1
|
Qi Z, Lin S, Yu Q, Ma R, Zhang K, Jiang W, Chen S, Mai Y, Fu QB. Human neutrophil defensin-1 binding increases histidine kinase activity of SaeS in Staphylococcus aureus. Biochem Biophys Rep 2025; 42:101982. [PMID: 40207086 PMCID: PMC11981803 DOI: 10.1016/j.bbrep.2025.101982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 03/06/2025] [Accepted: 03/17/2025] [Indexed: 04/11/2025] Open
Abstract
Human neutrophil defensin-1 (HNP-1) can specifically activate the SaeRS two-component system(TCS), which is essential for controlling virulence and immune evasion factors in Staphylococcus aureus. The reaction to HNP1 requires the transmembrane domain of SaeS (SaeS™), however the precise mechanism is yet unknown. In this work, we reconstructed the SaeS™ protein into bicelles and discovered that HNP1 can interact directly with SaeS™ using BiacoreT200, their binding significantly increases SaeS kinase activity and activated the SaeRS system subsequently. Staphylococcus aureus may exploit host-derived factors released by human immune cells to activate its two-component signal transduction system, thereby enhancing antimicrobial peptide resistance.
Collapse
Affiliation(s)
- Zhengfei Qi
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Shanghai Institute of Materia Medica, Zhongshan Institute for Drug Discovery, Chinese Academy of Sciences, Zhongshan, 528400, China
| | - Shuru Lin
- College of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
- Shanghai Institute of Materia Medica, Zhongshan Institute for Drug Discovery, Chinese Academy of Sciences, Zhongshan, 528400, China
| | - Quanxiang Yu
- Shanghai Institute of Materia Medica, Zhongshan Institute for Drug Discovery, Chinese Academy of Sciences, Zhongshan, 528400, China
| | - Rui Ma
- Shanghai Institute of Materia Medica, Zhongshan Institute for Drug Discovery, Chinese Academy of Sciences, Zhongshan, 528400, China
| | - Kexin Zhang
- Shanghai Institute of Materia Medica, Zhongshan Institute for Drug Discovery, Chinese Academy of Sciences, Zhongshan, 528400, China
| | - Wenqi Jiang
- Shanghai Institute of Materia Medica, Zhongshan Institute for Drug Discovery, Chinese Academy of Sciences, Zhongshan, 528400, China
| | - Shurong Chen
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Shanghai Institute of Materia Medica, Zhongshan Institute for Drug Discovery, Chinese Academy of Sciences, Zhongshan, 528400, China
| | - Yilin Mai
- Shanghai Institute of Materia Medica, Zhongshan Institute for Drug Discovery, Chinese Academy of Sciences, Zhongshan, 528400, China
| | - Qingshan Bill Fu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- College of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
- Shanghai Institute of Materia Medica, Zhongshan Institute for Drug Discovery, Chinese Academy of Sciences, Zhongshan, 528400, China
| |
Collapse
|
2
|
Pettygrove BA, Nygaard TK, Borgogna TR, Malachowa N, Gaur G, Salo SE, Pallister KB, Burroughs O, Robinson C, Gao A, Sturdevant DE, Ricklefs S, DeLeo FR, Otto M, Stewart PS, Voyich JM. Staphylococcus aureus SaeR/S-regulated factors overcome human complement-mediated inhibition of aggregation to evade neutrophil killing. Proc Natl Acad Sci U S A 2025; 122:e2412447122. [PMID: 40359050 DOI: 10.1073/pnas.2412447122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 03/25/2025] [Indexed: 05/15/2025] Open
Abstract
Staphylococcus aureus (S. aureus) is a frequent culprit in implant-associated infections and employs many virulence factors to escape killing by the host immune system. The specific immune evasion strategies used by small aggregates of S. aureus on a surface, precursors to mature biofilm, are still relatively unknown. Time-lapse confocal microscopy was leveraged to quantify interactions between S. aureus aggregates and human neutrophils in vitro and identify specific mechanisms of resistance to neutrophil killing. Surface-associated wild-type S. aureus rapidly formed small biofilm aggregates when grown in human serum. Conversely, aggregation was inhibited when the SaeR/S two-component gene regulatory system was deleted. Wild-type aggregates began to show individual and population-level resistance to neutrophil killing upon reaching sizes of approximately 50 to 75 µm2, whereas Δsae clusters failed to reach these sizes and were readily cleared. Aggregation of Δsae strains was impaired by serum complement, and this inhibition required complement proteins C3 and factor B, but not C4 or C5, suggesting that this activity primarily occurs at the level of the alternative pathway. Several complement-inhibiting genes regulated by SaeR/S were identified that collectively facilitate biofilm aggregate formation in human, but not murine serum. Finally, aggregation of two related opportunistic pathogens, Staphylococcus epidermidis and Enterococcus faecalis, was inhibited by serum. These data demonstrate a function of serum complement, the ability to inhibit bacterial aggregation, that is potently blocked by S. aureus through the production of multiple complement-interfering proteins that are regulated by the SaeR/S system.
Collapse
Affiliation(s)
- Brian A Pettygrove
- Center for Biofilm Engineering, Montana State University, Bozeman, MT 59717
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, MT 59717
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, Division of Intramural Research, National Institute of Allergy and Infection Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Tyler K Nygaard
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, MT 59717
| | - Timothy R Borgogna
- Center for Biofilm Engineering, Montana State University, Bozeman, MT 59717
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, MT 59717
| | - Natalia Malachowa
- Laboratory of Bacteriology, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840
| | - Gauri Gaur
- Center for Biofilm Engineering, Montana State University, Bozeman, MT 59717
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, MT 59717
| | - Shannon E Salo
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, Division of Intramural Research, National Institute of Allergy and Infection Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Kyler B Pallister
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, MT 59717
| | - Owen Burroughs
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, MT 59717
| | - Cassandra Robinson
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, MT 59717
| | - Annika Gao
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, MT 59717
| | - Daniel E Sturdevant
- Research Technologies Branch, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840
| | - Stacy Ricklefs
- Research Technologies Branch, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840
| | - Frank R DeLeo
- Laboratory of Bacteriology, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840
| | - Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, Division of Intramural Research, National Institute of Allergy and Infection Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Philip S Stewart
- Center for Biofilm Engineering, Montana State University, Bozeman, MT 59717
- Department of Chemical and Biological Engineering, Montana State University, Bozeman, MT 59717
| | - Jovanka M Voyich
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, MT 59717
| |
Collapse
|
3
|
Jobson ME, Tomlinson BR, Mustor EM, Felton EA, Weiss A, Caswell CC, Shaw LN. SSR42 is a novel regulator of cytolytic activity in Staphylococcus aureus. mBio 2025:e0077225. [PMID: 40340377 DOI: 10.1128/mbio.00772-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Accepted: 04/08/2025] [Indexed: 05/10/2025] Open
Abstract
SSR42 is the longest noncoding RNA in the Staphylococcus aureus cell and the second-most abundant transcript in the stationary-phase transcriptome, second only to RNAIII. It is highly conserved across strains and exhibits pronounced stability in stationary phase; however, the mechanism behind its regulatory role has yet to be fully elucidated. Herein, we used transcriptomic and proteomic approaches to probe the role of SSR42, revealing that it is a powerful, novel activator of the primary leukocidin LukAB. SSR42 is required for cytotoxicity toward, and escape from within, human neutrophils, and also mediates survival within human blood. We show that SSR42 wields this role via derepression by the peroxide repressor PerR in response to the presence of human neutrophils and governs lukAB induction in this niche. Importantly, this regulation is driven by direct RNA-RNA interaction, as we show binding of the 5' untranslated region (UTR) of the lukAB transcript with the 3' end of SSR42, which ultimately modulates transcript stability as well as translational activity. Finally, we demonstrate that this behavior is absolutely required for full virulence of S. aureus in murine models of both pneumonia and sepsis. Collectively, we present SSR42 as a pleiotropic regulatory RNA that acts as a nexus between environmental sensing and the regulation of pathogenesis, responding to environmental stimuli and host immune factors to bolster cytotoxic behavior and facilitate infection in S. aureus.IMPORTANCEStaphylococcus aureus is a master pathogen due to its formidable collection of virulence factors. These are tightly controlled by a diverse group of regulators that titrate their abundance to adapt to unique infectious niches. The role of regulatory RNAs in stress adaptation and pathogenesis is becoming increasingly more relevant in S. aureus. In this study, we provide the most comprehensive global analysis to date of just such a factor, SSR42. Specifically, we uncover that SSR42 is required for mediating cytotoxicity-one of the pillars of infection-in response to phagocytosis by human neutrophils. We find that SSR42 is induced by components of the host immune system and facilitates downstream activation of cytotoxic factors via RNA-RNA interactions. This illustrates that SSR42 forms a pivotal link between sensing the external environment and mediating resistance to oxidative stress while promoting virulence, solidifying it as a major global regulator in S. aureus.
Collapse
Affiliation(s)
- Mary-Elizabeth Jobson
- Department of Molecular Biosciences, University of South Florida, Tampa, Florida, USA
- Center for Antimicrobial Resistance, University of South Florida, Tampa, Florida, USA
| | - Brooke R Tomlinson
- Department of Molecular Biosciences, University of South Florida, Tampa, Florida, USA
| | - Emilee M Mustor
- Department of Molecular Biosciences, University of South Florida, Tampa, Florida, USA
- Center for Antimicrobial Resistance, University of South Florida, Tampa, Florida, USA
| | - Emily A Felton
- Department of Molecular Biosciences, University of South Florida, Tampa, Florida, USA
- Center for Antimicrobial Resistance, University of South Florida, Tampa, Florida, USA
| | - Andy Weiss
- Department of Molecular Biosciences, University of South Florida, Tampa, Florida, USA
| | - Clayton C Caswell
- Department of Biomedical Sciences and Pathobiology, Center for One Health Research, VA-MD College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
| | - Lindsey N Shaw
- Department of Molecular Biosciences, University of South Florida, Tampa, Florida, USA
- Center for Antimicrobial Resistance, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
4
|
Predtechenskaya M, Arbizzani CJ, Shomento SR, Borgogna TR, Voyich JM. Lung surfactant reduces Staphylococcus aureus cytotoxicity and protects host immune cells from membrane damage. Microbiol Spectr 2025:e0138624. [PMID: 40237467 DOI: 10.1128/spectrum.01386-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 01/20/2025] [Indexed: 04/18/2025] Open
Abstract
In this study, we identify that lung surfactant significantly reduces the cytotoxicity of Staphylococcus aureus (S. aureus) membrane-damaging toxins. Data demonstrate that natural surfactants from mice and rats and commercially available surfactant, Infasurf, protect human primary cells (neutrophils and peripheral blood mononuclear cells) from cytolytic activity caused by S. aureus supernatants. Supernatants from S. aureus grown in surfactant showed a significant reduction in plasma membrane damage against primary human cells as compared to supernatants grown without surfactant. This reduction was not due to a direct bactericidal effect of the surfactants on S. aureus growth. Rat and mouse surfactants downregulated the gene expression of saeR, the response regulator of the S. aureus two-component system SaeR/S that is responsible for the production of virulence factors which are important during lung infection and cause membrane damage in host cells. Rat and lung surfactants also reduced transcript abundance of SaeR/S-regulated genes lukF-PV, hla, and hlgA. Interestingly, the commercially available surfactant Infasurf did not recapitulate the effect of natural surfactants and did not decrease gene transcription of the virulence genes tested. These data suggest that components of natural surfactants protect lungs from S. aureus by suppressing S. aureus virulence factors and have implications for the role of surfactants in host defense against S. aureus.IMPORTANCEThis study explored the influence of lung surfactants on membrane-damaging Staphylococcus aureus (S. aureus) toxins. We demonstrate that natural and commercially available lung surfactants minimize the cytolytic capacity of S. aureus supernatants against primary human cells. Data indicate that cytolytic reduction by mouse and rat surfactants was partially due to surfactants reducing transcript abundance of virulence factors. This work identifies a novel role for surfactants and suggests their importance in modulating the severity of S. aureus lung infections.
Collapse
Affiliation(s)
- Maria Predtechenskaya
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, Montana, USA
| | - Corbin J Arbizzani
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, Montana, USA
| | - Sofia R Shomento
- University of Washington, School of Medicine, Seattle, Washington, USA
| | - Timothy R Borgogna
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, Montana, USA
| | - Jovanka M Voyich
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, Montana, USA
| |
Collapse
|
5
|
Sinha S, Murmu B, Roy AK, Balgote PJ, Sivaraman J. Targeting SufC ATPase in Staphylococcus aureus AR465: Insights from an in silico and molecular docking approach. J Microbiol Methods 2025; 232-234:107134. [PMID: 40250768 DOI: 10.1016/j.mimet.2025.107134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 04/20/2025]
Abstract
Staphylococcus aureus AR465 (S. aureus AR465) is a deadly pathogen that often inherits multidrug resistance, where the antibiotics become ineffective against it. The iron‑sulfur (FeS) cluster assembly pathway has the potential to serve as a new drug target, allowing for the modification of these molecules to be susceptible to oxidative conditions. Our study focuses on the preliminary stage of the FeS pathway inhibition by inhibiting the SufC protein, unlike previous studies that targeted the final stage. SufC has an Adenosine triphosphate (ATP) binding site. The main goal of this study is to inhibit the SufBCD complex of S. aureus AR465 to bind with other subunits to form an FeS cluster. The Sulfur Utilization Factor (SUF) system plays a massive role in the survival of this pathogen by producing electron carrier proteins which possess FeS cofactors. The SufC protein from the SufBCD system was chosen as the main target for the potential inhibitor molecules. SufC is an ATP-binding cassette (ABC) that transfers an FeS cluster to SufA, which then transports it to an apoprotein involved in electron transport processes. In this research, several drugs were selected which can block this particular stage of the FeS cluster formation pathway. The idea was to competitively inhibit the binding of ATP with the help of inhibitors so that it cannot bind to the desired site of SufC. Eventually, the inhibitor molecule blocks the transfer of the FeS cluster to a newly synthesized apo-protein and kills the pathogen.
Collapse
Affiliation(s)
- Sounak Sinha
- Department of Applied Microbiology, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Birsing Murmu
- Department of Applied Microbiology, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Arya Ketan Roy
- Department of Applied Microbiology, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Piyush Jagdish Balgote
- Department of Biotechnology, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Jayanthi Sivaraman
- Department of Biotechnology, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India.
| |
Collapse
|
6
|
Savin A, Anderson EE, Dyzenhaus S, Podkowik M, Shopsin B, Pironti A, Torres VJ. Staphylococcus aureus senses human neutrophils via PerR to coordinate the expression of the toxin LukAB. Infect Immun 2024; 92:e0052623. [PMID: 38235972 PMCID: PMC10863418 DOI: 10.1128/iai.00526-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 01/19/2024] Open
Abstract
Staphylococcus aureus is a gram-positive pathogen that poses a major health concern, in part due to its large array of virulence factors that allow infection and evasion of the immune system. One of these virulence factors is the bicomponent pore-forming leukocidin LukAB. The regulation of lukAB expression is not completely understood, especially in the presence of immune cells such as human polymorphonuclear neutrophils (hPMNs). Here, we screened for transcriptional regulators of lukAB during the infection of primary hPMNs. We uncovered that PerR, a peroxide sensor, is vital for hPMN-mediated induction of lukAB and that PerR upregulates cytotoxicity during the infection of hPMNs. Exposure of S. aureus to hydrogen peroxide (H2O2) alone also results in increased lukAB promoter activity, a phenotype dependent on PerR. Collectively, our data suggest that S. aureus uses PerR to sense the H2O2 produced by hPMNs to stimulate the expression of lukAB, allowing the bacteria to withstand these critical innate immune cells.IMPORTANCEStaphylococcus aureus utilizes a diverse set of virulence factors, such as leukocidins, to subvert human neutrophils, but how these toxins are regulated is incompletely defined. Here, we identified the peroxide-sensitive repressor, PerR, as a required protein involved in the induction of lukAB in the presence of primary human neutrophils, a phenotype directly linked to the ability of PerR to sense H2O2. Thus, we show that S. aureus coordinates sensing and resistance to oxidative stress with toxin production to promote pathogen survival.
Collapse
Affiliation(s)
- Avital Savin
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Department of Biology, New York University, New York, New York, USA
| | - Exene E. Anderson
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Sophie Dyzenhaus
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Magdalena Podkowik
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, New York, USA
- Division of Infectious Diseases, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Bo Shopsin
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, New York, USA
- Division of Infectious Diseases, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Alejandro Pironti
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, New York, USA
| | - Victor J. Torres
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Department of Host-Microbe Interactions, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
7
|
Gao P, Wei Y, Hou S, Lai PM, Liu H, Tai SSC, Tang VYM, Prakash PH, Sze KH, Chen JHK, Sun H, Li X, Kao RYT. SaeR as a novel target for antivirulence therapy against Staphylococcus aureus. Emerg Microbes Infect 2023; 12:2254415. [PMID: 37671453 PMCID: PMC10494732 DOI: 10.1080/22221751.2023.2254415] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 08/27/2023] [Accepted: 08/28/2023] [Indexed: 09/07/2023]
Abstract
Staphylococcus aureus is a major human pathogen responsible for a wide range of clinical infections. SaeRS is one of the two-component systems in S. aureus that modulate multiple virulence factors. Although SaeR is required for S. aureus to develop an infection, inhibitors have not been reported. Using an in vivo knockdown method, we demonstrated that SaeR is targetable for the discovery of antivirulence agent. HR3744 was discovered through a high-throughput screening utilizing a GFP-Lux dual reporter system driven by saeP1 promoter. The antivirulence efficacy of HR3744 was tested using Western blot, Quantitative Polymerase Chain Reaction, leucotoxicity, and haemolysis tests. In electrophoresis mobility shift assay, HR3744 inhibited SaeR-DNA probe binding. WaterLOGSY-NMR test showed HR3744 directly interacted with SaeR's DNA-binding domain. When SaeR was deleted, HR3744 lost its antivirulence property, validating the target specificity. Virtual docking and mutagenesis were used to confirm the target's specificity. When Glu159 was changed to Asn, the bacteria developed resistance to HR3744. A structure-activity relationship study revealed that a molecule with a slight modification did not inhibit SaeR, indicating the selectivity of HR3744. Interestingly, we found that SAV13, an analogue of HR3744, was four times more potent than HR3744 and demonstrated identical antivirulence properties and target specificity. In a mouse bacteraemia model, both HR3744 and SAV13 exhibited in vivo effectiveness. Collectively, we identified the first SaeR inhibitor, which exhibited in vitro and in vivo antivirulence properties, and proved that SaeR could be a novel target for developing antivirulence drugs against S. aureus infections.
Collapse
Affiliation(s)
- Peng Gao
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Yuanxin Wei
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Suying Hou
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Pok-Man Lai
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Han Liu
- Morningside Laboratory for Chemical Biology and Department of Chemistry, The University of Hong Kong, Hong Kong, People’s Republic of China
| | - Sherlock Shing Chiu Tai
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Victor Yat Man Tang
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Pradeep Halebeedu Prakash
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Kong-Hung Sze
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Jonathan Hon Kwan Chen
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Hongzhe Sun
- Morningside Laboratory for Chemical Biology and Department of Chemistry, The University of Hong Kong, Hong Kong, People’s Republic of China
| | - Xuechen Li
- Morningside Laboratory for Chemical Biology and Department of Chemistry, The University of Hong Kong, Hong Kong, People’s Republic of China
| | - Richard Yi-Tsun Kao
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| |
Collapse
|
8
|
Arya R, Kim T, Youn JW, Bae T, Kim KK. Identification of an antivirulence agent targeting the master regulator of virulence genes in Staphylococcus aureus. Front Cell Infect Microbiol 2023; 13:1268044. [PMID: 38029271 PMCID: PMC10644738 DOI: 10.3389/fcimb.2023.1268044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 10/10/2023] [Indexed: 12/01/2023] Open
Abstract
The emergence of bactericidal antibiotic-resistant strains has increased the demand for alternative therapeutic agents, such as antivirulence agents targeting the virulence regulators of pathogens. Staphylococcus aureus exoprotein expression (sae) locus, the master regulator of virulence gene expression in multiple drug-resistant S. aureus, is a promising therapeutic target. In this study, we screened a small-molecule library using a SaeRS green fluorescent protein (GFP)-reporter that responded to transcription controlled by the sae locus. We identified the compound, N-(2-methylcyclohexyl)-11-oxo-10,11-dihydrodibenzo[b,f][1,4]thiazepine-8-carboxamide (SKKUCS), as an efficient repressor of sae-regulated GFP activity. SKKUCS inhibited hemolysin production and reduced α-hemolysin-mediated cell lysis. Moreover, SKKUCS substantially reduced the expression levels of various virulence genes controlled by the master regulators, sae, and the accessory gene regulator (agr), demonstrating its potential as an antivirulence reagent targeting the key virulence regulators. Furthermore, autokinase inhibition assay and molecular docking suggest that SKKUCS inhibits the kinase activity of SaeS and potentially targets the active site of SaeS kinase, possibly inhibiting ATP binding. Next, we evaluated the efficacy and toxicity of SKKUCS in vivo using murine models of staphylococcal intraperitoneal and skin infections. Treatment with SKKUCS markedly increased animal survival and significantly decreased the bacterial burden in organs and skin lesion sizes. These findings highlight SKKUCS as a potential antivirulence drug for drug-resistant staphylococcal infections.
Collapse
Affiliation(s)
- Rekha Arya
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Institute of Antibacterial Resistance Research and Therapeutics, Suwon, Republic of Korea
- Department of Orthopedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Truc Kim
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Institute of Antibacterial Resistance Research and Therapeutics, Suwon, Republic of Korea
| | - Joo Won Youn
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Institute of Antibacterial Resistance Research and Therapeutics, Suwon, Republic of Korea
| | - Taeok Bae
- Department of Microbiology and Immunology, Indiana University School of Medicine-Northwest, Gary, IN, United States
| | - Kyeong Kyu Kim
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Institute of Antibacterial Resistance Research and Therapeutics, Suwon, Republic of Korea
| |
Collapse
|
9
|
Pivard M, Caldelari I, Brun V, Croisier D, Jaquinod M, Anzala N, Gilquin B, Teixeira C, Benito Y, Couzon F, Romby P, Moreau K, Vandenesch F. Complex Regulation of Gamma-Hemolysin Expression Impacts Staphylococcus aureus Virulence. Microbiol Spectr 2023; 11:e0107323. [PMID: 37347186 PMCID: PMC10434192 DOI: 10.1128/spectrum.01073-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 05/25/2023] [Indexed: 06/23/2023] Open
Abstract
Staphylococcus aureus gamma-hemolysin CB (HlgCB) is a core-genome-encoded pore-forming toxin that targets the C5a receptor, similar to the phage-encoded Panton-Valentine leucocidin (PVL). Absolute quantification by mass spectrometry of HlgCB in 39 community-acquired pneumonia (CAP) isolates showed considerable variations in the HlgC and HlgB yields between isolates. Moreover, although HlgC and HlgB are encoded on a single operon, their levels were dissociated in 10% of the clinical strains studied. To decipher the molecular basis for the variation in hlgCB expression and protein production among strains, different regulation levels were analyzed in representative clinical isolates and reference strains. Both the HlgCB level and the HlgC/HlgB ratio were found to depend on hlgC promoter activity and mRNA processing and translation. Strikingly, only one single nucleotide polymorphism (SNP) in the 5' untranslated region (UTR) of hlgCB mRNA strongly impaired hlgC translation in the USA300 strain, leading to a strong decrease in the level of HlgC but not in HlgB. Finally, we found that high levels of HlgCB synthesis led to mortality in a rabbit model of pneumonia, correlated with the implication of the role of HlgCB in severe S. aureus CAP. Taken together, this work illustrates the complexity of virulence factor expression in clinical strains and demonstrates a butterfly effect where subtle genomic variations have a major impact on phenotype and virulence. IMPORTANCE S. aureus virulence in pneumonia results in its ability to produce several virulence factors, including the leucocidin PVL. Here, we demonstrate that HlgCB, another leucocidin, which targets the same receptors as PVL, highly contributes to S. aureus virulence in pvl-negative strains. In addition, considerable variations in HlgCB quantities are observed among clinical isolates from patients with CAP. Biomolecular analyses have revealed that a few SNPs in the promoter sequences and only one SNP in the 5' UTR of hlgCB mRNA induce the differential expression of hlgCB, drastically impacting hlgC mRNA translation. This work illustrates the subtlety of regulatory mechanisms in bacteria, especially the sometimes major effects on phenotypes of single nucleotide variation in noncoding regions.
Collapse
Affiliation(s)
- Mariane Pivard
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS UMR5308, ENS de Lyon, Lyon, France
| | - Isabelle Caldelari
- Architecture et Réactivité de l’ARN, Université de Strasbourg, CNRS, IBMC, Strasbourg, France
| | - Virginie Brun
- Université Grenoble Alpes, Inserm, CEA, UMR BioSanté U1292, CNRS, CEA, Grenoble, France
- Université Grenoble Alpes, CEA, LETI, Clinatec, Grenoble, France
| | | | - Michel Jaquinod
- Université Grenoble Alpes, Inserm, CEA, UMR BioSanté U1292, CNRS, CEA, Grenoble, France
| | | | - Benoît Gilquin
- Université Grenoble Alpes, Inserm, CEA, UMR BioSanté U1292, CNRS, CEA, Grenoble, France
- Université Grenoble Alpes, CEA, LETI, Clinatec, Grenoble, France
| | - Chloé Teixeira
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS UMR5308, ENS de Lyon, Lyon, France
| | - Yvonne Benito
- Institut des Agents Infectieux, Hospices Civils de Lyon, Lyon, France
- Centre National de Référence des Staphylocoques, Hospices Civils de Lyon, Lyon, France
| | - Florence Couzon
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS UMR5308, ENS de Lyon, Lyon, France
| | - Pascale Romby
- Architecture et Réactivité de l’ARN, Université de Strasbourg, CNRS, IBMC, Strasbourg, France
| | - Karen Moreau
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS UMR5308, ENS de Lyon, Lyon, France
| | - François Vandenesch
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS UMR5308, ENS de Lyon, Lyon, France
- Institut des Agents Infectieux, Hospices Civils de Lyon, Lyon, France
- Centre National de Référence des Staphylocoques, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
10
|
Patel H, Rawat S. A genetic regulatory see-saw of biofilm and virulence in MRSA pathogenesis. Front Microbiol 2023; 14:1204428. [PMID: 37434702 PMCID: PMC10332168 DOI: 10.3389/fmicb.2023.1204428] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 05/30/2023] [Indexed: 07/13/2023] Open
Abstract
Staphylococcus aureus is one of the most common opportunistic human pathogens causing several infectious diseases. Ever since the emergence of the first methicillin-resistant Staphylococcus aureus (MRSA) strain decades back, the organism has been a major cause of hospital-acquired infections (HA-MRSA). The spread of this pathogen across the community led to the emergence of a more virulent subtype of the strain, i.e., Community acquired Methicillin resistant Staphylococcus aureus (CA-MRSA). Hence, WHO has declared Staphylococcus aureus as a high-priority pathogen. MRSA pathogenesis is remarkable because of the ability of this "superbug" to form robust biofilm both in vivo and in vitro by the formation of polysaccharide intercellular adhesin (PIA), extracellular DNA (eDNA), wall teichoic acids (WTAs), and capsule (CP), which are major components that impart stability to a biofilm. On the other hand, secretion of a diverse array of virulence factors such as hemolysins, leukotoxins, enterotoxins, and Protein A regulated by agr and sae two-component systems (TCS) aids in combating host immune response. The up- and downregulation of adhesion genes involved in biofilm formation and genes responsible for synthesizing virulence factors during different stages of infection act as a genetic regulatory see-saw in the pathogenesis of MRSA. This review provides insight into the evolution and pathogenesis of MRSA infections with a focus on genetic regulation of biofilm formation and virulence factors secretion.
Collapse
Affiliation(s)
| | - Seema Rawat
- Microbiology Laboratory, School of Life Sciences, Central University of Gujarat, Gandhinagar, Gujarat, India
| |
Collapse
|
11
|
Wittekind MA, Briaud P, Smith JL, Tennant JR, Carroll RK. The Small Protein ScrA Influences Staphylococcus aureus Virulence-Related Processes via the SaeRS System. Microbiol Spectr 2023; 11:e0525522. [PMID: 37154710 PMCID: PMC10269730 DOI: 10.1128/spectrum.05255-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 04/11/2023] [Indexed: 05/10/2023] Open
Abstract
Staphylococcus aureus is a Gram-positive commensal and opportunistic pathogen able to cause diseases ranging from mild skin infections to life-threatening endocarditis and toxic shock syndrome. The ability to cause such an array of diseases is due to the complex S. aureus regulatory network controlling an assortment of virulence factors, including adhesins, hemolysins, proteases, and lipases. This regulatory network is controlled by both protein and RNA elements. We previously identified a novel regulatory protein called ScrA, which, when overexpressed, leads to the increased activity and expression of the SaeRS regulon. In this study, we further explore the role of ScrA and examine the consequences to the bacterial cell of scrA gene disruption. These results demonstrate that scrA is required for several virulence-related processes, and in many cases, the phenotypes of the scrA mutant are inverse to those observed in cells overexpressing ScrA. Interestingly, while the majority of ScrA-mediated phenotypes appear to rely on the SaeRS system, our results also indicate that ScrA may also act independently of SaeRS when regulating hemolytic activity. Finally, using a murine model of infection, we demonstrate that scrA is required for virulence, potentially in an organ-specific manner. IMPORTANCE Staphylococcus aureus is the cause of several potentially life-threatening infections. An assortment of toxins and virulence factors allows such a wide range of infections. However, an assortment of toxins or virulence factors requires complex regulation to control expression under all of the different conditions encountered by the bacterium. Understanding the intricate web of regulatory systems allows the development of novel approaches to combat S. aureus infections. Here, we have shown that the small protein ScrA, which was previously identified by our laboratory, influences several virulence-related functions through the SaeRS global regulatory system. These findings add ScrA to the growing list of virulence regulators in S. aureus.
Collapse
Affiliation(s)
| | - Paul Briaud
- Department of Biological Sciences, Ohio University, Athens, Ohio, USA
| | - Jayanna L. Smith
- Department of Biological Sciences, Ohio University, Athens, Ohio, USA
| | - Julia R. Tennant
- Department of Biological Sciences, Ohio University, Athens, Ohio, USA
| | - Ronan K. Carroll
- Department of Biological Sciences, Ohio University, Athens, Ohio, USA
| |
Collapse
|
12
|
Langouët-Astrié C, Oshima K, McMurtry SA, Yang Y, Kwiecinski JM, LaRivière WB, Kavanaugh JS, Zakharevich I, Hansen KC, Shi D, Zhang F, Boguslawski KM, Perelman SS, Su G, Torres VJ, Liu J, Horswill AR, Schmidt EP. The influenza-injured lung microenvironment promotes MRSA virulence, contributing to severe secondary bacterial pneumonia. Cell Rep 2022; 41:111721. [PMID: 36450248 PMCID: PMC10082619 DOI: 10.1016/j.celrep.2022.111721] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 10/12/2022] [Accepted: 11/03/2022] [Indexed: 12/03/2022] Open
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.
Collapse
Affiliation(s)
| | - Kaori Oshima
- Division of Pulmonary Sciences and Critical Care, University of Colorado Denver, Aurora, CO 80045, USA
| | - Sarah A McMurtry
- Division of Pulmonary Sciences and Critical Care, University of Colorado Denver, Aurora, CO 80045, USA
| | - Yimu Yang
- Division of Pulmonary Sciences and Critical Care, University of Colorado Denver, Aurora, CO 80045, USA
| | - Jakub M Kwiecinski
- Department of Immunology & Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA; Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow 30387, Poland
| | - Wells B LaRivière
- Division of Pulmonary Sciences and Critical Care, University of Colorado Denver, Aurora, CO 80045, USA; Medical Scientist Training Program, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Jeffrey S Kavanaugh
- Department of Immunology & Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Igor Zakharevich
- Department of Biochemistry and Molecular Genetics, University of Colorado, Aurora, CO 80045, USA
| | - Kirk C Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado, Aurora, CO 80045, USA
| | - Deling Shi
- Department of Chemistry, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Fuming Zhang
- Department of Chemistry, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Kristina M Boguslawski
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Sofya S Perelman
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Gouwei Su
- University of North Carolina Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA
| | - Victor J Torres
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Jian Liu
- University of North Carolina Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA
| | - Alexander R Horswill
- Department of Immunology & Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Eric P Schmidt
- Division of Pulmonary Sciences and Critical Care, University of Colorado Denver, Aurora, CO 80045, USA; Department of Medicine, Massachusetts General Hospital, Boston, MA 02115, USA
| |
Collapse
|
13
|
Dou BB, Yang X, Yang FM, Yan K, Peng W, Tang J, Peng MZ, He QY, Chen HC, Yuan FY, Bei WC. The VraSR two-component signal transduction system contributes to the damage of blood-brain barrier during Streptococcus suis meningitis. Microb Pathog 2022; 172:105766. [PMID: 36087689 DOI: 10.1016/j.micpath.2022.105766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 10/31/2022]
Abstract
Streptococcus suis (S. suis) is an important zoonotic pathogen that can cause high morbidity and mortality in both humans and swine. As the most important life-threatening infection of the central nervous system (CNS), meningitis is an important syndrome of S. suis infection. The vancomycin resistance associated sensor/regulator (VraSR) is a critical two-component signal transduction system that affects the ability of S. suis to resist the host innate immune system and promotes its ability to adhere to brain microvascular endothelial cells (BMECs). Prior work also found mice infected with ΔvraSR had no obvious neurological symptoms, unlike mice infected with wild-type SC19. Whether and how VraSR participates in the development of S. suis meningitis remains unknown. Here, we found ΔvraSR-infected mice did not show obvious meningitis, compared with wild-type SC19-infected mice. Moreover, the proinflammatory cytokines and chemokines in serum and brains of ΔvraSR-infected mice, including IL-6, TNF-α, MCP-1 and IFN-γ, were significantly lower than wild-type infected group. Besides, blood-brain barrier (BBB) permeability also confirmed that the mutant had lower ability to disrupt BBB. Furthermore, in vivo and in vitro experiments showed that SC19 could increase BBB permeability by downregulating tight junction (TJ) proteins such as ZO-1, β-Catenin, Occludin, and Clauidn-5, compared with mutant ΔvraSR. These findings provide new insight into the influence of S. suis VraSR on BBB disruption during the pathogenic process of streptococcal meningitis, thereby offering potential targets for future preventative and therapeutic strategies against this disease.
Collapse
Affiliation(s)
- Bei-Bei Dou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Xia Yang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Feng-Ming Yang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Kang Yan
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Wei Peng
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Jia Tang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Ming-Zheng Peng
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Qi-Yun He
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Huan-Chun Chen
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; Hubei Hongshan Laboratory, Wuhan, Hubei, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; Guangxi Yangxiang Co., Ltd., Guangxi, 530015, China
| | - Fang-Yan Yuan
- Hubei Key Laboratory of Animal Embryo and Molecular Breeding, Institute of Animal Husbandry and Veterinary Sciences, Hubei Academy of Agricultural Sciences, Wuhan, 430064, China
| | - Wei-Cheng Bei
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; Hubei Hongshan Laboratory, Wuhan, Hubei, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; Guangxi Yangxiang Co., Ltd., Guangxi, 530015, China.
| |
Collapse
|
14
|
R. Borgogna T, M. Voyich J. Examining the Executioners, Influenza Associated Secondary Bacterial Pneumonia. Infect Dis (Lond) 2022. [DOI: 10.5772/intechopen.101666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Influenza infections typically present mild to moderate morbidities in immunocompetent host and are often resolved within 14 days of infection onset. Death from influenza infection alone is uncommon; however, antecedent influenza infection often leads to an increased susceptibility to secondary bacterial pneumonia. Bacterial pneumonia following viral infection exhibits mortality rates greater than 10-fold of those of influenza alone. Furthermore, bacterial pneumonia has been identified as the major contributor to mortality during each of the previous four influenza pandemics. Streptococcus pneumoniae, Staphylococcus aureus, Haemophilus influenzae, and Streptococcus pyogenes are the most prevalent participants in this pathology. Of note, these lung pathogens are frequently found as commensals of the upper respiratory tract. Herein we describe influenza-induced host-changes that lead to increased susceptibility to bacterial pneumonia, review virulence strategies employed by the most prevalent secondary bacterial pneumonia species, and highlight recent findings of bacterial sensing and responding to the influenza infected environment.
Collapse
|
15
|
Jain S, Bhowmick A, Jeong B, Bae T, Ghosh A. Unravelling the physiological roles of mazEF toxin-antitoxin system on clinical MRSA strain by CRISPR RNA-guided cytidine deaminase. J Biomed Sci 2022; 29:28. [PMID: 35524246 PMCID: PMC9077811 DOI: 10.1186/s12929-022-00810-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 04/22/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Curiosity on toxin-antitoxin modules has increased intensely over recent years as it is ubiquitously present in many bacterial genomes, including pathogens like Methicillin-resistant Staphylococcus aureus (MRSA). Several cellular functions of TA systems have been proposed however, their exact role in cellular physiology remains unresolved. METHODS This study aims to find out the impact of the mazEF toxin-antitoxin module on biofilm formation, pathogenesis, and antibiotic resistance in an isolated clinical ST239 MRSA strain, by constructing mazE and mazF mutants using CRISPR-cas9 base-editing plasmid (pnCasSA-BEC). Transcriptome analysis (RNA-seq) was performed for the mazE antitoxin mutant in order to identify the differentially regulated genes. The biofilm formation was also assessed for the mutant strains. Antibiogram profiling was carried out for both the generated mutants followed by murine experiment to determine the pathogenicity of the constructed strains. RESULTS For the first time our work showed, that MazF promotes cidA mediated cell death and lysis for biofilm formation without playing any significant role in host virulence as suggested by the murine experiment. Interestingly, the susceptibility to oxacillin, daptomycin and vancomycin was reduced significantly by the activated MazF toxin in the mazE mutant strain. CONCLUSIONS Our study reveals that activated MazF toxin leads to resistance to antibiotics like oxacillin, daptomycin and vancomycin. Therefore, in the future, any potential antibacterial drug can be designed to target MazF toxin against the problematic multi-drug resistant bug.
Collapse
Affiliation(s)
- Sonia Jain
- Infectious Disease and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, 700032, India.
| | - Arghya Bhowmick
- Department of Biochemistry, Bose Institute, EN Block, Sector-V, Kolkata, 700091, India
| | - Bohyun Jeong
- Department of Microbiology, Kosin University College of Medicine, Busan, 49267, South Korea
| | - Taeok Bae
- Department of Microbiology and Immunology, Indiana University, School of Medicine-Northwest, Gary, IN, 46408-1197, USA
| | - Abhrajyoti Ghosh
- Department of Biochemistry, Bose Institute, EN Block, Sector-V, Kolkata, 700091, India.
| |
Collapse
|
16
|
Pulia MS, Anderson J, Ye Z, Elsayed NS, Le T, Patitucci J, Ganta K, Hall M, Singh VK, Shukla SK. Expression of Staphylococcal Virulence Genes In Situ in Human Skin and Soft Tissue Infections. Antibiotics (Basel) 2022; 11:527. [PMID: 35453277 PMCID: PMC9032627 DOI: 10.3390/antibiotics11040527] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/08/2022] [Accepted: 04/10/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Staphylococcus aureus, the most common pathogen in skin and soft tissue infections (SSTI), harbors many well-characterized virulence genes. However, the expression of many of them in SSTIs is unknown. In this study, S. aureus virulence genes expressed in SSTI were investigated. METHODS Fifty-three subjects presenting to the outpatient's care and emergency departments with a purulent SSTI at two medical centers in Wisconsin, USA, were enrolled in the study. Total mRNA was extracted from the purulent or swab materials, made into cDNA and sequenced on MiSeq platform. The relative cDNA counts to gmk and identifications of the transcripts were carried out with respect to USA300 reference genome and using SAMTOOLS v.1.3 and BWA, respectively. RESULT A significantly higher cDNA count was observed for many of the virulence and regulatory gene transcripts in the pus samples compared to the swab samples relative to the cDNA counts for gmk, a housekeeping gene. They were for lukS-PV (18.6 vs. 14.2), isaA (13.4 vs. 8.5), ssaA (4.8 vs. 3.1), hlgC (1.4 vs. 1.33), atl (17.7 vs. 8.33), clfA (3.9 vs. 0.83), eno (6.04 vs. 3.16), fnbA (5.93 vs. 0.33), saeS (6.3 vs. 1.33), saeR (5.4 vs. 3.33) and agrC (5.6 vs. 1.5). CONCLUSIONS A relative increase in the transcripts of several toxins, adhesion and regulatory genes with respect to a gmk in purulent materials suggests their role in situ during SSTIs, perhaps in an orchestrated manner.
Collapse
Affiliation(s)
- Michael S. Pulia
- Department of Emergency Medicine, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53726, USA;
| | - Jennifer Anderson
- Integrated Research Development Center, Marshfield Clinic Research Institute, Marshfield, WI 54449, USA; (J.A.); (T.L.)
| | - Zhan Ye
- Bioinformatics Research Center, Marshfield Clinic Research Institute, Marshfield, WI 54449, USA; (Z.Y.); (J.P.)
| | - Noha S. Elsayed
- Center for Precision Medicine Research, Marshfield Clinic Research Institute, Marshfield, WI 54449, USA; (N.S.E.); (K.G.)
| | - Thao Le
- Integrated Research Development Center, Marshfield Clinic Research Institute, Marshfield, WI 54449, USA; (J.A.); (T.L.)
| | - Jacob Patitucci
- Bioinformatics Research Center, Marshfield Clinic Research Institute, Marshfield, WI 54449, USA; (Z.Y.); (J.P.)
| | - Krishna Ganta
- Center for Precision Medicine Research, Marshfield Clinic Research Institute, Marshfield, WI 54449, USA; (N.S.E.); (K.G.)
| | - Matthew Hall
- Department of Infectious Diseases, Marshfield Clinic Health System, Marshfield, WI 54449, USA;
| | - Vineet K. Singh
- Department of Microbiology and Immunology, Kirksville College of Osteopathic Medicine, A.T. Still, University of Health Sciences, Kirksville, MO 63501, USA;
| | - Sanjay K. Shukla
- Center for Precision Medicine Research, Marshfield Clinic Research Institute, Marshfield, WI 54449, USA; (N.S.E.); (K.G.)
| |
Collapse
|
17
|
Guo H, Tong Y, Cheng J, Abbas Z, Li Z, Wang J, Zhou Y, Si D, Zhang R. Biofilm and Small Colony Variants-An Update on Staphylococcus aureus Strategies toward Drug Resistance. Int J Mol Sci 2022; 23:ijms23031241. [PMID: 35163165 PMCID: PMC8835882 DOI: 10.3390/ijms23031241] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/16/2022] [Accepted: 01/20/2022] [Indexed: 02/07/2023] Open
Abstract
Recently, the drawbacks arising from the overuse of antibiotics have drawn growing public attention. Among them, drug-resistance (DR) and even multidrug-resistance (MDR) pose significant challenges in clinical practice. As a representative of a DR or MDR pathogen, Staphylococcus aureus can cause diversity of infections related to different organs, and can survive or adapt to the diverse hostile environments by switching into other phenotypes, including biofilm and small colony variants (SCVs), with altered physiologic or metabolic characteristics. In this review, we briefly describe the development of the DR/MDR as well as the classical mechanisms (accumulation of the resistant genes). Moreover, we use multidimensional scaling analysis to evaluate the MDR relevant hotspots in the recent published reports. Furthermore, we mainly focus on the possible non-classical resistance mechanisms triggered by the two important alternative phenotypes of the S. aureus, biofilm and SCVs, which are fundamentally caused by the different global regulation of the S. aureus population, such as the main quorum-sensing (QS) and agr system and its coordinated regulated factors, such as the SarA family proteins and the alternative sigma factor σB (SigB). Both the biofilm and the SCVs are able to escape from the host immune response, and resist the therapeutic effects of antibiotics through the physical or the biological barriers, and become less sensitive to some antibiotics by the dormant state with the limited metabolisms.
Collapse
|
18
|
Bleul L, Francois P, Wolz C. Two-Component Systems of S. aureus: Signaling and Sensing Mechanisms. Genes (Basel) 2021; 13:34. [PMID: 35052374 PMCID: PMC8774646 DOI: 10.3390/genes13010034] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 12/21/2021] [Accepted: 12/22/2021] [Indexed: 12/18/2022] Open
Abstract
Staphylococcus aureus encodes 16 two-component systems (TCSs) that enable the bacteria to sense and respond to changing environmental conditions. Considering the function of these TCSs in bacterial survival and their potential role as drug targets, it is important to understand the exact mechanisms underlying signal perception. The differences between the sensing of appropriate signals and the transcriptional activation of the TCS system are often not well described, and the signaling mechanisms are only partially understood. Here, we review present insights into which signals are sensed by histidine kinases in S. aureus to promote appropriate gene expression in response to diverse environmental challenges.
Collapse
Affiliation(s)
- Lisa Bleul
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, 72076 Tubingen, Germany;
- Cluster of Excellence EXC 2124 “Controlling Microbes to Fight Infections”, University of Tübingen, Elfriede-Aulhorn-Str. 6, 72076 Tubingen, Germany
| | - Patrice Francois
- Genomic Research Laboratory, Infectious Diseases Service, University Hospitals of Geneva University Medical Center, Michel Servet 1, CH-1211 Geneva, Switzerland;
| | - Christiane Wolz
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, 72076 Tubingen, Germany;
- Cluster of Excellence EXC 2124 “Controlling Microbes to Fight Infections”, University of Tübingen, Elfriede-Aulhorn-Str. 6, 72076 Tubingen, Germany
| |
Collapse
|
19
|
DeMars ZR, Krute CN, Ridder MJ, Gilchrist AK, Menjivar C, Bose JL. Fatty acids can inhibit Staphylococcus aureus SaeS activity at the membrane independent of alterations in respiration. Mol Microbiol 2021; 116:1378-1391. [PMID: 34626146 DOI: 10.1111/mmi.14830] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/06/2021] [Accepted: 10/06/2021] [Indexed: 12/29/2022]
Abstract
In Staphylococcus aureus, the two-component system SaeRS is responsible for regulating various virulence factors essential for the success of this pathogen. SaeRS can be stimulated by neutrophil-derived products but has also recently been shown to be inactivated by the presence of free fatty acids. A mechanism for how fatty acids negatively impacts SaeRS has not been described. We found that unsaturated fatty acids, as well as fatty acids not commonly found in Staphylococcal membranes, prevent the activation of SaeRS at a lower concentration than their saturated counterparts. These fatty acids can negatively impact SaeRS without altering the respiratory capacity of the bacterium. To uncover a potential mechanism for how fatty acids impact SaeRS function/activity, we utilized a naturally occurring point mutation found in S. aureus as well as chimeric SaeS proteins. Using these tools, we identified that the native transmembrane domains of SaeS dictate the transcriptional response to fatty acids in S. aureus. Our data support a model where free fatty acids alter the activity of the two-component system SaeRS directly through the sensor kinase SaeS and is dependent on the transmembrane domains of the protein.
Collapse
Affiliation(s)
- Zachary R DeMars
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Christina N Krute
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Miranda J Ridder
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Aubrey K Gilchrist
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Cindy Menjivar
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Jeffrey L Bose
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
20
|
Kretschmer D, Breitmeyer R, Gekeler C, Lebtig M, Schlatterer K, Nega M, Stahl M, Stapels D, Rooijakkers S, Peschel A. Staphylococcus aureus Depends on Eap Proteins for Preventing Degradation of Its Phenol-Soluble Modulin Toxins by Neutrophil Serine Proteases. Front Immunol 2021; 12:701093. [PMID: 34552584 PMCID: PMC8451722 DOI: 10.3389/fimmu.2021.701093] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 08/18/2021] [Indexed: 11/13/2022] Open
Abstract
Neutrophil granulocytes act as a first line of defense against pathogenic staphylococci. However, Staphylococcus aureus has a remarkable capacity to survive neutrophil killing, which distinguishes it from the less-pathogenic Staphylococcus epidermidis. Both species release phenol-soluble modulin (PSM) toxins, which activate the neutrophil formyl-peptide receptor 2 (FPR2) to promote neutrophil influx and phagocytosis, and which disrupt neutrophils or their phagosomal membranes at high concentrations. We show here that the neutrophil serine proteases (NSPs) neutrophil elastase, cathepsin G and proteinase 3, which are released into the extracellular space or the phagosome upon neutrophil FPR2 stimulation, effectively degrade PSMs thereby preventing their capacity to activate and destroy neutrophils. Notably, S. aureus, but not S. epidermidis, secretes potent NSP-inhibitory proteins, Eap, EapH1, EapH2, which prevented the degradation of PSMs by NSPs. Accordingly, a S. aureus mutant lacking all three NSP inhibitory proteins was less effective in activating and destroying neutrophils and it survived less well in the presence of neutrophils than the parental strain. We show that Eap proteins promote pathology via PSM-mediated FPR2 activation since murine intraperitoneal infection with the S. aureus parental but not with the NSP inhibitors mutant strain, led to a significantly higher bacterial load in the peritoneum and kidneys of mFpr2-/- compared to wild-type mice. These data demonstrate that NSPs can very effectively detoxify some of the most potent staphylococcal toxins and that the prominent human pathogen S. aureus has developed efficient inhibitors to preserve PSM functions. Preventing PSM degradation during infection represents an important survival strategy to ensure FPR2 activation.
Collapse
Affiliation(s)
- Dorothee Kretschmer
- Interfaculty Institute for Microbiology and Infection Medicine Tübingen (IMIT), Infection Biology, University of Tübingen, Tübingen, Germany.,German Center for Infection Research, Partner Site Tübingen, Tübingen, Germany.,Cluster of Excellence EXC2124 "Controlling Microbes to Fight Infections", Tübingen, Germany
| | - Ricarda Breitmeyer
- Interfaculty Institute for Microbiology and Infection Medicine Tübingen (IMIT), Infection Biology, University of Tübingen, Tübingen, Germany.,German Center for Infection Research, Partner Site Tübingen, Tübingen, Germany.,Cluster of Excellence EXC2124 "Controlling Microbes to Fight Infections", Tübingen, Germany
| | - Cordula Gekeler
- Interfaculty Institute for Microbiology and Infection Medicine Tübingen (IMIT), Infection Biology, University of Tübingen, Tübingen, Germany.,German Center for Infection Research, Partner Site Tübingen, Tübingen, Germany.,Cluster of Excellence EXC2124 "Controlling Microbes to Fight Infections", Tübingen, Germany
| | - Marco Lebtig
- Interfaculty Institute for Microbiology and Infection Medicine Tübingen (IMIT), Infection Biology, University of Tübingen, Tübingen, Germany.,German Center for Infection Research, Partner Site Tübingen, Tübingen, Germany.,Cluster of Excellence EXC2124 "Controlling Microbes to Fight Infections", Tübingen, Germany
| | - Katja Schlatterer
- Interfaculty Institute for Microbiology and Infection Medicine Tübingen (IMIT), Infection Biology, University of Tübingen, Tübingen, Germany.,German Center for Infection Research, Partner Site Tübingen, Tübingen, Germany.,Cluster of Excellence EXC2124 "Controlling Microbes to Fight Infections", Tübingen, Germany
| | - Mulugeta Nega
- Cluster of Excellence EXC2124 "Controlling Microbes to Fight Infections", Tübingen, Germany.,Interfaculty Institute for Microbiology and Infection Medicine Tübingen (IMIT), Microbial Genetics, University of Tübingen, Tübingen, Germany
| | - Mark Stahl
- Center for Plant Molecular Biology (ZMBP), University of Tübingen, Tübingen, Germany
| | - Daphne Stapels
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Suzan Rooijakkers
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Andreas Peschel
- Interfaculty Institute for Microbiology and Infection Medicine Tübingen (IMIT), Infection Biology, University of Tübingen, Tübingen, Germany.,German Center for Infection Research, Partner Site Tübingen, Tübingen, Germany.,Cluster of Excellence EXC2124 "Controlling Microbes to Fight Infections", Tübingen, Germany
| |
Collapse
|
21
|
Schilcher K, Horswill AR. Staphylococcal Biofilm Development: Structure, Regulation, and Treatment Strategies. Microbiol Mol Biol Rev 2020; 84:e00026-19. [PMID: 32792334 PMCID: PMC7430342 DOI: 10.1128/mmbr.00026-19] [Citation(s) in RCA: 380] [Impact Index Per Article: 76.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In many natural and clinical settings, bacteria are associated with some type of biotic or abiotic surface that enables them to form biofilms, a multicellular lifestyle with bacteria embedded in an extracellular matrix. Staphylococcus aureus and Staphylococcus epidermidis, the most frequent causes of biofilm-associated infections on indwelling medical devices, can switch between an existence as single free-floating cells and multicellular biofilms. During biofilm formation, cells first attach to a surface and then multiply to form microcolonies. They subsequently produce the extracellular matrix, a hallmark of biofilm formation, which consists of polysaccharides, proteins, and extracellular DNA. After biofilm maturation into three-dimensional structures, the biofilm community undergoes a disassembly process that leads to the dissemination of staphylococcal cells. As biofilms are dynamic and complex biological systems, staphylococci have evolved a vast network of regulatory mechanisms to modify and fine-tune biofilm development upon changes in environmental conditions. Thus, biofilm formation is used as a strategy for survival and persistence in the human host and can serve as a reservoir for spreading to new infection sites. Moreover, staphylococcal biofilms provide enhanced resilience toward antibiotics and the immune response and impose remarkable therapeutic challenges in clinics worldwide. This review provides an overview and an updated perspective on staphylococcal biofilms, describing the characteristic features of biofilm formation, the structural and functional properties of the biofilm matrix, and the most important mechanisms involved in the regulation of staphylococcal biofilm formation. Finally, we highlight promising strategies and technologies, including multitargeted or combinational therapies, to eradicate staphylococcal biofilms.
Collapse
Affiliation(s)
- Katrin Schilcher
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Alexander R Horswill
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Veterans Affairs Eastern Colorado Health Care System, Denver, Colorado, USA
| |
Collapse
|
22
|
Schilcher K, Horswill AR. Staphylococcal Biofilm Development: Structure, Regulation, and Treatment Strategies. Microbiol Mol Biol Rev 2020. [PMID: 32792334 DOI: 10.1128/mmbr.00026-19/asset/e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2023] Open
Abstract
In many natural and clinical settings, bacteria are associated with some type of biotic or abiotic surface that enables them to form biofilms, a multicellular lifestyle with bacteria embedded in an extracellular matrix. Staphylococcus aureus and Staphylococcus epidermidis, the most frequent causes of biofilm-associated infections on indwelling medical devices, can switch between an existence as single free-floating cells and multicellular biofilms. During biofilm formation, cells first attach to a surface and then multiply to form microcolonies. They subsequently produce the extracellular matrix, a hallmark of biofilm formation, which consists of polysaccharides, proteins, and extracellular DNA. After biofilm maturation into three-dimensional structures, the biofilm community undergoes a disassembly process that leads to the dissemination of staphylococcal cells. As biofilms are dynamic and complex biological systems, staphylococci have evolved a vast network of regulatory mechanisms to modify and fine-tune biofilm development upon changes in environmental conditions. Thus, biofilm formation is used as a strategy for survival and persistence in the human host and can serve as a reservoir for spreading to new infection sites. Moreover, staphylococcal biofilms provide enhanced resilience toward antibiotics and the immune response and impose remarkable therapeutic challenges in clinics worldwide. This review provides an overview and an updated perspective on staphylococcal biofilms, describing the characteristic features of biofilm formation, the structural and functional properties of the biofilm matrix, and the most important mechanisms involved in the regulation of staphylococcal biofilm formation. Finally, we highlight promising strategies and technologies, including multitargeted or combinational therapies, to eradicate staphylococcal biofilms.
Collapse
Affiliation(s)
- Katrin Schilcher
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Alexander R Horswill
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Veterans Affairs Eastern Colorado Health Care System, Denver, Colorado, USA
| |
Collapse
|
23
|
De Gregorio E, Esposito A, Vollaro A, De Fenza M, D’Alonzo D, Migliaccio A, Iula VD, Zarrilli R, Guaragna A. N-Nonyloxypentyl-l-Deoxynojirimycin Inhibits Growth, Biofilm Formation and Virulence Factors Expression of Staphylococcus aureus. Antibiotics (Basel) 2020; 9:E362. [PMID: 32604791 PMCID: PMC7344813 DOI: 10.3390/antibiotics9060362] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/15/2020] [Accepted: 06/24/2020] [Indexed: 12/14/2022] Open
Abstract
Staphylococcus aureus is one of the major causes of hospital- and community-associated bacterial infections throughout the world, which are difficult to treat due to the rising number of drug-resistant strains. New molecules displaying potent activity against this bacterium are urgently needed. In this study, d- and l-deoxynojirimycin (DNJ) and a small library of their N-alkyl derivatives were screened against S. aureus ATCC 29213, with the aim to identify novel candidates with inhibitory potential. Among them, N-nonyloxypentyl-l-DNJ (l-NPDNJ) proved to be the most active compound against S. aureus ATCC 29213 and its clinical isolates, with the minimum inhibitory concentration (MIC) value of 128 μg/mL. l-NPDNJ also displayed an additive effect with gentamicin and oxacillin against the gentamicin- and methicillin-resistant S. aureus isolate 00717. Sub-MIC values of l-NPDNJ affected S. aureus biofilm development in a dose-dependent manner, inducing a strong reduction in biofilm biomass. Moreover, real-time reverse transcriptase PCR analysis revealed that l-NPDNJ effectively inhibited at sub-MIC values the transcription of the spa, hla, hlb and sea virulence genes, as well as the agrA and saeR response regulator genes.
Collapse
Affiliation(s)
- Eliana De Gregorio
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy;
| | - Anna Esposito
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia, 80126 Naples, Italy; (A.E.); (M.D.F.); (D.D.)
| | - Adriana Vollaro
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy;
| | - Maria De Fenza
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia, 80126 Naples, Italy; (A.E.); (M.D.F.); (D.D.)
| | - Daniele D’Alonzo
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia, 80126 Naples, Italy; (A.E.); (M.D.F.); (D.D.)
| | - Antonella Migliaccio
- Department of Public Health, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy; (A.M.); (R.Z.)
| | - Vita Dora Iula
- Complex Operative Unit of Clinical Pathology, “Ospedale del Mare-ASL NA1 Centro”, 80131 Naples, Italy;
| | - Raffaele Zarrilli
- Department of Public Health, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy; (A.M.); (R.Z.)
| | - Annalisa Guaragna
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia, 80126 Naples, Italy; (A.E.); (M.D.F.); (D.D.)
| |
Collapse
|
24
|
Collins MM, Behera RK, Pallister KB, Evans TJ, Burroughs O, Flack C, Guerra FE, Pullman W, Cone B, Dankoff JG, Nygaard TK, Brinsmade SR, Voyich JM. The Accessory Gene saeP of the SaeR/S Two-Component Gene Regulatory System Impacts Staphylococcus aureus Virulence During Neutrophil Interaction. Front Microbiol 2020; 11:561. [PMID: 32390958 PMCID: PMC7189620 DOI: 10.3389/fmicb.2020.00561] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 03/16/2020] [Indexed: 01/04/2023] Open
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.
Collapse
Affiliation(s)
- Madison M. Collins
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| | - Ranjan K. Behera
- Department of Biology, Georgetown University, Washington, DC, United States
| | - Kyler B. Pallister
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| | - Tyler J. Evans
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| | - Owen Burroughs
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| | - Caralyn Flack
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| | - Fermin E. Guerra
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| | - Willis Pullman
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| | - Brock Cone
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| | - Jennifer G. Dankoff
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| | - Tyler K. Nygaard
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| | - Shaun R. Brinsmade
- Department of Biology, Georgetown University, Washington, DC, United States
| | - Jovanka M. Voyich
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| |
Collapse
|
25
|
Stochastic Expression of Sae-Dependent Virulence Genes during Staphylococcus aureus Biofilm Development Is Dependent on SaeS. mBio 2020; 11:mBio.03081-19. [PMID: 31937649 PMCID: PMC6960292 DOI: 10.1128/mbio.03081-19] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The intricate process of biofilm formation in the human pathogen Staphylococcus aureus involves distinct stages during which a complex mixture of matrix molecules is produced and modified throughout the developmental cycle. Early in biofilm development, a subpopulation of cells detaches from its substrate in an event termed "exodus" that is mediated by SaePQRS-dependent stochastic expression of a secreted staphylococcal nuclease, which degrades extracellular DNA within the matrix, causing the release of cells and subsequently allowing for the formation of metabolically heterogenous microcolonies. Since the SaePQRS regulatory system is involved in the transcriptional control of multiple S. aureus virulence factors, the expression of several additional virulence genes was examined within a developing biofilm by introducing fluorescent gene reporter plasmids into wild-type S. aureus and isogenic regulatory mutants and growing these strains in a microfluidic system that supplies the bacteria with a constant flow of media while simultaneously imaging developing biofilms in 5-min intervals. This study demonstrated that multiple virulence genes, including nuc, were expressed stochastically within a specialized subpopulation of cells in nascent biofilms. We demonstrated that virulence genes regulated by SaePQRS were stochastically expressed in nearly all strains examined whereas Agr-regulated genes were expressed more homogenously within maturing microcolonies. The commonly used Newman strain contains a variant of SaeS (SaeSP) that confers constitutive kinase activity to the protein and caused this strain to lack the stochastic expression pattern observed in other strain backgrounds. Importantly, repair of the SaeSP allele resulting in reversion to the well-conserved SaeS L allele found in other strains restored stochastic expression in this strain.IMPORTANCE Staphylococcus aureus is an important human pathogen capable of colonizing diverse tissue types and inducing severe disease in both immunocompromised and otherwise healthy individuals. Biofilm infections caused by this bacterial species are of particular concern because of their persistence, even in the face of intensive therapeutic intervention. The results of the current study demonstrate the stochastic nature of Sae-mediated virulence gene expression in S. aureus and indicate that this regulatory system may function as a "bistable switch" in a manner similar to that seen with regulators controlling competence gene expression in Bacillus subtilis and persister cell formation in Escherichia coli The results of this study provide a new perspective on the complex mechanisms utilized by S. aureus during the establishment of infections.
Collapse
|
26
|
Ibberson CB, Whiteley M. The Staphylococcus aureus Transcriptome during Cystic Fibrosis Lung Infection. mBio 2019; 10:e02774-19. [PMID: 31744924 PMCID: PMC6867902 DOI: 10.1128/mbio.02774-19] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 10/18/2019] [Indexed: 12/16/2022] Open
Abstract
Laboratory models have been invaluable for the field of microbiology for over 100 years and have provided key insights into core aspects of bacterial physiology such as regulation and metabolism. However, it is important to identify the extent to which these models recapitulate bacterial physiology within a human infection environment. Here, we performed transcriptomics (RNA-seq), focusing on the physiology of the prominent pathogen Staphylococcus aureusin situ in human cystic fibrosis (CF) infection. Through principal-component and hierarchal clustering analyses, we found remarkable conservation in S. aureus gene expression in the CF lung despite differences in the patient clinic, clinical status, age, and therapeutic regimen. We used a machine learning approach to identify an S. aureus transcriptomic signature of 32 genes that can reliably distinguish between S. aureus transcriptomes in the CF lung and in vitro The majority of these genes were involved in virulence and metabolism and were used to improve a common CF infection model. Collectively, these results advance our knowledge of S. aureus physiology during human CF lung infection and demonstrate how in vitro models can be improved to better capture bacterial physiology in infection.IMPORTANCE Although bacteria have been studied in infection for over 100 years, the majority of these studies have utilized laboratory and animal models that often have unknown relevance to the human infections they are meant to represent. A primary challenge has been to assess bacterial physiology in the human host. To address this challenge, we performed transcriptomics of S. aureus during human cystic fibrosis (CF) lung infection. Using a machine learning framework, we defined a "human CF lung transcriptome signature" that primarily included genes involved in metabolism and virulence. In addition, we were able to apply our findings to improve an in vitro model of CF infection. Understanding bacterial gene expression within human infection is a critical step toward the development of improved laboratory models and new therapeutics.
Collapse
Affiliation(s)
- Carolyn B Ibberson
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, USA
- Emory-Children's Cystic Fibrosis Center, Atlanta, Georgia, USA
- Center for Microbial Dynamics and Infection, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Marvin Whiteley
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, USA
- Emory-Children's Cystic Fibrosis Center, Atlanta, Georgia, USA
- Center for Microbial Dynamics and Infection, Georgia Institute of Technology, Atlanta, Georgia, USA
| |
Collapse
|
27
|
Sward EW, Fones EM, Spaan RR, Pallister KB, Haller BL, Guerra FE, Zurek OW, Nygaard TK, Voyich JM. Staphylococcus aureus SaeR/S-Regulated Factors Decrease Monocyte-Derived Tumor Necrosis Factor-α to Reduce Neutrophil Bactericidal Activity. J Infect Dis 2019; 217:943-952. [PMID: 29272502 DOI: 10.1093/infdis/jix652] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 12/17/2017] [Indexed: 12/11/2022] Open
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.
Collapse
Affiliation(s)
- Eli W Sward
- Department of Microbiology and Immunology, Montana State University, Bozeman
| | - Elizabeth M Fones
- Department of Microbiology and Immunology, Montana State University, Bozeman
| | - Russel R Spaan
- Department of Microbiology and Immunology, Montana State University, Bozeman
| | - Kyler B Pallister
- Department of Microbiology and Immunology, Montana State University, Bozeman
| | - Brandon L Haller
- Department of Microbiology and Immunology, Montana State University, Bozeman
| | - Fermin E Guerra
- Department of Microbiology and Immunology, Montana State University, Bozeman
| | - Oliwia W Zurek
- Infectious Disease Department, Genentech Inc, South San Francisco, California
| | - Tyler K Nygaard
- Department of Microbiology and Immunology, Montana State University, Bozeman
| | - Jovanka M Voyich
- Department of Microbiology and Immunology, Montana State University, Bozeman
| |
Collapse
|
28
|
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.
Collapse
|
29
|
Jenul C, Horswill AR. Regulation of Staphylococcus aureus Virulence. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0031-2018. [PMID: 30953424 PMCID: PMC6452892 DOI: 10.1128/microbiolspec.gpp3-0031-2018] [Citation(s) in RCA: 307] [Impact Index Per Article: 51.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Indexed: 01/15/2023] Open
Abstract
Staphylococcus aureus is a Gram-positive opportunistic pathogen that has evolved a complex regulatory network to control virulence. One of the main functions of this interconnected network is to sense various environmental cues and respond by altering the production of virulence factors necessary for survival in the host, including cell surface adhesins and extracellular enzymes and toxins. Of these S. aureus regulatory systems, one of the best studied is the accessory gene regulator (agr), which is a quorum-sensing system that senses the local concentration of a cyclic peptide signaling molecule. This system allows S. aureus to sense its own population density and translate this information into a specific gene expression pattern. Besides agr, this pathogen uses other two-component systems to sense specific cues and coordinates responses with cytoplasmic regulators of the SarA protein family and alternative sigma factors. These divergent regulatory systems integrate the various environmental and host-derived signals into a network that ensures optimal pathogen response to the changing conditions. This article gives an overview of the most important and best-studied S. aureus regulatory systems and summarizes the functions of these regulators during host interactions. The regulatory systems discussed include the agr quorum-sensing system; the SaeRS, SrrAB, and ArlRS two-component systems, the cytoplasmic SarA-family regulators (SarA, Rot, and MgrA); and the alternative sigma factors (SigB and SigH).
Collapse
Affiliation(s)
- Christian Jenul
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Alexander R Horswill
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| |
Collapse
|
30
|
Behera RK, Mlynek KD, Linz MS, Brinsmade SR. A Fluorescence-based Method to Study Bacterial Gene Regulation in Infected Tissues. J Vis Exp 2019:10.3791/59055. [PMID: 30855576 PMCID: PMC7295204 DOI: 10.3791/59055] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Bacterial virulence genes are often regulated at the transcriptional level by multiple factors that respond to different environmental signals. Some factors act directly on virulence genes; others control pathogenesis by adjusting the expression of downstream regulators or the accumulation of signals that affect regulator activity. While regulation has been studied extensively during in vitro growth, relatively little is known about how gene expression is adjusted during infection. Such information is important when a particular gene product is a candidate for therapeutic intervention. Transcriptional approaches like quantitative, real-time RT-PCR and RNA-Seq are powerful ways to examine gene expression on a global level but suffer from many technical challenges including low abundance of bacterial RNA compared to host RNA, and sample degradation by RNases. Evaluating regulation using fluorescent reporters is relatively easy and can be multiplexed with fluorescent proteins with unique spectral properties. The method allows for single-cell, spatiotemporal analysis of gene expression in tissues that exhibit complex three-dimensional architecture and physiochemical gradients that affect bacterial regulatory networks. Such information is lost when data are averaged over the bulk population. Herein, we describe a method for quantifying gene expression in bacterial pathogens in situ. The method is based on simple tissue processing and direct observation of fluorescence from reporter proteins. We demonstrate the utility of this system by examining the expression of Staphylococcus aureus thermonuclease (nuc), whose gene product is required for immune evasion and full virulence ex vivo and in vivo. We show that nuc-gfp is strongly expressed in renal abscesses and reveal heterogeneous gene expression due in part to apparent spatial regulation of nuc promoter activity in abscesses fully engaged with the immune response. The method can be applied to any bacterium with a manipulatable genetic system and any infection model, providing valuable information for preclinical studies and drug development.
Collapse
|
31
|
Stress-induced inactivation of the Staphylococcus aureus purine biosynthesis repressor leads to hypervirulence. Nat Commun 2019; 10:775. [PMID: 30770821 PMCID: PMC6377658 DOI: 10.1038/s41467-019-08724-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 01/18/2019] [Indexed: 11/09/2022] Open
Abstract
Staphylococcus aureus is a significant cause of human infection. Here, we demonstrate that mutations in the transcriptional repressor of purine biosynthesis, purR, enhance the pathogenic potential of S. aureus. Indeed, systemic infection with purR mutants causes accelerated mortality in mice, which is due to aberrant up-regulation of fibronectin binding proteins (FnBPs). Remarkably, purR mutations can arise upon exposure of S. aureus to stress, such as an intact immune system. In humans, naturally occurring anti-FnBP antibodies exist that, while not protective against recurrent S. aureus infection, ostensibly protect against hypervirulent S. aureus infections. Vaccination studies support this notion, where anti-Fnb antibodies in mice protect against purR hypervirulence. These findings provide a novel link between purine metabolism and virulence in S. aureus.
Collapse
|
32
|
Nygaard TK, Borgogna TR, Sward EW, Guerra FE, Dankoff JG, Collins MM, Pallister KB, Chen L, Kreiswirth BN, Voyich JM. Aspartic Acid Residue 51 of SaeR Is Essential for Staphylococcus aureus Virulence. Front Microbiol 2018; 9:3085. [PMID: 30619166 PMCID: PMC6302044 DOI: 10.3389/fmicb.2018.03085] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 11/29/2018] [Indexed: 11/13/2022] Open
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.
Collapse
Affiliation(s)
- Tyler K Nygaard
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| | - Timothy R Borgogna
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| | - Eli W Sward
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| | - Fermin E Guerra
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| | - Jennifer G Dankoff
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| | - Madison M Collins
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| | - Kyler B Pallister
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| | - Liang Chen
- Public Health Research Institute Tuberculosis Center, New Jersey Medical School, Rutgers University, Newark, NJ, United States
| | - Barry N Kreiswirth
- Public Health Research Institute Tuberculosis Center, New Jersey Medical School, Rutgers University, Newark, NJ, United States
| | - Jovanka M Voyich
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| |
Collapse
|
33
|
Venkatasubramaniam A, Kanipakala T, Ganjbaksh N, Mehr R, Mukherjee I, Krishnan S, Bae T, Aman MJ, Adhikari RP. A Critical Role for HlgA in Staphylococcus aureus Pathogenesis Revealed by A Switch in the SaeRS Two-Component Regulatory System. Toxins (Basel) 2018; 10:E377. [PMID: 30231498 PMCID: PMC6162840 DOI: 10.3390/toxins10090377] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Revised: 09/11/2018] [Accepted: 09/11/2018] [Indexed: 12/18/2022] Open
Abstract
Cytolytic pore-forming toxins including alpha hemolysin (Hla) and bicomponent leukotoxins play an important role in the pathogenesis of Staphylococcus aureus. These toxins kill the polymorphonuclear phagocytes (PMNs), disrupt epithelial and endothelial barriers, and lyse erythrocytes to provide iron for bacterial growth. The expression of these toxins is regulated by the two-component sensing systems Sae and Agr. Here, we report that a point mutation (L18P) in SaeS, the histidine kinase sensor of the Sae system, renders the S. aureus Newman hemolytic activity fully independent of Hla and drastically increases the PMN lytic activity. Furthermore, this Hla-independent activity, unlike Hla itself, can lyse human erythrocytes. The Hla-independent activity towards human erythrocytes was also evident in USA300, however, under strict agr control. Gene knockout studies revealed that this Hla-independent Sae-regulated activity was entirely dependent on gamma hemolysin A subunit (HlgA). In contrast, hemolytic activity of Newman towards human erythrocytes from HlgAB resistant donors was completely dependent on agr. The culture supernatant from Newman S. aureus could be neutralized by antisera against two vaccine candidates based on LukS and LukF subunits of Panton-Valentine leukocidin but not by an anti-Hla neutralizing antibody. These findings display the complex involvement of Sae and Agr systems in regulating the virulence of S. aureus and have important implications for vaccine and immunotherapeutics development for S. aureus disease in humans.
Collapse
Affiliation(s)
| | | | | | - Rana Mehr
- Integrated Biotherapeutics Inc., Rockville, MD 20850, USA.
| | | | | | - Taeok Bae
- Department of Microbiology and Immunology, Indiana University School of Medicine-Northwest, Gary, IN 46408, USA.
| | - M Javad Aman
- Integrated Biotherapeutics Inc., Rockville, MD 20850, USA.
| | | |
Collapse
|
34
|
Staphylococcus aureus biofilms release leukocidins to elicit extracellular trap formation and evade neutrophil-mediated killing. Proc Natl Acad Sci U S A 2018; 115:7416-7421. [PMID: 29941565 DOI: 10.1073/pnas.1721949115] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Bacterial biofilms efficiently evade immune defenses, greatly complicating the prognosis of chronic infections. How methicillin-resistant Staphylococcus aureus (MRSA) biofilms evade host immune defenses is largely unknown. This study describes some of the major mechanisms required for S. aureus biofilms to evade the innate immune response and provides evidence of key virulence factors required for survival and persistence of bacteria during chronic infections. Neutrophils are the most abundant white blood cells in circulation, playing crucial roles in the control and elimination of bacterial pathogens. Specifically, here we show that, unlike single-celled populations, S. aureus biofilms rapidly skew neutrophils toward neutrophil extracellular trap (NET) formation through the combined activity of leukocidins Panton-Valentine leukocidin and γ-hemolysin AB. By eliciting this response, S. aureus was able to persist, as the antimicrobial activity of released NETs was ineffective at clearing biofilm bacteria. Indeed, these studies suggest that NETs could inadvertently potentiate biofilm infections. Last, chronic infection in a porcine burn wound model clearly demonstrated that leukocidins are required for "NETosis" and facilitate bacterial survival in vivo.
Collapse
|
35
|
Nutritional Regulation of the Sae Two-Component System by CodY in Staphylococcus aureus. J Bacteriol 2018; 200:JB.00012-18. [PMID: 29378891 DOI: 10.1128/jb.00012-18] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 01/23/2018] [Indexed: 01/22/2023] Open
Abstract
Staphylococcus aureus subverts innate defenses during infection in part by killing host immune cells to exacerbate disease. This human pathogen intercepts host cues and activates a transcriptional response via the S. aureus exoprotein expression (SaeR/SaeS [SaeR/S]) two-component system to secrete virulence factors critical for pathogenesis. We recently showed that the transcriptional repressor CodY adjusts nuclease (nuc) gene expression via SaeR/S, but the mechanism remained unknown. Here, we identified two CodY binding motifs upstream of the sae P1 promoter, which suggested direct regulation by this global regulator. We show that CodY shares a binding site with the positive activator SaeR and that alleviating direct CodY repression at this site is sufficient to abrogate stochastic expression, suggesting that CodY represses sae expression by blocking SaeR binding. Epistasis experiments support a model that CodY also controls sae indirectly through Agr and Rot-mediated repression of the sae P1 promoter. We also demonstrate that CodY repression of sae restrains production of secreted cytotoxins that kill human neutrophils. We conclude that CodY plays a previously unrecognized role in controlling virulence gene expression via SaeR/S and suggest a mechanism by which CodY acts as a master regulator of pathogenesis by tying nutrient availability to virulence gene expression.IMPORTANCE Bacterial mechanisms that mediate the switch from a commensal to pathogenic lifestyle are among the biggest unanswered questions in infectious disease research. Since the expression of most virulence genes is often correlated with nutrient depletion, this implies that virulence is a response to the lack of nourishment in host tissues and that pathogens like S. aureus produce virulence factors in order to gain access to nutrients in the host. Here, we show that specific nutrient depletion signals appear to be funneled to the SaeR/S system through the global regulator CodY. Our findings reveal a strategy by which S. aureus delays the production of immune evasion and immune-cell-killing proteins until key nutrients are depleted.
Collapse
|
36
|
Kane TL, Carothers KE, Lee SW. Virulence Factor Targeting of the Bacterial Pathogen Staphylococcus aureus for Vaccine and Therapeutics. Curr Drug Targets 2018; 19:111-127. [PMID: 27894236 PMCID: PMC5957279 DOI: 10.2174/1389450117666161128123536] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 10/12/2016] [Accepted: 10/27/2016] [Indexed: 12/21/2022]
Abstract
BACKGROUND Staphylococcus aureus is a major bacterial pathogen capable of causing a range of infections in humans from gastrointestinal disease, skin and soft tissue infections, to severe outcomes such as sepsis. Staphylococcal infections in humans can be frequent and recurring, with treatments becoming less effective due to the growing persistence of antibiotic resistant S. aureus strains. Due to the prevalence of antibiotic resistance, and the current limitations on antibiotic development, an active and highly promising avenue of research has been to develop strategies to specifically inhibit the activity of virulence factors produced S. aureus as an alternative means to treat disease. OBJECTIVE In this review we specifically highlight several major virulence factors produced by S. aureus for which recent advances in antivirulence approaches may hold promise as an alternative means to treating diseases caused by this pathogen. Strategies to inhibit virulence factors can range from small molecule inhibitors, to antibodies, to mutant and toxoid forms of the virulence proteins. CONCLUSION The major prevalence of antibiotic resistant strains of S. aureus combined with the lack of new antibiotic discoveries highlight the need for vigorous research into alternative strategies to combat diseases caused by this highly successful pathogen. Current efforts to develop specific antivirulence strategies, vaccine approaches, and alternative therapies for treating severe disease caused by S. aureus have the potential to stem the tide against the limitations that we face in the post-antibiotic era.
Collapse
Affiliation(s)
- Trevor L. Kane
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556
- Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Katelyn E. Carothers
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556
- Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Shaun W. Lee
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556
- Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA
| |
Collapse
|
37
|
Balasubramanian D, Harper L, Shopsin B, Torres VJ. Staphylococcus aureus pathogenesis in diverse host environments. Pathog Dis 2017; 75:ftx005. [PMID: 28104617 DOI: 10.1093/femspd/ftx005] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 01/18/2017] [Indexed: 12/21/2022] Open
Abstract
Staphylococcus aureus is an eminent human pathogen that can colonize the human host and cause severe life-threatening illnesses. This bacterium can reside in and infect a wide range of host tissues, ranging from superficial surfaces like the skin to deeper tissues such as in the gastrointestinal tract, heart and bones. Due to its multifaceted lifestyle, S. aureus uses complex regulatory networks to sense diverse signals that enable it to adapt to different environments and modulate virulence. In this minireview, we explore well-characterized environmental and host cues that S. aureus responds to and describe how this pathogen modulates virulence in response to these signals. Lastly, we highlight therapeutic approaches undertaken by several groups to inhibit both signaling and the cognate regulators that sense and transmit these signals downstream.
Collapse
Affiliation(s)
- Divya Balasubramanian
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Lamia Harper
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Bo Shopsin
- Department of Medicine, Division of Infectious Diseases, New York University School of Medicine, New York, NY 10016 USA
| | - Victor J Torres
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| |
Collapse
|
38
|
Ericson ME, Subramanian C, Frank MW, Rock CO. Role of Fatty Acid Kinase in Cellular Lipid Homeostasis and SaeRS-Dependent Virulence Factor Expression in Staphylococcus aureus. mBio 2017; 8:e00988-17. [PMID: 28765222 PMCID: PMC5539427 DOI: 10.1128/mbio.00988-17] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 06/28/2017] [Indexed: 11/21/2022] Open
Abstract
The SaeRS two-component system is a master activator of virulence factor transcription in Staphylococcus aureus, but the cellular factors that control its activity are unknown. Fatty acid (FA) kinase is a two-component enzyme system required for extracellular FA uptake and SaeRS activity. Here, we demonstrate the existence of an intracellular nonesterified FA pool in S. aureus that is elevated in strains lacking FA kinase activity. SaeRS-mediated transcription is restored in FA kinase-negative strains when the intracellular FA pool is reduced either by growth with FA-depleted bovine serum albumin to extract the FA into the medium or by the heterologous expression of Neisseria gonorrhoeae acyl-acyl carrier protein synthetase to activate FA for phospholipid synthesis. These data show that FAs act as negative regulators of SaeRS signaling, and FA kinase activates SaeRS-dependent virulence factor production by lowering inhibitory FA levels. Thus, FA kinase plays a role in cellular lipid homeostasis by activating FA for incorporation into phospholipid, and it indirectly regulates SaeRS signaling by maintaining a low intracellular FA pool.IMPORTANCE The SaeRS two-component system is a master transcriptional activator of virulence factor production in response to the host environment in S. aureus, and strains lacking FA kinase have severely attenuated SaeRS-dependent virulence factor transcription. FA kinase is required for the activation of exogenous FAs, and it plays a role in cellular lipid homeostasis by recycling cellular FAs into the phospholipid biosynthetic pathway. Activation of the sensor kinase, SaeS, is mediated by its membrane anchor domain, and the FAs which accumulate in FA kinase knockout strains are potent inhibitors of SaeS-dependent signaling. This work identifies FAs as physiological effectors for the SaeRS system and reveals a connection between cellular lipid homeostasis and the regulation of virulence factor transcription. FA kinase is widely distributed in Gram-positive bacteria, suggesting similar roles for FA kinase in these organisms.
Collapse
Affiliation(s)
- Megan E Ericson
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Chitra Subramanian
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Matthew W Frank
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Charles O Rock
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
39
|
Guerra FE, Borgogna TR, Patel DM, Sward EW, Voyich JM. Epic Immune Battles of History: Neutrophils vs. Staphylococcus aureus. Front Cell Infect Microbiol 2017; 7:286. [PMID: 28713774 PMCID: PMC5491559 DOI: 10.3389/fcimb.2017.00286] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 06/12/2017] [Indexed: 12/23/2022] Open
Abstract
Neutrophils are the most abundant leukocytes in human blood and the first line of defense after bacteria have breached the epithelial barriers. After migration to a site of infection, neutrophils engage and expose invading microorganisms to antimicrobial peptides and proteins, as well as reactive oxygen species, as part of their bactericidal arsenal. Ideally, neutrophils ingest bacteria to prevent damage to surrounding cells and tissues, kill invading microorganisms with antimicrobial mechanisms, undergo programmed cell death to minimize inflammation, and are cleared away by macrophages. Staphylococcus aureus (S. aureus) is a prevalent Gram-positive bacterium that is a common commensal and causes a wide range of diseases from skin infections to endocarditis. Since its discovery, S. aureus has been a formidable neutrophil foe that has challenged the efficacy of this professional assassin. Indeed, proper clearance of S. aureus by neutrophils is essential to positive infection outcome, and S. aureus has developed mechanisms to evade neutrophil killing. Herein, we will review mechanisms used by S. aureus to modulate and evade neutrophil bactericidal mechanisms including priming, activation, chemotaxis, production of reactive oxygen species, and resolution of infection. We will also highlight how S. aureus uses sensory/regulatory systems to tailor production of virulence factors specifically to the triggering signal, e.g., neutrophils and defensins. To conclude, we will provide an overview of therapeutic approaches that may potentially enhance neutrophil antimicrobial functions.
Collapse
Affiliation(s)
- Fermin E Guerra
- Department of Microbiology and Immunology, Montana State UniversityBozeman, MT, United States
| | - Timothy R Borgogna
- Department of Microbiology and Immunology, Montana State UniversityBozeman, MT, United States
| | - Delisha M Patel
- Department of Microbiology and Immunology, Montana State UniversityBozeman, MT, United States
| | - Eli W Sward
- Department of Microbiology and Immunology, Montana State UniversityBozeman, MT, United States
| | - Jovanka M Voyich
- Department of Microbiology and Immunology, Montana State UniversityBozeman, MT, United States
| |
Collapse
|
40
|
Spaan AN, van Strijp JAG, Torres VJ. Leukocidins: staphylococcal bi-component pore-forming toxins find their receptors. Nat Rev Microbiol 2017; 15:435-447. [PMID: 28420883 DOI: 10.1038/nrmicro.2017.27] [Citation(s) in RCA: 244] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Staphylococcus aureus is a major bacterial pathogen that causes disease worldwide. The emergence of strains that are resistant to commonly used antibiotics and the failure of vaccine development have resulted in a renewed interest in the pathophysiology of this bacterium. Staphylococcal leukocidins are a family of bi-component pore-forming toxins that are important virulence factors. During the past five years, cellular receptors have been identified for all of the bi-component leukocidins. The identification of the leukocidin receptors explains the cellular tropism and species specificity that is exhibited by these toxins, which has important biological consequences. In this Review, we summarize the recent discoveries that have reignited interest in these toxins and provide an outlook for future research.
Collapse
Affiliation(s)
- András N Spaan
- Department of Medical Microbiology, University Medical Center Utrecht, Heidelberglaan 100, 3584CX Utrecht, The Netherlands
| | - Jos A G van Strijp
- Department of Medical Microbiology, University Medical Center Utrecht, Heidelberglaan 100, 3584CX Utrecht, The Netherlands
| | - Victor J Torres
- Department of Microbiology, New York University School of Medicine, 430 East 29th Street, 10016 New York, USA
| |
Collapse
|
41
|
Moormeier DE, Bayles KW. Staphylococcus aureus biofilm: a complex developmental organism. Mol Microbiol 2017; 104:365-376. [PMID: 28142193 DOI: 10.1111/mmi.13634] [Citation(s) in RCA: 330] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2017] [Indexed: 12/11/2022]
Abstract
Chronic biofilm-associated infections caused by Staphylococcus aureus often lead to significant increases in morbidity and mortality, particularly when associated with indwelling medical devices. This has triggered a great deal of research attempting to understand the molecular mechanisms that control S. aureus biofilm formation and the basis for the recalcitrance of these multicellular structures to antibiotic therapy. The purpose of this review is to summarize our current understanding of S. aureus biofilm development, focusing on the description of a newly-defined, five-stage model of biofilm development and the mechanisms required for each stage. Importantly, this model includes an alternate view of the processes involved in microcolony formation in S. aureus and suggests that these structures originate as a result of stochastically regulated metabolic heterogeneity and proliferation within a maturing biofilm population, rather than a subtractive process involving the release of cell clusters from a thick, unstructured biofilm. Importantly, it is proposed that this new model of biofilm development involves the genetically programmed generation of metabolically distinct subpopulations of cells, resulting in an overall population that is better able to adapt to rapidly changing environmental conditions.
Collapse
Affiliation(s)
- Derek E Moormeier
- Center for Staphylococcal Research, Department of Pathology & Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kenneth W Bayles
- Center for Staphylococcal Research, Department of Pathology & Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
42
|
VfrB Is a Key Activator of the Staphylococcus aureus SaeRS Two-Component System. J Bacteriol 2017; 199:JB.00828-16. [PMID: 28031278 DOI: 10.1128/jb.00828-16] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 12/14/2016] [Indexed: 12/22/2022] Open
Abstract
In previous studies, we identified the fatty acid kinase virulence factor regulator B (VfrB) as a potent regulator of α-hemolysin and other virulence factors in Staphylococcus aureus In this study, we demonstrated that VfrB is a positive activator of the SaeRS two-component regulatory system. Analysis of vfrB, saeR, and saeS mutant strains revealed that VfrB functions in the same pathway as SaeRS. At the transcriptional level, the promoter activities of SaeRS class I (coa) and class II (hla) target genes were downregulated during the exponential growth phase in the vfrB mutant, compared to the wild-type strain. In addition, saePQRS expression was decreased in the vfrB mutant strain, demonstrating a need for this protein in the autoregulation of SaeRS. The requirement for VfrB-mediated activation was circumvented when SaeS was constitutively active due to an SaeS (L18P) substitution. Furthermore, activation of SaeS via human neutrophil peptide 1 (HNP-1) overcame the dependence on VfrB for transcription from class I Sae promoters. Consistent with the role of VfrB in fatty acid metabolism, hla expression was decreased in the vfrB mutant with the addition of exogenous myristic acid. Lastly, we determined that aspartic acid residues D38 and D40, which are predicted to be key to VfrB enzymatic activity, were required for VfrB-mediated α-hemolysin production. Collectively, this study implicates VfrB as a novel accessory protein needed for the activation of SaeRS in S. aureusIMPORTANCE The SaeRS two-component system is a key regulator of virulence determinant production in Staphylococcus aureus Although the regulon of this two-component system is well characterized, the activation mechanisms, including the specific signaling molecules, remain elusive. Elucidating the complex regulatory circuit of SaeRS regulation is important for understanding how the system contributes to disease causation by this pathogen. To this end, we have identified the fatty acid kinase VfrB as a positive regulatory modulator of SaeRS-mediated transcription of virulence factors in S. aureus In addition to describing a new regulatory aspect of SaeRS, this study establishes a link between fatty acid kinase activity and virulence factor regulation.
Collapse
|
43
|
Schmidt F, Meyer T, Sundaramoorthy N, Michalik S, Surmann K, Depke M, Dhople V, Gesell Salazar M, Holtappels G, Zhang N, Bröker BM, Bachert C, Völker U. Characterization of human and Staphylococcus aureus proteins in respiratory mucosa by in vivo- and immunoproteomics. J Proteomics 2017; 155:31-39. [DOI: 10.1016/j.jprot.2017.01.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 11/28/2016] [Accepted: 01/13/2017] [Indexed: 10/20/2022]
|
44
|
Choby JE, Mike LA, Mashruwala AA, Dutter BF, Dunman PM, Sulikowski GA, Boyd JM, Skaar EP. A Small-Molecule Inhibitor of Iron-Sulfur Cluster Assembly Uncovers a Link between Virulence Regulation and Metabolism in Staphylococcus aureus. Cell Chem Biol 2016; 23:1351-1361. [PMID: 27773628 DOI: 10.1016/j.chembiol.2016.09.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 09/01/2016] [Accepted: 09/30/2016] [Indexed: 11/28/2022]
Abstract
The rising problem of antimicrobial resistance in Staphylococcus aureus necessitates the discovery of novel therapeutic targets for small-molecule intervention. A major obstacle of drug discovery is identifying the target of molecules selected from high-throughput phenotypic assays. Here, we show that the toxicity of a small molecule termed '882 is dependent on the constitutive activity of the S. aureus virulence regulator SaeRS, uncovering a link between virulence factor production and energy generation. A series of genetic, physiological, and biochemical analyses reveal that '882 inhibits iron-sulfur (Fe-S) cluster assembly most likely through inhibition of the Suf complex, which synthesizes Fe-S clusters. In support of this, '882 supplementation results in decreased activity of the Fe-S cluster-dependent enzyme aconitase. Further information regarding the effects of '882 has deepened our understanding of virulence regulation and demonstrates the potential for small-molecule modulation of Fe-S cluster assembly in S. aureus and other pathogens.
Collapse
Affiliation(s)
- Jacob E Choby
- Department of Pathology, Microbiology, & Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Graduate Program in Microbiology & Immunology, Vanderbilt University, Nashville, TN 37232, USA
| | - Laura A Mike
- Department of Pathology, Microbiology, & Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Ameya A Mashruwala
- Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, NJ 08901, USA
| | - Brendan F Dutter
- Department of Chemistry, Vanderbilt Institute for Chemical Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Paul M Dunman
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Gary A Sulikowski
- Department of Chemistry, Vanderbilt Institute for Chemical Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Jeffrey M Boyd
- Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, NJ 08901, USA.
| | - Eric P Skaar
- Department of Pathology, Microbiology, & Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Veterans Affairs Tennessee Valley Healthcare Services, Nashville, TN 37232, USA.
| |
Collapse
|
45
|
The Spl Serine Proteases Modulate Staphylococcus aureus Protein Production and Virulence in a Rabbit Model of Pneumonia. mSphere 2016; 1:mSphere00208-16. [PMID: 27747296 PMCID: PMC5061998 DOI: 10.1128/msphere.00208-16] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 09/23/2016] [Indexed: 01/09/2023] Open
Abstract
Staphylococcus aureus is a versatile human pathogen that produces an array of virulence factors, including several proteases. Of these, six proteases called the Spls are the least characterized. Previous evidence suggests that the Spls are expressed during human infection; however, their function is unknown. Our study shows that the Spls are required for S. aureus to cause disseminated lung damage during pneumonia. Further, we present the first example of a human protein cut by an Spl protease. Although the Spls were predicted not to cut staphylococcal proteins, we also show that an spl mutant has altered abundance of both secreted and surface-associated proteins. This work provides novel insight into the function of Spls during infection and their potential ability to degrade both staphylococcal and human proteins. The Spl proteases are a group of six serine proteases that are encoded on the νSaβ pathogenicity island and are unique to Staphylococcus aureus. Despite their interesting biochemistry, their biological substrates and functions in virulence have been difficult to elucidate. We found that an spl operon mutant of the community-associated methicillin-resistant S. aureus USA300 strain LAC induced localized lung damage in a rabbit model of pneumonia, characterized by bronchopneumonia observed histologically. Disease in the mutant-infected rabbits was restricted in distribution compared to that in wild-type USA300-infected rabbits. We also found that SplA is able to cleave the mucin 16 glycoprotein from the surface of the CalU-3 lung cell line, suggesting a possible mechanism for wild-type USA300 spreading pneumonia to both lungs. Investigation of the secreted and surface proteomes of wild-type USA300 and the spl mutant revealed multiple alterations in metabolic proteins and virulence factors. This study demonstrates that the Spls modulate S. aureus physiology and virulence, identifies a human target of SplA, and suggests potential S. aureus targets of the Spl proteases. IMPORTANCEStaphylococcus aureus is a versatile human pathogen that produces an array of virulence factors, including several proteases. Of these, six proteases called the Spls are the least characterized. Previous evidence suggests that the Spls are expressed during human infection; however, their function is unknown. Our study shows that the Spls are required for S. aureus to cause disseminated lung damage during pneumonia. Further, we present the first example of a human protein cut by an Spl protease. Although the Spls were predicted not to cut staphylococcal proteins, we also show that an spl mutant has altered abundance of both secreted and surface-associated proteins. This work provides novel insight into the function of Spls during infection and their potential ability to degrade both staphylococcal and human proteins.
Collapse
|
46
|
Liu Q, Yeo WS, Bae T. The SaeRS Two-Component System of Staphylococcus aureus. Genes (Basel) 2016; 7:genes7100081. [PMID: 27706107 PMCID: PMC5083920 DOI: 10.3390/genes7100081] [Citation(s) in RCA: 148] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 09/23/2016] [Indexed: 12/20/2022] Open
Abstract
In the Gram-positive pathogenic bacterium Staphylococcus aureus, the SaeRS twocomponent system (TCS) plays a major role in controlling the production of over 20 virulence factors including hemolysins, leukocidins, superantigens, surface proteins, and proteases. The SaeRS TCS is composed of the sensor histidine kinase SaeS, response regulator SaeR, and two auxiliary proteins SaeP and SaeQ. Since its discovery in 1994, the sae locus has been studied extensively, and its contributions to staphylococcal virulence and pathogenesis have been well documented and understood; however, the molecular mechanism by which the SaeRS TCS receives and processes cognate signals is not. In this article, therefore, we review the literature focusing on the signaling mechanism and its interaction with other global regulators.
Collapse
Affiliation(s)
- Qian Liu
- Department of Laboratory Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| | - Won-Sik Yeo
- Department of Microbiology and Immunology, Indiana University School of Medicine-Northwest, Gary, IN 46408, USA.
| | - Taeok Bae
- Department of Microbiology and Immunology, Indiana University School of Medicine-Northwest, Gary, IN 46408, USA.
| |
Collapse
|
47
|
Guerra FE, Addison CB, de Jong NWM, Azzolino J, Pallister KB, van Strijp JAG, Voyich JM. Staphylococcus aureus SaeR/S-regulated factors reduce human neutrophil reactive oxygen species production. J Leukoc Biol 2016; 100:1005-1010. [PMID: 27334228 DOI: 10.1189/jlb.4vmab0316-100rr] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 05/31/2016] [Indexed: 01/12/2023] Open
Abstract
Neutrophils are the first line of defense after a pathogen has breached the epithelial barriers, and unimpaired neutrophil functions are essential to clear infections. Staphylococcus aureus is a prevalent human pathogen that is able to withstand neutrophil killing, yet the mechanisms used by S. aureus to inhibit neutrophil clearance remain incompletely defined. The production of reactive oxygen species (ROS) is a vital neutrophil antimicrobial mechanism. Herein, we test the hypothesis that S. aureus uses the SaeR/S two-component gene regulatory system to produce virulence factors that reduce neutrophil ROS production. With the use of ROS probes, the temporal and overall production of neutrophil ROS was assessed during exposure to the clinically relevant S. aureus USA300 (strain LAC) and its isogenic mutant LACΔsaeR/S Our results demonstrated that SaeR/S-regulated factors do not inhibit neutrophil superoxide (O2-) production. However, subsequent neutrophil ROS production was significantly reduced during exposure to LAC compared with LACΔsaeR/S In addition, neutrophil H2O2 production was reduced significantly by SaeR/S-regulated factors by a mechanism independent of catalase. Consequently, the reduction in neutrophil H2O2 resulted in decreased production of the highly antimicrobial agent hypochlorous acid/hypochlorite anion (HOCl/-OCl). These findings suggest a new evasion strategy used by S. aureus to diminish a vital neutrophil antimicrobial mechanism.
Collapse
Affiliation(s)
- Fermin E Guerra
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana, USA
| | - Conrad B Addison
- School of Medicine, University of Washington, Seattle, Washington, USA; and
| | - Nienke W M de Jong
- Medical Microbiology University Medical Center Utrecht, Utrecht,Netherlands
| | - Joseph Azzolino
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana, USA
| | - Kyler B Pallister
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana, USA
| | - Jos A G van Strijp
- Medical Microbiology University Medical Center Utrecht, Utrecht,Netherlands
| | - Jovanka M Voyich
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana, USA;
| |
Collapse
|
48
|
Ramundo MS, Beltrame CO, Botelho AMN, Coelho LR, Silva-Carvalho MC, Ferreira-Carvalho BT, Nicolás MF, Guedes IA, Dardenne LE, O'Gara J, Figueiredo AMS. A unique SaeS allele overrides cell-density dependent expression of saeR and lukSF-PV in the ST30-SCCmecIV lineage of CA-MRSA. Int J Med Microbiol 2016; 306:367-80. [PMID: 27265234 DOI: 10.1016/j.ijmm.2016.05.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 05/06/2016] [Accepted: 05/08/2016] [Indexed: 12/11/2022] Open
Abstract
ST30 (CC30)-SCCmec IV (USA1100) is one of the most common community-acquired methicillin-resistant Staphylococcus aureus (CA-MRSA) lineages. ST30 isolates typically carry lukSF-PV genes encoding the Panton-Valentine leukocidin (PVL) and are responsible for outbreaks of invasive infections worldwide. In this study, twenty CC30 isolates were analyzed. All were very susceptible to non-β-lactam antimicrobials, 18/20 harbored the lukSF-PV genes, only 1/20 exhibited agr-rnaIII dysfunction, and the majority was not able to form biofilm on inert surfaces. Analysis of lukSF-PV temporal regulation revealed that opposite to other CA-MRSA isolates, these genes were more highly expressed in early log phase than in stationary phase. This inverted lukSF-PV temporal expression was associated with a similar pattern of saeRS expression in the ST30 isolates, namely high level expression in log phase and reduced expression in stationary phase. Reduced saeRS expression in stationary phase was associated with low expression levels of the sae regulators, agr and agr-upregulator sarA, which activate the stationary phase sae-P1 promoter and overexpression of agr-RNAIII restored the levels of saeR and lukSF-PV trancripts in stationary phase. Altered SaeRS activity in the ST30 isolates was attributed to amino acid substitutions (N227S, E268K and S351T) in the HTPase_c domain of SaeS (termed SaeS(SKT)). Complementation of a USA300 saeS mutant with the saeS(SKT) and saeS alleles under the direction of the log phase sae-P3 promoter revealed that saeR and lukSF-PV transcription levels were more significantly activated by saeS(SKT) than saeS. In summary our data identify a unique saeS allele (saeS(SKT)) which appears to override cell-density dependent SaeR and PVL expression in ST30 CA-MRSA isolates. Further studies to determine the contribution of saeS(SKT) allele to the pathogenesis of infections caused by ST30 isolates are merited.
Collapse
Affiliation(s)
- Mariana Severo Ramundo
- Universidade Federal do Rio de Janeiro, Instituto de Microbiologia Paulo de Góes, Rio de Janeiro, RJ, Brazil.
| | - Cristiana Ossaille Beltrame
- Universidade Federal do Rio de Janeiro, Instituto de Microbiologia Paulo de Góes, Rio de Janeiro, RJ, Brazil.
| | - Ana Maria Nunes Botelho
- Universidade Federal do Rio de Janeiro, Instituto de Microbiologia Paulo de Góes, Rio de Janeiro, RJ, Brazil.
| | - Leonardo Rocchetto Coelho
- Universidade Federal do Rio de Janeiro, Instituto de Microbiologia Paulo de Góes, Rio de Janeiro, RJ, Brazil.
| | - Maria Cicera Silva-Carvalho
- Universidade Federal do Rio de Janeiro, Instituto de Microbiologia Paulo de Góes, Rio de Janeiro, RJ, Brazil.
| | | | | | | | | | - James O'Gara
- School of Natural Sciences, Microbiology Department, NUI Galway, Galway, Ireland.
| | - Agnes Marie Sá Figueiredo
- Universidade Federal do Rio de Janeiro, Instituto de Microbiologia Paulo de Góes, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
49
|
Kiedrowski MR, Paharik AE, Ackermann LW, Shelton AU, Singh SB, Starner TD, Horswill AR. Development of an in vitro colonization model to investigate Staphylococcus aureus interactions with airway epithelia. Cell Microbiol 2016; 18:720-32. [PMID: 26566259 DOI: 10.1111/cmi.12543] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Revised: 11/03/2015] [Accepted: 11/04/2015] [Indexed: 12/27/2022]
Abstract
Staphylococcus aureus is a bacterial pathogen responsible for a wide range of diseases and is also a human commensal colonizing the upper respiratory tract. Strains belonging to the clonal complex group CC30 are associated with colonization, although the colonization state itself is not clearly defined. In this work, we developed a co-culture model with S. aureus colonizing the apical surface of polarized human airway epithelial cells. The S. aureus are grown at the air-liquid interface to allow an in-depth evaluation of a simulated colonization state. Exposure to wild-type, S. aureus bacteria or conditioned media killed airway epithelial cells within 1 day, while mutant S. aureus strains lacking alpha-toxin (hla) persisted on viable cells for at least 2 days. Recent S. aureus CC30 isolates are natural hla mutants, and we observed that these strains displayed reduced toxicity toward airway epithelial cells. Quantitative real-time polymerase chain reaction of known virulence factors showed the expression profile of S. aureus grown in co-culture correlates with results from previous human colonization studies. Microarray analysis indicated significant shifts in S. aureus physiology in the co-culture model toward lipid and amino acid metabolism. The development of the in vitro colonization model will enable further study of specific S. aureus interactions with the host epithelia.
Collapse
Affiliation(s)
- Megan R Kiedrowski
- Department of Microbiology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Alexandra E Paharik
- Department of Microbiology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Laynez W Ackermann
- Department of Microbiology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Annie U Shelton
- Department of Pediatrics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Sachinkumar B Singh
- Department of Pediatrics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Timothy D Starner
- Department of Pediatrics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Alexander R Horswill
- Department of Microbiology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| |
Collapse
|
50
|
Mueller EA, Merriman JA, Schlievert PM. Toxic shock syndrome toxin-1, not α-toxin, mediated Bundaberg fatalities. Microbiology (Reading) 2015; 161:2361-2368. [DOI: 10.1099/mic.0.000196] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Affiliation(s)
- Elizabeth A. Mueller
- Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Joseph A. Merriman
- Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Patrick M. Schlievert
- Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|