Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Sci Adv ; 8(45): eadd3221, 2022 Nov 11.
Article in English | MEDLINE | ID: mdl-36367946

ABSTRACT

Diabetic wounds have poor healing outcomes due to the presence of numerous pathogens and a dysregulated immune response. Group B Streptococcus (GBS) is commonly isolated from diabetic wound infections, but the mechanisms of GBS virulence during these infections have not been investigated. Here, we develop a murine model of GBS diabetic wound infection and, using dual RNA sequencing, demonstrate that GBS infection triggers an inflammatory response. GBS adapts to this hyperinflammatory environment by up-regulating virulence factors including those known to be regulated by the two-component system covRS, such as the surface protein pbsP, and the cyl operon, which is responsible for hemolysin/pigmentation production. We recover hyperpigmented/hemolytic GBS colonies from the murine diabetic wound, which we determined encode mutations in covR. We further demonstrate that GBS mutants in cylE and pbsP are attenuated in the diabetic wound. This foundational study provides insight into the pathogenesis of GBS diabetic wound infections.

2.
Cell Rep ; 41(9): 111721, 2022 11 29.
Article in English | MEDLINE | ID: mdl-36450248

ABSTRACT

Influenza infection is substantially worsened by the onset of secondary pneumonia caused by bacteria, such as methicillin-resistant Staphylococcus aureus (MRSA). The bidirectional interaction between the influenza-injured lung microenvironment and MRSA is poorly understood. By conditioning MRSA ex vivo in bronchoalveolar lavage fluid collected from mice at various time points of influenza infection, we found that the influenza-injured lung microenvironment dynamically induces MRSA to increase cytotoxin expression while decreasing metabolic pathways. LukAB, a SaeRS two-component system-dependent cytotoxin, is particularly important to the severity of post-influenza MRSA pneumonia. LukAB's activity is likely shaped by the post-influenza lung microenvironment, as LukAB binds to (and is activated by) heparan sulfate (HS) oligosaccharide sequences shed from the epithelial glycocalyx after influenza. Our findings indicate that post-influenza MRSA pneumonia is shaped by bidirectional host-pathogen interactions: host injury triggers changes in bacterial expression of toxins, the activity of which may be shaped by host-derived HS fragments.


Subject(s)
Coinfection , Influenza, Human , Methicillin-Resistant Staphylococcus aureus , Pneumonia, Bacterial , Animals , Mice , Humans , Influenza, Human/complications , Virulence , Pneumonia, Bacterial/complications , Cytotoxins , Heparitin Sulfate , Lung
3.
Microbiol Spectr ; 10(4): e0103922, 2022 08 31.
Article in English | MEDLINE | ID: mdl-35730963

ABSTRACT

Cephalexin and cefadroxil are oral first-generation cephalosporins used to treat methicillin-susceptible Staphylococcus aureus (MSSA) infections. Despite its shorter half-life, cephalexin is more frequently prescribed, although cefadroxil is an appealing alternative, given its slower clearance and possibility for less frequent dosing. We report comparative MIC distributions for cefadroxil and cephalexin, as well as for oxacillin, cephalothin, ceftaroline, and cefazolin, for 48 unique clinical MSSA isolates from pediatric patients with musculoskeletal infections. Both cefadroxil and cephalexin had MIC50 values of 2 µg/mL and MIC90 values of 4 µg/mL. MIC50s for oxacillin, cephalothin, and ceftaroline were ≤0.25 µg/mL, and cefazolin's MIC50 was 0.5 µg/mL. While cefadroxil and cephalexin MICs are higher than those for other active agents, the distributions of MICs for cefadroxil and cephalexin are statistically equivalent, suggesting similar in vitro MSSA activities. Cefadroxil should be further considered an alternative agent to cephalexin, although additional work is needed to identify the optimal dose and frequency of these antibiotics for the treatment of serious MSSA infections. IMPORTANCE Cephalexin and cefadroxil are oral antibiotics that are used to treat serious infections due to the bacteria MSSA. While cephalexin is used more commonly, cefadroxil is excreted from the body more slowly; this generally allows patients to take cefadroxil less frequently than cephalexin. In this study, we compared the abilities of cefadroxil, cephalexin, and several other representative intravenous antibiotics to inhibit the growth of MSSA in the laboratory. Bacterial samples were obtained from children with bone, joint, and/or muscle infections caused by MSSA. We found that cefadroxil and cephalexin inhibited the growth of MSSA at similar concentrations, suggesting similar antibacterial potencies. The selected intravenous antistaphylococcal antibiotics generally inhibited bacterial growth with lower antibiotic concentrations. Based on these results, cefadroxil should be further considered an alternative oral antibiotic to cephalexin, although future research is needed to identify the optimal dose and frequency of these antibiotics for serious infections.


Subject(s)
Cephalexin , Staphylococcal Infections , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Bacteria , Cefadroxil/therapeutic use , Cefazolin/pharmacology , Cefazolin/therapeutic use , Cephalexin/pharmacology , Cephalexin/therapeutic use , Cephalothin/therapeutic use , Child , Humans , Methicillin/therapeutic use , Microbial Sensitivity Tests , Oxacillin/therapeutic use , Staphylococcal Infections/drug therapy , Staphylococcus aureus
4.
Elife ; 112022 01 06.
Article in English | MEDLINE | ID: mdl-34989676

ABSTRACT

Implant-associated Staphylococcus aureus infections are difficult to treat because of biofilm formation. Bacteria in a biofilm are often insensitive to antibiotics and host immunity. Monoclonal antibodies (mAbs) could provide an alternative approach to improve the diagnosis and potential treatment of biofilm-related infections. Here, we show that mAbs targeting common surface components of S. aureus can recognize clinically relevant biofilm types. The mAbs were also shown to bind a collection of clinical isolates derived from different biofilm-associated infections (endocarditis, prosthetic joint, catheter). We identify two groups of antibodies: one group that uniquely binds S. aureus in biofilm state and one that recognizes S. aureus in both biofilm and planktonic state. Furthermore, we show that a mAb recognizing wall teichoic acid (clone 4497) specifically localizes to a subcutaneously implanted pre-colonized catheter in mice. In conclusion, we demonstrate the capacity of several human mAbs to detect S. aureus biofilms in vitro and in vivo.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Biofilms , Staphylococcus aureus/immunology , Animals , Catheter-Related Infections/immunology , Catheter-Related Infections/microbiology , Catheter-Related Infections/therapy , Humans , Male , Mice , Mice, Inbred BALB C , Staphylococcal Infections/microbiology , Teichoic Acids/immunology , Teichoic Acids/metabolism
5.
J Immunol ; 207(3): 966-973, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34290104

ABSTRACT

Neutrophils, polymorphonuclear leukocytes (PMN), play a critical role in the innate immune response to Staphylococcus aureus, a pathogen that continues to be associated with significant morbidity and mortality. Neutrophil extracellular trap (NET) formation is involved in ensnaring and killing of S. aureus, but this host-pathogen interaction also leads to host tissue damage. Importantly, NET components including neutrophil proteases are under consideration as therapeutic targets in a variety of disease processes. Although S. aureus lipoproteins are recognized to activate cells via TLRs, specific mechanisms of interaction with neutrophils are poorly delineated. We hypothesized that a lipoprotein-containing cell membrane preparation from methicillin-resistant S. aureus (MRSA-CMP) would elicit PMN activation, including NET formation. We investigated MRSA-CMP-elicited NET formation, regulated elastase release, and IL-8 production in human neutrophils. We studied PMN from healthy donors with or without a common single-nucleotide polymorphism in TLR1, previously demonstrated to impact TLR2/1 signaling, and used cell membrane preparation from both wild-type methicillin-resistant S. aureus and a mutant lacking palmitoylated lipoproteins (lgt). MRSA-CMP elicited NET formation, elastase release, and IL-8 production in a lipoprotein-dependent manner. TLR2/1 signaling was involved in NET formation and IL-8 production, but not elastase release, suggesting that MRSA-CMP-elicited elastase release is not mediated by triacylated lipoproteins. MRSA-CMP also primed neutrophils for enhanced NET formation in response to a subsequent stimulus. MRSA-CMP-elicited NET formation did not require Nox2-derived reactive oxygen species and was partially dependent on the activity of peptidyl arginine deiminase (PAD). In conclusion, lipoproteins from S. aureus mediate NET formation via TLR2/1 with clear implications for patients with sepsis.


Subject(s)
Cell Membrane/metabolism , Extracellular Traps/metabolism , Lipoproteins/metabolism , Methicillin-Resistant Staphylococcus aureus/metabolism , Neutrophils/immunology , Protein-Arginine Deiminase Type 1/metabolism , Staphylococcal Infections/immunology , Cells, Cultured , Humans , Interleukin-8/metabolism , Lipoproteins/genetics , Lipoylation , Methicillin-Resistant Staphylococcus aureus/genetics , Mutation/genetics , Pancreatic Elastase/metabolism , Polymorphism, Single Nucleotide , Signal Transduction/genetics , Toll-Like Receptor 1/genetics , Toll-Like Receptor 1/metabolism , Toll-Like Receptor 2/metabolism
6.
J Infect Dis ; 223(11): 1865-1869, 2021 06 04.
Article in English | MEDLINE | ID: mdl-32995850

ABSTRACT

Polymorphonuclear leukocytes (PMN) phagocytose and kill individual bacteria but are far less efficient when challenged with bacterial aggregates. Consequently, growth within a biofilm affords Staphylococcus aureus some protection but PMN penetrate S. aureus biofilms and phagocytose bacteria, suggesting that enzymes released through neutrophil degranulation degrade biofilms into fragments small enough for phagocytosis. Here we show that the capacity of PMN to invade biofilms depended largely on the activity of secreted cathepsin G.


Subject(s)
Cathepsin G , Neutrophils , Phagocytosis , Staphylococcal Infections , Staphylococcus aureus , Biofilms , Humans , Neutrophils/immunology
7.
J Allergy Clin Immunol ; 147(3): 955-966.e16, 2021 03.
Article in English | MEDLINE | ID: mdl-32634452

ABSTRACT

BACKGROUND: Staphylococcus aureus and Staphylococcus epidermidis are the most abundant bacteria found on the skin of patients with atopic dermatitis (AD). S aureus is known to exacerbate AD, whereas S epidermidis has been considered a beneficial commensal organism. OBJECTIVE: In this study, we hypothesized that S epidermidis could promote skin damage in AD by the production of a protease that damages the epidermal barrier. METHODS: The protease activity of S epidermidis isolates was compared with that of other staphylococcal species. The capacity of S epidermidis to degrade the barrier and induce inflammation was examined by using human keratinocyte tissue culture and mouse models. Skin swabs from atopic and healthy adult subjects were analyzed for the presence of S epidermidis genomic DNA and mRNA. RESULTS: S epidermidis strains were observed to produce strong cysteine protease activity when grown at high density. The enzyme responsible for this activity was identified as EcpA, a cysteine protease under quorum sensing control. EcpA was shown to degrade desmoglein-1 and LL-37 in vitro, disrupt the physical barrier, and induce skin inflammation in mice. The abundance of S epidermidis and expression of ecpA mRNA were increased on the skin of some patients with AD, and this correlated with disease severity. Another commensal skin bacterial species, Staphylococcus hominis, can inhibit EcpA production by S epidermidis. CONCLUSION: S epidermidis has commonly been regarded as a beneficial skin microbe, whereas S aureus has been considered deleterious. This study suggests that the overabundance of S epidermidis found on some atopic patients can act similarly to S aureus and damage the skin by expression of a cysteine protease.


Subject(s)
Bacterial Proteins/metabolism , Cysteine Proteases/metabolism , Dermatitis, Atopic/microbiology , Microbiota , Skin/microbiology , Staphylococcal Skin Infections/microbiology , Staphylococcus epidermidis/enzymology , Animals , Antimicrobial Cationic Peptides/metabolism , Cells, Cultured , DNA, Bacterial/genetics , Dermatitis, Atopic/pathology , Desmoglein 1/metabolism , Humans , Keratinocytes/microbiology , Keratinocytes/pathology , Mice , Mice, Inbred C57BL , Severity of Illness Index , Skin/pathology , Staphylococcal Skin Infections/pathology , Cathelicidins
8.
mSphere ; 4(4)2019 08 14.
Article in English | MEDLINE | ID: mdl-31413175

ABSTRACT

Methicillin-resistant Staphylococcus aureus (MRSA) infections impact all patient populations both in the community and in health care settings. Despite advances in our knowledge of MRSA virulence, little is known about the regulatory mechanisms of USA100 health care-associated MRSA isolates, which are the second most frequently identified MRSA isolates found in all infections. This work focused on the contribution of the USA100 agr type II quorum-sensing system to virulence and antibiotic resistance. From a MRSA strain collection, we selected 16 representative USA100 isolates, constructed mutants with Δagr mutations, and characterized selected strain pairs for virulence factor expression, murine skin infection, and antibiotic resistance. For each strain pair, hemolysis and extracellular protease expression were significantly greater in the wild-type (WT) strains than in the Δagr mutants. Similarly, mice challenged with the WT strains had larger areas of dermonecrosis and greater weight loss than those challenged with the Δagr mutants, demonstrating that the USA100 agr system regulates virulence. Although USA100 isolates exhibit a high level of antibiotic resistance, the WT and Δagr strain pairs showed no difference in MICs by MIC testing. However, in the presence of a sub-MIC of vancomycin, most of the USA100 Δagr mutants exhibited slower growth than the WT isolates, and a couple of the Δagr mutants also grew more slowly in the presence of a sub-MIC of cefoxitin. Altogether, our findings demonstrate that the USA100 agr system is a critical regulator of virulence, and it may have a contribution to the optimal survival of these MRSA strains in the presence of antibiotics.IMPORTANCE USA100 health care-associated MRSA isolates are highly antibiotic resistant and can cause invasive disease across all patient populations. Even though USA100 strains are some of the most frequently identified causes of infections, little is known about virulence regulation in these isolates. Our study demonstrates that the USA100 agr quorum-sensing system is important for the control of toxin and exoenzyme production and that the agr system has a key role in skin infection. In some USA100 isolates, the agr system is important for growth in the presence of low levels of antibiotics. Altogether, our findings demonstrate that the USA100 agr system is a critical regulator of virulence and that it may make a contribution to the optimal survival of these MRSA strains in the presence of antibiotics.


Subject(s)
Drug Resistance, Multiple, Bacterial , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Quorum Sensing , Virulence , Animals , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Humans , Methicillin-Resistant Staphylococcus aureus/genetics , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Staphylococcal Infections/microbiology , Trans-Activators/genetics , Virulence Factors/genetics
9.
mBio ; 10(3)2019 06 25.
Article in English | MEDLINE | ID: mdl-31239382

ABSTRACT

We developed a new approach that couples Southwestern blotting and mass spectrometry to discover proteins that bind extracellular DNA (eDNA) in bacterial biofilms. Using Staphylococcus aureus as a model pathogen, we identified proteins with known DNA-binding activity and uncovered a series of lipoproteins with previously unrecognized DNA-binding activity. We demonstrated that expression of these lipoproteins results in an eDNA-dependent biofilm enhancement. Additionally, we found that while deletion of lipoproteins had a minimal impact on biofilm accumulation, these lipoprotein mutations increased biofilm porosity, suggesting that lipoproteins and their associated interactions contribute to biofilm structure. For one of the lipoproteins, SaeP, we showed that the biofilm phenotype requires the lipoprotein to be anchored to the outside of the cellular membrane, and we further showed that increased SaeP expression correlates with more retention of high-molecular-weight DNA on the bacterial cell surface. SaeP is a known auxiliary protein of the SaeRS system, and we also demonstrated that the levels of SaeP correlate with nuclease production, which can further impact biofilm development. It has been reported that S. aureus biofilms are stabilized by positively charged cytoplasmic proteins that are released into the extracellular environment, where they make favorable electrostatic interactions with the negatively charged cell surface and eDNA. In this work we extend this electrostatic net model to include secreted eDNA-binding proteins and membrane-attached lipoproteins that can function as anchor points between eDNA in the biofilm matrix and the bacterial cell surface.IMPORTANCE Many bacteria are capable of forming biofilms encased in a matrix of self-produced extracellular polymeric substances (EPS) that protects them from chemotherapies and the host defenses. As a result of these inherent resistance mechanisms, bacterial biofilms are extremely difficult to eradicate and are associated with chronic wounds, orthopedic and surgical wound infections, and invasive infections, such as infective endocarditis and osteomyelitis. It is therefore important to understand the nature of the interactions between the bacterial cell surface and EPS that stabilize biofilms. Extracellular DNA (eDNA) has been recognized as an EPS constituent for many bacterial species and has been shown to be important in promoting biofilm formation. Using Staphylococcus aureus biofilms, we show that membrane-attached lipoproteins can interact with the eDNA in the biofilm matrix and promote biofilm formation, which suggests that lipoproteins are potential targets for novel therapies aimed at disrupting bacterial biofilms.


Subject(s)
Bacterial Proteins/metabolism , Biofilms , DNA-Binding Proteins/metabolism , Lipoproteins/metabolism , Staphylococcus aureus/genetics , Bacterial Proteins/genetics , Blotting, Southwestern , DNA, Bacterial/genetics , DNA-Binding Proteins/genetics , Extracellular Polymeric Substance Matrix/genetics , Lipoproteins/genetics , Mass Spectrometry , Staphylococcus aureus/physiology , Static Electricity
10.
Cell Rep ; 27(1): 187-198.e6, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30943400

ABSTRACT

Recurrent epidemics of drug-resistant Staphylococcus aureus illustrate the rapid lapse of antibiotic efficacy following clinical implementation. Over the last decade, community-associated methicillin-resistant S. aureus (MRSA) has emerged as a dominant cause of infections, and this problem is amplified by the hyper-virulent nature of these isolates. Herein, we report the discovery of a fungal metabolite, apicidin, as an innovative means to counter both resistance and virulence. Owing to its breadth and specificity as a quorum-sensing inhibitor, apicidin antagonizes all MRSA agr systems in a non-biocidal manner. In skin challenge experiments, the apicidin-mediated abatement of MRSA pathogenesis corresponds with quorum-sensing inhibition at in vivo sites of infection. Additionally, we show that apicidin attenuates MRSA-induced disease by potentiating innate effector responses, particularly through enhanced neutrophil accumulation and function at cutaneous challenge sites. Together, these results indicate that apicidin treatment represents a strategy to limit MRSA virulence and promote host defense.


Subject(s)
Immunity, Innate/drug effects , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Peptides, Cyclic/pharmacology , Quorum Sensing/drug effects , Staphylococcal Infections/immunology , Animals , Cells, Cultured , Drug Evaluation, Preclinical , Female , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Rabbits , Staphylococcal Infections/microbiology , Virulence/drug effects
11.
J Med Chem ; 59(19): 8879-8888, 2016 10 13.
Article in English | MEDLINE | ID: mdl-27585401

ABSTRACT

Competitive quorum sensing (QS) antagonism offers a novel strategy for attenuating current multidrug resistant staphylococcal infections. To this end, a series of 10 truncated analogues based on the parent autoinducing peptides (AIPs) of Staphylococcus lugdunensis (groups I and II) and Staphylococcus epidermidis (groups I-III) were sequentially assessed against a newly developed Staphylococcus lugdunensis group I QS reporter strain. The truncated analogues based upon Staphylococcus lugdunensis AIP-1 (1) and AIP-2 (2) displayed respective IC50 values of 0.2 ± 0.01 µM and 0.3 ± 0.01 µM, while the truncated analogue of the Staphylococcus epidermidis AIP-1 (3) elicited an IC50 value of 2.7 ± 0.1 µM. These findings demonstrate the potential of cognate and "crosstalk" competitive quorum sensing inhibition using truncated AIPs as a means of attenuating staphylococcal infections in species beyond Staphylococcus aureus.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/chemistry , Bacterial Proteins/pharmacology , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Quorum Sensing/drug effects , Staphylococcal Infections/drug therapy , Staphylococcus lugdunensis/drug effects , Drug Discovery , Humans , Models, Molecular , Staphylococcal Infections/microbiology , Staphylococcus epidermidis/chemistry , Staphylococcus lugdunensis/physiology
12.
J Microbiol Methods ; 127: 89-94, 2016 08.
Article in English | MEDLINE | ID: mdl-27237773

ABSTRACT

Drug resistant bacterial infections cause significant morbidity and mortality worldwide, and new strategies are needed for the treatment of these infections. The anti-virulence approach, which targets non-essential virulence factors in bacteria, has been proposed as one way to combat the problem of antibiotic resistance. Virulence in methicillin-resistant Staphylococcus aureus (MRSA) and many other Gram-positive bacterial pathogens is controlled by the quorum sensing system. Thus, there is excellent therapeutic potential for compounds that target this system. With this project, we have developed and validated a novel approach for measuring quorum sensing inhibition in vitro. Ultraperformance liquid chromatography coupled to mass spectrometry (UPLC-MS) was employed to directly measure one of the important outputs of the quorum sensing system in MRSA, auto-inducing peptide I (AIP I) in bacterial cultures. The method for AIP detection was validated and demonstrated limits of detection and quantification of range of 0.0035µM and 0.10µM, respectively. It was shown that the known quorum sensing inhibitor ambuic acid inhibited AIP I production by a clinically relevant strain of MRSA, with an IC50 value of 2.6±0.2µM. The new method performed similarly to previously published methods using GFP reporter assays, but has the advantage of being applicable without the need for engineering of a reporter strain. Additionally, the mass spectrometry-based method could be applicable in situations where interference by the inhibitor prevents the application of fluorescence-based methods.


Subject(s)
Bacterial Proteins/isolation & purification , Cyclohexanones/pharmacology , Mass Spectrometry/methods , Methicillin-Resistant Staphylococcus aureus/drug effects , Peptides, Cyclic/isolation & purification , Quorum Sensing , Gene Expression Regulation, Bacterial/drug effects , Green Fluorescent Proteins , Inhibitory Concentration 50 , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Methicillin-Resistant Staphylococcus aureus/physiology , Staphylococcal Infections/diagnosis , Staphylococcal Infections/microbiology , Virulence Factors
13.
J Biol Chem ; 291(24): 12556-12564, 2016 Jun 10.
Article in English | MEDLINE | ID: mdl-27129223

ABSTRACT

Staphylococci are commensal bacteria that colonize the epithelial surfaces of humans and many other mammals. These bacteria can also attach to implanted medical devices and develop surface-associated biofilm communities that resist clearance by host defenses and available chemotherapies. These communities are often associated with persistent staphylococcal infections that place a tremendous burden on the healthcare system. Understanding the regulatory program that controls staphylococcal biofilm development, as well as the environmental conditions that modulate this program, has been a focal point of research in recent years. A central regulator controlling biofilm development is a peptide quorum-sensing system, also called the accessory gene regulator or agr system. In the opportunistic pathogen Staphylococcus aureus, the agr system controls production of exo-toxins and exo-enzymes essential for causing infections, and simultaneously, it modulates the ability of this pathogen to attach to surfaces and develop a biofilm, or to disperse from the biofilm state. In this review, we explore advances on the interconnections between the agr quorum-sensing system and biofilm mechanisms, and topics covered include recent findings on how different environmental conditions influence quorum sensing, the impact on biofilm development, and ongoing questions and challenges in the field. As our understanding of the quorum sensing and biofilm interconnection advances, there are growing opportunities to take advantage of this knowledge and develop therapeutic approaches to control staphylococcal infections.


Subject(s)
Biofilms/growth & development , Environment , Quorum Sensing/physiology , Staphylococcal Infections/microbiology , Staphylococcus aureus/physiology , Gene Expression Regulation, Bacterial , Host-Pathogen Interactions , Humans , Hydrogen-Ion Concentration , Models, Genetic , Quorum Sensing/genetics , Staphylococcus aureus/genetics
14.
Antimicrob Agents Chemother ; 59(4): 2223-35, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25645827

ABSTRACT

Antibiotic-resistant pathogens are a global health threat. Small molecules that inhibit bacterial virulence have been suggested as alternatives or adjuncts to conventional antibiotics, as they may limit pathogenesis and increase bacterial susceptibility to host killing. Staphylococcus aureus is a major cause of invasive skin and soft tissue infections (SSTIs) in both the hospital and community settings, and it is also becoming increasingly antibiotic resistant. Quorum sensing (QS) mediated by the accessory gene regulator (agr) controls virulence factor production essential for causing SSTIs. We recently identified ω-hydroxyemodin (OHM), a polyhydroxyanthraquinone isolated from solid-phase cultures of Penicillium restrictum, as a suppressor of QS and a compound sought for the further characterization of the mechanism of action. At concentrations that are nontoxic to eukaryotic cells and subinhibitory to bacterial growth, OHM prevented agr signaling by all four S. aureus agr alleles. OHM inhibited QS by direct binding to AgrA, the response regulator encoded by the agr operon, preventing the interaction of AgrA with the agr P2 promoter. Importantly, OHM was efficacious in a mouse model of S. aureus SSTI. Decreased dermonecrosis with OHM treatment was associated with enhanced bacterial clearance and reductions in inflammatory cytokine transcription and expression at the site of infection. Furthermore, OHM treatment enhanced the immune cell killing of S. aureus in vitro in an agr-dependent manner. These data suggest that bacterial disarmament through the suppression of S. aureus QS may bolster the host innate immune response and limit inflammation.


Subject(s)
Anti-Bacterial Agents/pharmacology , Emodin/analogs & derivatives , Inflammation/prevention & control , Methicillin-Resistant Staphylococcus aureus/drug effects , Quorum Sensing/drug effects , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Animals , Bacterial Proteins/genetics , Cytokines/biosynthesis , Emodin/pharmacology , Humans , In Vitro Techniques , Inflammation/etiology , Inflammation/pathology , Leukocytes/microbiology , Mice , Models, Molecular , Rabbits , Staphylococcal Infections/pathology , Trans-Activators/genetics , Virulence Factors/metabolism
15.
Mol Microbiol ; 96(2): 388-404, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25612137

ABSTRACT

Staphylococcus aureus is a significant cause of chronic biofilm infections on medical implants. We investigated the biofilm regulatory cascade and discovered that the repressor of toxins (Rot) is part of this pathway. A USA300 community-associated methicillin-resistant S. aureus strain deficient in Rot was unable to form a biofilm using multiple different assays, and we found rot mutants in other strain lineages were also biofilm deficient. By performing a global analysis of transcripts and protein production controlled by Rot, we observed that all the secreted protease genes were up-regulated in a rot mutant, and we hypothesized that this regulation could be responsible for the biofilm phenotype. To investigate this question, we determined that Rot bound to the protease promoters, and we observed that activity levels of these enzymes, in particular the cysteine proteases, were increased in a rot mutant. By inactivating these proteases, biofilm capacity was restored to the mutant, demonstrating they are responsible for the biofilm negative phenotype. Finally, we tested the rot mutant in a mouse catheter model of biofilm infection and observed a significant reduction in biofilm burden. Thus S. aureus uses the transcription factor Rot to repress secreted protease levels in order to build a biofilm.


Subject(s)
Bacterial Proteins/metabolism , Biofilms , Repressor Proteins/metabolism , Staphylococcal Infections/microbiology , Staphylococcus aureus/physiology , Animals , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Humans , Male , Mice , Mice, Inbred C57BL , Peptide Hydrolases/genetics , Peptide Hydrolases/metabolism , Promoter Regions, Genetic , Repressor Proteins/genetics , Staphylococcus aureus/genetics
16.
J Leukoc Biol ; 97(4): 769-78, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25605871

ABSTRACT

Alcoholics are at increased risk of Staphylococcus aureus skin infection and serious sequelae, such as bacteremia and death. Despite the association between alcoholism and severe S. aureus skin infection, the impact of EtOH on anti-S. aureus cutaneous immunity has not been investigated in a model of chronic EtOH exposure. To test the hypothesis that EtOH enhances the severity of S. aureus skin infection, mice were fed EtOH for ≥12 weeks via the Meadows-Cook model of alcoholism and inoculated with S. aureus following epidermal abrasion. Evidence of exacerbated staphylococcal disease in EtOH-fed mice included: skin lesions that were larger and contained more organisms, greater weight loss, and increased bacterial dissemination. Infected EtOH-fed mice demonstrated poor maintenance and induction of PMN responses in skin and draining LNs, respectively. Additionally, altered PMN dynamics in the skin of these mice corresponded with reduced production of IL-23 and IL-1ß by CD11b(+) myeloid cells and IL-17 production by γδ T cells, with the latter defect occurring in the draining LNs as well. In addition, IL-17 restoration attenuated S. aureus-induced dermatopathology and improved bacterial clearance defects in EtOH-fed mice. Taken together, the findings show, in a novel model system, that the EtOH-induced increase in S. aureus-related injury/illness corresponds with defects in the IL-23/IL-17 inflammatory axis and poor PMN accumulation at the site of infection and draining LNs. These findings offer new information about the impact of EtOH on cutaneous host-defense pathways and provide a potential mechanism explaining why alcoholics are predisposed to S. aureus skin infection.


Subject(s)
Alcoholism/immunology , Ethanol/toxicity , Staphylococcal Skin Infections/immunology , Animals , Antimicrobial Cationic Peptides , Bacteremia/etiology , Bacterial Load , Cathelicidins/biosynthesis , Disease Susceptibility , Female , Hypersensitivity, Delayed/immunology , Immunocompromised Host , Interleukin-17/biosynthesis , Interleukin-17/therapeutic use , Interleukin-1beta/biosynthesis , Interleukin-23/biosynthesis , Lymph Nodes/metabolism , Mice , Mice, Inbred C57BL , Myeloid Cells/immunology , Myeloid Cells/metabolism , Neutrophil Infiltration , Receptors, Antigen, T-Cell, gamma-delta , Recombinant Proteins/therapeutic use , Skin/immunology , Skin/metabolism , Skin/pathology , Specific Pathogen-Free Organisms , Staphylococcal Skin Infections/drug therapy , Staphylococcal Skin Infections/etiology , Staphylococcal Skin Infections/microbiology , Staphylococcal Skin Infections/pathology , Staphylococcus aureus/immunology , Staphylococcus aureus/isolation & purification , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Toll-Like Receptor 2/biosynthesis , Weight Loss
17.
J Nat Prod ; 77(6): 1351-8, 2014 Jun 27.
Article in English | MEDLINE | ID: mdl-24911880

ABSTRACT

The endophytic fungus Penicillium restrictum was isolated from the stems of a milk thistle (Silybum marianum) plant. In culture, the fungus produced distinct red guttates, which have been virtually uninvestigated, particularly from the standpoint of chemistry. Hence, this study examined the chemical mycology of P. restrictum and, in doing so, uncovered a series of both known and new polyhydroxyanthraquinones (1-9). These compounds were quorum sensing inhibitors in a clinical isolate of methicillin-resistant Staphylococcus aureus (MRSA), with IC50 values ranging from 8 to 120 µM, suggesting antivirulence potential for the compounds. Moreover, the spatial and temporal distribution of the polyhydroxyanthraquinones was examined in situ via desorption electrospray ionization-mass spectrometry (DESI-MS) imaging, demonstrating the first application of this technique to a guttate-forming fungus and revealing both the concentration of secondary metabolites at the ventral surface of the fungus and their variance in colonies of differing ages.


Subject(s)
Anthraquinones/isolation & purification , Anthraquinones/pharmacology , Penicillium/chemistry , Spectrometry, Mass, Electrospray Ionization/methods , Anthraquinones/chemistry , Inhibitory Concentration 50 , Methicillin-Resistant Staphylococcus aureus/drug effects , Silybum marianum/microbiology , Molecular Structure , Quorum Sensing
18.
Infect Immun ; 81(11): 4053-62, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23959719

ABSTRACT

The autoinducer-2 (AI-2) quorum-sensing system has been linked to diverse phenotypes and regulatory changes in pathogenic bacteria. In the present study, we performed a molecular and biochemical characterization of the AI-2 system in Yersinia pestis, the causative agent of plague. In strain CO92, the AI-2 signal is produced in a luxS-dependent manner, reaching maximal levels of 2.5 µM in the late logarithmic growth phase, and both wild-type and pigmentation (pgm) mutant strains made equivalent levels of AI-2. Strain CO92 possesses a chromosomal lsr locus encoding factors involved in the binding and import of AI-2, and confirming this assignment, an lsr deletion mutant increased extracellular pools of AI-2. To assess the functional role of AI-2 sensing in Y. pestis, microarray studies were conducted by comparing Δpgm strain R88 to a Δpgm ΔluxS mutant or a quorum-sensing-null Δpgm ΔypeIR ΔyspIR ΔluxS mutant at 37°C. Our data suggest that AI-2 quorum sensing is associated with metabolic activities and oxidative stress genes that may help Y. pestis survive at the host temperature. This was confirmed by observing that the luxS mutant was more sensitive to killing by hydrogen peroxide, suggesting a potential requirement for AI-2 in evasion of oxidative damage. We also show that a large number of membrane protein genes are controlled by LuxS, suggesting a role for quorum sensing in membrane modeling. Altogether, this study provides the first global analysis of AI-2 signaling in Y. pestis and identifies potential roles for the system in controlling genes important to disease.


Subject(s)
Gene Expression Regulation, Bacterial , Homoserine/analogs & derivatives , Lactones/metabolism , Quorum Sensing , Yersinia pestis/genetics , Cell Membrane/physiology , Gene Deletion , Gene Expression Profiling , Genes, Bacterial , Homoserine/metabolism , Membrane Proteins/metabolism , Microarray Analysis , Oxidative Stress , Stress, Physiological , Yersinia pestis/physiology
19.
Article in English | MEDLINE | ID: mdl-23703543

ABSTRACT

Staphylococcus aureus infections acquired in hospitals now cause more deaths per annum in the US than does HIV/AIDS. Perhaps even more alarming is the rise in community associated methicillin-resistant S. aureus (CA-MRSA) infections, which have spread out of hospital settings and are infecting otherwise healthy individuals. The mechanism of enhanced pathogenesis in CA-MRSA remains unclear, but it has been postulated that high activity in the agr quorum-sensing system could be a contributing factor. The purpose of this study was to develop a quantitative method for analysis of autoinducing peptide I (AIP-I), the activating signal for the agr system in S. aureus. An effective method was developed using ultrahigh performance liquid chromatography (UHPLC) coupled to electrospray ionization mass spectrometry with an LTQ Orbitrap mass spectrometer. Relying on the exceptional resolving power and mass accuracy of this instrument configuration, it was possible to quantify AIP-I directly from the complex growth media of S. aureus cultures with a limit of detection (LOD) of 0.25µM and a linear dynamic range of 2.6 to 63µM. The method was then employed to monitor time-dependent production of AIP-I by S. aureus cultures, and it was observed that AIP-I production reached a maximum and leveled off after approximately 16h. Finally, it was determined that virulence of S. aureus was correlated with AIP-I production in some (but not all) strains analyzed.


Subject(s)
Bacterial Proteins/analysis , Chromatography, High Pressure Liquid/methods , Mass Spectrometry/methods , Peptides, Cyclic/analysis , Staphylococcus aureus/chemistry , Animals , Bacterial Proteins/metabolism , Bacterial Proteins/toxicity , Hemolysis/drug effects , Peptides, Cyclic/metabolism , Peptides, Cyclic/toxicity , Rabbits , Reproducibility of Results , Staphylococcus aureus/metabolism
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL