Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 925
Filtrar
1.
Nat Commun ; 15(1): 7062, 2024 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-39152136

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas , N-Acetilglucosaminiltransferasas , Yersinia pestis , Yersinia pestis/metabolismo , Yersinia pestis/genética , Yersinia pestis/patogenicidad , Yersinia pestis/enzimología , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , N-Acetilglucosaminiltransferasas/metabolismo , N-Acetilglucosaminiltransferasas/genética , Animales , Virulencia , Acetilglucosamina/metabolismo , Ratones , Antioxidantes/metabolismo , Procesamiento Proteico-Postraduccional , Peste/microbiología , Peste/metabolismo , Estrés Oxidativo , Glicosilación
2.
Nat Commun ; 15(1): 7266, 2024 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-39179552

RESUMEN

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.


Asunto(s)
Insectos Vectores , Peste , Xenopsylla , Yersinia pestis , Animales , Yersinia pestis/fisiología , Yersinia pestis/patogenicidad , Peste/transmisión , Peste/microbiología , Xenopsylla/microbiología , Insectos Vectores/microbiología , Femenino , Siphonaptera/microbiología , Humanos , Ratones , Masculino
3.
mBio ; 15(8): e0107524, 2024 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-38958447

RESUMEN

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.


Asunto(s)
Factores de Virulencia , Yersinia pestis , Yersinia pestis/genética , Yersinia pestis/metabolismo , Yersinia pestis/patogenicidad , Animales , Ratones , Virulencia , Factores de Virulencia/genética , Factores de Virulencia/metabolismo , Activadores Plasminogénicos/genética , Activadores Plasminogénicos/metabolismo , Femenino , Peste/microbiología , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Plásmidos/genética , Biopelículas/crecimiento & desarrollo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Evolución Molecular , Ratones Endogámicos BALB C , Modelos Animales de Enfermedad
4.
PLoS Biol ; 22(5): e3002625, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38771885

RESUMEN

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.


Asunto(s)
Pediculus , Peste , Yersinia pestis , Animales , Yersinia pestis/patogenicidad , Yersinia pestis/fisiología , Pediculus/microbiología , Pediculus/fisiología , Humanos , Peste/transmisión , Peste/microbiología , Insectos Vectores/microbiología , Insectos Vectores/parasitología , Mordeduras y Picaduras de Insectos/microbiología , Femenino , Masculino
5.
Nature ; 611(7935): 312-319, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36261521

RESUMEN

Infectious diseases are among the strongest selective pressures driving human evolution1,2. This includes the single greatest mortality event in recorded history, the first outbreak of the second pandemic of plague, commonly called the Black Death, which was caused by the bacterium Yersinia pestis3. This pandemic devastated Afro-Eurasia, killing up to 30-50% of the population4. To identify loci that may have been under selection during the Black Death, we characterized genetic variation around immune-related genes from 206 ancient DNA extracts, stemming from two different European populations before, during and after the Black Death. Immune loci are strongly enriched for highly differentiated sites relative to a set of non-immune loci, suggesting positive selection. We identify 245 variants that are highly differentiated within the London dataset, four of which were replicated in an independent cohort from Denmark, and represent the strongest candidates for positive selection. The selected allele for one of these variants, rs2549794, is associated with the production of a full-length (versus truncated) ERAP2 transcript, variation in cytokine response to Y. pestis and increased ability to control intracellular Y. pestis in macrophages. Finally, we show that protective variants overlap with alleles that are today associated with increased susceptibility to autoimmune diseases, providing empirical evidence for the role played by past pandemics in shaping present-day susceptibility to disease.


Asunto(s)
ADN Antiguo , Predisposición Genética a la Enfermedad , Inmunidad , Peste , Selección Genética , Yersinia pestis , Humanos , Aminopeptidasas/genética , Aminopeptidasas/inmunología , Peste/genética , Peste/inmunología , Peste/microbiología , Peste/mortalidad , Yersinia pestis/inmunología , Yersinia pestis/patogenicidad , Selección Genética/inmunología , Europa (Continente)/epidemiología , Europa (Continente)/etnología , Inmunidad/genética , Conjuntos de Datos como Asunto , Londres/epidemiología , Dinamarca/epidemiología
6.
Nature ; 606(7915): 718-724, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35705810

RESUMEN

The origin of the medieval Black Death pandemic (AD 1346-1353) has been a topic of continuous investigation because of the pandemic's extensive demographic impact and long-lasting consequences1,2. Until now, the most debated archaeological evidence potentially associated with the pandemic's initiation derives from cemeteries located near Lake Issyk-Kul of modern-day Kyrgyzstan1,3-9. These sites are thought to have housed victims of a fourteenth-century epidemic as tombstone inscriptions directly dated to 1338-1339 state 'pestilence' as the cause of death for the buried individuals9. Here we report ancient DNA data from seven individuals exhumed from two of these cemeteries, Kara-Djigach and Burana. Our synthesis of archaeological, historical and ancient genomic data shows a clear involvement of the plague bacterium Yersinia pestis in this epidemic event. Two reconstructed ancient Y. pestis genomes represent a single strain and are identified as the most recent common ancestor of a major diversification commonly associated with the pandemic's emergence, here dated to the first half of the fourteenth century. Comparisons with present-day diversity from Y. pestis reservoirs in the extended Tian Shan region support a local emergence of the recovered ancient strain. Through multiple lines of evidence, our data support an early fourteenth-century source of the second plague pandemic in central Eurasia.


Asunto(s)
Peste , Yersinia pestis , Arqueología , Cementerios , ADN Antiguo/análisis , ADN Bacteriano/análisis , Historia Medieval , Humanos , Kirguistán/epidemiología , Pandemias/historia , Filogenia , Peste/epidemiología , Peste/historia , Peste/microbiología , Yersinia pestis/clasificación , Yersinia pestis/patogenicidad
7.
Commun Biol ; 4(1): 1248, 2021 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-34728737

RESUMEN

Plague caused by Yersinia pestis is one of the deadliest diseases. However, many molecular mechanisms of bacterial virulence remain unclear. This study engaged in the discovery of small open reading frame (sORF)-encoded peptides (SEPs) in Y. pestis. An integrated proteogenomic pipeline was established, and an atlas containing 76 SEPs was described. Bioinformatic analysis indicated that 20% of these SEPs were secreted or localized to the transmembrane and that 33% contained functional domains. Two SEPs, named SEPs-yp1 and -yp2 and encoded in noncoding regions, were selected by comparative peptidomics analysis under host-specific environments and high-salinity stress. They displayed important roles in the regulation of antiphagocytic capability in a thorough functional assay. Remarkable attenuation of virulence in mice was observed in the SEP-deleted mutants. Further global proteomic analysis indicated that SEPs-yp1 and -yp2 affected the bacterial metabolic pathways, and SEP-yp1 was associated with the bacterial virulence by modulating the expression of key virulence factors of the Yersinia type III secretion system. Our study provides a rich resource for research on Y. pestis and plague, and the findings on SEP-yp1 and SEP-yp2 shed light on the molecular mechanism of bacterial virulence.


Asunto(s)
Proteínas Bacterianas/genética , Sistemas de Lectura Abierta/genética , Péptidos/genética , Factores de Virulencia/genética , Yersinia pestis/genética , Yersinia pestis/patogenicidad , Animales , Proteínas Bacterianas/metabolismo , Ratones , Péptidos/metabolismo , Proteogenómica
8.
Proc Natl Acad Sci U S A ; 118(44)2021 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-34716262

RESUMEN

Yersinia pestis causes human plague and colonizes both a mammalian host and a flea vector during its transmission cycle. A key barrier to bacterial infection is the host's ability to actively sequester key biometals (e.g., iron, zinc, and manganese) required for bacterial growth. This is referred to as nutritional immunity. Mechanisms to overcome nutritional immunity are essential virulence factors for bacterial pathogens. Y. pestis produces an iron-scavenging siderophore called yersiniabactin (Ybt) that is required to overcome iron-mediated nutritional immunity and cause lethal infection. Recently, Ybt has been shown to bind to zinc, and in the absence of the zinc transporter ZnuABC, Ybt improves Y. pestis growth in zinc-limited medium. These data suggest that, in addition to iron acquisition, Ybt may also contribute to overcoming zinc-mediated nutritional immunity. To test this hypothesis, we used a mouse model defective in iron-mediated nutritional immunity to demonstrate that Ybt contributes to virulence in an iron-independent manner. Furthermore, using a combination of bacterial mutants and mice defective in zinc-mediated nutritional immunity, we identified calprotectin as the primary barrier for Y. pestis to acquire zinc during infection and that Y. pestis uses Ybt to compete with calprotectin for zinc. Finally, we discovered that Y. pestis encounters zinc limitation within the flea midgut, and Ybt contributes to overcoming this limitation. Together, these results demonstrate that Ybt is a bona fide zinc acquisition mechanism used by Y. pestis to surmount zinc limitation during the infection of both the mammalian and insect hosts.


Asunto(s)
Fenoles/farmacología , Peste/metabolismo , Tiazoles/farmacología , Zinc/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Femenino , Expresión Génica/genética , Regulación Bacteriana de la Expresión Génica/genética , Hierro/metabolismo , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Fenoles/metabolismo , Peste/microbiología , Sideróforos/metabolismo , Tiazoles/metabolismo , Virulencia , Factores de Virulencia/metabolismo , Yersinia pestis/patogenicidad
9.
Int J Mol Sci ; 22(18)2021 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-34575967

RESUMEN

The bacterial pathogen, Yersinia pestis, has caused three historic pandemics and continues to cause small outbreaks worldwide. During infection, Y. pestis assembles a capsule-like protective coat of thin fibres of Caf1 subunits. This F1 capsular antigen has attracted much attention due to its clinical value in plague diagnostics and anti-plague vaccine development. Expression of F1 is tightly regulated by a transcriptional activator, Caf1R, of the AraC/XylS family, proteins notoriously prone to aggregation. Here, we have optimised the recombinant expression of soluble Caf1R. Expression from the native and synthetic codon-optimised caf1R cloned in three different expression plasmids was examined in a library of E. coli host strains. The functionality of His-tagged Caf1R was demonstrated in vivo, but insolubility was a problem with overproduction. High levels of soluble MBP-Caf1R were produced from codon optimised caf1R. Transcriptional-lacZ reporter fusions defined the PM promoter and Caf1R binding site responsible for transcription of the cafMA1 operon. Use of the identified Caf1R binding caf DNA sequence in an electrophoretic mobility shift assay (EMSA) confirmed correct folding and functionality of the Caf1R DNA-binding domain in recombinant MBP-Caf1R. Availability of functional recombinant Caf1R will be a valuable tool to elucidate control of expression of F1 and Caf1R-regulated pathophysiology of Y. pestis.


Asunto(s)
Antígenos Bacterianos/genética , Proteínas Bacterianas/genética , Peste/genética , Yersinia pestis/genética , Proteínas de Unión al ADN/genética , Genes araC/genética , Humanos , Operón/genética , Peste/microbiología , Peste/prevención & control , Plásmidos/genética , Factores de Transcripción/genética , Vacunas/genética , Yersinia pestis/patogenicidad
10.
Proc Natl Acad Sci U S A ; 118(36)2021 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-34465619

RESUMEN

The second plague pandemic started in Europe with the Black Death in 1346 and lasted until the 19th century. Based on ancient DNA studies, there is a scientific disagreement over whether the bacterium, Yersinia pestis, came into Europe once (Hypothesis 1) or repeatedly over the following four centuries (Hypothesis 2). Here, we synthesize the most updated phylogeny together with historical, archeological, evolutionary, and ecological information. On the basis of this holistic view, we conclude that Hypothesis 2 is the most plausible. We also suggest that Y. pestis lineages might have developed attenuated virulence during transmission, which can explain the convergent evolutionary signals, including pla decay, that appeared at the end of the pandemics.


Asunto(s)
Peste/epidemiología , Peste/etiología , Peste/genética , ADN Bacteriano/genética , Europa (Continente) , Genoma Bacteriano/genética , Genómica/métodos , Historia del Siglo XIX , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Pandemias/historia , Filogenia , Virulencia/genética , Yersinia pestis/genética , Yersinia pestis/patogenicidad
11.
mBio ; 12(4): e0135821, 2021 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-34340543

RESUMEN

Plague-causing Yersinia pestis is transmitted through regurgitation when it forms a biofilm-mediated blockage in the foregut of its flea vector. This biofilm is composed of an extracellular polysaccharide substance (EPS) produced when cyclic-di-GMP (c-di-GMP) levels are elevated. The Y. pestis diguanylate cyclase enzymes HmsD and HmsT synthesize c-di-GMP. HmsD is required for biofilm blockage formation but contributes minimally to in vitro biofilms. HmsT, however, is necessary for in vitro biofilms and contributes to intermediate rates of biofilm blockage. C-di-GMP synthesis is regulated at the transcriptional and posttranscriptional levels. In this, the global RNA chaperone, Hfq, posttranscriptionally represses hmsT mRNA translation. How c-di-GMP levels and biofilm blockage formation is modulated by nutritional stimuli encountered in the flea gut is unknown. Here, the RNA-binding regulator protein CsrA, which controls c-di-GMP-mediated biofilm formation and central carbon metabolism responses in many Gammaproteobacteria, was assessed for its role in Y. pestis biofilm formation. We determined that CsrA was required for markedly greater c-di-GMP and EPS levels when Y. pestis was cultivated on alternative sugars implicated in flea biofilm blockage metabolism. Our assays, composed of mobility shifts, quantification of mRNA translation, stability, and abundance, and epistasis analyses of a csrA hfq double mutant strain substantiated that CsrA represses hfq mRNA translation, thereby alleviating Hfq-dependent repression of hmsT mRNA translation. Additionally, a csrA mutant exhibited intermediately reduced biofilm blockage rates, resembling an hmsT mutant. Hence, we reveal CsrA-mediated control of c-di-GMP synthesis in Y. pestis as a tiered, posttranscriptional regulatory process that enhances biofilm blockage-mediated transmission from fleas. IMPORTANCE Yersinia pestis, the bacterial agent of bubonic plague, produces a c-di-GMP-dependent biofilm-mediated blockage of the flea vector foregut to facilitate its transmission by flea bite. However, the intricate molecular regulatory processes that underlie c-di-GMP-dependent biofilm formation and thus, biofilm-mediated blockage in response to the nutritional environment of the flea are largely undefined. This study provides a novel mechanistic understanding of how CsrA transduces alternative sugar metabolism cues to induce c-di-GMP-dependent biofilm formation required for efficient Y. pestis regurgitative transmission through biofilm-mediated flea foregut blockage. The Y. pestis-flea interaction represents a unique, biologically relevant, in vivo perspective on the role of CsrA in biofilm regulation.


Asunto(s)
Biopelículas/crecimiento & desarrollo , GMP Cíclico/análogos & derivados , Tracto Gastrointestinal/microbiología , Proteína de Factor 1 del Huésped/genética , ARN Mensajero/genética , Siphonaptera/microbiología , Yersinia pestis/metabolismo , Animales , GMP Cíclico/biosíntesis , Interacciones Huésped-Patógeno , ARN Mensajero/metabolismo , Siphonaptera/anatomía & histología , Yersinia pestis/patogenicidad
12.
PLoS Negl Trop Dis ; 15(8): e0009558, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34343197

RESUMEN

On 12 November 2019, one couple from the Sonid Left Qi (County) in the Inner Mongolia Autonomous Region was diagnosed with pneumonic plague in Beijing. The wife acquired the infection from her husband. Thereafter, two bubonic plague cases were identified in Inner Mongolia on November 16th and 24th. In this study, genome-wide single nucleotide polymorphism (SNP) analysis was used to identify the phylogenetic relationship of Yersinia pestis strains isolated in Inner Mongolia. Strains isolated from reservoirs in 2018 and 2019 in Inner Mongolia, together with the strain isolated from Patient C, were further clustered into 2.MED3m, and two novel lineages (2.MED3q, 2.MED3r) in the 2.MED3 population. According to the analysis of PCR-based molecular subtyping methods, such as the MLVA 14 scheme and seven SNP allele sequencing, Patients A/B and D were classified as 2.MED3m. In addition, strains from rodents living near the patients' residences were clustered into the same lineage as patients. Such observations indicated that human plague cases originated from local reservoirs. Corresponding phylogenetic analysis also indicated that rodent plague strains in different areas in Inner Mongolia belong to different epizootics rather than being caused by spreading from the same epizootic in Meriones unguiculatus in 2019.


Asunto(s)
Peste/epidemiología , Yersinia pestis/genética , Yersinia pestis/patogenicidad , Adulto , Animales , Beijing/epidemiología , China/epidemiología , Resultado Fatal , Femenino , Humanos , Masculino , Persona de Mediana Edad , Filogenia , Peste/etiología , Roedores/microbiología , Yersinia pestis/aislamiento & purificación
13.
Biomolecules ; 11(5)2021 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-34065940

RESUMEN

Plague-a deadly disease caused by the bacterium Yersinia pestis-is still an international public health concern. There are three main clinical forms: bubonic plague, septicemic plague, and pulmonary plague. In all three forms, the symptoms appear suddenly and progress very rapidly. Early antibiotic therapy is essential for countering the disease. Several classes of antibiotics (e.g., tetracyclines, fluoroquinolones, aminoglycosides, sulfonamides, chloramphenicol, rifamycin, and ß-lactams) are active in vitro against the majority of Y. pestis strains and have demonstrated efficacy in various animal models. However, some discrepancies have been reported. Hence, health authorities have approved and recommended several drugs for prophylactic or curative use. Only monotherapy is currently recommended; combination therapy has not shown any benefits in preclinical studies or case reports. Concerns about the emergence of multidrug-resistant strains of Y. pestis have led to the development of new classes of antibiotics and other therapeutics (e.g., LpxC inhibitors, cationic peptides, antivirulence drugs, predatory bacteria, phages, immunotherapy, host-directed therapy, and nutritional immunity). It is difficult to know which of the currently available treatments or therapeutics in development will be most effective for a given form of plague. This is due to the lack of standardization in preclinical studies, conflicting data from case reports, and the small number of clinical trials performed to date.


Asunto(s)
Antibacterianos/uso terapéutico , Inmunoterapia/métodos , Peste/tratamiento farmacológico , Vacunas/uso terapéutico , Yersinia pestis/efectos de los fármacos , Animales , Interacciones Microbiota-Huesped , Humanos , Peste/inmunología , Peste/microbiología , Peste/prevención & control , Yersinia pestis/inmunología , Yersinia pestis/patogenicidad
14.
Pathog Dis ; 79(4)2021 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-33734371

RESUMEN

The human pulmonary environment is complex, containing a matrix of cells, including fibroblasts, epithelial cells, interstitial macrophages, alveolar macrophages and neutrophils. When confronted with foreign material or invading pathogens, these cells mount a robust response. Nevertheless, many bacterial pathogens with an intracellular lifecycle stage exploit this environment for replication and survival. These include, but are not limited to, Coxiella burnetii, Legionella pneumophila, Yersinia pestis, Mycobacterium tuberculosis and Staphylococcus aureus. Currently, few human disease-relevant model systems exist for studying host-pathogen interactions during these bacterial infections in the lung. Here, we present two novel infection platforms, human alveolar macrophages (hAMs) and human precision-cut lung slices (hPCLS), along with an up-to-date synopsis of research using said models. Additionally, alternative uses for these systems in the absence of pathogen involvement are presented, such as tissue banking and further characterization of the human lung environment. Overall, hAMs and hPCLS allow novel human disease-relevant investigations that other models, such as cell lines and animal models, cannot completely provide.


Asunto(s)
Infecciones Bacterianas/microbiología , Interacciones Huésped-Patógeno/inmunología , Enfermedades Pulmonares/microbiología , Pulmón/microbiología , Macrófagos Alveolares/microbiología , Modelos Biológicos , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/patología , Coxiella burnetii/crecimiento & desarrollo , Coxiella burnetii/inmunología , Coxiella burnetii/patogenicidad , Humanos , Legionella pneumophila/crecimiento & desarrollo , Legionella pneumophila/inmunología , Legionella pneumophila/patogenicidad , Pulmón/inmunología , Pulmón/patología , Enfermedades Pulmonares/inmunología , Enfermedades Pulmonares/patología , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/patología , Microtomía , Mycobacterium tuberculosis/crecimiento & desarrollo , Mycobacterium tuberculosis/inmunología , Mycobacterium tuberculosis/patogenicidad , Cultivo Primario de Células , Staphylococcus aureus/crecimiento & desarrollo , Staphylococcus aureus/inmunología , Staphylococcus aureus/patogenicidad , Bancos de Tejidos , Técnicas de Cultivo de Tejidos , Yersinia pestis/crecimiento & desarrollo , Yersinia pestis/inmunología , Yersinia pestis/patogenicidad
15.
PLoS One ; 16(2): e0244615, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33571993

RESUMEN

According to the whole genome SNP analysis of 38 Yersinia pestis strains isolated in the foci of the Northern Caspian and Northern Aral Sea regions in the 20th-early 21st centuries, between 1912 and 2015, the spatial and temporal structure of the 2.MED population of a medieval biovar in this region was determined. A phylogenetic branch 2.MED4 was identified which preceded the 2.MED1 branch that diverged later. 2.MED1 strains became the etiological agent of high-mortality plague outbreaks that occurred in the Northern Caspian region at the beginning of the 20th century. Later in the 20th century, the 2.MED1 branch became widespread in the Caspian Sea region, Caucasus, and vast areas of Central Asia. Based on the data of phylogenetic analysis, as well as epidemiological and epizootiological data, we reconstructed the paths of spread of the 2.MED1 branch in the Northern Caspian Sea region and in the Northern subzone of the Central Asian deserts. It is shown, that the reason for the activation of plague foci in the Northern Caspian region in the second half of the 20th century after a long inter-epizootic period caused by cyclical climate warming was the return of 2.MED1 from the foci of the Northern Aral Sea region. This led to the formation of stable plague foci in the Northern Caspian Sea region and Pre-Caucasus, which manifested epizootic activity in the second half of the 20th and early 21st centuries.


Asunto(s)
Peste/epidemiología , Peste/historia , Yersinia pestis/genética , Evolución Biológica , Mar Caspio , ADN Bacteriano/genética , Evolución Molecular , Genotipo , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Kazajstán/epidemiología , Filogenia , Federación de Rusia/epidemiología , Uzbekistán/epidemiología , Yersinia pestis/metabolismo , Yersinia pestis/patogenicidad
16.
Mol Cell Proteomics ; 20: 100066, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33631294

RESUMEN

Plague is a zoonotic disease that primarily infects rodents via fleabite. Transmission from flea to host niches requires rapid adaption of Yersinia pestis to the outer environments to establish infection. Here, quantitative proteome and secretome analyses of Y. pestis grown under conditions mimicking the two typical niches, i.e., the mammalian host (Mh) and the flea vector (Fv), were performed to understand the adaption strategies of this deadly pathogen. A secretome of Y. pestis containing 308 proteins has been identified using TMT-labeling mass spectrometry analysis. Although some proteins are known to be secreted, such as the type III secretion substrates, PsaA and F1 antigen, most of them were found to be secretory proteins for the first time. Comparative proteomic analysis showed that membrane proteins, chaperonins and stress response proteins are significantly upregulated under the Mh condition, among which the previously uncharacterized proteins YP_3416∼YP_3418 are remarkable because they cannot only be secreted but also translocated into HeLa cells by Y. pestis. We further demonstrated that the purified YP_3416 and YP_3418 exhibited E3 ubiquitin ligase activity in in vitro ubiquitination assay and yp_3416∼3418 deletion mutant of Y. pestis showed significant virulence attenuation in mice. Taken together, our results represent the first Y. pestis secretome, which will promote the better understanding of Y. pestis pathogenesis, as well as the development of new strategies for treatment and prevention of plague.


Asunto(s)
Proteínas Bacterianas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Yersinia pestis/metabolismo , Yersinia pestis/patogenicidad , Animales , Proteínas Bacterianas/genética , Femenino , Células HeLa , Humanos , Ratones Endogámicos BALB C , Mutación , Peste , Proteómica , Secretoma , Ubiquitina-Proteína Ligasas/genética , Virulencia , Yersinia pestis/genética
17.
Infect Immun ; 89(2)2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33257531

RESUMEN

Yersinia pestis is a highly virulent pathogen and the causative agent of bubonic, septicemic, and pneumonic plague. Primary pneumonic plague caused by inhalation of respiratory droplets contaminated with Y. pestis is nearly 100% lethal within 4 to 7 days without antibiotic intervention. Pneumonic plague progresses in two phases, beginning with extensive bacterial replication in the lung with minimal host responsiveness, followed by the abrupt onset of a lethal proinflammatory response. The precise mechanisms by which Y. pestis is able to colonize the lung and survive two very distinct disease phases remain largely unknown. To date, a few bacterial virulence factors, including the Ysc type 3 secretion system, are known to contribute to the pathogenesis of primary pneumonic plague. The bacterial GTPase BipA has been shown to regulate expression of virulence factors in a number of Gram-negative bacteria, including Pseudomonas aeruginosa, Escherichia coli, and Salmonella enterica serovar Typhi. However, the role of BipA in Y. pestis has yet to be investigated. Here, we show that BipA is a Y. pestis virulence factor that promotes defense against early neutrophil-mediated bacterial killing in the lung. This work identifies a novel Y. pestis virulence factor and highlights the importance of early bacterial/neutrophil interactions in the lung during primary pneumonic plague.


Asunto(s)
Proteínas Bacterianas/fisiología , GTP Fosfohidrolasas/fisiología , Peste/inmunología , Peste/fisiopatología , Factores de Virulencia/fisiología , Yersinia pestis/inmunología , Yersinia pestis/patogenicidad , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Modelos Animales
18.
Diagn Microbiol Infect Dis ; 99(3): 115275, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33360431

RESUMEN

CRISPR-associated proteins that produce a signal in the presence of a target nucleic acid represent potentially powerful tools for diagnostics, but they also exhibit shortfalls that plague many CRISPR systems. For instance, not all targets elicit robust activity, which challenges the timely development of sensitive assays, and though many such tests have been reported, they often avoid discussion of the crRNA design and screening process. Here, motivated by the desire to detect the Yersinia pestis lcrV virulence gene, we detail the process involved in developing components for a CRISPR-based test that provides sensitive and specific identification of this sequence using Cas13a. This includes detailing the diversity of crRNA performance, identifying sequence that enable detection with attomolar sensitivity and species-level specificity, and presenting a method for simple streamlining of the crRNA screening process to allow for the high-throughput testing required for developing assay design rules in the future.


Asunto(s)
Antígenos Bacterianos/genética , Sistemas CRISPR-Cas/genética , Ensayos Analíticos de Alto Rendimiento/métodos , Patología Molecular/métodos , Proteínas Citotóxicas Formadoras de Poros/genética , ARN Bacteriano/genética , Yersinia pestis/genética , Línea Celular , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Humanos , Virulencia , Yersinia pestis/patogenicidad
19.
Ned Tijdschr Geneeskd ; 1642020 12 03.
Artículo en Holandés | MEDLINE | ID: mdl-33332041

RESUMEN

The plague epidemics wiped out large parts of the city population from the 15th to the 17th century in the Netherlands. The plague bacterium (Yersinia pestis) is transmitted to humans through infected rats and fleas and has been transferred from China to Europe via the trade routes over land and sea. Meetings were banned, plague victims were isolated at home or in pest houses, and ships quarantined. In the densely populated, poor neighborhoods of the cities, however, isolation and keeping distance were not feasible, which allowed the plague to rapidly spread. The lessons we have learned from the plague epidemics are timeless. Isolation, keeping your distance and quarantine were key principles and now apply again in the approach to the current Covid-19 pandemic. How effective these measures are depends on the social context in which they are applied.


Asunto(s)
COVID-19 , Pandemias , Distanciamiento Físico , Peste , Cuarentena , Animales , COVID-19/epidemiología , COVID-19/prevención & control , Reservorios de Enfermedades , Transmisión de Enfermedad Infecciosa/prevención & control , Vectores de Enfermedades , Historia del Siglo XV , Historia del Siglo XVI , Historia del Siglo XVII , Humanos , Países Bajos/epidemiología , Pandemias/historia , Pandemias/prevención & control , Peste/epidemiología , Peste/historia , Peste/microbiología , Peste/prevención & control , Cuarentena/historia , Cuarentena/métodos , SARS-CoV-2 , Yersinia pestis/patogenicidad
20.
PLoS Pathog ; 16(12): e1009092, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33284863

RESUMEN

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.


Asunto(s)
Peste/transmisión , Siphonaptera/fisiología , Yersinia pestis/metabolismo , Animales , Biopelículas/crecimiento & desarrollo , Brotes de Enfermedades , Progresión de la Enfermedad , Femenino , Insectos Vectores/fisiología , Ratones , Siphonaptera/metabolismo , Siphonaptera/microbiología , Yersinia pestis/patogenicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...