Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 367
Filtrar
1.
Microbiol Spectr ; 12(6): e0427823, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38712967

RESUMEN

Within the realm of Gram-negative bacteria, bacteriocins are secreted almost everywhere, and the most representative are colicin and pyocin, which are secreted by Escherichia coli and Pseudomonas aeruginosa, respectively. Signal peptides at the amino terminus of bacteriocins or ABC transporters can secrete bacteriocins, which then enter bacteria through cell membrane receptors and exert toxicity. In general, the bactericidal spectrum is usually narrow, killing only the kin or closely related species. Our previous research indicates that YPK_0952 is an effector of the third Type VI secretion system (T6SS-3) in Yersinia pseudotuberculosis. Next, we sought to determine its identity and characterize its toxicity. We found that YPK_0952 (a pyocin-like effector) can achieve intra-species and inter-species competitive advantages through both contact-dependent and contact-independent mechanisms mediated by the T6SS-3 while enhancing the intestinal colonization capacity of Y. pseudotuberculosis. We further identified YPK_0952 as a DNase dependent on Mg2+, Ni2+, Mn2+, and Co2+ bivalent metal ions, and the homologous immune protein YPK_0953 can inhibit its activity. In summary, YPK_0952 exerts toxicity by degrading nucleic acids from competing cells, and YPK_0953 prevents self-attack in Y. pseudotuberculosis.IMPORTANCEBacteriocins secreted by Gram-negative bacteria generally enter cells through specific interactions on the cell surface, resulting in a narrow bactericidal spectrum. First, we identified a new pyocin-like effector protein, YPK_0952, in the third Type VI secretion system (T6SS-3) of Yersinia pseudotuberculosis. YPK_0952 is secreted by T6SS-3 and can exert DNase activity through contact-dependent and contact-independent entry into nearby cells of the same and other species (e.g., Escherichia coli) to help Y. pseudotuberculosis to exert a competitive advantage and promote intestinal colonization. This discovery lays the foundation for an in-depth study of the different effector protein types within the T6SS and their complexity in competing interactions. At the same time, this study provides a new development for the toolbox of toxin/immune pairs for studying Gram-negative bacteriocin translocation.


Asunto(s)
Proteínas Bacterianas , Piocinas , Sistemas de Secreción Tipo VI , Infecciones por Yersinia pseudotuberculosis , Yersinia pseudotuberculosis , Yersinia pseudotuberculosis/metabolismo , Yersinia pseudotuberculosis/genética , Sistemas de Secreción Tipo VI/metabolismo , Sistemas de Secreción Tipo VI/genética , Piocinas/metabolismo , Infecciones por Yersinia pseudotuberculosis/microbiología , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Animales , Ratones , Humanos , Bacteriocinas/metabolismo , Antibacterianos/farmacología , Antibacterianos/metabolismo
2.
Methods Mol Biol ; 2674: 101-115, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37258963

RESUMEN

Pathogenic bacteria have evolved the ability to evade their host defenses and cause diseases. Virulence factors encompass a wide range of adaptations that allow pathogens to survive and proliferate in the hostile host environment during successful infection. In human pathogenic Yersinia species, the potent type III secretion system (T3SS) and other essential virulence factors are encoded on a virulence plasmid. Here, we investigated the bacterial growth rate and plasmid copy number following a Yersinia infection using droplet digital PCR (ddPCR). ddPCR is an exceptionally sensitive, highly precise, and cost-efficient method. It enables precise quantification even from very small amounts of target DNA. This method also enables analysis of complex samples with large amounts of interfering DNA, such as infected tissues or microbiome studies.


Asunto(s)
Infecciones por Yersinia pseudotuberculosis , Yersinia pseudotuberculosis , Humanos , Virulencia/genética , Yersinia pseudotuberculosis/genética , Variaciones en el Número de Copia de ADN , Infecciones por Yersinia pseudotuberculosis/diagnóstico , Infecciones por Yersinia pseudotuberculosis/microbiología , Plásmidos/genética , Factores de Virulencia/genética , Reacción en Cadena de la Polimerasa
3.
Nat Microbiol ; 8(4): 666-678, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36879169

RESUMEN

Granulomas are organized immune cell aggregates formed in response to chronic infection or antigen persistence. The bacterial pathogen Yersinia pseudotuberculosis (Yp) blocks innate inflammatory signalling and immune defence, inducing neutrophil-rich pyogranulomas (PGs) within lymphoid tissues. Here we uncover that Yp also triggers PG formation within the murine intestinal mucosa. Mice lacking circulating monocytes fail to form defined PGs, have defects in neutrophil activation and succumb to Yp infection. Yersinia lacking virulence factors that target actin polymerization to block phagocytosis and reactive oxygen burst do not induce PGs, indicating that intestinal PGs form in response to Yp disruption of cytoskeletal dynamics. Notably, mutation of the virulence factor YopH restores PG formation and control of Yp in mice lacking circulating monocytes, demonstrating that monocytes override YopH-dependent blockade of innate immune defence. This work reveals an unappreciated site of Yersinia intestinal invasion and defines host and pathogen drivers of intestinal granuloma formation.


Asunto(s)
Yersiniosis , Infecciones por Yersinia pseudotuberculosis , Yersinia pseudotuberculosis , Animales , Ratones , Monocitos , Infecciones por Yersinia pseudotuberculosis/genética , Infecciones por Yersinia pseudotuberculosis/microbiología , Yersinia pseudotuberculosis/genética , Factores de Virulencia/genética , Granuloma
4.
STAR Protoc ; 3(4): 101760, 2022 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-36219561

RESUMEN

Yersinia pseudotuberculosis (Yptb) is a bacterial pathogen that causes foodborne illness. Defense against the host antimicrobial gas, nitric oxide (NO), by the bacterial NO-detoxifying gene, hmp, promotes Yptb replication in mouse models of infection. Here, we detail the use of fluorescent signals as readouts for NO exposure within individual cells and subsequent detection of heterogeneity within a population, using single-cell imaging and analysis. This protocol quantifies NO exposure in culture, without capturing the full complexity of the host environment. For complete details on the use and execution of this protocol, please refer to Patel et al. (2021).


Asunto(s)
Infecciones por Yersinia pseudotuberculosis , Yersinia pseudotuberculosis , Animales , Ratones , Yersinia pseudotuberculosis/genética , Infecciones por Yersinia pseudotuberculosis/genética , Infecciones por Yersinia pseudotuberculosis/microbiología , Óxido Nítrico
5.
Microbiol Spectr ; 10(4): e0114522, 2022 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-35863020

RESUMEN

Yersinia pseudotuberculosis is an enteric pathogen causing mild enteritis that can lead to mesenteric adenitis in children and septicemia in elderly patients. Most cases are sporadic, but outbreaks have already been described in different countries. We report for the first time a Y. pseudotuberculosis clonal outbreak in France, that occurred in 2020. An epidemiological investigation based on food queries pointed toward the consumption of tomatoes as the suspected source of infection. The Yersinia National Reference Laboratory (YNRL) developed a new cgMLST scheme with 1,921 genes specific to Y. pseudotuberculosis that identified the clustering of isolates associated with the outbreak and allowed to perform molecular typing in real time. In addition, this method allowed to retrospectively identify isolates belonging to this cluster from earlier in 2020. This method, which does not require specific bioinformatic skills, is now used systematically at the YNRL and proves to display an excellent discriminatory power and is available to the scientific community. IMPORTANCE We describe in here a novel core-genome MLST method that allowed to identify in real time, and for the first time in France, a Y. pseudotuberculosis clonal outbreak that took place during the summer 2020 in Corsica. Our method allows to support epidemiological and microbiological investigations to establish a link between patients infected with closely associated Y. pseudotuberculosis isolates, and to identify the potential source of infection. In addition, we made this method available for the scientific community.


Asunto(s)
Infecciones por Yersinia pseudotuberculosis , Yersinia pseudotuberculosis , Anciano , Niño , Brotes de Enfermedades , Humanos , Tipificación de Secuencias Multilocus/métodos , Estudios Retrospectivos , Yersinia pseudotuberculosis/genética , Infecciones por Yersinia pseudotuberculosis/epidemiología , Infecciones por Yersinia pseudotuberculosis/microbiología
6.
Infect Immun ; 90(8): e0016722, 2022 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-35862700

RESUMEN

Fluorescence dilution approaches can detect bacterial cell division events and can detect if there are differential rates of cell division across individual cells within a population. This approach typically involves inducing expression of a fluorescent protein and then tracking partitioning of fluorescence into daughter cells. However, fluorescence can be diluted very quickly within a rapidly replicating population, such as pathogenic bacterial populations replicating within host tissues. To overcome this limitation, we have generated two revTetR reporter constructs, where either mCherry or yellow fluorescent protein (YFP) is constitutively expressed and repressed by addition of tetracyclines, resulting in fluorescence dilution within defined time frames. We show that fluorescent signals are diluted in replicating populations and that signal accumulates in growth-inhibited populations, including during nitric oxide (NO) exposure. Furthermore, we show that tetracyclines can be delivered to the mouse spleen during Yersinia pseudotuberculosis infection and defined a drug concentration that results in even exposure of cells to tetracyclines. We then used this system to visualize bacterial cell division within defined time frames postinfection. revTetR-mCherry allowed us to detect slow-growing cells in response to NO in culture; however, this strain had a growth defect within mouse tissues, which complicated results. To address this issue, we constructed revTetR-YFP using the less toxic YFP and showed that heightened NO exposure correlated with heightened YFP signal, indicating decreased cell division rates within this subpopulation in vivo. This revTetR reporter will provide a critical tool for future studies to identify and isolate slowly replicating bacterial subpopulations from host tissues.


Asunto(s)
Infecciones por Yersinia pseudotuberculosis , Yersinia pseudotuberculosis , Animales , División Celular , Ratones , Óxido Nítrico/metabolismo , Bazo/microbiología , Tetraciclinas , Yersinia pseudotuberculosis/genética , Infecciones por Yersinia pseudotuberculosis/microbiología
7.
J Mol Biol ; 434(18): 167667, 2022 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-35667470

RESUMEN

The type III secretion system (T3SS) is indispensable for successful host cell infection by many Gram-negative pathogens. The molecular syringe delivers effector proteins that suppress the host immune response. Synthesis of T3SS components in Yersinia pseudotuberculosis relies on host body temperature, which induces the RNA thermometer (RNAT)-controlled translation of lcrF coding for a virulence master regulator that activates transcription of the T3SS regulon. The assembly of the secretion machinery follows a strict coordinated succession referred to as outside-in assembly, in which the membrane ring complex and the export apparatus represent the nucleation points. Two components essential for the initial assembly are YscJ and YscT. While YscJ connects the membrane ring complex with the export apparatus in the inner membrane, YscT is required for a functional export apparatus. Previous transcriptome-wide RNA structuromics data suggested the presence of unique intercistronic RNATs upstream of yscJ and yscT. Here, we show by reporter gene fusions that both upstream regions confer translational control. Moreover, we demonstrate the temperature-induced opening of the Shine-Dalgarno region, which facilitates ribosome binding, by in vitro structure probing and toeprinting methods. Rationally designed thermostable RNAT variants of the yscJ and yscT thermometers confirmed their physiological relevance with respect to T3SS assembly and host infection. Since we have shown in a recent study that YopN, the gatekeeper of type III secretion, also is under RNAT control, it appears that the synthesis, assembly and functionality of the Yersinia T3S machinery is coordinated by RNA-based temperature sensors at multiple levels.


Asunto(s)
Temperatura Corporal , Interacciones Huésped-Patógeno , ARN Bacteriano , Sistemas de Secreción Tipo III , Infecciones por Yersinia pseudotuberculosis , Yersinia pseudotuberculosis , Proteínas Bacterianas/genética , Regulación Bacteriana de la Expresión Génica , Humanos , ARN Bacteriano/química , Transactivadores/genética , Sistemas de Secreción Tipo III/genética , Sistemas de Secreción Tipo III/metabolismo , Yersinia pseudotuberculosis/genética , Yersinia pseudotuberculosis/patogenicidad , Infecciones por Yersinia pseudotuberculosis/microbiología
8.
Bull Exp Biol Med ; 172(6): 725-728, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35503586

RESUMEN

One of the mechanisms underlying the appearance of chronic infections is transition of pathogens into a non-culturable state, which is largely associated with the use of antibiotics. We studied ultrastructure of dormant bacteria Yersinia pseudotuberculosis obtained from the vegetative form of strain 512 by inhibition with kanamycin. On the model of the causative agent of pseudotuberculosis we showed that transition of prokaryotes to a dormant state occurs through apoptosis of bacteria. Fragmentation and condensation of chromatin with the formation of electron-dense fibrils, clumps and large conglomerates characteristic of apoptosis were found in the nucleoid zone of the cytoplasm of inhibited bacterial cells. These results are of great importance for understanding the mechanisms of the existence of pathogens in different conditions, as well as for identifying the causative agents of infectious diseases.


Asunto(s)
Infecciones por Yersinia pseudotuberculosis , Yersinia pseudotuberculosis , Antibacterianos , Humanos , Yersinia , Yersinia pseudotuberculosis/ultraestructura , Infecciones por Yersinia pseudotuberculosis/microbiología
9.
Vet Microbiol ; 269: 109424, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35429816

RESUMEN

A Yersinia pseudotuberculosis outbreak was diagnosed in a male turkey flock in Finland. Y. pseudotuberculosis is a quite rare zoonotic bacterium, which typically causes enteritis in humans and sudden death in animals. In this study, osteomyelitis was diagnosed in small, lame, 11- to 12-wk-old male turkeys. Lameness and slower growth among the turkeys was observed on the farm. During pathological examination, multiple lesions were found in the metaphyseal and physeal areas of the femurs, tibiotarsi, and tarsometatarsi, with multifocal to coalescing mixed heterophilic/granulomatous necrotizing osteomyelitis. Y. pseudotuberculosis was isolated from the femoral and tibiotarsal bones or from the joints of six lame turkeys sent for necropsy. The isolation required homogenizing of lesion tissue in phosphate-mannitol-peptone broth, which was cultured directly - and, if needed, after cold enrichment - on selective cefsulodin-irgasan-novobiocin agar. Whole-genome sequencing was used for identification and typing. All isolates belonged to bio/serotype 1/O:1a and sequence type ST42 (Achtman scheme), which is commonly reported in both human and animal Y. pseudotuberculosis infections in Europe. The isolates from all six turkeys showed only one to two allele differences in the core genome comparison, indicating a common source of infection. All asymptomatic turkeys were slaughtered at the age of 17 weeks. Whole and partial carcass condemnation rates at the slaughterhouse were high, but no macroscopic changes in the skeletal system were found, showing that food chain information is essential. This study confirms earlier findings that Y. pseudotuberculosis can cause osteomyelitis in fattening turkeys, leading to lameness. Food chain information is essential for slaughterhouse operations, to protect the workers and emphasize good working hygiene during slaughter.


Asunto(s)
Osteomielitis , Infecciones por Yersinia pseudotuberculosis , Yersinia pseudotuberculosis , Animales , Cojera Animal , Masculino , Osteomielitis/epidemiología , Osteomielitis/veterinaria , Pavos , Yersinia pseudotuberculosis/genética , Infecciones por Yersinia pseudotuberculosis/epidemiología , Infecciones por Yersinia pseudotuberculosis/microbiología , Infecciones por Yersinia pseudotuberculosis/veterinaria
10.
J Vet Diagn Invest ; 34(2): 334-338, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35037547

RESUMEN

A concurrent outbreak of infection with Yersinia pseudotuberculosis occurred in adult captive African lions (Panthera leo). Two 17-y-old male lions and one 14-y-old female lion developed respiratory distress, lethargy, ataxia, and hyporexia. Within 3-5 d of the onset of clinical signs, one male and the female lion died and were submitted for postmortem examination. Macroscopically, the liver and spleen had multifocal-to-coalescing, semi-firm, pale-tan nodules throughout the parenchyma. The lungs were non-collapsed and marked by petechiae. Histologic examination identified lytic, necrosuppurative foci in the liver, spleen, lungs, and kidney, with abundant intralesional gram-negative coccobacilli in the male lion. Similar findings were seen in the female lion in the liver, spleen, kidney, and mesenteric lymph node; however, the intralesional bacterial colonies were more pleomorphic, comprising rod and filamentous morphologies. Aerobic bacterial culture of the liver, spleen, and lung revealed Y. pseudotuberculosis growth. The source of infection is unknown, and an epidemiologic study was performed. Sources to be considered are from the predation of rodent and/or bird reservoirs, or contaminated soil or water. Mortality associated with Y. pseudotuberculosis has been described in an African lion cub, however, to our knowledge, Y. pseudotuberculosis has not been reported in adult African lions, and this is only the second report of Y. pseudotuberculosis with aberrant bacterial morphology observed histologically.


Asunto(s)
Leones , Infecciones por Yersinia pseudotuberculosis , Yersinia pseudotuberculosis , Animales , Brotes de Enfermedades/veterinaria , Femenino , Masculino , Bazo/patología , Infecciones por Yersinia pseudotuberculosis/epidemiología , Infecciones por Yersinia pseudotuberculosis/microbiología , Infecciones por Yersinia pseudotuberculosis/veterinaria
11.
PLoS Pathog ; 17(11): e1009650, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34767606

RESUMEN

Many bacterial pathogens use a type III secretion system (T3SS) as molecular syringe to inject effector proteins into the host cell. In the foodborne pathogen Yersinia pseudotuberculosis, delivery of the secreted effector protein cocktail through the T3SS depends on YopN, a molecular gatekeeper that controls access to the secretion channel from the bacterial cytoplasm. Here, we show that several checkpoints adjust yopN expression to virulence conditions. A dominant cue is the host body temperature. A temperature of 37°C is known to induce the RNA thermometer (RNAT)-dependent synthesis of LcrF, a transcription factor that activates expression of the entire T3SS regulon. Here, we uncovered a second layer of temperature control. We show that another RNAT silences translation of the yopN mRNA at low environmental temperatures. The long and short 5'-untranslated region of both cellular yopN isoforms fold into a similar secondary structure that blocks ribosome binding. The hairpin structure with an internal loop melts at 37°C and thereby permits formation of the translation initiation complex as shown by mutational analysis, in vitro structure probing and toeprinting methods. Importantly, we demonstrate the physiological relevance of the RNAT in the faithful control of type III secretion by using a point-mutated thermostable RNAT variant with a trapped SD sequence. Abrogated YopN production in this strain led to unrestricted effector protein secretion into the medium, bacterial growth arrest and delayed translocation into eukaryotic host cells. Cumulatively, our results show that substrate delivery by the Yersinia T3SS is under hierarchical surveillance of two RNATs.


Asunto(s)
Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , ARN Bacteriano/metabolismo , Sistemas de Secreción Tipo III/metabolismo , Virulencia , Infecciones por Yersinia pseudotuberculosis/microbiología , Yersinia pseudotuberculosis/metabolismo , Proteínas Bacterianas/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Fagocitosis , Transporte de Proteínas , ARN Bacteriano/genética , Infecciones por Yersinia pseudotuberculosis/metabolismo
12.
Cell ; 184(23): 5715-5727.e12, 2021 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-34717799

RESUMEN

The enteric nervous system (ENS) controls several intestinal functions including motility and nutrient handling, which can be disrupted by infection-induced neuropathies or neuronal cell death. We investigated possible tolerance mechanisms preventing neuronal loss and disruption in gut motility after pathogen exposure. We found that following enteric infections, muscularis macrophages (MMs) acquire a tissue-protective phenotype that prevents neuronal loss, dysmotility, and maintains energy balance during subsequent challenge with unrelated pathogens. Bacteria-induced neuroprotection relied on activation of gut-projecting sympathetic neurons and signaling via ß2-adrenergic receptors (ß2AR) on MMs. In contrast, helminth-mediated neuroprotection was dependent on T cells and systemic production of interleukin (IL)-4 and IL-13 by eosinophils, which induced arginase-expressing MMs that prevented neuronal loss from an unrelated infection located in a different intestinal region. Collectively, these data suggest that distinct enteric pathogens trigger a state of disease or tissue tolerance that preserves ENS number and functionality.


Asunto(s)
Sistema Nervioso Entérico/microbiología , Sistema Nervioso Entérico/parasitología , Infecciones/microbiología , Infecciones/parasitología , Neuronas/patología , Neuroprotección , Especificidad de Órganos , Yersinia pseudotuberculosis/fisiología , Animales , Eosinófilos/metabolismo , Células Madre Hematopoyéticas/metabolismo , Inmunidad , Infecciones/inmunología , Interleucina-13/metabolismo , Interleucina-4/metabolismo , Macrófagos/metabolismo , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Strongyloides/fisiología , Estrongiloidiasis/genética , Estrongiloidiasis/inmunología , Estrongiloidiasis/parasitología , Transcriptoma/genética , Infecciones por Yersinia pseudotuberculosis/genética , Infecciones por Yersinia pseudotuberculosis/inmunología , Infecciones por Yersinia pseudotuberculosis/microbiología
13.
Proc Natl Acad Sci U S A ; 118(28)2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34260403

RESUMEN

Injection of effector proteins to block host innate immune signaling is a common strategy used by many pathogenic organisms to establish an infection. For example, pathogenic Yersinia species inject the acetyltransferase YopJ into target cells to inhibit NF-κB and MAPK signaling. To counteract this, detection of YopJ activity in myeloid cells promotes the assembly of a RIPK1-caspase-8 death-inducing platform that confers antibacterial defense. While recent studies revealed that caspase-8 cleaves the pore-forming protein gasdermin D to trigger pyroptosis in macrophages, whether RIPK1 activates additional substrates downstream of caspase-8 to promote host defense is unclear. Here, we report that the related gasdermin family member gasdermin E (GSDME) is activated upon detection of YopJ activity in a RIPK1 kinase-dependent manner. Specifically, GSDME promotes neutrophil pyroptosis and IL-1ß release, which is critical for anti-Yersinia defense. During in vivo infection, IL-1ß neutralization increases bacterial burden in wild-type but not Gsdme-deficient mice. Thus, our study establishes GSDME as an important mediator that counteracts pathogen blockade of innate immune signaling.


Asunto(s)
Inmunidad Innata , Macrófagos/metabolismo , Proteínas de Neoplasias/metabolismo , Neutrófilos/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Transducción de Señal , Yersinia pseudotuberculosis/fisiología , Células 3T3 , Animales , Citocinas/metabolismo , Interacciones Huésped-Patógeno/inmunología , Interleucina-1beta/metabolismo , Ratones , Ratones Endogámicos C57BL , Piroptosis , Infecciones por Yersinia pseudotuberculosis/inmunología , Infecciones por Yersinia pseudotuberculosis/microbiología
14.
PLoS Pathog ; 17(7): e1009284, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34214139

RESUMEN

It is now well appreciated that members of pathogenic bacterial populations exhibit heterogeneity in growth rates and metabolic activity, and it is known this can impact the ability to eliminate all members of the bacterial population during antibiotic treatment. It remains unclear which pathways promote slowed bacterial growth within host tissues, primarily because it has been difficult to identify and isolate slow growing bacteria from host tissues for downstream analyses. To overcome this limitation, we have developed a novel variant of TIMER, a slow-folding fluorescent protein, named DsRed42, to identify subsets of slowly dividing bacteria within host tissues. The original TIMER folds too slowly for fluorescence accumulation in quickly replicating bacterial species (Escherichia coli, Yersinia pseudotuberculosis), however DsRed42 accumulates red fluorescence in late stationary phase cultures of E. coli and Y. pseudotuberculosis. We show DsRed42 signal also accumulates during exposure to sources of nitric oxide (NO), suggesting DsRed42 signal detects growth-arrested bacterial cells. In a mouse model of Y. pseudotuberculosis deep tissue infection, DsRed42 signal was detected, and primarily accumulates in bacteria expressing markers of stationary phase growth. There was no significant overlap between DsRed42 signal and NO-exposed subpopulations of bacteria within host tissues, suggesting NO stress was transient, allowing bacteria to recover from this stress and resume replication. This novel DsRed42 variant represents a tool that will enable additional studies of slow-growing subpopulations of bacteria, specifically within bacterial species that quickly divide.


Asunto(s)
Proteínas Luminiscentes , Técnicas Microbiológicas , Yersinia pseudotuberculosis/crecimiento & desarrollo , Animales , Proliferación Celular , Ratones , Mutagénesis Sitio-Dirigida , Infecciones por Yersinia pseudotuberculosis/microbiología
15.
Microbiol Spectr ; 9(1): e0049621, 2021 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-34319170

RESUMEN

Prostaglandin E2 (PGE2) is an essential immunomodulatory lipid released by cells in response to infection with many bacteria, yet its function in macrophage-mediated bacterial clearance is poorly understood. Yersinia overall inhibits the inflammatory circuit, but its effect on PGE2 production is unknown. We hypothesized that one of the Yersinia effector proteins is responsible for the inhibition of PGE2 biosynthesis. We identified that yopB-deficient Y. enterocolitica and Y. pseudotuberculosis deficient in the secretion of virulence proteins via a type 3 secretion system (T3SS) failed to inhibit PGE2 biosynthesis in macrophages. Consistently, COX-2-mediated PGE2 biosynthesis is upregulated in cells treated with heat-killed or T3SS-deficient Y. pseudotuberculosis but diminished in the presence of a MAPK/ERK inhibitor. Mutants expressing catalytically inactive YopJ induce similar levels of PGE2 as heat-killed or ΔyopB Y. pseudotuberculosis, reversed by YopJ complementation. Shotgun proteomics discovered host pathways regulated in a YopJ-mediated manner, including pathways regulating PGE2 synthesis and oxidative phosphorylation. Consequently, this study identified that YopJ-mediated inhibition of MAPK signal transduction serves as a mechanism targeting PGE2, an alternative means of inflammasome inhibition by Yersinia. Finally, we showed that EP4 signaling supports macrophage function in clearing intracellular bacteria. In summary, our unique contribution was to determine a bacterial virulence factor that targets COX-2 transcription, thereby enhancing the intracellular survival of yersiniae. Future studies should investigate whether PGE2 or its stable synthetic derivatives could serve as a potential therapeutic molecule to improve the outcomes of specific bacterial infections. Since other pathogens encode YopJ homologs, this mechanism is expected to be present in other infections. IMPORTANCE PGE2 is a critical immunomodulatory lipid, but its role in bacterial infection and pathogen clearance is poorly understood. We previously demonstrated that PGE2 leads to macrophage polarization toward the M1 phenotype and stimulates inflammasome activation in infected macrophages. Finally, we also discovered that PGE2 improved the clearance of Y. enterocolitica. The fact that Y. enterocolitica hampers PGE2 secretion in a type 3 secretion system (T3SS)-dependent manner and because PGE2 appears to assist macrophage in the clearance of this bacterium indicates that targeting of the eicosanoid pathway by Yersinia might be an adaption used to counteract host defenses. Our study identified a mechanism used by Yersinia that obstructs PGE2 biosynthesis in human macrophages. We showed that Y. pseudotuberculosis interferes with PGE2 biosynthesis by using one of its T3SS effectors, YopJ. Specifically, YopJ targets the host COX-2 enzyme responsible for PGE2 biosynthesis, which happens in a MAPK/ER-dependent manner. Moreover, in a shotgun proteomics study, we also discovered other pathways that catalytically active YopJ targets in the infected macrophages. YopJ was revealed to play a role in limiting host LPS responses, including repression of EGR1 and JUN proteins, which control transcriptional activation of proinflammatory cytokine production such as interleukin-1ß. Since YopJ has homologs in other bacterial species, there are likely other pathogens that target and inhibit PGE2 biosynthesis. In summary, our study's unique contribution was to determine a bacterial virulence factor that targets COX-2 transcription. Future studies should investigate whether PGE2 or its stable synthetic derivatives could serve as a potential therapeutic target.


Asunto(s)
Proteínas Bacterianas/inmunología , Ciclooxigenasa 2/inmunología , Dinoprostona/inmunología , Quinasas MAP Reguladas por Señal Extracelular/inmunología , Macrófagos/inmunología , Quinasas de Proteína Quinasa Activadas por Mitógenos/inmunología , Infecciones por Yersinia pseudotuberculosis/microbiología , Yersinia pseudotuberculosis/inmunología , Animales , Proteínas Bacterianas/genética , Ciclooxigenasa 2/genética , Quinasas MAP Reguladas por Señal Extracelular/genética , Interacciones Huésped-Patógeno , Humanos , Activación de Macrófagos , Ratones , Ratones Endogámicos BALB C , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Transducción de Señal , Yersinia pseudotuberculosis/genética , Infecciones por Yersinia pseudotuberculosis/inmunología
16.
mBio ; 12(3): e0063321, 2021 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-34060331

RESUMEN

The iron-sulfur cluster coordinating transcription factor IscR is important for the virulence of Yersinia pseudotuberculosis and a number of other bacterial pathogens. However, the IscR regulon has not yet been defined in any organism. To determine the Yersinia IscR regulon and identify IscR-dependent functions important for virulence, we employed chromatin immunoprecipitation sequencing (ChIP-Seq) and RNA sequencing (RNA-Seq) of Y. pseudotuberculosis expressing or lacking iscR following iron starvation conditions, such as those encountered during infection. We found that IscR binds to the promoters of genes involved in iron homeostasis, reactive oxygen species metabolism, and cell envelope remodeling and regulates expression of these genes in response to iron depletion. Consistent with our previous work, we also found that IscR binds in vivo to the promoter of the Ysc type III secretion system (T3SS) master regulator LcrF, leading to regulation of T3SS genes. Interestingly, comparative genomic analysis suggested over 93% of IscR binding sites were conserved between Y. pseudotuberculosis and the related plague agent Yersinia pestis. Surprisingly, we found that the IscR positively regulated sufABCDSE Fe-S cluster biogenesis pathway was required for T3SS activity. These data suggest that IscR regulates the T3SS in Yersinia through maturation of an Fe-S cluster protein critical for type III secretion, in addition to its known role in activating T3SS genes through LcrF. Altogether, our study shows that iron starvation triggers IscR to coregulate multiple, distinct pathways relevant to promoting bacterial survival during infection. IMPORTANCE How bacteria adapt to the changing environment within the host is critical for their ability to survive and cause disease. For example, the mammalian host severely restricts iron availability to limit bacterial growth, referred to as nutritional immunity. Here, we show that pathogenic Yersinia use the iron-sulfur (Fe-S) cluster regulator IscR, a factor critical for pathogenesis, to sense iron availability and regulate multiple pathways known or predicted to contribute to virulence. Under low iron conditions that mimic those Yersinia encounter during infection, IscR levels increase, leading to modulation of genes involved in iron metabolism, stress resistance, cell envelope remodeling, and subversion of host defenses. These data suggest that IscR senses nutritional immunity to coordinate processes important for bacterial survival within the mammalian host.


Asunto(s)
Proteínas Bacterianas/genética , Regulación Bacteriana de la Expresión Génica/genética , Genoma Bacteriano , Genómica/métodos , Factores de Virulencia/genética , Yersinia pseudotuberculosis/genética , Yersinia pseudotuberculosis/patogenicidad , Proteínas Bacterianas/metabolismo , Sitios de Unión , Humanos , Hierro/metabolismo , Regiones Promotoras Genéticas , Virulencia , Yersinia pestis/genética , Yersinia pseudotuberculosis/metabolismo , Infecciones por Yersinia pseudotuberculosis/microbiología
17.
Infect Immun ; 89(8): e0026521, 2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-34031132

RESUMEN

Primary infection of C57BL/6 mice with the bacterial pathogen Yersinia pseudotuberculosis elicits an unusually large H-2Kb-restricted CD8+ T cell response to the endogenous and protective bacterial epitope YopE69-77. To better understand the basis for this large response, the model OVA257-264 epitope was inserted into YopE in Y. pseudotuberculosis and antigen-specific CD8+ T cells in mice were characterized after foodborne infection with the resulting strain. The epitope YopE69-77 elicited significantly larger CD8+ T cell populations in the small intestine, mesenteric lymph nodes (MLNs), spleen, and liver between 7 and 30 days postinfection, despite residing in the same protein and having an affinity for H-2Kb similar to that of OVA257-264. YopE-specific CD8+ T cell precursors were ∼4.6 times as abundant as OVA-specific precursors in the MLNs, spleens, and other lymph nodes of naive mice, explaining the dominance of YopE69-77 over OVA257-264 at early infection times. However, other factors contributed to this dominance, as the ratio of YopE-specific to OVA-specific CD8+ T cells increased between 7 and 30 days postinfection. We also compared the YopE-specific and OVA-specific CD8+ T cells generated during infection for effector and memory phenotypes. Significantly higher percentages of YopE-specific cells were characterized as short-lived effectors, while higher percentages of OVA-specific cells were memory precursor effectors at day 30 postinfection in spleen and liver. Our results suggest that a large precursor number contributes to the dominance and effector and memory functions of CD8+ T cells generated in response to the protective YopE69-77 epitope during Y. pseudotuberculosis infection of C57BL/6 mice.


Asunto(s)
Antígenos Bacterianos/inmunología , Linfocitos T CD8-positivos/inmunología , Interacciones Huésped-Patógeno/inmunología , Especificidad del Receptor de Antígeno de Linfocitos T , Infecciones por Yersinia pseudotuberculosis/inmunología , Infecciones por Yersinia pseudotuberculosis/microbiología , Yersinia pseudotuberculosis/inmunología , Animales , Linfocitos T CD8-positivos/metabolismo , Modelos Animales de Enfermedad , Epítopos de Linfocito T/inmunología , Memoria Inmunológica , Ratones , Ratones Endogámicos C57BL , Infecciones por Yersinia pseudotuberculosis/transmisión
18.
Virulence ; 12(1): 638-653, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33550901

RESUMEN

Adhesins facilitate bacterial colonization and invasion of host tissues and are considered virulence factors, but their impact on immune-mediated damage as a driver of pathogenesis remains unclear. Yersinia pseudotuberculosis encodes for a multivalent adhesion molecule (MAM), a mammalian cell entry (MCE) family protein and adhesin. MAMs are widespread in Gram-negative bacteria and enable enteric bacteria to colonize epithelial tissues. Their role in bacterial interactions with the host innate immune system and contribution to pathogenicity remains unclear. Here, we investigated howY. pseudotuberculosis MAM contributes to pathogenesis during infection of the Galleria mellonella insect model. We show that Y. pseudotuberculosis MAM is required for efficient bacterial binding and uptake by hemocytes, the host phagocytes. Y. pseudotuberculosis interactions with insect and mammalian phagocytes are determined by bacterial and host factors. Loss of MAM, and deficient microbe-phagocyte interaction, increased pathogenesis in G. mellonella. Diminished phagocyte association also led to increased bacterial clearance. Furthermore, Y. pseudotuberculosis that failed to engage phagocytes hyperactivated humoral immune responses, most notably melanin production. Despite clearing the pathogen, excessive melanization also increased phagocyte death and host mortality. Our findings provide a basis for further studies investigating how microbe- and host-factors integrate to drive pathogenesis in a tractable experimental system.


Asunto(s)
Interacciones Huésped-Patógeno , Larva/microbiología , Mariposas Nocturnas/microbiología , Fagocitos/microbiología , Fagocitos/patología , Yersinia pseudotuberculosis/metabolismo , Adhesinas Bacterianas , Animales , Hemocitos , Mariposas Nocturnas/citología , Fagocitos/inmunología , Factores de Virulencia , Yersinia pseudotuberculosis/inmunología , Yersinia pseudotuberculosis/patogenicidad , Infecciones por Yersinia pseudotuberculosis/microbiología
19.
Infect Immun ; 89(4)2021 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-33495272

RESUMEN

Pathogenic Yersinia spp. depend on the activity of a potent virulence plasmid-encoded ysc/yop type 3 secretion system (T3SS) to colonize hosts and cause disease. It was recently shown that Yersinia pseudotuberculosis upregulates the virulence plasmid copy number (PCN) during infection and that the resulting elevated gene dose of plasmid-encoded T3SS genes is essential for virulence. When and how this novel regulatory mechanism is deployed and regulates the replication of the virulence plasmid during infection is unknown. In the present study, we applied droplet digital PCR (ddPCR) to investigate the dynamics of Y. pseudotuberculosis virulence PCN variations and growth rates in infected mouse organs. We demonstrated that both PCN and growth varied in different tissues and over time throughout the course of infection, indicating that the bacteria adapted to discrete microenvironments during infection. The PCN was highest in Peyer's patches and cecum during the clonal invasive phase of the infection, while the highest growth rates were found in the draining mesenteric lymph nodes. In deeper, systemic organs, the PCN was lower and more modest growth rates were recorded. Our study indicates that increased gene dosage of the plasmid-encoded T3SS genes is most important early in the infection during invasion of the host. The described ddPCR approach will greatly simplify analyses of PCN, growth dynamics, and bacterial loads in infected tissues and will be readily applicable to other infection models.


Asunto(s)
Carga Bacteriana , Variaciones en el Número de Copia de ADN , Plásmidos/genética , Infecciones por Yersinia pseudotuberculosis/microbiología , Yersinia pseudotuberculosis/fisiología , Animales , Proteínas Bacterianas/genética , Modelos Animales de Enfermedad , Femenino , Ratones , Especificidad de Órganos , Reacción en Cadena en Tiempo Real de la Polimerasa , Virulencia , Factores de Virulencia/genética , Infecciones por Yersinia pseudotuberculosis/diagnóstico
20.
Nat Commun ; 12(1): 423, 2021 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-33462232

RESUMEN

Bacterial type VI secretion systems (T6SSs) inject toxic effectors into adjacent eukaryotic and prokaryotic cells. It is generally thought that this process requires physical contact between the two cells. Here, we provide evidence of contact-independent killing by a T6SS-secreted effector. We show that the pathogen Yersinia pseudotuberculosis uses a T6SS (T6SS-3) to secrete a nuclease effector that kills other bacteria in vitro and facilitates gut colonization in mice. The effector (Tce1) is a small protein that acts as a Ca2+- and Mg2+-dependent DNase, and its toxicity is inhibited by a cognate immunity protein, Tci1. As expected, T6SS-3 mediates canonical, contact-dependent killing by directly injecting Tce1 into adjacent cells. In addition, T6SS-3 also mediates killing of neighboring cells in the absence of cell-to-cell contact, by secreting Tce1 into the extracellular milieu. Efficient contact-independent entry of Tce1 into target cells requires proteins OmpF and BtuB in the outer membrane of target cells. The discovery of a contact-independent, long-range T6SS toxin delivery provides a new perspective for understanding the physiological roles of T6SS in competition. However, the mechanisms mediating contact-independent uptake of Tce1 by target cells remain unclear.


Asunto(s)
Toxinas Bacterianas/metabolismo , Desoxirribonucleasas/metabolismo , Sistemas de Secreción Tipo VI/metabolismo , Infecciones por Yersinia pseudotuberculosis/patología , Yersinia pseudotuberculosis/patogenicidad , Animales , Proteínas de la Membrana Bacteriana Externa/metabolismo , Toxinas Bacterianas/genética , Toxinas Bacterianas/aislamiento & purificación , Toxinas Bacterianas/toxicidad , Desoxirribonucleasas/genética , Desoxirribonucleasas/aislamiento & purificación , Desoxirribonucleasas/toxicidad , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Mutagénesis , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/toxicidad , Yersinia pseudotuberculosis/metabolismo , Infecciones por Yersinia pseudotuberculosis/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...