Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
Proc Natl Acad Sci U S A ; 114(35): 9439-9444, 2017 08 29.
Article in English | MEDLINE | ID: mdl-28808028

ABSTRACT

Staphylococcus aureus is highly adapted to its host and has evolved many strategies to resist opsonization and phagocytosis. Even after uptake by neutrophils, S. aureus shows resistance to killing, which suggests the presence of phagosomal immune evasion molecules. With the aid of secretome phage display, we identified a highly conserved protein that specifically binds and inhibits human myeloperoxidase (MPO), a major player in the oxidative defense of neutrophils. We have named this protein "staphylococcal peroxidase inhibitor" (SPIN). To gain insight into inhibition of MPO by SPIN, we solved the cocrystal structure of SPIN bound to a recombinant form of human MPO at 2.4-Å resolution. This structure reveals that SPIN acts as a molecular plug that prevents H2O2 substrate access to the MPO active site. In subsequent experiments, we observed that SPIN expression increases inside the neutrophil phagosome, where MPO is located, compared with outside the neutrophil. Moreover, bacteria with a deleted gene encoding SPIN showed decreased survival compared with WT bacteria after phagocytosis by neutrophils. Taken together, our results demonstrate that S. aureus secretes a unique proteinaceous MPO inhibitor to enhance survival by interfering with MPO-mediated killing.


Subject(s)
Peroxidase/antagonists & inhibitors , Bacterial Proteins , Gene Expression Regulation, Bacterial , Humans , Models, Molecular , Neutrophils/physiology , Phagocytosis , Protein Binding , Protein Conformation , Staphylococcus aureus/metabolism , Up-Regulation
2.
Infect Immun ; 87(12)2019 12.
Article in English | MEDLINE | ID: mdl-31548325

ABSTRACT

The ability of human neutrophils to clear newly attached Staphylococcus aureus bacteria from a serum-coated glass surface was examined in vitro using time-lapse confocal scanning laser microscopy. Quantitative image analysis was used to measure the temporal change in bacterial biomass, neutrophil motility, and fraction of the surface area policed by neutrophils. In control experiments in which the surface was inoculated with bacteria but no neutrophils were added, prolific bacterial growth was observed. Neutrophils were able to control bacterial growth but only consistently when the neutrophil/bacterium number ratio exceeded approximately 1. When preattached bacteria were given a head start and allowed to grow for 3 h prior to neutrophil addition, neutrophils were unable to maintain control of the nascent biofilm. In these head-start experiments, aggregates of bacterial biofilm with areas of 50 µm2 or larger formed, and the growth of such aggregates continued even when multiple neutrophils attacked a cluster. These results suggest a model for the initiation of a biofilm infection in which a delay in neutrophil recruitment to an abiotic surface allows surface-attached bacteria time to grow and form aggregates that become protected from neutrophil clearance. Results from a computational model of the neutrophil-biofilm surface contest supported this conceptual model and highlighted the stochastic nature of the interaction. Additionally, we observed that both neutrophil motility and clearance of bacteria were impaired when oxygen tension was reduced to 0% or 2% O2.


Subject(s)
Biofilms/growth & development , Neutrophils/immunology , Prostheses and Implants/microbiology , Prosthesis-Related Infections/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Anaerobiosis , Computational Biology , Computer Simulation , Humans , Immune Evasion/immunology , Microscopy, Confocal , Prosthesis-Related Infections/microbiology , Staphylococcus aureus/growth & development
3.
J Infect Dis ; 218(5): 809-813, 2018 07 24.
Article in English | MEDLINE | ID: mdl-29668950

ABSTRACT

Staphylococcus aureus is a predominant cause of fatal pneumonia following influenza A virus (IAV) infection. Herein we investigate the influence of antecedent IAV infection on S. aureus virulence gene expression. Using a murine model, comparing the USA300 and USA300ΔsaeR/S strains, we demonstrate that S. aureus pathogenesis following IAV infection is SaeR/S dependent. Furthermore, we show that IAV modulates the lung environment to rapidly up-regulate S. aureus virulence factors containing the SaeR-binding domain. Data demonstrate that the pathogen response to IAV infection impacts host outcome and provides evidence that the ability of S. aureus to sense and respond to the lung environment determines severity of pneumonia.


Subject(s)
Bacterial Proteins/metabolism , Orthomyxoviridae Infections/complications , Pneumonia, Staphylococcal/immunology , Protein Kinases/metabolism , Staphylococcus aureus/pathogenicity , Transcription Factors/metabolism , Animals , Bacterial Proteins/genetics , Disease Models, Animal , Female , Gene Deletion , Male , Mice, Inbred BALB C , Orthomyxoviridae Infections/pathology , Pneumonia, Staphylococcal/genetics , Protein Kinases/genetics , Staphylococcus aureus/genetics , Transcription Factors/genetics
4.
J Infect Dis ; 217(6): 943-952, 2018 03 05.
Article in English | MEDLINE | ID: mdl-29272502

ABSTRACT

Background: The ability of Staphylococcus aureus to evade killing by human neutrophils significantly contributes to disease progression. In this study, we characterize an influential role for the S. aureus SaeR/S 2-component gene regulatory system in suppressing monocyte production of tumor necrosis factor alpha (TNF-α) to subsequently influence human neutrophil priming. Methods: Using flow cytometry and TNF-α specific enzyme-linked immunosorbent assays we identify the primary cellular source of TNF-α in human blood and in purified peripheral blood mononuclear cells (PBMCs) during interaction with USA300 and an isogenic saeR/S deletion mutant (USA300∆saeR/S). Assays with conditioned media from USA300 and USA300∆saeR/S exposed PBMCs were used to investigate priming on neutrophil bactericidal activity. Results: TNF-α production from monocytes was significantly reduced following challenge with USA300 compared to USA300∆saeR/S. We observed that priming of neutrophils using conditioned medium from peripheral blood mononuclear cells stimulated with USA300∆saeR/S significantly increased neutrophil bactericidal activity against USA300 relative to unprimed neutrophils and neutrophils primed with USA300 conditioned medium. The increased neutrophil bactericidal activity was associated with enhanced reactive oxygen species production that was significantly influenced by elevated TNF-α concentrations. Conclusions: Our findings identify an immune evasion strategy used by S. aureus to impede neutrophil priming and subsequent bactericidal activity.


Subject(s)
Bacterial Proteins/pharmacology , Methicillin-Resistant Staphylococcus aureus , Monocytes/metabolism , Neutrophils/immunology , Protein Kinases/pharmacology , Transcription Factors/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Bacterial Proteins/metabolism , Cells, Cultured , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Methicillin-Resistant Staphylococcus aureus/immunology , Monocytes/drug effects , Neutrophils/drug effects , Neutrophils/metabolism , Protein Kinases/metabolism , Transcription Factors/metabolism
5.
Molecules ; 23(7)2018 07 10.
Article in English | MEDLINE | ID: mdl-29996498

ABSTRACT

Essential oils (EOs) were obtained by hydrodistillation of various parts of Ferula ovina (Boiss.) Boiss., Ferula iliensis Krasn. ex. Korovin, and Ferula akitschkensis B. Fedtsch. ex Koso-Pol., collected in the flowering/budding and fruiting stages. Eight samples of EOs isolated from F. ovina and four samples from F. akitsckensis were analyzed by gas chromatography⁻mass spectrometry (GC-MS). The major constituents of F. ovina EOs were α-pinene (6.9⁻47.8%), ß-pinene (1.5⁻7.1%), sabinene (0.1⁻20.5%), ß-phellandrene (0⁻6.5%), trans-verbenol (0.9⁻7.4%), eremophilene (3.1⁻12%), and 6Z-2,5,5,10-tetramethyl-undeca-2,6,9-trien-8-one (0⁻13.7%). The major constituents of F. akitsckensis EOs were α-pinene (0⁻46.2%), ß-pinene (0⁻47.9%), sabinene (0⁻28.3%), eremophilene (0⁻10.6), ß-caryophyllene (0⁻7.5%), himachalen-7-ol (0⁻28.2%), and an himachalol derivative (0⁻8.3%). Samples of EOs from F. ovina, F. iliensis, and F. akitsckensis were evaluated for antibacterial activity against methicillin-resistant Staphylococcus aureus (MRSA) pulse-field gel electrophoresis type USA300 (LAC). EOs from F. ovina exhibited the highest antibacterial activity compared to samples from other Ferula spp., with the most potent EOs being isolated from roots at the flowering and fruiting stages and stems at the fruiting stage (IC50 values of 19.1, 20.9, and 22.9 µg/mL, respectively). Although EOs demonstrated concentration-dependent inhibition of MRSA growth, analysis of the major constituents (α-pinene, ß-pinene, and sabinene) showed that they had low activity, suggesting that other components were likely responsible for the observed bioactivity of the unfractionated EOs. Indeed, correlation of the GC-MS data with antibacterial activity suggested that the putative components responsible for antibacterial activity were, either individually or in combination, eremophilene and trans-verbenol. Overall, these results suggest that the EOs from F. ovina could have potential for use as alternative remedies for the treatment of infectious diseases caused by MRSA.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Ferula/chemistry , Methicillin-Resistant Staphylococcus aureus/drug effects , Oils, Volatile/chemistry , Oils, Volatile/pharmacology , Linear Models , Microbial Sensitivity Tests
6.
Infect Immun ; 85(5)2017 05.
Article in English | MEDLINE | ID: mdl-28264907

ABSTRACT

Studies of the human pathogen group A Streptococcus (GAS) define the carrier phenotype to be an increased ability to adhere to and persist on epithelial surfaces and a decreased ability to cause disease. We tested the hypothesis that a single amino acid change (Arg135Gly) in a highly conserved sensor kinase (LiaS) of a poorly defined GAS regulatory system contributes to a carrier phenotype through increased pilus production. When introduced into an emm serotype-matched invasive strain, the carrier allele (the gene encoding the LiaS protein with an arginine-to-glycine change at position 135 [liaSR135G]) recapitulated a carrier phenotype defined by an increased ability to adhere to mucosal surfaces and a decreased ability to cause disease. Gene transcript analyses revealed that the liaS mutation significantly altered transcription of the genes encoding pilus in the presence of bacitracin. Elimination of pilus production in the isogenic carrier mutant decreased its ability to colonize the mouse nasopharynx and to adhere to and be internalized by cultured human epithelial cells and restored the virulence phenotype in a mouse model of necrotizing fasciitis. We also observed significantly reduced survival of the isogenic carrier mutant compared to that of the parental invasive strain after exposure to human neutrophils. Elimination of pilus in the isogenic carrier mutant increased the level of survival after exposure to human neutrophils to that for the parental invasive strain. Together, our data demonstrate that the carrier mutation (liaSR135G) affects pilus expression. Our data suggest new mechanisms of pilus gene regulation in GAS and that the invasiveness associated with pilus gene regulation in GAS differs from the enhanced invasiveness associated with increased pilus production in other bacterial pathogens.


Subject(s)
Carrier State/microbiology , Fimbriae, Bacterial/genetics , Histidine Kinase/genetics , Mutation, Missense , Organelle Biogenesis , Streptococcal Infections/microbiology , Streptococcus pyogenes/pathogenicity , Animals , Bacterial Adhesion , Cells, Cultured , Epithelial Cells/microbiology , Female , Gene Expression Profiling , Host-Pathogen Interactions , Humans , Mice , Microbial Viability , Nasopharynx/microbiology , Neutrophils/immunology , Neutrophils/microbiology , Streptococcus pyogenes/physiology , Transcription, Genetic
7.
Proc Natl Acad Sci U S A ; 111(19): E2037-45, 2014 May 13.
Article in English | MEDLINE | ID: mdl-24782537

ABSTRACT

Two-component systems (TCSs) are highly conserved across bacteria and are used to rapidly sense and respond to changing environmental conditions. The human pathogen Staphylococcus aureus uses the S. aureus exoprotein expression (sae) TCS to sense host signals and activate transcription of virulence factors essential to pathogenesis. Despite its importance, the mechanism by which the histidine kinase SaeS recognizes specific host stimuli is unknown. After mutagenizing the predicted extracellular loop of SaeS, we discovered one methionine residue (M31) was essential for the ability of S. aureus to transcribe sae target genes, including hla, lukAB/lukGH, and hlgA. This single M31A mutation also significantly reduced cytotoxicity in human neutrophils to levels observed in cells following interaction with ΔsaeS. Another important discovery was that mutation of two aromatic anchor residues (W32A and F33A) disrupted the normal basal signaling of SaeS in the absence of inducing signals, yet both mutant kinases had appropriate activation of effector genes following exposure to neutrophils. Although the transcriptional profile of aromatic mutation W32A was consistent with that of WT in response to human α-defensin 1, mutant kinase F33A did not properly transcribe the γ-toxin genes in response to this stimulus. Taken together, our results provide molecular evidence for how SaeS recognizes host signals and triggers activation of select virulence factors to facilitate evasion of innate immunity. These findings have important implications for signal transduction in prokaryotes and eukaryotes due to conservation of aromatic anchor residues across both of these domains and the important role they play in sensor protein structure and function.


Subject(s)
Neutrophils/microbiology , Protein Kinases/genetics , Protein Kinases/immunology , Staphylococcus aureus/enzymology , Staphylococcus aureus/genetics , Amino Acid Sequence , Bacterial Proteins , Cell Membrane/metabolism , Enzyme Activation , Immunity, Innate/immunology , Molecular Sequence Data , Neutrophils/immunology , Protein Kinases/chemistry , Protein Structure, Tertiary , Signal Transduction/immunology , Staphylococcus aureus/pathogenicity , Virulence
8.
Phytother Res ; 30(8): 1287-97, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27215200

ABSTRACT

Epilobium angustifolium is a medicinal plant used around the world in traditional medicine for the treatment of many disorders and ailments. Experimental studies have demonstrated that Epilobium extracts possess a broad range of pharmacological and therapeutic effects, including antioxidant, anti-proliferative, anti-inflammatory, antibacterial, and anti-aging properties. Flavonoids and ellagitannins, such as oenothein B, are among the compounds considered to be the primary biologically active components in Epilobium extracts. In this review, we focus on the biological properties and the potential clinical usefulness of oenothein B, flavonoids, and other polyphenols derived from E. angustifolium. Understanding the biochemical properties and therapeutic effects of polyphenols present in E. angustifolium extracts will benefit further development of therapeutic treatments from this plant. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Epilobium/chemistry , Plant Extracts/pharmacology , Plants, Medicinal/chemistry , Polyphenols/chemistry , Humans
9.
J Infect Dis ; 212(6): 934-8, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-25722299

ABSTRACT

While Staphylococcus aureus accelerates human neutrophil cell death, the underlying host- and pathogen-derived mechanisms remain incompletely defined. Previous studies demonstrated that the S. aureus SaeR/S sensory system is essential for pathogen survival following neutrophil phagocytosis. Herein, we demonstrate that the SaeR/S system promoted accelerated cell death, suppressed phosphorylation of nuclear factor-κB, and reduced interleukin-8 (IL-8) production in human neutrophils. Treatment of neutrophils with recombinant IL-8 significantly reduced bacterial burden and apoptosis. Our findings demonstrate a mechanism by which S. aureus suppresses the early neutrophil-derived IL-8 response to disrupt cell fate and promote disease.


Subject(s)
Cell Death/physiology , Interleukin-8/metabolism , Neutrophils/physiology , Staphylococcus aureus/physiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cells, Cultured , Gene Expression Regulation/physiology , Humans , Interleukin-8/genetics , NF-kappa B/metabolism , Protein Kinases/genetics , Protein Kinases/metabolism , Transcription Factors
10.
Infect Immun ; 82(12): 5293-307, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25287924

ABSTRACT

The important human pathogen Streptococcus pyogenes (group A Streptococcus [GAS]) produces a hyaluronic acid (HA) capsule that plays critical roles in immune evasion. Previous studies showed that the hasABC operon encoding the capsule biosynthesis enzymes is under the control of a single promoter, P1, which is negatively regulated by the two-component regulatory system CovR/S. In this work, we characterize the sequence upstream of P1 and identify a novel regulatory region controlling transcription of the capsule biosynthesis operon in the M1 serotype strain MGAS2221. This region consists of a promoter, P2, which initiates transcription of a novel small RNA, HasS, an intrinsic transcriptional terminator that inefficiently terminates HasS, permitting read-through transcription of hasABC, and a putative promoter which lies upstream of P2. Electrophoretic mobility shift assays, quantitative reverse transcription-PCR, and transcriptional reporter data identified CovR as a negative regulator of P2. We found that the P1 and P2 promoters are completely repressed by CovR, and capsule expression is regulated by the putative promoter upstream of P2. Deletion of hasS or of the terminator eliminates CovR-binding sequences, relieving repression and increasing read-through, hasA transcription, and capsule production. Sequence analysis of 44 GAS genomes revealed a high level of polymorphism in the HasS sequence region. Most of the HasS variations were located in the terminator sequences, suggesting that this region is under strong selective pressure. We discovered that the terminator deletion mutant is highly resistant to neutrophil-mediated killing and is significantly more virulent in a mouse model of GAS invasive disease than the wild-type strain. Together, these results are consistent with the naturally occurring mutations in this region modulating GAS virulence.


Subject(s)
Bacterial Capsules/metabolism , Hyaluronic Acid/biosynthesis , Operon , Regulatory Elements, Transcriptional , Streptococcus pyogenes/genetics , Transcription, Genetic , Animals , Blood Bactericidal Activity , Disease Models, Animal , Electrophoretic Mobility Shift Assay , Female , Gene Expression Profiling , Genes, Reporter , Genetic Variation , Humans , Immune Evasion , Mice, Inbred BALB C , Real-Time Polymerase Chain Reaction , Sequence Deletion , Streptococcal Infections/immunology , Streptococcal Infections/microbiology , Streptococcal Infections/pathology , Virulence
11.
Infect Immun ; 81(4): 1316-24, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23381999

ABSTRACT

Several prominent bacterial pathogens secrete nuclease (Nuc) enzymes that have an important role in combating the host immune response. Early studies of Staphylococcus aureus Nuc attributed its regulation to the agr quorum-sensing system. However, recent microarray data have indicated that nuc is under the control of the SaeRS two-component system, which is a major regulator of S. aureus virulence determinants. Here we report that the nuc gene is directly controlled by the SaeRS two-component system through reporter fusion, immunoblotting, Nuc activity measurements, promoter mapping, and binding studies, and additionally, we were unable identify a notable regulatory link to the agr system. The observed SaeRS-dependent regulation was conserved across a wide spectrum of representative S. aureus isolates. Moreover, with community-associated methicillin-resistant S. aureus (CA MRSA) in a mouse model of peritonitis, we observed in vivo expression of Nuc activity in an SaeRS-dependent manner and determined that Nuc is a virulence factor that is important for in vivo survival, confirming the enzyme's role as a contributor to invasive disease. Finally, natural polymorphisms were identified in the SaeRS proteins, one of which was linked to Nuc regulation in a CA MRSA USA300 endocarditis isolate. Altogether, our findings demonstrate that Nuc is an important S. aureus virulence factor and part of the SaeRS regulon.


Subject(s)
Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Micrococcal Nuclease/biosynthesis , Protein Kinases/metabolism , Staphylococcus aureus/pathogenicity , Virulence Factors/biosynthesis , Animals , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred BALB C , Microbial Viability , Peritonitis/microbiology , Peritonitis/pathology , Regulon , Staphylococcus aureus/genetics , Transcription Factors
12.
Antimicrob Agents Chemother ; 57(1): 241-7, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23114775

ABSTRACT

Methicillin-resistant Staphylococcus aureus (MRSA) has become a major source of infection in hospitals and in the community. Increasing antibiotic resistance in S. aureus strains has created a need for alternative therapies to treat disease. A component of the licorice root Glycyrrhiza spp., 18ß-glycyrrhetinic acid (GRA), has been shown to have antiviral, antitumor, and antibacterial activity. This investigation explores the in vitro and in vivo effects of GRA on MRSA pulsed-field gel electrophoresis (PFGE) type USA300. GRA exhibited bactericidal activity at concentrations exceeding 0.223 µM. Upon exposure of S. aureus to sublytic concentrations of GRA, we observed a reduction in expression of key virulence genes, including saeR and hla. In murine models of skin and soft tissue infection, topical GRA treatment significantly reduced skin lesion size and decreased the expression of saeR and hla genes. Our investigation demonstrates that at high concentrations GRA is bactericidal to MRSA and at sublethal doses it reduces virulence gene expression in S. aureus both in vitro and in vivo.


Subject(s)
Anti-Bacterial Agents/pharmacology , Gene Expression Regulation, Bacterial/drug effects , Glycyrrhetinic Acid/analogs & derivatives , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Soft Tissue Infections/drug therapy , Staphylococcal Skin Infections/drug therapy , Animals , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Female , Glycyrrhetinic Acid/pharmacology , Hemolysin Proteins/antagonists & inhibitors , Hemolysin Proteins/genetics , Hemolysin Proteins/metabolism , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/growth & development , Mice , Microbial Sensitivity Tests , Skin/drug effects , Skin/microbiology , Skin/pathology , Soft Tissue Infections/microbiology , Soft Tissue Infections/pathology , Staphylococcal Skin Infections/microbiology , Staphylococcal Skin Infections/pathology , Transcription Factors , Virulence
13.
J Bacteriol ; 194(16): 4355-65, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22685286

ABSTRACT

Staphylococcus aureus is a significant human pathogen that is capable of infecting a wide range of host tissues. This bacterium is able to evade the host immune response by utilizing a repertoire of virulence factors. These factors are tightly regulated by various two-component systems (TCS) and transcription factors. Previous studies have suggested that transcriptional regulation of a subset of immunomodulators, known as the staphylococcal superantigen-like proteins (Ssls), is mediated by the master regulators accessory gene regulator (Agr) TCS, S. aureus exoprotein expression (Sae) TCS, and Rot. Here we demonstrate that Rot and SaeR, the response regulator of the Sae TCS, synergize to coordinate the activation of the ssl promoters. We have determined that both transcription factors are required, but that neither is sufficient, for promoter activation. This regulatory scheme is mediated by direct binding of both transcription factors to the ssl promoters. We also demonstrate that clinically relevant methicillin-resistant S. aureus (MRSA) strains respond to neutrophils via the Sae TCS to upregulate the expression of ssls. Until now, Rot and the Sae TCS have been proposed to work in opposition of one another on their target genes. This is the first example of these two regulators working in concert to activate promoters.


Subject(s)
Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Protein Kinases/metabolism , Repressor Proteins/metabolism , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Superantigens/biosynthesis , Humans , Transcription Factors
14.
Mol Microbiol ; 79(3): 814-25, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21255120

ABSTRACT

Staphylococcus aureus is an important pathogen that continues to be a significant global health threat because of the prevalence of methicillin-resistant S. aureus strains (MRSA). The pathogenesis of this organism is partly attributed to the production of a large repertoire of cytotoxins that target and kill innate immune cells, which provide the first line of defence against S. aureus infection. Here we demonstrate that leukocidin A/B (LukAB) is required and sufficient for the ability of S. aureus, including MRSA, to kill human neutrophils, macrophages and dendritic cells. LukAB targets the plasma membrane of host cells resulting in cellular swelling and subsequent cell death. We found that S. aureus lacking lukAB are severely impaired in their ability to kill phagocytes during bacteria-phagocyte interaction, which in turn renders the lukAB-negative staphylococci more susceptible to killing by neutrophils. Notably, we show that lukAB is expressed in vivo within abscesses in a murine infection model and that it contributes significantly to pathogenesis of MRSA in an animal host. Collectively, these results extend our understanding of how S. aureus avoids phagocyte-mediated clearance, and underscore LukAB as an important factor that contributes to staphylococcal pathogenesis.


Subject(s)
Cytotoxins/metabolism , Staphylococcus aureus/pathogenicity , Cell Death , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cytoprotection , HL-60 Cells , Humans , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Phagocytes/cytology , Phagocytes/metabolism , Phagocytes/ultrastructure
15.
Infect Immun ; 79(6): 2404-11, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21422178

ABSTRACT

Streptococcus pyogenes is an important human pathogen with an expansive repertoire of verified and putative virulence factors. Here we demonstrate that a mutant deficient in the production of the streptococcal ADP-ribosyltransferase SpyA generates lesions of reduced size in a subcutaneous mouse infection model. At early stages of infection, when the difference in lesion size is first established, inflamed tissue isolated from lesions of mice infected with spyA mutant bacteria has higher levels of mRNA encoding the chemokines CXCL1 and CCL2 than does tissue isolated from mice infected with wild-type bacteria. In addition, at these early times, the mRNA levels for the gene encoding the intermediate filament vimentin are higher in the mutant-infected tissue. As wound resolution progresses, mRNA levels of the gene encoding matrix metallopeptidase 2 are lower in mutant-infected tissue. Furthermore, we demonstrate that the spyA mutant is internalized more efficiently than wild-type bacteria by HeLa cells. We conclude that SpyA contributes to streptococcal pathogenesis in the mouse subcutaneous infection model. Our observations suggest that the presence of SpyA delays wound healing in this model.


Subject(s)
ADP Ribose Transferases/physiology , Streptococcal Infections/microbiology , Streptococcus pyogenes/pathogenicity , Virulence Factors/physiology , ADP Ribose Transferases/metabolism , Animals , Blotting, Western , Chemokine CCL2/physiology , Chemokine CXCL1/physiology , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Female , HeLa Cells , Humans , Mice , Neutrophils/physiology , Reverse Transcriptase Polymerase Chain Reaction , Streptococcus pyogenes/enzymology , Virulence Factors/metabolism
16.
J Infect Dis ; 201(2): 241-54, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20001858

ABSTRACT

This investigation examines the role of the SaeR/S 2-component system in USA300, a prominent circulating clone of community-associated methicillin-resistant Staphylococcus aureus. Using a saeR/S isogenic deletion mutant of USA300 (USA300DeltasaeR/S) in murine models of sepsis and soft-tissue infection revealed that this sensory system is critical to pathogenesis of USA300 during both superficial and invasive infection. Oligonucleotide microarray and real-time reverse-transcriptase polymerase chain reaction identified numerous extracellular virulence genes that are down-regulated in USA300DeltasaeR/S. Unexpectedly, an up-regulation of mecA and mecR1 corresponded to increased methicillin resistance in USA300DeltasaeR/S. 5'-RACE analysis defined transcript start sites for sbi, efb, mecA, lukS-PV, hlb, SAUSA300_1975, and hla, to underscore a conserved consensus sequence within promoter regions of genes under strong SaeR/S transcriptional regulation. Electrophoretic mobility shift assay experiments illustrated direct binding of SaeR(His) to promoter regions containing the conserved consensus sequence. Collectively, the findings of this investigation demonstrate that SaeR/S directly interacts with virulence gene promoters to significantly influence USA300 pathogenesis.


Subject(s)
Bacterial Proteins/genetics , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Promoter Regions, Genetic/genetics , Virulence Factors/metabolism , Animals , Bacterial Proteins/physiology , Community-Acquired Infections/microbiology , Disease Models, Animal , Electrophoretic Mobility Shift Assay , Gene Expression Profiling , Humans , Mice , Oligonucleotide Array Sequence Analysis , Sequence Deletion , Soft Tissue Infections/microbiology , Transcription Factors , Up-Regulation
17.
Front Microbiol ; 12: 785182, 2021.
Article in English | MEDLINE | ID: mdl-35095798

ABSTRACT

The goal of this study was to quantify the variability of confocal laser scanning microscopy (CLSM) time-lapse images of early colonizing biofilms to aid in the design of future imaging experiments. To accomplish this a large imaging dataset consisting of 16 independent CLSM microscopy experiments was leveraged. These experiments were designed to study interactions between human neutrophils and single cells or aggregates of Staphylococcus aureus (S. aureus) during the initial stages of biofilm formation. Results suggest that in untreated control experiments, variability differed substantially between growth phases (i.e., lag or exponential). When studying the effect of an antimicrobial treatment (in this case, neutrophil challenge), regardless of the inoculation level or of growth phase, variability changed as a frown-shaped function of treatment efficacy (i.e., the reduction in biofilm surface coverage). These findings were used to predict the best experimental designs for future imaging studies of early biofilms by considering differing (i) numbers of independent experiments; (ii) numbers of fields of view (FOV) per experiment; and (iii) frame capture rates per hour. A spreadsheet capable of assessing any user-specified design is included that requires the expected mean log reduction and variance components from user-generated experimental results. The methodology outlined in this study can assist researchers in designing their CLSM studies of antimicrobial treatments with a high level of statistical confidence.

18.
Microbiol Spectr ; 9(2): e0088821, 2021 10 31.
Article in English | MEDLINE | ID: mdl-34704790

ABSTRACT

Staphylococcus aureus is an important human pathogen that can cause a variety of diseases ranging from mild superficial skin infections to life-threatening conditions like necrotizing pneumonia, endocarditis, and septicemia. Polymorphonuclear leukocytes (PMNs; neutrophils in particular herein) are essential for host defense against S. aureus infections, and the microbe is phagocytosed readily. Most ingested bacteria are killed, but some S. aureus strains-such as the epidemic USA300 strain-have an enhanced ability to cause PMN lysis after phagocytosis. Although progress has been made, the mechanism for lysis after phagocytosis of S. aureus remains incompletely determined. Here, we tested the hypothesis that disruption of phagosome integrity and escape of S. aureus from the PMN phagosome into the cytoplasm precedes PMN lysis. We used USA300 wild-type and isogenic deletion strains to evaluate and/or verify the role of selected S. aureus molecules in this cytolytic process. Compared to the wild-type USA300 strain, Δagr, Δhla, ΔlukGH, and Δpsm strains each caused significantly less lysis of human PMNs 3 h and/or 6 h after phagocytosis, consistent with previous studies. Most notably, confocal microscopy coupled with selective permeabilization assays demonstrated that phagosome membrane integrity is largely maintained prior to PMN lysis after S. aureus phagocytosis. We conclude that PMN lysis does not require escape of S. aureus from the phagosome to the cytoplasm and that these are independent phenomena. The findings are consistent with the ability of S. aureus (via selected molecules) to trigger lysis of human PMNs by an undetermined signaling mechanism. IMPORTANCE S. aureus strain USA300 has the ability to cause rapid lysis of human neutrophils after phagocytosis. Although this phenomenon likely contributes to the success of USA300 as a human pathogen, our knowledge of the mechanism remains incomplete. Here, we used a selective permeabilization assay coupled with confocal microscopy to demonstrate that USA300 is contained within human neutrophil phagosomes until the point of host cell lysis. Thus, consistent with a process in macrophages, S. aureus fails to escape into the neutrophil cytoplasm prior to cytolysis.


Subject(s)
Cell Death/physiology , Neutrophils/immunology , Neutrophils/microbiology , Phagosomes/microbiology , Staphylococcus aureus/immunology , Humans , Phagocytosis/immunology , Signal Transduction/immunology , Staphylococcal Infections/immunology
19.
Biomaterials ; 275: 120775, 2021 08.
Article in English | MEDLINE | ID: mdl-34243039

ABSTRACT

Biofilms that form on implanted medical devices cause recalcitrant infections. The early events enabling contaminating bacteria to evade immune clearance, before a mature biofilm is established, are poorly understood. Live imaging in vitro demonstrated that Staphylococcus aureus sparsely inoculated on an abiotic surface can go undiscovered by human neutrophils, grow, and form aggregates. Small (~50 µm2) aggregates of attached bacteria resisted killing by human neutrophils, resulting in neutrophil lysis and bacterial persistence. In vivo, neutrophil recruitment to a peritoneal implant was spatially heterogenous, with some bacterial aggregates remaining undiscovered by neutrophils after 24 h. Intravital imaging in mouse skin revealed that attached S. aureus aggregates grew and remained undiscovered by neutrophils for up to 3 h. These results suggest a model in which delayed recruitment of neutrophils to an abiotic implant presents a critical window in which bacteria establish a nascent biofilm and acquire tolerance to neutrophil killing.


Subject(s)
Staphylococcal Infections , Staphylococcus aureus , Animals , Biofilms , Immune Evasion , Mice , Neutrophil Infiltration , Neutrophils
20.
N Engl J Med ; 357(16): 1608-19, 2007 Oct 18.
Article in English | MEDLINE | ID: mdl-17881745

ABSTRACT

BACKGROUND: The hyper-IgE syndrome (or Job's syndrome) is a rare disorder of immunity and connective tissue characterized by dermatitis, boils, cyst-forming pneumonias, elevated serum IgE levels, retained primary dentition, and bone abnormalities. Inheritance is autosomal dominant; sporadic cases are also found. METHODS: We collected longitudinal clinical data on patients with the hyper-IgE syndrome and their families and assayed the levels of cytokines secreted by stimulated leukocytes and the gene expression in resting and stimulated cells. These data implicated the signal transducer and activator of transcription 3 gene (STAT3) as a candidate gene, which we then sequenced. RESULTS: We found increased levels of proinflammatory gene transcripts in unstimulated peripheral-blood neutrophils and mononuclear cells from patients with the hyper-IgE syndrome, as compared with levels in control cells. In vitro cultures of mononuclear cells from patients that were stimulated with lipopolysaccharide, with or without interferon-gamma, had higher tumor necrosis factor alpha levels than did identically treated cells from unaffected persons (P=0.003). In contrast, the cells from patients with the hyper-IgE syndrome generated lower levels of monocyte chemoattractant protein 1 in response to the presence of interleukin-6 (P=0.03), suggesting a defect in interleukin-6 signaling through its downstream mediators, one of which is STAT3. We identified missense mutations and single-codon in-frame deletions in STAT3 in 50 familial and sporadic cases of the hyper-IgE syndrome. Eighteen discrete mutations, five of which were hot spots, were predicted to directly affect the DNA-binding and SRC homology 2 (SH2) domains. CONCLUSIONS: Mutations in STAT3 underlie sporadic and dominant forms of the hyper-IgE syndrome, an immunodeficiency syndrome involving increased innate immune response, recurrent infections, and complex somatic features.


Subject(s)
Job Syndrome/genetics , Mutation, Missense , STAT3 Transcription Factor/genetics , Sequence Deletion , Adult , Aged , Aged, 80 and over , Cytokines/blood , Female , Gene Expression Profiling , Humans , Interleukin-6/physiology , Leukocytes/immunology , Male , Oligonucleotide Array Sequence Analysis , Pedigree , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL