Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
1.
PLoS Biol ; 22(5): e3002625, 2024 May.
Article in English | MEDLINE | ID: mdl-38771885

ABSTRACT

Yersinia pestis, the causative agent of plague, is a highly lethal vector-borne pathogen responsible for killing large portions of Europe's population during the Black Death of the Middle Ages. In the wild, Y. pestis cycles between fleas and rodents; occasionally spilling over into humans bitten by infectious fleas. For this reason, fleas and the rats harboring them have been considered the main epidemiological drivers of previous plague pandemics. Human ectoparasites, such as the body louse (Pediculus humanus humanus), have largely been discounted due to their reputation as inefficient vectors of plague bacilli. Using a membrane-feeder adapted strain of body lice, we show that the digestive tract of some body lice become chronically infected with Y. pestis at bacteremia as low as 1 × 105 CFU/ml, and these lice routinely defecate Y. pestis. At higher bacteremia (≥1 × 107 CFU/ml), a subset of the lice develop an infection within the Pawlowsky glands (PGs), a pair of putative accessory salivary glands in the louse head. Lice that developed PG infection transmitted Y. pestis more consistently than those with bacteria only in the digestive tract. These glands are thought to secrete lubricant onto the mouthparts, and we hypothesize that when infected, their secretions contaminate the mouthparts prior to feeding, resulting in bite-based transmission of Y. pestis. The body louse's high level of susceptibility to infection by gram-negative bacteria and their potential to transmit plague bacilli by multiple mechanisms supports the hypothesis that they may have played a role in previous human plague pandemics and local outbreaks.


Subject(s)
Pediculus , Plague , Yersinia pestis , Animals , Yersinia pestis/pathogenicity , Yersinia pestis/physiology , Pediculus/microbiology , Pediculus/physiology , Humans , Plague/transmission , Plague/microbiology , Insect Vectors/microbiology , Insect Vectors/parasitology , Insect Bites and Stings/microbiology , Female , Male
2.
PLoS Pathog ; 18(12): e1010996, 2022 12.
Article in English | MEDLINE | ID: mdl-36520713

ABSTRACT

Yersinia pestis, the bacterial agent of plague, is enzootic in many parts of the world within wild rodent populations and is transmitted by different flea vectors. The ecology of plague is complex, with rodent hosts exhibiting varying susceptibilities to overt disease and their fleas exhibiting varying levels of vector competence. A long-standing question in plague ecology concerns the conditions that lead to occasional epizootics among susceptible rodents. Many factors are involved, but a major one is the transmission efficiency of the flea vector. In this study, using Oropsylla montana (a ground squirrel flea that is a major plague vector in the western United States), we comparatively quantified the efficiency of the two basic modes of flea-borne transmission. Transmission efficiency by the early-phase mechanism was strongly affected by the host blood source. Subsequent biofilm-dependent transmission by blocked fleas was less influenced by host blood and was more efficient. Mathematical modeling predicted that early-phase transmission could drive an epizootic only among highly susceptible rodents with certain blood characteristics, but that transmission by blocked O. montana could do so in more resistant hosts irrespective of their blood characteristics. The models further suggested that for most wild rodents, exposure to sublethal doses of Y. pestis transmitted during the early phase may restrain rapid epizootic spread by increasing the number of immune, resistant individuals in the population.


Subject(s)
Plague , Siphonaptera , Yersinia pestis , Animals , Insect Vectors/microbiology , Siphonaptera/microbiology , Rodentia
3.
PLoS Pathog ; 17(10): e1009995, 2021 10.
Article in English | MEDLINE | ID: mdl-34648607

ABSTRACT

Yersinia murine toxin (Ymt) is a phospholipase D encoded on a plasmid acquired by Yersinia pestis after its recent divergence from a Yersinia pseudotuberculosis progenitor. Despite its name, Ymt is not required for virulence but acts to enhance bacterial survival in the flea digestive tract. Certain Y. pestis strains circulating in the Bronze Age lacked Ymt, suggesting that they were not transmitted by fleas. However, we show that the importance of Ymt varies with host blood source. In accordance with the original description, Ymt greatly enhanced Y. pestis survival in fleas infected with bacteremic mouse, human, or black rat blood. In contrast, Ymt was much less important when fleas were infected using brown rat blood. A Y. pestis Ymt- mutant infected fleas nearly as well as the Ymt+ parent strain after feeding on bacteremic brown rat blood, and the mutant was transmitted efficiently by flea bite during the first weeks after infection. The protective function of Ymt correlated with red blood cell digestion kinetics in the flea gut. Thus, early Y. pestis strains that lacked Ymt could have been maintained in flea-brown rat transmission cycles, and perhaps in other hosts with similar blood characteristics. Acquisition of Ymt, however, served to greatly expand the range of hosts that could support flea-borne plague.


Subject(s)
Bacterial Toxins/metabolism , Plague/transmission , Siphonaptera/microbiology , Yersinia pestis/genetics , Yersinia pestis/metabolism , Animals , Humans , Insect Vectors/microbiology , Mice , Plasmids , Rats , Virulence
4.
J Biol Chem ; 297(5): 101322, 2021 11.
Article in English | MEDLINE | ID: mdl-34688666

ABSTRACT

The salivary glands of the flea Xenopsylla cheopis, a vector of the plague bacterium, Yersinia pestis, express proteins and peptides thought to target the hemostatic and inflammatory systems of its mammalian hosts. Past transcriptomic analyses of salivary gland tissue revealed the presence of two similar peptides (XC-42 and XC-43) having no extensive similarities to any other deposited sequences. Here we show that these peptides specifically inhibit coagulation of plasma and the amidolytic activity of α-thrombin. XC-43, the smaller of the two peptides, is a fast, tight-binding inhibitor of thrombin with a dissociation constant of less than 10 pM. XC-42 exhibits similar selectivity as well as kinetic and binding properties. The crystal structure of XC-43 in complex with thrombin shows that despite its substrate-like binding mode, XC-43 is not detectably cleaved by thrombin and that it interacts with the thrombin surface from the enzyme catalytic site through the fibrinogen-binding exosite I. The low rate of hydrolysis was verified in solution experiments with XC-43, which show the substrate to be largely intact after 2 h of incubation with thrombin at 37 °C. The low rate of XC-43 cleavage by thrombin may be attributable to specific changes in the catalytic triad observable in the crystal structure of the complex or to extensive interactions in the prime sites that may stabilize the binding of cleavage products. Based on the increased arterial occlusion time, tail bleeding time, and blood coagulation parameters in rat models of thrombosis XC-43 could be valuable as an anticoagulant.


Subject(s)
Anticoagulants/chemistry , Antithrombins/chemistry , Insect Proteins/chemistry , Salivary Glands/chemistry , Salivary Proteins and Peptides/chemistry , Thrombin , Xenopsylla/chemistry , Animals , Humans , Rats , Thrombin/antagonists & inhibitors , Thrombin/chemistry , Xenopsylla/metabolism
5.
PLoS Pathog ; 16(12): e1009092, 2020 12.
Article in English | MEDLINE | ID: mdl-33284863

ABSTRACT

Yersinia pestis can be transmitted by fleas during the first week after an infectious blood meal, termed early-phase or mass transmission, and again after Y. pestis forms a cohesive biofilm in the flea foregut that blocks normal blood feeding. We compared the transmission efficiency and the progression of infection after transmission by Oropsylla montana fleas at both stages. Fleas were allowed to feed on mice three days after an infectious blood meal to evaluate early-phase transmission, or after they had developed complete proventricular blockage. Transmission was variable and rather inefficient by both modes, and the odds of early-phase transmission was positively associated with the number of infected fleas that fed. Disease progression in individual mice bitten by fleas infected with a bioluminescent strain of Y. pestis was tracked. An early prominent focus of infection at the intradermal flea bite site and dissemination to the draining lymph node(s) soon thereafter were common features, but unlike what has been observed in intradermal injection models, this did not invariably lead to further systemic spread and terminal disease. Several of these mice resolved the infection without progression to terminal sepsis and developed an immune response to Y. pestis, particularly those that received an intermediate number of early-phase flea bites. Furthermore, two distinct types of terminal disease were noted: the stereotypical rapid onset terminal disease within four days, or a prolonged onset preceded by an extended, fluctuating infection of the lymph nodes before eventual systemic dissemination. For both modes of transmission, bubonic plague rather than primary septicemic plague was the predominant disease outcome. The results will help to inform mathematical models of flea-borne plague dynamics used to predict the relative contribution of the two transmission modes to epizootic outbreaks that erupt periodically from the normal enzootic background state.


Subject(s)
Plague/transmission , Siphonaptera/physiology , Yersinia pestis/metabolism , Animals , Biofilms/growth & development , Disease Outbreaks , Disease Progression , Female , Insect Vectors/physiology , Mice , Siphonaptera/metabolism , Siphonaptera/microbiology , Yersinia pestis/pathogenicity
6.
Annu Rev Microbiol ; 71: 215-232, 2017 09 08.
Article in English | MEDLINE | ID: mdl-28886687

ABSTRACT

Interest in arthropod-borne pathogens focuses primarily on how they cause disease in humans. How they produce a transmissible infection in their arthropod host is just as critical to their life cycle, however. Yersinia pestis adopts a unique life stage in the digestive tract of its flea vector, characterized by rapid formation of a bacterial biofilm that is enveloped in a complex extracellular polymeric substance. Localization and adherence of the biofilm to the flea foregut is essential for transmission. Here, we review the molecular and genetic mechanisms of these processes and present a comparative evaluation and updated model of two related transmission mechanisms.


Subject(s)
Adaptation, Biological , Biofilms/growth & development , Insect Vectors/microbiology , Plague/microbiology , Plague/transmission , Siphonaptera/microbiology , Yersinia pestis/physiology , Animals , Disease Transmission, Infectious , Gastrointestinal Tract/microbiology , Yersinia pestis/genetics
7.
Infect Immun ; 89(1)2020 12 15.
Article in English | MEDLINE | ID: mdl-33077628

ABSTRACT

Bubonic plague results when Yersinia pestis is deposited in the skin via the bite of an infected flea. Bacteria then traffic to the draining lymph node (dLN) where they replicate to large numbers. Without treatment, this infection can result in highly fatal septicemia. Several plague vaccine candidates are currently at various stages of development, but no licensed vaccine is available in the United States. Though polyclonal and monoclonal antibodies (Ab) can provide complete protection against bubonic plague in animal models, the mechanisms responsible for this antibody-mediated immunity (AMI) to Y. pestis remain poorly understood. Here, we examine the effects of Ab opsonization on Y. pestis interactions with phagocytes in vitro and in vivo Opsonization of Y. pestis with polyclonal antiserum modestly increased phagocytosis/killing by an oxidative burst of murine neutrophils in vitro Intravital microscopy (IVM) showed increased association of Ab-opsonized Y. pestis with neutrophils in the dermis in a mouse model of bubonic plague. IVM of popliteal LNs after intradermal (i.d.) injection of bacteria in the footpad revealed increased Y. pestis-neutrophil interactions and increased neutrophil crawling and extravasation in response to Ab-opsonized bacteria. Thus, despite only having a modest effect in in vitro assays, opsonizing Ab had a dramatic effect in vivo on Y. pestis-neutrophil interactions in the dermis and dLN very early after infection. These data shed new light on the importance of neutrophils in AMI to Y. pestis and may provide a new correlate of protection for evaluation of plague vaccine candidates.


Subject(s)
Antibodies, Bacterial/immunology , Host-Pathogen Interactions/immunology , Neutrophils/immunology , Neutrophils/metabolism , Plague/etiology , Plague/pathology , Yersinia pestis/immunology , Animals , Antibody-Dependent Cell Cytotoxicity/immunology , Complement System Proteins/immunology , Complement System Proteins/metabolism , Cytokines/metabolism , Disease Models, Animal , Immunity, Innate , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymph Nodes/pathology , Mice , Reactive Oxygen Species/metabolism , Skin/immunology , Skin/metabolism , Skin/microbiology , Skin/pathology , Type III Secretion Systems/immunology , Type III Secretion Systems/metabolism
8.
PLoS Pathog ; 14(1): e1006859, 2018 01.
Article in English | MEDLINE | ID: mdl-29357385

ABSTRACT

Fleas can transmit Yersinia pestis by two mechanisms, early-phase transmission (EPT) and biofilm-dependent transmission (BDT). Transmission efficiency varies among flea species and the results from different studies have not always been consistent. One complicating variable is the species of rodent blood used for the infectious blood meal. To gain insight into the mechanism of EPT and the effect that host blood has on it, fleas were fed bacteremic mouse, rat, guinea pig, or gerbil blood; and the location and characteristics of the infection in the digestive tract and transmissibility of Y. pestis were assessed 1 to 3 days after infection. Surprisingly, 10-28% of two rodent flea species fed bacteremic rat or guinea pig blood refluxed a portion of the infected blood meal into the esophagus within 24 h of feeding. We term this phenomenon post-infection esophageal reflux (PIER). In contrast, PIER was rarely observed in rodent fleas fed bacteremic mouse or gerbil blood. PIER correlated with the accumulation of a dense mixed aggregate of Y. pestis, red blood cell stroma, and oxyhemoglobin crystals that filled the proventriculus. At their next feeding, fleas with PIER were 3-25 times more likely to appear partially blocked, with fresh blood retained within the esophagus, than were fleas without PIER. Three days after feeding on bacteremic rat blood, groups of Oropsylla montana transmitted significantly more CFU than did groups infected using mouse blood, and this enhanced transmission was biofilm-dependent. Our data support a model in which EPT results from regurgitation of Y. pestis from a partially obstructed flea foregut and that EPT and BDT can sometimes temporally overlap. The relative insolubility of the hemoglobin of rats and Sciurids and the slower digestion of their blood appears to promote regurgitative transmission, which may be one reason why these rodents are particularly prominent in plague ecology.


Subject(s)
Blood/microbiology , Gastrointestinal Tract/microbiology , Insect Vectors/microbiology , Plague/blood , Plague/transmission , Siphonaptera/microbiology , Yersinia pestis/physiology , Animals , Gastrointestinal Transit/physiology , Gerbillinae , Guinea Pigs , Mice , Rats , Time Factors
9.
PLoS Pathog ; 11(3): e1004734, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25781984

ABSTRACT

Yersinia pestis, the causative agent of plague, is typically transmitted by the bite of an infected flea. Many aspects of mammalian innate immune response early after Y. pestis infection remain poorly understood. A previous study by our lab showed that neutrophils are the most prominent cell type recruited to the injection site after intradermal needle inoculation of Y. pestis, suggesting that neutrophil interactions with Y. pestis may be important in bubonic plague pathogenesis. In the present study, we developed new tools allowing for intravital microscopy of Y. pestis in the dermis of an infected mouse after transmission by its natural route of infection, the bite of an infected flea. We found that uninfected flea bites typically induced minimal neutrophil recruitment. The magnitude of neutrophil response to flea-transmitted Y. pestis varied considerably and appeared to correspond to the number of bacteria deposited at the bite site. Macrophages migrated towards flea bite sites and interacted with small numbers of flea-transmitted bacteria. Consistent with a previous study, we observed minimal interaction between Y. pestis and dendritic cells; however, dendritic cells did consistently migrate towards flea bite sites containing Y. pestis. Interestingly, we often recovered viable Y. pestis from the draining lymph node (dLN) 1 h after flea feeding, indicating that the migration of bacteria from the dermis to the dLN may be more rapid than previously reported. Overall, the innate cellular host responses to flea-transmitted Y. pestis differed from and were more variable than responses to needle-inoculated bacteria. This work highlights the importance of studying the interactions between fleas, Y. pestis and the mammalian host to gain a better understanding of the early events in plague pathogenesis.


Subject(s)
Dendritic Cells/immunology , Macrophages/immunology , Neutrophils/immunology , Plague/immunology , Skin/immunology , Animals , Disease Models, Animal , Insect Vectors , Mice , Mice, Inbred C57BL , Neutrophil Infiltration/immunology , Plague/transmission , Siphonaptera/microbiology , Yersinia pestis/immunology
10.
Proc Natl Acad Sci U S A ; 111(52): 18709-14, 2014 Dec 30.
Article in English | MEDLINE | ID: mdl-25453069

ABSTRACT

The arthropod-borne transmission route of Yersinia pestis, the bacterial agent of plague, is a recent evolutionary adaptation. Yersinia pseudotuberculosis, the closely related food-and water-borne enteric species from which Y. pestis diverged less than 6,400 y ago, exhibits significant oral toxicity to the flea vectors of plague, whereas Y. pestis does not. In this study, we identify the Yersinia urease enzyme as the responsible oral toxin. All Y. pestis strains, including those phylogenetically closest to the Y. pseudotuberculosis progenitor, contain a mutated ureD allele that eliminated urease activity. Restoration of a functional ureD was sufficient to make Y. pestis orally toxic to fleas. Conversely, deletion of the urease operon in Y. pseudotuberculosis rendered it nontoxic. Enzymatic activity was required for toxicity. Because urease-related mortality eliminates 30-40% of infective flea vectors, ureD mutation early in the evolution of Y. pestis was likely subject to strong positive selection because it significantly increased transmission potential.


Subject(s)
Bacterial Proteins , Evolution, Molecular , Gene Silencing , Insect Vectors/microbiology , Urease , Xenopsylla/microbiology , Yersinia pestis , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Humans , Mutation , Plague/enzymology , Plague/genetics , Plague/pathology , Plague/transmission , Urease/genetics , Urease/metabolism , Yersinia pestis/enzymology , Yersinia pestis/genetics , Yersinia pestis/pathogenicity , Yersinia pseudotuberculosis/enzymology , Yersinia pseudotuberculosis/genetics , Yersinia pseudotuberculosis/pathogenicity
11.
Infect Immun ; 84(7): 1932-40, 2016 07.
Article in English | MEDLINE | ID: mdl-27160296

ABSTRACT

The plague bacillus Yersinia pestis is unique among the pathogenic Enterobacteriaceae in utilizing an arthropod-borne transmission route. Transmission by fleabite is a recent evolutionary adaptation that followed the divergence of Y. pestis from the closely related food- and waterborne enteric pathogen Yersinia pseudotuberculosis A combination of population genetics, comparative genomics, and investigations of Yersinia-flea interactions have disclosed the important steps in the evolution and emergence of Y. pestis as a flea-borne pathogen. Only a few genetic changes, representing both gene gain by lateral transfer and gene loss by loss-of-function mutation (pseudogenization), were fundamental to this process. The emergence of Y. pestis fits evolutionary theories that emphasize ecological opportunity in adaptive diversification and rapid emergence of new species.


Subject(s)
Biological Evolution , Communicable Diseases, Emerging , Plague/microbiology , Plague/transmission , Siphonaptera/microbiology , Yersinia pestis/physiology , Adaptation, Biological , Animals , Evolution, Molecular , Genetic Variation , Humans , Insect Vectors/microbiology , Plague/epidemiology
12.
Proc Natl Acad Sci U S A ; 109(25): 9857-62, 2012 Jun 19.
Article in English | MEDLINE | ID: mdl-22679291

ABSTRACT

Bacterial pathogens are becoming increasingly resistant to antibiotics. As an alternative therapeutic strategy, phage therapy reagents containing purified viral lysins have been developed against gram-positive organisms but not against gram-negative organisms due to the inability of these types of drugs to cross the bacterial outer membrane. We solved the crystal structures of a Yersinia pestis outer membrane transporter called FyuA and a bacterial toxin called pesticin that targets this transporter. FyuA is a ß-barrel membrane protein belonging to the family of TonB dependent transporters, whereas pesticin is a soluble protein with two domains, one that binds to FyuA and another that is structurally similar to phage T4 lysozyme. The structure of pesticin allowed us to design a phage therapy reagent comprised of the FyuA binding domain of pesticin fused to the N-terminus of T4 lysozyme. This hybrid toxin kills specific Yersinia and pathogenic E. coli strains and, importantly, can evade the pesticin immunity protein (Pim) giving it a distinct advantage over pesticin. Furthermore, because FyuA is required for virulence and is more common in pathogenic bacteria, the hybrid toxin also has the advantage of targeting primarily disease-causing bacteria rather than indiscriminately eliminating natural gut flora.


Subject(s)
Bacteriophages/metabolism , Gram-Negative Bacteria/virology , Mucoproteins/metabolism , Bacterial Proteins/chemistry , Bacteriocins/chemistry , Bacteriophages/physiology , Cell Membrane/metabolism , Cryoelectron Microscopy , Models, Molecular , Mucoproteins/chemistry , Protein Conformation , Protein Engineering , Protein Transport , Receptors, Cell Surface/chemistry
13.
Microbiology (Reading) ; 160(Pt 2): 396-405, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24222617

ABSTRACT

YadB and YadC are putative trimeric autotransporters present only in the plague bacterium Yersinia pestis and its evolutionary predecessor, Yersinia pseudotuberculosis. Previously, yadBC was found to promote invasion of epithelioid cells by Y. pestis grown at 37 °C. In this study, we found that yadBC also promotes uptake of 37 °C-grown Y. pestis by mouse monocyte/macrophage cells. We tested whether yadBC might be required for lethality of the systemic stage of plague in which the bacteria would be pre-adapted to mammalian body temperature before colonizing internal organs and found no requirement for early colonization or growth over 3 days. We tested the hypothesis that YadB and YadC function on ambient temperature-grown Y. pestis in the flea vector or soon after infection of the dermis in bubonic plague. We found that yadBC did not promote uptake by monocyte/macrophage cells if the bacteria were grown at 28 °C, nor was there a role of yadBC in colonization of fleas by Y. pestis grown at 21 °C. However, the presence of yadBC did promote recoverability of the bacteria from infected skin for 28 °C-grown Y. pestis. Furthermore, the gene for the proinflammatory chemokine CXCL1 was upregulated in expression if the infecting Y. pestis lacked yadBC but not if yadBC was present. Also, yadBC was not required for recoverability if the bacteria were grown at 37 °C. These findings imply that thermally induced virulence properties dominate over effects of yadBC during plague but that yadBC has a unique function early after transmission of Y. pestis to skin.


Subject(s)
Adhesins, Bacterial/biosynthesis , Monocytes/immunology , Monocytes/microbiology , Yersinia pestis/radiation effects , Animals , Bacterial Load , Cells, Cultured , Disease Models, Animal , Mice , Phenotype , Plague/microbiology , Plague/pathology , Skin/microbiology , Skin/pathology , Temperature , Yersinia pestis/isolation & purification , Yersinia pestis/physiology
14.
Microbiology (Reading) ; 160(Pt 11): 2517-2525, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25187626

ABSTRACT

Plague, caused by Yersinia pestis, is characterized by quiescent periods punctuated by rapidly spreading epizootics. The classical 'blocked flea' paradigm, by which a blockage forms in the flea's proventriculus on average 1-2 weeks post-infection (p.i.), forces starving fleas to take multiple blood meals, thus increasing opportunities for transmission. Recently, the importance of early-phase transmission (EPT), which occurs prior to blockage formation, has been emphasized during epizootics. Whilst the physiological and molecular mechanisms of blocked flea transmission are well characterized, the pathogen-vector interactions have not been elucidated for EPT. Within the blocked flea model, Yersinia murine toxin (Ymt) has been shown to be important for facilitating colonization of the midgut within the flea. One proposed mechanism of EPT is the regurgitation of infectious material from the flea midgut during feeding. Such a mechanism would require bacteria to colonize and survive for at least brief periods in the midgut, a process that is mediated by Ymt. Two key bridging vectors of Y. pestis to humans, Oropsylla montana (Siphonaptera: Ceratophyllidae) or Xenopsylla cheopis (Siphonaptera: Pulicidae), were used in our study to test this hypothesis. Fleas were infected with a mutant strain of Y. pestis containing a non-functional ymt that was shown previously to be incapable of colonizing the midgut and were then allowed to feed on SKH-1 mice 3 days p.i. Our results show that Ymt was not required for EPT by either flea species.


Subject(s)
Bacterial Toxins/metabolism , Insect Vectors/microbiology , Plague/transmission , Siphonaptera/microbiology , Xenopsylla/microbiology , Yersinia pestis/metabolism , Animals , Humans , Insect Vectors/physiology , Mice , Plague/microbiology , Rats , Rats, Sprague-Dawley , Siphonaptera/physiology , Virulence , Xenopsylla/physiology , Yersinia pestis/genetics , Yersinia pestis/pathogenicity
15.
mBio ; 15(6): e0012424, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38722159

ABSTRACT

Transmission of Yersinia pestis by fleas depends on the formation of condensed bacterial aggregates embedded within a gel-like matrix that localizes to the proventricular valve in the flea foregut and interferes with normal blood feeding. This is essentially a bacterial biofilm phenomenon, which at its end stage requires the production of a Y. pestis exopolysaccharide that bridges the bacteria together in a cohesive, dense biofilm that completely blocks the proventriculus. However, bacterial aggregates are evident within an hour after a flea ingests Y. pestis, and the bacterial exopolysaccharide is not required for this process. In this study, we characterized the biochemical composition of the initial aggregates and demonstrated that the yersinia murine toxin (Ymt), a Y. pestis phospholipase D, greatly enhances rapid aggregation following infected mouse blood meals. The matrix of the bacterial aggregates is complex, containing large amounts of protein and lipid (particularly cholesterol) derived from the flea's blood meal. A similar incidence of proventricular aggregation occurred after fleas ingested whole blood or serum containing Y. pestis, and intact, viable bacteria were not required. The initial aggregation of Y. pestis in the flea gut is likely due to a spontaneous physical process termed depletion aggregation that occurs commonly in environments with high concentrations of polymers or other macromolecules and particles such as bacteria. The initial aggregation sets up subsequent binding aggregation mediated by the bacterially produced exopolysaccharide and mature biofilm that results in proventricular blockage and efficient flea-borne transmission. IMPORTANCE: Yersinia pestis, the bacterial agent of plague, is maintained in nature in mammal-flea-mammal transmission cycles. After a flea feeds on a mammal with septicemic plague, the bacteria rapidly coalesce in the flea's digestive tract to form dense aggregates enveloped in a viscous matrix that often localizes to the foregut. This represents the initial stage of biofilm development that potentiates transmission of Y. pestis when the flea later bites a new host. The rapid aggregation likely occurs via a depletion-aggregation mechanism, a non-canonical first step of bacterial biofilm development. We found that the biofilm matrix is largely composed of host blood proteins and lipids, particularly cholesterol, and that the enzymatic activity of a Y. pestis phospholipase D (Ymt) enhances the initial aggregation. Y. pestis transmitted by flea bite is likely associated with this host-derived matrix, which may initially shield the bacteria from recognition by the host's intradermal innate immune response.


Subject(s)
Biofilms , Phospholipase D , Siphonaptera , Yersinia pestis , Yersinia pestis/enzymology , Phospholipase D/metabolism , Siphonaptera/microbiology , Biofilms/growth & development , Plague/microbiology , Plague/transmission , Extracellular Polymeric Substance Matrix/chemistry , Extracellular Polymeric Substance Matrix/microbiology , Extracellular Polymeric Substance Matrix/ultrastructure , Polysaccharides/metabolism , Microscopy, Electron, Transmission , Proteome/metabolism , Animals , Mice , Lipids/analysis
16.
J Bacteriol ; 195(9): 1920-30, 2013 May.
Article in English | MEDLINE | ID: mdl-23435973

ABSTRACT

Transmission of Yersinia pestis is greatly enhanced after it forms a bacterial biofilm in the foregut of the flea vector that interferes with normal blood feeding. Here we report that the ability to produce a normal foregut-blocking infection depends on induction of the Y. pestis PhoP-PhoQ two-component regulatory system in the flea. Y. pestis phoP-negative mutants achieved normal infection rates and bacterial loads in the flea midgut but produced a less cohesive biofilm both in vitro and in the flea and had a greatly reduced ability to localize to and block the flea foregut. Thus, not only is the PhoP-PhoQ system induced in the flea gut environment, but also this induction is required to produce a normal transmissible infection. The altered biofilm phenotype in the flea was not due to lack of PhoPQ-dependent or PmrAB-dependent addition of aminoarabinose to the Y. pestis lipid A, because an aminoarabinose-deficient mutant that is highly sensitive to cationic antimicrobial peptides had a normal phenotype in the flea digestive tract. In addition to enhancing transmissibility, induction of the PhoP-PhoQ system in the arthropod vector prior to transmission may preadapt Y. pestis to resist the initial encounter with the mammalian innate immune response.


Subject(s)
Arthropod Vectors/microbiology , Bacterial Proteins/metabolism , Plague/microbiology , Plague/transmission , Siphonaptera/microbiology , Yersinia pestis/metabolism , Animals , Bacterial Proteins/genetics , Female , Gene Expression Regulation, Bacterial , Humans , Male , Mice , Plague/parasitology , Virulence , Yersinia pestis/genetics , Yersinia pestis/pathogenicity
17.
Infect Immun ; 81(11): 4041-52, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23959716

ABSTRACT

Yersinia pestis carries homologues of the toxin complex (Tc) family proteins, which were first identified in other Gram-negative bacteria as having potent insecticidal activity. The Y. pestis Tc proteins are neither toxic to fleas nor essential for survival of the bacterium in the flea, even though tc gene expression is highly upregulated and much more of the Tc proteins YitA and YipA are produced in the flea than when Y. pestis is grown in vitro. We show that Tc(+) and Tc(-) Y. pestis strains are transmitted equivalently from coinfected fleas, further demonstrating that the Tc proteins have no discernible role, either positive or negative, in transmission by the flea vector. Tc proteins did, however, confer Y. pestis with increased resistance to killing by polymorphonuclear leukocytes (PMNs). Resistance to killing was not the result of decreased PMN viability or increased intracellular survival but instead correlated with a Tc protein-dependent resistance to phagocytosis that was independent of the type III secretion system (T3SS). Correspondingly, we did not detect T3SS-dependent secretion of the native Tc proteins YitA and YipA or the translocation of YitA- or YipA-ß-lactamase fusion proteins into CHO-K1 (CHO) cells or human PMNs. Thus, although highly produced by Y. pestis within the flea and related to insecticidal toxins, the Tc proteins do not affect interaction with the flea or transmission. Rather, the Y. pestis Tc proteins inhibit phagocytosis by mouse PMNs, independent of the T3SS, and may be important for subverting the mammalian innate immune response immediately following transmission from the flea.


Subject(s)
Bacterial Toxins/immunology , Host-Pathogen Interactions , Immune Evasion , Neutrophils/immunology , Neutrophils/microbiology , Phagocytosis , Yersinia pestis/immunology , Yersinia pestis/pathogenicity , Animals , Cells, Cultured , Cricetinae , Humans , Mice , Siphonaptera/microbiology
18.
Infect Immun ; 81(9): 3163-72, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23774602

ABSTRACT

Na(+)/H(+) antiporters are ubiquitous membrane proteins that play a central role in the ion homeostasis of cells. In this study, we examined the possible role of Na(+)/H(+) antiport in Yersinia pestis virulence and found that Y. pestis strains lacking the major Na(+)/H(+) antiporters, NhaA and NhaB, are completely attenuated in an in vivo model of plague. The Y. pestis derivative strain lacking the nhaA and nhaB genes showed markedly decreased survival in blood and blood serum ex vivo. Complementation of either nhaA or nhaB in trans restored the survival of the Y. pestis nhaA nhaB double deletion mutant in blood. The nhaA nhaB double deletion mutant also showed inhibited growth in an artificial serum medium, Opti-MEM, and a rich LB-based medium with Na(+) levels and pH values similar to those for blood. Taken together, these data strongly suggest that intact Na(+)/H(+) antiport is indispensable for the survival of Y. pestis in the bloodstreams of infected animals and thus might be regarded as a promising noncanonical drug target for infections caused by Y. pestis and possibly for those caused by other blood-borne bacterial pathogens.


Subject(s)
Sodium-Hydrogen Exchangers/genetics , Sodium-Hydrogen Exchangers/metabolism , Virulence/genetics , Yersinia pestis/genetics , Yersinia pestis/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Female , Mice , Plague/genetics , Plague/metabolism , Plague/microbiology , Sequence Deletion/genetics , Sheep/blood , Sheep/microbiology
19.
J Leukoc Biol ; 114(1): 42-52, 2023 07 01.
Article in English | MEDLINE | ID: mdl-36992528

ABSTRACT

Neutrophils represent a first line of defense against a wide variety of microbial pathogens. Transduction with an estrogen receptor-Hoxb8 transcription factor fusion construct conditionally immortalizes myeloid progenitor cells (NeutPro) capable of differentiation into neutrophils. This system has been very useful for generating large numbers of murine neutrophils for in vitro and in vivo studies. However, some questions remain as to how closely neutrophils derived from these immortalized progenitors reflect primary neutrophils. Here we describe our experience with NeutPro-derived neutrophils as it relates to our studies of Yersinia pestis pathogenesis. NeutPro neutrophils have circular or multilobed nuclei, similar to primary bone marrow neutrophils. Differentiation of neutrophils from NeutPro cells leads to increased expression of CD11b, GR1, CD62L, and Ly6G. However, the NeutPro neutrophils expressed lower levels of Ly6G than bone marrow neutrophils. NeutPro neutrophils produced reactive oxygen species at slightly lower levels than bone marrow neutrophils, and the 2 cell types phagocytosed and killed Y. pestis in vitro to a similar degree. To further demonstrate their utility, we used a nonviral method for nuclear delivery of CRISPR/Cas9 guide RNA complexes to delete genes of interest in NeutPro cells. In summary, we have found these cells to be morphologically and functionally equivalent to primary neutrophils and useful for in vitro assays related to studies of bacterial pathogenesis.


Subject(s)
Homeodomain Proteins , Neutrophils , Mice , Animals , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Neutrophils/metabolism , Receptors, Estrogen/metabolism , CRISPR-Cas Systems , Cell Differentiation , Myeloid Progenitor Cells
20.
Commun Biol ; 6(1): 1280, 2023 12 18.
Article in English | MEDLINE | ID: mdl-38110569

ABSTRACT

The salivary glands of hematophagous arthropods contain pharmacologically active molecules that interfere with host hemostasis and immune responses, favoring blood acquisition and pathogen transmission. Exploration of the salivary gland composition of the rat flea, Xenopsylla cheopis, revealed several abundant acid phosphatase-like proteins whose sequences lacked one or two of their presumed catalytic residues. In this study, we undertook a comprehensive characterization of the tree most abundant X. cheopis salivary acid phosphatase-like proteins. Our findings indicate that the three recombinant proteins lacked the anticipated catalytic activity and instead, displayed the ability to bind different biogenic amines and leukotrienes with high affinity. Moreover, X-ray crystallography data from the XcAP-1 complexed with serotonin revealed insights into their binding mechanisms.


Subject(s)
Siphonaptera , Xenopsylla , Rats , Animals , Siphonaptera/physiology , Acid Phosphatase , Salivary Proteins and Peptides/genetics , Biogenic Amines , Leukotrienes
SELECTION OF CITATIONS
SEARCH DETAIL