Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 263
Filtrar
1.
Microbiol Spectr ; 9(1): e0000521, 2021 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-34132578

RESUMEN

Current methods for screening small molecules that inhibit the plasmid pCD1-encoded Yersinia pestis type III secretion system (T3SS) include lengthy growth curves followed by multistep luminescence assays or Western blot assays to detect secretion, or lack thereof, of effector proteins. The goal of this research was to develop a novel disk diffusion assay on magnesium oxalate (MOX) agar as a simple way to evaluate the susceptibility of Y. pestis to type III secretion system inhibitors. MOX agar produces distinct Y. pestis growth characteristics based on the bacteria's ability or inability to secrete effector proteins; small, barely visible colonies are observed when secretion is activated versus larger, readily visible colonies when secretion is inhibited. Wild-type Y. pestis was diluted and spread onto a MOX agar plate. Disks containing 20 µl of various concentrations of imidocarb dipropionate, a known Y. pestis T3SS inhibitor, or distilled water (dH2O) were placed on the plate. After incubation at 37°C for 48 h, visible colonies of Y. pestis were observed surrounding the disks with imidocarb dipropionate, suggesting that T3S was inhibited. The diameter of the growth of colonies surrounding the disks increased as the concentration of the T3SS inhibitor increased. Imidocarb dipropionate was also able to inhibit Y. pestis strains lacking effector Yops and Yop chaperones, suggesting that they are not necessary for T3S inhibition. This disk diffusion assay is a feasible and useful method for testing the susceptibility of Y. pestis to type III secretion system inhibitors and has the potential to be used in a clinical setting. IMPORTANCE Disk diffusion assays have traditionally been used as a simple and effective way to screen compounds for antibacterial activity and to determine the susceptibility of pathogens to antibiotics; however, they are limited to detecting growth inhibition only. Consequently, antimicrobial agents that inhibit virulence factors, but not growth, would not be detected. Therefore, we developed a disk diffusion assay that could detect inhibition of bacterial virulence factors, specifically, type III secretion systems (T3SSs), needle-like structures used by several pathogenic bacteria to inject host cells with effector proteins and cause disease. We demonstrate that magnesium oxalate (MOX) agar can be used in a disk diffusion assay to detect inhibition of the T3SS of Yersinia pestis, the causative agent of bubonic plague, by small-molecule inhibitors. This assay may be useful for screening additional small molecules that target bacterial T3SSs or testing the susceptibility of patient-derived samples to drugs that target T3SSs.


Asunto(s)
Antibacterianos/farmacología , Proteínas Bacterianas/antagonistas & inhibidores , Pruebas Antimicrobianas de Difusión por Disco/métodos , Ácido Oxálico/farmacología , Sistemas de Secreción Tipo III/antagonistas & inhibidores , Yersinia pestis/efectos de los fármacos , Proteínas Bacterianas/metabolismo , Pruebas Antimicrobianas de Difusión por Disco/instrumentación , Humanos , Peste/microbiología , Sistemas de Secreción Tipo III/metabolismo , Yersinia pestis/crecimiento & desarrollo , Yersinia pestis/metabolismo
2.
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
3.
Biotechnol Prog ; 37(2): e3103, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33190425

RESUMEN

Yersiniabactin (Ybt) is a hybrid polyketide-nonribosomal complex natural product also known as a siderophore for its iron chelation properties. The native producer of Ybt, Yersinia pestis, is a priority pathogen responsible for the plague in which the siderophore properties of Ybt are used to sequester iron and other metal species upon host infection. Alternatively, the high metal binding properties of Ybt enable a plethora of potentially valuable applications benefiting from metal remediation and/or recovery. For these applications, a surrogate production source is highly preferred relative to the pathogenic native host. In this work, we present a modification to the heterologous Escherichia coli production system established for Ybt biosynthesis. In particular, the multiple plasmids originally used to express the genetic pathway required for Ybt biosynthesis were consolidated to a single, copy-amplifiable plasmid. In so doing, plasmid stability was improved from ~30% to ≥80% while production values maintained at 20-30% of the original system, which resulted in titers of 0.5-3 mg/L from shake flask vessels.


Asunto(s)
Escherichia coli/metabolismo , Hierro/metabolismo , Fenoles/metabolismo , Plásmidos/genética , Sideróforos/biosíntesis , Tiazoles/metabolismo , Yersinia pestis/metabolismo , Escherichia coli/genética , Escherichia coli/crecimiento & desarrollo , Plásmidos/química , Plásmidos/metabolismo , Yersinia pestis/genética , Yersinia pestis/crecimiento & desarrollo
4.
PLoS Negl Trop Dis ; 14(9): e0008688, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32946437

RESUMEN

Yersinia pestis, the causative agent of plague, is a highly lethal pathogen transmitted by the bite of infected fleas. Once ingested by a flea, Y. pestis establish a replicative niche in the gut and produce a biofilm that promotes foregut colonization and transmission. The rat flea Xenopsylla cheopis is an important vector to several zoonotic bacterial pathogens including Y. pestis. Some fleas naturally clear themselves of infection; however, the physiological and immunological mechanisms by which this occurs are largely uncharacterized. To address this, RNA was extracted, sequenced, and distinct transcript profiles were assembled de novo from X. cheopis digestive tracts isolated from fleas that were either: 1) not fed for 5 days; 2) fed sterile blood; or 3) fed blood containing ~5x108 CFU/ml Y. pestis KIM6+. Analysis and comparison of the transcript profiles resulted in identification of 23 annotated (and 11 unknown or uncharacterized) digestive tract transcripts that comprise the early transcriptional response of the rat flea gut to infection with Y. pestis. The data indicate that production of antimicrobial peptides regulated by the immune-deficiency pathway (IMD) is the primary flea immune response to infection with Y. pestis. The remaining infection-responsive transcripts, not obviously associated with the immune response, were involved in at least one of 3 physiological themes: 1) alterations to chemosensation and gut peristalsis; 2) modification of digestion and metabolism; and 3) production of chitin-binding proteins (peritrophins). Despite producing several peritrophin transcripts shortly after feeding, including a subset that were infection-responsive, no thick peritrophic membrane was detectable by histochemistry or electron microscopy of rat flea guts for the first 24 hours following blood-feeding. Here we discuss the physiological implications of rat flea infection-responsive transcripts, the function of X. cheopis peritrophins, and the mechanisms by which Y. pestis may be cleared from the flea gut.


Asunto(s)
Tracto Gastrointestinal/microbiología , Transcriptoma , Xenopsylla/microbiología , Yersinia pestis/genética , Yersinia pestis/metabolismo , Animales , Biopelículas , Epitelio/microbiología , Epitelio/patología , Femenino , Tracto Gastrointestinal/patología , Perfilación de la Expresión Génica , Insectos Vectores/microbiología , Peste/microbiología , Peste/veterinaria , Ratas , Análisis de Secuencia de ARN , Yersinia pestis/crecimiento & desarrollo , Yersinia pestis/aislamiento & purificación
5.
J Bacteriol ; 202(20)2020 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-32778558

RESUMEN

Cohesion of biofilms made by Yersinia pestis and Yersinia pseudotuberculosis has been attributed solely to an extracellular polysaccharide matrix encoded by the hms genes (Hms-dependent extracellular matrix [Hms-ECM]). However, mutations in the Y. pseudotuberculosis BarA/UvrY/CsrB regulatory cascade enhance biofilm stability without dramatically increasing Hms-ECM production. We found that treatment with proteinase K enzyme effectively destabilized Y. pseudotuberculosiscsrB mutant biofilms, suggesting that cell-cell interactions might be mediated by protein adhesins or extracellular matrix proteins. We identified an uncharacterized trimeric autotransporter lipoprotein (YPTB2394), repressed by csrB, which has been referred to as YadE. Biofilms made by a ΔyadE mutant strain were extremely sensitive to mechanical disruption. Overexpression of yadE in wild-type Y. pseudotuberculosis increased biofilm cohesion, similar to biofilms made by csrB or uvrY mutants. We found that the Rcs signaling cascade, which represses Hms-ECM production, activated expression of yadE The yadE gene appears to be functional in Y. pseudotuberculosis but is a pseudogene in modern Y. pestis strains. Expression of functional yadE in Y. pestis KIM6+ weakened biofilms made by these bacteria. This suggests that although the YadE autotransporter protein increases Y. pseudotuberculosis biofilm stability, it may be incompatible with the Hms-ECM production that is essential for Y. pestis biofilm production in fleas. Inactivation of yadE in Y. pestis may be another instance of selective gene loss in the evolution of flea-borne transmission by this species.IMPORTANCE The evolution of Yersinia pestis from its Y. pseudotuberculosis ancestor involved gene acquisition and gene losses, leading to differences in biofilm production. Characterizing the unique biofilm features of both species may provide better understanding of how each adapts to its specific niches. This study identifies a trimeric autotransporter, YadE, that promotes biofilm stability of Y. pseudotuberculosis but which has been inactivated in Y. pestis, perhaps because it is not compatible with the Hms polysaccharide that is crucial for biofilms inside fleas. We also reveal that the Rcs signaling cascade, which represses Hms expression, activates YadE in Y. pseudotuberculosis The ability of Y. pseudotuberculosis to use polysaccharide or YadE protein for cell-cell adhesion may help it produce biofilms in different environments.


Asunto(s)
Proteínas Bacterianas/metabolismo , Biopelículas/crecimiento & desarrollo , Regulación Bacteriana de la Expresión Génica , Yersinia pestis/crecimiento & desarrollo , Yersinia pseudotuberculosis/crecimiento & desarrollo , Animales , Proteínas Bacterianas/genética , Seudogenes , Selección Genética , Siphonaptera/microbiología , Sistemas de Secreción Tipo V/metabolismo , Yersinia pestis/genética , Yersinia pseudotuberculosis/genética , Infecciones por Yersinia pseudotuberculosis/microbiología , Infecciones por Yersinia pseudotuberculosis/transmisión
6.
J Bacteriol ; 202(11)2020 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-32205462

RESUMEN

While alternating between insects and mammals during its life cycle, Yersinia pestis, the flea-transmitted bacterium that causes plague, regulates its gene expression appropriately to adapt to these two physiologically disparate host environments. In fleas competent to transmit Y. pestis, low-GC-content genes y3555, y3551, and y3550 are highly transcribed, suggesting that these genes have a highly prioritized role in flea infection. Here, we demonstrate that y3555, y3551, and y3550 are transcribed as part of a single polycistronic mRNA comprising the y3555, y3554, y3553, y355x, y3551, and y3550 genes. Additionally, y355x-y3551-y3550 compose another operon, while y3550 can be also transcribed as a monocistronic mRNA. The expression of these genes is induced by hyperosmotic salinity stress, which serves as an explicit environmental stimulus that initiates transcriptional activity from the predicted y3550 promoter. Y3555 has homology to pyridoxal 5'-phosphate (PLP)-dependent aromatic aminotransferases, while Y3550 and Y3551 are homologous to the Rid protein superfamily (YjgF/YER057c/UK114) members that forestall damage caused by reactive intermediates formed during PLP-dependent enzymatic activity. We demonstrate that y3551 specifically encodes an archetypal RidA protein with 2-aminoacrylate deaminase activity but Y3550 lacks Rid deaminase function. Heterologous expression of y3555 generates a critical aspartate requirement in a Salmonella entericaaspC mutant, while its in vitro expression, and specifically its heterologous coexpression with y3550, enhances the growth rate of an Escherichia coli ΔaspC ΔtyrB mutant in a defined minimal amino acid-supplemented medium. Our data suggest that the y3555, y3551, and y3550 genes operate cooperatively to optimize aromatic amino acid metabolism and are induced under conditions of hyperosmotic salinity stress.IMPORTANCE Distinct gene repertoires are expressed during Y. pestis infection of its flea and mammalian hosts. The functions of many of these genes remain predicted or unknown, necessitating their characterization, as this may provide a better understanding of Y. pestis specialized biological adaptations to the discrete environments of its two hosts. This study provides functional context to adjacently clustered horizontally acquired genes predominantly expressed in the flea host by deciphering their fundamental processes with regard to (i) transcriptional organization, (ii) transcription activation signals, and (iii) biochemical function. Our data support a role for these genes in osmoadaptation and aromatic amino acid metabolism, highlighting these as preferential processes by which Y. pestis gene expression is modulated during flea infection.


Asunto(s)
Aminoácidos Aromáticos/metabolismo , Siphonaptera/microbiología , Yersinia pestis/metabolismo , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Transferencia de Gen Horizontal , Operón , Yersinia pestis/genética , Yersinia pestis/crecimiento & desarrollo
7.
Artículo en Inglés | MEDLINE | ID: mdl-32015027

RESUMEN

Pneumonic plague, caused by the Gram-negative bacteria Yersinia pestis, is an invasive, rapidly progressing disease with poor survival rates. Following inhalation of Y. pestis, bacterial invasion of the lungs and a tissue-damaging inflammatory response allows vascular spread of the infection. Consequently, primary pneumonic plague is a multiorgan disease involving sepsis and necrosis of immune tissues and the liver, as well as bronchopneumonia and rampant bacterial growth. Given the likely role of the hyperinflammatory response in accelerating the destruction of tissue, in this work we evaluated the therapeutic potential of the inducible cytoprotective enzyme heme oxygenase 1 (HO-1) against primary pneumonic plague. On its own, the HO-1 inducer cobalt protoporphyrin IX (CoPP) provided mice protection from lethal challenge with Y. pestis CO92 with improved pulmonary bacterial clearance and a dampened inflammatory response compared to vehicle-treated mice. Furthermore, CoPP treatment combined with doxycycline strongly enhanced protection in a rat aerosol challenge model. Compared to doxycycline alone, CoPP treatment increased survival, with a 3-log decrease in median bacterial titer recovered from the lungs and the general absence of a systemic hyperinflammatory response. In contrast, treatment with the HO-1 inhibitor SnPP had no detectable impact on doxycycline efficacy. The combined data indicate that countering inflammatory toxicity by therapeutically inducing HO-1 is effective in reducing the rampant growth of Y. pestis and preventing pneumonic plague.


Asunto(s)
Antibacterianos/uso terapéutico , Doxiciclina/uso terapéutico , Hemo-Oxigenasa 1/metabolismo , Peste/prevención & control , Protoporfirinas/uso terapéutico , Yersinia pestis/efectos de los fármacos , Aerosoles , Animales , Bronconeumonía/microbiología , Bronconeumonía/patología , Modelos Animales de Enfermedad , Quimioterapia Combinada , Femenino , Hemo-Oxigenasa 1/genética , Humanos , Pulmón/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Peste/tratamiento farmacológico , Peste/microbiología , Ratas , Ratas Sprague-Dawley , Yersinia pestis/crecimiento & desarrollo
8.
Methods Mol Biol ; 2010: 69-84, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31177432

RESUMEN

Laser scanning microscopy (LSM) is a technology that allows for direct observations of host-pathogen interactions during infection. Two of the most available forms of LSM are confocal and two-photon LSM. In addition to high resolution and contrast, these two technologies also provide high excitation penetrance in unsectioned samples. High penetrance allows for imaging of layers of tissue that are difficult to image with other more conventional microscopy approaches. Thus, confocal and two-photon LSM open the possibility of observing infection in a three-dimensional context, where the natural architecture of a tissue is preserved. Few studies have used LSM technology to gain insights into Yersinia pestis pathogenesis in the mammalian host. The use of LSM in the plague field has an enormous potential for the discovery of the mechanisms that lie behind key aspects of pathogenesis such as colonization, dissemination, and tissue damage. This chapter provides guidance for the implementation of confocal or two-photon LSM to study Y. pestis interactions with the host in unsectioned tissues. This document provides specific instructions applied to imaging of Y. pestis, and also discusses relevant aspects of imaging, such as the operation of laser scanning microscopes and the use of fluorescent probes.


Asunto(s)
Microscopía Confocal/métodos , Peste/patología , Yersinia pestis/aislamiento & purificación , Animales , Modelos Animales de Enfermedad , Diseño de Equipo , Ratones , Microscopía Confocal/instrumentación , Imagen Óptica/métodos , Coloración y Etiquetado/métodos , Fijación del Tejido/métodos , Yersinia pestis/crecimiento & desarrollo
9.
Methods Mol Biol ; 2010: 167-178, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31177438

RESUMEN

With the limited availability of genomic sequence information and no established methods for genetic knockdowns or the creation of transgenic fleas and flea cell lines, we have adopted Drosophila melanogaster as a model for the study of the insect life cycle of Yersinia pestis. Infection of Drosophila larvae can be used to model early colonization of fleas, while the established embryonic cell lines can be used to model insect-pathogen interactions that underlie the unique capacity of Y. pestis to colonize the gut of its flea host. In this chapter, we present the methods we developed for infection of Drosophila in vivo and in vitro.


Asunto(s)
Drosophila melanogaster/microbiología , Insectos Vectores/microbiología , Peste/microbiología , Yersinia pestis/crecimiento & desarrollo , Animales , Línea Celular , Larva/microbiología , Peste/transmisión
10.
Methods Mol Biol ; 2010: 181-196, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31177439

RESUMEN

Yersinia pestis is able to survive and replicate within macrophages, while also being able to live in the extracellular milieu of the host. Assays that facilitate better understanding of how Y. pestis survives intracellularly and subverts normal host antimicrobial defenses require the ability to monitor intracellular Y. pestis survival and replication. In this chapter three different assays for monitoring intracellular survival and replication will be described, along with the formulas and methods to quantify and present the acquired data. These assays are fundamental to answering a multitude of questions pertaining to which bacterial factors are important for intracellular survival. Additionally, these assays can be used, with modifications, for other intracellular pathogens of interest. The first assay discussed will be the conventional bacterial enumeration assay, which quantifies bacterial numbers directly through a classic colony forming units (CFU) assay. Quantifying bacterial burden through CFU determination allows for differentiation between intracellular/cell-associated bacteria and extracellular bacteria. However, CFU determination is laborious, does not allow for direct kinetic monitoring of bacterial growth, and is difficult to adapt to high throughput assays. Bioluminescence bioreporters that use luciferase to monitor bacterial numbers allow for simple, plate reader-based, real-time kinetic monitoring of bacterial growth that is amendable to high throughput techniques. Finally, we will describe live cell microscopy using fluorescent bioreporters, which allows for monitoring of bacterial replication in individual cells and the possibility to visualize interactions between bacterial and host proteins during intracellular infection.


Asunto(s)
Macrófagos/microbiología , Peste/patología , Yersinia pestis/fisiología , Animales , Recuento de Colonia Microbiana/métodos , Humanos , Mediciones Luminiscentes/métodos , Macrófagos/patología , Ratones , Peste/microbiología , Células RAW 264.7 , Yersinia pestis/crecimiento & desarrollo
12.
Artículo en Inglés | MEDLINE | ID: mdl-31069175

RESUMEN

Yersinia pestis is the etiological agent of the notorious plague that has claimed millions of deaths in history. Of the four known Y. pestis biovars (Antiqua, Medievalis, Orientalis, and Microtus), Microtus strains are unique for being highly virulent in mice but avirulent in humans. Here, human peripheral lymphocytes were infected with the fully virulent 141 strain or the Microtus strain 201, and their transcriptomes were determined and compared. The most notable finding was that robust responses in the pathways for cytokine-cytokine receptor interaction, chemokine signaling pathway, Toll-like receptor signaling and Jak-STAT signaling were induced at 2 h post infection (hpi) in the 201- but not the 141-infected lymphocytes, suggesting that human lymphocytes might be able to constrain infections caused by strain 201 but not 141. Consistent with the transcriptome results, much higher IFN-γ and IL-1ß were present in the supernatants from the 201-infected lymphocytes, while inflammatory inhibitory IL-10 levels were higher in the 141-infected lymphocytes. The expressions of CSTD and SLC11A1, both of which are functional components of the lysosome, increased in the 201-infected human macrophage-like U937 cells. Further assessment of the survival rate of the 201 bacilli in the U937 cells and murine macrophage RAW 264.7 cells revealed no viable bacteria in the U937 cells at 32 hpi.; however, about 5-10% of the bacteria were still alive in the RAW264.7 cells. Our results indicate that human macrophages can clear the intracellular Y. pestis 201 bacilli more efficiently than murine macrophages, probably by interfering with critical host immune responses, and this could partially account for the host-specific pathogenicity of Y. pestis Microtus strains.


Asunto(s)
Linfocitos/inmunología , Linfocitos/microbiología , Macrófagos/inmunología , Macrófagos/microbiología , Yersinia pestis/crecimiento & desarrollo , Yersinia pestis/inmunología , Animales , Células Cultivadas , Citocinas/metabolismo , Perfilación de la Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Ratones , Viabilidad Microbiana
13.
Sci Rep ; 9(1): 2501, 2019 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-30792499

RESUMEN

Reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR) is a very sensitive widespread technique considered as the gold standard to explore transcriptional variations. While a particular methodology has to be followed to provide accurate results many published studies are likely to misinterpret results due to lack of minimal quality requirements. Yersinia pestis is a highly pathogenic bacterium responsible for plague. It has been used to propose a ready-to-use and complete approach to mitigate the risk of technical biases in transcriptomic studies. The selection of suitable reference genes (RGs) among 29 candidates was performed using four different methods (GeNorm, NormFinder, BestKeeper and the Delta-Ct method). An overall comprehensive ranking revealed that 12 following candidate RGs are suitable for accurate normalization: gmk, proC, fabD, rpoD, nadB, rho, thrA, ribD, mutL, rpoB, adk and tmk. Some frequently used genes like 16S RNA had even been found as unsuitable to study Y. pestis. This methodology allowed us to demonstrate, under different temperatures and states of growth, significant transcriptional changes of six efflux pumps genes involved in physiological aspects as antimicrobial resistance or virulence. Previous transcriptomic studies done under comparable conditions had not been able to highlight these transcriptional modifications. These results highlight the importance of validating RGs prior to the normalization of transcriptional expression levels of targeted genes. This accurate methodology can be extended to any gene of interest in Y. pestis. More generally, the same workflow can be applied to identify and validate appropriate RGs in other bacteria to study transcriptional variations.


Asunto(s)
Proteínas Bacterianas/genética , Perfilación de la Expresión Génica/normas , Yersinia pestis/crecimiento & desarrollo , Biología Computacional , Regulación Bacteriana de la Expresión Génica , Estándares de Referencia , Temperatura , Flujo de Trabajo , Yersinia pestis/genética
14.
Infect Immun ; 87(4)2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30642901

RESUMEN

Yersinia pestis causes bubonic, pneumonic, and septicemic plague. Although no longer responsible for pandemic outbreaks, pneumonic plague continues to be a challenge for medical treatment and has been classified as a reemerging disease in some parts of the world. In the early stage of infection, inflammatory responses are believed to be suppressed by Y. pestis virulence factors in order to prevent clearance, while later, the hyperactivation of inflammation contributes to the progression of disease. In this work, we sought to identify the host factors that mediate this process and studied the role of the Toll/interleukin 1 (IL-1) receptor adapter and major inflammatory mediator myeloid differentiation primary response 88 (MyD88) in pneumonic plague. We show that pulmonary challenge of Myd88-/- mice with wild-type (WT) Y. pestis results in significant loss of pro- and anti-inflammatory cytokines and chemokines, especially gamma interferon (IFN-γ) and KC, in the lungs compared to that in WT mice. Bacterial growth in the lungs occurred more rapidly in the WT mice, however, indicating a role for the MyD88 response in facilitating the primary lung infection. Nevertheless, Myd88-/- mice were more sensitive to lethality from secondary septicemic plague. Together these findings indicate a central role for MyD88 during the biphasic inflammatory response to pulmonary Y. pestis infection. In the early phase, low-level MyD88-dependent chemokine expression limits initial growth but facilitates Y. pestis access to a protected replicative niche. The later hyperinflammatory phase is partially MyD88 dependent and ineffective in the lungs but controls systemic infection and reduces the progression of secondary septicemic plague.


Asunto(s)
Pulmón/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Peste/metabolismo , Peste/microbiología , Yersinia pestis/crecimiento & desarrollo , Animales , Quimiocinas/genética , Quimiocinas/metabolismo , Citocinas/genética , Citocinas/metabolismo , Femenino , Humanos , Pulmón/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Factor 88 de Diferenciación Mieloide/genética , Peste/genética , Virulencia , Yersinia pestis/genética , Yersinia pestis/metabolismo , Yersinia pestis/patogenicidad
15.
BMC Microbiol ; 18(1): 218, 2018 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-30563467

RESUMEN

BACKGROUND: For Yersinia pestis, Burkholderia pseudomallei, and Burkholderia mallei, conventional broth microdilution (BMD) is considered the gold standard for antimicrobial susceptibility testing (AST) and, depending on the species, requires an incubation period of 16-20 h, or 24-48 h according to the Clinical and Laboratory Standards Institute (CLSI) guidelines. After a diagnosis of plague, melioidosis or glanders during an outbreak or after an exposure event, the timely distribution of appropriate antibiotics for treatment or post-exposure prophylaxis of affected populations could reduce mortality rates. RESULTS: Herein, we developed and evaluated a rapid, automated susceptibility test for these Gram-negative bacterial pathogens based on time-lapse imaging of cells incubating in BMD microtitre drug panels using an optical screening instrument (oCelloScope). In real-time, the instrument screened each inoculated well containing broth with various concentrations of antibiotics published by CLSI for primary testing: ciprofloxacin (CIP), doxycycline (DOX) and gentamicin (GEN) for Y. pestis; imipenem (IPM), ceftazidime (CAZ) and DOX for B. mallei; and IPM, DOX, CAZ, amoxicillin-clavulanic acid (AMC) and trimethoprim-sulfamethoxazole (SXT) for B. pseudomallei. Based on automated growth kinetic data, the time required to accurately determine susceptibility decreased by ≥70% for Y. pestis and ≥ 50% for B. mallei and B. pseudomallei compared to the times required for conventional BMD testing. Susceptibility to GEN, IPM and DOX could be determined in as early as three to six hours. In the presence of CAZ, susceptibility based on instrument-derived growth values could not be determined for the majority of B. pseudomallei and B. mallei strains tested. Time-lapse video imaging of these cultures revealed that the formation of filaments in the presence of this cephalosporin at inhibitory concentrations was detected as growth. Other ß-lactam-induced cell morphology changes, such as the formation of spheroplasts and rapid cell lysis, were also observed and appear to be strain- and antibiotic concentration-dependent. CONCLUSIONS: A rapid, functional AST was developed and real-time video footage captured ß-lactam-induced morphologies of wild-type B. mallei and B. pseudomallei strains in broth. Optical screening reduced the time to results required for AST of three Gram-negative biothreat pathogens using clinically relevant, first-line antibiotics compared to conventional BMD.


Asunto(s)
Antibacterianos/farmacología , Burkholderia mallei/efectos de los fármacos , Burkholderia pseudomallei/efectos de los fármacos , Pruebas de Sensibilidad Microbiana/métodos , Imagen de Lapso de Tiempo/métodos , Yersinia pestis/efectos de los fármacos , beta-Lactamas/farmacología , Burkholderia mallei/citología , Burkholderia mallei/crecimiento & desarrollo , Burkholderia mallei/fisiología , Burkholderia pseudomallei/crecimiento & desarrollo , Burkholderia pseudomallei/fisiología , Ciprofloxacina/farmacología , Doxiciclina/farmacología , Muermo/microbiología , Humanos , Imipenem/farmacología , Melioidosis/microbiología , Peste/microbiología , Yersinia pestis/citología , Yersinia pestis/crecimiento & desarrollo , Yersinia pestis/fisiología
16.
Artículo en Inglés | MEDLINE | ID: mdl-30333962

RESUMEN

Biofilm formation is critical for blocking flea foregut and hence for transmission of Y. pestis by flea biting. In this study, we identified the regulatory role of the AraC-family transcriptional regulator BfvR (YPO1737 in strain CO92) in biofilm formation and virulence of Yersinia pestis biovar Microtus. Crystal violet staining, Caenorhabditis elegans biofilm assay, colony morphology assay, intracellular c-di-GMP concentration determination, and BALB/c mice challenge were employed to reveal that BfvR enhanced Y. pestis biofilm formation while repressed its virulence in mice. Further molecular biological assays demonstrated that BfvR directly stimulated the expression of hmsHFRS, waaAE-coaD, and hmsCDE, which, in turn, affected the production of exopolysaccharide, LPS, and c-di-GMP, respectively. In addition, BfvR directly and indirectly repressed psaABC and psaEF transcription, respectively. We concluded that the modulation of biofilm- and virulence-related genes by BfvR led to increased biofilm formation and reduced virulence of Y. pestis biovar Microtus.


Asunto(s)
Antígenos Bacterianos/metabolismo , Biopelículas/crecimiento & desarrollo , Regulación Bacteriana de la Expresión Génica , Factores de Transcripción/metabolismo , Yersinia pestis/crecimiento & desarrollo , Yersinia pestis/metabolismo , Animales , Caenorhabditis elegans/microbiología , GMP Cíclico/análogos & derivados , GMP Cíclico/análisis , Modelos Animales de Enfermedad , Redes Reguladoras de Genes , Genes Reguladores , Ratones Endogámicos BALB C , Peste/microbiología , Peste/patología , Polisacáridos Bacterianos/metabolismo , Análisis de Supervivencia , Virulencia , Yersinia pestis/genética
17.
BMC Microbiol ; 18(1): 46, 2018 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-29855259

RESUMEN

BACKGROUND: Yersinia pseudotuberculosis is a zoonotic pathogen, causing mild gastrointestinal infection in humans. From this comparatively benign pathogenic species emerged the highly virulent plague bacillus, Yersinia pestis, which has experienced significant genetic divergence in a relatively short time span. Much of our knowledge of Yersinia spp. evolution stems from genomic comparison and gene expression studies. Here we apply transposon-directed insertion site sequencing (TraDIS) to describe the essential gene set of Y. pseudotuberculosis IP32953 in optimised in vitro growth conditions, and contrast these with the published essential genes of Y. pestis. RESULTS: The essential genes of an organism are the core genetic elements required for basic survival processes in a given growth condition, and are therefore attractive targets for antimicrobials. One such gene we identified is yptb3665, which encodes a peptide deformylase, and here we report for the first time, the sensitivity of Y. pseudotuberculosis to actinonin, a deformylase inhibitor. Comparison of the essential genes of Y. pseudotuberculosis with those of Y. pestis revealed the genes whose importance are shared by both species, as well as genes that were differentially required for growth. In particular, we find that the two species uniquely rely upon different iron acquisition and respiratory metabolic pathways under similar in vitro conditions. CONCLUSIONS: The discovery of uniquely essential genes between the closely related Yersinia spp. represent some of the fundamental, species-defining points of divergence that arose during the evolution of Y. pestis from its ancestor. Furthermore, the shared essential genes represent ideal candidates for the development of novel antimicrobials against both species.


Asunto(s)
Genes Esenciales , Mutagénesis Insercional/métodos , Yersinia pestis/crecimiento & desarrollo , Yersinia pseudotuberculosis/crecimiento & desarrollo , Proteínas Bacterianas/genética , Elementos Transponibles de ADN , Evolución Molecular , Especiación Genética , Humanos , Análisis de Secuencia de ADN , Yersinia pestis/genética , Yersinia pseudotuberculosis/genética
18.
Microbes Infect ; 20(3): 166-175, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29180033

RESUMEN

Yersinia pestis has evolved from Yersinia pseudotuberculosis serotype O:1b. A typical Y. pestis contains three plasmids: pCD1, pMT1 and pPCP1. However, some isolates only harbor pCD1 (pCD1+-mutant). Y. pestis and Y. pseudotuberculosis share a common plasmid (pCD1 or pYV), but little is known about whether Y. pseudotuberculosis exhibited plague-inducing potential before it was evolved into Y. pestis. Here, the luxCDABE::Tn5::kan was integrated into the chromosome of the pCD1+-mutant, Y. pseudotuberculosis or Escherichia coli K12 to construct stable bioluminescent strains for investigation of their dissemination in mice by bioluminescence imaging technology. After subcutaneous infection, the pCD1+-mutant entered the lymph nodes, followed by the liver and spleen, and, subsequently, the lungs, causing pathological changes in these organs. Y. pseudotuberculosis entered the lymph nodes, but not the liver, spleen and lungs. It also resided in the lymph nodes for several days, but did not cause lymphadenitis or pathological lesions. By contrast, E. coli K12-lux was not isolatable from mouse lymph nodes, liver, spleen and lungs. These results indicate that the pCD1+-mutant can cause typical bubonic and pneumonic plague-like diseases, and Y. pestis has inherited lymphoid tissue tropism from its ancestor rather than acquiring these properties independently.


Asunto(s)
Rastreo Celular , Mediciones Luminiscentes , Peste/microbiología , Yersinia pestis/fisiología , Yersinia pseudotuberculosis/fisiología , Animales , Modelos Animales de Enfermedad , Escherichia coli/genética , Escherichia coli/crecimiento & desarrollo , Escherichia coli/patogenicidad , Hígado/microbiología , Hígado/patología , Pulmón/microbiología , Pulmón/patología , Tejido Linfoide/microbiología , Tejido Linfoide/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Peste/patología , Plásmidos/genética , Bazo/microbiología , Bazo/patología , Tropismo Viral , Virulencia , Yersinia pestis/genética , Yersinia pestis/crecimiento & desarrollo , Yersinia pseudotuberculosis/genética , Yersinia pseudotuberculosis/crecimiento & desarrollo
19.
J Clin Microbiol ; 56(1)2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29070654

RESUMEN

Plague is caused by Yersinia pestis and is not commonly encountered in clinics, although natural plague foci are widely distributed around the world. Y. pestis has been listed as a category A bioterrorism agent. A neglected diagnosis will cause severe consequences. Therefore, this minireview briefly introduces the current understanding on Y. pestis and then focuses on practical aspects of plague, including clinical manifestations, diagnosis, treatment, and prevention, to alert clinicians about this notorious disease.


Asunto(s)
Peste , Yersinia pestis/aislamiento & purificación , Animales , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Armas Biológicas , Humanos , Técnicas Microbiológicas , Peste/diagnóstico , Peste/tratamiento farmacológico , Peste/prevención & control , Peste/transmisión , Roedores/microbiología , Siphonaptera/microbiología , Yersinia pestis/efectos de los fármacos , Yersinia pestis/crecimiento & desarrollo
20.
Sheng Wu Gong Cheng Xue Bao ; 33(9): 1513-1524, 2017 Sep 25.
Artículo en Chino | MEDLINE | ID: mdl-28956397

RESUMEN

Yersinia pestis, the cause of plague, is transmitted by flea bite. Y. pestis forms a biofilm in the proventriculus of its flea vector to enhance transmission. Biofilm formation in Y. pestis is positively regulated by the intracellular levels of the second messenger cyclic diguanylate (c-di-GMP). The c-di-GMP in Y. pestis is synthesized by two diguanylate cyclases (DGC), HmsT and HmsD, and degraded by phosphodiesterase (PDE), HmsP. Here we summarized the regulators that modulate c-di-GMP metabolism and biofilm formation in Y. pestis and discussed their regulatory mechanism.


Asunto(s)
Biopelículas , GMP Cíclico/análogos & derivados , Sistemas de Mensajero Secundario , Yersinia pestis/crecimiento & desarrollo , Animales , Proteínas Bacterianas , GMP Cíclico/metabolismo , Proteínas de Escherichia coli , Liasas de Fósforo-Oxígeno , Siphonaptera/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...