Skip to main content
Advertisement
  • Loading metrics

Anthrax toxin component, Protective Antigen, protects insects from bacterial infections

  • Saleem Alameh ,

    Contributed equally to this work with: Saleem Alameh, Gloria Bartolo

    Roles Formal analysis, Investigation, Methodology, Validation, Writing – original draft, Writing – review & editing

    Affiliation Henry E. Riggs School of Applied Life Sciences, Keck Graduate Institute, Claremont, California, United States of America

  • Gloria Bartolo ,

    Contributed equally to this work with: Saleem Alameh, Gloria Bartolo

    Roles Formal analysis, Investigation, Methodology, Validation, Writing – review & editing

    Affiliation Henry E. Riggs School of Applied Life Sciences, Keck Graduate Institute, Claremont, California, United States of America

  • Summer O’Brien,

    Roles Formal analysis, Investigation, Methodology, Validation, Writing – review & editing

    Affiliation Henry E. Riggs School of Applied Life Sciences, Keck Graduate Institute, Claremont, California, United States of America

  • Elizabeth A. Henderson,

    Roles Formal analysis, Investigation, Methodology, Validation, Writing – review & editing

    Affiliation Henry E. Riggs School of Applied Life Sciences, Keck Graduate Institute, Claremont, California, United States of America

  • Leandra O. Gonzalez,

    Roles Formal analysis, Investigation, Methodology, Validation, Writing – review & editing

    Affiliation Henry E. Riggs School of Applied Life Sciences, Keck Graduate Institute, Claremont, California, United States of America

  • Stella Hartmann,

    Roles Formal analysis, Investigation, Methodology, Validation, Writing – review & editing

    Affiliation Henry E. Riggs School of Applied Life Sciences, Keck Graduate Institute, Claremont, California, United States of America

  • Christopher P. Klimko,

    Roles Formal analysis, Investigation, Methodology, Validation, Writing – review & editing

    Affiliation Bacteriology Division, US Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland, United States of America

  • Jennifer L. Shoe,

    Roles Formal analysis, Investigation, Methodology, Validation, Writing – review & editing

    Affiliation Bacteriology Division, US Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland, United States of America

  • Christopher K. Cote,

    Roles Formal analysis, Investigation, Methodology, Resources, Supervision, Validation, Writing – review & editing

    Affiliation Bacteriology Division, US Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland, United States of America

  • Laurence K. Grill,

    Roles Formal analysis, Methodology, Resources, Supervision, Writing – review & editing

    Affiliation Henry E. Riggs School of Applied Life Sciences, Keck Graduate Institute, Claremont, California, United States of America

  • Anastasia Levitin ,

    Roles Conceptualization, Data curation, Formal analysis, Methodology, Project administration, Resources, Supervision, Writing – review & editing

    anastasia_levitin@kgi.edu (AL); mikhail_shilman@kgi.edu (MMS)

    Affiliation Henry E. Riggs School of Applied Life Sciences, Keck Graduate Institute, Claremont, California, United States of America

  • Mikhail Martchenko Shilman

    Roles Conceptualization, Data curation, Formal analysis, Funding acquisition, Methodology, Project administration, Resources, Supervision, Writing – original draft, Writing – review & editing

    anastasia_levitin@kgi.edu (AL); mikhail_shilman@kgi.edu (MMS)

    Affiliation Henry E. Riggs School of Applied Life Sciences, Keck Graduate Institute, Claremont, California, United States of America

Abstract

Anthrax is a major zoonotic disease of wildlife, and in places like West Africa, it can be caused by Bacillus anthracis in arid nonsylvatic savannahs, and by B. cereus biovar anthracis (Bcbva) in sylvatic rainforests. Bcbva-caused anthrax has been implicated in as much as 38% of mortality in rainforest ecosystems, where insects can enhance the transmission of anthrax-causing bacteria. While anthrax is well-characterized in mammals, its transmission by insects points to an unidentified anthrax-resistance mechanism in its vectors. In mammals, a secreted anthrax toxin component, 83 kDa Protective Antigen (PA83), binds to cell-surface receptors and is cleaved by furin into an evolutionary-conserved PA20 and a pore-forming PA63 subunits. We show that PA20 increases the resistance of Drosophila flies and Culex mosquitoes to bacterial challenges, without directly affecting the bacterial growth. We further show that the PA83 loop known to be cleaved by furin to release PA20 from PA63 is, in part, responsible for the PA20-mediated protection. We found that PA20 binds directly to the Toll activating peptidoglycan-recognition protein-SA (PGRP-SA) and that the Toll/NF-κB pathway is necessary for the PA20-mediated protection of infected flies. This effect of PA20 on innate immunity may also exist in mammals: we show that PA20 binds to human PGRP-SA ortholog. Moreover, the constitutive activity of Imd/NF-κB pathway in MAPKK Dsor1 mutant flies is sufficient to confer the protection from bacterial infections in a manner that is independent of PA20 treatment. Lastly, Clostridium septicum alpha toxin protects flies from anthrax-causing bacteria, showing that other pathogens may help insects resist anthrax. The mechanism of anthrax resistance in insects has direct implications on insect-mediated anthrax transmission for wildlife management, and with potential for applications, such as reducing the sensitivity of pollinating insects to bacterial pathogens.

Author summary

Gram-positive Bacillus anthracis and B. cereus biovar anthracis cause anthrax in mammals by secreting anthrax toxin. Recent studies report the devastating ecological effects anthrax has on the wildlife in areas like tropical rainforests, where in the past 26 years, anthrax had been implicated in 38% of wildlife mortality, and where insects can act as the vectors for anthrax transmission. The anthrax-resistance mechanism in its insect vectors has not been identified. We discovered that one of anthrax toxin components reduces the sensitivity of insects to anthrax-causing bacilli and other bacteria, and that insect immunity Toll/NF-κB and Imd/NF-κB pathways play a role in this toxin-mediated protection. The toxin-mediated protection of insects would benefit the anthrax-causing bacteria by extending the lifespan of vectors and enhancing the pathogen transmission. Anthrax toxin component activation of NF-κB has also been found to exists in humans. Moreover, the activation of the innate NF-κB-dependent immunity by this anthrax toxin component has potential implications in protecting pollinating insects from bacterial pathogens.

Introduction

Anthrax is a major zoonotic disease of wildlife caused by the Gram-positive bacterium Bacillus anthracis or by the closely related B. cereus biovar anthracis (Bcbva), which combines the chromosomal background of B. cereus with the toxin-encoding pXO1 and the capsule-encoding pXO2 plasmids of B. anthracis [1]. Bcbva causes “sylvatic anthrax”, a prevalent and persistent cause of death in a broad range of mammalian hosts in the rainforest ecosystem of Taï National Park (TNP), Côte d'Ivoire. Sylvatic anthrax was responsible for as much as 38% of wildlife mortality observed over 26 years in TNP and is predicted to accelerate the decline and possibly result in the local extinction of chimpanzee populations [2, 3]. Moreover, viable spores of Bcbva were detected in blow flies sampled inside and outside the chimpanzee habitats in TNP [2]. In addition to TNP, Bcbva has been isolated from dead or sick mammals in other West and Central Africa regions, such as Cameroon, Central African Republic, and Democratic Republic of Congo [4], as well as in regions outside of Africa, such as Texas and Louisiana, USA [5].

In contrast, B. anthracis, which is distributed globally [6], causes “classical anthrax” that is most commonly observed in arid nonsylvatic ecosystems, such as African savannahs in Krüger National Park [7], South Africa and Etosha National Park, Namibia [8, 9]. In these ecosystems, major anthrax outbreaks display strong seasonal variation, where the mortality coincides with rainfall cycles and the wet season [79]. In these nonsylvatic ecosystems, it has also been suggested that flies spread B. anthracis spores from carcasses through the environment, potentially contributing to anthrax transmission [10, 11].

In nature, insects can serve as the vectors for mechanical anthrax transmission to humans, cattle, and the mammalian wildlife [2, 6, 10, 1214]. Biting flies have been shown to acquire anthrax-causing bacilli from infected animals and directly transmit them to other mammals [14]. Similarly, non-biting flies feed on the bodily fluids of infected carcasses and deposit contaminated feces or vomit on nearby vegetation, creating a reservoir for herbivores to potentially contract anthrax while grazing [7]. Other insects, such as mosquitoes and ticks, may also contribute to anthrax transmission [1517], raising the interesting question as to how insect vectors tolerate anthrax-mediated toxicity.

Insects depend on their innate immunity for protection against pathogens. During an infection, insects produce antimicrobial peptides (AMP), while circulating hemocytes phagocytose invading microbes [18]. Two classical signaling pathways, the Toll and the immune deficiency (Imd) pathways, mediate broad-spectrum AMP responses. The Toll pathway is primarily activated by lysine (lys)-type peptidoglycan found in most Gram-positive bacterial cell walls, which interacts with insect PGRP-SA [19]. In contrast, the Imd pathway is mostly activated in response to diaminopimelate (dap)-type peptidoglycans found in all Gram-negative bacteria and Gram-positive Bacillus species, which interact with insect PGRP-LC and PGRP-LE [20]. Interestingly, it was recently shown that the sensing of the bacterial cell wall type is not as pathway-specific as previously thought: as long as the bacterial wall is accessible, both Toll and Imd pathways can detect lys- and dap-peptidoglycan, since dap is a derivative of lys [21]. Activation of these pathways leads to the induction of nuclear factor-κB (NF-κB) transcription factors, which promote the transcription of AMPs, whose expression levels could be regulated by both pathways, and with many of them showing broad-spectrum antimicrobial activity [2225].

Both Bcbva and B. anthracis secrete the toxin encoded by plasmid pXO1, which can ultimately kill mammals [2]. The pXO1 plasmid encodes three toxin components that injure mammalian cells: protective antigen (PA), lethal factor (LF), and edema factor (EF) [26]. During anthrax infection, 83 kDa PA (PA83) binds to mammalian cell-surface receptors and is cleaved by host furin proteases into 20 kDa (PA20) and 63 kDa (PA63) subunits [27]. Following the cleavage, PA20 dissociates from PA63, and receptor-bound PA63 multimerize, forming a pre-pore, which binds EF and LF before endocytosis [28, 29]. In the acidified endosome, the PA63 pore allows EF and LF to escape into the cytosol to exert their deleterious effects [30]. Although PA20 is evolutionary-conserved [31], its function is poorly understood: in addition to blocking the multimerization of PA83, it may play a role in activating adaptive and innate immunity responses. During anthrax, the major adaptive immune response is mounted against PA83, and several epitopes within PA20 have been identified in PA83-immunized mammalian hosts [3235]. In addition to inducing adaptive immunity, PA20 may also activate innate immunity: the host cell exposure to PA83 or PA20 was shown to activate NF-κB and pro-inflammatory cytokines, such as Interleukins [3638].

It was previously observed that several insect species, such as blow flies, horn flies, house flies, and mosquitoes are insensitive to B. anthracis oral exposure [11, 15, 17, 39]. In contrast, insects are sensitive to non-anthracis strains of closely related B. cereus [4042]. Therefore, we hypothesize that anthrax toxin components may contribute to insects’ resistance to bacilli, as insects feed on anthrax-infected mammals or their carcasses. Previous studies have established Drosophila melanogaster as a fly model to study the effects of genetically expressed LF and EF [43]. Here, we discovered that when administered orally, PA20 reduces the sensitivity of Drosophila flies and Culex mosquitoes to insecticidal bacteria, and further study the mechanism by which insects resist anthrax. We propose that this toxin-dependent phenomenon may provide insects with a counterbalance to the adverse effects of insecticidal bacteria, thereby facilitating the spread of anthrax.

Results

PA83 protects infected flies from B. cereus infection

B. anthracis and Bcbva belong to a sensu lato group of bacteria, called B. cereus, which comprises seven closely related species that includes B. cereus sensu stricto [44] (referred to herein as B. cereus). The phylogenetic cluster that includes B. anthracis and Bcbva also includes B. cereus strain ATCC 10987 [44, 45]. While the chromosome of B. cereus strain ATCC 10987 shares close homology to the B. anthracis genome (complete nucleotide sequence identity of 94%) and the Bcbva genome, this strain lacks anthrax toxin orthologues on its pXO1-like plasmid pBc10987 [4446]. This, together with the fact that specific isolates of B. cereus cause sylvatic anthrax in tropical rainforests [14, 46], presented an opportunity to test the effects of anthrax toxin components on the sensitivity of Drosophila to this Bcbva-related bacterium.

When feeding on anthrax-infected mammals, insects can be exposed to anthrax toxin components. We observed that the oral exposure of flies to a mix of purified components PA83, LF, and EF in the absence of the bacterial challenge did not alter their survival (S1 Fig). In the presence of the anthrax toxin components, flies showed increased survival time (quantified as the increased median survival time) to B. cereus infection by 25 hours, P < 0.0001 (Fig 1A). While neither EF nor LF affected the sensitivity of flies to B. cereus (Fig 1B and 1C), PA83 alone delayed the death of infected flies (delay of the median survival time by 26 hours, P < 0.0001) (Fig 1D). In addition to flies aged 4–5 days (Fig 1), the protective effect of PA83 is also observed in flies of various ages (delay of the median survival time by 34 hours, P < 0.0001) (S2 Fig). These results show that protein regions located within PA83 cause the delay in bacilli-induced death in Drosophila.

thumbnail
Fig 1. Anthrax toxin component PA83 protects Drosophila from Bacillus cereus.

The discovery of PA-mediated reduction in sensitivity of flies to B. cereus infection. Male Oregon-R wild type (WT) flies were challenged with B. cereus in the absence or presence of one or more anthrax toxin components, including a combination of all toxin components (A), EF (B), LF (C), and PA83 (D). P values indicate statistical significance compared to the bacteria-only condition.

https://doi.org/10.1371/journal.ppat.1008836.g001

The effect of PA 1β13−1α1 loop and PA20 on insects’ sensitivity to bacterial infections

PA83 contains four functional domains (Fig 2A): the host receptor-binding domains 2 and 4 (PAD2 and PAD4), the multimerization domain 3 (PAD3), and the PA20-containing domain 1 (PAD1) [29]. After furin cleavage, the portion of PAD1 that remains on PA63 is called domain 1’ (PAD1’). To investigate the region within PA83 responsible for the protection of infected flies, we recombinantly expressed all PA83 domains (Fig 2B). We observed that PA20-containing PAD1, but not PAD1’, PAD2, PAD3, nor PAD4, protected flies from B. cereus infection (delay of the median survival time by 37 hours, P < 0.0001) (Fig 2C).

thumbnail
Fig 2. Structure-function analysis of PA83.

(A) Domain organization in the linear and tertiary structure or PA83. The domains and the corresponding amino acids are indicated in the linear structure of PA83. The full tertiary structure of monomeric PA83 is shown using iCn3D (Protein Data Bank ID: 4H2A). The regions in blue, purple, and yellow represent the domains 2, 3, and 4 portions of PA63, while the green region represents the portion of domain 1 (aka domain 1’) that remains part of PA63 after furin cleavage, and the red region represents PA20. Furin cleavage occurs in the 1β13−1α1 loop composed of amino acids 181–200. The amino acid sequence of the PA83 loop 1β13−1α1 connecting PA20 to domain 1’ is also shown. After furin cleavage (the furin cleavage site is shaded gray), amino acids 181–192 are retained by PA20 (shown in the second line labeled PA2013−1α1 loop). (B) Western blot showing the ability of the polyclonal anti-PA83 antibody to recognize recombinantly expressed and purified PA domains 1, 1’, 2, 3, and 4. (C) The effect of the PA domains 1, 1’, 2, 3, and 4 (PAD1, PAD1’, PAD2, PAD3, and PAD4) on the sensitivity of Drosophila to B. cereus. WT flies of both genders were pooled and challenged with B. cereus with or without individual PA domains. (D) The effect of the 1β13−1α1 loop on the sensitivity of Drosophila to B. cereus. WT flies were challenged with B. cereus with or without PA20, PA83 loop 181–200, or PA20 loop 181–192. (E) WT flies were challenged with B. cereus in the absence or presence of furin-processed PA20, PA63, or their combination. P as in Fig 1.

https://doi.org/10.1371/journal.ppat.1008836.g002

Like insects, plants rely only on innate immunity to protect them from pathogens. In plants, programmed cell death, or necrotic lesions, often occur at the site of infection to prevent the systemic spread of invading microorganisms [47]. Previously we observed that amino acids 181–200 of PA83 caused necrotic lesions in Nicotiana benthamiana [48], suggesting that they trigger an innate immune response in plants. This peptide forms an unstructured loop (between β-sheet 13 and α-helix 1, referred to as the “1β13−1α1 loop”) within domain 1 of PA83 [29], which contains the furin-cleavage site [49] (Fig 2A). The loop residues 181–192 remain on PA20 after furin processing. We assessed the ability of the entire loop (residues 181–200) and the furin-cleaved portion (residues 181–192) of the 1β13−1α1 loop to reduce the sensitivity of flies to bacterial infections. To some degree, both peptides delayed the death of B. cereus-infected flies (delay of the median survival time 11 hours, where some flies survive for 29 hours longer, P < 0.0001) (Fig 2D) while also not directly altering bacterial growth (S3 Fig). Therefore, the residues within the PA2013−1α1 loop are, in part, responsible for the PA83-mediated reduction in the sensitivity of flies to bacterial infections.

When feeding on anthrax-infected mammals, insects can be exposed to PA in its native PA83 state, as well as in the furin-cleaved state, PA63 and PA20. Further testing revealed that within PA83, it was PA20 and not PA63 responsible for the reduction in fly sensitivity to B. cereus: PA20 protected infected flies without and with PA63 (delay of the median survival time by 24 hours, P < 0.0001) (Fig 2E).

PA20 protection is broad-spectrum and insect-directed

We tested the sensitivity of D. melanogaster to oral administration of the toxigenic Sterne strain of B. anthracis harboring the pXO1 plasmid, and the ΔSterne strain, a pXO1-cured derivative of the Sterne strain. While flies were sensitive to both B. anthracis strains in the vegetative state, surprisingly, the Sterne strain was less pathogenic than the toxin-negative ΔSterne strain (delay of the median survival time by 21 hours, P < 0.0001) (Fig 3A). Moreover, we investigated the sensitivity of flies to spores of the same B. anthracis strains. Interestingly, while uninfected flies feeding solely on sucrose can survive for 10–14 days, the oral exposure to B. anthracis spores extended the longevity of flies, with the Sterne strain extending the life significantly longer than the ΔSterne strain (delay of the median survival time by 57 hours, P < 0.0001) (Fig 3A). These data show that the toxin-containing strain of B. anthracis is less pathogenic to flies and extends the survival of flies, compared to the toxin-negative strain of bacteria.

thumbnail
Fig 3. PA20 protects Drosophila from Bacillus anthracis.

(A) WT flies were orally challenged with B. anthracis Sterne and plasmid-cured ΔSterne strains in vegetative and spore states. P values in (A) indicate the statistical significance of Sterne vs. ΔSterne for vegetative and spore states. (B-C) WT flies were orally challenged with vegetative cells of the B. anthracis Sterne (B) and ΔSterne (C) strains with or without PA20. P as in Fig 1. (D) Male Culex quinquefasciatus (Culex) were orally challenged with vegetative B. anthracis Sterne or ΔSterne strains with or without PA20. P as in Fig 1. (E) C. quinquefasciatus (Culex) mosquitoes were orally challenged with spores of B. anthracis Sterne or ΔSterne strains.

https://doi.org/10.1371/journal.ppat.1008836.g003

When feeding on anthrax-infected mammals, insects can be exposed to pXO1-containing bacteria, such as Bcbva and B. anthracis cells, as well as secreted toxins components, including PA20. We show that exogenously added PA20 protects flies from the Sterne strain (Fig 3B) and ΔSterne strain (Fig 3C). PA20 delayed the median survival time of Sterne strain infected flies by 19 hours (P < 0.0001) and of ΔSterne strain infected flies by 16 hours (P < 0.0001), although some flies were protected by as long as 25–70 hours.

Mosquitoes may also contribute to anthrax transmission [15, 17] and were previously found to be insensitive to anthrax toxin [50]. We tested whether PA20-mediated protection occurs in B. anthracis-infected mosquitoes, Culex quinquefasciatus. Interestingly while mosquitoes were sensitive to the vegetative Sterne and ΔSterne strains of B. anthracis, in contrast to Drosophila, their sensitivity was similar to both strains (Fig 3D). Moreover, in contrast to our fruit fly experiments, oral exposure to B. anthracis spores did not affect the longevity of mosquitoes (Fig 3E). Nevertheless, PA20 significantly protected Culex from both strains of bacteria and delayed the median survival time of Sterne strain infected mosquitoes by 102 hours (P < 0.0001) (Fig 3D) and of ΔSterne strain infected mosquitoes by 96 hours (P < 0.0001) (Fig 3D). Collectively, these results show that PA20 causes the delay of B. anthracis-induced death in Drosophila and Culex.

The effects of PA20 extend beyond bacteria of the B. cereus group, as it enhanced the survival of flies exposed to related B. subtilis and unrelated Gram-negative Serratia liquefaciens and Escherichia coli. PA20 extended the median survival time of B. subtilis infected flies by 24 hours (P < 0.0001), S. liquefaciens infected flies by 22 hours (P < 0.0001), and E. coli infected flies by 25 hours (P < 0.0001) although some flies were protected by as long as 35–47 hours (Fig 4A–4C). As with the wild-type flies infected with B. cereus (S2 Fig), we observed that a group of flies of various ages infected with S. liquefaciens was protected by PA83 (delay of the median survival time by 152 hours, P < 0.0001) (S4 Fig). To test whether PA20 exerted its protective effects by suppressing the growth of bacteria, we measured the growth rates of bacteria in the presence or the absence of PA20. PA20 did not affect the growth of B. cereus and S. liquefaciens in liquid culture (Fig 4D and 4E) or on solid media (Fig 4F and 4G), suggesting that PA20 reduces the sensitivity of flies to insecticidal bacteria through an insect-oriented mechanism.

thumbnail
Fig 4. PA20 protection is broad-spectrum and insect-directed.

(A-C) The effect of PA20 on the sensitivity of flies to other bacteria. WT flies were infected with B. subtilis (A), S. liquefaciens (B), and E. coli (C) without or with PA20. P as in Fig 1. (D-G) PA20 does not alter the growth of B. cereus and S. liquefaciens. (D-E) The ability of PA20 to affect the rate of B. cereus (D) and S. liquefaciens (E) growth in LB and TSB, respectively. The bacterial growth was measured in a liquid medium in a 96-well plate with and without 1 μg/mL of PA20. Each data point shown indicates the mean ± SD value obtained in triplicate assays done in a representative experiment. (F-G) Agar diffusion susceptibility assay of B. cereus (F) and S. liquefaciens (G). LB and TSB plates were treated with 1 μL of toxins right after the spreading of B. cereus and S. liquefaciens cultures, respectively, and left to incubate overnight. Spots below a-g contain a 1 μL spot of: 1 μg/mL PA83 (a), PA63 (b), PA20 (c), LF (d), EF (e), PBS (f), and 10 mM levofloxacin (g).

https://doi.org/10.1371/journal.ppat.1008836.g004

PGRP-SA binds to PA20 and is necessary for PA20-mediated protection

Lys-type peptidoglycans are recognized by a circulating extracellular heterodimer receptor consisting of peptidoglycan-recognition protein-SA (PGRP-SA) and GNBP1 [19, 51] (Fig 5A). This peptidoglycan binding triggers a proteolytic cascade that includes ModSP, Grass, Sphe, Psh, and SPE [18]. Ultimately, SPE cleaves pro-Spätzle to release the mature Toll ligand, Spätzle (Spz), and trigger the subsequent intracellular Toll signaling leading to the degradation of the inhibitor of κB, cactus, the activation of NF-κB (Dif), and the transcription of AMPs.

thumbnail
Fig 5. Fly PGRP-SA interacts with PA20 and is necessary for PA20-mediated protection.

(A) A model of PA20-mediated phenomenon. During mammalian anthrax infection (left panel), anthrax PA83 binds to host CMG2 and TEM8 cell surface receptors. PA83 is then cleaved by cell surface mammalian furin proteases, yielding PA20 and PA63, with the latter remaining attached to the host cell receptor. PA63 then multimerizes and binds to the circulating LF or EF molecules. This PA pre-pore is endocytosed with the help of clathrin. Upon entry into the cell, the PA pre-pore becomes the PA-pore. This change allows LF and EF to escape into the cell cytosol. Once in the cytosol, EF functions as an adenyl cyclase resulting in edema. Conversely, LF cleaves MAPKK and Nlrp1, causing caspase-mediated pyroptosis. Upon contact with anthrax infected animals or their carcasses, scavenging insects are likely exposed to PA20. PA20, once ingested by Drosophila, may interact with the Toll pathway to reduce the sensitivity of flies to bacterial challenge (center panel). Two pathways trigger the expression of antimicrobial peptides in Drosophila: the Toll and Imd pathways. During the bacterial challenge, Toll is activated by bacterial lys-type peptidoglycan (PGN) by binding to the PGRP-SA/GNBP1 heterodimer and undergoing enzymatic modification. This results in Toll activation by SPZ ligand binding. Concurrently or separately, Toll is also known to be activated by the circulating virulence factor detection protein, Psh, which also works through SPZ to induce Toll activation. The Imd pathway responds primarily to bacterial dap-type peptidoglycans such as that found in B. cereus and S. liquefaciens. Both Toll and Imd ultimately activate NF-κB transcription factors, which induce the transcription of AMPs that act in a broad-spectrum manner. PA20, once ingested by Drosophila, works through Toll and likely PGRP-SA to reduce the sensitivity of flies to bacterial challenge. The effect of PA20 on the Toll/NF-κB pathway may also occur on the homologous mammalian pathway (right panel). The vaccination with PA83 and human cell exposures to PA83 or PA20 are known to induce the Toll-like receptors-mediated activation of NF-κB and inflammatory cytokines, such as Interleukins. (B-C) ELISA assays showing the interaction between D. melanogaster PGRP-SA (B) or human PGRP-S (C) with PA20 or peptidoglycans (PGN). PGRP-SA or PGRP-S are added to wells with immobilized PA20 or PGNs. Anti-T7 antibody is used to detect T7-tagged PGRP-SA, and anti-PGRP-S antibody is used to detect PGRP-S. (D) PGRP-S pull-down assay. PGRP-S binds to and co-precipitates with the partially soluble PA20 or the insoluble B. subtilis peptidoglycan (PGN). The supernatant (s) or pellet (p) samples from the pull-down assay were analyzed by the Western blot using anti-PGRP-S (top) or anti-PA (bottom) polyclonal antibodies. (E) The effect of PA20 on PGRP-SA mutant flies. Drosophila mutants were challenged with B. cereus with or without PA20.

https://doi.org/10.1371/journal.ppat.1008836.g005

The five amino acid consensus sequence of lys-type peptides recognized by PGRP-SA is AQKA/SA/S [52]. The PA20 portion of the 1β13−1α1 loop contains a similar sequence: 186-KQKSS-190. To test whether the protective capacity of PA20 might be due to its ability to bind to PGRP-SA (Fig 5A), we used purified recombinant D. melanogaster PGRP-SA and its human orthologue PGRP-S, as they share 60% sequence homology and the peptidoglycan-binding site is closely conserved between the two proteins [53, 54]. Moreover, just like PGRP-SA, PGRP-S binds to lys- and dap-type peptidoglycans (dap is a derivative of lys) [54]. We performed an ELISA, where PRGP-SA or PGRP-S was added to plates coated with PA20 or peptidoglycans. PRGP-SA (Fig 5B) and PGRP-S (Fig 5C) bound PA20 and peptidoglycans and were detected with anti-PGRP antibodies.

To validate this observation, we analyzed the ability of PGRP-S to bind to PA20 in a co-precipitation assay, which relied on the observation that dap-type peptidoglycan and PA20 are partially insoluble. We incubated PGRP-S with either peptidoglycan or PA20, followed by centrifugation and washing of pellets. Western blot analysis demonstrated that PGRP-S bound to PA20 and peptidoglycan, and thus, was pulled down into pellets (Fig 5D). Moreover, PA20 did not affect the sensitivity of PGRP-SA loss-of-function mutant flies to B. cereus, showing that this gene is necessary for the PA20-mediated phenomenon (Fig 5E). Collectively, these results suggest that PA20 may exert its protective effects in Drosophila by binding to PGRP-SA (Fig 5A).

The Toll pathway is necessary, and the Imd pathway is sufficient for PA20-mediated protection

We investigated whether the Toll pathway, the Imd pathway, and the phagosomal ROS production are involved in PA20-mediated protection of infected flies. Toll pathway GNBP1, ModSP, Grass, Sphe, Psh, SPE, Spz, and cactus loss-of-function mutants were infected with B. cereus in the absence and the presence of PA20 (Fig 6A–6H). PA20 did not protect any of the Toll-pathway mutants infected with B. cereus. We confirmed the inability of PA20 to protect SPE and Spz mutants challenged with S. liquefaciens (S5A and S5B Fig). We tested the ability of a known immunosuppressive inhibitor of NF-κB, cortisone acetate [55], to sensitize flies to B. cereus and suppress PA20-mediated protection. The addition of a minimal immunosuppressive concentration of cortisone acetate (20 mM) to the feeding medium increased the sensitivity of Drosophila to B. cereus and negated the protective effects of PA20 (Fig 6I and S6 Fig) without affecting the growth of B. cereus (S3 Fig).

thumbnail
Fig 6. The effect of the Toll and Imd pathways and phagosomal ROS on PA20-mediated protection.

The effect of PA20 on the Toll pathway mutants. Gram-Negative Bacteria Binding Protein 1 (GNBP1) (A), Modular Serine Protease (ModSP) (B), Gram-positive Specific Serine protease (Grass) (C), Spheroide (Sphe) (D), Persephone (Psh) (E), Spätzle Processing Enzyme (SPE) (F), Spätzle (Spz) (G), and cactus (H). Drosophila mutants were challenged with B. cereus with or without PA20. WT (I) and (J) mutant flies were infected with B. cereus, without or with 1 μg/mL PA20 or 20 mM Cortisone acetate. Female WT and Dsor1 (K) mutant flies were tested with and without PA20. P as in Fig 1. (L) Measuring reactive oxygen species (ROS) consisting of hydrogen peroxide, peroxynitrite, hydroxyl radicals, nitric oxide, and peroxy radicals in Drosophila macrophage-like plasmatocytes S2 cells in the absence and the presence of PA20. Radicals were measured using ROS-ID Total ROS/Superoxide Detection Kit (Enzo Life Sciences). Radicals produced by S2 cells were measured in the absence or the presence of B. cereus and various concentrations of PA20 (0.25 to 20 μg/mL).

https://doi.org/10.1371/journal.ppat.1008836.g006

We evaluated whether Imd is necessary for PA20-mediated protection by testing the ability of PA20 to reduce the sensitivity of Imd-deficient mutant flies to bacterial infections. The results revealed that PA20 was able to delay B. cereus-induced (Fig 6J) and S. liquefaciens-induced (S5C Fig) mortality of Imd mutants.

We then evaluated whether the activity of the Imd pathway is sufficient for PA20-mediated protection. One of D. melanogaster’s mitogen-activated protein kinase kinases, Dsor1, acts as a suppressor of the Imd pathway, and the downregulation of Dsor1 mimics the induction of the Imd pathway by microbes, even in the absence of an immune challenge [56]. Dsor1 blocks the Imd pathway by activating Pirk, which, in turn, interacts directly with PGRP-LC and PGRP-LE and disrupts their interaction with Imd [57] (Fig 5A). We tested the ability of PA20 to reduce the sensitivity of Dsor1 mutant flies to B. cereus infection. We observed that these mutant flies are less sensitive to B. cereus infection in the absence of PA20, and that the addition of PA20 has no effect on Dsor1 mutant fly sensitivity (Fig 6K). We confirmed the inability of PA83 and PA20 to protect Dsor1 mutant flies infected with S. liquefaciens (S5D and S5E Fig). Collectively, these data demonstrate that while Imd is not necessary for the PA20-mediated phenomenon, the constitutive activity of the Imd/NF-κB pathway in Dsor1 mutant flies is sufficient to negate the protective effects of PA20.

We examined whether PA20 induced the production of reactive oxygen species (ROS) in Drosophila macrophage-like plasmatocytes, S2 cells. We measured the total ROS and separately superoxide radicals produced by S2 in the absence or the presence of B. cereus and various concentrations of PA20 (0.25 to 20 μg/mL) (Fig 6L and S7 Fig). Although B. cereus increased the abundance of ROS in S2, the addition of PA20 did not further alter the levels of these species. Our overall results demonstrate that the Toll pathway is necessary and the Imd pathway is sufficient but not necessary for the PA20-mediated phenomenon, and that PA20 does not affect phagosomal ROS production.

Toxins of other bacterial pathogens may help flies resist anthrax

During the infection, anthrax-causing bacilli may co-exist with other environmental and commensal pathogenic agents. Consequently, anthrax-exposed insects can also be co-exposed to toxins of other bacteria. We investigated whether exotoxins of eight pathogenic bacteria affect the sensitivity of flies to B. anthracis Sterne strain (Fig 7A–7K). We observed that Clostridium septicum alpha toxin reduces the sensitivity of flies to B. anthracis Sterne strain infection (delay of the median survival time by 43 hours, P < 0.0001) (Fig 7A), demonstrating that toxins from other microorganisms may help flies resist anthrax.

thumbnail
Fig 7. The effect of various bacterial toxins on the sensitivity of Drosophila to Bacillus anthracis Sterne strain.

Male Oregon-R wild type (WT) flies were orally challenged with vegetative B. anthracis Sterne strain in the absence or presence of bacterial toxins: Clostridium septicum alpha toxin (A), C. difficile binary toxin subunit A (B) and toxins B (C) and A (D), C. perfringens epsilon toxin (E), pertussis toxin (F), botulinum neurotoxins A (G) and B (H) (heavy chains), cholera toxin (I), Pasteurella toxin (J), and diphtheria toxin (K). All toxins were tested orally at 1 μg/ml in a sucrose solution. The effect of each toxin on fly survival was tested in the absence of bacteria as a control. P as in Fig 1.

https://doi.org/10.1371/journal.ppat.1008836.g007

Discussion

While biting and non-biting flies have been known to be capable of transmitting B. anthracis since the 19th century [5860], the mechanism of insect-mediated anthrax transmission is unknown. Recent studies emphasized that anthrax and anthrax-containing insects pose a significant ecological threat in West African ecosystems [24, 711]. When flies feed on anthrax-infected or deceased mammals, they invariably ingest both B. anthracis and circulating anthrax toxin components, including PA20 [37]. B. anthracis is known to remain in the insect’s gut for an extended time or as part of the insect microbiota for life [7]. While the potential benefit of this co-existence has not been studied, we propose a mechanism by which the vector-borne pathogen may benefit. Bacillus species, which naturally reside in the soil, are relatively nonmotile and benefit from a suitable vector to assist bacterial distribution. The reduced insect sensitivity would benefit mammalian pathogens by allowing microbes to further their reach.

Our study exposed insects to vegetative B. anthracis for days, while in previous studies blow flies, horn flies, house flies, mosquitoes were found to be insensitive to this pathogen after short exposures for several hours [11, 15, 17, 39], possibly explaining the difference in the insect sensitivity between studies. Moreover, we demonstrate that Drosophila and Culex are insensitive to B. anthracis spores, which is similar to previous observations that B. anthracis spores are not lethal to flies, mosquitoes [11, 15, 17, 39], and nematodes [61, 62]. Our results revealed that flies and mosquitoes respond differently to spores. Spores from either the Sterne or ΔSterne strains extended the longevity of Drosophila, with toxin-containing Sterne strain extending the life longer than the ΔSterne strain, whereas the longevity of mosquitoes was not affected by spores of either strain (Fig 3). It is possible that in our experimental conditions, uninfected flies can succumb to environmental microorganisms after 10–14 days, and the oral exposure to B. anthracis spores extended the longevity of flies by stimulating their innate immunity. Since it was previously shown that PA is detected on the surface of B. anthracis spores [63, 64], we hypothesize that the presence of this surface PA proteins could further activate the innate immunity of flies and extend their longevity, as we observed in this study. The reason for the lack of the extension of longevity in mosquitoes could be that the cause of death of mosquitoes may be different from flies or because spores do not affect the immunity the same way they do in flies. Moreover, our results revealed that unlike Drosophila, mosquitoes sensitivity was similar to Sterne and ΔSterne strains. We conjecture this could occur due to mosquitoes’ comparatively superior immune system, but this observed difference was not further explored. Future studies should investigate the toxin-dependent and -independent interaction of B. anthracis with flies and mosquitoes.

This work describes a new function of PA20, the domain that previously has only been known to prevent self-assembly of PA83 in solution. PA20 has also been known to contribute to the adaptive immunogenicity of PA83 in mammals, as it contains at least three immunogenic epitopes [32]. One such PA20 epitope is the 1β13−1α1 loop shown to affect insect and plant innate immunity in this and previous studies [48]. The results described above led us to propose a novel mechanism by which PA20 reduces the sensitivity of flies to insecticidal bacterial pathogens by utilizing Drosophila’s Toll pathway (Fig 5A). Both Bacillus and Serratia species of bacteria are known to possess dap-type peptidoglycans and thus are known inducers of the Imd pathway [52]. Toll pathway is required for PA20 function, and PA20 may directly interact with Drosophila PGRP-SA to activate Toll, while Bacillus bacteria activate Imd, which would result in the transcription of AMPs [23]. Independently and together, Toll and Imd pathways result in the expression of AMPs that are known to be broad-spectrum in their effects and may possess the ability to act additively and/or synergistically [23, 65]. PA20 may be providing insects with an added benefit, resulting in the extension in lifespan during the bacterial challenge and allowing pathogens to further their reach. Alternatively, as dap is a structural derivative of lys, and since both Toll and Imd pathways can detect lys- and dap-peptidoglycan [21, 54], it is possible that PA20 may bind to Imd-activating PGRP-LC and PGRP-LE, thus leading to the activation of both Toll and Imd pathways. Although we show that the Imd pathway is not necessary, its activity is sufficient to negate the protective effects of PA20.

Drosophila GNBP-1 and PGRP-SA form a functional heterodimer. Fly PGRP-SA binds predominantly to lys-type peptidoglycan, but also binds to dap-type peptidoglycan [19]. In contrast, GNBP-1 only binds to lys-type peptidoglycan [19]. GNBP-1 is known to hydrolyze Gram-positive peptidoglycan, while PGRP-SA binds peptidoglycan fragments (muropeptides). GNBP-1 presents a hydrolyzed form of peptidoglycan for sensing by PGRP-SA, and this tripartite interaction between these proteins and peptidoglycan fragments is essential for downstream signaling. Future studies should establish whether GNBP-1 and PGRP-SA form a tripartite interaction and whether this leads to the activation of the downstream Toll pathway.

In Drosophila, at least seven AMPs (plus their isoforms) have been described: Diptericins (DptA and DptB), Cecropins (CecA1, CecA2, CecB, CecC, and Cecψ1), Drosocin (Dro), Attacins (AttA, AttB, AttC, and AttD), Drosomycins (Drs, Dro2, Dro3, Dro4, Dro5, Dro5, and Drsl1), Metchnikowin (Mtk), and Defensin (Def), with many of them showing broad-spectrum antimicrobial activity against Gram-negative and Gram-positive bacteria [2325]. The expression of many of them has been shown to be regulated at the transcriptional level by both the Toll and the Imd pathways and by both Gram-negative and Gram-positive bacteria, where the time of AMPs expression post-infection varies for each peptide [2325]. Moreover, the tissue expression of AMPs follows a complex pattern that is specific for each peptide, where individual AMPs can be expressed in the digestive tract, salivary glands, labellar glands, respiratory tract, seminal receptacle, spermatheca, calyx, and oviduct [24]. Future studies should determine: i) the identity of AMPs activated in response to the exposure of flies to PA20, ii) the tissues where AMPs are expressed in response to PA20, and iii) the timing when AMPs are expressed in response to PA20.

In addition to AMP and hemocyte responses, fly intestinal epithelia exposed to pore-forming toxins undergo an evolutionarily conserved process of thinning (purging) followed by the rapid recovery of their initial thickness [66]. We argue that while PA is a pore-forming toxin component, the effect of PA20 is independent of pore formation, as it lacks PA domain 3, which is located within PA63 and is necessary for PA63-multimerization [67]. We propose that the mechanism of action by which the pore-forming Clostridium septicum alpha toxin provides flies protection from Bacillus anthracis Sterne strain infection is by inducing the purging of fly intestinal epithelia, as described by Lee et al. [66]. Future studies should explore this hypothesis.

The effect of PA20 on the Toll/NF-κB pathway observed in this study is consistent with the impact PA83 and PA20 have on the homologous mammalian pathway (Fig 5A, right panel). PA83 is the central antigen in the FDA-approved anthrax vaccine, BioThrax [68]. The vaccination with PA83 [38] and human cell exposures to PA83 [36] or PA20 [37] lead to the Toll-like receptors-dependent activation of NF-κB and a subsequent upregulation in the expression of pro-inflammatory cytokines, especially Interleukin-6 (IL-6), IL-6 receptor, IL-1β, and Tumor necrosis factor alpha (TNF-α). Thus, in addition to an established paradigm of PA83 activating adaptive immunity in mammals, this toxin component may also affect innate immunity. BioThrax is approved for use as both a pre-exposure vaccination and post-exposure prophylaxis [69, 70], which is consistent with the potential that its efficacy is partly based on an unrecognized effect on the human innate immune system, as presented here.

While our study may help understand the mechanism through which insects tolerate anthrax, resulting in their greater opportunity to transmit the anthrax-causing bacteria, it further suggests that PA20 could be used beneficially in agriculture. Since fruit flies and mosquitoes are known to act as plant pollinators [71, 72], PA20 should be further evaluated for its ability to reduce the sensitivity of pollinating insects to bacterial pathogens, such as Serratia and Bacillus species [7375].

Materials and methods

Chemicals and reagents

Anthrax toxin components used: PA83, PA63, and LF were a kind gift from Kenneth Bradley (University of California, Los Angeles). Toxins (product numbers) purchased from List Biological Laboratories were: EF (178A), diphtheria toxin (150), Clostridium septicum alpha toxin (116L), C. perfringens epsilon toxin (126A), C. difficile toxins A (152C), B (155B), and binary toxin subunit A(157A), Pasteurella toxin (156), pertussis toxin (181), botulinum neurotoxins A (612A) and B (622A) (heavy chains), and cholera toxin (100B). PA20 was recombinantly expressed using the methods described below. Purified recombinant T7-tagged D. melanogaster PGRP-SA protein was a kind gift from Sérgio Raposo Filipe (Universidade Nova de Lisboa). Chemical/reagents (source, catalog #) are: cortisone acetate (TCI, C0389), human recombinant PGRP-S (also called PGLYRP1) (BioVendor, RD172316100), polyclonal mouse anti-PGRP-S antibody (Abnova, H00008993-B01P), monoclonal mouse Anti-T7 tag antibody (Sigma-Aldrich, T8823), peptidoglycan from B. subtilis and Staphylococcus aureus (Sigma-Aldrich, 69554 and 77140, respectively), polyclonal goat anti-PA antibodies (List Biological Laboratories, 771B), rabbit anti-goat-HRP secondary antibody (Invitrogen, 81–1620), and goat anti-mouse-HRP secondary antibody (Bio-Rad, 172–1011). PA83 and PA2013−1α1 loop peptides were custom synthesized with purity higher than 95% and lyophilized by LifeTein using the method described below and resuspended in PBS at 1 mg/mL.

PA20 peptides were synthesized by LifeTein on ChemMatrix Rink Amide resin, using standard Fmoc synthesis protocol with DIC/Cl-HOBt coupling, on an APEX 396 automatic synthesizer. The resin was swollen in N,N-Dimethylformamide (DMF) for 30 minutes, treated with 20 v% Piperidine-DMF for 8 minutes to remove the Fmoc protecting group, at 50°C, and washed with DMF for three times. For the coupling reaction, the resin was added with Fmoc-protected amino acid, Cl-HOBt, DIC, and NMP. The mixture was vortexed for 20 minutes at 50°C. Afterward, the resin was washed with DMF once. The cycle of deprotection and coupling steps was repeated until the last amino acid residue was assembled. After the final Fmoc protecting group was removed, the resin was treated with 20 v% acetic Anhydride-NMP for 20 minutes. The resin was then washed with DMF, DCM, and dried with air. The peptides were cleaved using a TFA cocktail (95 v% TFA, 2.5 v% water, and 2.5 v% TIS) for three hours. Crude peptides were precipitated by adding ice-chilled anhydrous ethyl ether, washed with anhydrous ethyl ether three times, and dried in vacuo. The crude peptides were HPLC purified.

Drosophila rearing

D. melanogaster fly strains were maintained at 25 ˚C with 12-hour light/dark cycles and fed standard cornmeal-molasses-agar fly medium with yeast flakes. Wild-type (WT) experiments were conducted with Oregon-R Drosophila (Bloomington Drosophila Stock Center (BDSC) stock # 5) aged 4–5 days. Some experiments were performed with wild-type flies of various ages. All mutant flies were of various ages at the time of experimentation. Mutant fly strains used (and their BDSC stock #) are: Imd (55711), cactus (34501), SPE (33926), Spz (3115), GNBP1 (18150), Sphe (29227), Psh (52877), PGRP-SA (58589), ModSP (55717), Grass (67099) and Dsor1 (5545). For Dsor1 experiments, only female flies were tested because male flies are short-lived post-eclosure (S8 Fig).

Mosquito rearing

Culex quinquefasciatus mosquitoes were obtained from a colony maintained by Benzon Research (Carlisle, PA, USA). Culex larvae were fed daily with 0.025 g/L of a 3:1 mixture of bovine liver powder and brewer’s yeast (Benzon Research). Mosquito larvae were maintained at 200 larvae per 1 liter of nanopore water in 35.6 cm x 27.9 cm x 8.3 cm plastic trays. Mosquitoes were reared and maintained at 28°C, 12:12 hours light:dark diurnal cycle at 80% relative humidity in 30 × 30 × 30-cm cages. Mosquitoes were provided with 10% sucrose ad libitum for maintenance. For experiments, mosquitoes aged 3–4 days were used.

Bacterial strains and spore preparation

B. cereus (ATCC 10987), B. subtilis (strain 168), Escherichia coli (K-12 MDS42), and S. liquefaciens (ATCC 27592) were used as the infective agents for Drosophila survival assays. B. cereus and B. subtilis were incubated on Lysogeny Broth (LB) at 37°C while S. liquefaciens was grown on Tryptic Soy Broth at 30°C. Overnight cultures were made in the respective media and temperatures at 180 rpm for 16–18 hours.

Strains of B. anthracis used included the toxigenic Sterne and toxin-negative ΔSterne strains. Spores were prepared and purified from solid agar (NBY-Mn) cultures of the strains and purified as described [76, 77]. Medium components were from BD-Difco, and the NBY-Mn medium was composed of nutrient broth (8 g/L) (BD 23400); yeast extract (3 g/L) (BD 244020); MnSO4·H2O (25 mg/L) (Sigma-Aldrich, M7634); and agar (15 g/L) (Aldon Corp, AA0075). Spores were used only if >95% were refractile (ungerminated) as determined by phase microscopy and heat-resistance. Spore preparation included two centrifugations in density gradient medium (58 mL Hypaque-76™, Nycomed into 42 mL WFI) accompanied by extensive washing in sterile water for injection (WFI). The spores were activated by heating at 65°C for 30 min just before use in assays [76].

Oral feeding survival assay

Flies or mosquitoes were infected according to the bacterial intestinal infection methods previously employed by Nehme et al. [78] with minor modifications. To prepare the Drosophila or Culex vials for infection, three pre-cut extra-thick Whatman blotting papers (Bio-Rad Laboratories, 1703965) were stacked at the bottom of either 25x95 mm diameter polystyrene Drosophila vials or 55x55x102 mm specimen bottles for fly and mosquito infections, respectively, following by capping with a foam plug. Overnight bacterial cultures were centrifuged, and the bacterial pellets were resuspended in either a 50 mM or 10% sucrose solution for fly or mosquito experiments, respectively.

Insects were exposed orally to anthrax toxin components, domains, or peptides, each at a concentration of 1 μg/mL, which falls within the known 0.01–100 μg/mL range of the concentration of plasma-circulating toxin components in infected mammals during the late stage of anthrax infection [7988]. All other bacterial toxins were tested at the same concentration.

Flies and mosquitoes were exposed orally to approximately 2.6 X 109 cells/mL, as previous studies have shown that the concentration of plasma-circulating bacteria in infected mammals during the late stage of anthrax infection is 109−1010 cells/mL [80, 81, 88]. Oral exposure was chosen because it represents a route by which flies are exposed to anthrax toxin components and bacilli in a natural environment [18]. All experiments were performed with 4–5 day old (post-eclosion) wild type male flies and mosquitoes to reduce any potential variability in sensitivity to bacterial infections due to confounding gender and age differences.

A final optical density at 600 nm (OD600) was 3.3 for Bacilli species and E. coli (2.6 X 109 cells/mL), as well as OD600 of 4.6 for S. liquefaciens (3.7 X 109 cells/mL). Depending on the experimental condition, toxin components and cortisone acetate were also added to bacterial sucrose solutions before adding to Drosophila vials and mosquito bottles. Finally, 2.5 or 10 mL of the respective solution was pipetted onto the Whatman paper in each Drosophila vial or mosquito bottle. Flies and mosquitoes were anesthetized using CO2, separated by gender, and placed ten at a time into their respective vials or bottles and incubated at 30°C. The survival of insects was recorded a minimum of twice per day.

PA domains expression and purification

A gene strand containing a pelB leader sequence, the amino-terminal of PA20 (residues 30–192), a 6-His tag, and 20 bp vector overlaps at the 5’ and 3’ end was ordered from Eurofins Genomics. PA83 amino acids 1–29 were not included in the expression, because they form a signal peptide that is cleaved off of the mature protein [89]. Digested pSX2 expression vector (Scarab Genomics) and the pelB-PA20-6His gene strand were assembled using Gibson assembly.

PA domains 2, 3, and 4 fusion proteins were designed to include the PA domain, a C terminal InaD PDZ domain (amino acids 2–98, GenBank accession no. 1IHJ_A, cys53ala mutation), a C-terminal 6xHis tag, and 3’ and 5’ overlaps for insertion into the pSX2. The gene strands were cloned into KpnI and SacI-digested pSX2 using the NEBuilder HiFi DNA Assembly Cloning Kit (New England Biolabs, E5520S).

The sequences for the His-tagged PAD1 and PAD1’-InaD fusion proteins were PCR amplified with vector overlaps to IPTG-inducible expression vector containing maltose-binding protein (MBP) with a C-terminal PreScission protease cleavage site (pET His6 MBP prescission LIC cloning vector (HMPKS, plasmid #29721)). The vector was digested using XhoI and SspI, and the PCR-amplified inserts were cloned in using the NEBuilder HiFi DNA Assembly Cloning Kit.

All constructed plasmids were transformed into chemically competent Escherichia coli DH5α cells for plasmid preparations. Purified PA20, PAD2, PAD3, and PAD4 plasmids were then transformed into MDS42 E. coli for expression. Purified PAD1 and PAD1’ plasmids were transformed into chemically-competent T7 Express E. coli (New England Biolabs, C2566I) for protein expression. His-tagged PA20, PAD1, and PAD1’ were purified using Ni-NTA resin (Thermo Fisher Scientific, 88221), and PAD2, PAD3, and PAD4 were purified on HisTrap HP columns (GE Healthcare, GE29-0510-21).

MBP was cleaved from the MBP- PAD1, and -PAD1’ fusion proteins in solution using the Pierce HRV 3C Protease Kit (Thermo Scientific, 88946). The HRV3C protease was removed using Pierce Glutathione Agarose (Thermo Scientific, 16100) and Pierce Centrifuge Columns (Thermo Scientific, 89898). MBP was removed using amylose resin (New England Biolabs, E8021S), followed by dialysis and concentration of proteins using Vivaspin Centrifugal Concentrators (Sartorius, VS0101). Protein concentration was determined using the Pierce BCA Protein Assay Kit with bovine serum albumin as a standard (Thermo Scientific, 23225).

All PA domains were analyzed via SDS-PAGE and anti-PA83 Western blotting to confirm the molecular weight using polyclonal goat anti-PA83 and the secondary rabbit anti-goat IgG antibodies. Chemiluminescence of bands and their relative intensities were revealed using Azure c500 (Azure Biosystems, Dublin, CA).

Bacterial growth assays

For the agar-containing media assays, the OD600 of overnight cultures of bacteria was measured. The OD600 values were converted to cells/mL according to McFarland’s scale [90]. 6 X 108 bacterial cells were added to solid media on 25 cm Petri dishes. One μL of toxin component or cortisone acetate was then pipetted onto the plate surface. The plates were then incubated for 24 hours at the temperatures appropriate for each bacterium. One μL of 10 mM levofloxacin (Cayman Chemical, 20382) or phosphate-buffered saline (PBS) (Corning, 21-040-CV) were included as controls.

In the liquid media experiments, the bacterial overnight culture was resuspended in a new liquid bacterial medium to OD600 of 0.1. One hundred μL of the bacterial solution were then added into wells of a 96-well plate, followed by the addition of PA20 to specific wells to a concentration of 1 μg/mL. The bacteria were incubated in 96-well plates at appropriate temperatures with constant shaking. The OD600 was determined every 610 seconds for 700 minutes by a microplate reader (Molecular Devices, Spectra Max 384 PLUS).

PRGP-S pull-down assay

This assay was performed according to the procedure of Yoshida et al. [91] with minor modifications. Approximately 1 mg of insoluble dap-type peptidoglycan from B. subtilis was dispersed in 1 mL of PBS and centrifuged at 12,600 x g for 5 min. This process was repeated three times, and the sedimented peptidoglycan was resuspended in 1 mL of PBS. Ten μL (0.5 mg/mL) of recombinant human PGRP-S was mixed with 100 μg of peptidoglycan suspension or PA20, and incubated overnight at 4°C. The mixture was centrifuged at 12,600 x g for 5 min, and the pellets were washed three times with 300 μL PBS and resuspended in 200 μL Laemmli sample buffer (VWR, 89230–104). Both the pellets and the supernatants were analyzed by Western blot using anti-PA83 and anti-PGRP-S antibodies.

PGRP-SA and PRGP-S binding ELISAs

PA20, washed dap-type peptidoglycan, or washed lys-type peptidoglycan were diluted to 10 μg/mL in bicarbonate buffer, and 50 μL was added to flat-bottom, high-binding, half-area 96-well plates (Corning, 29442–318) in triplicates and incubated overnight at 4°C. The next day, the wells were rinsed with PBST and PBS, then blocked with 100 μL blocking buffer (5% non-fat milk in PBS (PGRP-SA) or 5% BSA [Roche, 10738328103] in PBS (PGRP-S)) for 2 hours at room temperature. Wells were washed two times with PBST, and two times with PBS. Fifty μL of PRGP-SA with a T7 tag or PGRP-S diluted in PBS to 200 ng/μL was added to each well, and the plates were incubated for 2 hours at room temperature. The wells were washed as previously described, and 50 μL of anti-T7 monoclonal antibody (Sigma) diluted to 6.7 μg/mL in PBST and incubated overnight at 4°C. For the PGRP-S plate, anti-PRGP-S polyclonal antibody diluted to 0.5 μg/mL in 5% BSA added to each well and incubated for 2 hours at room temperature. The wells were washed as above, and horseradish peroxidase-conjugated anti-mouse secondary antibody, diluted in PBST for PGRP-SA and 5% BSA for PGRP-S, was added to each well and incubated for 2 hours at room temperature. Following PBST and PBS washes, 50 μL of o-Phenylenediamine dihydrochloride (Sigma, P1526) dissolved in deionized water was added to each well, and the plate was incubated for 30 minutes at room temperature. The absorbance at 450 nm was measured using a microplate reader (Molecular Devices, Spectra Max 384 PLUS).

Measurement of reactive oxygen species in S2 cells

Reactive oxygen species (ROS) consisting of hydrogen peroxide, peroxynitrite, hydroxyl radicals, nitric oxide, and peroxy radicals, and separately superoxide radicals were measured using ROS-ID Total ROS/Superoxide Detection Kit (Enzo Life Sciences, ENZ-51010). S2 cells (Expression Systems, 94-005F) were collected by centrifugation at 400 x g for 5 minutes and resuspended in ESF 921 medium (Expression Systems, 96-001-01). Cells were seeded at 500,000 cells per well in 50 μL of resuspension medium into clear 96-well plates with black chimneys. Dilutions of PA20 were performed in separate 12 channel basins with LB or LB containing B. cereus overnight culture at 1.6 OD600. PA20 was tested at 22, 2, 1, 0.5, and 0.25 μg/mL. A condition with no PA20 was included with every assay. The oxidative stress detection reagent and the superoxide detection reagent were reconstituted in anhydrous DMF (VWR, 97064–586) to yield 5 mM stock solutions and stored at -20°C. Detection reagents were added at 0.04% of sample preparation. Fifty μL of sample preparation reagent was added to the 96-well containing S2 cells and then incubated at 27°C in the dark for 30 minutes. Fluorescence was measured with bottom reading for two different wavelengths. Total ROS was measured at excitation 488 nm, cutoff 515 nm, and emission 520 nm. Superoxide detection was measured at excitation 550 nm, cutoff 610 nm, and emission 610 nm (Molecular Devices, SpectraMax Gemini XPS/EM Microplate Reader).

Data analysis

Data analysis was conducted using GraphPad Prism software. All P-values reported are products of the respective positive control to a single experimental condition using two statistical analyses: the Log-rank (Mantel-Cox) and the Gehan-Breslow-Wilcoxon tests. An alpha of 0.05 was deemed the threshold for significance. We report P values adjusted by the Bonferroni correction. The delay in median survival was reported. Since the chance of dying in a small-time interval was not the same early in the study and late in the study, the values for the 95% CI of the ratio of median survivals were not meaningful and were not reported. Flies that died within the first 24 hours of the assay were censored from statistical analysis and considered to have died due to non-infective causes. Each insect experiment shown is representative of at least three independent experiments.

Supporting information

S1 Fig. The effect of anthrax toxins on the longevity of flies.

In the absence of bacterial challenge, oral administration of anthrax toxins does not affect fly survival: flies were fed a 50 mM sucrose solution (WT condition) or a solution containing anthrax toxin components PA83, PA63, PA20, LF and EF, resuspended in 50 mM sucrose.

https://doi.org/10.1371/journal.ppat.1008836.s001

(TIF)

S2 Fig. PA83 protects unaged Drosophila from Bacillus cereus.

Male WT flies of various ages were challenged with B. cereus in the absence or presence of PA83. P as in Fig 1.

https://doi.org/10.1371/journal.ppat.1008836.s002

(TIF)

S3 Fig. PA2013−1α1 loop does not alter the growth of B. cereus.

Evaluation of the effects of cortisone and PA8313−1α1 loop. Agar diffusion susceptibility assay of B. cereus grown on LB solid medium. Plates were spread with 50 μL of bacterial overnight culture diluted to an OD600 of 0.1 and subsequently spotted with 1 μL of the following reagents. The plate was left to incubate overnight at 37 ˚C. Spots below a through e contain a 1 μL spot of: 20 mM cortisone acetate (a), 1 μg/mL of PA8313−1α1 loop (aa 181–200), (b) 1 μg/mL of PA2013−1α1 loop (aa 181–192) (c), PBS (d), 10 mM levofloxacin (e).

https://doi.org/10.1371/journal.ppat.1008836.s003

(TIF)

S4 Fig. PA83 protects unaged Drosophila from Serratia liquefaciens.

Male WT flies of various ages were challenged with S. liquefaciens in the absence or presence of PA83. P as in Fig 1.

https://doi.org/10.1371/journal.ppat.1008836.s004

(TIF)

S5 Fig. The effect of PA20 on the sensitivity of fly mutants to S. liquefaciens.

The effect of PA20 or PA83 on the Toll and Imd pathways mutants. SPE (A), SPZ (B), Imd (C), Dsor1 (with PA83) (D), and Dsor1 (with PA20) (E) mutant flies were fed a 50 mM sucrose solution in the presence or the absence of S. liquefaciens, or S. liquefaciens with 1 μg/mL of PA20 or PA83. Flies were maintained at 30°C and monitored for death a minimum of twice daily and expressed as percent survival. P as in Fig 1.

https://doi.org/10.1371/journal.ppat.1008836.s005

(TIF)

S6 Fig. Determination of minimal immunosuppressive concentration of cortisone acetate.

Titration assays revealed that 20 mM cortisone acetate added to the feeding medium was the minimum concentration sufficient to immunosuppress Drosophila (Fig 6I), as 10mM did not alter the sensitivity of Drosophila to B. cereus. Wild type flies were fed a 50 mM sucrose solution. Some conditions included B. cereus, which was resuspended in 50 mM sucrose solution, or a condition containing an additional 10 mM cortisone acetate. Flies were maintained at 30°C and monitored for death a minimum of twice daily and expressed as percent survival.

https://doi.org/10.1371/journal.ppat.1008836.s006

(TIF)

S7 Fig. Determining the effect of PA20 on superoxide production in S2 cells.

Measuring superoxide radicals in S2 cells in the absence and the presence of PA20. Superoxides were measured using ROS-ID Total ROS/Superoxide Detection Kit (Enzo Life Sciences). Superoxides produced by S2 cells were measured in the absence or the presence of B. cereus and various concentrations of PA20 (0.25 to 20 μg/mL).

https://doi.org/10.1371/journal.ppat.1008836.s007

(TIF)

S8 Fig. Determination of the longevity of Dsor1 male and female flies.

Dsor1 male and female flies were fed a 50mM sucrose solution (Dsor1 M/F). Flies were maintained at 30°C and monitored for death a minimum of twice daily and expressed as percent survival. Note, no bacterium was included in this experiment.

https://doi.org/10.1371/journal.ppat.1008836.s008

(TIF)

Acknowledgments

We thank Kenneth Bradley for a kind gift of anthrax toxin components, and Sérgio Raposo Filipe and Gonçalo Covas for a kind gift of fly PGRP-SA protein.

References

  1. 1. Brezillon C, Haustant M, Dupke S, Corre JP, Lander A, Franz T, et al. Capsules, toxins and AtxA as virulence factors of emerging Bacillus cereus biovar anthracis. PLoS Negl Trop Dis. 2015;9(4):e0003455. Epub 2015/04/02. pmid:25830379; PubMed Central PMCID: PMC4382292.
  2. 2. Hoffmann C, Zimmermann F, Biek R, Kuehl H, Nowak K, Mundry R, et al. Persistent anthrax as a major driver of wildlife mortality in a tropical rainforest. Nature. 2017;548(7665):82–6. Epub 2017/08/05. pmid:28770842.
  3. 3. Leendertz FH, Ellerbrok H, Boesch C, Couacy-Hymann E, Matz-Rensing K, Hakenbeck R, et al. Anthrax kills wild chimpanzees in a tropical rainforest. Nature. 2004;430(6998):451–2. Epub 2004/07/23. pmid:15269768.
  4. 4. Antonation KS, Grutzmacher K, Dupke S, Mabon P, Zimmermann F, Lankester F, et al. Bacillus cereus Biovar Anthracis Causing Anthrax in Sub-Saharan Africa-Chromosomal Monophyly and Broad Geographic Distribution. PLoS Negl Trop Dis. 2016;10(9):e0004923. Epub 2016/09/09. pmid:27607836. PubMed Central PMCID: PMC5015827.
  5. 5. Hoffmaster AR, Hill KK, Gee JE, Marston CK, De BK, Popovic T, et al. Characterization of Bacillus cereus isolates associated with fatal pneumonias: strains are closely related to Bacillus anthracis and harbor B. anthracis virulence genes. J Clin Microbiol. 2006;44(9):3352–60. Epub 2006/09/07. pmid:16954272; PubMed Central PMCID: PMC1594744.
  6. 6. Carlson CJ, Kracalik IT, Ross N, Alexander KA, Hugh-Jones ME, Fegan M, et al. The global distribution of Bacillus anthracis and associated anthrax risk to humans, livestock and wildlife. Nat Microbiol. 2019. Epub 2019/05/16. pmid:31086311.
  7. 7. De Vos V, Turnbull PC. Anthrax. In: Coetzer JA, Thomson GR, Tustin RC, eds. Infectious diseases of livestock, with special reference to Southern Africa. 2 ed. Cape Town: Oxford University Press Southern Africa; 2004.
  8. 8. Beyer W, Bellan S, Eberle G, Ganz HH, Getz WM, Haumacher R, et al. Distribution and molecular evolution of bacillus anthracis genotypes in Namibia. PLoS Negl Trop Dis. 2012;6(3):e1534. Epub 2012/03/14. pmid:22413024; PubMed Central PMCID: PMC3295808.
  9. 9. Lindeque PM, Turnbull PC. Ecology and epidemiology of anthrax in the Etosha National Park, Namibia. Onderstepoort J Vet Res. 1994;61(1):71–83. Epub 1994/03/01. pmid:7898901.
  10. 10. Blackburn JK, Van Ert M, Mullins JC, Hadfield TL, Hugh-Jones ME. The necrophagous fly anthrax transmission pathway: empirical and genetic evidence from wildlife epizootics. Vector Borne Zoonotic Dis. 2014;14(8):576–83. Epub 2014/07/30. pmid:25072988.
  11. 11. Fasanella A, Scasciamacchia S, Garofolo G, Giangaspero A, Tarsitano E, Adone R. Evaluation of the house fly Musca domestica as a mechanical vector for an anthrax. PLoS One. 2010;5(8):e12219. Epub 2010/09/03. pmid:20808920; PubMed Central PMCID: PMC2923185.
  12. 12. Gogarten JF, Düx A, Mubemba B, Pléh K, Hoffmann C, Mielke A, et al. Tropical rainforest flies carrying pathogens form stable associations with social nonhuman primates. Mol Ecol. 2019;28(18):4242–58. Epub 2019/06/10. pmid:31177585.
  13. 13. Zohdy S, Schwartz TS. Shoo fly don't bother me: Flies track social primates and carry viable anthrax. Mol Ecol. 2019;28(18):4135–7. Epub 2019/09/29. pmid:31559659.
  14. 14. WHO. Anthrax in humans and animals. 4th ed. ed. Geneva, Switzerland: World Health Organization; 2008. 12 p.
  15. 15. Turell MJ, Knudson GB. Mechanical transmission of Bacillus anthracis by stable flies (Stomoxys calcitrans) and mosquitoes (Aedes aegypti and Aedes taeniorhynchus). Infect Immun. 1987;55(8):1859–61. Epub 1987/08/01. pmid:3112013; PubMed Central PMCID: PMC260614.
  16. 16. Stiles CW. Isolation of the Bacillus anthracis from Spinose Ear Ticks Ornithodorus megnini. American Journal of Veterinary Research. 1944;5(17):318–9.
  17. 17. Morris H. Blood-sucking insects as transmitters of anthrax or charbon: Agricultural Experiment Station of the Louisiana State University and A. & M. College; 1918.
  18. 18. Buchon N, Silverman N, Cherry S. Immunity in Drosophila melanogaster—from microbial recognition to whole-organism physiology. Nat Rev Immunol. 2014;14(12):796–810. Epub 2014/11/26. pmid:25421701; PubMed Central PMCID: PMC6190593.
  19. 19. Wang L, Weber AN, Atilano ML, Filipe SR, Gay NJ, Ligoxygakis P. Sensing of Gram-positive bacteria in Drosophila: GNBP1 is needed to process and present peptidoglycan to PGRP-SA. EMBO J. 2006;25(20):5005–14. Epub 2006/10/07. pmid:17024181; PubMed Central PMCID: PMC1618108.
  20. 20. Iatsenko I, Kondo S, Mengin-Lecreulx D, Lemaitre B. PGRP-SD, an Extracellular Pattern-Recognition Receptor, Enhances Peptidoglycan-Mediated Activation of the Drosophila Imd Pathway. Immunity. 2016;45(5):1013–23. Epub 2016/11/17. pmid:27851910.
  21. 21. Vaz F, Kounatidis I, Covas G, Parton RM, Harkiolaki M, Davis I, et al. Accessibility to Peptidoglycan Is Important for the Recognition of Gram-Positive Bacteria in Drosophila. Cell Rep. 2019;27(8):2480–92.e6. Epub 2019/05/23. pmid:31116990; PubMed Central PMCID: PMC6533200.
  22. 22. Lemaitre B. The road to Toll. Nat Rev Immunol. 2004;4(7):521–7. Epub 2004/07/02. pmid:15229471.
  23. 23. Tzou P, Reichhart JM, Lemaitre B. Constitutive expression of a single antimicrobial peptide can restore wild-type resistance to infection in immunodeficient Drosophila mutants. Proc Natl Acad Sci U S A. 2002;99(4):2152–7. Epub 2002/02/21. pmid:11854512; PubMed Central PMCID: PMC122334.
  24. 24. Imler JL, Bulet P. Antimicrobial peptides in Drosophila: structures, activities and gene regulation. Chem Immunol Allergy. 2005;86:1–21. Epub 2005/06/25. pmid:15976485.
  25. 25. Lemaitre B, Reichhart JM, Hoffmann JA. Drosophila host defense: differential induction of antimicrobial peptide genes after infection by various classes of microorganisms. Proc Natl Acad Sci U S A. 1997;94(26):14614–9. Epub 1998/02/07. pmid:9405661; PubMed Central PMCID: PMC25070.
  26. 26. Mock M, Fouet A. Anthrax. Annu Rev Microbiol. 2001;55:647–71. Epub 2001/09/07. pmid:11544370.
  27. 27. Bradley KA, Mogridge J, Mourez M, Collier RJ, Young JA. Identification of the cellular receptor for anthrax toxin. Nature. 2001;414(6860):225–9. Epub 2001/11/09. pmid:11700562.
  28. 28. Kintzer AF, Thoren KL, Sterling HJ, Dong KC, Feld GK, Tang, II, et al. The protective antigen component of anthrax toxin forms functional octameric complexes. J Mol Biol. 2009;392(3):614–29. Epub 2009/07/25. pmid:19627991; PubMed Central PMCID: PMC2742380.
  29. 29. Petosa C, Collier RJ, Klimpel KR, Leppla SH, Liddington RC. Crystal structure of the anthrax toxin protective antigen. Nature. 1997;385(6619):833–8. Epub 1997/02/27. pmid:9039918.
  30. 30. Naik S, Brock S, Akkaladevi N, Tally J, McGinn-Straub W, Zhang N, et al. Monitoring the kinetics of the pH-driven transition of the anthrax toxin prepore to the pore by biolayer interferometry and surface plasmon resonance. Biochemistry. 2013;52(37):6335–47. Epub 2013/08/24. pmid:23964683; PubMed Central PMCID: PMC3790466.
  31. 31. Rigden DJ, Mello LV, Galperin MY. The PA14 domain, a conserved all-beta domain in bacterial toxins, enzymes, adhesins and signaling molecules. Trends Biochem Sci. 2004;29(7):335–9. Epub 2004/07/09. pmid:15236739.
  32. 32. Crowe SR, Ash LL, Engler RJ, Ballard JD, Harley JB, Farris AD, et al. Select human anthrax protective antigen epitope-specific antibodies provide protection from lethal toxin challenge. J Infect Dis. 2010;202(2):251–60. Epub 2010/06/11. pmid:20533877; PubMed Central PMCID: PMC2891133.
  33. 33. Abboud N, De Jesus M, Nakouzi A, Cordero RJ, Pujato M, Fiser A, et al. Identification of linear epitopes in Bacillus anthracis protective antigen bound by neutralizing antibodies. J Biol Chem. 2009;284(37):25077–86. Epub 2009/07/21. pmid:19617628; PubMed Central PMCID: PMC2757211.
  34. 34. Kwok WW, Yang J, James E, Bui J, Huston L, Wiesen AR, et al. The anthrax vaccine adsorbed vaccine generates protective antigen (PA)-Specific CD4+ T cells with a phenotype distinct from that of naive PA T cells. Infect Immun. 2008;76(10):4538–45. Epub 2008/08/06. pmid:18678674; PubMed Central PMCID: PMC2546819.
  35. 35. Laughlin EM, Miller JD, James E, Fillos D, Ibegbu CC, Mittler RS, et al. Antigen-specific CD4+ T cells recognize epitopes of protective antigen following vaccination with an anthrax vaccine. Infect Immun. 2007;75(4):1852–60. Epub 2007/02/07. pmid:17283103; PubMed Central PMCID: PMC1865708.
  36. 36. Triantafilou M, Uddin A, Maher S, Charalambous N, Hamm TS, Alsumaiti A, et al. Anthrax toxin evades Toll-like receptor recognition, whereas its cell wall components trigger activation via TLR2/6 heterodimers. Cell Microbiol. 2007;9(12):2880–92. Epub 2007/07/27. pmid:17651447.
  37. 37. Hammamieh R, Ribot WJ, Abshire TG, Jett M, Ezzell J. Activity of the Bacillus anthracis 20 kDa protective antigen component. BMC Infect Dis. 2008;8:124. Epub 2008/09/24. pmid:18808698; PubMed Central PMCID: PMC2564935.
  38. 38. Rogers JV, Casbohm SL, Giannunzio LF, Sinnott LT, Rust SW, Brys AM, et al. Transcriptional analysis of protective antigen-stimulated PBMC from non-human primates vaccinated with the anthrax vaccine absorbed. Vaccine. 2006;24(17):3609–17. Epub 2006/02/24. pmid:16494973.
  39. 39. von Terzi B, Turnbull PC, Bellan SE, Beyer W. Failure of Sterne- and Pasteur-like strains of Bacillus anthracis to replicate and survive in the urban bluebottle blow fly Calliphora vicina under laboratory conditions. PLoS One. 2014;9(1):e83860. Epub 2014/01/07. pmid:24392098; PubMed Central PMCID: PMC3879289.
  40. 40. Bartolo G, Gonzalez LO, Alameh S, Valencia CA, Martchenko Shilman M. Identification of glucocorticoid receptor in Drosophila melanogaster. BMC Microbiol. 2020;20(1):161. Epub 2020/06/17. pmid:32539689; PubMed Central PMCID: PMC7296755.
  41. 41. Ma J, Benson AK, Kachman SD, Jacobsen DJ, Harshman LG. Drosophila melanogaster Selection for Survival after Infection with Bacillus cereus Spores: Evolutionary Genetic and Phenotypic Investigations of Respiration and Movement. Int J Evol Biol. 2013;2013:576452. Epub 2013/05/02. pmid:23634317; PubMed Central PMCID: PMC3619139.
  42. 42. Ramírez-Camejo L, García-Alicea M, Maldonado-Morales G, Bayman P. Probiotics may protect Drosophila from infection by Aspergillus flavus. Int J Pharm Sci Res. 2017;8(4):1624–32.
  43. 43. Guichard A, Park JM, Cruz-Moreno B, Karin M, Bier E. Anthrax lethal factor and edema factor act on conserved targets in Drosophila. Proc Natl Acad Sci U S A. 2006;103(9):3244–9. Epub 2006/02/04. pmid:16455799; PubMed Central PMCID: PMC1413899.
  44. 44. Kolsto AB, Tourasse NJ, Okstad OA. What sets Bacillus anthracis apart from other Bacillus species? Annu Rev Microbiol. 2009;63:451–76. Epub 2009/06/12. pmid:19514852.
  45. 45. Rasko DA, Ravel J, Okstad OA, Helgason E, Cer RZ, Jiang L, et al. The genome sequence of Bacillus cereus ATCC 10987 reveals metabolic adaptations and a large plasmid related to Bacillus anthracis pXO1. Nucleic Acids Res. 2004;32(3):977–88. Epub 2004/02/13. pmid:14960714; PubMed Central PMCID: PMC373394.
  46. 46. Klee SR, Brzuszkiewicz EB, Nattermann H, Bruggemann H, Dupke S, Wollherr A, et al. The genome of a Bacillus isolate causing anthrax in chimpanzees combines chromosomal properties of B. cereus with B. anthracis virulence plasmids. PLoS One. 2010;5(7):e10986. Epub 2010/07/17. pmid:20634886; PubMed Central PMCID: PMC2901330.
  47. 47. Mandadi KK, Scholthof KB. Plant immune responses against viruses: how does a virus cause disease? Plant Cell. 2013;25(5):1489–505. Epub 2013/05/28. pmid:23709626; PubMed Central PMCID: PMC3694688.
  48. 48. McComb RC, Ho CL, Bradley KA, Grill LK, Martchenko M. Presentation of peptides from Bacillus anthracis protective antigen on Tobacco Mosaic Virus as an epitope targeted anthrax vaccine. Vaccine. 2015;33(48):6745–51. Epub 2015/10/31. pmid:26514421.
  49. 49. Liu S, Moayeri M, Leppla SH. Anthrax lethal and edema toxins in anthrax pathogenesis. Trends Microbiol. 2014;22(6):317–25. Epub 2014/04/02. pmid:24684968; PubMed Central PMCID: PMC4041834.
  50. 50. Turell MJ, Middlebrook JL. Mosquito inoculation: an alternative bioassay for toxins. Toxicon. 1988;26(11):1089–94. Epub 1988/01/01. pmid:2907687.
  51. 51. Ganesan S, Aggarwal K, Paquette N, Silverman N. NF-kappaB/Rel proteins and the humoral immune responses of Drosophila melanogaster. Curr Top Microbiol Immunol. 2011;349:25–60. Epub 2010/09/21. pmid:20852987. PubMed Central PMCID: PMC3083852.
  52. 52. Schleifer KH, Kandler O. Peptidoglycan types of bacterial cell walls and their taxonomic implications. Bacteriol Rev. 1972;36(4):407–77. Epub 1972/12/01. pmid:4568761; PubMed Central PMCID: PMC408328.
  53. 53. Guan R, Wang Q, Sundberg EJ, Mariuzza RA. Crystal structure of human peptidoglycan recognition protein S (PGRP-S) at 1.70 A resolution. J Mol Biol. 2005;347(4):683–91. Epub 2005/03/17. pmid:15769462.
  54. 54. Liu C, Xu Z, Gupta D, Dziarski R. Peptidoglycan recognition proteins: a novel family of four human innate immunity pattern recognition molecules. J Biol Chem. 2001;276(37):34686–94. Epub 2001/07/20. pmid:11461926.
  55. 55. Stephens RS, Chen WJ, Kuo CC. Effects of corticosteroids and cyclophosphamide on a mouse model of Chlamydia trachomatis pneumonitis. Infect Immun. 1982;35(2):680–4. Epub 1982/02/01. pmid:7056581; PubMed Central PMCID: PMC351095.
  56. 56. Ragab A, Buechling T, Gesellchen V, Spirohn K, Boettcher AL, Boutros M. Drosophila Ras/MAPK signalling regulates innate immune responses in immune and intestinal stem cells. Embo j. 2011;30(6):1123–36. Epub 2011/02/08. pmid:21297578; PubMed Central PMCID: PMC3061042.
  57. 57. Kleino A, Myllymäki H, Kallio J, Vanha-aho LM, Oksanen K, Ulvila J, et al. Pirk is a negative regulator of the Drosophila Imd pathway. J Immunol. 2008;180(8):5413–22. Epub 2008/04/09. pmid:18390723.
  58. 58. Budd W. Observations on the Occurrence of Malignant Pustule in England: Illustrated by Numerous Fatal Cases. British Medical Journal. 1863;1(114):237–41. PMC2324801.
  59. 59. Greenberg B. Fliesand Disease. Princeton: Princeton University Press; 1973.
  60. 60. Greenberg B. Flies and Disease. Princeton: Princeton University Press; 1971.
  61. 61. Kho MF, Bellier A, Balasubramani V, Hu Y, Hsu W, Nielsen-LeRoux C, et al. The pore-forming protein Cry5B elicits the pathogenicity of Bacillus sp. against Caenorhabditis elegans. PLoS One. 2011;6(12):e29122. Epub 2012/01/05. pmid:22216181; PubMed Central PMCID: PMC3245237.
  62. 62. Schelkle B, Choi Y, Baillie LW, Richter W, Buyuk F, Celik E, et al. Caenorhabditis elegans Predation on Bacillus anthracis: Decontamination of Spore Contaminated Soil with Germinants and Nematodes. Front Microbiol. 2017;8:2601. Epub 2018/01/31. pmid:29379472; PubMed Central PMCID: PMC5770795.
  63. 63. Cote CK, Rossi CA, Kang AS, Morrow PR, Lee JS, Welkos SL. The detection of protective antigen (PA) associated with spores of Bacillus anthracis and the effects of anti-PA antibodies on spore germination and macrophage interactions. Microb Pathog. 2005;38(5–6):209–25. Epub 2005/06/01. pmid:15925272.
  64. 64. Welkos S, Little S, Friedlander A, Fritz D, Fellows P. The role of antibodies to Bacillus anthracis and anthrax toxin components in inhibiting the early stages of infection by anthrax spores. Microbiology. 2001;147(Pt 6):1677–85. Epub 2001/06/08. pmid:11390699.
  65. 65. Hanson MA, Dostálová A, Ceroni C, Poidevin M, Kondo S, Lemaitre B. Synergy and remarkable specificity of antimicrobial peptides in vivo using a systematic knockout approach. Elife. 2019;8. Epub 2019/02/26. pmid:30803481; PubMed Central PMCID: PMC6398976.
  66. 66. Lee KZ, Lestradet M, Socha C, Schirmeier S, Schmitz A, Spenle C, et al. Enterocyte Purge and Rapid Recovery Is a Resilience Reaction of the Gut Epithelium to Pore-Forming Toxin Attack. Cell Host Microbe. 2016;20(6):716–30. Epub 2016/11/28. pmid:27889464.
  67. 67. Miller CJ, Elliott JL, Collier RJ. Anthrax protective antigen: prepore-to-pore conversion. Biochemistry. 1999;38(32):10432–41. Epub 1999/08/11. pmid:10441138.
  68. 68. McComb RC, Martchenko M. Neutralizing antibody and functional mapping of Bacillus anthracis protective antigen-The first step toward a rationally designed anthrax vaccine. Vaccine. 2016;34(1):13–9. Epub 2015/11/28. pmid:26611201.
  69. 69. Longstreth J, Skiadopoulos MH, Hopkins RJ. Licensure strategy for pre- and post-exposure prophylaxis of biothrax vaccine: the first vaccine licensed using the FDA animal rule. Expert Rev Vaccines. 2016;15(12):1467–79. Epub 2016/10/30. pmid:27792416.
  70. 70. FDA. BioThrax (Anthrax Vaccine Adsorbed) Emergent BioSolutions 2015. Available from: https://www.fda.gov/media/71954/download.
  71. 71. Lahondère C, Vinauger C, Okubo RP, Wolff GH, Chan JK, Akbari OS, et al. The olfactory basis of orchid pollination by mosquitoes. Proc Natl Acad Sci U S A. 2020;117(1):708–16. Epub 2019/12/25. pmid:31871198. PubMed Central PMCID: PMC6955360.
  72. 72. Ihara M, Furutani S, Shigetou S, Shimada S, Niki K, Komori Y, et al. Cofactor-enabled functional expression of fruit fly, honeybee, and bumblebee nicotinic receptors reveals picomolar neonicotinoid actions. Proc Natl Acad Sci U S A. 2020. Epub 2020/07/03. pmid:32611810.
  73. 73. Burritt NL, Foss NJ, Neeno-Eckwall EC, Church JO, Hilger AM, Hildebrand JA, et al. Sepsis and Hemocyte Loss in Honey Bees (Apis mellifera) Infected with Serratia marcescens Strain Sicaria. PLoS One. 2016;11(12):e0167752. Epub 2016/12/22. pmid:28002470; PubMed Central PMCID: PMC5176276.
  74. 74. Raymann K, Coon KL, Shaffer Z, Salisbury S, Moran NA. Pathogenicity of Serratia marcescens Strains in Honey Bees. MBio. 2018;9(5). Epub 2018/10/12. pmid:30301854; PubMed Central PMCID: PMC6178626.
  75. 75. Evans JD, Schwarz RS. Bees brought to their knees: microbes affecting honey bee health. Trends Microbiol. 2011;19(12):614–20. Epub 2011/10/29. pmid:22032828.
  76. 76. Welkos SL, Cote CK, Rea KM, Gibbs PH. A microtiter fluorometric assay to detect the germination of Bacillus anthracis spores and the germination inhibitory effects of antibodies. J Microbiol Methods. 2004;56(2):253–65. Epub 2004/01/28. pmid:14744454.
  77. 77. Pomerantsev AP, Chang Z, Rappole C, Leppla SH. Identification of three noncontiguous regions on Bacillus anthracis plasmid pXO1 that are important for its maintenance. J Bacteriol. 2014;196(16):2921–33. Epub 2014/06/11. pmid:24914182; PubMed Central PMCID: PMC4135633.
  78. 78. Nehme NT, Liégeois S, Kele B, Giammarinaro P, Pradel E, Hoffmann JA, et al. A model of bacterial intestinal infections in Drosophila melanogaster. PLoS pathogens. 2007;3(11):e173–e. Epub 2007/11/23. pmid:18039029.
  79. 79. Mabry R, Brasky K, Geiger R, Carrion R Jr., Hubbard GB, Leppla S, et al. Detection of anthrax toxin in the serum of animals infected with Bacillus anthracis by using engineered immunoassays. Clin Vaccine Immunol. 2006;13(6):671–7. Epub 2006/06/09. pmid:16760326; PubMed Central PMCID: PMC1489546.
  80. 80. Weiss S, Kobiler D, Levy H, Pass A, Ophir Y, Rothschild N, et al. Antibiotics cure anthrax in animal models. Antimicrob Agents Chemother. 2011;55(4):1533–42. Epub 2011/01/26. pmid:21263056; PubMed Central PMCID: PMC3067169.
  81. 81. Kobiler D, Weiss S, Levy H, Fisher M, Mechaly A, Pass A, et al. Protective antigen as a correlative marker for anthrax in animal models. Infect Immun. 2006;74(10):5871–6. Epub 2006/09/22. pmid:16988266; PubMed Central PMCID: PMC1594923.
  82. 82. Walsh JJ, Pesik N, Quinn CP, Urdaneta V, Dykewicz CA, Boyer AE, et al. A case of naturally acquired inhalation anthrax: clinical care and analyses of anti-protective antigen immunoglobulin G and lethal factor. Clin Infect Dis. 2007;44(7):968–71. Epub 2007/03/08. pmid:17342650.
  83. 83. Boyer AE, Quinn CP, Woolfitt AR, Pirkle JL, McWilliams LG, Stamey KL, et al. Detection and quantification of anthrax lethal factor in serum by mass spectrometry. Anal Chem. 2007;79(22):8463–70. Epub 2007/10/13. pmid:17929949.
  84. 84. Tang S, Moayeri M, Chen Z, Harma H, Zhao J, Hu H, et al. Detection of anthrax toxin by an ultrasensitive immunoassay using europium nanoparticles. Clin Vaccine Immunol. 2009;16(3):408–13. Epub 2009/01/09. pmid:19129473; PubMed Central PMCID: PMC2650879.
  85. 85. Stearns-Kurosawa DJ, Lupu F, Taylor FB Jr., Kinasewitz G, Kurosawa S. Sepsis and pathophysiology of anthrax in a nonhuman primate model. Am J Pathol. 2006;169(2):433–44. Epub 2006/08/01. pmid:16877346; PubMed Central PMCID: PMC1698797.
  86. 86. Mytle N, Hopkins RJ, Malkevich NV, Basu S, Meister GT, Sanford DC, et al. Evaluation of intravenous anthrax immune globulin for treatment of inhalation anthrax. Antimicrob Agents Chemother. 2013;57(11):5684–92. Epub 2013/08/28. pmid:23979731 PubMed Central PMCID: PMC3811278.
  87. 87. Boyer AE, Quinn CP, Hoffmaster AR, Kozel TR, Saile E, Marston CK, et al. Kinetics of lethal factor and poly-D-glutamic acid antigenemia during inhalation anthrax in rhesus macaques. Infect Immun. 2009;77(8):3432–41. Epub 2009/06/10. pmid:19506008; PubMed Central PMCID: PMC2715684.
  88. 88. Yamamoto BJ, Shadiack AM, Carpenter S, Sanford D, Henning LN, O'Connor E, et al. Efficacy Projection of Obiltoxaximab for Treatment of Inhalational Anthrax across a Range of Disease Severity. Antimicrob Agents Chemother. 2016;60(10):5787–95. Epub 2016/07/20. pmid:27431222; PubMed Central PMCID: PMC5038317.
  89. 89. Welkos SL, Lowe JR, Eden-McCutchan F, Vodkin M, Leppla SH, Schmidt JJ. Sequence and analysis of the DNA encoding protective antigen of Bacillus anthracis. Gene. 1988;69(2):287–300. Epub 1988/09/30. pmid:3148491.
  90. 90. McFarland J. The nephelometer: an instrument for estimating the number of bacteria in suspensions used for calculating the opsonic index and for vaccines. Journal of the American Medical Association. 1907;XLIX(14):1176–8.
  91. 91. Yoshida H, Kinoshita K, Ashida M. Purification of a peptidoglycan recognition protein from hemolymph of the silkworm, Bombyx mori. J Biol Chem. 1996;271(23):13854–60. Epub 1996/06/07. pmid:8662762.