Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
J Sex Med ; 19(6): 1024-1031, 2022 06.
Article in English | MEDLINE | ID: mdl-35414488

ABSTRACT

BACKGROUND: Delayed infection, thought to be due to gradual biofilm formation, remains a feared complication after inflatable penile prosthesis (IPP) insertion. Understanding and preventing biofilm formation is necessary to prevent infections. AIM: To develop an in vitro model and compare growth of biofilm by different bacteria on IPPs and evaluate the anti-infective efficacy of the Coloplast Titan and AMS 700 InhibiZone. METHODS: Sterile IPPs (Coloplast) were cut into rings and incubated with S. epidermidis, S. aureus, P. aeruginosa, A. baumannii, or K. pneumoniae cultures in tryptic soy broth (TSB) (4 hour) to ensure adequate bacteria attachment, and then in only TSB (120 hours) to allow for biofilm formation. Rings were fixed with ethanol and biofilm measured by spectrophotometer (OD570) after crystal violet staining. This methodology was repeated for S. epidermidis and P. aeruginosa with Coloplast rings dipped in 10 ml of a 10 mg/ml Rifampin, 1 mg/ml Gentamicin, and deionized water solution and undipped AMS InhibiZone rings. Crystal violet assay (OD570) was repeated after incubation within bacteria (2 hour), and then only TSB (120 hours). OUTCOMES: The primary outcome of the study was OD570 readings, indirectly measuring biofilm mass on implant rings. RESULTS: S. epidermidis, S. aureus, A. baumannii, P. aeruginosa, and K. pneumoniae all formed significant biofilm. P. aeruginosa showed the strongest predilection to grow biofilm on IPPs. P. aeruginosa also formed significant biofilm on antibiotic-treated Coloplast and AMS rings, while S. epidermidis was inhibited. No significant difference was found in biofilm inhibition between the implants. CLINICAL TRANSLATION: Our findings suggest gram-negative bacteria may form biofilm more proficiently and quickly on IPPs than gram-positive organisms. Commonly used antibiotic treatments on IPPs may be effective against S. epidermidis but not against P. aeruginosa biofilm formation. STRENGTHS & LIMITATIONS: This is the first study comparing biofilm formation by different bacteria organisms on IPPs and the inhibitive ability of Coloplast and AMS implants against biofilm formation. Clinical data on organisms responsible for infected IPPs is needed to determine the clinical relevance of our findings. CONCLUSION: Our novel in vitro model of biofilm formation of IPPs evaluated the effect of a gentamicin/rifampin antibiotic dip on Coloplast Titan implants and the anti-infective capacity of the minocycline/rifampin precoated AMS 700 InhibiZone against S. epidermidis and P. aeruginosa. P. aeruginosa was able to grow on both antibiotic-treated implants, with no significant difference, and should continue to be a specific target of investigation to reduce delayed post-operative IPP infections. Narasimman M, Ory J, Bartra SS, et al. Evaluation of Bacteria in a Novel In Vitro Biofilm Model of Penile Prosthesis. J Sex Med 2022;19:1024-1031.


Subject(s)
Penile Prosthesis , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Biofilms , Gentamicins/pharmacology , Gentian Violet , Humans , Rifampin/therapeutic use , Staphylococcus aureus
3.
Microbiol Spectr ; 9(1): e0000521, 2021 09 03.
Article in English | MEDLINE | ID: mdl-34132578

ABSTRACT

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.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Disk Diffusion Antimicrobial Tests/methods , Oxalic Acid/pharmacology , Type III Secretion Systems/antagonists & inhibitors , Yersinia pestis/drug effects , Bacterial Proteins/metabolism , Disk Diffusion Antimicrobial Tests/instrumentation , Humans , Plague/microbiology , Type III Secretion Systems/metabolism , Yersinia pestis/growth & development , Yersinia pestis/metabolism
4.
Microb Pathog ; 154: 104852, 2021 May.
Article in English | MEDLINE | ID: mdl-33762201

ABSTRACT

Cellular Perforin-2 (MPEG1) is a pore-forming MACPF family protein that plays a critical role in the defense against bacterial pathogens. Macrophages, neutrophils, and several other cell types that are part of the front line of innate defenses constitutively express high levels of Perforin-2; whereas, most other cell types must be induced to express Perforin-2 by interferons (α, ß and γ) and/or PAMPs such as LPS. In this study, we demonstrate that many bacterial pathogens can limit the expression of Perforin-2 in cells normally inducible for Perforin-2 expression, while ordinarily commensal or non-pathogenic bacteria triggered high levels of Perforin-2 expression in these same cell types. The mechanisms by which pathogens suppress Perforin-2 expression was explored further using Salmonella enterica serovar Typhimurium and cultured MEFs as well as intestinal epithelial cell lines. These studies identified multiple factors required to minimize the expression of Perforin-2 in cell types inducible for Perforin-2 expression. These included the PmrAB and PhoPQ two-component systems, select LPS modification enzymes and the Type III secretion effector protein AvrA.


Subject(s)
Lipopolysaccharides , Salmonella typhimurium , Bacterial Proteins/genetics , Epithelial Cells , Fibroblasts , Perforin/genetics , Serogroup
5.
Biophys J ; 120(3): 453-462, 2021 02 02.
Article in English | MEDLINE | ID: mdl-33359463

ABSTRACT

Understanding microbe-host interactions at the molecular level is a major goal of fundamental biology and therapeutic drug development. Structural biology strives to capture biomolecular structures in action, but the samples are often highly simplified versions of the complex native environment. Here, we present an Escherichia coli model system that allows us to probe the structure and function of Ail, the major surface protein of the deadly pathogen Yersinia pestis. We show that cell surface expression of Ail produces Y. pestis virulence phenotypes in E. coli, including resistance to human serum, cosedimentation of human vitronectin, and pellicle formation. Moreover, isolated bacterial cell envelopes, encompassing inner and outer membranes, yield high-resolution solid-state NMR spectra that reflect the structure of Ail and reveal Ail sites that are sensitive to the bacterial membrane environment and involved in the interactions with human serum components. The data capture the structure and function of Ail in a bacterial outer membrane and set the stage for probing its interactions with the complex milieu of immune response proteins present in human serum.


Subject(s)
Yersinia pestis , Bacterial Outer Membrane Proteins , Escherichia coli , Humans , Virulence , Virulence Factors
6.
Front Immunol ; 11: 550946, 2020.
Article in English | MEDLINE | ID: mdl-33042139

ABSTRACT

Perforin-2 (P-2) is an antimicrobial protein with unique properties to kill intracellular bacteria. Gamma delta (GD) T cells, as the major T cell population in epithelial tissues, play a central role in protective and pathogenic immune responses in the skin. However, the tissue-specific mechanisms that control the innate immune response and the effector functions of GD T cells, especially the cross-talk with commensal organisms, are not very well understood. We hypothesized that the most prevalent skin commensal microorganism, Staphylococcus epidermidis, may play a role in regulating GD T cell-mediated cutaneous responses. We analyzed antimicrobial protein P-2 expression in human skin at a single cell resolution using an amplified fluorescence in situ hybridization approach to detect P-2 mRNA in combination with immunophenotyping. We show that S. epidermidis activates GD T cells and upregulates P-2 in human skin ex vivo in a cell-specific manner. Furthermore, P-2 upregulation following S. epidermidis stimulation correlates with increased ability of skin cells to kill intracellular Staphylococcus aureus. Our findings are the first to reveal that skin commensal bacteria induce P-2 expression, which may be utilized beneficially to modulate host innate immune responses and protect from skin infections.


Subject(s)
Immunity, Innate , Pore Forming Cytotoxic Proteins/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Staphylococcal Skin Infections/immunology , Staphylococcal Skin Infections/metabolism , Staphylococcus epidermidis/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Biomarkers , Cytokines/metabolism , Cytotoxicity, Immunologic , Fibroblasts/metabolism , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immunomodulation , Inflammation Mediators/metabolism , Keratinocytes/immunology , Keratinocytes/metabolism , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphocyte Count , Pore Forming Cytotoxic Proteins/genetics , Staphylococcal Skin Infections/microbiology
7.
J Bacteriol ; 202(15)2020 07 09.
Article in English | MEDLINE | ID: mdl-32424009

ABSTRACT

Chlamydia trachomatis Scc4 (formerly CT663) engages the transcription machinery and the pathogenic type III secretion system (T3SS). Both machines are required for Chlamydia infection. These requirements and the limited ability for genetic manipulation in Chlamydia have hampered dissection of Scc4's contributions. Here, by developing bacterial systems that permit the controlled expression and stable maintenance of Scc4, we assess Scc4's effects on chlamydial growth phenotype, secretion, and the patterns of T3SS gene expression. Expressing Scc4 in Escherichia coli lacking a T3SS injectisome causes a growth defect. This deficiency is rescued by overexpressing the ß-subunit of RNA polymerase (RNAP) or by exploiting sigma 70 (σ70) (homologous to chlamydial σ66) mutants that strengthen the interaction between σ70 region 4 and the ß-flap, confirming Scc4's distinction as a module of RNAP holoenzyme capable of modulating transcription. Yersinia pestis expressing Scc4 sustains a functional T3SS, through which CopN secretion is boosted by cooption of Scc4 and Scc1. Finally, conditional expression of Scc4 in C. trachomatis results in fast expansion of the Chlamydia-containing vacuole and accelerated chlamydial development, coupled to selective up- or downregulation of gene expression from different T3SS genes. This work reveals, for the first time, the context-dependent action of Scc4 linking it to diverse protein networks in bacteria. It establishes that Scc4, when overexpressed, exerts incredible effects on chlamydial development by reinforcing control of the T3SS.IMPORTANCE The T3SS is a key virulence factor required for C. trachomatis infection. The control of the T3SS has not been well studied in this obligate intracellular pathogen. Here, we show that Scc4 plays a major role for precise control of the pathogenic T3SS at the levels of gene expression and effector secretion through genetically separable protein networks, allowing a fast adaptive mode of C. trachomatis development during infection in human epithelial cells.


Subject(s)
Bacterial Proteins/metabolism , Chlamydia trachomatis/metabolism , Type III Secretion Systems/metabolism , Bacterial Proteins/genetics , Chlamydia Infections/microbiology , Chlamydia trachomatis/genetics , Chlamydia trachomatis/growth & development , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Gene Expression Regulation, Bacterial , Humans , Protein Transport , Sigma Factor/genetics , Sigma Factor/metabolism , Type III Secretion Systems/genetics
8.
Mol Microbiol ; 114(3): 510-520, 2020 09.
Article in English | MEDLINE | ID: mdl-32462782

ABSTRACT

The outer membrane is a key virulence determinant of gram-negative bacteria. In Yersinia pestis, the deadly agent that causes plague, the protein Ail and lipopolysaccharide (LPS)6 enhance lethality by promoting resistance to human innate immunity and antibiotics, enabling bacteria to proliferate in the human host. Their functions are highly coordinated. Here we describe how they cooperate to promote pathogenesis. Using a multidisciplinary approach, we identify mutually constructive interactions between Ail and LPS that produce an extended conformation of Ail at the membrane surface, cause thickening and rigidification of the LPS membrane, and collectively promote Y. pestis survival in human serum, antibiotic resistance, and cell envelope integrity. The results highlight the importance of the Ail-LPS assembly as an organized whole, rather than its individual components, and provide a handle for targeting Y. pestis pathogenesis.


Subject(s)
Bacterial Outer Membrane Proteins/immunology , Bacterial Outer Membrane Proteins/metabolism , Lipopolysaccharides/immunology , Lipopolysaccharides/metabolism , Virulence Factors/immunology , Virulence Factors/metabolism , Yersinia pestis/immunology , Yersinia pestis/metabolism , Amino Acid Motifs , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial , Humans , Microbial Sensitivity Tests , Molecular Dynamics Simulation , Mutation , Plague/immunology , Plague/microbiology , Protein Binding , Protein Conformation , Yersinia pestis/drug effects
9.
J Immunol ; 204(8): 2242-2256, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32161097

ABSTRACT

Type I IFNs play a complex role in determining the fate of microbial pathogens and may also be deleterious to the host during bacterial and viral infections. Upon ligand binding, a receptor proximal complex consisting of IFN-α and -ß receptors 1 and 2 (IFNAR1, IFNAR2, respectively), tyrosine kinase 2 (Tyk2), Jak1, and STAT2 are assembled and promote the phosphorylation of STAT1 and STAT2. However, how the IFNARs proximal complex is assembled upon binding to IFN is poorly understood. In this study, we show that the membrane-associated pore-forming protein Perforin-2 (P2) is critical for LPS-induced endotoxic shock in wild-type mice. Type I IFN-mediated JAK-STAT signaling is severely impaired, and activation of MAPKs and PI3K signaling pathways are delayed in P2-deficient mouse bone marrow-derived macrophages, mouse embryonic fibroblasts (MEFs), and human HeLa cells upon IFN stimulation. The P2 N-glycosylated extracellular membrane attack complex/perforin domain and the P2 domain independently associate with the extracellular regions of IFNAR1 and IFNAR2, respectively, in resting MEFs. In addition, the P2 cytoplasmic tail domain mediated the constitutive interaction between STAT2 and IFNAR2 in resting MEFs, an interaction that is dependent on the association of the extracellular regions of P2 and IFNAR2. Finally, the constitutive association of P2 with both receptors and STAT2 is critical for the receptor proximal complex assembly and reciprocal transphosphorylation of Jak1 and Tyk2 as well as the phosphorylation and activation of STAT1 and STAT2 upon IFN-ß stimulation.


Subject(s)
Interferon Type I/immunology , Interferon Type I/metabolism , Pore Forming Cytotoxic Proteins/metabolism , Signal Transduction/immunology , Animals , Cells, Cultured , HeLa Cells , Humans , Lipopolysaccharides , Mice , Mice, Knockout , Shock, Septic/chemically induced , Shock, Septic/immunology
10.
Microb Pathog ; 141: 103993, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31988008

ABSTRACT

Yersinia pestis, a Gram-negative bacterium, is the etiologic agent of plague. A hallmark of Y. pestis infection is the organism's ability to rapidly disseminate through an animal host. Y. pestis expresses the outer membrane protein, Ail (Attachment invasion locus), which is associated with host invasion and serum resistance. However, whether Ail plays a role in host dissemination remains unclear. In this study, C57BL/6J mice were challenged with a defined Y. pestis strain, KimD27, or an isogenic ail-deleted mutant derived from KimD27 via metacarpal paw pad inoculation, nasal drops, orogastric infection, or tail vein injection to mimic bubonic, pneumonic, oral, or septicemic plague, respectively. Our results showed that ail-deleted Y. pestis KimD27 lost the ability to invade host cells, leading to failed host dissemination in the pneumonic and oral plague models but not in the bubonic or septicemic plague models, which do not require invasiveness. Therefore, this study demonstrated that whether Ail plays a role in Y. pestis pathogenesis depends on the infection route.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Plague/microbiology , Virulence Factors/metabolism , Virulence , Yersinia pestis , Animals , Bacterial Proteins/metabolism , Disease Models, Animal , Lung/microbiology , Mice , Mice, Inbred C57BL , Mouth/microbiology , Yersinia pestis/metabolism , Yersinia pestis/pathogenicity
11.
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
12.
Sci Adv ; 5(9): eaax5068, 2019 09.
Article in English | MEDLINE | ID: mdl-31535027

ABSTRACT

Vitronectin (Vn) is a major component of blood that controls many processes central to human biology. It is a drug target and a key factor in cell and tissue engineering applications, but despite long-standing efforts, little is known about the molecular basis for its functions. Here, we define the domain organization of Vn, report the crystal structure of its carboxyl-terminal domain, and show that it harbors the binding site for the Yersinia pestis outer membrane protein Ail, which recruits Vn to the bacterial cell surface to evade human host defenses. Vn forms a single four-bladed ß/α-propeller that serves as a hub for multiple functions. The structure explains key features of native Vn and provides a blueprint for understanding and targeting this essential human protein.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Virulence Factors/metabolism , Vitronectin/metabolism , Yersinia pestis/metabolism , Amino Acid Sequence , Bacterial Outer Membrane Proteins/chemistry , Binding Sites , Crystallography, X-Ray , Humans , Protein Binding , Protein Conformation , Sequence Homology , Virulence Factors/chemistry , Vitronectin/chemistry
13.
Article in English | MEDLINE | ID: mdl-30854334

ABSTRACT

Yersinia pestis, the causative agent of plague, possesses a number of virulence mechanisms that allows it to survive and proliferate during its interaction with the host. To discover additional infection-specific Y. pestis factors, a transposon site hybridization (TraSH)-based genome-wide screen was employed to identify genomic regions required for its survival during cellular infection. In addition to several well-characterized infection-specific genes, this screen identified three chromosomal genes (y3397, y3399, and y3400), located in an apparent operon, that promoted successful infection. Each of these genes is predicted to encode a leucine-rich repeat family protein with or without an associated ubiquitin E3 ligase domain. These genes were designated Yersinia leucine-rich repeat gene A (ylrA), B (ylrB), and C (ylrC). Engineered strains with deletions of y3397 (ylrC), y3399 (ylrB), or y3400 (ylrA), exhibited infection defects both in cultured cells and in the mouse. C-terminal FLAG-tagged YlrA, YlrB, and YlrC were secreted by Y. pestis in the absence but not the presence of extracellular calcium and deletions of the DNA sequences encoding the predicted N-terminal type III secretion signals of YlrA, YlrB, and YlrC prevented their secretion, indicating that these proteins are substrates of the type III secretion system (T3SS). Further strengthening the connection with the T3SS, YlrB was readily translocated into HeLa cells and expression of the YlrA and YlrC proteins in yeast inhibited yeast growth, indicating that these proteins may function as anti-host T3S effector proteins.


Subject(s)
Host-Pathogen Interactions , Plague/physiopathology , Type III Secretion Systems/metabolism , Virulence Factors/metabolism , Yersinia pestis/pathogenicity , Animals , Biological Transport , Chromosomes, Bacterial , Disease Models, Animal , Gene Deletion , Genes, Bacterial , Genetic Testing , HeLa Cells , Humans , Mice , Models, Theoretical , RAW 264.7 Cells , Virulence , Virulence Factors/genetics , Yersinia pestis/genetics
14.
Front Immunol ; 10: 96, 2019.
Article in English | MEDLINE | ID: mdl-30915064

ABSTRACT

Yersinia pestis, a Gram-negative bacterium and the etiologic agent of plague, has evolved from Yersinia pseudotuberculosis, a cause of a mild enteric disease. However, the molecular and biological mechanisms of how Y. pseudotuberculosis evolved to such a remarkably virulent pathogen, Y. pestis, are not clear. The ability to initiate a rapid bacterial dissemination is a characteristic hallmark of Y. pestis infection. A distinguishing characteristic between the two Yersinia species is that Y. pseudotuberculosis strains possess an O-antigen of lipopolysaccharide (LPS) while Y. pestis has lost the O-antigen during evolution and therefore exposes its core LPS. In this study, we showed that Y. pestis utilizes its core LPS to interact with SIGNR1 (CD209b), a C-type lectin receptor on antigen presenting cells (APCs), leading to bacterial dissemination to lymph nodes, spleen and liver, and the initiation of a systemic infection. We therefore propose that the loss of O-antigen represents a critical step in the evolution of Y. pseudotuberculosis into Y. pestis in terms of hijacking APCs, promoting bacterial dissemination and causing the plague.


Subject(s)
Cell Adhesion Molecules/immunology , Host-Pathogen Interactions/immunology , Lectins, C-Type/immunology , Lipopolysaccharides/immunology , Plague/immunology , Receptors, Cell Surface/immunology , Yersinia pestis/physiology , Animals , Antigen-Presenting Cells/immunology , Cell Adhesion Molecules/genetics , Cell Line , Female , HeLa Cells , Humans , Lectins, C-Type/genetics , Macrophages/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Receptors, Cell Surface/genetics , Yersinia pseudotuberculosis/physiology , Yersinia pseudotuberculosis Infections/immunology
15.
J Immunol ; 201(9): 2710-2720, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30249808

ABSTRACT

Perforin-2, the product of the MPEG1 gene, limits the spread and dissemination of bacterial pathogens in vivo. It is highly expressed in murine and human phagocytes, and macrophages lacking Perforin-2 are compromised in their ability to kill phagocytosed bacteria. In this study, we used Salmonella enterica serovar Typhimurium as a model intracellular pathogen to elucidate the mechanism of Perforin-2's bactericidal activity. In vitro Perforin-2 was found to facilitate the degradation of Ags contained within the envelope of phagocytosed bacteria. In contrast, degradation of a representative surface Ag was found to be independent of Perforin-2. Consistent with our in vitro results, a protease-sensitive, periplasmic superoxide dismutase (SodCII) contributed to the virulence of S. Typhimurium in Perforin-2 knockout but not wild-type mice. In aggregate, our studies indicate that Perforin-2 breaches the envelope of phagocytosed bacteria, facilitating the delivery of proteases and other antimicrobial effectors to sites within the bacterial cell.


Subject(s)
Pore Forming Cytotoxic Proteins/immunology , Salmonella Infections, Animal/immunology , Animals , Cell Wall , Mice , Mice, Knockout , Phagocytosis/immunology , Pore Forming Cytotoxic Proteins/metabolism , Salmonella Infections, Animal/metabolism , Salmonella typhimurium
16.
Microbiology (Reading) ; 164(3): 338-348, 2018 03.
Article in English | MEDLINE | ID: mdl-29458689

ABSTRACT

The needle structures of type III secretion (T3S) systems are formed by the secretion and polymerization of a needle subunit protein, YscF in Yersinia pestis. A subset of T3S systems employ unique heterodimeric chaperones, YscE and YscG in Y. pestis, to prevent the polymerization of needle subunits within the bacterial cell. We demonstrate that the YscE/YscG chaperone is also required for stable YscF expression and for secretion of YscF. Overexpression of a functional maltose-binding protein (MBP)-YscG hybrid protein stabilized cytoplasmic YscF but YscF was not secreted in the absence of YscE. Furthermore, a YscE mutant protein was identified that functioned with YscG to stabilize cytosolic YscF; however, YscF was not secreted. These findings confirm a role for the YscE/YscG chaperone in YscF secretion and suggest that YscE may have a specific role in this process. Recent studies have shown that YscF deleted of its N-terminal 15 residues is still secreted and functional, suggesting that YscF may not require an N-terminal secretion signal. However, we demonstrate that YscF contains an N-terminal secretion signal and that a functional N-terminal signal is required for YscF secretion.


Subject(s)
Bacterial Proteins/metabolism , Membrane Proteins/metabolism , Molecular Chaperones/metabolism , Protein Sorting Signals/genetics , Type III Secretion Systems/metabolism , Yersinia pestis/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Cytoplasm/metabolism , Gene Expression , Gene Expression Regulation, Bacterial , Membrane Proteins/genetics , Molecular Chaperones/genetics , Mutation , Protein Binding , Protein Multimerization , Yersinia pestis/genetics
17.
Methods Mol Biol ; 1531: 111-119, 2017.
Article in English | MEDLINE | ID: mdl-27837486

ABSTRACT

Numerous bacterial pathogens employ specialized protein secretion machineries to directly inject anti-host proteins, termed effector proteins, into eukaryotic cells. Effector proteins carrying small phosphorylatable tags can be used to detect and quantify effector protein injection. Here, we describe the use of the ELK- and GSK-tags to detect the translocation of the Y. pestis YopE effector protein into RAW 264.7 macrophage-like cells using immunoblot analysis with phospho-specific antibodies.


Subject(s)
Antibodies, Phospho-Specific , Bacterial Proteins/metabolism , Bacterial Secretion Systems/metabolism , Epitopes , Recombinant Fusion Proteins/metabolism , Amino Acid Sequence , Animals , Antibodies, Phospho-Specific/immunology , Bacterial Proteins/chemistry , Blotting, Western , Cell Line , Epitopes/chemistry , Epitopes/immunology , Gene Order , Mice , Phosphorylation , Plasmids/genetics , Protein Transport , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/immunology , Yersinia pestis/immunology , Yersinia pestis/metabolism
18.
Microbiology (Reading) ; 161(11): 2174-2183, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26377177

ABSTRACT

Yersinia pestis, the agent of plague, requires the Ail (attachment invasion locus) outer membrane protein to survive in the blood and tissues of its mammalian hosts. Ail is important for both attachment to host cells and for resistance to complement-dependent bacteriolysis. Previous studies have shown that Ail interacts with components of the extracellular matrix, including fibronectin, laminin and heparan sulfate proteoglycans, and with the complement inhibitor C4b-binding protein. Here, we demonstrate that Ail-expressing Y. pestis strains bind vitronectin - a host protein with functions in cell attachment, fibrinolysis and inhibition of the complement system. The Ail-dependent recruitment of vitronectin resulted in efficient cleavage of vitronectin by the outer membrane Pla (plasminogen activator protease). Escherichia coli DH5α expressing Y. pestis Ail bound vitronectin, but not heat-treated vitronectin. The ability of Ail to directly bind vitronectin was demonstrated by ELISA using purified refolded Ail in nanodiscs.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Host-Pathogen Interactions , Virulence Factors/metabolism , Vitronectin/metabolism , Yersinia pestis/metabolism , Bacterial Proteins/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Hydrolysis , Plasminogen Activators/metabolism , Protein Binding
19.
Elife ; 42015 Sep 24.
Article in English | MEDLINE | ID: mdl-26402460

ABSTRACT

Perforin-2 (MPEG1) is a pore-forming, antibacterial protein with broad-spectrum activity. Perforin-2 is expressed constitutively in phagocytes and inducibly in parenchymal, tissue-forming cells. In vitro, Perforin-2 prevents the intracellular replication and proliferation of bacterial pathogens in these cells. Perforin-2 knockout mice are unable to control the systemic dissemination of methicillin-resistant Staphylococcus aureus (MRSA) or Salmonella typhimurium and perish shortly after epicutaneous or orogastric infection respectively. In contrast, Perforin-2-sufficient littermates clear the infection. Perforin-2 is a transmembrane protein of cytosolic vesicles -derived from multiple organelles- that translocate to and fuse with bacterium containing vesicles. Subsequently, Perforin-2 polymerizes and forms large clusters of 100 Å pores in the bacterial surface with Perforin-2 cleavage products present in bacteria. Perforin-2 is also required for the bactericidal activity of reactive oxygen and nitrogen species and hydrolytic enzymes. Perforin-2 constitutes a novel and apparently essential bactericidal effector molecule of the innate immune system.


Subject(s)
Immunity, Innate , Pore Forming Cytotoxic Proteins/metabolism , Salmonella Infections, Animal/immunology , Salmonella typhimurium/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Animals , Cells, Cultured , Disease Models, Animal , Mice, Knockout , Microbial Viability , Phagocytes , Salmonella typhimurium/drug effects , Salmonella typhimurium/physiology , Staphylococcus aureus/drug effects , Staphylococcus aureus/physiology , Survival Analysis , Vacuoles/microbiology
20.
J Mol Biol ; 427(19): 3096-109, 2015 Sep 25.
Article in English | MEDLINE | ID: mdl-26259880

ABSTRACT

The type III secretion system (T3SS) is essential in the pathogenesis of Yersinia pestis, the causative agent of plague. A small protein, LcrG, functions as a chaperone to the tip protein LcrV, and the LcrG-LcrV interaction is important in regulating protein secretion through the T3SS. The atomic structure of the LcrG family is currently unknown. However, because of its predicted helical propensity, many have suggested that the LcrG family forms a coiled-coil structure. Here, we show by NMR and CD spectroscopy that LcrG lacks a tertiary structure and it consists of three partially folded α-helices spanning residues 7-38, 41-46, and 58-73. NMR titrations of LcrG with LcrV show that the entire length of a truncated LcrG (residues 7-73) is involved in binding to LcrV. However, there is regional variation in how LcrG binds to LcrV. The C-terminal region of a truncated LcrG (residues 52-73) shows tight binding interaction with LcrV while the N-terminal region (residues 7-51) shows weaker interaction with LcrV. This suggests that there are at least two binding events when LcrG binds to LcrV. Biological assays and mutagenesis indicate that the C-terminal region of LcrG (residues 52-73) is important in blocking protein secretion through the T3SS. Our results reveal structural and mechanistic insights into the atomic conformation of LcrG and how it binds to LcrV.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Plague/microbiology , Pore Forming Cytotoxic Proteins/chemistry , Pore Forming Cytotoxic Proteins/metabolism , Type III Secretion Systems/metabolism , Yersinia pestis/metabolism , Amino Acid Sequence , Animals , Antigens, Bacterial/chemistry , Humans , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Molecular Dynamics Simulation , Molecular Sequence Data , Protein Binding , Protein Folding , Protein Structure, Secondary , Sequence Alignment , Type III Secretion Systems/chemistry , Yersinia pestis/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...