Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
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
2.
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
3.
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
4.
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
5.
Biochemistry ; 49(13): 2834-42, 2010 Apr 06.
Article in English | MEDLINE | ID: mdl-20180543

ABSTRACT

The heme-binding proteins Shp and HtsA of Streptococcus pyogenes are part of the heme acquisition machinery in which Shp directly transfers its heme to HtsA. Mutagenesis and spectroscopic analyses were performed to identify the heme axial ligands in HtsA and to characterize axial mutants of HtsA. Replacements of the M79 and H229 residues, not the other methionine and histidine residues, with alanine convert UV-vis spectra of HtsA with a low-spin, hexacoordinate heme iron into spectra of high-spin heme complexes. Ferrous M79A and H229A HtsA mutants possess magnetic circular dichroism (MCD) spectra that are similar with those of proteins with pentacoordinate heme iron. Ferric M79A HtsA displays UV-vis, MCD, and resonance Raman (RR) spectra that are typical of a hexacoordinate heme iron with histidine and water ligands. In contrast, ferric H229A HtsA has UV-vis, MCD, and RR spectra that represent a pentacoordinate heme iron complex with a methionine axial ligand. Imidazole readily forms a low-spin hexacoordinate adduct with M79A HtsA with a K(d) of 40.9 muM but not with H229A HtsA, and cyanide binds to M79A and H229A with K(d) of 0.5 and 19.1 microM, respectively. The ferrous mutants rapidly bind CO and form simple CO complexes. These results establish the H229 and M79 residues as the axial ligands of the HtsA heme iron, indicate that the M79 side is more accessible to the solvent than the H229 side of the bound heme in HtsA, and provide unique spectral features for a protein with pentacoordinate, methionine-ligated heme iron. These findings will facilitate elucidation of the molecular mechanism and structural basis for rapid and direct heme transfer from Shp to HtsA.


Subject(s)
Carrier Proteins/chemistry , Heme/metabolism , Hemeproteins/chemistry , Streptococcus pyogenes/chemistry , Bacterial Proteins , Carbon Monoxide/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Heme-Binding Proteins , Hemeproteins/genetics , Hemeproteins/metabolism , Ligands , Mutation, Missense , Spectrum Analysis
6.
J Vis Exp ; (155)2020 01 03.
Article in English | MEDLINE | ID: mdl-31957747

ABSTRACT

Staphylococcus aureus is capable of secreting a wide range of leukocidins that target and disrupt the membrane integrity of polymorphonuclear leukocytes (PMNs or neutrophils). This protocol describes both the purification of human PMNs and the quantification of S. aureus cytotoxicity against PMNs in three different sections. Section 1 details the isolation of PMNs and serum from human blood using density centrifugation. Section 2 tests the cytotoxicity of extracellular proteins produced by S. aureus against these purified human PMNs. Section 3 measures the cytotoxicity against human PMNs following the phagocytosis of live S. aureus. These procedures measure disruption of PMN plasma membrane integrity by S. aureus leukocidins using flow cytometry analysis of PMNs treated with propidium iodide, a DNA binding fluorophore that is cell membrane impermeable. Collectively, these methods have the advantage of rapidly testing S. aureus cytotoxicity against primary human PMNs and can be easily adapted to study other aspects of host-pathogen interactions.


Subject(s)
Neutrophils/cytology , Neutrophils/microbiology , Staphylococcus aureus/physiology , Bacterial Proteins/metabolism , Cell Death , Cell Separation , Flow Cytometry , Humans , Phagocytosis , Propidium/metabolism
7.
Front Microbiol ; 11: 561, 2020.
Article in English | MEDLINE | ID: mdl-32390958

ABSTRACT

Staphylococcus aureus (S. aureus) causes a range of diseases ranging from superficial skin and soft-tissue infections to invasive and life-threatening conditions (Klevens et al., 2007; Kobayashi et al., 2015). S. aureus utilizes the Sae sensory system to adapt to neutrophil challenge. Although the roles of the SaeR response regulator and its cognate sensor kinase SaeS have been demonstrated to be critical for surviving neutrophil interaction and for causing infection, the roles for the accessory proteins SaeP and SaeQ remain incompletely defined. To characterize the functional role of these proteins during innate immune interaction, we generated isogenic deletion mutants lacking these accessory genes in USA300 (USA300ΔsaeP and USA300ΔsaeQ). S. aureus survival was increased following phagocytosis of USA300ΔsaeP compared to USA300 by neutrophils. Additionally, secreted extracellular proteins produced by USA300ΔsaeP cells caused significantly more plasma membrane damage to human neutrophils than extracellular proteins produced by USA300 cells. Deletion of saeQ resulted in a similar phenotype, but effects did not reach significance during neutrophil interaction. The enhanced cytotoxicity of USA300ΔsaeP cells toward human neutrophils correlated with an increased expression of bi-component leukocidins known to target these immune cells. A saeP and saeQ double mutant (USA300ΔsaePQ) showed a significant increase in survival following neutrophil phagocytosis that was comparable to the USA300ΔsaeP single mutant and increased the virulence of USA300 during murine bacteremia. These data provide evidence that SaeP modulates the Sae-mediated response of S. aureus against human neutrophils and suggest that saeP and saeQ together impact pathogenesis in vivo.

8.
Front Microbiol ; 9: 3085, 2018.
Article in English | MEDLINE | ID: mdl-30619166

ABSTRACT

Staphylococcus aureus is a common Gram-positive bacteria that is a major cause of human morbidity and mortality. The SaeR/S two-component sensory system of S. aureus is important for virulence gene transcription and pathogenesis. However, the influence of SaeR phosphorylation on virulence gene transcription is not clear. To determine the importance of potential SaeR phosphorylation sites for S. aureus virulence, we generated genomic alanine substitutions at conserved aspartic acid residues in the receiver domain of the SaeR response regulator in clinically significant S. aureus pulsed-field gel electrophoresis (PFGE) type USA300. Transcriptional analysis demonstrated a dramatic reduction in the transcript abundance of various toxins, adhesins, and immunomodulatory proteins for SaeR with an aspartic acid to alanine substitution at residue 51. These findings corresponded to a significant decrease in cytotoxicity against human erythrocytes and polymorphonuclear leukocytes, the ability to block human myeloperoxidase activity, and pathogenesis during murine soft-tissue infection. Analysis of SaeR sequences from over 8,000 draft S. aureus genomes revealed that aspartic acid residue 51 is 100% conserved. Collectively, these results demonstrate that aspartic acid residue 51 of SaeR is essential for S. aureus virulence and underscore a conserved target for novel antimicrobial strategies that treat infection caused by this pathogen.

9.
BMC Microbiol ; 6: 82, 2006 Sep 28.
Article in English | MEDLINE | ID: mdl-17007644

ABSTRACT

BACKGROUND: Heme is a preferred iron source of bacterial pathogens. Streptococcus equi subspecies equi is a bacterial pathogen that causes strangles in horses. Whether S. equi has a heme acquisition transporter is unknown. RESULTS: An S. equi genome database was blasted with the heme binding proteins Shp and HtsA of Streptococcus pyogenes, and found that S. equi has the homologue of Shp (designated SeShp) and HtsA (designated SeHtsA). Tag-free recombinant SeShp and SeHtsA and 6xHis-tagged SeHtsA (SeHtsAHis) were prepared and characterized. Purified holoSeShp and holoSeHtsA bind Fe(II)-protoporphyrin IX (heme) and Fe(III)-protoporphyrin IX (hemin) in a 1:1 stoichiometry, respectively, and are designated hemoSeShp and hemiSeHtsA. HemiSeShp and hemiSeHtsAHis can be reconstituted from apoSeShp and apoSeHtsAHis and hemin. HemoSeShp is stable in air and can be oxidized to hemiSeShp by ferricyanide. HemiSeHtsA can be reduced into hemoSeHtsA, which autoxidizes readily. HemoSeShp rapidly transfers its heme to apoSeHtsAHis. In addition, hemoSeShp can also transfer its heme to apoHtsA, and hemoShp is able to donate heme to apoSeHtsAHis. CONCLUSION: The primary structures, optical properties, oxidative stability, and in vitro heme transfer reaction of SeShp and SeHtsA are very similar to those of S. pyogenes Shp and HtsA. The data suggest that the putative cell surface protein SeShp and lipoprotein SeHtsA are part of the machinery to acquire heme in S. equi. The results also imply that the structure, function, and functional mechanism of the heme acquisition machinery are conserved in S. equi and S. pyogenes.


Subject(s)
Bacterial Proteins/metabolism , Carrier Proteins/metabolism , Heme/metabolism , Hemeproteins/metabolism , Streptococcus equi/metabolism , Amino Acid Sequence , Bacterial Proteins/chemistry , Carrier Proteins/chemistry , Gene Expression Regulation, Bacterial , Genome, Bacterial , Heme-Binding Proteins , Hemeproteins/chemistry , Molecular Sequence Data , Protein Binding , Streptococcus equi/classification
10.
PLoS One ; 11(10): e0164410, 2016.
Article in English | MEDLINE | ID: mdl-27711145

ABSTRACT

Staphylococcus aureus is a leading cause of human infections worldwide. The pathogen produces numerous molecules that can interfere with recognition and binding by host innate immune cells, an initial step required for the ingestion and subsequent destruction of microbes by phagocytes. To better understand the interaction of this pathogen with human immune cells, we compared the association of S. aureus and S. epidermidis with leukocytes in human blood. We found that a significantly greater proportion of B cells associated with S. epidermidis relative to S. aureus. Complement components and complement receptors were important for the binding of B cells with S. epidermidis. Experiments using staphylococci inactivated by ultraviolet radiation and S. aureus isogenic deletion mutants indicated that S. aureus secretes molecules regulated by the SaeR/S two-component system that interfere with the ability of human B cells to bind this bacterium. We hypothesize that the relative inability of B cells to bind S. aureus contributes to the microbe's success as a human pathogen.


Subject(s)
B-Lymphocytes/metabolism , Staphylococcus aureus/metabolism , Staphylococcus epidermidis/metabolism , B-Lymphocytes/cytology , Complement System Proteins/metabolism , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Microscopy, Fluorescence , Phagocytosis/physiology , Protein Binding , Staphylococcus aureus/genetics , Staphylococcus aureus/radiation effects , Staphylococcus epidermidis/genetics , Staphylococcus epidermidis/radiation effects , Ultraviolet Rays
11.
PLoS One ; 10(9): e0138084, 2015.
Article in English | MEDLINE | ID: mdl-26359669

ABSTRACT

In addition to the well characterized function of chemokines in mediating the homing and accumulation of leukocytes to tissues, some chemokines also exhibit potent antimicrobial activity. Little is known of the potential role of chemokines in bovine mammary gland health and disease. The chemokine CCL28 has previously been shown to play a key role in the homing and accumulation of IgA antibody secreting cells to the lactating murine mammary gland. CCL28 has also been shown to act as an antimicrobial peptide with activity demonstrated against a wide range of pathogens including bacteria, fungi and protozoans. Here we describe the cloning and function of bovine CCL28 and document the concentration of this chemokine in bovine milk. Bovine CCL28 was shown to mediate cellular chemotaxis via the CCR10 chemokine receptor and exhibited antimicrobial activity against a variety of bovine mastitis causing organisms. The concentration of bovine CCL28 in milk was found to be highly correlated with the lactation cycle. Highest concentrations of CCL28 were observed soon after parturition, with levels decreasing over time. These results suggest a potential role for CCL28 in the prevention/resolution of bovine mastitis.


Subject(s)
Anti-Bacterial Agents/pharmacology , Chemokines, CC/metabolism , Mastitis, Bovine/microbiology , Milk/immunology , Receptors, CCR10/metabolism , Animals , Anti-Bacterial Agents/metabolism , Bacteria/drug effects , COS Cells , Cattle , Chemokines, CC/genetics , Chemokines, CC/pharmacology , Chemotaxis , Chlorocebus aethiops , Cloning, Molecular , Female , Gene Expression Regulation , Mastitis, Bovine/immunology
12.
J Innate Immun ; 6(1): 21-30, 2014.
Article in English | MEDLINE | ID: mdl-23816635

ABSTRACT

The ability of Staphylococcus aureus to infect tissues is dependent on precise control of virulence through gene-regulatory systems. While the SaeR/S two-component system has been shown to be a major regulator of S. aureus virulence, the influence of the host environment on SaeR/S-regulated genes (saeR/S targets) remains incompletely defined. Using QuantiGene 2.0 transcriptional assays, we examined expression of genes with the SaeR binding site in USA300 exposed to human and mouse neutrophils and host-derived peptides and during subcutaneous skin infection. We found that only some of the saeR/S targets, as opposed to the entire SaeR/S virulon, were activated within 5 and 10 min of interacting with human neutrophils as well as α-defensin. Furthermore, mouse neutrophils promoted transcription of saeR/S targets despite lacking α-defensin, and the murine skin environment elicited a distinctive expression profile of saeR/S targets. These findings indicate that saeR/S-mediated transcription is unique to and dependent on specific host stimuli. By using isogenic USA300ΔsaeR/S and USA300Δagr knockout strains, we also determined that SaeR/S is the major regulator of virulence factors, while Agr, a quorum-sensing two-component system, has moderate influence on transcription of the saeR/S targets under the tested physiological conditions.


Subject(s)
Bacterial Proteins/metabolism , Neutrophils/immunology , Skin/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Animals , Gene Expression Regulation, Bacterial , Gene Knockout Techniques , Host Specificity , Host-Pathogen Interactions , Humans , Immunity, Innate , Mice , Microarray Analysis , Neutrophils/microbiology , Skin/microbiology , Staphylococcus aureus/pathogenicity , Trans-Activators , Transcription Factors , Transcriptome , Virulence/genetics , alpha-Defensins/metabolism
13.
J Leukoc Biol ; 94(5): 971-9, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24026286

ABSTRACT

This investigation examines the influence of α-toxin (Hla) expression by CA-MRSA on host immune cell integrity and cytokine expression during infection of human blood. Flow cytometry analysis of human blood infected by Staphylococcus aureus PFGE type USA300 or a USA300Δhla demonstrated that Hla expression significantly increased plasma membrane permeability of human CD14(+) monocytes. The increased susceptibility of human CD14(+) monocytes to Hla toxicity paralleled the high cell-surface expression on these cell types of ADAM10. USA300 rapidly associated with PMNs and monocytes but not T cells following inoculation of human blood. Transcription analysis indicated a strong up-regulation of proinflammatory cytokine transcription following infection of human blood by USA300 and USA300Δhla. CBAs and ELISAs determined that IL-6, IL-10, TNF-α, IFN-γ, IL-1ß, IL-8, and IL-4 are significantly up-regulated during the initial phases of human blood infection by USA300 relative to mock-infected blood but failed to distinguish any significant differences in secreted cytokine protein concentrations during infection by USA300Δhla relative to USA300. Collectively, these findings demonstrate that expression of Hla by USA300 has a significant impact on human CD14(+) monocyte plasma membrane integrity but is not exclusively responsible for the proinflammatory cytokine profile induced by USA300 during the initial stages of human blood infection.


Subject(s)
Bacteremia/immunology , Bacterial Toxins/pharmacology , Cell Membrane Permeability/drug effects , Cytokines/genetics , Hemolysin Proteins/pharmacology , Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections/immunology , ADAM Proteins/blood , ADAM10 Protein , Amyloid Precursor Protein Secretases/blood , Humans , Lipopolysaccharide Receptors/blood , Membrane Proteins/blood
14.
PLoS One ; 7(5): e36532, 2012.
Article in English | MEDLINE | ID: mdl-22574180

ABSTRACT

This investigation examines the influence of alpha-toxin (Hla) during USA300 infection of human leukocytes. Survival of an USA300 isogenic deletion mutant of hla (USA300Δhla) in human blood was comparable to the parental wild-type strain and polymorphonuclear leukocyte (PMN) plasma membrane permeability caused by USA300 did not require Hla. Flow cytometry analysis of peripheral blood mononuclear cells (PBMCs) following infection by USA300, USA300Δhla, and USA300Δhla transformed with a plasmid over-expressing Hla (USA300Δhla Comp) demonstrated this toxin plays a significant role inducing plasma membrane permeability of CD14(+), CD3(+), and CD19(+) PBMCs. Rapid plasma membrane permeability independent of Hla was observed for PMNs, CD14(+) and CD19(+) PBMCs following intoxication with USA300 supernatant while the majority of CD3(+) PBMC plasma membrane permeability induced by USA300 required Hla. Addition of recombinant Hla to USA300Δhla supernatant rescued CD3(+) and CD19(+) PBMC plasma membrane permeability generated by USA300 supernatant. An observed delay in plasma membrane permeability caused by Hla in conjunction with Annexin V binding and ApoBrdU Tunel assays examining PBMCs intoxicated with recombinant Hla or infected with USA300, USA300Δhla, USA300Δhla Comp, and USA300ΔsaeR/S suggest Hla induces programmed cell death of monocytes, B cells, and T cells that results in plasma membrane permeability. Together these findings underscore the importance of Hla during S. aureus infection of human tissue and specifically demonstrate Hla activity during USA300 infection triggers programmed cell death of human monocytes, T cells and B cells that leads to plasma membrane permeability.


Subject(s)
Apoptosis/drug effects , Bacterial Toxins/toxicity , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Methicillin-Resistant Staphylococcus aureus/physiology , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , B-Lymphocytes/microbiology , Bacterial Toxins/genetics , Cell Line , Cell Membrane Permeability/drug effects , Culture Media, Conditioned/metabolism , Humans , Leukocytes, Mononuclear/microbiology , Monocytes/cytology , Monocytes/drug effects , Monocytes/microbiology , Neutrophils/cytology , Neutrophils/drug effects , Neutrophils/microbiology , Recombinant Proteins/genetics , Recombinant Proteins/toxicity , Sequence Deletion , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/microbiology
15.
J Infect Dis ; 199(11): 1698-706, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19374556

ABSTRACT

Methicillin-resistant Staphylococcus aureus is problematic both in hospitals and in the community. Currently, we have limited understanding of mechanisms of innate immune evasion used by S. aureus. To that end, we created an isogenic deletion mutant in strain MW2 (USA400) of the saeR/S 2-component gene regulatory system and studied its role in mouse models of pathogenesis and during human neutrophil interaction. In this study, we demonstrate that saeR/S plays a distinct role in S. aureus pathogenesis and is vital for virulence of MW2 in a mouse model of sepsis. Moreover, deletion of saeR/S significantly impaired survival of MW2 in human blood and after neutrophil phagocytosis. Microarray analysis revealed that SaeR/S of MW2 influences expression of a wide variety of genes with diverse biological functions. These data provide new insight into how virulence is regulated in S. aureus and associates a specific staphylococcal gene-regulatory system with invasive staphylococcal disease.


Subject(s)
Bacterial Proteins/genetics , Immunity, Innate/genetics , Protein Kinases/genetics , Staphylococcal Infections/immunology , Staphylococcus aureus/genetics , Staphylococcus aureus/immunology , Animals , Disease Models, Animal , Mice , Mutagenesis , Neutrophils/microbiology , Neutrophils/physiology , Phagocytosis , Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sepsis/microbiology , Sequence Deletion , Staphylococcus aureus/isolation & purification , Staphylococcus aureus/pathogenicity , Transcription Factors , Virulence
16.
Curr Opin Infect Dis ; 21(2): 147-52, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18317037

ABSTRACT

PURPOSE OF REVIEW: In recent years there has been an increase in the incidence of community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA) infections in healthy individuals, the cause of which is largely unknown. CA-MRSA primarily causes skin and soft-tissue infections but certain strains are also associated with unusually severe pathology. The purpose of this review is to provide a critical analysis of our current knowledge of virulence factors contributing to skin and soft-tissue infections caused by CA-MRSA. RECENT FINDINGS: Isolates classified as pulsed-field gel electrophoresis type USA300 have emerged as the predominant CA-MRSA genotype and in most geographic areas account for 97% or more of CA-MRSA infections. Recent key studies, such as those reporting the complete genome sequence of USA300, and the discovery of cytolytic peptides that contribute significantly to CA-MRSA virulence, lead the way for future investigations. SUMMARY: Although we have only a cursory understanding of the molecular mechanisms of CA-MRSA virulence, studies using clinically relevant CA-MRSA isolates are beginning to identify virulence determinants specific to this pathogen. Identifying CA-MRSA virulence determinants and the concerted regulation of these factors will foster development of vaccines and therapeutics designed to control CA-MRSA skin infections.


Subject(s)
Methicillin Resistance , Staphylococcal Skin Infections/microbiology , Staphylococcus aureus , Virulence Factors/genetics , Community-Acquired Infections/microbiology , Humans , Leukocidins/genetics , Staphylococcal Skin Infections/drug therapy , Staphylococcus aureus/genetics , Staphylococcus aureus/immunology , Staphylococcus aureus/pathogenicity , Virulence/physiology
17.
J Biol Chem ; 281(30): 20761-20771, 2006 Jul 28.
Article in English | MEDLINE | ID: mdl-16717094

ABSTRACT

The heme-binding proteins Shp and HtsA are part of the heme acquisition machinery found in Streptococcus pyogenes. The hexacoordinate heme (Fe(II)-protoporphyrin IX) or hemochrome form of holoShp (hemoShp) is stable in air in Tris-HCl buffer, pH 8.0, binds to apoHtsA with a K(d) of 120 +/- 18 microm, and transfers its heme to apoHtsA with a rate constant of 28 +/- 6s(-1) at 25 degrees C, pH 8.0. The hemoHtsA product then autoxidizes to the hexacoordinate hemin (Fe(III)-protoporphyrin IX) or hemichrome form (hemiHtsA) with an apparent rate constant of 0.017 +/- 0.002 s(-1). HemiShp also rapidly transfers hemin to apoHtsA through a hemiShp.apoHtsA complex (K(d) = 48 +/- 7 microM) at a rate approximately 40,000 times greater than the rate of simple hemin dissociation from hemiShp into solvent (k(transfer) = 43 +/- 3s(-1) versus k(-hemin) = 0.0003 +/- 0.00006 s(-1)). The rate constants for hemin binding to and dissociation from HtsA (k'(hemin) approximately 80 microm(-1) s(-1), k(-hemin) = 0.0026 +/- 0.0002 s(-1)) are 50- and 10-fold greater than the corresponding rate constants for Shp (k(hemin) approximately 1.6 microM(-1) s(-1), k(-hemin) = 0.0003 s(-1)), which implies that HtsA has a more accessible active site. However, the affinity of apoHtsA for hemin (k(hemin) approximately 31,000 microm(-1)) is roughly 5-fold greater than that of apoShp (k(hemin) approximately 5,300 microM(-1)), accounting for the net transfer from Shp to HstA. These results support a direct, rapid, and affinity-driven mechanism of heme and hemin transfer from the cell surface receptor Shp to the ATP-binding cassette transporter system.


Subject(s)
Heme/chemistry , Hemeproteins/physiology , Streptococcus pyogenes/metabolism , Adenosine Triphosphate/chemistry , Cell Membrane/metabolism , Electron Spin Resonance Spectroscopy , Escherichia coli/metabolism , Hemeproteins/genetics , Hydrogen-Ion Concentration , Kinetics , Membrane Transport Proteins/chemistry , Models, Chemical , Oxygen/chemistry , Oxygen/metabolism , Recombinant Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL