Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Proc Natl Acad Sci U S A ; 119(2)2022 01 11.
Article in English | MEDLINE | ID: mdl-34969677

ABSTRACT

Hemachromatosis (iron-overload) increases host susceptibility to siderophilic bacterial infections that cause serious complications, but the underlying mechanisms remain elusive. The present study demonstrates that oral infection with hyperyersiniabactin (Ybt) producing Yersinia pseudotuberculosis Δfur mutant (termed Δfur) results in severe systemic infection and acute mortality to hemochromatotic mice due to rapid disruption of the intestinal barrier. Transcriptome analysis of Δfur-infected intestine revealed up-regulation in cytokine-cytokine receptor interactions, the complement and coagulation cascade, the NF-κB signaling pathway, and chemokine signaling pathways, and down-regulation in cell-adhesion molecules and Toll-like receptor signaling pathways. Further studies indicate that dysregulated interleukin (IL)-1ß signaling triggered in hemachromatotic mice infected with Δfur damages the intestinal barrier by activation of myosin light-chain kinases (MLCK) and excessive neutrophilia. Inhibiting MLCK activity or depleting neutrophil infiltration reduces barrier disruption, largely ameliorates immunopathology, and substantially rescues hemochromatotic mice from lethal Δfur infection. Moreover, early intervention of IL-1ß overproduction can completely rescue hemochromatotic mice from the lethal infection.


Subject(s)
Hemochromatosis/metabolism , Intestines/metabolism , Yersinia pseudotuberculosis Infections/metabolism , Yersinia pseudotuberculosis/metabolism , Animals , Bacterial Proteins/genetics , Calcium-Binding Proteins/metabolism , Cytokines/metabolism , Inflammation , Interleukin-1beta/metabolism , Intestines/pathology , Mice , Myosin-Light-Chain Kinase/metabolism , NF-kappa B/metabolism , Repressor Proteins/genetics , Siderophores/metabolism , Signal Transduction , Transcriptome , Yersinia pseudotuberculosis/genetics
2.
PLoS Pathog ; 17(11): e1009650, 2021 11.
Article in English | MEDLINE | ID: mdl-34767606

ABSTRACT

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.


Subject(s)
Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , RNA, Bacterial/metabolism , Type III Secretion Systems/metabolism , Virulence , Yersinia pseudotuberculosis Infections/microbiology , Yersinia pseudotuberculosis/metabolism , Bacterial Proteins/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Phagocytosis , Protein Transport , RNA, Bacterial/genetics , Yersinia pseudotuberculosis Infections/metabolism
3.
PLoS Pathog ; 15(12): e1008001, 2019 12.
Article in English | MEDLINE | ID: mdl-31869388

ABSTRACT

The enteropathogen Yersinia pseudotuberculosis and the related plague agent Y. pestis require the Ysc type III secretion system (T3SS) to subvert phagocyte defense mechanisms and cause disease. Yet type III secretion (T3S) in Yersinia induces growth arrest and innate immune recognition, necessitating tight regulation of the T3SS. Here we show that Y. pseudotuberculosis T3SS expression is kept low under anaerobic, iron-rich conditions, such as those found in the intestinal lumen where the Yersinia T3SS is not required for growth. In contrast, the Yersinia T3SS is expressed under aerobic or anaerobic, iron-poor conditions, such as those encountered by Yersinia once they cross the epithelial barrier and encounter phagocytic cells. We further show that the [2Fe-2S] containing transcription factor, IscR, mediates this oxygen and iron regulation of the T3SS by controlling transcription of the T3SS master regulator LcrF. IscR binds directly to the lcrF promoter and, importantly, a mutation that prevents this binding leads to decreased disseminated infection of Y. pseudotuberculosis but does not perturb intestinal colonization. Similar to E. coli, Y. pseudotuberculosis uses the Fe-S cluster occupancy of IscR as a readout of oxygen and iron conditions that impact cellular Fe-S cluster homeostasis. We propose that Y. pseudotuberculosis has coopted this system to sense entry into deeper tissues and induce T3S where it is required for virulence. The IscR binding site in the lcrF promoter is completely conserved between Y. pseudotuberculosis and Y. pestis. Deletion of iscR in Y. pestis leads to drastic disruption of T3S, suggesting that IscR control of the T3SS evolved before Y. pestis split from Y. pseudotuberculosis.


Subject(s)
Iron/metabolism , Oxygen/metabolism , Type III Secretion Systems/metabolism , Yersinia pseudotuberculosis Infections/immunology , Animals , Bacterial Proteins/metabolism , Bacterial Secretion Systems/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial/genetics , Promoter Regions, Genetic/genetics , Transcription, Genetic , Yersinia/metabolism , Yersinia pseudotuberculosis/pathogenicity , Yersinia pseudotuberculosis Infections/metabolism
4.
Methods Mol Biol ; 2010: 211-229, 2019.
Article in English | MEDLINE | ID: mdl-31177441

ABSTRACT

Many Gram-negative pathogens produce a type III secretion system capable of intoxicating eukaryotic cells with immune-modulating effector proteins. Fundamental to this injection process is the prior secretion of two translocator proteins destined for injectisome translocon pore assembly within the host cell plasma membrane. It is through this pore that effectors are believed to travel to gain access to the host cell interior. Yersinia species especially pathogenic to humans and animals assemble this translocon pore utilizing two hydrophobic translocator proteins-YopB and YopD. Although a full molecular understanding of the biogenesis, function and regulation of this translocon pore and subsequent effector delivery into host cells remains elusive, some of what we know about these processes can be attributed to studies of bacterial infections of erythrocytes. Herein we describe the methodology of erythrocyte infections by Yersinia, and how analysis of the resultant contact-dependent hemolysis can serve as a relative measurement of YopB- and YopD-dependent translocon pore formation.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Erythrocytes/microbiology , Yersinia Infections/pathology , Yersinia/physiology , Animals , Bacterial Outer Membrane Proteins/analysis , Erythrocytes/pathology , Hemolysis , Humans , Sheep , Sheep Diseases/metabolism , Sheep Diseases/microbiology , Sheep Diseases/pathology , Type III Secretion Systems/analysis , Type III Secretion Systems/metabolism , Yersinia Infections/metabolism , Yersinia Infections/microbiology , Yersinia Infections/veterinary , Yersinia pseudotuberculosis/physiology , Yersinia pseudotuberculosis Infections/metabolism , Yersinia pseudotuberculosis Infections/microbiology , Yersinia pseudotuberculosis Infections/pathology , Yersinia pseudotuberculosis Infections/veterinary
5.
J Med Microbiol ; 68(2): 143-147, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30648936

ABSTRACT

BACKGROUND: Despite being a well-known but seldom encountered zoonotic pathogen, diagnosis of Yersinia pseudotuberculosis is not necessarily easy. Infected patients occasionally present with various symptoms resembling Kawasaki disease; thus discriminating the two in the acute phase is challenging. In addition to bacterial culture and serology, novel detection methods based on loop-mediated isothermal amplification (LAMP) are reported in the literature. However, the clinical utility of LAMP-based methods in comparison with the other methods is scarcely documented in the literature. AIM: To clarify the clinical utility of a LAMP-based method in the diagnosis of Yersinia pseudotuberculosis infection. METHODOLOGY: Inpatients admitted due to suspected Yersinia pseudotuberculosis infection during April 2008 through March 2015 were enrolled. Results of the LAMP-based method as well as culture and serology were collected and compared. RESULTS: Among 16 eligible cases, serology proved positive in 13 (81.3 %) cases, LAMP in eight (50 %) cases, and bacterial culture in four (25 %) cases. No significant difference among the three methods could be proved statistically. Although serology was the most sensitive method, it is known to miss cases such as young patients, whereas LAMP could complement all three cases missed by serology. Furthermore, LAMP can return the test result within a few hours from specimen receipt, whereas serology and bacterial culture requires days to weeks of time. CONCLUSION: Although second to serology in sensitivity, the LAMP-based method proved its utility in making rapid diagnosis, and serving a complementary role to serology.


Subject(s)
Nucleic Acid Amplification Techniques/methods , Yersinia pseudotuberculosis Infections/diagnosis , Yersinia pseudotuberculosis/isolation & purification , Agglutination Tests , Child , Child, Preschool , DNA, Bacterial/chemistry , DNA, Bacterial/isolation & purification , Feces/microbiology , Female , Humans , Infant , Male , Nucleic Acid Amplification Techniques/standards , Time Factors , Yersinia pseudotuberculosis/genetics , Yersinia pseudotuberculosis Infections/immunology , Yersinia pseudotuberculosis Infections/metabolism
6.
Sci Rep ; 8(1): 14186, 2018 09 21.
Article in English | MEDLINE | ID: mdl-30242257

ABSTRACT

In this study we show Yersinia pseudotuberculosis secretes membrane vesicles (MVs) that contain different proteins and virulence factors depending on the strain. Although MVs from Y. pseudotuberculosis YPIII and ATCC 29833 had many proteins in common (68.8% of all the proteins identified), those located in the outer membrane fraction differed significantly. For instance, the MVs from Y. pseudotuberculosis YPIII harbored numerous Yersinia outer proteins (Yops) while they were absent in the ATCC 29833 MVs. Another virulence factor found solely in the YPIII MVs was the cytotoxic necrotizing factor (CNFy), a toxin that leads to multinucleation of host cells. The ability of YPIII MVs to transport this toxin and its activity to host cells was verified using HeLa cells, which responded in a dose-dependent manner; nearly 70% of the culture was multinucleated after addition of 5 µg/ml of the purified YPIII MVs. In contrast, less than 10% were multinucleated when the ATCC 29833 MVs were added. Semi-quantification of CNFy within the YPIII MVs found this toxin is present at concentrations of 5 ~ 10 ng per µg of total MV protein, a concentration that accounts for the cellular responses seen.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Extracellular Vesicles/metabolism , Virulence Factors/metabolism , Yersinia pseudotuberculosis Infections/metabolism , Yersinia pseudotuberculosis Infections/microbiology , Yersinia pseudotuberculosis/pathogenicity , Biological Transport/physiology , Cell Line, Tumor , HeLa Cells , Humans , Virulence/physiology
7.
J Biol Chem ; 293(22): 8691-8692, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29858390

ABSTRACT

In a new paper, the protein InvD from Yersinia pseudotuberculosis, a zoonotic pathogen, is shown to assist late-stage invasion of intestinal epithelia. Remarkably, InvD acts by binding the Fab region of IgG or IgA. It straddles adjacent light-chain and heavy-chain variable domains, but its binding is different from that of antigens in that complementarity-determining regions do not participate. Structure determination revealed that its Fab-interacting domain adopts an immunoglobulin-like fold, fused to the preceding immunoglobulin-like domain and carried on a long stalk anchored to the bacterial outer membrane. Possible roles of this unusual host-pathogen interaction include avoidance of clearance from the intestine by secretory IgA.


Subject(s)
Adhesins, Bacterial/metabolism , Antibodies/metabolism , Immunoglobulin Fab Fragments/metabolism , Intestines/microbiology , Yersinia pseudotuberculosis Infections/microbiology , Yersinia pseudotuberculosis/pathogenicity , Adhesins, Bacterial/chemistry , Animals , Antibodies/immunology , Humans , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Variable Region , Intestines/immunology , Intestines/pathology , Yersinia pseudotuberculosis/immunology , Yersinia pseudotuberculosis Infections/metabolism , Yersinia pseudotuberculosis Infections/pathology
8.
J Biol Chem ; 293(22): 8672-8690, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29535184

ABSTRACT

Yersinia pseudotuberculosis is a Gram-negative bacterium and zoonotic pathogen responsible for a wide range of diseases, ranging from mild diarrhea, enterocolitis, lymphatic adenitis to persistent local inflammation. The Y. pseudotuberculosis invasin D (InvD) molecule belongs to the invasin (InvA)-type autotransporter proteins, but its structure and function remain unknown. In this study, we present the first crystal structure of InvD, analyzed its expression and function in a murine infection model, and identified its target molecule in the host. We found that InvD is induced at 37 °C and expressed in vivo 2-4 days after infection, indicating that InvD is a virulence factor. During infection, InvD was expressed in all parts of the intestinal tract, but not in deeper lymphoid tissues. The crystal structure of the C-terminal adhesion domain of InvD revealed a distinct Ig-related fold that, apart from the canonical ß-sheets, comprises various modifications of and insertions into the Ig-core structure. We identified the Fab fragment of host-derived IgG/IgA antibodies as the target of the adhesion domain. Phage display panning and flow cytometry data further revealed that InvD exhibits a preferential binding specificity toward antibodies with VH3/VK1 variable domains and that it is specifically recruited to a subset of B cells. This finding suggests that InvD modulates Ig functions in the intestine and affects direct interactions with a subset of cell surface-exposed B-cell receptors. In summary, our results provide extensive insights into the structure of InvD and its specific interaction with the target molecule in the host.


Subject(s)
Adhesins, Bacterial/metabolism , Antibodies/metabolism , Immunoglobulin Fab Fragments/metabolism , Intestines/microbiology , Yersinia pseudotuberculosis Infections/microbiology , Yersinia pseudotuberculosis/pathogenicity , Adhesins, Bacterial/chemistry , Amino Acid Sequence , Animals , Antibodies/immunology , Bacterial Adhesion , Female , Immunoglobulin Fab Fragments/immunology , Intestines/immunology , Intestines/pathology , Mice , Mice, Inbred BALB C , Models, Molecular , Protein Conformation , Sequence Homology , Virulence , Yersinia pseudotuberculosis/immunology , Yersinia pseudotuberculosis Infections/metabolism , Yersinia pseudotuberculosis Infections/pathology
9.
Article in English | MEDLINE | ID: mdl-29520342

ABSTRACT

Despite the mammalian host actively sequestering iron to limit pathogenicity, heme (or hemin when oxidized) and hemoproteins serve as important sources of iron for many bloodborne pathogens. The HmuRSTUV hemin uptake system allows Yersinia species to uptake and utilize hemin and hemoproteins as iron sources. HmuR is a TonB-dependent outer membrane receptor for hemin and hemoproteins. HmuTUV comprise a inner membrane ABC transporter that transports hemin and hemoproteins from the periplasmic space into the bacterial cytoplasm, where it is degraded by HmuS. Here we show that hmuSTUV but not hmuR are expressed under iron replete conditions, whereas hmuR as well as hmuSTUV are expressed under iron limiting conditions, suggesting complex transcriptional control. Indeed, expression of hmuSTUV in the presence of inorganic iron, but not in the presence of hemin, requires the global regulator IscR acting from a promoter in the intergenic region between hmuR and hmuS. This effect of IscR appears to be direct by binding a site mapped by DNaseI footprinting. In contrast, expression of hmuR under iron limiting conditions requires derepression of the ferric uptake regulator Fur acting from the hmuR promoter, as Fur binding upstream of hmuR was demonstrated biochemically. Differential expression by both Fur and IscR would facilitate maximal hemin uptake and utilization when iron and heme availability is low while maintaining the capacity for periplasmic removal and cytosolic detoxification of heme under a wider variety of conditions. We also demonstrate that a Y. pseudotuberculosis ΔiscR mutant has a survival defect when incubated in whole blood, in which iron is sequestered by heme-containing proteins. Surprisingly, this phenotype was independent of the Hmu system, the type III secretion system, complement, and the ability of Yersinia to replicate intracellularly. These results suggest that IscR regulates multiple virulence factors important for Yersinia survival and growth in mammalian tissues and reveal a surprising complexity of heme uptake expression and function under differing conditions of iron.


Subject(s)
Heme/metabolism , Hemin/genetics , Iron/metabolism , Yersinia pseudotuberculosis Infections/metabolism , Yersinia pseudotuberculosis Infections/microbiology , Yersinia pseudotuberculosis/physiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Base Sequence , Binding Sites , Gene Expression Regulation, Bacterial , Gene Order , Genetic Loci , Mutation , Promoter Regions, Genetic , Protein Binding , Transcription, Genetic , Yersinia pseudotuberculosis Infections/blood
10.
J Biol Chem ; 293(4): 1466-1479, 2018 01 26.
Article in English | MEDLINE | ID: mdl-29197826

ABSTRACT

Yersinia pseudotuberculosis is a foodborne pathogenic bacterium that causes acute gastrointestinal illness, but its mechanisms of infection are incompletely described. We examined how host cell sterol composition affected Y. pseudotuberculosis uptake. To do this, we depleted or substituted cholesterol in human MDA-MB-231 epithelial cells with various alternative sterols. Decreasing host cell cholesterol significantly reduced pathogen internalization. When host cell cholesterol was substituted with various sterols, only desmosterol and 7-dehydrocholesterol supported internalization. This specificity was not due to sterol dependence of bacterial attachment to host cells, which was similar with all sterols studied. Because a key step in Y. pseudotuberculosis internalization is interaction of the bacterial adhesins invasin and YadA with host cell ß1 integrin, we compared the sterol dependence of wildtype Y. pseudotuberculosis internalization with that of Δinv, ΔyadA, and ΔinvΔyadA mutant strains. YadA deletion decreased bacterial adherence to host cells, whereas invasin deletion had no effect. Nevertheless, host cell sterol substitution had a similar effect on internalization of these bacterial deletion strains as on the wildtype bacteria. The ΔinvΔyadA double mutant adhered least to cells and so was not significantly internalized. The sterol structure dependence of Y. pseudotuberculosis internalization differed from that of endocytosis, as monitored using antibody-clustered ß1 integrin and previous studies on other proteins, which had a more permissive sterol dependence. This study suggests that agents could be designed to interfere with internalization of Yersinia without disturbing endocytosis.


Subject(s)
Bacterial Adhesion , Dehydrocholesterols/metabolism , Integrin beta1/metabolism , Yersinia pseudotuberculosis Infections/metabolism , Yersinia pseudotuberculosis/metabolism , Cell Line, Tumor , Female , Gene Deletion , Humans , Yersinia pseudotuberculosis/genetics , Yersinia pseudotuberculosis/pathogenicity , Yersinia pseudotuberculosis Infections/genetics , Yersinia pseudotuberculosis Infections/pathology
11.
J Biol Chem ; 292(8): 3299-3311, 2017 02 24.
Article in English | MEDLINE | ID: mdl-28039361

ABSTRACT

Many pathogenic Gram-negative bacteria use the type III secretion system (T3SS) to deliver effector proteins into eukaryotic host cells. In Yersinia, the switch to secretion of effector proteins is induced first after intimate contact between the bacterium and its eukaryotic target cell has been established, and the T3SS proteins YscP and YscU play a central role in this process. Here we identify the molecular details of the YscP binding site on YscU by means of nuclear magnetic resonance (NMR) spectroscopy. The binding interface is centered on the C-terminal domain of YscU. Disrupting the YscU-YscP interaction by introducing point mutations at the interaction interface significantly reduced the secretion of effector proteins and HeLa cell cytotoxicity. Interestingly, the binding of YscP to the slowly self-cleaving YscU variant P264A conferred significant protection against autoproteolysis. The YscP-mediated inhibition of YscU autoproteolysis suggests that the cleavage event may act as a timing switch in the regulation of early versus late T3SS substrates. We also show that YscUC binds to the inner rod protein YscI with a dissociation constant (Kd ) of 3.8 µm and with 1:1 stoichiometry. The significant similarity among different members of the YscU, YscP, and YscI families suggests that the protein-protein interactions discussed in this study are also relevant for other T3SS-containing Gram-negative bacteria.


Subject(s)
Protein Interaction Maps , Type III Secretion Systems/metabolism , Yersinia pseudotuberculosis Infections/metabolism , Yersinia pseudotuberculosis/metabolism , HeLa Cells , Humans , Models, Molecular , Substrate Specificity , Type III Secretion Systems/chemistry , Yersinia pseudotuberculosis/chemistry , Yersinia pseudotuberculosis Infections/microbiology
12.
Infect Immun ; 85(2)2017 02.
Article in English | MEDLINE | ID: mdl-27872237

ABSTRACT

Signaling through Toll-like receptors (TLRs), the main receptors in innate immunity, is essential for the defense of mucosal surfaces. It was previously shown that systemic TLR5 stimulation by bacterial flagellin induces an immediate, transient interleukin-22 (IL-22)-dependent antimicrobial response to bacterial or viral infections of the mucosa. This process was dependent on the activation of type 3 innate lymphoid cells (ILCs). The objective of the present study was to analyze the effects of flagellin treatment in a murine model of oral infection with Yersinia pseudotuberculosis (an invasive, Gram-negative, enteropathogenic bacterium that targets the small intestine). We found that systemic administration of flagellin significantly increased the survival rate after intestinal infection (but not systemic infection) by Y. pseudotuberculosis This protection was associated with a low bacterial count in the gut and the spleen. In contrast, no protection was afforded by administration of the TLR4 agonist lipopolysaccharide, suggesting the presence of a flagellin-specific effect. Lastly, we found that TLR5- and MyD88-mediated signaling was required for the protective effects of flagellin, whereas neither lymphoid cells nor IL-22 was involved.


Subject(s)
Flagellin/immunology , Interleukins/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Yersinia pseudotuberculosis Infections/immunology , Yersinia pseudotuberculosis Infections/metabolism , Yersinia pseudotuberculosis/immunology , Animals , Disease Models, Animal , Female , Flagellin/administration & dosage , Interleukins/genetics , Intestinal Mucosa/microbiology , Lipopolysaccharides/immunology , Mice , Mice, Knockout , Recombinant Fusion Proteins , Signal Transduction , Toll-Like Receptors/metabolism , Yersinia pseudotuberculosis Infections/microbiology , Yersinia pseudotuberculosis Infections/mortality , Interleukin-22
13.
PLoS One ; 11(6): e0157092, 2016.
Article in English | MEDLINE | ID: mdl-27275606

ABSTRACT

Antimicrobial chemokines (AMCs) are a recently described family of host defense peptides that play an important role in protecting a wide variety of organisms from bacterial infection. Very little is known about the bacterial targets of AMCs or factors that influence bacterial susceptibility to AMCs. In an effort to understand how bacterial pathogens resist killing by AMCs, we screened Yersinia pseudotuberculosis transposon mutants for those with increased binding to the AMCs CCL28 and CCL25. Mutants exhibiting increased binding to AMCs were subjected to AMC killing assays, which revealed their increased sensitivity to chemokine-mediated cell death. The majority of the mutants exhibiting increased binding to AMCs contained transposon insertions in genes related to lipopolysaccharide biosynthesis. A particularly strong effect on susceptibility to AMC mediated killing was observed by disruption of the hldD/waaF/waaC operon, necessary for ADP-L-glycero-D-manno-heptose synthesis and a complete lipopolysaccharide core oligosaccharide. Periodate oxidation of surface carbohydrates also enhanced AMC binding, whereas enzymatic removal of surface proteins significantly reduced binding. These results suggest that the structure of Y. pseudotuberculosis LPS greatly affects the antimicrobial activity of AMCs by shielding a protein ligand on the bacterial cell surface.


Subject(s)
Antimicrobial Cationic Peptides/pharmacology , Chemokines, CC/pharmacology , Drug Resistance, Bacterial , Lipopolysaccharides , Operon , Yersinia pseudotuberculosis , Humans , Lipopolysaccharides/biosynthesis , Lipopolysaccharides/genetics , Yersinia pseudotuberculosis/enzymology , Yersinia pseudotuberculosis/genetics , Yersinia pseudotuberculosis/growth & development , Yersinia pseudotuberculosis Infections/genetics , Yersinia pseudotuberculosis Infections/metabolism
14.
PLoS One ; 10(8): e0133298, 2015.
Article in English | MEDLINE | ID: mdl-26275291

ABSTRACT

Microbial pathogens and host immune cells each initiate events following their interaction in an attempt to drive the outcome to their respective advantage. Here we show that the bacterial pathogen Yersinia pseudotuberculosis sustains itself on the surface of a macrophage by forming acidic fluid-accessible compartments that are partially bounded by the host cell plasma membrane. These Yersinia-containing acidic compartments (YACs) are bereft of the early endosomal marker EEA1 and the lysosomal antigen LAMP1 and readily form on primary macrophages as well as macrophage-like cell lines. YAC formation requires the presence of the Yersinia virulence plasmid which encodes a type III secretion system. Unexpectedly, we found that the initial formation of YACs did not require translocation of the type III effectors into the host cell cytosol; however, the duration of YACs was markedly greater in infections using translocation-competent Y. pseudotuberculosis strains as well as strains expressing the effector YopJ. Furthermore, it was in this translocation- and YopJ-dependent phase of infection that the acidic environment was critical for Y. pseudotuberculosis survival during its interaction with macrophages. Our findings indicate that during its extracellular phase of infection Y. pseudotuberculosis initiates and then, by a separate mechanism, stabilizes the formation of a highly intricate structure on the surface of the macrophage that is disengaged from the endocytic pathway.


Subject(s)
Macrophages/metabolism , Macrophages/microbiology , Yersinia pseudotuberculosis/physiology , Animals , Cell Line , Cells, Cultured , Mice , Yersinia pseudotuberculosis Infections/metabolism
15.
PLoS Pathog ; 11(7): e1005020, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26134274

ABSTRACT

Type VI secretion systems (T6SSs) are widespread multi-component machineries that translocate effectors into either eukaryotic or prokaryotic cells, for virulence or for interbacterial competition. Herein, we report that the T6SS-4 from Yersinia pseudotuberculosis displays an unexpected function in the transportation of Zn2+ to combat diverse stresses and host immunity. Environmental insults such as oxidative stress induce the expression of T6SS-4 via OxyR, the transcriptional factor that also regulates many oxidative response genes. Zinc transportation is achieved by T6SS-4-mediated translocation of a novel Zn2+-binding protein substrate YezP (YPK_3549), which has the capacity to rescue the sensitivity to oxidative stress exhibited by T6SS-4 mutants when added to extracellular milieu. Disruption of the classic zinc transporter ZnuABC together with T6SS-4 or yezP results in mutants that almost completely lost virulence against mice, further highlighting the importance of T6SS-4 in resistance to host immunity. These results assigned an unconventional role to T6SSs, which will lay the foundation for studying novel mechanisms of metal ion uptake by bacteria and the role of this process in their resistance to host immunity and survival in harmful environments.


Subject(s)
Adaptation, Physiological/immunology , Type VI Secretion Systems/metabolism , Yersinia pseudotuberculosis Infections/metabolism , Yersinia pseudotuberculosis/metabolism , Yersinia pseudotuberculosis/pathogenicity , Animals , Blotting, Western , Electrophoretic Mobility Shift Assay , Mice , Polymerase Chain Reaction , Real-Time Polymerase Chain Reaction , Stress, Physiological/immunology , Type VI Secretion Systems/immunology , Yersinia pseudotuberculosis Infections/immunology , Zinc/metabolism
16.
PLoS Pathog ; 9(11): e1003746, 2013.
Article in English | MEDLINE | ID: mdl-24244167

ABSTRACT

Some isolates of Yersinia pseudotuberculosis produce the cytotoxic necrotizing factor (CNFY), but the functional consequences of this toxin for host-pathogen interactions during the infection are unknown. In the present study we show that CNFY has a strong influence on virulence. We demonstrate that the CNFY toxin is thermo-regulated and highly expressed in all colonized lymphatic tissues and organs of orally infected mice. Most strikingly, we found that a cnfY knock-out variant of a naturally toxin-expressing Y. pseudotuberculosis isolate is strongly impaired in its ability to disseminate into the mesenteric lymph nodes, liver and spleen, and has fully lost its lethality. The CNFY toxin contributes significantly to the induction of acute inflammatory responses and to the formation of necrotic areas in infected tissues. The analysis of the host immune response demonstrated that presence of CNFY leads to a strong reduction of professional phagocytes and natural killer cells in particular in the spleen, whereas loss of the toxin allows efficient tissue infiltration of these immune cells and rapid killing of the pathogen. Addition of purified CNFY triggers formation of actin-rich membrane ruffles and filopodia, which correlates with the activation of the Rho GTPases, RhoA, Rac1 and Cdc42. The analysis of type III effector delivery into epithelial and immune cells in vitro and during the course of the infection further demonstrated that CNFY enhances the Yop translocation process and supports a role for the toxin in the suppression of the antibacterial host response. In summary, we highlight the importance of CNFY for pathogenicity by showing that this toxin modulates inflammatory responses, protects the bacteria from attacks of innate immune effectors and enhances the severity of a Yersinia infection.


Subject(s)
Bacterial Toxins/metabolism , Neuropeptides/metabolism , Yersinia pseudotuberculosis Infections/metabolism , Yersinia pseudotuberculosis/metabolism , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Bacterial Toxins/genetics , Enzyme Activation/genetics , Humans , Mice , Mice, Inbred BALB C , Neuropeptides/genetics , Protein Transport , Yersinia pseudotuberculosis/genetics , Yersinia pseudotuberculosis Infections/genetics , Yersinia pseudotuberculosis Infections/pathology , cdc42 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/genetics , rho GTP-Binding Proteins/genetics , rhoA GTP-Binding Protein
17.
PLoS Pathog ; 9(6): e1003415, 2013.
Article in English | MEDLINE | ID: mdl-23818844

ABSTRACT

Yersinia delivers Yops into numerous types of cultured cells, but predominantly into professional phagocytes and B cells during animal infection. The basis for this cellular tropism during animal infection is not understood. This work demonstrates that efficient and specific Yop translocation into phagocytes by Yersinia pseudotuberculosis (Yptb) is a multi-factorial process requiring several adhesins and host complement. When WT Yptb or a multiple adhesin mutant strain, ΔailΔinvΔyadA, colonized tissues to comparable levels, ΔailΔinvΔyadA translocated Yops into significantly fewer cells, demonstrating that these adhesins are critical for translocation into high numbers of cells. However, phagocytes were still selectively targeted for translocation, indicating that other bacterial and/or host factors contribute to this function. Complement depletion showed that complement-restricted infection by ΔailΔinvΔyadA but not WT, indicating that adhesins disarm complement in mice either by prevention of opsonophagocytosis or by suppressing production of pro-inflammatory cytokines. Furthermore, in the absence of the three adhesins and complement, the spectrum of cells targeted for translocation was significantly altered, indicating that Yersinia adhesins and complement direct Yop translocation into neutrophils during animal infection. In summary, these findings demonstrate that in infected tissues, Yersinia uses adhesins both to disarm complement-dependent killing and to efficiently translocate Yops into phagocytes.


Subject(s)
Adhesins, Bacterial/metabolism , Complement System Proteins/metabolism , Phagocytes/metabolism , Yersinia pseudotuberculosis Infections/metabolism , Yersinia pseudotuberculosis/metabolism , Adhesins, Bacterial/genetics , Animals , Complement System Proteins/genetics , Mice , Phagocytes/microbiology , Phagocytes/pathology , Protein Transport/genetics , Yersinia pseudotuberculosis/genetics , Yersinia pseudotuberculosis Infections/genetics , Yersinia pseudotuberculosis Infections/pathology
18.
PLoS Pathog ; 8(8): e1002828, 2012.
Article in English | MEDLINE | ID: mdl-22876175

ABSTRACT

A highly conserved virulence plasmid encoding a type III secretion system is shared by the three Yersinia species most pathogenic for mammals. Although factors encoded on this plasmid enhance the ability of Yersinia to thrive in their mammalian hosts, the loss of this virulence plasmid does not eliminate growth or survival in host organs. Most notably, yields of viable plasmid-deficient Yersinia pseudotuberculosis (Yptb) are indistinguishable from wild-type Yptb within mesenteric lymph nodes. To identify chromosomal virulence factors that allow for plasmid-independent survival during systemic infection of mice, we generated transposon insertions in plasmid-deficient Yptb, and screened a library having over 20,000 sequence-identified insertions. Among the previously uncharacterized loci, insertions in mrtAB, an operon encoding an ABC family transporter, had the most profound phenotype in a plasmid-deficient background. The absence of MrtAB, however, had no effect on growth in the liver and spleen of a wild type strain having an intact virulence plasmid, but caused a severe defect in colonization of the mesenteric lymph nodes. Although this result is consistent with lack of expression of the type III secretion system by Wt Yptb in the mesenteric lymph nodes, a reporter for YopE indicated that expression of the system was robust. We demonstrate that the ATPase activity of MrtB is required for growth in mice, indicating that transport activity is required for virulence. Indeed, MrtAB appears to function as an efflux pump, as the ATPase activity enhances resistance to ethidium bromide while increasing sensitivity to pyocyanin, consistent with export across the inner membrane.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Bacterial Proteins/metabolism , Lymph Nodes/microbiology , Mesentery/microbiology , RNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Virulence Factors/metabolism , Yersinia pseudotuberculosis Infections/metabolism , Yersinia pseudotuberculosis/pathogenicity , ATP-Binding Cassette Transporters/genetics , Animals , Bacterial Proteins/genetics , Lymph Nodes/metabolism , Lymph Nodes/pathology , Mesentery/metabolism , Mesentery/pathology , Mice , Mice, Inbred BALB C , RNA-Binding Proteins/genetics , Transcription Factors/genetics , Virulence Factors/genetics , Yersinia pseudotuberculosis/genetics , Yersinia pseudotuberculosis/metabolism , Yersinia pseudotuberculosis Infections/genetics , Yersinia pseudotuberculosis Infections/pathology
19.
Proc Natl Acad Sci U S A ; 109(27): E1830-8, 2012 Jul 03.
Article in English | MEDLINE | ID: mdl-22691497

ABSTRACT

The cycle inhibiting factors (Cifs) are a family of translocated effector proteins, found in diverse pathogenic bacteria, that interfere with the host cell cycle by catalyzing the deamidation of a specific glutamine residue (Gln40) in NEDD8 and the related protein ubiquitin. This modification prevents recycling of neddylated cullin-RING ligases, leading to stabilization of various cullin-RING ligase targets, and also prevents polyubiquitin chain formation. Here, we report the crystal structures of two Cif/NEDD8 complexes, revealing a conserved molecular interface that defines enzyme/substrate recognition. Mutation of residues forming the interface suggests that shape complementarity, rather than specific individual interactions, is a critical feature for complex formation. We show that Cifs from diverse bacteria bind NEDD8 in vitro and conclude that they will all interact with their substrates in the same way. The "occluding loop" in Cif gates access to Gln40 by forcing a conformational change in the C terminus of NEDD8. We used native PAGE to follow the activity of Cif from the human pathogen Yersinia pseudotuberculosis and selected variants, and the position of Gln40 in the active site has allowed us to propose a catalytic mechanism for these enzymes.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Photorhabdus/enzymology , Ubiquitins/chemistry , Ubiquitins/metabolism , Yersinia pseudotuberculosis/enzymology , Amino Acid Sequence , Bacterial Proteins/genetics , Catalytic Domain , Crystallization , Glutamine/genetics , HeLa Cells , Host-Parasite Interactions/physiology , Humans , Molecular Sequence Data , Mutagenesis/physiology , NEDD8 Protein , Oncogene Protein p21(ras)/metabolism , Photorhabdus/genetics , Polyubiquitin/metabolism , Protein Binding/physiology , Protein Structure, Tertiary , Ubiquitins/genetics , Virulence Factors/chemistry , Virulence Factors/genetics , Virulence Factors/metabolism , Yersinia pseudotuberculosis/genetics , Yersinia pseudotuberculosis Infections/metabolism , Yersinia pseudotuberculosis Infections/microbiology
20.
Cell Host Microbe ; 11(4): 337-51, 2012 Apr 19.
Article in English | MEDLINE | ID: mdl-22520462

ABSTRACT

Yersinia pseudotuberculosis is an enteropathogenic bacteria that disrupts the intestinal barrier and invades its host through gut-associated lymphoid tissue and Peyer's patches (PP). We show that the Y. pseudotuberculosis effector YopJ induces intestinal barrier dysfunction by subverting signaling of the innate immune receptor Nod2, a phenotype that can be reversed by pretreating with the Nod2 ligand muramyl-dipeptide. YopJ, but not the catalytically inactive mutant YopJ(C172A), acetylates critical sites in the activation loops of the RICK and TAK1 kinases, which are central mediators of Nod2 signaling, and decreases the affinity of Nod2 for RICK. Concomitantly, Nod2 interacts with and activates caspase-1, resulting in increased levels of IL-1ß. Finally, IL-1ß within PP plays an essential role in inducing intestinal barrier dysfunction. Thus, YopJ alters intestinal permeability and promotes the dissemination of Yersinia as well as commensal bacteria by exploiting the mucosal inflammatory response.


Subject(s)
Bacterial Proteins/metabolism , Caspase 1/metabolism , Intestines/enzymology , MAP Kinase Kinase Kinases/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Yersinia pseudotuberculosis Infections/enzymology , Yersinia pseudotuberculosis/metabolism , Animals , Bacterial Proteins/genetics , Caspase 1/genetics , Cell Line , Female , Humans , Intestinal Mucosa/metabolism , Intestines/microbiology , MAP Kinase Kinase Kinases/genetics , Male , Mice , Mice, Inbred C57BL , Nod2 Signaling Adaptor Protein/genetics , Receptor-Interacting Protein Serine-Threonine Kinase 2/genetics , Signal Transduction , Yersinia pseudotuberculosis/genetics , Yersinia pseudotuberculosis Infections/genetics , Yersinia pseudotuberculosis Infections/metabolism , Yersinia pseudotuberculosis Infections/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL
...