WO2022051406A1 - Use of vitellogenin for defining and testing novel immunogens in insects - Google Patents

Use of vitellogenin for defining and testing novel immunogens in insects Download PDF

Info

Publication number
WO2022051406A1
WO2022051406A1 PCT/US2021/048727 US2021048727W WO2022051406A1 WO 2022051406 A1 WO2022051406 A1 WO 2022051406A1 US 2021048727 W US2021048727 W US 2021048727W WO 2022051406 A1 WO2022051406 A1 WO 2022051406A1
Authority
WO
WIPO (PCT)
Prior art keywords
paenibacillus
honey bee
inactivated
rvg
pathogen
Prior art date
Application number
PCT/US2021/048727
Other languages
French (fr)
Inventor
Huw HUGHES
Original Assignee
Dalan Animal Health, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Dalan Animal Health, Inc. filed Critical Dalan Animal Health, Inc.
Priority to US18/043,723 priority Critical patent/US20240018183A1/en
Priority to EP21865059.6A priority patent/EP4208150A4/en
Publication of WO2022051406A1 publication Critical patent/WO2022051406A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/16Extraction; Separation; Purification by chromatography
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/43504Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates
    • C07K14/43563Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates from insects
    • C07K14/43572Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates from insects from bees
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics

Definitions

  • Vitellogenin is a precursor protein of egg yolk normally in the blood or hemolymph only of females that is used as a biomarker in vertebrates of exposure to environmental estrogens which stimulate elevated levels in males as well as females.
  • the protein product of the gene is classified as a glycolipoprotein, having properties of a sugar, fat and protein. It is the precursor of the lipoproteins and phosphoproteins that make up most of the protein content of yolk.
  • Vitellogenin has been shown to bind to various proteins and transport them to ovaries in queen bees, where they are then passed on through eggs to newly hatched larvae. These proteins are capable of activating the non-specific insect immune mechanisms in insects, with the net result of a protective effect against pathogens, such as American foulbrood (AFB).
  • American foulbrood is a fatal bacterial disease of honey bee brood caused by the spore forming bacterium Paenibacillus larvae (see htps://beeaware.org.aU/archive-pest/american-foulbrood/#ad-image-0).
  • this disclosure provides the methods and compositions for the identification, isolation, preparation and use of these protective proteins and fragments thereof.
  • the protection-inducing proteins an immunogenic fragments thereof from honey bee pathogens.
  • recombinant vitellogenin is produced in an insect viral expression system, such as a baculovirus expression system.
  • the rVg is bound to a sepharose CL column or other chromatographic support using cyanogen bromide or other covalent linking mechanism known to those skilled in the art.
  • rVg may be bound to an ion exchange (IEX) column at favourable conditions (optimal pH, salt).
  • IEX ion exchange
  • Suitable methods for inactivation include for example, sonication, followed by solubilization in non-ionic detergents such as Brij, Triton, octyl-glucoside and others known in the art.
  • Honey bee pathogens are those responsible for American foulbrood, European foulbrood, deformed wing virus, varroa mite infestations and other viral, bacterial and parasitic diseases of honey bees. Solubilized protein mixtures of these pathogens are applied to the solid phase with rVg, and proteins that bind to rVg are eluted using a salt gradient and/or pH gradient. The rVg is covalently bound to the column and the proteins of interest are eluted.
  • the rVg can be ionically bound to IEX gels, the rVg-protein mixture is eluted depending on binding characteristics, ionic strength, etc. Purified proteins or protein- rVg complexes are then identified using standard methods such as electrophoresis, HPLC, MS, etc.
  • rVg is used in the place of antibodies to detect protein in high throughput methods such as phage display, high throughput screening of the entire pathogen genome, and other methods known in the art. Labeled rVg or a polyclonal antibody is used to detect the proteins of interest, and then they may be further characterized. This method allows for the rapid screening of multiple disease agents extremely quickly.
  • vaccines and bacterins identified by these methods. After the proteins of interest are identified, they are partially sequenced, and the genes expressing such proteins are isolated, and the proteins are produced in a suitable eukaryotic or prokaryotic expression system (baculovirus, CHO, HEK, E. coll. Saccharomyces. and others known in the art).
  • the recombinant proteins can be added to feed for queen bees as is described in U.S. Patent No. 10,994,001, or may be introduced directly into hives in a suitable feed substance. Immunity in honey bees and larvae is generated and assessed as described in U.S. Patent No. 10,994,001.
  • Toll-like receptor (TLR) binding CpGs may be added to the vaccines or bacterins to enhance effector mechanisms.
  • FIG. 1 shows a one dimensional gel using the below methods. 2 different blocking agents were assessed. Superblock was chosen as it appeared to provide a clearer image with less background staining. From Left to right: Vitellogenin blots with PF block, Vitellogenin blot with Superblock, Detection antibodies alone (i.e. anti-His tag and anti- mouse antibodies) and Coomassie stained (protein) gel.
  • FIG. 2 shows a two dimensional gel. Methods are described below. 2D pattern following staining with detection antibodies only (no vitellogenin).
  • FIG. 3 shows a 2D gel pattern following staining with vitellogenin and detection antibodies. The circled spots are those that were detected uniquely by vitellogenin, i.e. did not bind to either of the detection antibodies.
  • FIG. 4 is a Coomassie stained gel indicating the proteins detected by specific vitellogenin staining.
  • FIG. 5 shows a map of an exemplary recombinant vitellogenin construct.
  • a cell includes a plurality of cells, including mixtures thereof.
  • compositions and methods include the recited elements, but not excluding others.
  • Consisting essentially of when used to define compositions and methods, shall mean excluding other elements of any essential significance to the combination for the stated purpose. Thus, a composition consisting essentially of the elements as defined herein would not exclude trace contaminants from the isolation and purification method and pharmaceutically acceptable carriers, such as phosphate buffered saline, preservatives and the like.
  • Consisting of’ shall mean excluding more than trace elements of other ingredients and substantial method steps for administering the compositions of this invention or process steps to produce a composition or achieve an intended result. Embodiments defined by each of these transition terms are within the scope of this invention.
  • isolated refers to molecules separated from other DNAs or RNAs, respectively, that are present in the natural source of the macromolecule.
  • isolated as used herein also refers to a nucleic acid or peptide that is substantially free of cellular material, viral material, or culture medium when produced by recombinant DNA techniques, or chemical precursors or other chemicals when chemically synthesized.
  • an “isolated nucleic acid” is meant to include nucleic acid fragments which are not naturally occurring as fragments and would not be found in the natural state.
  • isolated is also used herein to refer to cells or polypeptides which are isolated from other cellular proteins or tissues. Isolated polypeptides is meant to encompass both purified and recombinant polypeptides.
  • Cells “host cells” or “recombinant host cells” are terms used interchangeably herein. It is understood that such terms refer not only to the particular subject cell but to the progeny or potential progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term as used herein.
  • Host cells can be prokaryotic or eukaryotic.
  • Prokaryotic cells that usually lack a nucleus or any other membrane-bound organelles and are divided into two domains, bacteria and archaea. In addition to chromosomal DNA, these cells can also contain genetic information in a circular loop called an episome. Bacterial cells are very small, roughly the size of an animal mitochondrion (about 1-2 pm in diameter and 10 pm long). Prokaryotic cells feature three major shapes: rod shaped, spherical, and spiral. Instead of going through elaborate replication processes like eukaryotes, bacterial cells divide by binary fission. Examples include but are not limited to Bacillus bacteria, E. coli bacterium, and Salmonella bacterium.
  • Eukaryotic cells comprise all of the life kingdoms except monera. They can be easily distinguished through a membrane-bound nucleus. Animals, plants, fungi, and protists are eukaryotes or organisms whose cells are organized into complex structures by internal membranes and a cytoskeleton. The most characteristic membrane-bound structure is the nucleus. Unless specifically recited, the term “host” includes a eukaryotic host, including, for example, yeast, higher plant, insect and mammalian cells.
  • Non-limiting examples of eukaryotic cells or hosts include simian, bovine, porcine, murine, rat, avian, reptilian and human, e.g., HEK293 cells, Chinese Hamster Ovary (CHO) cells, CHO-S cells, CHO-K1 cells, 293T cells, HeLa cells, Baby hamster kidney (BHK) cells, and Sf9 cells.
  • Sf9 cells are a clonal isolate of Spodopter frugiperda. They were originally isolated from ovarian tissue and are commonly used in insect cell culture for recombinant protein production using baculovirus. These cells are commercially available from American Type Culture Collection (ATCC) under CRL-1711.
  • DHIOBac can serve as a host for a recombinant pFastBac vector containing a cloned gene of interest.
  • ThermoFisher Scientific provides these cells as MAX Efficiency DHIOBac Competent Cells.
  • These DHIOBac cells harbor a baculovirus shuttle vector (bMON14272) and a helper plasmid (pMON7142), and are capable of supporting site-specific recombination between pFastBac and bMON14272 to generate high molecular weight bacmids that can then be amplified, purified, and used for insect cell transfection and subsequent gene expression.
  • Kanamycin resistance for bacmid selection and maintenance is conferred by bMON14272, and tetracycline resistance by pMON7124. These cells are commercially available from ThermoFisher Scientific (htps://www.thermofisher.com/order/catalog/product/10361012#/10361012, last accessed on August 28, 2021).
  • a baculovirus-insect expression system utilizes recombinant baculovirus (insect viruses) and their ability to manufacture high yields of biologically active proteins from insect cells. These systems are known in the art and commercially available. See, e.g., Jarvis, Baculovirus-insect cell expression systems, Methods Enzymol. (2009)463: 191-222; Scholz, J., Suppmann, S., A new single-step protocol for rapid baculovirus-driven protein production in insect cells. BMC Biotechnol. Vol.
  • ELISA enzyme-linked immunosorbent assay
  • EIA enzyme immunoassay
  • antigens e.g., antigens from the sample to be tested are attached to a surface.
  • a matching antibody is applied over the surface so it can bind the antigen.
  • This antibody is linked to an enzyme and then any unbound antibodies are removed.
  • a substance containing the enzyme's substrate is added. If there was binding the subsequent reaction produces a detectable signal, most commonly a color change.
  • Performing an ELISA involves at least one antibody with specificity for a particular antigen.
  • the sample with an unknown amount of antigen is immobilized on a solid support (e.g., a polystyrene microtiter plate) either non- specifically (via adsorption to the surface) or specifically (via capture by another antibody specific to the same antigen, in a "sandwich” ELISA).
  • the detection antibody is added, forming a complex with the antigen.
  • the detection antibody can be covalently linked to an enzyme or label or can itself be detected by a secondary antibody that is linked to an enzyme through bioconjugation.
  • the plate is typically washed with a mild detergent solution to remove any proteins or antibodies that are non-specifically bound.
  • the plate is developed by adding an enzymatic substrate to produce a signal, which indicates the quantity of antigen in the sample.
  • ELISA can also be performed in other forms of ligand binding assays.
  • the technique essentially requires any ligating reagent that can be immobilized on the solid phase along with a detection reagent that will bind specifically and use an enzyme to generate a signal that can be properly quantified. In between the washes, only the ligand and its specific binding counterparts remain specifically bound or "immimosorbed" by antigen- antibody interactions or other interactions (protein-protein, e.g., Vitellogenin binding to a bee pathogen to the solid phase, while the nonspecific or unbound components are washed away.
  • protein-protein e.g., Vitellogenin binding to a bee pathogen to the solid phase
  • “Amplify” “amplifying” or “amplification” of a polynucleotide sequence includes methods such as traditional cloning methodologies, PCR, ligation amplification (or ligase chain reaction, LCR) or other amplification methods. These methods are known and practiced in the art. See, e.g., U.S. Patent Nos. 4,683,195 and 4,683,202 and Innis et al. (1990) Mol. Cell Biol. 10(11):5977-5982 (for PCR); and Wu et al. (1989) Genomics 4:560- 569 (for LCR).
  • the PCR procedure describes a method of gene amplification which is comprised of (i) sequence-specific hybridization of primers to specific genes within a DNA sample (or library), (ii) subsequent amplification involving multiple rounds of annealing, elongation, and denaturation using a DNA polymerase, and (iii) screening the PCR products for a band of the correct size.
  • the primers used are oligonucleotides of sufficient length and appropriate sequence to provide initiation of polymerization, i.e. each primer is specifically designed to be complementary to each strand of the genomic locus to be amplified.
  • Reagents and hardware for conducting PCR are commercially available. Primers useful to amplify sequences from a particular region are preferably complementary to, and hybridize specifically to sequences in the target region or in its flanking regions. Nucleic acid sequences generated by amplification may be sequenced directly. Alternatively the amplified sequence(s) may be cloned prior to sequence analysis. A method for the direct cloning and sequence analysis of enzymatically amplified genomic segments is known in the art.
  • genotype refers to the specific allelic composition of an entire cell, a certain gene or a specific polynucleotide region of a genome, whereas the term “phenotype’ refers to the detectable outward manifestations of a specific genotype.
  • gene refers to a nucleic acid molecule comprising an open reading frame and including at least one exon and (optionally) an intron sequence.
  • a gene may also refer to a polymorphic or a mutant form or allele of a gene.
  • Homology or “identity” or “similarity” refers to sequence similarity between two peptides or between two nucleic acid molecules. Homology can be determined by comparing a position in each sequence which may be aligned for purposes of comparison. When a position in the compared sequence is occupied by the same base or amino acid, then the molecules are homologous at that position. A degree of homology between sequences is a function of the number of matching or homologous positions shared by the sequences. An “unrelated” or “non-homologous” sequence shares less than 40% identity, though preferably less than 25% identity, with one of the sequences of the present invention.
  • a polynucleotide or polynucleotide region (or a polypeptide or polypeptide region) has a certain percentage (for example, 60 %, 65 %, 70 %, 75 %, 80 %, 85 %, 90 %, 95 %, 98 % or 99 %) of “sequence identity” to another sequence means that, when aligned, that percentage of bases (or amino acids) are the same in comparing the two sequences.
  • This alignment and the percent homology or sequence identity can be determined using software programs known in the art, for example those described in Ausubel et al. eds. (2007) Current Protocols in Molecular Biology. Preferably, default parameters are used for alignment.
  • One alignment program is BLAST, using default parameters.
  • Biologically equivalent polynucleotides are those having the specified percent homology and encoding a polypeptide having the same or similar biological activity.
  • an equivalent nucleic acid refers to a nucleic acid having a nucleotide sequence having a certain degree of homology with the nucleotide sequence of the nucleic acid or complement thereof.
  • a homolog of a double stranded nucleic acid is intended to include nucleic acids having a nucleotide sequence which has a certain degree of homology with or with the complement thereof.
  • homologs of nucleic acids are capable of hybridizing to the nucleic acid or complement thereof.
  • rhe term interact or contacting is also meant to include “binding” interactions between molecules. Interactions may be, for example, protein-protein, protein-nucleic acid, or nucleic acid-nucleic acid in nature.
  • Hybridization refers to a reaction in which one or more polynucleotides react to form a hybridization complex that is stabilized via hydrogen bonding between the bases of the nucleotide residues.
  • the hydrogen bonding may occur by Watson-Crick base pairing, Hoogstein binding, or in any other sequence-specific manner.
  • the complex may comprise two strands forming a duplex structure, three or more strands forming a multi-stranded complex, a single self-hybridizing strand, or any combination of these.
  • a hybridization reaction may constitute a step in a more extensive process, such as the initiation of a PCR reaction, or the enzymatic cleavage of a polynucleotide by a ribozyme.
  • Hybridization reactions can be performed under conditions of different “stringency”. In general, a low stringency hybridization reaction is carried out at about 40°C in about 10 x SSC or a solution of equivalent ionic strength/temperature. A moderate stringency hybridization is typically performed at about 50°C in about 6 x SSC, and a high stringency hybridization reaction is generally performed at about 60°C in about 1 x SSC.
  • Hybridization reactions can also be performed under “physiological conditions” which is well known to one of skill in the art.
  • a non-limiting example of a physiological condition is the temperature, ionic strength, pH and concentration of Mg 2+ normally found in a cell.
  • a double-stranded polynucleotide can be “complementary” or “homologous” to another polynucleotide, if hybridization can occur between one of the strands of the first polynucleotide and the second.
  • “Complementarity” or “homology” is quantifiable in terms of the proportion of bases in opposing strands that are expected to form hydrogen bonding with each other, according to generally accepted base-pairing rules.
  • mismatches refers to hybridized nucleic acid duplexes which are not 100% homologous. The lack of total homology may be due to deletions, insertions, inversions, substitutions or frameshift mutations.
  • oligonucleotide refers to polynucleotides such as deoxyribonucleic acid (DNA), and, where appropriate, ribonucleic acid (RNA).
  • DNA deoxyribonucleic acid
  • RNA ribonucleic acid
  • Deoxyribonucleotides include deoxyadenosine, deoxycytidine, deoxyguanosine, and deoxythymidine.
  • nucleotide of a nucleic acid which can be DNA or an RNA
  • adenosine cytidine
  • guanosine thymidine
  • thymidine a nucleotide having a uracil base
  • polynucleotide and “oligonucleotide” are used interchangeably and refer to a polymeric form of nucleotides of any length, either deoxyribonucleotides or ribonucleotides or analogs thereof. Polynucleotides can have any three-dimensional structure and may perform any function, known or unknown.
  • polynucleotides a gene or gene fragment (for example, a probe, primer, EST or SAGE tag), exons, introns, messenger RNA (mRNA), transfer RNA, ribosomal RNA, ribozymes, cDNA, dsRNA, siRNA, miRNA, recombinant polynucleotides, branched polynucleotides, plasmids, vectors, isolated DNA of any sequence, isolated RNA of any sequence, nucleic acid probes and primers.
  • a polynucleotide can comprise modified nucleotides, such as methylated nucleotides and nucleotide analogs.
  • modifications to the nucleotide structure can be imparted before or after assembly of the polynucleotide.
  • the sequence of nucleotides can be interrupted by non-nucleotide components.
  • a polynucleotide can be further modified after polymerization, such as by conjugation with a labeling component.
  • the term also refers to both double- and single-stranded molecules. Unless otherwise specified or required, any embodiment of this invention that is a polynucleotide encompasses both the double-stranded form and each of two complementary single-stranded forms known or predicted to make up the double-stranded form.
  • a polynucleotide is composed of a specific sequence of four nucleotide bases: adenine (A); cytosine (C); guanine (G); thymine (T); and uracil (U) for thymine when the polynucleotide is RNA.
  • A adenine
  • C cytosine
  • G guanine
  • T thymine
  • U uracil
  • polynucleotide sequence is the alphabetical representation of a polynucleotide molecule. This alphabetical representation can be input into databases in a computer having a central processing unit and used for bioinformatics applications such as functional genomics and homology searching.
  • polymorphism refers to the coexistence of more than one form of a gene or portion thereof.
  • a polymorphic region can be a single nucleotide, the identity of which differs in different alleles.
  • the term “carrier” encompasses any of the standard carriers, such as a phosphate buffered saline solution, buffers, water, and emulsions, such as an oil/water or water/oil emulsion, and various types of wetting agents.
  • the compositions also can include stabilizers and preservatives.
  • stabilizers and adjuvants see Sambrook and Russell (2001), supra. Those skilled in the art will know many other suitable carriers for binding polynucleotides, or will be able to ascertain the same by use of routine experimentation.
  • the carrier is a buffered solution such as, but not limited to, a PCR buffer solution.
  • a “gene delivery vehicle” is defined as any molecule that can carry inserted polynucleotides into a host cell.
  • Examples of gene delivery vehicles are liposomes, biocompatible polymers, including natural polymers and synthetic polymers; lipoproteins; polypeptides; polysaccharides; lipopolysaccharides; artificial viral envelopes; metal particles; and bacteria, or viruses, such as baculovirus, adenovirus and retrovirus, bacteriophage, cosmid, plasmid, fungal vectors and other recombination vehicles typically used in the art which have been described for expression in a variety of eukaryotic and prokaryotic hosts, and may be used for gene therapy as well as for simple protein expression.
  • Gene delivery are terms referring to the introduction of an exogenous polynucleotide (sometimes referred to as a “transgene”) into a host cell, irrespective of the method used for the introduction.
  • exogenous polynucleotide sometimes referred to as a “transgene”
  • transgene an exogenous polynucleotide
  • Such methods include a variety of well-known techniques such as vector-mediated gene transfer (by, e.g., viral infection, sometimes called transduction), transfection, transformation or various other protein-based or lipid-based gene delivery complexes) as well as techniques facilitating the delivery of “naked” polynucleotides (such as electroporation, “gene gun” delivery and various other techniques used for the introduction of polynucleotides).
  • transfected, transduced or transformed may be used interchangeably herein to indicate the presence of exogenous polynucleotides or the expressed polypeptide therefrom in a cell.
  • the introduced polynucleotide may be stably or transiently maintained in the host cell. Stable maintenance typically requires that the introduced polynucleotide either contains an origin of replication compatible with the host cell or integrates into a replicon of the host cell such as an extrachromosomal replicon (e.g., a plasmid) or a nuclear or mitochondrial chromosome.
  • a number of vectors are known to be capable of mediating transfer of genes to mammalian cells, as is known in the art and described herein.
  • the term “express” refers to the production of a gene product.
  • the gene product is a polypeptide or protein.
  • the gene product is a mRNA, a tRNA, a rRNA, a miRNA, a dsRNA, or a siRNA.
  • a cell that “stably expresses” an exogenous polypeptide is one that continues to express a polypeptide encoded by an exogenous gene introduced into the cell either after replication if the cell is dividing or for longer than a day, up to about a week, up to about two weeks, up to three weeks, up to four weeks, for several weeks, up to a month, up to two months, up to three months, for several months, up to a year or more.
  • a “viral vector” is defined as a recombinantly produced virus or viral particle that comprises a polynucleotide to be delivered into a host cell, either in vivo, ex vivo or in vitro.
  • viral vectors include retroviral vectors, lentiviral vectors, adenovirus vectors, adeno-associated virus vectors, alphavirus vectors and the like.
  • Alphavirus vectors such as Semliki Forest virus-based vectors and Sindbis virus-based vectors, have also been developed for use in gene therapy and immunotherapy. See, Schlesinger and Dubensky (1999) Curr. Opin. Biotechnol. 5:434-439 and Ying, et al. (1999) Nat. Med. 5(7):823-827.
  • a vector construct refers to the polynucleotide comprising the retroviral genome or part thereof, and a therapeutic gene.
  • retroviral mediated gene transfer or “retroviral transduction” carries the same meaning and refers to the process by which a gene or nucleic acid sequences are stably transferred into the host cell by virtue of the virus entering the cell and integrating its genome into the host cell genome.
  • the virus can enter the host cell via its normal mechanism of infection or be modified such that it binds to a different host cell surface receptor or ligand to enter the cell.
  • Retroviruses carry their genetic information in the form of RNA; however, once the virus infects a cell, the RNA is reverse-transcribed into the DNA form which integrates into the genomic DNA of the infected cell.
  • the integrated DNA form is called a provirus.
  • retroviral vector refers to a viral particle capable of introducing exogenous nucleic acid into a cell through a viral or viral-like entry mechanism.
  • a “lentiviral vector” is a type of retroviral vector well-known in the art that has certain advantages in transducing nondividing cells as compared to other retroviral vectors. See, Trono D. (2002) Lentiviral vectors, New York: Spring-Verlag Berlin Heidelberg.
  • a vector construct refers to the polynucleotide comprising the viral genome or part thereof, and a transgene.
  • Ads adenoviruses
  • Ads are a relatively well characterized, homogenous group of viruses, including over 50 serotypes. See, e.g., International PCT Application No. WO 95/27071. Ads do not require integration into the host cell genome. Recombinant Ad derived vectors, particularly those that reduce the potential for recombination and generation of wild-type virus, have also been constructed. See, International PCT Application Nos.
  • Wild-type AAV has high infectivity and specificity integrating into the host cell’s genome. See, Hermonat and Muzyczka (1984) Proc. Natl. Acad. Sci. USA 81 :6466- 6470 and Lebkowski, et al. (1988) Mol. Cell. Biol. 8:3988-3996.
  • Vectors that contain both a promoter and a cloning site into which a polynucleotide can be operatively linked are well known in the art. Such vectors are capable of transcribing RNA in vitro or in vivo, and are commercially available from sources such as Stratagene (La Jolla, CA) and Promega Biotech (Madison, WI). In order to optimize expression and/or in vitro transcription, it may be necessary to remove, add or alter 5’ and/or 3’ untranslated portions of the clones to eliminate extra, potential inappropriate alternative translation initiation codons or other sequences that may interfere with or reduce expression, either at the level of transcription or translation. Alternatively, consensus ribosome binding sites can be inserted immediately 5’ of the start codon to enhance expression.
  • Under transcriptional control is a term well understood in the art and indicates that transcription of a polynucleotide sequence, usually a DNA sequence, depends on its being operatively linked to an element which contributes to the initiation of, or promotes, transcription. “Operatively linked” intends the polynucleotides are arranged in a manner that allows them to function in a cell.
  • a “probe” when used in the context of polynucleotide manipulation refers to an oligonucleotide that is provided as a reagent to detect a target potentially present in a sample of interest by hybridizing with the target.
  • a probe will comprise a label or a means by which a label can be attached, either before or subsequent to the hybridization reaction. Suitable labels are described and exemplified herein.
  • a “primer” is a short polynucleotide, generally with a free 3’ -OH group that binds to a target or “template” potentially present in a sample of interest by hybridizing with the target, and thereafter promoting polymerization of a polynucleotide complementary to the target.
  • a “polymerase chain reaction” (“PCR”) is a reaction in which replicate copies are made of a target polynucleotide using a “pair of primers” or a “set of primers” consisting of an “upstream” and a “downstream” primer, and a catalyst of polymerization, such as a DNA polymerase, and typically a thermally-stable polymerase enzyme.
  • a primer can also be used as a probe in hybridization reactions, such as Southern or Northern blot analyses. Sambrook et al., supra.
  • the primers may optionall contain detectable labels and are exemplified and described herein.
  • label intends a directly or indirectly detectable compound or composition that is conjugated directly or indirectly to the composition to be detected, e.g., polynucleotide or protein such as an antibody so as to generate a "labeled" composition.
  • the term also includes sequences conjugated to the polynucleotide that will provide a signal upon expression of the inserted sequences, such as green fluorescent protein (GFP) and the like.
  • the label may be detectable by itself (e.g. radioisotope labels or fluorescent labels) or, in the case of an enzymatic label, may catalyze chemical alteration of a substrate compound or composition which is detectable.
  • the labels can be suitable for small scale detection or more suitable for high-throughput screening.
  • suitable labels include, but are not limited to radioisotopes, fluorochromes, chemiluminescent compounds, dyes, and proteins, including enzymes.
  • the label may be simply detected or it may be quantified.
  • a response that is simply detected generally comprises a response whose existence merely is confirmed, whereas a response that is quantified generally comprises a response having a quantifiable (e.g., numerically reportable) value such as an intensity, polarization, and/or other property.
  • the detectable response may be generated directly using a luminophore or fluorophore associated with an assay component actually involved in binding, or indirectly using a luminophore or fluorophore associated with another (e.g., reporter or indicator) component.
  • luminescent labels that produce signals include, but are not limited to bioluminescence and chemiluminescence. Detectable luminescence response generally comprises a change in, or an occurrence of, a luminescence signal. Suitable methods and luminophores for luminescently labeling assay components are known in the art and described for example in Haugland, Richard P. (1996) Handbook of Fluorescent Probes and Research Chemicals (6 th ed.). Examples of luminescent probes include, but are not limited to, aequorin and luciferases.
  • fluorescent labels include, but are not limited to, fluorescein, rhodamine, tetramethylrhodamine, eosin, erythrosin, coumarin, methyl-coumarins, pyrene, Malacite green, stilbene, Lucifer Yellow, Cascade Blue.TM., and Texas Red.
  • suitable optical dyes are described in the Haugland, Richard P. (1996) Handbook of Fluorescent Probes and Research Chemicals (6 th ed.).
  • the fluorescent label is functionalized to facilitate covalent attachment to a cellular component present in or on the surface of the cell or tissue such as a cell surface marker.
  • Suitable functional groups including, but not are limited to, isothiocyanate groups, amino groups, haloacetyl groups, maleimides, succinimidyl esters, and sulfonyl halides, all of which may be used to attach the fluorescent label to a second molecule.
  • the choice of the functional group of the fluorescent label will depend on the site of attachment to either a linker, the agent, the marker, or the second labeling agent.
  • Attachment of the fluorescent label may be either directly to the cellular component or compound or alternatively, can by via a linker.
  • Suitable binding pairs for use in indirectly linking the fluorescent label to the intermediate include, but are not limited to, antigens/antibodies, e.g., rhodamine/anti-rhodamine, biotin/avidin and biotin/strepavidin.
  • solid support refers to non-aqueous surfaces such as “culture plates” “gene chips” or “microarrays.”
  • gene chips or microarrays can be used for diagnostic and therapeutic purposes by a number of techniques known to one of skill in the art.
  • oligonucleotides are attached and arrayed on a gene chip for determining the DNA sequence by the hybridization approach, such as that outlined in U.S. Patent Nos.: 6,025,136 and 6,018,041.
  • the polynucleotides of this invention can be modified to probes, which in turn can be used for detection of a genetic sequence.
  • Such techniques have been described, for example, in U.S. Patent Nos.: 5,968,740 and 5,858,659.
  • a probe also can be attached or affixed to an electrode surface for the electrochemical detection of nucleic acid sequences such as described by Kayem et al. U.S. Patent No. 5,952,172 and by Kelley et al. (1999) Nucleic Acids Res. 27:4830-4837.
  • Vitellogenin is a precursor of egg yolk that transports protein and some lipid from the liver through the blood to the growing oocytes where it becomes part of the yolk. Normally, it is only found in the blood or hemolymph of females, and can therefore be used as a biomarker in vertebrates of exposure to environmental estrogens which stimulate elevated levels in males as well as females. "Vitellogenin” is a synonymous term for the gene and the expressed protein. The protein product is classified as a glycolipoprotein, having properties of a sugar, fat and protein.
  • Vg polynucleotide and protein sequences can be found in UniProt database, e.g., https://www.uniprot.org/uniprot/Q868N5 and https://www.uniprot.org/uniprot/Q868N5, last accessed on August 28, 2021 , and the sequences provided herein.
  • composition is intended to mean a combination of active agent and another compound or composition, inert (for example, a detectable agent or label) or active, such as an adjuvant.
  • a “pharmaceutical composition” is intended to include the combination of an active agent with a carrier, inert or active, making the composition suitable for diagnostic or therapeutic use in vitro, in vivo or ex vivo.
  • the term “pharmaceutically acceptable carrier” encompasses any of the standard pharmaceutical carriers, such as a phosphate buffered saline solution, water, and emulsions, such as an oil/water or water/oil emulsion, and various types of wetting agents.
  • the compositions also can include stabilizers and preservatives.
  • stabilizers and adjuvants see Martin (1975) Remington’s Pharm. Sci., 15th Ed. (Mack Publ. Co., Easton).
  • An “artificial diet” intends a honey bee diet with an antigen or fragment thereof as described herein.
  • Substantially homogeneous describes a population of cells in which more than about 50%, or alternatively more than about 60 %, or alternatively more than 70 %, or alternatively more than 75 %, or alternatively more than 80%, or alternatively more than 85 %, or alternatively more than 90%, or alternatively, more than 95 %, of the cells are of the same or similar phenotype.
  • Phenotype can be determined by a pre-selected cell surface marker or other marker, e.g. myosin or actin or the expression of a gene or protein, e.g. a calcium handling protein, a t-tubule protein or alternatively, a calcium pump protein.
  • the substantially homogenous population have a decreased (e.g., less than about 95%, or alternatively less than about 90%, or alternatively less than about 80%, or alternatively less than about 75%, or alternatively less than about 70%, or alternatively less than about 65%, or alternatively less than about 60%, or alternatively less than about 55%, or alternatively less than about 50%) of the normal level of expression than the wild-type counterpart cell or tissue.
  • a decreased e.g., less than about 95%, or alternatively less than about 90%, or alternatively less than about 80%, or alternatively less than about 75%, or alternatively less than about 70%, or alternatively less than about 65%, or alternatively less than about 60%, or alternatively less than about 55%, or alternatively less than about 50%
  • American foulbrood or “American foulbrood disease” or “AFB” as used herein, refers to a fatal bacterial disease of honeybee brood caused by the spore forming bacterium Paenibacillus larvae. Since Paenibacillus larvae causes American fouldbrood disease in honeybees, Paenibacillus larvae is referred to herein as a “disease- causing” bacterium or a “disease species.”
  • non-disease causing” species or “non-disease species” as used herein refer to species of bacteria which may or may not be pathogenic but which do not cause the disease being targeted.
  • exemplary non-disease Paenibacillus species which may be found for example in the environment, but not on humans or in human wounds may include e.g., Paenibacillus alvei, Paenibacillus dendritiformis Paenibacillus amylolyticus, Paenibacillus campinasensis, Paenibacillus chondr oitinus, Paenibacillus chungangensis, Paenibacillus doosanensis, Paenibacillus glucanolyticus, Paenibacillus humicus, Paenibacillus lactis, Paenibacillus lautus, Pa
  • Paenibacillus phoenicis Paenibacillus polymyxa, Paenibacillus popilliae. Paenibacillus pulde ungensis, Paenibacillus residui, Paenibacillus stellife, Paenibacillus thiaminolyticus, Paenibacillus validus, and Paenibacillus xylanisolvens.
  • Paenibacillus alvei and Paenibacillus dendritiformis are pathogenic and will inflict a brood disease on larvae but the disease is not American Foulbrood caused by Paenibacillus larvae and are thus non-disease forming species.
  • honey bee refers to is any bee which is a member of the genus Apis, primarily distinguished by the production and storage of honey and the construction of perennial, colonial nests from wax.
  • honey bees include but are not limited to Apis andreniformis and Apis florea in subgenus Micrapis, Apis dorsata in subgenus Megapis, and Apis cerana, Apis koschevnikovi, Apis mellifera and Apis nigrocincta in subgenus Apis.
  • bee colony refers to a social unit of bees, e.g., honeybees comprising a colony.
  • the social unit can be of any system organization utilized by bees, which has the purpose of facilitating survival of the group or colony.
  • a "bee colony” consists of several thousand bees that cooperate in nest building, food collection, and brood rearing.
  • Each member of a "bee colony” has a definite task to perform, and it takes the combined efforts of the entire colony to survive and reproduce.
  • a colony typically comprises a single queen, thousands of workers, and hundreds of drones during late spring and summer. Thus, a bee colony is a “population of honeybees.”
  • a “honeybee colony” peaks from late spring to summer and reaches a low point in winter.
  • the social structure of the colony is maintained by the queen and workers and depends on an effective system of communication.
  • domesticated honeybees are cultivated in “beehives” or “honeybee hives.”
  • beehive or “honeybee hive” refers to a structure that functions as a habitation for a colony of bees, e.g., a colony of honeybees.
  • honey bee is any bee that is a member of the genus Apis. Two species of honey bees, A. mellifer, A. cernan indica, A. andrreniformis, A. florea, A. koschevnikovie, and A. nigrocincta are examples of such.
  • the term “nurse bee” as used herein intends are the bees that feed the worker larvae worker jelly that is secreted from glands that produce royal jelly.
  • brood intends the three developmental stages in bees, which are collectively known as brood. Bees begin in eggs, which hatch to become larvae (plural). The larvae is legless and is specialized to eat.
  • prophylactic or “vaccine” refers to an agent that acts to prevent a disease e.g., a honeybee disease, such as e.g., Foulbrood caused by the bacterium Paenibacillus larvae in the brood.
  • a honeybee disease such as e.g., Foulbrood caused by the bacterium Paenibacillus larvae in the brood.
  • the term “vaccinate” as used herein, refers to means for producing immunity against a disease e.g., producing immunity to Paenibacillus larvae so as to prevent a disease or condition from occurring (prophylactic treatment) or inhibiting the disease from spreading (slowing or arresting its development) in the brood, larvae, progeny or colony.
  • treatment intends to raise an immune response in the queen which is then passed on to her progeny.
  • the term “raise an immune response” intends that the treatment the vaccine or treatment produces the non-disease causing antigens in the ovaries of the honey bee queen or to the developing eggs.
  • An “antigen/unif ’ intends the number of cells or antigenic fragments of the non- disease pathogen.
  • dose intends the amount provided to the queen bee, the worker bee, nurse bee or larvae in one feeding or unit amount of food.
  • composition prepared by or isolated from these methods are provided herein as well as use thereof in preventive and therapeutic methods.
  • a method to isolate a honey bee antigen that could be useful as a vaccine, the method comprising, or consisting essentially of, or yet further consisting of, contacting purified or recombinant vitellogenin (rVg) with a solid support under conditions to form rVg bound to the solid support; contacting a solubilized inactivated honey bee pathogen with the rVg bound to the solid support; and then eluting the inactivated honey bee pathogens from the solid support to isolate the honey bee antigen.
  • rVg purified or recombinant vitellogenin
  • antigen intends complete or full length proteins and fragments thereof. As is apparent to the skilled artisan wash steps to remove unbound rVg and antigen can be added to the method.
  • the solid support is selected from a chromatographic support, a sepharose CL column (Pharmacia), or an ion exchange column. These are commercially available from BioRad, , Pall Corp., Thermofischer and Pharmacia.
  • the solid support can be a Sepharose CL column or other chromatographic support using cyanogen bromide or other covalent linking mechanism known to those skilled in the art.
  • rVg may be bound to an ion exchange (IEX) column at favourable conditions (optimal pH, salt). These methods are known in the art and will vary with the rVg and the solid support.
  • solubilized protein mixtures of these pathogens are applied to the solid phase or antigens or fragment thereof with rVg, and proteins that bind to rVg are eluted using an appropriate method depending on the antigen and column.
  • a salt gradient and/or pH gradient is used to elute the proteins bound to the rVg.
  • the rVg is covalently bound to the column and the proteins of interest are eluted.
  • the rVg can be ionically bound to ion exchange chromatography gels (IEX gels), the rVg- protein mixture is eluted depending on binding characteristics, ionic strength, etc. Purified proteins or protein-rVg complexes are then identified using standard methods such as electrophoresis, high performance liquid chromatography (HPLC), mass spectrometry (MS), and/or protein sequencing, etc.
  • HPLC high performance liquid chromatography
  • MS mass spectrometry
  • the method can further comprise, or consist essentially of, or yet further consist of, determining the identity of the isolated inactivated honey bee pathogens. This can be accomplished by sequencing the antigens using well- known techniques that will determine the antigen and the polynucleotide encoding the antigen.
  • the isolated antigen is expressed in a recombinant cell system for expression and/or multiplication of the antigen.
  • Also provided herein is a method to isolate a honey bee antigen that could be useful as a vaccine, the method comprising, or consisting essentially of, or yet further consisting of, binding an anti-His tag or anti-Vg antibody with a solubilized inactivated honey bee pathogen and detecting any antibody bound to the inactivated honey bee pathogen, wherein the antibody or inactivated honey bee pathogen is detectably labeled.
  • the antibody or inactivated honey bee pathogen is detectably labeled.
  • labels are known in the art and described herein.
  • Anti-His tag antibodies are commercially available from Cell Signaling Technology, Invitrogen, ThermoFisher, and other commercial vendors.
  • Anti-Vg antibodies are made using purified or rVg or fragments thereof using conventional techniques well-known in the art.
  • the rVg for use in the methods can be expressed in a prokaryotic or eukaryotic cell system, e.g., baculovirus, CHO, HEK, E. coli. Saccharomyces. and others known in the art.
  • recombinant vitellogenin (rVg) is produced in an insect viral expression system, such as a baculovirus expression system.
  • the baculovirus expression system is the DHIOBac system.
  • Vg protein or fragment can be used in the method.
  • One non-limiting example is the rVg having the amino acid sequence of SEQ ID NO: 1, or a fragment or an equivalent thereof, with and without the His tag.
  • the polynucleotide encoding SEQ ID NO: 1 is provided in SEQ ID NO: 2.
  • detectable labels can be used, e.g., a radioisotope labels or a fluorescent label or, in the case of an enzymatic label, it may catalyze chemical alteration of a substrate compound or composition which is detectable.
  • suitable labels include, but are not limited to radioisotopes, fluorochromes, chemiluminescent compounds, dyes, and proteins, including enzymes.
  • the label may be simply detected or it may be quantified.
  • a response that is simply detected generally comprises a response whose existence merely is confirmed, whereas a response that is quantified generally comprises a response having a quantifiable (e.g., numerically reportable) value such as an intensity, polarization, and/or other property.
  • the detectable response can be generated directly using a luminophore or fluorophore associated with an assay component actually involved in binding, or indirectly using a luminophore or fluorophore associated with another (e.g., reporter or indicator) component.
  • honey bee pathogens antigens and fragments thereof
  • the solid support or antigen or fragment thereof Prior to contacting the honey bee pathogens (antigens and fragments thereof) with the solid support or antigen or fragment thereof, they are inactivated and then solubilized so that internal and trans-membrane proteins are in solution. Suitable methods for inactivation include for example, sonication, followed by solubilization in non-ionic detergents such as Brij, Triton, octyl-glucoside and others known in the art.
  • the antigen contacted with the solid support can comprise one or multiple types of antigens, e.g., from the same or different pathogen.
  • Non-limiting examples of honey bee antigens for use in the method are those responsible for American foulbrood, European foulbrood, deformed wing virus, Varroa mite infestations and other viral, bacterial and parasitic diseases of honey bees.
  • Examples of such include, without limitation, Paenibacillus larvae, Paenibacillus alvei, Paenibacillus dendritiformis Paenibacillus amylolyticus, Paenibacillus campinasensis, Paenibacillus chondr oitinus, Paenibacillus chungangensis, Paenibacillus doosanensis, Paenibacillus glucanolyticus, Paenibacillus humicus, Paenibacillus lactis, Paenibacillus lautus, Paenibacillus lentimorbus, Paenibacillus maceran, Paenibacillus macerans-like, Paenibacillus macquariensis, Paenibacillus motobuensis, Paenibacillus pabuli, Paenibacillus phoenicis, Paenibacillus polymyxa, Paenibacillus popilliae
  • the methods can further comprise, or consist essentially of, or yet further consist of, determining the identity of the isolated inactivated honey bee pathogens. This can be accomplished by sequencing the antigens using well-known techniques that will determine the antigen and the polynucleotide encoding the antigen.
  • the isolated antigen is expressed in a recombinant cell system for expression and/or multiplication of the antigen.
  • rVg is used in the place of antibodies to detect protein in high throughput methods such as phage display, high throughput screening of the entire pathogen genome, and other methods known in the art. Labeled rVg or an anti-His tag antibodyis used to detect the proteins of interest, and then they may be further characterized. This method allows for the rapid screening of multiple disease agents extremely quickly.
  • vaccines and bacterins identified by these methods. After the proteins of interest are identified, they are partially sequenced, and the genes expressing such proteins are isolated, and the proteins are produced in a suitable eukaryotic or prokaryotic expression system (baculovirus, CHO, HEK, E. coli, Saccharomyces, and others known in the art).
  • the recombinant proteins can be added to feed for queen bees as is described in U.S. Patent No. 10,994,001, or may be introduced directly into hives in a suitable feed substance. Immunity in honey bees and larvae is generated and assessed as described in U.S. Patent No. 10,994,001.
  • Toll-like receptor (TLR) binding CpGs may be added to the vaccines or bacterins to enhance effector mechanisms.
  • compositions comprising, or consisting essentially of, or yet further consisting of, the immunogenic proteins and a carrier.
  • the composition comprises, or consisting essentially of, or yet further consisting of, the proteins or immunogenic fragments thereof and a carrier.
  • a carrier can be a solid or a liquid carrier and can include preservatives, insect nutrients, or other coloring agents as necessary.
  • the carrier is an insect food, such as a queen bee wafer or sugar feed.
  • compositions of the present invention are easily administered or fed to insects.
  • the edible composition may optionally comprise one or more acceptable (e.g. pharmaceu-tically acceptable) agents selected from the group consisting of carrier(s) (e.g.
  • composition consists of only bacteria, fungi, fragments thereof and/or spores thereof and water.
  • the compositions or artificial diet may be used for example in solid, semisolid or liquid form, such as in the form of patties, syrups, drenches, dustings, pastes, tablets, pellets, capsules, solutions, emulsions, suspensions or like.
  • the composition is for oral administration.
  • the term "prevent” or “preventing” refers to feeding or administration of microbes to an insect for purposes which include not only 100% or complete prevention but also partial prophylaxis and therefore also amelioration or alleviation of disorders or symptoms related to microbial infections. Preventive effect may be assessed e.g. by monitoring the symptoms mentioned in any of the tables of Figures 6-9 of U.S. Patent No. 10,994,001, incorporated herein by reference. In this respect, the present disclosure can provide any amount of increase e.g. in the survival data compared to untreated controls.
  • the methods further comprise feeding a honey bee queen or worker bee an artificial insect diet comprising antigen or fragment thereof isolated by the above methods.
  • the compositions are useful as vaccines.
  • the amount of antigen in the artificial diet is 1 to 20% by weight.
  • the artificial diet can be administered to the queen beefor administration one, two or three times a year.
  • the honey bee queen prior is feed the artificial diet prior to egg laying. After consumption of the diet, the queen bee and/or the hive is challenged to determine protective or therapeutic response, as noted herein and U.S. Patent No. 10,994,001, incorporated herein by reference.
  • Also provided herein is a method selected from: treating, raising an immune response or immunizing a honeybee larvae, honeybee, honeybee hatched from a treated queen bee, honeybee population or honeybee hive against AFB, the method comprising, or alternatively consisting essentially of, or yet further consisting essentially of, administering an effective amount of the composition as described herein to a nurse bees or the honeybee queen or honey bee larvae thereby treating, raising an immune response, or immunizing the queen bee and the honeybee larvae produced by the queen bee.
  • Sf9 cells were grown in Sf-900 II SFM Expression Medium (Life Technologies, Catalog Number: 10902-088). The cells were maintained in Erlenmeyer Flasks at 27 °C in an orbital shaker. One day before transfection, the cells were seeded at an appropriate density in 6 wells CORNING-COSTAR. On the day of transfection, DNA and Transfection Reagent (PROMEGA, Catalog Number:E2691) were mixed at an optimal ratio and then added into the plate with cells ready for transfection. Cells were incubated in Sf-900 II SFM for 5-7 days at 27 °C before harvest. The supernatant was collected after centrifugation and designated as Pl viral stock. P2 was amplified for later infection. The expression was analyzed by Western blot.
  • the anti-His antibody used was a monoclonal Mouse-anti-His antibody (GenScript, Cat.No.A00186).
  • Paenibacillus larvae strain was isolated from NY state bee hive. The strain has been tested found to be a pure strain of Paenibacillus larvae strain (99.4% similarity to Paenibacillus larvae strain DSM 7030) and found to be the ERIC I Pathotype.
  • P. larvae strain was grown on enriched Mueller-Hinton MYPGP agar medium, for 2-6 days, harvested and then snap-frozen at -80°C and shipped for protein analysis on dry ice, with a temperature control monitor, to avoid any proteolysis.
  • the four cell pellet was lysed in 225 ⁇ l of of Osmotic Lysis Buffer containing protease inhibitor stock, nuclease stock, and phosphatase inhibitor stocks (I and II), 75 pl SDS Boiling Buffer without ⁇ -mercaptoethanol, and 100 mg of washed glass beads (Sigma G9268, mesh size 425-6000 microns) as described by lazwinski (SDS-polyacrylamide gel electrophoresis. Trends in Biochemical Science, 25, 590-592. .Jazwinski. S.M. (1990)).
  • the sample was vortexed 2 minutes, sonicated 15 minutes, vortexed again for 2 minutes, and placed in a dry bath at 95°C for 10 minutes.
  • the sample was centrifuged, and the supernatants was combined into one tube.
  • the protein concentration (4.72 ⁇ g/ ⁇ l) was then determined using the BCA assay (Smith et. al. Anal. Biochem. 150: 76-85, 1985, and Pierce Chemical Co., Rockford, IL).
  • the sample was then lyophilized, and redissolved to 2.66 mg/ml in 1 : 1 diluted SDS Boiling Buffer: Urea Sample Buffer before loading (see 2D gels below).
  • Isoelectric focusing was carried out in a glass tube of inner diameter 3.3 mm using 2.0% 4-8 mix (Serva, Heidelberg, Germany) for 20,000 volt- hrs.
  • One pg of an IEF internal standard, tropomyosin, was added to each sample. This protein migrates as a doublet with lower polypeptide spot of MW 33,000 and pl 5.2; an arrow on the stained gels marks its position.
  • the enclosed tube gel pH gradient plot for this set of Servalytes was determined with a surface pH electrode.
  • each tube gel was sealed to the top of a stacking gel that overlaid a 10% acrylamide slab gel (1.0 mm thick).
  • SDS slab gel electrophoresis was carried out for about 5 hrs at 25 mA/gel.
  • the following proteins (Millipore Sigma) were used as molecular weight standards: myosin (220,000), phosphorylase A (94,000), catalase (60,000), actin (43,000), carbonic anhydrase (29,000), and lysozyme (14,000). These standards appear as bands at the basic edge of the Coomassie Brilliant Blue R-250 stained 10% acrylamide slab gel. The gel was dried between sheets of cellophane paper with the acid edge to the left.
  • the blot was then placed in tertiary antibody (anti-Mouse IgG-HRP [GE, Cat# NA931V, Lot# 17205275] diluted 1 :2,000 in SBT) for two hours, rinsed as above, treated with ECL, and exposed to x-ray
  • tertiary antibody anti-Mouse IgG-HRP [GE, Cat# NA931V, Lot# 17205275] diluted 1 :2,000 in SBT
  • the blot was blocked for two hours in Superblock (Thermofi scher) containing 0.05M Tris buffer, pH 6.8 (SBT). The blot was then incubated overnight in primary solution (Vitellogenin (GenScript, Lot # U5329GB010-3/P4GE001)) diluted to 0.1 ⁇ g/ml in SBT) and rinsed 3 x 10 minutes in TTBS. The blot was then placed in secondary solution (THE His Tag [GenScript, Cat# A00186-100, Lot# 17K001367] diluted to 0.02 ⁇ g/ml in SBT) for two hours and rinsed 3 x 10 minutes in TTBS.
  • TCE His Tag [GenScript, Cat# A00186-100, Lot# 17K001367] diluted to 0.02 ⁇ g/ml in SBT
  • the blot was then placed in tertiary antibody (anti-Mouse IgG-HRP [GE, Cat# NA931V, Lot# 17205275] diluted 1 :2,000 in SBT) for two hours, rinsed as above, treated with ECL, and exposed to x-ray film.
  • tertiary antibody anti-Mouse IgG-HRP [GE, Cat# NA931V, Lot# 17205275] diluted 1 :2,000 in SBT
  • FIG. 2 shows the construct of the one dimensional electrophoresis.
  • FIG. 2 and FIG. 3 indicate the results of detection of protein binding by vitellogenin. Using simple visual overlay at least 10 spots can be detected that bind uniquely to vitellogenin. In FIG. 4, these are identified on the Coomassie stained gel.
  • Honeybee rVG binds specifically to multiple proteins detected. It is noteworthy that there are some proteins that appear in very small concentrations, show a high degree of binding (e.g. H, I, J in the 2D gels above). These tended to be lower MW proteins between 14 and 29 MW. There also appears to be a stronger signal at ⁇ 60 and 94 MW and between 94 and 220 MW.
  • Known virulence factors for P. larvae include enolase (MW 82-200 kDa, depending on isoform), and a chitinase (49 kDa).
  • enolase MW 82-200 kDa, depending on isoform
  • chitinase 49 kDa
  • C3 mART C3 mART which has a MW between 20 and 25 kDa. Further analysis of the proteins bound by vitellogenin will identify those proteins that are involved in immunity.
  • SEQ ID NO: 2 DNA Sequence Encoding Vg (Signal Peptide is underlined and is an optional element for the DNA Sequence).

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Zoology (AREA)
  • Insects & Arthropods (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Toxicology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Analytical Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Oncology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Communicable Diseases (AREA)
  • Peptides Or Proteins (AREA)

Abstract

Provided herein are novel solutions for the identification and measurement of protective proteins to treat and prevent honey bee infections.

Description

USE OF VITELLOGENIN FOR DEFINING AND TESTING
NOVEL IMMUNOGENS IN INSECTS
CROSS-REFERENCE TO RELATED APPLICATIONS
[1] This application claims priority under 35 U.S.C. §119(e) to U.S. provisional application Serial No. 63/074,365, filed September 3, 2020, the content of which is incorporated herein by reference in its entirety.
BACKGROUND
[2] Vitellogenin (VTG or less popularly known as VG) is a precursor protein of egg yolk normally in the blood or hemolymph only of females that is used as a biomarker in vertebrates of exposure to environmental estrogens which stimulate elevated levels in males as well as females. The protein product of the gene is classified as a glycolipoprotein, having properties of a sugar, fat and protein. It is the precursor of the lipoproteins and phosphoproteins that make up most of the protein content of yolk.
[3] Vitellogenin has been shown to bind to various proteins and transport them to ovaries in queen bees, where they are then passed on through eggs to newly hatched larvae. These proteins are capable of activating the non-specific insect immune mechanisms in insects, with the net result of a protective effect against pathogens, such as American foulbrood (AFB). American foulbrood (AFB) is a fatal bacterial disease of honey bee brood caused by the spore forming bacterium Paenibacillus larvae (see htps://beeaware.org.aU/archive-pest/american-foulbrood/#ad-image-0). It is not a stress related disease and can infect the strongest to the weakest colony in an apiary. Infected brood usually die at the pre-pupal or pupal stage. The disease is unable to be cured, meaning that destruction of infected colonies and hives or irradiation of infected material is the only current means to manage AFB. Thus a need exists in the art for therapeutic and preventive intervention. This disclosure satisfies this need and provides related advantages as well. SUMMARY OF THE DISCLOSURE
[4] Provided herein are novel solutions for the identification and measurement of these protective proteins to treat and prevent AFB.
[5] To that end, this disclosure provides the methods and compositions for the identification, isolation, preparation and use of these protective proteins and fragments thereof.
[6] In one aspect, the protection-inducing proteins an immunogenic fragments thereof from honey bee pathogens.
[7] In one embodiment, provided herein is a method for identification of proteins. In one aspect, recombinant vitellogenin (rVg) is produced in an insect viral expression system, such as a baculovirus expression system. The rVg is bound to a sepharose CL column or other chromatographic support using cyanogen bromide or other covalent linking mechanism known to those skilled in the art. Alternatively, rVg may be bound to an ion exchange (IEX) column at favourable conditions (optimal pH, salt). Honey bee pathogens are inactivated and then solubilized so that internal and trans-membrane proteins are in solution. Suitable methods for inactivation include for example, sonication, followed by solubilization in non-ionic detergents such as Brij, Triton, octyl-glucoside and others known in the art. Honey bee pathogens are those responsible for American foulbrood, European foulbrood, deformed wing virus, varroa mite infestations and other viral, bacterial and parasitic diseases of honey bees. Solubilized protein mixtures of these pathogens are applied to the solid phase with rVg, and proteins that bind to rVg are eluted using a salt gradient and/or pH gradient. The rVg is covalently bound to the column and the proteins of interest are eluted. The rVg can be ionically bound to IEX gels, the rVg-protein mixture is eluted depending on binding characteristics, ionic strength, etc. Purified proteins or protein- rVg complexes are then identified using standard methods such as electrophoresis, HPLC, MS, etc.
[8] In another aspect, provided herein is a proteomics method for the identification of antigenic proteins. In this method, rVg is used in the place of antibodies to detect protein in high throughput methods such as phage display, high throughput screening of the entire pathogen genome, and other methods known in the art. Labeled rVg or a polyclonal antibody is used to detect the proteins of interest, and then they may be further characterized. This method allows for the rapid screening of multiple disease agents extremely quickly.
[9] In a yet further aspect, provided herein are vaccines and bacterins identified by these methods. After the proteins of interest are identified, they are partially sequenced, and the genes expressing such proteins are isolated, and the proteins are produced in a suitable eukaryotic or prokaryotic expression system (baculovirus, CHO, HEK, E. coll. Saccharomyces. and others known in the art). The recombinant proteins can be added to feed for queen bees as is described in U.S. Patent No. 10,994,001, or may be introduced directly into hives in a suitable feed substance. Immunity in honey bees and larvae is generated and assessed as described in U.S. Patent No. 10,994,001. Toll-like receptor (TLR) binding CpGs may be added to the vaccines or bacterins to enhance effector mechanisms.
[10] Further provided is a novel test system for detecting and quantitating immunogens. Standard methods described and approved universally by regulatory bodies are indirect ELISA methods. In this method, 2 antibodies are used - one to trap the protein(s) of interest to a solid phase, and the second (usually a monoclonal) to detect the protein(s) of interest. The secondary antibody may be labeled with a fluorescent or enzyme label, and the specific binding quantified and compared to a standard. This novel method describes the use of rVg to detect immunogenic protein(s) of interest rather than a secondary antibody. The advantage of this method is that it would be detecting known immunogenic proteins, peptides, lids and polysaccharides rather than surrogate molecules that may or may not correlate with immunity.
BRIEF DESCRIPTION OF THE DRAWINGS
[11] FIG. 1 shows a one dimensional gel using the below methods. 2 different blocking agents were assessed. Superblock was chosen as it appeared to provide a clearer image with less background staining. From Left to right: Vitellogenin blots with PF block, Vitellogenin blot with Superblock, Detection antibodies alone (i.e. anti-His tag and anti- mouse antibodies) and Coomassie stained (protein) gel.
[12] FIG. 2 shows a two dimensional gel. Methods are described below. 2D pattern following staining with detection antibodies only (no vitellogenin). [13] FIG. 3 shows a 2D gel pattern following staining with vitellogenin and detection antibodies. The circled spots are those that were detected uniquely by vitellogenin, i.e. did not bind to either of the detection antibodies.
[14] FIG. 4 is a Coomassie stained gel indicating the proteins detected by specific vitellogenin staining.
[15] FIG. 5 shows a map of an exemplary recombinant vitellogenin construct.
DETAILED DESCRIPTION
Definitions
[16] As used herein and in the appended claims, singular articles such as "a" and "an" and "the" and similar referents in the context of describing the elements are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context.
[17] As used herein, “about” is understood by persons of ordinary skill in the art and may vary to some extent depending upon the context in which it is used. If there are uses of the term which are not clear to persons of ordinary skill in the art given the context in which the term "about" is used, "about" will mean up to plus or minus 10% of the particular term.
[18] The term "exemplary" as used herein refers to "serving as an example, instance, or illustration," and not "preferred" or "advantageous over other embodiments."
[19] Unless defined otherwise, technical and scientific terms used herein have the same meaning as commonly understood by a person of ordinary skill in the art. In particular, this disclosure utilizes routine techniques in the field of honeybee husbandry.
[20] All numerical designations, e.g., pH, temperature, time, concentration, and molecular weight, including ranges, are approximations which are varied ( + ) or ( - ) by increments of 0.1 or 1 where appropriate. It is to be understood, although not always explicitly stated that all numerical designations are preceded by the term “about”. The term “about” also includes the exact value “X” in addition to minor increments of “X” such as “X + 0.1 or 1” or “X - 0.1 or 1,” where appropriate. It also is to be understood, although not always explicitly stated, that the reagents described herein are merely exemplary and that equivalents of such are known in the art.
[21] As will be understood by one skilled in the art, for any and all purposes, all ranges disclosed herein also encompass any and all possible subranges and combinations of subranges thereof. Furthermore, as will be understood by one skilled in the art, a range includes each individual member.
[22] As used in the specification and claims, the singular form “a”, “an” and “the” include plural references unless the context clearly dictates otherwise. For example, the term “a cell” includes a plurality of cells, including mixtures thereof.
[23] As used herein, the term “comprising” is intended to mean that the compositions and methods include the recited elements, but not excluding others. “Consisting essentially of’ when used to define compositions and methods, shall mean excluding other elements of any essential significance to the combination for the stated purpose. Thus, a composition consisting essentially of the elements as defined herein would not exclude trace contaminants from the isolation and purification method and pharmaceutically acceptable carriers, such as phosphate buffered saline, preservatives and the like. “Consisting of’ shall mean excluding more than trace elements of other ingredients and substantial method steps for administering the compositions of this invention or process steps to produce a composition or achieve an intended result. Embodiments defined by each of these transition terms are within the scope of this invention.
[24] The term “isolated” as used herein with respect to cells, nucleic acids, such as DNA or RNA, refers to molecules separated from other DNAs or RNAs, respectively, that are present in the natural source of the macromolecule. The term “isolated” as used herein also refers to a nucleic acid or peptide that is substantially free of cellular material, viral material, or culture medium when produced by recombinant DNA techniques, or chemical precursors or other chemicals when chemically synthesized. Moreover, an “isolated nucleic acid” is meant to include nucleic acid fragments which are not naturally occurring as fragments and would not be found in the natural state. The term “isolated” is also used herein to refer to cells or polypeptides which are isolated from other cellular proteins or tissues. Isolated polypeptides is meant to encompass both purified and recombinant polypeptides.
[25] “ Cells,” “host cells” or “recombinant host cells” are terms used interchangeably herein. It is understood that such terms refer not only to the particular subject cell but to the progeny or potential progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term as used herein. Host cells can be prokaryotic or eukaryotic.
[26] “Prokaryotic cells” that usually lack a nucleus or any other membrane-bound organelles and are divided into two domains, bacteria and archaea. In addition to chromosomal DNA, these cells can also contain genetic information in a circular loop called an episome. Bacterial cells are very small, roughly the size of an animal mitochondrion (about 1-2 pm in diameter and 10 pm long). Prokaryotic cells feature three major shapes: rod shaped, spherical, and spiral. Instead of going through elaborate replication processes like eukaryotes, bacterial cells divide by binary fission. Examples include but are not limited to Bacillus bacteria, E. coli bacterium, and Salmonella bacterium.
[27] “Eukaryotic cells” comprise all of the life kingdoms except monera. They can be easily distinguished through a membrane-bound nucleus. Animals, plants, fungi, and protists are eukaryotes or organisms whose cells are organized into complex structures by internal membranes and a cytoskeleton. The most characteristic membrane-bound structure is the nucleus. Unless specifically recited, the term “host” includes a eukaryotic host, including, for example, yeast, higher plant, insect and mammalian cells. Non-limiting examples of eukaryotic cells or hosts include simian, bovine, porcine, murine, rat, avian, reptilian and human, e.g., HEK293 cells, Chinese Hamster Ovary (CHO) cells, CHO-S cells, CHO-K1 cells, 293T cells, HeLa cells, Baby hamster kidney (BHK) cells, and Sf9 cells. Sf9 cells are a clonal isolate of Spodopter frugiperda. They were originally isolated from ovarian tissue and are commonly used in insect cell culture for recombinant protein production using baculovirus. These cells are commercially available from American Type Culture Collection (ATCC) under CRL-1711.
[28] DHIOBac can serve as a host for a recombinant pFastBac vector containing a cloned gene of interest. ThermoFisher Scientific provides these cells as MAX Efficiency DHIOBac Competent Cells. These DHIOBac cells harbor a baculovirus shuttle vector (bMON14272) and a helper plasmid (pMON7142), and are capable of supporting site-specific recombination between pFastBac and bMON14272 to generate high molecular weight bacmids that can then be amplified, purified, and used for insect cell transfection and subsequent gene expression. Kanamycin resistance for bacmid selection and maintenance is conferred by bMON14272, and tetracycline resistance by pMON7124. These cells are commercially available from ThermoFisher Scientific (htps://www.thermofisher.com/order/catalog/product/10361012#/10361012, last accessed on August 28, 2021).
[29] A baculovirus-insect expression system utilizes recombinant baculovirus (insect viruses) and their ability to manufacture high yields of biologically active proteins from insect cells. These systems are known in the art and commercially available. See, e.g., Jarvis, Baculovirus-insect cell expression systems, Methods Enzymol. (2009)463: 191-222; Scholz, J., Suppmann, S., A new single-step protocol for rapid baculovirus-driven protein production in insect cells. BMC Biotechnol. Vol. 17, 83 (2017), https://doi.org/10.1186/s12896-017-0400-3; Lemaitre, R.P., et al., FlexiBAC: a versatile, open-source baculovirus vector system for protein expression, secretion, and proteolytic processing, BMC Biotechnol, Vol. 19, 20 (2019); https://doi.org/10.1186/sl2896-019-0512- z; www.sinobiological.com/services/baculovirus-insect-protein-expression- service?gclid=Cj0KCQjwvaeJBhCvARIsABgTDM6Yn3HdX7g7D1c5kmbFTkQWxHTLO nqrQ4ww,zvXsfYH1DGNa_lr665AaAos7EALw_wcB and expressionsystems.com/product/insect-ceIls, each last accessed on August 28, 2021.
[30] "ELISA" (enzyme-linked immunosorbent assay) is a plate-based assay technique designed for detecting and quantifying soluble substances such as peptides, proteins, antibodies, and hormones. Other names, such as enzyme immunoassay (EIA), are also used to describe the same technology. In the most simple form of an ELISA, antigens, e.g., antigens from the sample to be tested are attached to a surface. Then, a matching antibody is applied over the surface so it can bind the antigen. This antibody is linked to an enzyme and then any unbound antibodies are removed. In the final step, a substance containing the enzyme's substrate is added. If there was binding the subsequent reaction produces a detectable signal, most commonly a color change. Performing an ELISA involves at least one antibody with specificity for a particular antigen. The sample with an unknown amount of antigen is immobilized on a solid support (e.g., a polystyrene microtiter plate) either non- specifically (via adsorption to the surface) or specifically (via capture by another antibody specific to the same antigen, in a "sandwich" ELISA). After the antigen is immobilized, the detection antibody is added, forming a complex with the antigen. The detection antibody can be covalently linked to an enzyme or label or can itself be detected by a secondary antibody that is linked to an enzyme through bioconjugation. Between each step, the plate is typically washed with a mild detergent solution to remove any proteins or antibodies that are non-specifically bound. After the final wash step, the plate is developed by adding an enzymatic substrate to produce a signal, which indicates the quantity of antigen in the sample.
[31] ELISA can also be performed in other forms of ligand binding assays. In one aspect, the technique essentially requires any ligating reagent that can be immobilized on the solid phase along with a detection reagent that will bind specifically and use an enzyme to generate a signal that can be properly quantified. In between the washes, only the ligand and its specific binding counterparts remain specifically bound or "immimosorbed" by antigen- antibody interactions or other interactions (protein-protein, e.g., Vitellogenin binding to a bee pathogen to the solid phase, while the nonspecific or unbound components are washed away.
[32] “Amplify” “amplifying” or “amplification” of a polynucleotide sequence includes methods such as traditional cloning methodologies, PCR, ligation amplification (or ligase chain reaction, LCR) or other amplification methods. These methods are known and practiced in the art. See, e.g., U.S. Patent Nos. 4,683,195 and 4,683,202 and Innis et al. (1990) Mol. Cell Biol. 10(11):5977-5982 (for PCR); and Wu et al. (1989) Genomics 4:560- 569 (for LCR). In general, the PCR procedure describes a method of gene amplification which is comprised of (i) sequence-specific hybridization of primers to specific genes within a DNA sample (or library), (ii) subsequent amplification involving multiple rounds of annealing, elongation, and denaturation using a DNA polymerase, and (iii) screening the PCR products for a band of the correct size. The primers used are oligonucleotides of sufficient length and appropriate sequence to provide initiation of polymerization, i.e. each primer is specifically designed to be complementary to each strand of the genomic locus to be amplified.
[33] Reagents and hardware for conducting PCR are commercially available. Primers useful to amplify sequences from a particular region are preferably complementary to, and hybridize specifically to sequences in the target region or in its flanking regions. Nucleic acid sequences generated by amplification may be sequenced directly. Alternatively the amplified sequence(s) may be cloned prior to sequence analysis. A method for the direct cloning and sequence analysis of enzymatically amplified genomic segments is known in the art. [34] The term “genotype” refers to the specific allelic composition of an entire cell, a certain gene or a specific polynucleotide region of a genome, whereas the term “phenotype’ refers to the detectable outward manifestations of a specific genotype.
[35] As used herein, the term “gene” or “recombinant gene” refers to a nucleic acid molecule comprising an open reading frame and including at least one exon and (optionally) an intron sequence. A gene may also refer to a polymorphic or a mutant form or allele of a gene.
[36] “Homology” or “identity” or “similarity” refers to sequence similarity between two peptides or between two nucleic acid molecules. Homology can be determined by comparing a position in each sequence which may be aligned for purposes of comparison. When a position in the compared sequence is occupied by the same base or amino acid, then the molecules are homologous at that position. A degree of homology between sequences is a function of the number of matching or homologous positions shared by the sequences. An “unrelated” or “non-homologous” sequence shares less than 40% identity, though preferably less than 25% identity, with one of the sequences of the present invention.
[37] A polynucleotide or polynucleotide region (or a polypeptide or polypeptide region) has a certain percentage (for example, 60 %, 65 %, 70 %, 75 %, 80 %, 85 %, 90 %, 95 %, 98 % or 99 %) of “sequence identity” to another sequence means that, when aligned, that percentage of bases (or amino acids) are the same in comparing the two sequences. This alignment and the percent homology or sequence identity can be determined using software programs known in the art, for example those described in Ausubel et al. eds. (2007) Current Protocols in Molecular Biology. Preferably, default parameters are used for alignment. One alignment program is BLAST, using default parameters. In particular, programs are BLASTN and BLASTP, using the following default parameters: Genetic code = standard; filter = none; strand = both; cutoff = 60; expect = 10; Matrix = BLOSUM62; Descriptions = 50 sequences; sort by = HIGH SCORE; Databases = non-redundant, GenBank + EMBL + DDBJ + PDB + GenBank CDS translations + SwissProtein + SPupdate + PIR. Details of these programs can be found at the following Internet address: http://www.ncbi.nlm.nih.gov/blast/Blast.cgi, last accessed on May 21, 2008. Biologically equivalent polynucleotides are those having the specified percent homology and encoding a polypeptide having the same or similar biological activity. [38] The term “an equivalent nucleic acid” refers to a nucleic acid having a nucleotide sequence having a certain degree of homology with the nucleotide sequence of the nucleic acid or complement thereof. A homolog of a double stranded nucleic acid is intended to include nucleic acids having a nucleotide sequence which has a certain degree of homology with or with the complement thereof. In one aspect, homologs of nucleic acids are capable of hybridizing to the nucleic acid or complement thereof.
[39] The term “contacting” or interact” as used herein is meant to include detectable interactions between molecules, such as can be detected using, for example, a hybridization assay, an antibody binding assay such as ELIA. As such, rhe term interact or contacting is also meant to include “binding” interactions between molecules. Interactions may be, for example, protein-protein, protein-nucleic acid, or nucleic acid-nucleic acid in nature.
[40] “Hybridization” refers to a reaction in which one or more polynucleotides react to form a hybridization complex that is stabilized via hydrogen bonding between the bases of the nucleotide residues. The hydrogen bonding may occur by Watson-Crick base pairing, Hoogstein binding, or in any other sequence-specific manner. The complex may comprise two strands forming a duplex structure, three or more strands forming a multi-stranded complex, a single self-hybridizing strand, or any combination of these. A hybridization reaction may constitute a step in a more extensive process, such as the initiation of a PCR reaction, or the enzymatic cleavage of a polynucleotide by a ribozyme.
[41] Hybridization reactions can be performed under conditions of different “stringency”. In general, a low stringency hybridization reaction is carried out at about 40°C in about 10 x SSC or a solution of equivalent ionic strength/temperature. A moderate stringency hybridization is typically performed at about 50°C in about 6 x SSC, and a high stringency hybridization reaction is generally performed at about 60°C in about 1 x SSC.
Hybridization reactions can also be performed under “physiological conditions” which is well known to one of skill in the art. A non-limiting example of a physiological condition is the temperature, ionic strength, pH and concentration of Mg2+ normally found in a cell.
[42] When hybridization occurs in an antiparallel configuration between two single-stranded polynucleotides, the reaction is called “annealing” and those polynucleotides are described as “complementary”. A double-stranded polynucleotide can be “complementary” or “homologous” to another polynucleotide, if hybridization can occur between one of the strands of the first polynucleotide and the second. “Complementarity” or “homology” (the degree that one polynucleotide is complementary with another) is quantifiable in terms of the proportion of bases in opposing strands that are expected to form hydrogen bonding with each other, according to generally accepted base-pairing rules.
[43] The term “mismatches” refers to hybridized nucleic acid duplexes which are not 100% homologous. The lack of total homology may be due to deletions, insertions, inversions, substitutions or frameshift mutations.
[44] As used herein, the term “oligonucleotide” refers to polynucleotides such as deoxyribonucleic acid (DNA), and, where appropriate, ribonucleic acid (RNA). The term should also be understood to include, as equivalents, derivatives, variants and analogs of either RNA or DNA made from nucleotide analogs, and, as applicable to the embodiment being described, single (sense or antisense) and double-stranded polynucleotides. Deoxyribonucleotides include deoxyadenosine, deoxycytidine, deoxyguanosine, and deoxythymidine. For purposes of clarity, when referring herein to a nucleotide of a nucleic acid, which can be DNA or an RNA, the terms “adenosine”, “cytidine”, “guanosine”, and “thymidine” are used. It is understood that if the nucleic acid is RNA, a nucleotide having a uracil base is uridine.
[45] The terms “polynucleotide” and “oligonucleotide” are used interchangeably and refer to a polymeric form of nucleotides of any length, either deoxyribonucleotides or ribonucleotides or analogs thereof. Polynucleotides can have any three-dimensional structure and may perform any function, known or unknown. The following are non-limiting examples of polynucleotides: a gene or gene fragment (for example, a probe, primer, EST or SAGE tag), exons, introns, messenger RNA (mRNA), transfer RNA, ribosomal RNA, ribozymes, cDNA, dsRNA, siRNA, miRNA, recombinant polynucleotides, branched polynucleotides, plasmids, vectors, isolated DNA of any sequence, isolated RNA of any sequence, nucleic acid probes and primers. A polynucleotide can comprise modified nucleotides, such as methylated nucleotides and nucleotide analogs. If present, modifications to the nucleotide structure can be imparted before or after assembly of the polynucleotide. The sequence of nucleotides can be interrupted by non-nucleotide components. A polynucleotide can be further modified after polymerization, such as by conjugation with a labeling component. The term also refers to both double- and single-stranded molecules. Unless otherwise specified or required, any embodiment of this invention that is a polynucleotide encompasses both the double-stranded form and each of two complementary single-stranded forms known or predicted to make up the double-stranded form.
[46] A polynucleotide is composed of a specific sequence of four nucleotide bases: adenine (A); cytosine (C); guanine (G); thymine (T); and uracil (U) for thymine when the polynucleotide is RNA. Thus, the term “polynucleotide sequence” is the alphabetical representation of a polynucleotide molecule. This alphabetical representation can be input into databases in a computer having a central processing unit and used for bioinformatics applications such as functional genomics and homology searching. The term “polymorphism” refers to the coexistence of more than one form of a gene or portion thereof. A portion of a gene of which there are at least two different forms, i.e., two different nucleotide sequences, is referred to as a “polymorphic region of a gene”. A polymorphic region can be a single nucleotide, the identity of which differs in different alleles.
[47] As used herein, the term “carrier” encompasses any of the standard carriers, such as a phosphate buffered saline solution, buffers, water, and emulsions, such as an oil/water or water/oil emulsion, and various types of wetting agents. The compositions also can include stabilizers and preservatives. For examples of carriers, stabilizers and adjuvants, see Sambrook and Russell (2001), supra. Those skilled in the art will know many other suitable carriers for binding polynucleotides, or will be able to ascertain the same by use of routine experimentation. In one aspect of the invention, the carrier is a buffered solution such as, but not limited to, a PCR buffer solution.
[48] A “gene delivery vehicle” is defined as any molecule that can carry inserted polynucleotides into a host cell. Examples of gene delivery vehicles are liposomes, biocompatible polymers, including natural polymers and synthetic polymers; lipoproteins; polypeptides; polysaccharides; lipopolysaccharides; artificial viral envelopes; metal particles; and bacteria, or viruses, such as baculovirus, adenovirus and retrovirus, bacteriophage, cosmid, plasmid, fungal vectors and other recombination vehicles typically used in the art which have been described for expression in a variety of eukaryotic and prokaryotic hosts, and may be used for gene therapy as well as for simple protein expression.
[49] “Gene delivery,” “gene transfer,” and the like as used herein, are terms referring to the introduction of an exogenous polynucleotide (sometimes referred to as a “transgene”) into a host cell, irrespective of the method used for the introduction. Such methods include a variety of well-known techniques such as vector-mediated gene transfer (by, e.g., viral infection, sometimes called transduction), transfection, transformation or various other protein-based or lipid-based gene delivery complexes) as well as techniques facilitating the delivery of “naked” polynucleotides (such as electroporation, “gene gun” delivery and various other techniques used for the introduction of polynucleotides). Unless otherwise specified, the term transfected, transduced or transformed may be used interchangeably herein to indicate the presence of exogenous polynucleotides or the expressed polypeptide therefrom in a cell. The introduced polynucleotide may be stably or transiently maintained in the host cell. Stable maintenance typically requires that the introduced polynucleotide either contains an origin of replication compatible with the host cell or integrates into a replicon of the host cell such as an extrachromosomal replicon (e.g., a plasmid) or a nuclear or mitochondrial chromosome. A number of vectors are known to be capable of mediating transfer of genes to mammalian cells, as is known in the art and described herein.
[50] The term “express” refers to the production of a gene product. In some embodiments, the gene product is a polypeptide or protein. In some embodiments, the gene product is a mRNA, a tRNA, a rRNA, a miRNA, a dsRNA, or a siRNA.
[51] A cell that “stably expresses” an exogenous polypeptide is one that continues to express a polypeptide encoded by an exogenous gene introduced into the cell either after replication if the cell is dividing or for longer than a day, up to about a week, up to about two weeks, up to three weeks, up to four weeks, for several weeks, up to a month, up to two months, up to three months, for several months, up to a year or more.
[52] A “viral vector” is defined as a recombinantly produced virus or viral particle that comprises a polynucleotide to be delivered into a host cell, either in vivo, ex vivo or in vitro. Examples of viral vectors include retroviral vectors, lentiviral vectors, adenovirus vectors, adeno-associated virus vectors, alphavirus vectors and the like. Alphavirus vectors, such as Semliki Forest virus-based vectors and Sindbis virus-based vectors, have also been developed for use in gene therapy and immunotherapy. See, Schlesinger and Dubensky (1999) Curr. Opin. Biotechnol. 5:434-439 and Ying, et al. (1999) Nat. Med. 5(7):823-827.
[53] In aspects where gene transfer is mediated by a retroviral vector, a vector construct refers to the polynucleotide comprising the retroviral genome or part thereof, and a therapeutic gene. As used herein, “retroviral mediated gene transfer” or “retroviral transduction” carries the same meaning and refers to the process by which a gene or nucleic acid sequences are stably transferred into the host cell by virtue of the virus entering the cell and integrating its genome into the host cell genome. The virus can enter the host cell via its normal mechanism of infection or be modified such that it binds to a different host cell surface receptor or ligand to enter the cell. Retroviruses carry their genetic information in the form of RNA; however, once the virus infects a cell, the RNA is reverse-transcribed into the DNA form which integrates into the genomic DNA of the infected cell. The integrated DNA form is called a provirus. As used herein, retroviral vector refers to a viral particle capable of introducing exogenous nucleic acid into a cell through a viral or viral-like entry mechanism. A “lentiviral vector” is a type of retroviral vector well-known in the art that has certain advantages in transducing nondividing cells as compared to other retroviral vectors. See, Trono D. (2002) Lentiviral vectors, New York: Spring-Verlag Berlin Heidelberg.
[54] In aspects where gene transfer is mediated by a DNA viral vector, such as an adenovirus (Ad) or adeno-associated virus (AAV), a vector construct refers to the polynucleotide comprising the viral genome or part thereof, and a transgene. Adenoviruses (Ads) are a relatively well characterized, homogenous group of viruses, including over 50 serotypes. See, e.g., International PCT Application No. WO 95/27071. Ads do not require integration into the host cell genome. Recombinant Ad derived vectors, particularly those that reduce the potential for recombination and generation of wild-type virus, have also been constructed. See, International PCT Application Nos. WO 95/00655 and WO 95/11984. Wild-type AAV has high infectivity and specificity integrating into the host cell’s genome. See, Hermonat and Muzyczka (1984) Proc. Natl. Acad. Sci. USA 81 :6466- 6470 and Lebkowski, et al. (1988) Mol. Cell. Biol. 8:3988-3996.
[55] Vectors that contain both a promoter and a cloning site into which a polynucleotide can be operatively linked are well known in the art. Such vectors are capable of transcribing RNA in vitro or in vivo, and are commercially available from sources such as Stratagene (La Jolla, CA) and Promega Biotech (Madison, WI). In order to optimize expression and/or in vitro transcription, it may be necessary to remove, add or alter 5’ and/or 3’ untranslated portions of the clones to eliminate extra, potential inappropriate alternative translation initiation codons or other sequences that may interfere with or reduce expression, either at the level of transcription or translation. Alternatively, consensus ribosome binding sites can be inserted immediately 5’ of the start codon to enhance expression.
[56] “Under transcriptional control” is a term well understood in the art and indicates that transcription of a polynucleotide sequence, usually a DNA sequence, depends on its being operatively linked to an element which contributes to the initiation of, or promotes, transcription. “Operatively linked” intends the polynucleotides are arranged in a manner that allows them to function in a cell.
[57] A “probe” when used in the context of polynucleotide manipulation refers to an oligonucleotide that is provided as a reagent to detect a target potentially present in a sample of interest by hybridizing with the target. Usually, a probe will comprise a label or a means by which a label can be attached, either before or subsequent to the hybridization reaction. Suitable labels are described and exemplified herein.
[58] A “primer” is a short polynucleotide, generally with a free 3’ -OH group that binds to a target or “template” potentially present in a sample of interest by hybridizing with the target, and thereafter promoting polymerization of a polynucleotide complementary to the target. A “polymerase chain reaction” (“PCR”) is a reaction in which replicate copies are made of a target polynucleotide using a “pair of primers” or a “set of primers” consisting of an “upstream” and a “downstream” primer, and a catalyst of polymerization, such as a DNA polymerase, and typically a thermally-stable polymerase enzyme. Methods for PCR are well known in the art, and taught, for example in M. MacPherson et al. (1991) PCR: A Practical Approach, IRL Press at Oxford University Press. All processes of producing replicate copies of a polynucleotide, such as PCR or gene cloning, are collectively referred to herein as “replication.” A primer can also be used as a probe in hybridization reactions, such as Southern or Northern blot analyses. Sambrook et al., supra. The primers may optionall contain detectable labels and are exemplified and described herein.
[59] As used herein, the term "label" intends a directly or indirectly detectable compound or composition that is conjugated directly or indirectly to the composition to be detected, e.g., polynucleotide or protein such as an antibody so as to generate a "labeled" composition. The term also includes sequences conjugated to the polynucleotide that will provide a signal upon expression of the inserted sequences, such as green fluorescent protein (GFP) and the like. The label may be detectable by itself (e.g. radioisotope labels or fluorescent labels) or, in the case of an enzymatic label, may catalyze chemical alteration of a substrate compound or composition which is detectable. The labels can be suitable for small scale detection or more suitable for high-throughput screening. As such, suitable labels include, but are not limited to radioisotopes, fluorochromes, chemiluminescent compounds, dyes, and proteins, including enzymes. The label may be simply detected or it may be quantified. A response that is simply detected generally comprises a response whose existence merely is confirmed, whereas a response that is quantified generally comprises a response having a quantifiable (e.g., numerically reportable) value such as an intensity, polarization, and/or other property. In luminescence or fluoresecence assays, the detectable response may be generated directly using a luminophore or fluorophore associated with an assay component actually involved in binding, or indirectly using a luminophore or fluorophore associated with another (e.g., reporter or indicator) component.
[60] Examples of luminescent labels that produce signals include, but are not limited to bioluminescence and chemiluminescence. Detectable luminescence response generally comprises a change in, or an occurrence of, a luminescence signal. Suitable methods and luminophores for luminescently labeling assay components are known in the art and described for example in Haugland, Richard P. (1996) Handbook of Fluorescent Probes and Research Chemicals (6th ed.). Examples of luminescent probes include, but are not limited to, aequorin and luciferases.
[61] Examples of suitable fluorescent labels include, but are not limited to, fluorescein, rhodamine, tetramethylrhodamine, eosin, erythrosin, coumarin, methyl-coumarins, pyrene, Malacite green, stilbene, Lucifer Yellow, Cascade Blue.TM., and Texas Red. Other suitable optical dyes are described in the Haugland, Richard P. (1996) Handbook of Fluorescent Probes and Research Chemicals (6th ed.).
[62] In another aspect, the fluorescent label is functionalized to facilitate covalent attachment to a cellular component present in or on the surface of the cell or tissue such as a cell surface marker. Suitable functional groups, including, but not are limited to, isothiocyanate groups, amino groups, haloacetyl groups, maleimides, succinimidyl esters, and sulfonyl halides, all of which may be used to attach the fluorescent label to a second molecule. The choice of the functional group of the fluorescent label will depend on the site of attachment to either a linker, the agent, the marker, or the second labeling agent.
[63] Attachment of the fluorescent label may be either directly to the cellular component or compound or alternatively, can by via a linker. Suitable binding pairs for use in indirectly linking the fluorescent label to the intermediate include, but are not limited to, antigens/antibodies, e.g., rhodamine/anti-rhodamine, biotin/avidin and biotin/strepavidin.
[64] The phrase “solid support” refers to non-aqueous surfaces such as “culture plates” “gene chips” or “microarrays.” Such gene chips or microarrays can be used for diagnostic and therapeutic purposes by a number of techniques known to one of skill in the art. In one technique, oligonucleotides are attached and arrayed on a gene chip for determining the DNA sequence by the hybridization approach, such as that outlined in U.S. Patent Nos.: 6,025,136 and 6,018,041. The polynucleotides of this invention can be modified to probes, which in turn can be used for detection of a genetic sequence. Such techniques have been described, for example, in U.S. Patent Nos.: 5,968,740 and 5,858,659. A probe also can be attached or affixed to an electrode surface for the electrochemical detection of nucleic acid sequences such as described by Kayem et al. U.S. Patent No. 5,952,172 and by Kelley et al. (1999) Nucleic Acids Res. 27:4830-4837.
[65] Vitellogenin (Vg) is a precursor of egg yolk that transports protein and some lipid from the liver through the blood to the growing oocytes where it becomes part of the yolk. Normally, it is only found in the blood or hemolymph of females, and can therefore be used as a biomarker in vertebrates of exposure to environmental estrogens which stimulate elevated levels in males as well as females. "Vitellogenin" is a synonymous term for the gene and the expressed protein. The protein product is classified as a glycolipoprotein, having properties of a sugar, fat and protein. Vg polynucleotide and protein sequences can be found in UniProt database, e.g., https://www.uniprot.org/uniprot/Q868N5 and https://www.uniprot.org/uniprot/Q868N5, last accessed on August 28, 2021 , and the sequences provided herein.
[66] A “composition” is intended to mean a combination of active agent and another compound or composition, inert (for example, a detectable agent or label) or active, such as an adjuvant.
[67] A “pharmaceutical composition” is intended to include the combination of an active agent with a carrier, inert or active, making the composition suitable for diagnostic or therapeutic use in vitro, in vivo or ex vivo.
[68] As used herein, the term “pharmaceutically acceptable carrier” encompasses any of the standard pharmaceutical carriers, such as a phosphate buffered saline solution, water, and emulsions, such as an oil/water or water/oil emulsion, and various types of wetting agents. The compositions also can include stabilizers and preservatives. For examples of carriers, stabilizers and adjuvants, see Martin (1975) Remington’s Pharm. Sci., 15th Ed. (Mack Publ. Co., Easton).
[69] An “artificial diet” intends a honey bee diet with an antigen or fragment thereof as described herein.
[70] "Substantially homogeneous" describes a population of cells in which more than about 50%, or alternatively more than about 60 %, or alternatively more than 70 %, or alternatively more than 75 %, or alternatively more than 80%, or alternatively more than 85 %, or alternatively more than 90%, or alternatively, more than 95 %, of the cells are of the same or similar phenotype. Phenotype can be determined by a pre-selected cell surface marker or other marker, e.g. myosin or actin or the expression of a gene or protein, e.g. a calcium handling protein, a t-tubule protein or alternatively, a calcium pump protein. In another aspects, the substantially homogenous population have a decreased (e.g., less than about 95%, or alternatively less than about 90%, or alternatively less than about 80%, or alternatively less than about 75%, or alternatively less than about 70%, or alternatively less than about 65%, or alternatively less than about 60%, or alternatively less than about 55%, or alternatively less than about 50%) of the normal level of expression than the wild-type counterpart cell or tissue.
[71] The term “American foulbrood” or “American foulbrood disease” or “AFB” as used herein, refers to a fatal bacterial disease of honeybee brood caused by the spore forming bacterium Paenibacillus larvae. Since Paenibacillus larvae causes American fouldbrood disease in honeybees, Paenibacillus larvae is referred to herein as a “disease- causing” bacterium or a “disease species.”
[72] The terms “non-disease causing” species or “non-disease species” as used herein refer to species of bacteria which may or may not be pathogenic but which do not cause the disease being targeted. For example, with respect to American foulbrood disease which is caused by Paenibacillus larvae, exemplary non-disease Paenibacillus species, which may be found for example in the environment, but not on humans or in human wounds may include e.g., Paenibacillus alvei, Paenibacillus dendritiformis Paenibacillus amylolyticus, Paenibacillus campinasensis, Paenibacillus chondr oitinus, Paenibacillus chungangensis, Paenibacillus doosanensis, Paenibacillus glucanolyticus, Paenibacillus humicus, Paenibacillus lactis, Paenibacillus lautus, Paenibacillus lentimorbus, Paenibacillus mace ran, Paenibacillus macerans-like, Paenibacillus macquariensis, Paenibacillus motobuensis, Paenibacillus pabuli. Paenibacillus phoenicis, Paenibacillus polymyxa, Paenibacillus popilliae. Paenibacillus pulde ungensis, Paenibacillus residui, Paenibacillus stellife, Paenibacillus thiaminolyticus, Paenibacillus validus, and Paenibacillus xylanisolvens. By way of example, Paenibacillus alvei and Paenibacillus dendritiformis are pathogenic and will inflict a brood disease on larvae but the disease is not American Foulbrood caused by Paenibacillus larvae and are thus non-disease forming species.
[73] The term “honey bee” as used herein refers to is any bee which is a member of the genus Apis, primarily distinguished by the production and storage of honey and the construction of perennial, colonial nests from wax. For example, two species of honey bees, namely A. mellifera or A. cerana indica, are often maintained by beekeepers. Honey bees include but are not limited to Apis andreniformis and Apis florea in subgenus Micrapis, Apis dorsata in subgenus Megapis, and Apis cerana, Apis koschevnikovi, Apis mellifera and Apis nigrocincta in subgenus Apis.
[74] The term “bee colony” or “honeybee colony” as used herein, refers to a social unit of bees, e.g., honeybees comprising a colony. The social unit can be of any system organization utilized by bees, which has the purpose of facilitating survival of the group or colony. Typically, a "bee colony" consists of several thousand bees that cooperate in nest building, food collection, and brood rearing. Each member of a "bee colony" has a definite task to perform, and it takes the combined efforts of the entire colony to survive and reproduce. A colony typically comprises a single queen, thousands of workers, and hundreds of drones during late spring and summer. Thus, a bee colony is a “population of honeybees.”
[75] Typically, a “honeybee colony” peaks from late spring to summer and reaches a low point in winter. The social structure of the colony is maintained by the queen and workers and depends on an effective system of communication. Domesticated honeybees are cultivated in “beehives” or “honeybee hives.” Thus, the term “beehive” or “honeybee hive” refers to a structure that functions as a habitation for a colony of bees, e.g., a colony of honeybees.
[76] As used herein, the term “honey bee” is any bee that is a member of the genus Apis. Two species of honey bees, A. mellifer, A. cernan indica, A. andrreniformis, A. florea, A. koschevnikovie, and A. nigrocincta are examples of such. [77] The term “nurse bee” as used herein intends are the bees that feed the worker larvae worker jelly that is secreted from glands that produce royal jelly.
[78] The term “worker bee” as used herein intends any female (eusocial) bee that lacks the full reproductive capacity of the colony’s queen bee.
[79] The term “brood” intends the three developmental stages in bees, which are collectively known as brood. Bees begin in eggs, which hatch to become larvae (plural). The larvae is legless and is specialized to eat.
[80] The term “prophylactic” or “vaccine” refers to an agent that acts to prevent a disease e.g., a honeybee disease, such as e.g., Foulbrood caused by the bacterium Paenibacillus larvae in the brood.
[81] The term “vaccinate” as used herein, refers to means for producing immunity against a disease e.g., producing immunity to Paenibacillus larvae so as to prevent a disease or condition from occurring (prophylactic treatment) or inhibiting the disease from spreading (slowing or arresting its development) in the brood, larvae, progeny or colony.
[82] The term “treatment” intends to raise an immune response in the queen which is then passed on to her progeny.
[83] The term “raise an immune response” intends that the treatment the vaccine or treatment produces the non-disease causing antigens in the ovaries of the honey bee queen or to the developing eggs.
[84] An “antigen/unif ’ intends the number of cells or antigenic fragments of the non- disease pathogen.
[85] The term “dose” intends the amount provided to the queen bee, the worker bee, nurse bee or larvae in one feeding or unit amount of food.
[86] Modes For Carrying Out the Disclosure
[87] Any composition prepared by or isolated from these methods are provided herein as well as use thereof in preventive and therapeutic methods.
[88] Provided herein is a method to isolate a honey bee antigen that could be useful as a vaccine, the method comprising, or consisting essentially of, or yet further consisting of, contacting purified or recombinant vitellogenin (rVg) with a solid support under conditions to form rVg bound to the solid support; contacting a solubilized inactivated honey bee pathogen with the rVg bound to the solid support; and then eluting the inactivated honey bee pathogens from the solid support to isolate the honey bee antigen. As used herein, the term antigen intends complete or full length proteins and fragments thereof. As is apparent to the skilled artisan wash steps to remove unbound rVg and antigen can be added to the method.
[89] In one aspect, the solid support is selected from a chromatographic support, a sepharose CL column (Pharmacia), or an ion exchange column. These are commercially available from BioRad, , Pall Corp., Thermofischer and Pharmacia.
[90] The solid support can be a Sepharose CL column or other chromatographic support using cyanogen bromide or other covalent linking mechanism known to those skilled in the art. Alternatively, rVg may be bound to an ion exchange (IEX) column at favourable conditions (optimal pH, salt). These methods are known in the art and will vary with the rVg and the solid support.
[91] As noted above, solubilized protein mixtures of these pathogens are applied to the solid phase or antigens or fragment thereof with rVg, and proteins that bind to rVg are eluted using an appropriate method depending on the antigen and column. In one aspect, a salt gradient and/or pH gradient is used to elute the proteins bound to the rVg. The rVg is covalently bound to the column and the proteins of interest are eluted. In one aspect, the rVg can be ionically bound to ion exchange chromatography gels (IEX gels), the rVg- protein mixture is eluted depending on binding characteristics, ionic strength, etc. Purified proteins or protein-rVg complexes are then identified using standard methods such as electrophoresis, high performance liquid chromatography (HPLC), mass spectrometry (MS), and/or protein sequencing, etc.
[92] After eluting the isolated antigens, the method can further comprise, or consist essentially of, or yet further consist of, determining the identity of the isolated inactivated honey bee pathogens. This can be accomplished by sequencing the antigens using well- known techniques that will determine the antigen and the polynucleotide encoding the antigen. In a further aspect, the isolated antigen is expressed in a recombinant cell system for expression and/or multiplication of the antigen.
[93] Also provided herein is a method to isolate a honey bee antigen that could be useful as a vaccine, the method comprising, or consisting essentially of, or yet further consisting of, binding an anti-His tag or anti-Vg antibody with a solubilized inactivated honey bee pathogen and detecting any antibody bound to the inactivated honey bee pathogen, wherein the antibody or inactivated honey bee pathogen is detectably labeled.
[94] In one aspect, the antibody or inactivated honey bee pathogen is detectably labeled. Such labels are known in the art and described herein. Anti-His tag antibodies are commercially available from Cell Signaling Technology, Invitrogen, ThermoFisher, and other commercial vendors. Anti-Vg antibodies are made using purified or rVg or fragments thereof using conventional techniques well-known in the art.
[95] The rVg for use in the methods can be expressed in a prokaryotic or eukaryotic cell system, e.g., baculovirus, CHO, HEK, E. coli. Saccharomyces. and others known in the art. In one aspect, recombinant vitellogenin (rVg) is produced in an insect viral expression system, such as a baculovirus expression system. In one aspect, the baculovirus expression system is the DHIOBac system.
[96] Any Vg protein or fragment can be used in the method. One non-limiting example is the rVg having the amino acid sequence of SEQ ID NO: 1, or a fragment or an equivalent thereof, with and without the His tag. The polynucleotide encoding SEQ ID NO: 1 is provided in SEQ ID NO: 2. When the His tag is not used, other detectable labels can be used, e.g., a radioisotope labels or a fluorescent label or, in the case of an enzymatic label, it may catalyze chemical alteration of a substrate compound or composition which is detectable. As such, suitable labels include, but are not limited to radioisotopes, fluorochromes, chemiluminescent compounds, dyes, and proteins, including enzymes. The label may be simply detected or it may be quantified. A response that is simply detected generally comprises a response whose existence merely is confirmed, whereas a response that is quantified generally comprises a response having a quantifiable (e.g., numerically reportable) value such as an intensity, polarization, and/or other property. In luminescence or fluoresecence assays, the detectable response can be generated directly using a luminophore or fluorophore associated with an assay component actually involved in binding, or indirectly using a luminophore or fluorophore associated with another (e.g., reporter or indicator) component.
[97] Prior to contacting the honey bee pathogens (antigens and fragments thereof) with the solid support or antigen or fragment thereof, they are inactivated and then solubilized so that internal and trans-membrane proteins are in solution. Suitable methods for inactivation include for example, sonication, followed by solubilization in non-ionic detergents such as Brij, Triton, octyl-glucoside and others known in the art. The antigen contacted with the solid support can comprise one or multiple types of antigens, e.g., from the same or different pathogen.
[98] Non-limiting examples of honey bee antigens for use in the method are those responsible for American foulbrood, European foulbrood, deformed wing virus, Varroa mite infestations and other viral, bacterial and parasitic diseases of honey bees. Examples of such include, without limitation, Paenibacillus larvae, Paenibacillus alvei, Paenibacillus dendritiformis Paenibacillus amylolyticus, Paenibacillus campinasensis, Paenibacillus chondr oitinus, Paenibacillus chungangensis, Paenibacillus doosanensis, Paenibacillus glucanolyticus, Paenibacillus humicus, Paenibacillus lactis, Paenibacillus lautus, Paenibacillus lentimorbus, Paenibacillus maceran, Paenibacillus macerans-like, Paenibacillus macquariensis, Paenibacillus motobuensis, Paenibacillus pabuli, Paenibacillus phoenicis, Paenibacillus polymyxa, Paenibacillus popilliae, Paenibacillus puldeungensis, Paenibacillus residui, Paenibacillus stellife, Paenibacillus thiaminolyticus, Paenibacillus validus, or Paenibacillus xylanisolvens.
[99] After isolation of the antigen or fragment thereof (e.g., eluting the isolated antigens) the methods can further comprise, or consist essentially of, or yet further consist of, determining the identity of the isolated inactivated honey bee pathogens. This can be accomplished by sequencing the antigens using well-known techniques that will determine the antigen and the polynucleotide encoding the antigen. In a further aspect, the isolated antigen is expressed in a recombinant cell system for expression and/or multiplication of the antigen.
[100] In another aspect, provided herein is a proteomics method for the identification of antigenic proteins. In this method, rVg is used in the place of antibodies to detect protein in high throughput methods such as phage display, high throughput screening of the entire pathogen genome, and other methods known in the art. Labeled rVg or an anti-His tag antibodyis used to detect the proteins of interest, and then they may be further characterized. This method allows for the rapid screening of multiple disease agents extremely quickly.
[101] In a yet further aspect, provided herein are vaccines and bacterins identified by these methods. After the proteins of interest are identified, they are partially sequenced, and the genes expressing such proteins are isolated, and the proteins are produced in a suitable eukaryotic or prokaryotic expression system (baculovirus, CHO, HEK, E. coli, Saccharomyces, and others known in the art). The recombinant proteins can be added to feed for queen bees as is described in U.S. Patent No. 10,994,001, or may be introduced directly into hives in a suitable feed substance. Immunity in honey bees and larvae is generated and assessed as described in U.S. Patent No. 10,994,001. Toll-like receptor (TLR) binding CpGs may be added to the vaccines or bacterins to enhance effector mechanisms.
[102] In a specific embodiment, provided herein is a composition, comprising, or consisting essentially of, or yet further consisting of, the immunogenic proteins and a carrier. In one aspect, the composition comprises, or consisting essentially of, or yet further consisting of, the proteins or immunogenic fragments thereof and a carrier. A carrier can be a solid or a liquid carrier and can include preservatives, insect nutrients, or other coloring agents as necessary. In one specific embodiment, the carrier is an insect food, such as a queen bee wafer or sugar feed.
[103] Compositions of the present invention are easily administered or fed to insects. In addition to bacteria, fungi, fragments and/or spores thereof, the edible composition may optionally comprise one or more acceptable (e.g. pharmaceu-tically acceptable) agents selected from the group consisting of carrier(s) (e.g. water, glucose or lactose), adjuvant(s), excipient(s), auxiliary excipient(s), anti- septic(s), stabilizing, thickening or coloring agent(s), perfume(s), binding agent(s), filling agent(s), lubricating agent(s), suspending agent(s), sweetener(s), flavoring agent(s), gelatinizer(s), anti- oxi dant(s), preservative(s), buffer(s), pH regulator(s), wetting agent(s) and components normally found in corresponding products. However, in a very specific embodiment of the disclosure only bacteria, fungi, fragments thereof and/or spores thereof are needed in the composition. In a further specific embodiment the composition consists of only bacteria, fungi, fragments thereof and/or spores thereof and water.
[104] In one specific embodiment of the invention, the compositions or artificial diet may be used for example in solid, semisolid or liquid form, such as in the form of patties, syrups, drenches, dustings, pastes, tablets, pellets, capsules, solutions, emulsions, suspensions or like. Preferably the composition is for oral administration. [105] As used herein, the term "prevent" or "preventing" refers to feeding or administration of microbes to an insect for purposes which include not only 100% or complete prevention but also partial prophylaxis and therefore also amelioration or alleviation of disorders or symptoms related to microbial infections. Preventive effect may be assessed e.g. by monitoring the symptoms mentioned in any of the tables of Figures 6-9 of U.S. Patent No. 10,994,001, incorporated herein by reference. In this respect, the present disclosure can provide any amount of increase e.g. in the survival data compared to untreated controls.
[106] In one aspect, the methods further comprise feeding a honey bee queen or worker bee an artificial insect diet comprising antigen or fragment thereof isolated by the above methods. The compositions are useful as vaccines. In one aspect, the amount of antigen in the artificial diet is 1 to 20% by weight. The artificial diet can be administered to the queen beefor administration one, two or three times a year. In one aspect, the honey bee queen prior is feed the artificial diet prior to egg laying. After consumption of the diet, the queen bee and/or the hive is challenged to determine protective or therapeutic response, as noted herein and U.S. Patent No. 10,994,001, incorporated herein by reference.
[107] Also provided herein is a method selected from: treating, raising an immune response or immunizing a honeybee larvae, honeybee, honeybee hatched from a treated queen bee, honeybee population or honeybee hive against AFB, the method comprising, or alternatively consisting essentially of, or yet further consisting essentially of, administering an effective amount of the composition as described herein to a nurse bees or the honeybee queen or honey bee larvae thereby treating, raising an immune response, or immunizing the queen bee and the honeybee larvae produced by the queen bee.
Experimental Methods
[108] Preparation of Recombinant Vitellogenin. The recombinant vitellogenin was produced by Genscript, Inc. (Genscript, see https://www.genscript.com/?src=google&gclid=Cj0KCQjwvaeJBhCvARIsABgTDM419ret qScAAlyWup5xcTOXQC5muC9o4cZ7wSVpn9b62LHArbj7rqQaAvBZEALw_wcB, last accessed on August 28, 2021) using their proprietary methods, vector technologies and expression systems. The target DNA sequence of honey bee VG was synthesized and then subcloned into a target vector for insect cell expression. The map of the construct is shown in FIG. 5. DH10Bac was used for the recombinant bacmid (rbacmid) generation. The positive rbacmid containing VG sequence gene was confirmed by polymerase chain reaction (PCR).
Virus Generation
[109] Sf9 cells were grown in Sf-900 II SFM Expression Medium (Life Technologies, Catalog Number: 10902-088). The cells were maintained in Erlenmeyer Flasks at 27 °C in an orbital shaker. One day before transfection, the cells were seeded at an appropriate density in 6 wells CORNING-COSTAR. On the day of transfection, DNA and Transfection Reagent (PROMEGA, Catalog Number:E2691) were mixed at an optimal ratio and then added into the plate with cells ready for transfection. Cells were incubated in Sf-900 II SFM for 5-7 days at 27 °C before harvest. The supernatant was collected after centrifugation and designated as Pl viral stock. P2 was amplified for later infection. The expression was analyzed by Western blot.
Antigen Expression
[110] The 0.5 L Sf9 cell culture were infected by Passage 2 virus. Cells were incubated in Sf-900II SFM(IX) for 3 days at 27 °C before harvest. The expression was analyzed by Western blot.
Purification and Analysis
[111] Cell culture was centrifuged and followed by filtration. Filtered medium supernatant was incubated with Ni-NTA to capture the target protein. Higher purity fractions were pooled and followed by 0.22 pm filter sterilization. Proteins were analyzed by SDS-PAGE and Western blot by using standard protocols for molecular weight and purity measurements. The primary antibody for Western blot was Mouse-anti-His mAb (GenScript, Cat.No.A00186). The concentration was determined by Bradford protein assay with BSA as a standard.
Anti-His-tag Antibody
[112] The anti-His antibody used was a monoclonal Mouse-anti-His antibody (GenScript, Cat.No.A00186).
Paenibacillus larvae Strain [113] Paenibacillus larvae strain was isolated from NY state bee hive. The strain has been tested found to be a pure strain of Paenibacillus larvae strain (99.4% similarity to Paenibacillus larvae strain DSM 7030) and found to be the ERIC I Pathotype.
Growth Medium
[114] P. larvae strain was grown on enriched Mueller-Hinton MYPGP agar medium, for 2-6 days, harvested and then snap-frozen at -80°C and shipped for protein analysis on dry ice, with a temperature control monitor, to avoid any proteolysis.
[115] Analysis of P. larvae Vittellogenin Western blot
[116] Gel loading and sample preparation are given in Table 1 (see 2D gel analysis, below).
Table 1
Figure imgf000029_0001
2D Gel Loading and Sample Preparation.
[117] The four cell pellet was lysed in 225 μl of of Osmotic Lysis Buffer containing protease inhibitor stock, nuclease stock, and phosphatase inhibitor stocks (I and II), 75 pl SDS Boiling Buffer without β-mercaptoethanol, and 100 mg of washed glass beads (Sigma G9268, mesh size 425-6000 microns) as described by lazwinski (SDS-polyacrylamide gel electrophoresis. Trends in Biochemical Science, 25, 590-592. .Jazwinski. S.M. (1990)). The sample was vortexed 2 minutes, sonicated 15 minutes, vortexed again for 2 minutes, and placed in a dry bath at 95°C for 10 minutes. The sample was centrifuged, and the supernatants was combined into one tube. The protein concentration (4.72 μg/μl) was then determined using the BCA assay (Smith et. al. Anal. Biochem. 150: 76-85, 1985, and Pierce Chemical Co., Rockford, IL). The sample was then lyophilized, and redissolved to 2.66 mg/ml in 1 : 1 diluted SDS Boiling Buffer: Urea Sample Buffer before loading (see 2D gels below).
Electrophoresis and Western Blot
[118] Two-dimensional electrophoresis was performed according to the carrier ampholine method of isoelectric focusing (O'Farrell, P.H., J. Biol. Chem. 250: 4007-4021, 1975; Burgess-Cassler, A., Johansen, J., Santek, D., Ide J., and Kendrick N., Clin. Chem. 35: 2297, 1989; Kendrick, N., C.C. Darie, M. Hoelter, G. Powers, and J. Johansen, Adv Exp Med Biol, 2019. 1140: p. 563-574). Isoelectric focusing was carried out in a glass tube of inner diameter 3.3 mm using 2.0% 4-8 mix (Serva, Heidelberg, Germany) for 20,000 volt- hrs. One pg of an IEF internal standard, tropomyosin, was added to each sample. This protein migrates as a doublet with lower polypeptide spot of MW 33,000 and pl 5.2; an arrow on the stained gels marks its position. The enclosed tube gel pH gradient plot for this set of Servalytes was determined with a surface pH electrode.
[119] After equilibration for 10 min in buffer "O" (10% glycerol, 50 mM dithiothreitol, 2.3% SDS and 0.0625 M tris, pH 6.8), each tube gel was sealed to the top of a stacking gel that overlaid a 10% acrylamide slab gel (1.0 mm thick). SDS slab gel electrophoresis was carried out for about 5 hrs at 25 mA/gel. The following proteins (Millipore Sigma) were used as molecular weight standards: myosin (220,000), phosphorylase A (94,000), catalase (60,000), actin (43,000), carbonic anhydrase (29,000), and lysozyme (14,000). These standards appear as bands at the basic edge of the Coomassie Brilliant Blue R-250 stained 10% acrylamide slab gel. The gel was dried between sheets of cellophane paper with the acid edge to the left.
[120] After slab gel electrophoresis, duplicate gels for blotting were placed in transfer buffer (10 mM CAPS, pH 11.0, 10% MeOH) and transblotted onto PVDF membrane overnight at 225 mA and approximately 100 volts/ two gels. The same proteins as above (Millipore Sigma) were used as molecular weight standards. These standards appear as bands at the basic edge of the Coomassie Brilliant Blue R-250-stained membrane.
Western Blot Methods
[121] The blots were destained in 100% methanol and rinsed briefly in Tween-20 tris buffer saline (TTBS).
His Tag & Anti-Mouse IgG-HRP Only (No Vitellogenin), Super Block, LF1473 #12 (See 2D gels, in the Figures). [122] The blot was blocked overnight in Super Block (ThermoFisher, Cat # 37535) containing 0.05% Tween-20 (SBT). The blot was then placed in secondary solution (THE His Tag [GenScript, Cat# A00186-100, Lot# 17K001367] diluted to 0.02 μg/ml in SBT) for two hours and rinsed 3 x 10 minutes in TTBS. The blot was then placed in tertiary antibody (anti-Mouse IgG-HRP [GE, Cat# NA931V, Lot# 17205275] diluted 1 :2,000 in SBT) for two hours, rinsed as above, treated with ECL, and exposed to x-ray
Vitellogenin, Super Block, LF1473 #13 (see 2Dgels, below)
[123] The blot was blocked for two hours in Superblock (Thermofi scher) containing 0.05M Tris buffer, pH 6.8 (SBT). The blot was then incubated overnight in primary solution (Vitellogenin (GenScript, Lot # U5329GB010-3/P4GE001)) diluted to 0.1 μg/ml in SBT) and rinsed 3 x 10 minutes in TTBS. The blot was then placed in secondary solution (THE His Tag [GenScript, Cat# A00186-100, Lot# 17K001367] diluted to 0.02 μg/ml in SBT) for two hours and rinsed 3 x 10 minutes in TTBS. The blot was then placed in tertiary antibody (anti-Mouse IgG-HRP [GE, Cat# NA931V, Lot# 17205275] diluted 1 :2,000 in SBT) for two hours, rinsed as above, treated with ECL, and exposed to x-ray film.
Experimental Results
[124] FIG. 2 shows the construct of the one dimensional electrophoresis.
[125] The ID (molecular weight) gel analysis indicated that there were a number of proteins that may bind specifically to Vitellogenin. These were initially identified by the red arrows in FIG. 1. Proteins that bound non-specifically to the either the His-tag or anti- mouse Ig antibodies are indicated by yellow arrows. These data led to analysis using 2D gels.
2D Gel analysis.
[126] FIG. 2 and FIG. 3 indicate the results of detection of protein binding by vitellogenin. Using simple visual overlay at least 10 spots can be detected that bind uniquely to vitellogenin. In FIG. 4, these are identified on the Coomassie stained gel.
Experimental Discussion
Non-Specific binding
[127] It appears that there is a considerable amount of non-specific binding - either by the anti-HIS tag or by the anti-HRP. Low level non-specific binding appeared to be at a MW -35-50, with stronger signals at MW between 14 and 29. rVg specific binding
[128] Honeybee rVG binds specifically to multiple proteins detected. It is noteworthy that there are some proteins that appear in very small concentrations, show a high degree of binding (e.g. H, I, J in the 2D gels above). These tended to be lower MW proteins between 14 and 29 MW. There also appears to be a stronger signal at ~60 and 94 MW and between 94 and 220 MW.
Virulence factors of PL
[129] Known virulence factors for P. larvae include enolase (MW 82-200 kDa, depending on isoform), and a chitinase (49 kDa). A variety of toxins have also been proposed as virulence factors, such as C3 mART which has a MW between 20 and 25 kDa. Further analysis of the proteins bound by vitellogenin will identify those proteins that are involved in immunity.
Equivalents
[130] It is to be understood that while the invention has been described in conjunction with the above embodiments, that the foregoing description and examples are intended to illustrate and not limit the scope of the invention. Other aspects, advantages and modifications within the scope of the invention will be apparent to those skilled in the art to which the invention pertains.
[131] It should be understood that although the present disclosure has been specifically disclosed by specific embodiments and optional features, modification, improvement and variation of the embodiments therein herein disclosed may be resorted to by those skilled in the art, and that such modifications, improvements and variations are considered to be within the scope of this disclosure. The materials, methods, and examples provided here are representative of particular embodiments, are exemplary, and are not intended as limitations on the scope of the disclosure.
[132] The scope of the disclosure has been described broadly and generically herein. Each of the narrower species and subgeneric groupings falling within the generic disclosure also form part of the disclosure. This includes the generic description with a proviso or negative limitation removing any subject matter from the genus, regardless of whether or not the excised material is specifically recited herein. [133] In addition, where features or aspects of the invention are described in terms of Markush groups, those skilled in the art will recognize that the invention is also thereby described in terms of any individual member or subgroup of members of the Markush group.
[134] All publications, patent applications, patents, and other references mentioned herein are expressly incorporated by reference in their entirety, to the same extent as if each were incorporated by reference individually. In case of conflict, the present specification, including definitions, will control.
SEQ ID NO: 1 Protein sequence of Vg
> U2111FG010 -1 (Vitellogenin, C-His, His tag is underlined, which is an optional element for the protein)
Protein Length=1778 MW=202109.1 Predicted pl=6.82
MLLLLTLLLFAGTVAADFQHNWQVGNEYTYLVRSRTLTSLGDLSDVHTGILIKALL TVQAKDSNVLAAKVWNGQYARVQQSMPDGWETE
ISDQMLELRDLPISGKPFQIRMKHGLIRDLIVDRDVPTWEVNILKSIVGQLQVDTQGE NAVKVNSVQVPTDDEPYASFKAMEDSVGGKC
EVLYDIAPLSDFVIHRSPELVPMPTLKGDGRHMEVIKIKNFDNCDQRINYHFGMTDN SRLEPGTNKNGKFF SRS STSRIVISESLKHFT
IQSSVTTSKMMVSPRLYDRQNGLVLSRMNLTLAKMEKTSKPLPMVDNPESTGNLV YIYNNPF SD VEERRVSKT AMNSNQI VSDNSLS S S
EEKLKQDILNLRTDIS S S S S SIS S SEENDF WQPKPTLED APQNSLLPNF VGYKGKHIG KSGKVDVINAAKELIFQIANELEDASNIPVH
ATLEKFMILCNLMRTMNRKQISELESNMQISPNELKPNDKSQVIKQNTWTVFRDAIT QTGTGPAFLTIKEWIERGTTKSMEAANIMSKL
PKTVRTPTDSYIRSFFELLQNPKVSNEQFLNTAATLSFCEMIHNAQVNKRSIHNNYP VHTFGRLTSKHDNSLYDEYIPFLERELRKAHQ
EKDSPRIQTYIMALGMIGEPKILSVFEPYLEGKQQMTVFQRTLMVGSLGKLTETNPK LARSVLYKIYLNTMESHEVRCTAVFLLMKTNP
PLSMLQRMAEFTKLDTNRQVNSAVKSTIQSLMKLKSPEWKDLAKKARSVNHLLTH HEYDYELSRGYIDEKILENQNIITHMILNYVGSE DSVIPRILYLTWYSSNGDIKVPSTKVLAMISSVKSFMELSLRSVKDRETIISAAEKIAE
ELKIVPEELVPLEGNLMINNKYALKFFPFD
KHILDKLPTLISNYIEAVKEGKFMNVNMLDTYESVHSFPTETGLPFVYTFNVIKLTK
TSGTVQAQINPDFAFIVNSNLRLTFSKNVQGR
VGFVTPFEHRHFISGIDSNLHVYAPLKISLDVNTPKGNMQWKIWPMKGEEKSRLFH
YSVVPFVSNHDILNLRPLSMEKGTRPMIPDDNT
SLALPKNEGPFRLNVETAKTNEEMWELIDTEKLTDRLPYPWTMDNERYVKVDMY
MNLEGEQKDPVIFSTSFDSKVMTRPDTDSENWTPK
MMAVEPTDKQANSKTRRQEMMREAGRGIESAKSYVVDVRVHVPGESESETVLTL
AWSESNVESKGRLLGFWRVEMPRSNADYEVCIGSQ
IMVSPETLLSYDEKMDQKPKMDFNVDIRYGKNCGKGERIDMNGKLRQSPRLKELV
GATSIIKDCVEDMKRGNKILRTCQKAVVLSMLLD
EVDISMEVPSDALIALYSQGLFSLSEIDNLDVSLDVSNPKNAGKKKIDVRAKLNEYL
DKADVIVNTPIMDAHFKDVKLSDFGFSTEDIL
DTADEDLLINNVFYEDETSCMLDKTRAQTFDGKDYPLRLGPCWHAVMTTYPRINP
DNHNEKLHIPKDKSVSVLSRENEAGQKEVKVLLG
SDKIKFVPGTTSQPEVFVNGEKIVVSRNKAYQKVEENEIIFEIYKMGDRFIGLTSDKF
DVSLALDGERVMLKASEDYRYSVRGLCGNFD
HDSTNDFVGPKNCLFRKPEHFVASYALISNQCEGDSLNVAKSLQDHDCIRQERTQQ
RNVISDSESGRLDTEMSTWGYHHNVNKHCTIHR
TQVKETDDKICFTMRPVVSCASGCTAVETKSKPYKFHCMEKNEAAMKLKKRIEKG
ANPDLSOKPVSTTEELTVPFVCKAHHHHHHHH* *
SEQ ID NO: 2 DNA Sequence Encoding Vg (Signal Peptide is underlined and is an optional element for the DNA Sequence).
GAATTCGCCGCCACCATGCTGCTGCTGCTGACTCTGCTGCTGTTCGCTGGTACCG
TGGCTGCCGACTTCCAGCACAACTGGCAGG
TCGGCAACGAGTACACCTACCTGGTGCGCTCTCGTACCCTGACTTCACTGGGCG
ACCTGTCCGACGTCCACACTGGAATCCTGAT
CAAGGCTCTGCTGACCGTGCAGGCCAAGGACTCTAACGTCCTGGCTGCCAAAGT GTGGAACGGCCAGTACGCTCGTGTGCAGCAG
TCCATGCCCGACGGATGGGAGACTGAAATCAGCGACCAGATGCTGGAACTGCG
TGACCTGCCAATCTCAGGCAAGCCTTTCCAGA
TCAGGATGAAGCACGGACTGATCAGGGACCTGATCGTCGACAGAGACGTGCCA
ACCTGGGAGGTGAACATCCTGAAGTCTATCGT
CGGTCAGCTGCAGGTGGACACTCAGGGCGAAAACGCTGTGAAGGTCAACTCAG
TCCAGGTCCCCACCGACGACGAGCCATACGCT
TCCTTCAAGGCCATGGAAGACAGCGTCGGTGGCAAGTGCGAGGTGCTGTACGA
CATCGCCCCTCTGTCTGACTTCGTCATCCACC
GTTCACCCGAACTGGTGCCAATGCCTACCCTGAAGGGAGACGGTAGGCACATG
GAGGTCATCAAGATCAAGAACTTCGACAACTG
CGACCAGAGAATCAACTACCACTTCGGTATGACTGACAACAGCCGCCTGGAAC
CAGGTACCAACAAGAACGGCAAGTTCTTCAGC
CGCTCCAGCACTTCTCGTATCGTGATCTCCGAGAGCCTGAAGCACTTCACCATC
CAATCTTCAGTCACCACTTCAAAGATGATGG
TGTCCCCTAGGCTGTACGACAGACAGAACGGTCTGGTCCTGTCCCGTATGAACC
TGACTCTGGCTAAGATGGAAAAGACCTCTAA
GCCCCTGCCAATGGTCGACAACCCTGAGTCAACTGGCAACCTGGTGTACATCTA
CAACAACCCCTTCAGCGACGTCGAGGAACGC
CGTGTGAGCAAGACCGCCATGAACTCTAACCAGATCGTGTCAGACAACTCCCTG
TCCAGCTCTGAGGAAAAGCTGAAACAGGACA
TCCTGAACCTGAGGACTGACATCTCATCCAGCTCTTCATCCATCAGCTCTTCAGA
GGAAAACGACTTCTGGCAGCCTAAGCCCAC
CCTGGAGGACGCTCCACAGAACTCCCTGCTGCCTAACTTCGTGGGCTACAAGGG
AAAGCACATCGGCAAGAGCGGCAAGGTGGAC
GTCATCAACGCTGCCAAGGAACTGATCTTCCAGATCGCTAACGAACTGGAAGA
CGCCTCCAACATCCCAGTCCACGCCACTCTGG
AGAAGTTCATGATCCTGTGCAACCTGATGCGCACCATGAACCGTAAGCAGATCT
CAGAGCTGGAATCCAACATGCAGATCTCTCC TAACGAACTGAAGCCCAACGACAAGTCACAGGTCATCAAGCAGAACACCTGGA
CTGTGTTCAGAGACGCTATCACCCAGACTGGC
ACCGGACCTGCCTTCCTGACTATCAAGGAATGGATCGAGCGCGGTACCACTAAG
TCTATGGAGGCTGCCAACATCATGTCAAAGC
TGCCCAAGACCGTGAGGACTCCAACCGACAGCTACATCAGATCTTTCTTCGAAC
TGCTGCAGAACCCTAAGGTGTCCAACGAGCA
GTTCCTGAACACTGCTGCCACCCTGAGCTTCTGCGAGATGATCCACAACGCTCA
GGTCAACAAGAGAAGCATCCACAACAACTAC
CCCGTGCACACTTTCGGCCGCCTGACCAGCAAGCACGACAACTCTCTGTACGAC
GAATACATCCCTTTCCTGGAGAGGGAACTGA
GAAAGGCCCACCAGGAGAAGGACTCCCCCCGTATCCAGACCTACATCATGGCT
CTGGGAATGATCGGTGAACCAAAGATCCTGAG
CGTGTTCGAACCTTACCTGGAGGGAAAGCAGCAGATGACTGTCTTCCAGAGGA
CCCTGATGGTGGGCTCTCTGGGAAAGCTGACT
GAAACCAACCCCAAGCTGGCCCGTTCTGTCCTGTACAAGATCTACCTGAACACT
ATGGAATCACACGAGGTCAGGTGCACTGCTG
TGTTCCTGCTGATGAAGACCAACCCTCCCCTGTCAATGCTGCAGCGTATGGCCG
AGTTCACTAAGCTGGACACCAACAGGCAGGT
CAACTCAGCTGTGAAGTCCACCATCCAGAGCCTGATGAAGCTGAAGTCCCCAG
AGTGGAAGGACCTGGCTAAGAAGGCCAGAAGC
GTGAACCACCTGCTGACTCACCACGAATACGACTACGAGCTGTCCCGCGGCTAC
ATCGACGAAAAGATCCTGGAGAACCAGAACA
TCATCACCCACATGATCCTGAACTACGTCGGCAGCGAGGACTCTGTGATCCCAC
GCATCCTGTACCTGACTTGGTACTCCAGCAA
CGGAGACATCAAGGTCCCTTCTACCAAGGTGCTGGCTATGATCTCTTCAGTCAA
GTCATTCATGGAACTGTCACTGAGGTCCGTG
AAGGACAGAGAAACCATCATCAGCGCTGCCGAGAAGATCGCCGAGGAACTGAA
GATCGTCCCAGAGGAACTGGTGCCTCTGGAGG
GCAACCTGATGATCAACAACAAGTACGCTCTGAAGTTCTTCCCATTCGACAAGC ACATCCTGGACAAGCTGCCTACTCTGATCTC
CAACTACATCGAAGCCGTCAAGGAGGGAAAGTTCATGAACGTGAACATGCTGG
ACACCTACGAAAGCGTGCACTCTTTCCCTACT
GAAACCGGACTGCCCTTCGTCTACACTTTCAACGTGATCAAGCTGACTAAGACC
AGCGGTACCGTCCAGGCTCAGATCAACCCTG
ACTTCGCCTTCATCGTGAACTCCAACCTGAGGCTGACTTTCAGCAAGAACGTCC
AGGGTAGAGTCGGCTTCGTGACCCCCTTCGA
GCACCGCCACTTCATCTCTGGTATCGACTCAAACCTGCACGTCTACGCTCCCCTG
AAGATCTCCCTGGACGTGAACACCCCAAAG
GGAAACATGCAGTGGAAGATCTGGCCTATGAAGGGAGAGGAAAAGTCAAGACT
GTTCCACTACTCCGTGGTCCCCTTCGTGAGCA
ACCACGACATCCTGAACCTGCGCCCACTGTCCATGGAAAAGGGTACTCGTCCCA
TGATCCCAGACGACAACACCAGCCTGGCCCT
GCCCAAGAACGAAGGCCCATTCCGCCTGAACGTCGAGACTGCTAAGACCAACG
AGGAAATGTGGGAACTGATCGACACTGAGAAG
CTGACCGACAGACTGCCATACCCTTGGACCATGGACAACGAGCGCTACGTGAA
GGTCGACATGTACATGAACCTGGAGGGCGAAC
AGAAGGACCCCGTCATCTTCTCTACTTCATTCGACTCCAAGGTCATGACTCGTCC
AGACACCGACAGCGAAAACTGGACCCCTAA
GATGATGGCTGTGGAGCCCACTGACAAGCAGGCCAACTCTAAGACCAGGAGAC
AGGAAATGATGAGGGAGGCTGGTAGAGGCATC
GAGTCAGCCAAGTCCTACGTGGTCGACGTGAGAGTCCACGTGCCCGGAGAGTC
CGAAAGCGAGACTGTCCTGACCCTGGCTTGGT
CTGAATCAAACGTGGAGTCTAAGGGAAGACTGCTGGGATTCTGGAGAGTGGAA
ATGCCACGTTCAAACGCCGACTACGAGGTCTG
CATCGGCTCACAGATCATGGTGTCCCCAGAAACCCTGCTGTCCTACGACGAGAA
GATGGACCAGAAGCCTAAGATGGACTTCAAC
GTCGACATCCGTTACGGAAAGAACTGCGGAAAGGGAGAGAGGATCGACATGAA
CGGCAAGCTGCGCCAGTCCCCTCGTCTGAAGG AACTGGTCGGCGCTACTAGCATCATCAAGGACTGCGTGGAGGACATGAAGCGC
GGTAACAAGATCCTGCGTACCTGCCAGAAGGC
CGTGGTCCTGTCTATGCTGCTGGACGAAGTCGACATCAGCATGGAGGTGCCCTC
TGACGCTCTGATCGCCCTGTACTCACAGGGA
CTGTTCTCCCTGAGCGAAATCGACAACCTGGACGTCTCCCTGGACGTGAGCAAC
CCAAAGAACGCTGGCAAGAAGAAGATCGACG
TGCGCGCCAAGCTGAACGAGTACCTGGACAAGGCTGACGTCATCGTGAACACT
CCTATCATGGACGCCCACTTCAAGGACGTGAA
GCTGTCAGACTTCGGCTTCTCCACTGAAGACATCCTGGACACCGCTGACGAGGA
CCTGCTGATCAACAACGTCTTCTACGAAGAC
GAAACCTCCTGCATGCTGGACAAGACTCGTGCCCAGACCTTCGACGGAAAGGA
CTACCCTCTGAGGCTGGGTCCATGCTGGCACG
CTGTGATGACCACTTACCCTCGTATCAACCCCGACAACCACAACGAAAAGCTGC
ACATCCCTAAGGACAAGTCTGTCTCAGTGCT
GTCCAGGGAAAACGAGGCTGGCCAGAAGGAGGTCAAGGTGCTGCTGGGATCTG
ACAAGATCAAGTTCGTGCCAGGTACCACTTCA
CAGCCTGAAGTCTTCGTGAACGGAGAGAAGATCGTGGTCAGCCGTAACAAGGC
CTACCAGAAGGTCGAGGAAAACGAAATCATCT
TCGAGATCTACAAGATGGGTGACCGCTTCATCGGTCTGACTTCTGACAAGTTCG
ACGTCTCACTGGCCCTGGACGGTGAACGCGT
GATGCTGAAGGCTTCCGAGGACTACAGGTACAGCGTGAGAGGACTGTGCGGTA
ACTTCGACCACGACTCCACCAACGACTTCGTG
GGCCCCAAGAACTGCCTGTTCCGCAAGCCAGAACACTTCGTCGCTAGCTACGCC
CTGATCTCTAACCAGTGCGAGGGAGACTCTC
TGAACGTGGCTAAGTCACTGCAGGACCACGACTGCATCCGCCAGGAGCGTACC
CAGCAGAGAAACGTCATCTCCGACAGCGAATC
TGGTCGCCTGGACACTGAGATGTCTACCTGGGGCTACCACCACAACGTCAACAA
GCACTGCACTATCCACAGAACCCAGGTGAAG
GAAACTGACGACAAGATCTGCTTCACCATGCGCCCCGTGGTCAGCTGCGCTTCT GGATGCACTGCCGTGGAAACCAAGTCAAAGC
CATACAAGTTCCACTGCATGGAAAAGAACGAGGCTGCCATGAAGCTGAAGAAG
CGTATCGAGAAGGGTGCCAACCCCGACCTGTC
ACAGAAGCCAGTCTCCACCACTGAGGAACTGACCGTCCCCTTCGTGTGCAAGGC
TCACCATCATCACCACCACCACCACTAATGAAAGCTT

Claims

WHAT IS CLAIMED IS:
1. A method to isolate a honey bee antigen comprising: contacting recombinant vitellogenin (rVg) with a solid support to form rVg to the solid support; contacting a solubilized inactivated honey bee pathogen with the rVg bound to the solid support; eluting the inactivated honey bee pathogens to isolate the honey bee antigen.
2. The method of claim 1, further comprising determining the identity of the isolated inactivated honey bee pathogens.
2. The method of claim 1, wherein the rVg is produced in an insect expression system.
3. The method of claim 2, wherein the expression system is a baculovirus -insect cell system.
4. The method of claim 1 or 2, wherein the rVg has the amino acid sequence of SEQ ID NO: 1, or an equivalent thereof.
5. The method of any one of claims 1 to 4, wherein the solid support is selected from a chromatographic support, a sepharose CL column, or an ion exchange column.
6. The method of any one of claims 1 to 5, wherein the honey bee pathogens are inactivated by sonication or solubilization in non-ionic detergents.
7. The method of any one of claims 1 to 6, wherein the honey bee pathogen is selected from one or more of Paenibacillus larvae, Paenibacillus alvei, Paenibacillus dendritiformis Paenibacillus amylolyticus, Paenibacillus campinasensis, Paenibacillus chondr oitinus, Paenibacillus chungangensis, Paenibacillus doosanensis, Paenibacillus glucanolyticus, Paenibacillus humicus, Paenibacillus lactis, Paenibacillus lautus, Paenibacillus lentimorbus, Paenibacillus maceran, Paenibacillus macerans-hke, Paenibacillus macquariensis, Paenibacillus motobuensis, Paenibacillus pabuli, Paenibacillus phoenicis, Paenibacillus polymyxa, Paenibacillus popilliae, Paenibacillus puldeungensis, Paenibacillus residui, Paenibacillus stellife, Paenibacillus thiaminolyticus, Paenibacillus validus, or Paenibacillus xylanisolvens .
8. The method of claim 7, wherein more than one honey bee pathogen is contacted with the column.
38
9. The method of any one of claims 1 to 8, wherein the identity of the isolated inactivated honey bee pathogens are determined by a method comprising a combination electrophoresis, high performance liquid chromatography, mass spectrometry, and/or protein sequencing.
10. A method to identify a honey bee antigen comprising: binding an anti-His tag or anti-Vg antibody with a solubilized inactivated honey bee pathogen and detecting any antibody bound to the inactivated honey bee pathogen, wherein the antibody or inactivated honey bee pathogen is detectably labeled.
11. The method of claim 10, further comprising determining the identity of the isolated inactivated honey bee pathogens.
12. The method of claim 10 or 11, wherein the rVg is produced in an insect expression system, optionally baculovirus-Sf9.
13. The method of claim 12, wherein the insect is a baculovirus.
14. The method of claim 11 or 12, wherein the rVg has the amino acid sequence of SEQ ID NO: 1, or an equivalent thereof.
15. The method of any one of claims 10 to 14, wherein the honey bee pathogens are inactivated by sonication or solubilization in non-ionic detergents.
16. The method of any one of claims 10 to 15, wherein the honey bee pathogen is selected from one or more of Paenibacillus larvae, Paenibacillus alvei, Paenibacillus dendritiformis Paenibacillus amylolyticus, Paenibacillus campinasensis, Paenibacillus chondr oitinus, Paenibacillus chungangensis, Paenibacillus doosanensis, Paenibacillus glucanolyticus, Paenibacillus humicus, Paenibacillus lactis, Paenibacillus lautus, Paenibacillus lentimorbus, Paenibacillus maceran, Paenibacillus macerans-W , Paenibacillus macquariensis, Paenibacillus motobuensis, Paenibacillus pabuli, Paenibacillus phoenicis, Paenibacillus polymyxa, Paenibacillus popilliae, Paenibacillus puldeungensis, Paenibacillus residui, Paenibacillus stellife, Paenibacillus thiaminolyticus, Paenibacillus validus, or Paenibacillus xylanisolvens.
17. The method of any one of claims 10 to 16, wherein the identity of the isolated inactivated honey bee pathogen is determined by a method comprising a combination of electrophoresis, high performance liquid chromatography, mass spectrometry, and/or protein sequencing.
39
18. The method of any of claims 1 to 17, wherein the inactivated honey bee pathogen is detectably labeled.
40
PCT/US2021/048727 2020-09-03 2021-09-01 Use of vitellogenin for defining and testing novel immunogens in insects WO2022051406A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US18/043,723 US20240018183A1 (en) 2020-09-03 2021-09-01 Use of vitellogenin for defining and testing novel immunogens in insects
EP21865059.6A EP4208150A4 (en) 2020-09-03 2021-09-01 Use of vitellogenin for defining and testing novel immunogens in insects

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202063074365P 2020-09-03 2020-09-03
US63/074,365 2020-09-03

Publications (1)

Publication Number Publication Date
WO2022051406A1 true WO2022051406A1 (en) 2022-03-10

Family

ID=80491476

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2021/048727 WO2022051406A1 (en) 2020-09-03 2021-09-01 Use of vitellogenin for defining and testing novel immunogens in insects

Country Status (3)

Country Link
US (1) US20240018183A1 (en)
EP (1) EP4208150A4 (en)
WO (1) WO2022051406A1 (en)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160032252A1 (en) * 2013-04-15 2016-02-04 The University Of Warwick Strain of deformed wing virus (dwv)

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3325007A1 (en) * 2015-07-24 2018-05-30 Helsingin Yliopisto Edible vaccination against microbial pathogens

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160032252A1 (en) * 2013-04-15 2016-02-04 The University Of Warwick Strain of deformed wing virus (dwv)

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
MARHEINEKE, K ET AL.: "Lipid Composition of Spodoptera Frugiperda (sf9) and Trichoplusia Ni (tn) Insect Cells Used for Baculovirus Infection", FEBS LETTERS., vol. 441, no. 1, 11 December 1998 (1998-12-11), pages 49 - 52, XP071238590, DOI: 10.1016/s0014-5793(98)01523-3 *
NOSE YOSHIAKI, MIN LEE JAE, UENO TAKATOSHI, HATAKEYAMA MASATSUGU, OISHI KUGAO: "Cloning of cDNA for vitellogenin of the parasitoid wasp, Pimpla nipponica (Hymenoptera: Apocrita: Ichneumonidae): vitellogenin primary structure and evolutionary considerations", INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY, vol. 27, no. 12, December 1997 (1997-12-01), pages 1047 - 1056, XP055914494, DOI: 10.1016/s0965-1748(97)00091-x *
See also references of EP4208150A4 *

Also Published As

Publication number Publication date
US20240018183A1 (en) 2024-01-18
EP4208150A1 (en) 2023-07-12
EP4208150A4 (en) 2024-04-24

Similar Documents

Publication Publication Date Title
Jurat-Fuentes et al. Reduced levels of membrane-bound alkaline phosphatase are common to lepidopteran strains resistant to Cry toxins from Bacillus thuringiensis
Hansen et al. Transcription properties of a cell type–specific TATA-binding protein, TRF
Houdusse et al. The many roles of myosins in filopodia, microvilli and stereocilia
Qin et al. Cloning and characterization of a member of the hsp70 gene family from Locusta migratoria, a highly thermotolerant insect
Hirayama et al. The occurrence of two types of hemopexin-like protein in medaka and differences in their affinity to heme
Egge et al. Age-onset phosphorylation of a minor actin variant promotes intestinal barrier dysfunction
Huang et al. The mitochondrial Ca2+ uniporter complex (MCUC) of Trypanosoma brucei is a hetero-oligomer that contains novel subunits essential for Ca2+ uptake
KR20040088077A (en) Promotion or Inhibition of Angiogenesis and Cardiovascularization
Gim et al. Drosophila Med6 is required for elevated expression of a large but distinct set of developmentally regulated genes
Gal et al. Alterations in the levels of glycogen and glycogen synthase transcripts during desiccation in the insect-killing nematode Steinernema feltiae IS-6
Chen et al. Quantitative expression analysis of a Glyptapanteles indiensis polydnavirus protein tyrosine phosphatase gene in its natural lepidopteran host, Lymantria dispar
US20240018183A1 (en) Use of vitellogenin for defining and testing novel immunogens in insects
Christophides et al. Two medfly promoters that have originated by recent gene duplication drive distinct sex, tissue and temporal expression patterns
Prag et al. Molecular analysis of muskelin identifies a conserved discoidin-like domain that contributes to protein self-association
Okumura‐Noji et al. CD36‐related protein in Schistosoma japonicum: candidate mediator of selective cholesteryl ester uptake from high‐density lipoprotein for egg maturation
Chen et al. NHE1 gene associated with avian leukosis virus subgroup J infection in chicken
Zhang et al. Molecular and immunohistochemical analyses of cardiac troponin T during cardiac development in the Mexican axolotl, Ambystoma mexicanum
Kou et al. Identification of β‐chain of FoF1‐ATPase in apoptotic cell population induced by Microplitis bicoloratus bracovirus and its role in the development of Spodoptera litura
Saunders et al. Expression of GIMAP1, a GTPase of the immunity-associated protein family, is not up-regulated in malaria
KR20220021906A (en) gene therapy monitoring
Liu et al. Expression, purification, and enzymatic characterization of Bombyx mori nucleopolyhedrovirus DNA polymerase
Yamada et al. The endosomal TbTpr86/TbUsp7/SkpZ (TUS) complex controls surface protein abundance in trypanosomes
RU2813731C2 (en) New fish coronavirus
Cao et al. Transfer of the zp3a gene results in changes in egg adhesiveness and buoyancy in transgenic zebrafish
JP4606766B2 (en) Therapeutic or preventive agent for diseases related to glucose metabolism

Legal Events

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

Ref document number: 21865059

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2021865059

Country of ref document: EP

Effective date: 20230403