Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 997
Filter
1.
Science ; 385(6715): 1305-1310, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39298587

ABSTRACT

Rodents are ubiquitous and typically unwelcome dwellers in human habitats worldwide, infesting homes, farm fields, and agricultural stores and potentially shedding disease-causing microbes into the most human-occupied of spaces. Of the vertebrate animal taxa that share pathogens with us, rodents are the most abundant and diverse, with hundreds of species of confirmed zoonotic hosts, some of which have nearly global distributions. However, only 12% of rodent species are known to be sources of pathogens that also infect people, and those rodents that do are now recognized as tending to share a suite of predictable traits. Here, we characterize those traits and explore them in the context of three emerging or reemerging rodent-borne zoonotic diseases of people: Lassa fever, Lyme disease, and plague.


Subject(s)
Lassa Fever , Lyme Disease , Plague , Rodentia , Zoonoses , Animals , Lassa Fever/transmission , Lassa Fever/epidemiology , Humans , Zoonoses/transmission , Zoonoses/epidemiology , Plague/transmission , Plague/epidemiology , Plague/microbiology , Lyme Disease/transmission , Lyme Disease/microbiology , Lyme Disease/epidemiology , Communicable Diseases, Emerging/epidemiology , Communicable Diseases, Emerging/veterinary , Communicable Diseases, Emerging/transmission , Disease Reservoirs
2.
Commun Biol ; 7(1): 1013, 2024 Aug 18.
Article in English | MEDLINE | ID: mdl-39155318

ABSTRACT

Yersinia pestis has been infecting humans since the Late Neolithic (LN). Whether those early infections were isolated zoonoses or initiators of a pandemic remains unclear. We report Y. pestis infections in two individuals (of 133) from the LN necropolis at Warburg (Germany, 5300-4900 cal BP). Our analyses show that the two genomes belong to distinct strains and reflect independent infection events. All LN genomes known today (n = 4) are basal in the phylogeny and represent separate lineages that probably originated in different animal hosts. In the LN, an opening of the landscape resulted in the introduction of new rodent species, which may have acted as Y. pestis reservoirs. Coincidentally, the number of dogs increased, possibly leading to Y. pestis infections in canines. Indeed, we detect Y. pestis in an LN dog. Collectively, our data suggest that Y. pestis frequently entered human settlements at the time without causing significant outbreaks.


Subject(s)
Dog Diseases , Phylogeny , Plague , Yersinia pestis , Animals , Yersinia pestis/genetics , Yersinia pestis/isolation & purification , Dogs/microbiology , Plague/microbiology , Plague/epidemiology , Plague/history , Plague/transmission , Humans , Dog Diseases/microbiology , Germany/epidemiology , Genome, Bacterial , History, Ancient
3.
Nat Commun ; 15(1): 7062, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39152136

ABSTRACT

Post-translational addition of O-linked N-acetylglucosamine (O-GlcNAc) to proteins is commonly associated with a variety of stress responses and cellular processes in eukaryotes, but its potential roles in bacteria are unclear. Here, we show that protein HmwC acts as an O-GlcNAc transferase (OGT) responsible for O-GlcNAcylation of multiple proteins in Yersinia pestis, a flea-borne pathogen responsible for plague. We identify 64 O-GlcNAcylated proteins (comprising 65 sites) with differential abundance under conditions mimicking the mammalian host (Mh) and flea vector (Fv) environments. Deletion of hmwC, encoding a putative OGT, structurally distinct from any existing member of the GT41 family, results in reduced O-GlcNAcylation, reduced growth, and alterations in virulence properties and survival under stress. Purified HmwC can modify target proteins in vitro using UDP-GlcNAc as sugar donor. One of the target proteins, OsdY, promotes Y. pestis survival under oxidative stress conditions. Thus, our results support that regulation of antioxidative responses through O-GlcNAcylation may be a conserved process shared by prokaryotes and eukaryotes.


Subject(s)
Bacterial Proteins , N-Acetylglucosaminyltransferases , Yersinia pestis , Yersinia pestis/metabolism , Yersinia pestis/genetics , Yersinia pestis/pathogenicity , Yersinia pestis/enzymology , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , N-Acetylglucosaminyltransferases/metabolism , N-Acetylglucosaminyltransferases/genetics , Animals , Virulence , Acetylglucosamine/metabolism , Mice , Antioxidants/metabolism , Protein Processing, Post-Translational , Plague/microbiology , Plague/metabolism , Oxidative Stress , Glycosylation
4.
Emerg Infect Dis ; 30(9): 1850-1864, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39173663

ABSTRACT

Plague is a deadly zoonosis that still poses a threat in many regions of the world. We combined epidemiologic, host, and vector surveillance data collected during 1961-1980 from the Araripe Plateau focus in northeastern Brazil with ecologic, geoclimatic, and Yersinia pestis genomic information to elucidate how these factors interplay in plague activity. We identified well-delimited plague hotspots showing elevated plague risk in low-altitude areas near the foothills of the plateau's concave sectors. Those locations exhibited distinct precipitation and vegetation coverage patterns compared with the surrounding areas. We noted a seasonal effect on plague activity, and human cases linearly correlated with precipitation and rodent and flea Y. pestis positivity rates. Genomic characterization of Y. pestis strains revealed a foundational strain capable of evolving into distinct genetic variants, each linked to temporally and spatially constrained plague outbreaks. These data could identify risk areas and improve surveillance in other plague foci within the Caatinga biome.


Subject(s)
Plague , Yersinia pestis , Plague/epidemiology , Plague/microbiology , Brazil/epidemiology , Yersinia pestis/genetics , Humans , Animals , Epidemics , Siphonaptera/microbiology , Genome, Bacterial , Genomics/methods , Seasons
5.
Nat Commun ; 15(1): 7266, 2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39179552

ABSTRACT

Yersinia pestis, the causative agent of plague, is endemic in certain regions due to a stable transmission cycle between rodents and their associated fleas. In addition, fleas are believed to serve as reservoirs that can occasionally cause enzootic plague cycles and explosive epizootic outbreaks that increase human exposure. However, transmission by fleas is inefficient and associated with a shortened lifespan of the flea and rodent hosts, indicating that there remain significant gaps in our understanding of the vector-animal cycle of Y. pestis. Here, we show that laboratory-reared, infected fleas (Xenopsylla cheopis) can transmit viable Y. pestis from adults to eggs, and the bacteria can be passed through all subsequent life stages of the flea. Thus, our data raise the possibility that transovarial transmission in fleas might contribute to the persistence of Y. pestis in the environment without detectable plague activity in mammals.


Subject(s)
Insect Vectors , Plague , Xenopsylla , Yersinia pestis , Animals , Yersinia pestis/physiology , Yersinia pestis/pathogenicity , Plague/transmission , Plague/microbiology , Xenopsylla/microbiology , Insect Vectors/microbiology , Female , Siphonaptera/microbiology , Humans , Mice , Male
6.
J Bacteriol ; 206(9): e0017324, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39158280

ABSTRACT

The adaptation of Yersinia pestis, the flea-borne plague agent, to fluctuating environmental conditions is essential for the successful colonization of the flea vector. A previous comparative transcriptomic analysis showed that the Cpx pathway of Y. pestis is up-regulated in infected fleas. The CpxAR two-component system is a component of the envelope stress response and is critical for maintaining the integrity of the cell. Here, a phenotypic screening revealed a survival defect of the cpxAR mutant to oxidative stress and copper. The measured copper concentration in the digestive tract contents of fed fleas increased fourfold during the digestive process. By direct analysis of phosphorylation of CpxR by a Phos-Tag gel approach, we demonstrated that biologically relevant concentrations of copper triggered the system. Then, a competitive challenge highlighted the role of the CpxAR system in bacterial fitness during flea infection. Lastly, an in vitro sequential exposure to copper and then H2O2 to mimic the flea suggests a model in which, within the insect digestive tract, the CpxAR system would be triggered by copper, establishing an oxidative stress response. IMPORTANCE: The bacterium Yersinia pestis is the agent of flea-borne plague. Our knowledge of the mechanisms used by the plague bacillus to infect the flea vector is limited. The up-regulation of the envelope stress response under the control of the Cpx signaling pathway was previously shown in a transcriptomic study. Here, our in vivo and in vitro approaches suggest a model in which Y. pestis uses the CpxAR phosphorelay system to sense and respond to the copper present in the flea gut, thereby optimizing the flea gut colonization. In other words, the system is essential for bacterial fitness in the flea.


Subject(s)
Bacterial Proteins , Gastrointestinal Tract , Gene Expression Regulation, Bacterial , Signal Transduction , Siphonaptera , Yersinia pestis , Animals , Yersinia pestis/genetics , Yersinia pestis/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Siphonaptera/microbiology , Gastrointestinal Tract/microbiology , Plague/microbiology , Copper/metabolism , Copper/pharmacology , Oxidative Stress , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/pharmacology , Insect Vectors/microbiology , Protein Kinases
7.
Appl Environ Microbiol ; 90(8): e0099524, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39023266

ABSTRACT

Surveillance for animal plague was conducted in the Marmota himalayana plague focus of the Qinghai-Tibet Plateau from 2020 to 2023. A 22.89% positive rate of serum F1 antibody was detected in live-caught marmots, alongside a 43.40% incidence of Yersinia pestis isolation from marmot carcasses. Marmot carcasses infected with plague exhibited a significantly higher spleen-somatic index (P < 0.05). Twenty-one Y. pestis-specific phages were isolated, among which one Y. pestis lytic phage (AKS2022HT87GU_phi) was isolated from the bone marrow of a marmot carcass (no. AKS2022HT87) and was found to be symbiotic with Y. pestis. Microscopy revealed the coexistence of lysed and non-lysed colonies of Y. pestis AKS2022HT87. Genome-wide analysis showed that certain strains of the Y. pestis AKS2022HT87 carried phage DNA fragments consistent with phage AKS2022HT87GU_phi. The rare symbiotic relationship between a lytic phage and Y. pestis observed in vitro was highlighted in this study, laying the basis for further exploring the relationship between Y. pestis and its bacteriophages.IMPORTANCEBacteriophages and host bacteria commonly coexist in vivo or in soil environments through complex and interdependent microbial interactions. However, recapitulating this symbiotic state remains challenging in vitro due to limited medium nutrients. In this work, the natural symbiosis between Yersinia pestis and specific phages has been discovered in a Marmota himalayana specimen. Epidemiological analysis presented the characteristics of the Y. pestis and specific phages in the area with a strong plague epidemic. Crucially, comparative genomics has been conducted to analyze the genetic changes in both the Y. pestis and phages over different periods, revealing the dynamic and evolving nature of their symbiosis. These are the critical steps to study the mechanism of the symbiosis.


Subject(s)
Bacteriophages , Marmota , Plague , Symbiosis , Yersinia pestis , Yersinia pestis/virology , Marmota/microbiology , Marmota/virology , Plague/microbiology , Animals , Bacteriophages/isolation & purification , Bacteriophages/physiology , Bacteriophages/genetics , China
8.
Nature ; 632(8023): 114-121, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38987589

ABSTRACT

In the period between 5,300 and 4,900 calibrated years before present (cal. BP), populations across large parts of Europe underwent a period of demographic decline1,2. However, the cause of this so-called Neolithic decline is still debated. Some argue for an agricultural crisis resulting in the decline3, others for the spread of an early form of plague4. Here we use population-scale ancient genomics to infer ancestry, social structure and pathogen infection in 108 Scandinavian Neolithic individuals from eight megalithic graves and a stone cist. We find that the Neolithic plague was widespread, detected in at least 17% of the sampled population and across large geographical distances. We demonstrate that the disease spread within the Neolithic community in three distinct infection events within a period of around 120 years. Variant graph-based pan-genomics shows that the Neolithic plague genomes retained ancestral genomic variation present in Yersinia pseudotuberculosis, including virulence factors associated with disease outcomes. In addition, we reconstruct four multigeneration pedigrees, the largest of which consists of 38 individuals spanning six generations, showing a patrilineal social organization. Lastly, we document direct genomic evidence for Neolithic female exogamy in a woman buried in a different megalithic tomb than her brothers. Taken together, our findings provide a detailed reconstruction of plague spread within a large patrilineal kinship group and identify multiple plague infections in a population dated to the beginning of the Neolithic decline.


Subject(s)
Farmers , Genomics , Pedigree , Plague , Population Dynamics , Yersinia pestis , Female , Humans , Male , Cemeteries/history , Farmers/history , Genome, Bacterial/genetics , History, Ancient , Phylogeny , Plague/epidemiology , Plague/history , Plague/microbiology , Plague/mortality , Scandinavian and Nordic Countries/epidemiology , Time Factors , Virulence Factors/genetics , Yersinia pestis/genetics , Yersinia pestis/isolation & purification
9.
mBio ; 15(8): e0107524, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-38958447

ABSTRACT

Yersinia pestis has recently evolved into a highly lethal flea-borne pathogen through the pseudogenization of extensive genes and the acquisition of exogenous plasmids. Particularly noteworthy are the newly acquired pPCP1 and pMT1 plasmids, which encode the virulence determinants Pla and Yersinia murine toxin (Ymt), crucial for subcutaneous infection and survival within flea vector of Y. pestis, respectively. This study reveals that Pla can cleave Ymt at K299 both in vivo and in vitro. Y. pestis expressing YmtK299A displays enhanced in vitro biofilm formation and increased blood survival, indicating significant roles of Pla-mediated Ymt cleavage in these phenotypes. Intriguingly, although both the ancestral form of Pla and the prevalent Pla-I259T variant in modern Y. pestis strains are capable of cleaving Ymt at K299, the cleavage efficiency of Pla-I259T is only half that of the ancestral variant. In subcutaneous infection, mice infected with Δymt::ymt-K299A show significantly prolonged survival compared to those infected with Δymt::ymt. Similarly, infection with Δpla::pla-I259T also results in extended survival compared to Δpla::pla infection. These data demonstrate that the I259T substitution of Pla mitigates the enhanced virulence of Y. pestis in mice caused by Pla-mediated Ymt cleavage, thereby prolonging the survival period of infected animals and potentially conferring advantages on the transmission of Y. pestis to the next host. These findings deepen our understanding of the intricate interplay between two newly acquired plasmids and shed light on the positive selection of the Pla-I259T mutation, providing new insights into the virulence dynamics and transmission mechanisms of Y. pestis. IMPORTANCE: The emergence of Y. pestis as a highly lethal pathogen is driven by extensive gene pseudogenization and acquisition of exogenous plasmids pPCP1 and pMT1. However, the interplay between these two plasmids during evolution remains largely unexplored. Our study reveals intricate interactions between Ymt and Pla, two crucial virulence determinants encoded on these plasmids. Pla-mediated cleavage of Ymt significantly decreases Y. pestis survival in mouse blood and enhances its virulence in mice. The prevalent Pla-I259T variant in modern strains displays reduced Ymt cleavage, thereby extending the survival of infected animals and potentially increasing strain transmissibility. Our findings shed light on the nuanced evolution of Y. pestis, wherein reduced cleavage efficiency is a positive selection force, shaping the pathogen's natural trajectory.


Subject(s)
Virulence Factors , Yersinia pestis , Yersinia pestis/genetics , Yersinia pestis/metabolism , Yersinia pestis/pathogenicity , Animals , Mice , Virulence , Virulence Factors/genetics , Virulence Factors/metabolism , Plasminogen Activators/genetics , Plasminogen Activators/metabolism , Female , Plague/microbiology , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Plasmids/genetics , Biofilms/growth & development , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Evolution, Molecular , Mice, Inbred BALB C , Disease Models, Animal
10.
Cell Immunol ; 403-404: 104856, 2024.
Article in English | MEDLINE | ID: mdl-39002222

ABSTRACT

Yersinia pestis is the causative agent of bubonic, septicemic and pneumonic plague. The historical importance and potential of plague to re-emerge as a threat worldwide are indisputable. The most severe manifestion of plague is pneumonic plague, which results in disease that is 100% lethal without treatment. Y. pestis suppresses host immune responses early in the lung to establish infection. The later stages of infection see the rapid onset of hyperinflammatory responses that prove lethal. The study of Y. pestis host/pathogen interactions have largely been investigated during bubonic plague and with attenuated strains in cell culture models. There remains a somewhat limited understanding of the interactions between virulent Y. pestis and immune populations in the lung that drive severe disease. In this review we give a broad overview of the progression of pneumonic plague and highlighting how Y. pestis interfaces with host innate immune populations in the lung to cause lethal disease.


Subject(s)
Host-Pathogen Interactions , Immunity, Innate , Lung , Plague , Yersinia pestis , Yersinia pestis/immunology , Yersinia pestis/pathogenicity , Plague/immunology , Plague/microbiology , Humans , Immunity, Innate/immunology , Lung/immunology , Lung/microbiology , Animals , Host-Pathogen Interactions/immunology , Virulence/immunology
11.
Mol Biol Rep ; 51(1): 722, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38829419

ABSTRACT

BACKGROUND: Yersinia pestis is a bacterium that causes the disease plague. It has caused the deaths of many people throughout history. The bacterium possesses several virulence factors (pPla, pFra, and PYV). PFra plasmid encodes fraction 1 (F1) capsular antigen. F1 protein protects the bacterium against host immune cells through phagocytosis process. This protein is specific for Y. pestis. Many diagnostic techniques are based on molecular and serological detection and quantification of F1 protein in different food and clinical samples. Aptamers are small nucleic acid sequences that can act as specific ligands for many targets.This study, aimed to isolate the high-affinity ssDNA aptamers against F1 protein. METHODS AND RESULTS: In this study, SELEX was used as the main strategy in screening aptamers. Moreover, enzyme-linked aptamer sorbent assay (ELASA) and surface plasmon resonance (SPR) were used to determine the affinity and specificity of obtained aptamers to F1 protein. The analysis showed that among the obtained aptamers, the three aptamers of Yer 21, Yer 24, and Yer 25 were selected with a KD value of 1.344E - 7, 2.004E - 8, and 1.68E - 8 M, respectively. The limit of detection (LoD) was found to be 0.05, 0.076, and 0.033 µg/ml for Yer 21, Yer 24, and Yer 25, respectively. CONCLUSION: This study demonstrated that the synthesized aptamers could serve as effective tools for detecting and analyzing the F1 protein, indicating their potential value in future diagnostic applications.


Subject(s)
Aptamers, Nucleotide , Bacterial Proteins , SELEX Aptamer Technique , Yersinia pestis , Yersinia pestis/genetics , SELEX Aptamer Technique/methods , Bacterial Proteins/genetics , Surface Plasmon Resonance/methods , Humans , Plague/diagnosis , Plague/microbiology , Antigens, Bacterial
12.
PLoS Negl Trop Dis ; 18(6): e0012252, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38935608

ABSTRACT

BACKGROUND: Plague, a zoonotic disease caused by Yersinia pestis, was responsible for 3 historical human pandemics that killed millions of people. It remains endemic in rodent populations in Africa, Asia, North America, and South America but human plague is rare in most of these locations. However, human plague is still highly prevalent in Madagascar, which typically records a significant part of all annual global cases. This has afforded an opportunity to study contemporary human plague in detail using various typing methods for Y. pestis. AIM: This review aims to summarize the methods that have been used to type Y. pestis in Madagascar along with the major discoveries that have been made using these approaches. METHODS: Pubmed and Google Scholar were used to search for the keywords: "typing Yersinia pestis Madagascar," "evolution Yersinia pestis Madagascar," and "diversity Yersinia pestis Madagascar." Eleven publications were relevant to our topic and further information was retrieved from references cited in those publications. RESULTS: The history of Y. pestis typing in Madagascar can be divided in 2 periods: the pre-genomics and genomics eras. During the pre-genomics era, ribotyping, direct observation of plasmid content and plasmid restriction fragment length polymorphisms (RFLP) were employed but only revealed a limited amount of diversity among Malagasy Y. pestis strains. Extensive diversity only started to be revealed in the genomics era with the use of clustered regularly interspaced palindromic repeats (CRISPR), multiple-locus variable number tandem repeats (VNTR) analysis (MLVA), and single-nucleotide polymorphisms (SNPs) discovered from whole genome sequences. These higher-resolution genotyping methods have made it possible to highlight the distribution and persistence of genotypes in the different plague foci of Madagascar (Mahajanga and the Central and Northern Highlands) by genotyping strains from the same locations across years, to detect transfers between foci, to date the emergence of genotypes, and even to document the transmission of antimicrobial resistant (AMR) strains during a pneumonic plague outbreak. Despite these discoveries, there still remain topics that deserve to be explored, such as the contribution of horizontal gene transfer to the evolution of Malagasy Y. pestis strains and the evolutionary history of Y. pestis in Madagascar. CONCLUSIONS: Genotyping of Y. pestis has yielded important insights on plague in Madagascar, particularly since the advent of whole-genome sequencing (WGS). These include a better understanding of plague persistence in the environment, antimicrobial AMR and multi-drug resistance in Y. pestis, and the person-to-person spread of pneumonic plague. Considering that human plague is still a significant public health threat in Madagascar, these insights can be useful for controlling and preventing human plague in Madagascar and elsewhere, and also are relevant for understanding the historical pandemics and the possible use of Y. pestis as a biological weapon.


Subject(s)
Plague , Yersinia pestis , Yersinia pestis/genetics , Yersinia pestis/classification , Yersinia pestis/isolation & purification , Madagascar/epidemiology , Plague/microbiology , Plague/epidemiology , Humans , Animals , Genotype , Genotyping Techniques/methods
13.
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
14.
Virus Res ; 346: 199395, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38782263

ABSTRACT

The plague, caused by Yersinia pestis, is a natural focal disease and the presence of Y. pestis in the environment is a critical ecological concern worldwide. The role of Y. pestis phages in the ecological life cycle of the plague is crucial. Previously, a temperature-sensitive phage named vB_YpM_HQ103 was isolated from plague foci in Yunnan province, China. Upon infecting the EV76 strain of Y. pestis, vB_YpM_HQ103 exhibits lysogenic behavior at 21 °C and lytic behavior at 37 °C. Various methods including continuous passage lysogenic tests, in vitro lysis tests, comparative genomic assays, fluorescence quantitative PCR and receptor identification tests were employed to demonstrate that the lysogenic life cycle of this phage is applicable to wild Y. pestis strains; its lysogeny is pseudolysogenic (carrying but not integrating), allowing it to replicate and proliferate within Y. pestis. Furthermore, we have identified the outer membrane protein OmpA of Y. pestis as the receptor for phage infection. In conclusion, our research provides insight into the characteristics and receptors of a novel Y. pestis phage infection with a pseudolysogenic cycle. The findings of this study enhance our understanding of Y. pestis phages and plague microecology, offering valuable insights for future studies on the conservation and genetic evolution of Y. pestis in nature.


Subject(s)
Bacteriophages , Genome, Viral , Lysogeny , Plague , Yersinia pestis , Yersinia pestis/virology , Yersinia pestis/genetics , Bacteriophages/genetics , Bacteriophages/isolation & purification , Bacteriophages/classification , Bacteriophages/physiology , Plague/microbiology , China , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism
15.
Viruses ; 16(5)2024 05 08.
Article in English | MEDLINE | ID: mdl-38793629

ABSTRACT

Plague is an endemic infectious disease caused by Yersinia pestis. In this study, we isolated fourteen phages with similar sequence arrangements to phage 186; these phages exhibited different lytic abilities in Enterobacteriaceae strains. To illustrate the phylogenetic relationships and evolutionary relationships between previously designated 186-type phages, we analysed the complete sequences and important genes of the phages, including whole-genome average nucleotide identity (ANI) and collinearity comparison, evolutionary analysis of four conserved structural genes (V, T, R, and Q genes), and analysis of the regulatory genes (cI, apl, and cII) and integrase gene (int). Phylogenetic analysis revealed that thirteen of the newly isolated phages belong to the genus Eganvirus and one belongs to the genus Felsduovirus in the family Peduoviridae, and these Eganvirus phages can be roughly clustered into three subgroups. The topological relationships exhibited by the whole-genome and structural genes seemed similar and stable, while the regulatory genes presented different topological relationships with the structural genes, and these results indicated that there was some homologous recombination in the regulatory genes. These newly isolated 186-type phages were mostly isolated from dogs, suggesting that the resistance of Canidae to Y. pestis infection may be related to the wide distribution of phages with lytic capability.


Subject(s)
Bacteriophages , Genome, Viral , Phylogeny , Yersinia pestis , Yersinia pestis/virology , Yersinia pestis/genetics , Bacteriophages/genetics , Bacteriophages/classification , Bacteriophages/isolation & purification , Animals , Evolution, Molecular , Dogs , Plague/microbiology
16.
PLoS Negl Trop Dis ; 18(5): e0012167, 2024 May.
Article in English | MEDLINE | ID: mdl-38701065

ABSTRACT

BACKGROUND: Plague, caused by the bacterium Yersinia pestis, is a zoonotic disease that poses considerable threats to human health. Nucleic acid tests are crucial for plague surveillance and the rapid detection of Y. pestis. However, inhibitors in complex samples such as soil and animal tissues often hamper nucleic acid detection, leading to a reduced rate of identifying low concentrations of Y. pestis. To address this challenge, we developed a sensitive and specific droplet digital polymerase chain reaction (ddPCR) assay for detecting Y. pestis DNA from soil and animal tissue samples. METHODS: Three genes (ypo2088, caf1, and pla) from Y. pestis were used to develop a multi-target ddPCR assay. The limits of detection (LoD), reproducibility, and specificity were assessed for bacterial genomic DNA samples. The ability of the assay to detect low concentrations of Y. pestis DNA from simulated soil and mouse liver tissue samples was respectively evaluated and compared with that of quantitative real-time PCR (qPCR). RESULTS: The results showed that the ddPCR LoDs ranged from 6.2 to 15.4 copies/reaction for the target genes, with good reproducibility and high specificity for Y. pestis. By testing 130 soil and mouse liver tissue samples spiked with Y. pestis, the ddPCR assay exhibited a better sensitivity than that of the qPCR assay used in the study, with LoDs of 102 colony forming units (CFU)/100 mg soil and 103 CFU/20 mg liver. Moreover, the assay presented good quantitative linearity (R2 = 0.99) for Y. pestis at 103-106 CFU/sample for soil and liver samples. CONCLUSION: The ddPCR assay presented good performance for detecting Y. pestis DNA from soil and mouse tissue samples, showing great potential for improving the detection rate of low concentrations of Y. pestis in plague surveillance and facilitating the early diagnosis of plague cases.


Subject(s)
Plague , Sensitivity and Specificity , Soil Microbiology , Yersinia pestis , Yersinia pestis/genetics , Yersinia pestis/isolation & purification , Animals , Plague/diagnosis , Plague/microbiology , Mice , Polymerase Chain Reaction/methods , DNA, Bacterial/genetics , Reproducibility of Results , Bacterial Proteins/genetics , Liver/microbiology , Limit of Detection , Humans
17.
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
18.
Antonie Van Leeuwenhoek ; 117(1): 61, 2024 Mar 23.
Article in English | MEDLINE | ID: mdl-38520511

ABSTRACT

Yersinia pestis, the causative agent of plague, is a highly virulent bacterium that poses a significant threat to human health. Preserving this bacterium in a viable state is crucial for research and diagnostic purposes. This paper presents and evaluates a simple lyophilization protocol for the long-term storage of Y. pestis strains from Fiocruz-CYP, aiming to explore its impact on viability and long-term stability, while replacing the currently used methodologies. The lyophilization tests were conducted using the non-virulent Y. pestis strain EV76, subjected to the lyophilization process under vacuum conditions. Viability assessment was performed to evaluate the effects of lyophilization and storage conditions on Y. pestis under multiple temperature conditions (- 80 °C, - 20 °C, 4-8 °C and room temperature). The lyophilization protocol employed in this study consistently demonstrated its efficacy in maintaining high viability rates for Y. pestis samples in a up to one year follow-up. The storage temperature that consistently exhibited the highest recovery rates was - 80 °C, followed by - 20 °C and 4-8 °C. Microscopic analysis of the post-lyophilized cultures revealed preserved morphological features, consistent with viable bacteria. The high viability rates observed in the preserved samples indicate the successful preservation of Y. pestis using this protocol. Overall, the presented lyophilization protocol provides a valuable tool for the long-term storage of Y. pestis, offering stability, viability, and functionality. By refining the currently used methods of lyophilization, this protocol can improve long-term preservation for Y. pestis strains collections, facilitating research efforts, diagnostic procedures, and the development of preventive and therapeutic strategies against plague.


Subject(s)
Plague , Yersinia pestis , Humans , Plague/microbiology , Brazil , Freeze Drying , Temperature
19.
PLoS Negl Trop Dis ; 18(3): e0012036, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38452122

ABSTRACT

Plague is a flea-borne fatal disease caused by the bacterium Yersinia pestis, which persists in rural Madagascar. Although fleas parasitizing rats are considered the primary vectors of Y. pestis, the human flea, Pulex irritans, is abundant in human habitations in Madagascar, and has been found naturally infected by the plague bacterium during outbreaks. While P. irritans may therefore play a role in plague transmission if present in plague endemic areas, the factors associated with infestation and human exposure within such regions are little explored. To determine the socio-ecological risk factors associated with P. irritans infestation in rural households in plague-endemic areas of Madagascar, we used a mixed-methods approach, integrating results from P. irritans sampling, a household survey instrument, and an observational checklist. Using previously published vectorial capacity data, the minimal P. irritans index required for interhuman bubonic plague transmission was modeled to determine whether household infestations were enough to pose a plague transmission risk. Socio-ecological risk factors associated with a high P. irritans index were then identified for enrolled households using generalized linear models. Household flea abundance was also modeled using the same set of predictors. A high P. irritans index occurred in approximately one third of households and was primarily associated with having a traditional dirt floor covered with a plant fiber mat. Interventions targeting home improvement and livestock housing management may alleviate flea abundance and plague risk in rural villages experiencing high P. irritans infestation. As plague-control resources are limited in developing countries such as Madagascar, identifying the household parameters and human behaviors favoring flea abundance, such as those identified in this study, are key to developing preventive measures that can be implemented at the community level.


Subject(s)
Flea Infestations , Plague , Siphonaptera , Yersinia pestis , Humans , Animals , Rats , Plague/microbiology , Madagascar/epidemiology , Siphonaptera/microbiology , Flea Infestations/epidemiology , Risk Factors
20.
Virulence ; 15(1): 2316439, 2024 12.
Article in English | MEDLINE | ID: mdl-38389313

ABSTRACT

The genus Yersinia includes human, animal, insect, and plant pathogens as well as many symbionts and harmless bacteria. Within this genus are Yersinia enterocolitica and the Yersinia pseudotuberculosis complex, with four human pathogenic species that are highly related at the genomic level including the causative agent of plague, Yersinia pestis. Extensive laboratory, field work, and clinical research have been conducted to understand the underlying pathogenesis and zoonotic transmission of these pathogens. There are presently more than 500 whole genome sequences from which an evolutionary footprint can be developed that details shared and unique virulence properties. Whereas the virulence of Y. pestis now seems in apparent homoeostasis within its flea transmission cycle, substantial evolutionary changes that affect transmission and disease severity continue to ndergo apparent selective pressure within the other Yersiniae that cause intestinal diseases. In this review, we will summarize the present understanding of the virulence and pathogenesis of Yersinia, highlighting shared mechanisms of virulence and the differences that determine the infection niche and disease severity.


Subject(s)
Plague , Yersinia Infections , Yersinia pestis , Animals , Humans , Yersinia/genetics , Virulence/genetics , Yersinia pestis/genetics , Plague/microbiology , Yersinia Infections/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL