanXRQChr08g0219051
chlorophyll A-B binding protein



Helianthuus annuus

HanXRQChr12g0370841
chlorophyll A-B binding protein



Helianthuus annuus

HanXRQChr02g0053151
chlorophyll A-B binding protein



Helianthuus annuus

HanXRQChr02g0053161
chlorophyll A-B binding protein



Helianthuus annuus

HanXRQCPg0580051
cytochrome f



Helianthuus annuus

HanXRQChr01g0020341
cytochrome f



Helianthuus annuus

HanXRQChr10g0283571
cytochrome f



Helianthuus annuus

HanXRQChr10g0284261
cytochrome f



Helianthuus annuus

HanXRQChr10g0289281
cytochrome f



Helianthuus annuus

HanXRQChr10g0318181
cytochrome f



Helianthuus annuus

HanXRQChr11g0326841
cytochrome f



Helianthuus annuus

HanXRQChr15g0497521
cytochrome f



Helianthuus annuus

HanXRQChr06g0163851
ribosomal protein



Helianthuus annuus

HanXRQChr09g0252071
ribosomal protein



Helianthuus annuus

HanXRQChr12g0374041
ribosomal protein



Helianthuus annuus

HanXRQChr04g0128141
ribosomal protein



Helianthuus annuus

HanXRQChr05g0163131
ribosomal protein



Helianthuus annuus

HanXRQChr03g0076971
ribosomal protein



Helianthuus annuus

HanXRQChr05g0159851
ribosomal protein



Helianthuus annuus

HanXRQChr05g0159971
ribosomal protein



Helianthuus annuus

HanXRQChr11g0324631
ribosomal protein



Helianthuus annuus

HanXRQChr13g0408051
ribosomal protein



Helianthuus annuus

HanXRQChr03g0089331
ribosomal protein



Helianthuus annuus

HanXRQChr13g0419951
ribosomal protein



Helianthuus annuus

HanXRQChr15g0497041
ribosomal protein



Helianthuus annuus

HanXRQChr16g0499761
ribosomal protein



Helianthuus annuus

HanXRQChr04g0106961
ribosomal protein



Helianthuus annuus

HanXRQChr06g0175811
ribosomal protein



Helianthuus annuus

HanXRQChr04g0122771
ribosomal protein



Helianthuus annuus

HanXRQChr09g0245691
ribosomal protein



Helianthuus annuus

HanXRQChr16g0520021
ribosomal protein



Helianthuus annuus

HanXRQChr03g0060471
ribosomal protein



Helianthuus annuus

HanXRQChr14g0429531
ribosomal protein



Helianthuus annuus

HanXRQChr06g0171911
ribosomal protein



Helianthuus annuus

HanXRQChr15g0479091
ribosomal protein



Helianthuus annuus

HanXRQChr15g0479101
ribosomal protein



Helianthuus annuus

HanXRQChr17g0543641
ribosomal protein



Helianthuus annuus

HanXRQChr17g0543661
ribosomal protein



Helianthuus annuus

HanXRQChr04g0105831
ribosomal protein



Helianthuus annuus

HanXRQChr09g0258341
ribosomal protein



Helianthuus annuus

HanXRQChr10g0287141
ribosomal protein



Helianthuus annuus

HanXRQChr15g0463911
ribosomal protein



Helianthuus annuus

HanXRQChr03g0076171
ribosomal protein



Helianthuus annuus

HanXRQChr05g0159291
ribosomal protein



Helianthuus annuus

HanXRQChr13g0407551
ribosomal protein



Helianthuus annuus

HanXRQChr12g0380701
ribosomal protein



Helianthuus annuus

HanXRQChr15g0477271
ribosomal protein



Helianthuus annuus

HanXRQChr17g0545211
ribosomal protein



Helianthuus annuus

HanXRQChr17g0570741
ribosomal protein



Helianthuus annuus

HanXRQChr17g0570761
ribosomal protein



Helianthuus annuus

HanXRQChr02g0044021
ribosomal protein



Helianthuus annuus

HanXRQChr05g0152871
ribosomal protein



Helianthuus annuus

HanXRQChr01g0012781
ribosomal protein



Helianthuus annuus

HanXRQChr08g0230861
ribosomal protein



Helianthuus annuus

HanXRQChr13g0391831
ribosomal protein



Helianthuus annuus

HanXRQChr11g0337791
bifunctional trypsin/alpha-amylase inhibitor



Helianthuus annuus

HanXRQChr10g0312371
2-oxoacid dehydrogenase acyltransferase



Helianthuus annuus

HanXRQChr09g0276191
acid phosphatase (class B)



Helianthuus annuus

HanXRQChr05g0142271
aldose-1-epimerase



Helianthuus annuus

HanXRQChr14g0439791
alpha-D-phosphohexomutase



Helianthuus annuus

HanXRQChr09g0251071
alpha-L-fucosidase



Helianthuus annuus

HanXRQChr05g0147371
annexin



Helianthuus annuus

HanXRQChr09g0247561
Asp protease (Peptidase family A1)



Helianthuus annuus

HanXRQChr13g0409681
berberine-bridge enzyme (S)-reticulin: oxygen oxido-reductase



Helianthuus annuus

HanXRQChr10g0295971
beta-hydroxyacyl-(acyl-carrier-protein) dehydratase



Helianthuus annuus

HanXRQChr13g0412571
carbohydrate esterase family 13 - CE13 (pectin acylesterase - PAE)



Helianthuus annuus

HanXRQChr12g0360101
carbohydrate esterase family 8 - CE8 (pectin methylesterase - PME)



Helianthuus annuus

HanXRQChr01g0019231
carbonic anhydrase



Helianthuus annuus

HanXRQChr02g0036611
cellular retinaldehyde binding/alpha-tocopherol transport



Helianthuus annuus

HanXRQChr10g0313581
chaperonin Cpn60



Helianthuus annuus

HanXRQChr09g0251791
chlathrin



Helianthuus annuus

HanXRQChr11g0329811
chlorophyll A-B binding protein



Helianthuus annuus

HanXRQChr13g0398861
cobalamin (vitamin B12)-independent methionine synthase



Helianthuus annuus

HanXRQChr10g0298981
cyclophilin



Helianthuus annuus

HanXRQChr04g0103281
Cys protease (papain family)



Helianthuus annuus

HanXRQChr09g0268361
cytochrome P450



Helianthuus annuus

HanXRQChr17g0535591
dirigent protein



Helianthuus annuus

HanXRQChr03g0065901
expansin



Helianthuus annuus

HanXRQChr11g0336761
expressed protein (cupin domain, seed storage protein




domain)



Helianthuus annuus

HanXRQChr10g0280931
expressed protein (cupin domain, seed storage protein




domain)



Helianthuus annuus

HanXRQChr10g0288971
expressed protein (cupin domain, seed storage protein




domain)



Helianthuus annuus

HanXRQChr12g0380361
expressed protein (cupin domain, seed storage protein




domain)



Helianthuus annuus

HanXRQChr09g0254381
expressed protein (cupin domain, seed storage protein




domain)



Helianthuus annuus

HanXRQChr04g0112711
expressed protein (cupin domain, seed storage protein




domain)



Helianthuus annuus

HanXRQChr07g0196131
expressed protein (cupin domain, seed storage protein




domain)



Helianthuus annuus

HanXRQChr10g0301281
expressed protein (cupin domain, seed storage protein




domain)



Helianthuus annuus

HanXRQChr10g0301931
expressed protein (cupin domain, seed storage protein




domain)



Helianthuus annuus

HanXRQChr13g0404461
expressed protein (cupin domain)



Helianthuus annuus

HanXRQChr01g0015821
expressed protein (DUF642)



Helianthuus annuus

HanXRQChr03g0065301
expressed protein (Gnk2-homologous domain, antifungal




protein of Ginkgo seeds)



Helianthuus annuus

HanXRQChr03g0068311
expressed protein (LRR domains)



Helianthuus annuus

HanXRQChr10g0291371
expressed protein (LRR domains)



Helianthuus annuus

HanXRQChr03g0075061
fasciclin-like arabinogalactan protein (FLA)



Helianthuus annuus

HanXRQChr08g0221961
ferritin



Helianthuus annuus

HanXRQChr09g0257521
FMN-dependent dehydrogenase



Helianthuus annuus

HanXRQChr14g0441641
fructose-bisphosphate aldolase



Helianthuus annuus

HanXRQChr10g0312621
germin



Helianthuus annuus

HanXRQChr09g0244271
glucose-methanol-choline oxidoreductase



Helianthuus annuus

HanXRQChr03g0061571
glutamate synthase



Helianthuus annuus

HanXRQChr05g0144801
glyceraldehyde 3-phosphate dehydrogenase



Helianthuus annuus

HanXRQChr17g0550211
glycerophosphoryl diester phosphodiesterase



Helianthuus annuus

HanXRQChr06g0175391
glycoside hydrolase family 16 - GH16 (endoxyloglucan




transferase)



Helianthuus annuus

HanXRQChr11g0351571
glycoside hydrolase family 17 - GH17 (beta-1,3-glucosidase)



Helianthuus annuus

HanXRQChr05g0141461
glycoside hydrolase family 18 - GH18



Helianthuus annuus

HanXRQChr09g0276721
glycoside hydrolase family 19 - GH19



Helianthuus annuus

HanXRQChr02g0046191
glycoside hydrolase family 2 - GH2



Helianthuus annuus

HanXRQChr16g0524981
glycoside hydrolase family 20 - GH20




(N-acetyl-beta-glucosaminidase)



Helianthuus annuus

HanXRQChr11g0322851
glycoside hydrolase family 27 - GH27




(alpha-galactosidase/melibiase)



Helianthuus annuus

HanXRQChr10g0293191
glycoside hydrolase family 3 - GH3



Helianthuus annuus

HanXRQChr16g0511881
glycoside hydrolase family 31 - GH31 (alpha-xylosidase)



Helianthuus annuus

HanXRQChr14g0461441
glycoside hydrolase family 32 - GH32 (vacuolar invertase)



Helianthuus annuus

HanXRQChr13g0423671
glycoside hydrolase family 35 - GH35 (beta-galactosidase)



Helianthuus annuus

HanXRQChr10g0319301
glycoside hydrolase family 35 - GH35 (beta-galactosidase)



Helianthuus annuus

HanXRQChr09g0256531
glycoside hydrolase family 38 - GH38 (alpha-mannosidase)



Helianthuus annuus

HanXRQChr11g0320901
glycoside hydrolase family 5 - GH5 (glucan-1,3-beta




glucosidase)



Helianthuus annuus

HanXRQChr05g0130491
glycoside hydrolase family 51 - GH51




(alpha-arabinofuranosidase)



Helianthuus annuus

HanXRQChr10g0314191
glycoside hydrolase family 79 - GH79




(endo-beta-glucuronidase/heparanase



Helianthuus annuus

HanXRQChr13g0397411
homologous to A. thaliana PMR5 (Powdery Mildew




Resistant) (carbohydrate acylation)



Helianthuus annuus

HanXRQChr14g0444681
inhibitor family I3 (Kunitz-P family)



Helianthuus annuus

HanXRQChr14g0445181
lactate/malate dehydrogenase



Helianthuus annuus

HanXRQChr17g0564111
lectin (D-mannose)



Helianthuus annuus

HanXRQChr17g0558861
lectin (PAN-2 domain)



Helianthuus annuus

HanXRQChr02g0039251
lipase acylhydrolase (GDSL family)



Helianthuus annuus

HanXRQChr01g0000161
lipid transfer protein/trypsin-alpha amylase inhibitor



Helianthuus annuus

HanXRQChr02g0047121
mannose-binding lectin



Helianthuus annuus

HanXRQChr10g0303361
mitochondrial carrier protein



Helianthuus annuus

HanXRQChr15g0489551
multicopper oxidase



Helianthuus annuus

HanXRQChr05g0135581
neutral/alkaline nonlysosomal ceramidase



Helianthuus annuus

HanXRQChr01g0017621
nucleoside diphosphate kinase



Helianthuus annuus

HanXRQChr10g0295991
peroxidase



Helianthuus annuus

HanXRQChr13g0398251
peroxiredoxin



Helianthuus annuus

HanXRQChr11g0333171
phosphate-induced (phi) protein 1



Helianthuus annuus

HanXRQChr03g0060421
phosphodiesterase/nucleotide pyrophosphatase/phosphate




transferase



Helianthuus annuus

HanXRQChr03g0078011
phosphofructokinase



Helianthuus annuus

HanXRQChr13g0408831
phosphoglycerate kinase



Helianthuus annuus

HanXRQChr10g0286701
phosphoglycerate mutase



Helianthuus annuus

HanXRQChr06g0171591
photosystem II PsbP, oxygen evolving complex



Helianthuus annuus

HanXRQChr14g0434951
plastid lipid-associated protein/fibrillin conserved domain



Helianthuus annuus

HanXRQChr05g0146621
plastocyanin (blue copper binding protein)



Helianthuus annuus

HanXRQChr11g0330251
polyphenol oxidase



Helianthuus annuus

HanXRQChr04g0094541
proteasome A-type subunit



Helianthuus annuus

HanXRQChr03g0081271
proteasome B-type subunit



Helianthuus annuus

HanXRQChr12g0356851
purple acid phosphatase



Helianthuus annuus

HanXRQChr15g0485781
pyridoxal phosphate-dependent transferase



Helianthuus annuus

HanXRQChr11g0336791
ribosomal protein



Helianthuus annuus

HanXRQChr11g0330521
ribosomal protein



Helianthuus annuus

HanXRQChr11g0326801
ribulose bisphosphate carboxylase, large subunit



Helianthuus annuus

HanXRQChr16g0523951
ribulose-1,5-bisphosphate carboxylase small subunit



Helianthuus annuus

HanXRQChr01g0022151
S-adenosyl-L-homocysteine hydrolase



Helianthuus annuus

HanXRQChr14g0454811
S-adenosylmethionine synthetase



Helianthuus annuus

HanXRQChr04g0109991
SCP-like extracellular protein (PR-1)



Helianthuus annuus

HanXRQChr03g0072241
Ser carboxypeptidase (Peptidase family S10)



Helianthuus annuus

HanXRQChr12g0377221
Ser protease (subtilisin) (Peptidase family S8)



Helianthuus annuus

HanXRQChr02g0055581
superoxide dismutase



Helianthuus annuus

HanXRQChr15g0493261
thaumatin (PR5)



Helianthuus annuus

HanXRQChr16g0532531
transketolase



Helianthuus annuus

HanXRQChr07g0197421
translation elongation factor EFTu/EF1A



Helianthuus annuus

HanXRQChr06g0173951
translationally controlled tumour protein








Claims
  • 1. A method for producing plant messenger packs (PMPs), the method comprising: (a) providing a pectin-rich preparation from a plant comprising extracellular vesicles (EVs), the preparation having a turbidity of 0.8 AU or greater at an absorbance of 650 nm;(b) treating the preparation to reduce the turbidity of the preparation or a fraction thereof; and(c) separating PMPs from the preparation or fraction thereof, thereby producing PMPs.
  • 2. A method for producing plant messenger packs (PMPs), the method comprising: (a) providing a pectin-rich preparation from a plant comprising EVs;(b) treating the preparation with an agent that reduces pectin gelation;(c) concentrating the preparation, wherein the viscosity of the concentrated preparation is reduced by at least 10% relative to a concentrated preparation that has not been treated with the agent that reduces pectin gelation; and(d) separating PMPs from the preparation or fraction thereof, thereby producing PMPs.
  • 3. A method for producing plant messenger packs (PMPs), the method comprising: (a) providing a pectin-rich preparation from a plant comprising EVs;(b) treating the preparation to reduce high molecular weight pectin in the preparation or a fraction thereof; and(c) separating PMPs from the preparation or fraction thereof, thereby producing PMPs.
  • 4. A method for producing plant messenger packs (PMPs), the method comprising: (a) providing a pectin-rich preparation from a plant comprising EVs;(b) contacting the preparation or a fraction thereof with a chelating agent; and(c) separating PMPs from the chelated preparation or fraction thereof, thereby producing PMPs.
  • 5. A method for manufacturing PMPs, the method comprising: (a) processing at least 500 g of a pectin-rich plant or plant part comprising EVs into a preparation;(b) contacting the preparation or a fraction thereof with a chelating agent; and(c) processing the chelated preparation or fraction thereof to separate PMPs, wherein the contacting is performed in an amount and for a time sufficient to reduce high molecular weight pectin in the chelated preparation or fraction thereof by at least 10%.
  • 6. The method of claim 5, wherein the processing of step (c) comprises separating the PMPs from the chelated preparation or fraction thereof.
  • 7. The method of any one of claims 4-6, wherein the chelating agent reduces gelation of pectin in the chelated preparation or fraction thereof.
  • 8. The method any one of claims 4-7, wherein the chelating agent is EDTA or EGTA.
  • 9. The method of claim 8, wherein the EDTA or EGTA is in a solution with MES, Tris, or PBS.
  • 10. The method of any one of claims 1-9, further comprising treating the preparation with a pectinase enzyme.
  • 11. A method for producing PMPs, the method comprising: (a) providing a pectin-rich preparation from a plant comprising EVs;(b) contacting the preparation or a fraction thereof with a pectinase enzyme; and(c) separating PMPs from the preparation or fraction thereof, thereby producing PMPs.
  • 12. The method of claim 10 or 11, further comprising removal or inactivation of the pectinase enzyme.
  • 13. The method of any one of claims 1-12, wherein the pectin concentration in the preparation is at least 0.1%.
  • 14. The method of any one of claims 1-13, wherein the PMPs of step (c) are concentrated at least 10× relative to the preparation of step (a).
  • 15. The method of any one of claims 1-14, wherein the separating or processing comprises centrifugation.
  • 16. The method of claim 15, wherein the centrifugation is differential centrifugation.
  • 17. The method of any one of claims 1-16, wherein the separating or processing comprises one or more filtration steps.
  • 18. The method of claim 17, wherein the one or more filtration steps comprise tangential flow filtration.
  • 19. The method of claim 18, wherein the tangential flow filtration comprises exchanging the volume of the preparation at least 10 times.
  • 20. The method of claim 17, wherein the one or more filtration steps comprise size exclusion chromatography.
  • 21. The method of claim 17, wherein the one or more filtration steps comprise tangential flow filtration and size exclusion chromatography.
  • 22. The method of any one of claims 1-17, wherein the separating or processing comprises one, two, or all three of centrifugation, tangential flow filtration, and size exclusion chromatography.
  • 23. The method of any one of claims 1-22, wherein the separating or processing comprises one or more of a wash step, dilution, pH modification, dialysis, and removal of contaminants.
  • 24. The method of any one of claims 1-23, wherein pectin concentration in the PMPs of step (c) is reduced by at least 10% relative to PMPs produced from a preparation that has not been treated.
  • 25. The method of any one of claims 1-24, wherein providing the preparation comprises processing a plant or a plant part to release PMPs.
  • 26. The method of claim 25, wherein the processing comprises blending a plant or a plant part.
  • 27. The method of claim 26, wherein the plant part is a juice sac of a grapefruit or lemon.
  • 28. The method of claim 25, wherein the processing comprises mashing a plant or a plant part through a strainer.
  • 29. The method of claim 25, wherein the processing comprises cold pressing a plant or a plant part.
  • 30. The method of any one of claims 1-29, wherein the preparation is obtained from a pectin-rich plant or a pectin-rich plant part.
  • 31. The method of any one of claims 25, 26, or 28-30, wherein the plant is a citrus plant.
  • 32. The method of claim 31, wherein the citrus plant is a grapefruit or lemon.
  • 33. The method of any one of claims 25, 26, or 28-30, wherein the plant is a flowering plant.
  • 34. The method of any one of claims 25, 26, or 28-30, wherein the plant is a vegetable.
  • 35. The method of any one of claims 1-34, wherein the viscosity of the preparation is monitored.
  • 36. The method of any one of claims 1-35, wherein the viscosity of the preparation is reduced by at least 5% relative to a preparation that has not been treated.
  • 37. The method of any one of claims 1-36, comprising formulating the PMPs produced in step (c) with a carrier.
  • 38. The method of claim 37, wherein the carrier is an agriculturally acceptable carrier.
  • 39. The method of claim 38, wherein the PMPs are formulated for delivery to a plant.
  • 40. The method of claim 39, wherein the carrier is a pharmaceutically acceptable carrier.
  • 41. The method of claim 40, wherein the PMPs are formulated for administration to a human.
  • 42. The method of any one of claims 37-41, wherein the PMPs are formulated with a liquid, a solid, an aerosol, a paste, a gel, or a gas composition.
  • 43. The method of any one of claims 37-42, wherein the PMPs are stable for at least 24 hours, 48 hours, seven days, or 30 days.
  • 44. The method of any one of claims 37-43, wherein the PMPs are stable at a temperature of at least 4° C.
  • 45. The method of any one of claims 37-44, wherein the PMPs are at a concentration of at least 1, 10, 50, 100, or 250 μg PMP protein/ml.
  • 46. The method of any one of claims 1-45, comprising loading the PMPs with a heterologous functional agent.
  • 47. The method of claim 46, wherein the heterologous functional agent is a heterologous agricultural agent.
  • 48. The method of claim 47, wherein the heterologous agricultural agent is a pesticidal agent.
  • 49. The method of claim 47, wherein the heterologous agricultural agent is a fertilizing agent.
  • 50. The method of claim 47, wherein the heterologous agricultural agent is an herbicidal agent.
  • 51. The method of claim 47, wherein the heterologous agricultural agent is a plant-modifying agent.
  • 52. The method of claim 46, wherein the heterologous functional agent is a heterologous therapeutic agent.
  • 53. The method of claim 46, wherein the heterologous functional agent comprises an antifungal agent, an antibacterial agent, a virucidal agent, an anti-viral agent, an insecticidal agent, a nematicidal agent, an antiparasitic agent, or an insect repellent.
  • 54. A PMP composition comprising a plurality of PMPs, wherein the PMPs are produced by a process comprising the steps of: (a) providing a pectin-rich preparation from a plant comprising extracellular vesicles (EVs), the preparation having a turbidity of 0.8 AU or greater at an absorbance of 650 nm;(b) treating the preparation to reduce the turbidity of the preparation or a fraction thereof, and(c) separating PMPs from the preparation or fraction thereof, thereby producing PMPs.
  • 55. A PMP composition comprising a plurality of PMPs, wherein the PMPs are produced by a process comprising the steps of: (a) providing a pectin-rich preparation from a plant comprising EVs;(b) treating the preparation with an agent that reduces pectin gelation;(c) concentrating the preparation, wherein the viscosity of the concentrated preparation is reduced by at least 10% relative to a concentrated preparation that has not been treated with the agent that reduces pectin gelation; and(d) separating PMPs from the preparation or fraction thereof, thereby producing PMPs.
  • 56. A PMP composition comprising a plurality of PMPs, wherein the PMPs are produced by a process comprising the steps of: (a) providing a pectin-rich preparation from a plant comprising EVs;(b) treating the preparation to reduce high molecular weight pectin in the preparation or a fraction thereof; and(c) separating PMPs from the preparation or fraction thereof, thereby producing PMPs.
  • 57. A PMP composition comprising a plurality of PMPs, wherein the PMPs are produced by a process comprising the steps of: (a) providing a pectin-rich preparation from a plant comprising EVs;(b) contacting the preparation or a fraction thereof with a chelating agent; and(c) separating PMPs from the chelated preparation or fraction thereof, threby producing PMPs.
  • 58. A PMP composition comprising a plurality of PMPs, wherein the PMPs are produced by a process comprising the steps of: (a) processing at least 500 g of a pectin-rich plant or plant part comprising EVs into a preparation;(b) contacting the preparation or a fraction thereof with a chelating agent; and(c) processing the chelated preparation or fraction thereof to separate PMPs, wherein the contacting is performed in an amount and for a time sufficient to reduce high molecular weight pectin in the chelated preparation or fraction thereof by at least 10%.
  • 59. The PMP composition of claim 58, wherein the processing of step (c) comprises separating the PMPs from the chelated preparation or fraction thereof.
  • 60. The PMP composition of any one of claims 57-59, wherein the chelating agent reduces polymerization of pectin in the chelated preparation or fraction thereof.
  • 61. The PMP composition of any one of claims 57-60, wherein the chelating agent is EDTA or EGTA.
  • 62. The PMP composition of claim 61, wherein the EDTA or EGTA is in a solution with MES, Tris, or PBS.
  • 63. The PMP composition of any one of claims 54-62, further comprising treating the preparation with a pectinase enzyme.
  • 64. A PMP composition comprising a plurality of PMPs, wherein the PMPs are produced by a process comprising the steps of: (a) providing a pectin-rich preparation from a plant comprising EVs;(b) contacting the preparation or a fraction thereof with a pectinase enzyme; and(c) separating PMPs from the preparation or fraction thereof, thereby producing PMPs.
  • 65. The PMP composition of claim 63 or 64, further comprising removal or inactivation of the pectinase enzyme.
  • 66. The PMP composition of any one of claims 54-65, wherein the PMP composition further comprises a carrier.
  • 67. The PMP composition of claim 66, wherein the carrier is an agriculturally acceptable carrier.
  • 68. The PMP composition of claim 66, wherein the carrier is a pharmaceutically acceptable carrier.
  • 69. The PMP composition of any one of claims 54-68, wherein the composition is formulated as a liquid, a solid, an aerosol, a paste, a gel, or a gas composition.
  • 70. The PMP composition of any one of claims 54-69, wherein the PMP composition is stable for at least 24 hours, 48 hours, seven days, or 30 days.
  • 71. The PMP composition of any one of claims 54-70, wherein the PMP composition is stable at a temperature of at least 4° C.
  • 72. The PMP composition of any one of claims 54-71, wherein the PMPs in the composition are at a concentration of at least 1, 10, 50, 100, or 250 μg PMP protein/ml.
  • 73. A method of increasing the fitness of a plant, the method comprising delivering to the plant an effective amount of the PMP composition of any one of claims 54-72, wherein the method increases the fitness of the plant relative to an untreated plant.
  • 74. A method of decreasing the fitness of a plant pest, the method comprising delivering to the plant pest an effective amount of the PMP composition of any one of claims 54-72, wherein the method decreases the fitness of the plant pest relative to an untreated plant pest.
  • 75. A method of treating an infection in an animal in need thereof, the method comprising administering to the animal an effective amount of the PMP composition of any one of claims 54-72.
  • 76. A method of decreasing the fitness of a pathogen, the method comprising delivering to the pathogen an effective amount of the PMP composition of any one of claims 54-72, wherein the method is effective to decrease the fitness of the pathogen relative to an untreated pathogen.
  • 77. A method of decreasing the fitness of an animal pathogen vector, the method comprising delivering to the vector an effective amount of the PMP composition of any one of claims 54-72, wherein the method decreases the fitness of the vector relative to an untreated vector.
  • 78. A method for producing plant messenger packs (PMPs), the method comprising: (a) providing a pectin-rich preparation from a plant comprising EVs;(b) (i) treating the preparation to reduce the turbidity of the preparation or a fraction thereof; (ii) treating the preparation to reduce the viscosity of the preparation or a fraction thereof; (iii) treating the preparation to reduce high molecular weight pectin in the preparation or a fraction thereof; (iv) contacting the preparation or a fraction thereof with a chelating agent; or (v) contacting the preparation or a fraction thereof with a pectinase enzyme;(c) intermittently or continuously measuring the viscosity of the preparation or fraction thereof during step (b);(d) ending step (b) when the viscosity of the preparation or fraction thereof is below a predetermined level that informs that the preparation or fraction thereof of step (e) will have reduced gelation relative to a preparation or fraction thereof that has not been treated; and(e) separating PMPs from the preparation or fraction thereof.
  • 79. The method of claim 78, wherein viscosity is measured in-process during step (b).
  • 80. The method of claim 78, wherein viscosity is measured intermittently during step (b).
  • 81. The method of claim 78, wherein viscosity is measured continuously during at least a portion of step (b).
  • 82. The method of claim 78, wherein viscosity is measured continuously during step (b).
  • 83. The method of any one of claims 78-82, wherein the predetermined level of viscosity is 1.4 cP when viscosity is measured at 20° C.
  • 84. The method of any one of claims 78-83, wherein the temperature of the composition during step (b) is 20° C.
  • 85. A method for producing plant messenger packs (PMPs), the method comprising: (a) providing a pectin-rich preparation from a plant comprising EVs;(b) (i) treating the preparation to reduce the turbidity of the preparation or a fraction thereof; (ii) treating the preparation to reduce the viscosity of the preparation or a fraction thereof; (iii) treating the preparation to reduce high molecular weight pectin in the preparation or a fraction thereof; (iv) contacting the preparation or a fraction thereof with a chelating agent; or (v) contacting the preparation or a fraction thereof with a pectinase enzyme;(c) intermittently or continuously measuring the turbidity of the preparation or fraction thereof during step (b);(d) ending step (b) when the turbidity of the preparation or fraction thereof is below a predetermined level that informs that the preparation or fraction thereof of step (e) will have reduced gelation relative to a preparation or fraction thereof that has not been treated; and(e) separating PMPs from the preparation or fraction thereof.
  • 86. The method of claim 85, wherein turbidity is measured in-process during step (b).
  • 87. The method of claim 85, wherein turbidity is measured intermittently during step (b).
  • 88. The method of claim 85, wherein turbidity is measured continuously during at least a portion of step (b).
  • 89. The method of claim 85, wherein turbidity is measured continuously during step (b).
  • 90. The method of any one of claims 85-89, wherein the predetermined level of turbidity is 0.8 AU at an absorbance of 650 nm.
PCT Information
Filing Document Filing Date Country Kind
PCT/US2020/029886 4/24/2020 WO 00
Provisional Applications (2)
Number Date Country
62848466 May 2019 US
62838930 Apr 2019 US