1
|
Komorowicz E, Farkas VJ, Szabó L, Cherrington S, Thelwell C, Kolev K. DNA and histones impair the mechanical stability and lytic susceptibility of fibrin formed by staphylocoagulase. Front Immunol 2023; 14:1233128. [PMID: 37662916 PMCID: PMC10470048 DOI: 10.3389/fimmu.2023.1233128] [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: 06/01/2023] [Accepted: 07/31/2023] [Indexed: 09/05/2023] Open
Abstract
Background Staphylocoagulase (SCG) is a virulence factor of Staphylococcus aureus, one of the most lethal pathogens of our times. The complex of SCG with prothrombin (SCG/ProT) can clot fibrinogen, and SCG/ProT-induced fibrin and plasma clots have been described to show decreased mechanical and lytic resistance, which may contribute to septic emboli from infected cardiac vegetations. At infection sites, neutrophils can release DNA and histones, as parts of neutrophil extracellular traps (NETs), which in turn favor thrombosis, inhibit fibrinolysis and strengthen clot structure. Objectives To characterize the combined effects of major NET-components (DNA, histone H1 and H3) on SCG/ProT-induced clot structure, mechanical and lytic stability. Methods Recombinant SCG was used to clot purified fibrinogen and plasma. The kinetics of formation and lysis of fibrin and plasma clots containing H1 or core histones+/-DNA were followed by turbidimetry. Fibrin structure and mechanical stability were characterized with scanning electron microscopy, pressure-driven permeation, and oscillation rheometry. Results Histones and DNA favored the formation of thicker fibrin fibers and a more heterogeneous clot structure including high porosity with H1 histone, whereas low porosity with core histones and DNA. As opposed to previous observations with thrombin-induced clots, SCG/ProT-induced fibrin was not mechanically stabilized by histones. Similarly to thrombin-induced clots, the DNA-histone complexes prolonged fibrinolysis with tissue-type plasminogen activator (up to 2-fold). The anti-fibrinolytic effect of the DNA and DNA-H3 complex was observed in plasma clots too. Heparin (low molecular weight) accelerated the lysis of SCG/ProT-clots from plasma, even if DNA and histones were also present. Conclusions In the interplay of NETs and fibrin formed by SCG, DNA and histones promote structural heterogeneity in the clots, and fail to stabilize them against mechanical stress. The DNA-histone complexes render the SCG-fibrin more resistant to lysis and thereby less prone to embolization.
Collapse
Affiliation(s)
- Erzsébet Komorowicz
- Institute of Biochemistry and Molecular Biology, Department of Biochemistry, Semmelweis University, Budapest, Hungary
| | - Veronika J. Farkas
- Institute of Biochemistry and Molecular Biology, Department of Biochemistry, Semmelweis University, Budapest, Hungary
| | - László Szabó
- Institute of Biochemistry and Molecular Biology, Department of Biochemistry, Semmelweis University, Budapest, Hungary
- Plasma Chemistry Research Group, Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences, Budapest, Hungary
| | - Sophie Cherrington
- South Mimms Laboratories, Medicines and Healthcare Products Regulatory Agency, Potters Bar, United Kingdom
| | - Craig Thelwell
- South Mimms Laboratories, Medicines and Healthcare Products Regulatory Agency, Potters Bar, United Kingdom
| | - Krasimir Kolev
- Institute of Biochemistry and Molecular Biology, Department of Biochemistry, Semmelweis University, Budapest, Hungary
| |
Collapse
|
2
|
Hsieh RC, Liu R, Burgin DJ, Otto M. Understanding mechanisms of virulence in MRSA: implications for antivirulence treatment strategies. Expert Rev Anti Infect Ther 2023; 21:911-928. [PMID: 37501364 DOI: 10.1080/14787210.2023.2242585] [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: 05/15/2023] [Accepted: 07/26/2023] [Indexed: 07/29/2023]
Abstract
INTRODUCTION Methicillin-resistant Staphylococcus aureus (MRSA) is a widespread pathogen, often causing recurrent and deadly infections in the hospital and community. Many S. aureus virulence factors have been suggested as potential targets for antivirulence therapy to decrease the threat of diminishing antibiotic availability. Antivirulence methods hold promise due to their adjunctive and prophylactic potential and decreased risk for selective pressure. AREAS COVERED This review describes the dominant virulence mechanisms exerted by MRSA and antivirulence therapeutics that are currently undergoing testing in clinical or preclinical stages. We also discuss the advantages and downsides of several investigational antivirulence approaches, including the targeting of bacterial transporters, host-directed therapy, and quorum-sensing inhibitors. For this review, a systematic search of literature on PubMed, Google Scholar, and Web of Science for relevant search terms was performed in April and May 2023. EXPERT OPINION Vaccine and antibody strategies have failed in clinical trials and could benefit from more basic science-informed approaches. Antivirulence-targeting approaches need to be set up better to meet the requirements of drug development, rather than only providing limited results to provide 'proof-of-principle' translational value of pathogenesis research. Nevertheless, there is great potential of such strategies and potential particular promise for novel probiotic approaches.
Collapse
Affiliation(s)
- Roger C Hsieh
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases (NIAID), U.S. National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Ryan Liu
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases (NIAID), U.S. National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Dylan J Burgin
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases (NIAID), U.S. National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases (NIAID), U.S. National Institutes of Health (NIH), Bethesda, Maryland, USA
| |
Collapse
|
3
|
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: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [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
|
4
|
Schwermann N, Winstel V. Functional diversity of staphylococcal surface proteins at the host-microbe interface. Front Microbiol 2023; 14:1196957. [PMID: 37275142 PMCID: PMC10232760 DOI: 10.3389/fmicb.2023.1196957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 04/21/2023] [Indexed: 06/07/2023] Open
Abstract
Surface proteins of Gram-positive pathogens are key determinants of virulence that substantially shape host-microbe interactions. Specifically, these proteins mediate host invasion and pathogen transmission, drive the acquisition of heme-iron from hemoproteins, and subvert innate and adaptive immune cell responses to push bacterial survival and pathogenesis in a hostile environment. Herein, we briefly review and highlight the multi-facetted roles of cell wall-anchored proteins of multidrug-resistant Staphylococcus aureus, a common etiological agent of purulent skin and soft tissue infections as well as severe systemic diseases in humans. In particular, we focus on the functional diversity of staphylococcal surface proteins and discuss their impact on the variety of clinical manifestations of S. aureus infections. We also describe mechanistic and underlying principles of staphylococcal surface protein-mediated immune evasion and coupled strategies S. aureus utilizes to paralyze patrolling neutrophils, macrophages, and other immune cells. Ultimately, we provide a systematic overview of novel therapeutic concepts and anti-infective strategies that aim at neutralizing S. aureus surface proteins or sortases, the molecular catalysts of protein anchoring in Gram-positive bacteria.
Collapse
Affiliation(s)
- Nicoletta Schwermann
- Research Group Pathogenesis of Bacterial Infections, TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture Between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - Volker Winstel
- Research Group Pathogenesis of Bacterial Infections, TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture Between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
5
|
Muñoz-Wolf N, Ward RW, Hearnden CH, Sharp FA, Geoghegan J, O’Grady K, McEntee CP, Shanahan KA, Guy C, Bowie AG, Campbell M, Roces C, Anderluzzi G, Webb C, Perrie Y, Creagh E, Lavelle EC. Non-canonical inflammasome activation mediates the adjuvanticity of nanoparticles. Cell Rep Med 2023; 4:100899. [PMID: 36652908 PMCID: PMC9873954 DOI: 10.1016/j.xcrm.2022.100899] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 07/24/2022] [Accepted: 12/19/2022] [Indexed: 01/19/2023]
Abstract
The non-canonical inflammasome sensor caspase-11 and gasdermin D (GSDMD) drive inflammation and pyroptosis, a type of immunogenic cell death that favors cell-mediated immunity (CMI) in cancer, infection, and autoimmunity. Here we show that caspase-11 and GSDMD are required for CD8+ and Th1 responses induced by nanoparticulate vaccine adjuvants. We demonstrate that nanoparticle-induced reactive oxygen species (ROS) are size dependent and essential for CMI, and we identify 50- to 60-nm nanoparticles as optimal inducers of ROS, GSDMD activation, and Th1 and CD8+ responses. We reveal a division of labor for IL-1 and IL-18, where IL-1 supports Th1 and IL-18 promotes CD8+ responses. Exploiting size as a key attribute, we demonstrate that biodegradable poly-lactic co-glycolic acid nanoparticles are potent CMI-inducing adjuvants. Our work implicates ROS and the non-canonical inflammasome in the mode of action of polymeric nanoparticulate adjuvants and establishes adjuvant size as a key design principle for vaccines against cancer and intracellular pathogens.
Collapse
Affiliation(s)
- Natalia Muñoz-Wolf
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 D02 R590, Ireland,Translational & Respiratory Immunology Lab, Department of Clinical Medicine, School of Medicine, Trinity Biomedical Sciences Institute, Dublin D02 R590, Ireland,Clinical Medicine Tallaght University Hospital, Dublin D24 NR04, Ireland
| | - Ross W. Ward
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 D02 R590, Ireland
| | - Claire H. Hearnden
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 D02 R590, Ireland
| | - Fiona A. Sharp
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 D02 R590, Ireland
| | - Joan Geoghegan
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland,Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Katie O’Grady
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 D02 R590, Ireland
| | - Craig P. McEntee
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 D02 R590, Ireland
| | - Katharine A. Shanahan
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute (TBSI), Trinity College Dublin, Dublin D02 R590, Ireland
| | - Coralie Guy
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute (TBSI), Trinity College Dublin, Dublin D02 R590, Ireland
| | - Andrew G. Bowie
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute (TBSI), Trinity College Dublin, Dublin D02 R590, Ireland
| | - Matthew Campbell
- Smurfit Institute of Genetics, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland
| | - Carla.B. Roces
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK
| | - Giulia Anderluzzi
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK
| | - Cameron Webb
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK
| | - Yvonne Perrie
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK
| | - Emma Creagh
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute (TBSI), Trinity College Dublin, Dublin D02 R590, Ireland
| | - Ed C. Lavelle
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 D02 R590, Ireland,Centre for Research on Adaptive Nanostructures and Nanodevices (CRANN) & Advanced Materials Bio-Engineering Research Centre (AMBER), Trinity College Dublin, Dublin D02 PN40, Ireland,Corresponding author
| |
Collapse
|
6
|
Szafraniec GM, Szeleszczuk P, Dolka B. Review on skeletal disorders caused by Staphylococcus spp. in poultry. Vet Q 2022; 42:21-40. [PMID: 35076352 PMCID: PMC8843168 DOI: 10.1080/01652176.2022.2033880] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 12/03/2021] [Accepted: 01/21/2022] [Indexed: 12/17/2022] Open
Abstract
Lameness or leg weakness is the main cause of poor poultry welfare and serious economic losses in meat-type poultry production worldwide. Disorders related to the legs are often associated with multifactorial aetiology which makes diagnosis and proper treatment difficult. Among the infectious agents, bacteria of genus Staphylococcus are one of the most common causes of bone infections in poultry and are some of the oldest bacterial infections described in poultry. Staphylococci readily infect bones and joints and are associated with bacterial chondronecrosis with osteomyelitis (BCO), spondylitis, arthritis, tendinitis, tenosynovitis, osteomyelitis, turkey osteomyelitis complex (TOC), bumblefoot, dyschondroplasia with osteomyelitis and amyloid arthropathy. Overall, 61 staphylococcal species have been described so far, and 56% of them (34/61) have been isolated from clinical cases in poultry. Although Staphylococcus aureus is the principal cause of poultry staphylococcosis, other Staphylococcus species, such as S. agnetis, S. cohnii, S. epidermidis, S. hyicus, S. simulans, have also been isolated from skeletal lesions. Antimicrobial treatment of staphylococcosis is usually ineffective due to the location and type of lesion, as well as the possible occurrence of multidrug-resistant strains. Increasing demand for antibiotic-free farming has contributed to the use of alternatives to antibiotics. Other prevention methods, such as better management strategies, early feed restriction or use of slow growing broilers should be implemented to avoid rapid growth rate, which is associated with locomotor problems. This review aims to summarise and address current knowledge on skeletal disorders associated with Staphylococcus spp. infection in poultry.
Collapse
Affiliation(s)
- Gustaw M. Szafraniec
- Department of Pathology and Veterinary Diagnostics, Institute of Veterinary Medicine, Warsaw University of Life Sciences – SGGW, Warsaw, Poland
| | - Piotr Szeleszczuk
- Department of Pathology and Veterinary Diagnostics, Institute of Veterinary Medicine, Warsaw University of Life Sciences – SGGW, Warsaw, Poland
| | - Beata Dolka
- Department of Pathology and Veterinary Diagnostics, Institute of Veterinary Medicine, Warsaw University of Life Sciences – SGGW, Warsaw, Poland
| |
Collapse
|
7
|
Tatara AM, Gandhi RG, Mooney DJ, Nelson SB. Antiplatelet therapy for Staphylococcus aureus bacteremia: Will it stick? PLoS Pathog 2022; 18:e1010240. [PMID: 35143595 PMCID: PMC8830658 DOI: 10.1371/journal.ppat.1010240] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Staphylococcus aureus bacteremia (SAB) remains a clinically challenging infection despite extensive investigation. Repurposing medications approved for other indications is appealing as clinical safety profiles have already been established. Ticagrelor, a reversible adenosine diphosphate receptor antagonist that prevents platelet aggregation, is indicated for patients suffering from acute coronary syndrome (ACS). However, some clinical data suggest that patients treated with ticagrelor are less likely to have poor outcomes due to S. aureus infection. There are several potential mechanisms by which ticagrelor may affect S. aureus virulence. These include direct antibacterial activity, up-regulation of the innate immune system through boosting platelet-mediated S. aureus killing, and prevention of S. aureus adhesion to host tissues. In this Pearl, we review the clinical data surrounding ticagrelor and infection as well as explore the evidence surrounding these proposed mechanisms of action. While more evidence is needed before antiplatelet medications formally become part of the arsenal against S. aureus infection, these potential mechanisms represent exciting pathways to target in the host/pathogen interface.
Collapse
Affiliation(s)
- Alexander M. Tatara
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts, United States of America
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, United States of America
| | - Ronak G. Gandhi
- Department of Pharmacy, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - David J. Mooney
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts, United States of America
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, United States of America
| | - Sandra B. Nelson
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
8
|
Xia X, Liu J, Huang L, Zhang X, Deng Y, Li F, Liu Z, Huang R. Molecular Details of Actinomycin D-Treated MRSA Revealed via High-Dimensional Data. Mar Drugs 2022; 20:md20020114. [PMID: 35200643 PMCID: PMC8878686 DOI: 10.3390/md20020114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/18/2022] [Accepted: 01/27/2022] [Indexed: 02/04/2023] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is highly concerning as a principal infection pathogen. The investigation of higher effective natural anti-MRSA agents from marine Streptomyces parvulus has led to the isolation of actinomycin D, that showed potential anti-MRSA activity with MIC and MBC values of 1 and 8 μg/mL, respectively. Proteomics-metabolomics analysis further demonstrated a total of 261 differential proteins and 144 differential metabolites induced by actinomycin D in MRSA, and the co-mapped correlation network of omics, indicated that actinomycin D induced the metabolism pathway of producing the antibiotic sensitivity in MRSA. Furthermore, the mRNA expression levels of the genes acnA, ebpS, clfA, icd, and gpmA related to the key differential proteins were down-regulated measured by qRT-PCR. Molecular docking predicted that actinomycin D was bound to the targets of the two key differential proteins AcnA and Icd by hydrogen bonds and interacted with multiple amino acid residues of the proteins. Thus, these findings will provide a basic understanding to further investigation of actinomycin D as a potential anti-MRSA agent.
Collapse
Affiliation(s)
- Xuewei Xia
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China; (X.X.); (L.H.); (Y.D.); (F.L.); (Z.L.)
| | - Jun Liu
- Laboratory of Pathogenic Biology, Guangdong Medical University, Zhanjiang 524023, China;
| | - Li Huang
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China; (X.X.); (L.H.); (Y.D.); (F.L.); (Z.L.)
| | - Xiaoyong Zhang
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China;
| | - Yunqin Deng
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China; (X.X.); (L.H.); (Y.D.); (F.L.); (Z.L.)
| | - Fengming Li
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China; (X.X.); (L.H.); (Y.D.); (F.L.); (Z.L.)
| | - Zhiyuan Liu
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China; (X.X.); (L.H.); (Y.D.); (F.L.); (Z.L.)
| | - Riming Huang
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China; (X.X.); (L.H.); (Y.D.); (F.L.); (Z.L.)
- Correspondence:
| |
Collapse
|
9
|
Negrón O, Hur WS, Prasad J, Paul DS, Rowe SE, Degen JL, Abrahams SR, Antoniak S, Conlon BP, Bergmeier W, Hӧӧk M, Flick MJ. Fibrin(ogen) engagement of S. aureus promotes the host antimicrobial response and suppression of microbe dissemination following peritoneal infection. PLoS Pathog 2022; 18:e1010227. [PMID: 35041705 PMCID: PMC8797238 DOI: 10.1371/journal.ppat.1010227] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/28/2022] [Accepted: 12/22/2021] [Indexed: 12/22/2022] Open
Abstract
The blood-clotting protein fibrin(ogen) plays a critical role in host defense against invading pathogens, particularly against peritoneal infection by the Gram-positive microbe Staphylococcus aureus. Here, we tested the hypothesis that direct binding between fibrin(ogen) and S. aureus is a component of the primary host antimicrobial response mechanism and prevention of secondary microbe dissemination from the peritoneal cavity. To establish a model system, we showed that fibrinogen isolated from FibγΔ5 mice, which express a mutant form lacking the final 5 amino acids of the fibrinogen γ chain (termed fibrinogenγΔ5), did not support S. aureus adherence when immobilized and clumping when in suspension. In contrast, purified wildtype fibrinogen supported robust adhesion and clumping that was largely dependent on S. aureus expression of the receptor clumping factor A (ClfA). Following peritoneal infection with S. aureus USA300, FibγΔ5 mice displayed worse survival compared to WT mice coupled to reduced bacterial killing within the peritoneal cavity and increased dissemination of the microbes into circulation and distant organs. The failure of acute bacterial killing, but not enhanced dissemination, was partially recapitulated by mice infected with S. aureus USA300 lacking ClfA. Fibrin polymer formation and coagulation transglutaminase Factor XIII each contributed to killing of the microbes within the peritoneal cavity, but only elimination of polymer formation enhanced systemic dissemination. Host macrophage depletion or selective elimination of the fibrin(ogen) β2-integrin binding motif both compromised local bacterial killing and enhanced S. aureus systemic dissemination, suggesting fibrin polymer formation in and of itself was not sufficient to retain S. aureus within the peritoneal cavity. Collectively, these findings suggest that following peritoneal infection, the binding of S. aureus to stabilized fibrin matrices promotes a local, macrophage-mediated antimicrobial response essential for prevention of microbe dissemination and downstream host mortality. The Gram-positive bacterium Staphylococcus aureus (S. aureus) produces a number of soluble and surface-associated proteins that bind the host coagulation protein fibrinogen. The contribution of fibrinogen-S. aureus binding through the fibrinogen receptor clumping factor A (ClfA) in peritoneal infection has not been defined. Elimination of the binding motif on fibrinogen for ClfA or deletion of ClfA from S. aureus significantly reduced S. aureus-fibrinogen binding and bacterial clumping in solution. In a mouse model of peritonitis, loss of these activities resulted in diminished bacterial killing, increased bacterial dissemination, and worsened host survival. Although fibrin polymer formation and fibrin(ogen)-macrophage binding are mechanistically linked to the local antimicrobial response, fibrin formation in and of itself is not sufficient to suppress microbe dissemination. These discoveries have identified important components of the fibrin(ogen)-dependent host antimicrobial response against S. aureus, providing further understanding of this physiological response to infection which could uncover potential therapeutic strategies for peritonitis patients.
Collapse
Affiliation(s)
- Oscar Negrón
- Department of Pathology and Laboratory Medicine, UNC Blood Research Center, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Woosuk S. Hur
- Department of Pathology and Laboratory Medicine, UNC Blood Research Center, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Joni Prasad
- Division of Experimental Hematology, Cincinnati Children’s Hospital Medical Center and University of Cincinnati School of Medicine, Cincinnati, Ohio, United States of America
| | - David S. Paul
- Department of Biochemistry, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Sarah E. Rowe
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Jay L. Degen
- Division of Experimental Hematology, Cincinnati Children’s Hospital Medical Center and University of Cincinnati School of Medicine, Cincinnati, Ohio, United States of America
| | - Sara R. Abrahams
- Department of Pathology and Laboratory Medicine, UNC Blood Research Center, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Silvio Antoniak
- Department of Pathology and Laboratory Medicine, UNC Blood Research Center, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Brian P. Conlon
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Wolfgang Bergmeier
- Department of Biochemistry, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Magnus Hӧӧk
- Center of Infectious and Inflammatory Diseases, Texas A&M Health Sciences Center, Houston, Texas, United States of America
| | - Matthew J. Flick
- Department of Pathology and Laboratory Medicine, UNC Blood Research Center, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
10
|
Contribution of Coagulase and Its Regulator SaeRS to Lethality of CA-MRSA 923 Bacteremia. Pathogens 2021; 10:pathogens10111396. [PMID: 34832552 PMCID: PMC8623987 DOI: 10.3390/pathogens10111396] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/18/2021] [Accepted: 10/25/2021] [Indexed: 11/16/2022] Open
Abstract
Coagulase is a critical factor for distinguishing Staphylococcus aureus and coagulase-negative Staphylococcus. Our previous studies demonstrated that the null mutation of coagulase (coa) or its direct regulator, SaeRS, significantly enhanced the ability of S. aureus (CA-MRSA 923) to survive in human blood in vitro. This led us to further investigate the role of coagulase and its direct regulator, SaeRS, in the pathogenicity of CA-MRSA 923 in bacteremia during infection. In this study, we found that the null mutation of coa significantly decreased the mortality of CA-MRSA 923; moreover, the single null mutation of saeRS and the double deletion of coa/saeRS abolished the virulence of CA-MRSA 923. Moreover, the mice infected with either the saeRS knockout or the coa/saeRS double knockout mutant exhibited fewer histological lesions and less neutrophils infiltration in the infected kidneys compared to those infected with the coa knockout mutant or their parental control. Furthermore, we examined the impact of coa and saeRS on bacterial survival in vitro. The null mutation of coa had no impact on bacterial survival in mice blood, whereas the deletion mutation of saeRS or coa/saeRS significantly enhanced bacterial survival in mice blood. These data indicate that SaeRS plays a key role in the lethality of CA-MRSA 923 bacteremia, and that coagulase is one of the important virulence factors that is regulated by SaeRS and contributes to the pathogenicity of CA-MRSA 923.
Collapse
|
11
|
Schwarz C, Töre Y, Hoesker V, Ameling S, Grün K, Völker U, Schulze PC, Franz M, Faber C, Schaumburg F, Niemann S, Hoerr V. Host-pathogen interactions of clinical S. aureus isolates to induce infective endocarditis. Virulence 2021; 12:2073-2087. [PMID: 34490828 PMCID: PMC8425731 DOI: 10.1080/21505594.2021.1960107] [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] [Indexed: 01/29/2023] Open
Abstract
To evaluate potential pathomechanisms in the induction of infective endocarditis (IE), 34 Staphylococcus aureus (S. aureus) isolates, collected from patients with S. aureus endocarditis and from healthy individuals were investigated both in vitro and in vivo. S. aureus isolates were tested in vitro for their cytotoxicity, invasion and the association with platelets. Virulence factor expression profiles and cellular response were additionally investigated and tested for correlation with the ability of S. aureus to induce vegetations on the aortic valves in vivo. In an animal model of IE valvular conspicuity was assessed by in vivo magnetic resonance imaging at 9.4 T, histology and enrichment gene expression analysis. All S. aureus isolates tested in vivo caused a reliable infection and inflammation of the aortic valves, but could not be differentiated and categorized according to the measured in vitro virulence profiles and cytotoxicity. Results from in vitro assays did not correlate with the severity of IE. However, the isolates differed substantially in the activation and inhibition of pathways connected to the extracellular matrix and inflammatory response. Thus, comprehensive approaches of host-pathogen interactions and corresponding immune pathways are needed for the evaluation of the pathogenic capacity of bacteria. An improved understanding of the interaction between virulence factors and immune response in S. aureus infective endocarditis would offer novel possibilities for the development of therapeutic strategies and specific diagnostic imaging markers.
Collapse
Affiliation(s)
- Christian Schwarz
- Translational Research Imaging Center, Clinic for Radiology, University Hospital Muenster, Muenster, Germany
| | - Yasemin Töre
- Translational Research Imaging Center, Clinic for Radiology, University Hospital Muenster, Muenster, Germany
| | - Vanessa Hoesker
- Translational Research Imaging Center, Clinic for Radiology, University Hospital Muenster, Muenster, Germany
| | - Sabine Ameling
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Katja Grün
- Department of Internal Medicine I, Jena University Hospital, Jena, Germany
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | | | - Marcus Franz
- Department of Internal Medicine I, Jena University Hospital, Jena, Germany
| | - Cornelius Faber
- Translational Research Imaging Center, Clinic for Radiology, University Hospital Muenster, Muenster, Germany
| | - Frieder Schaumburg
- Institute of Medical Microbiology, University Hospital Muenster, Muenster, Germany
| | - Silke Niemann
- Institute of Medical Microbiology, University Hospital Muenster, Muenster, Germany
| | - Verena Hoerr
- Translational Research Imaging Center, Clinic for Radiology, University Hospital Muenster, Muenster, Germany.,Institute of Medical Microbiology, Jena University Hospital - Friedrich Schiller University Jena, Jena, Germany.,Medical Physics Group, Institute of Diagnostic and Interventional Radiology, Jena University Hospital - Friedrich Schiller University Jena, Jena, Germany
| |
Collapse
|
12
|
Abstract
Staphylococcus aureus interacts with fibrinogen in plasma to form macroscopic clumps of cells. A simple and rapid slide agglutination test using rabbit plasma has been employed in clinical labs to distinguish S. aureus from most coagulase-negative Staphylococci. The method described here is a quantitative clumping assay in which S. aureus cells are mixed with either plasma or purified fibrinogen, and clumps are allowed to sediment out of solution. Clearing of the overlying solution is monitored over time by measuring the optical density at 600 nm and comparing these values to the initial turbidity. This simple assay can be used to study regulation and expression of various cell wall-anchored adhesins.
Collapse
|
13
|
Lerche CJ, Schwartz F, Theut M, Fosbøl EL, Iversen K, Bundgaard H, Høiby N, Moser C. Anti-biofilm Approach in Infective Endocarditis Exposes New Treatment Strategies for Improved Outcome. Front Cell Dev Biol 2021; 9:643335. [PMID: 34222225 PMCID: PMC8249808 DOI: 10.3389/fcell.2021.643335] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 05/04/2021] [Indexed: 12/13/2022] Open
Abstract
Infective endocarditis (IE) is a life-threatening infective disease with increasing incidence worldwide. From early on, in the antibiotic era, it was recognized that high-dose and long-term antibiotic therapy was correlated to improved outcome. In addition, for several of the common microbial IE etiologies, the use of combination antibiotic therapy further improves outcome. IE vegetations on affected heart valves from patients and experimental animal models resemble biofilm infections. Besides the recalcitrant nature of IE, the microorganisms often present in an aggregated form, and gradients of bacterial activity in the vegetations can be observed. Even after appropriate antibiotic therapy, such microbial formations can often be identified in surgically removed, infected heart valves. Therefore, persistent or recurrent cases of IE, after apparent initial infection control, can be related to biofilm formation in the heart valve vegetations. On this background, the present review will describe potentially novel non-antibiotic, antimicrobial approaches in IE, with special focus on anti-thrombotic strategies and hyperbaric oxygen therapy targeting the biofilm formation of the infected heart valves caused by Staphylococcus aureus. The format is translational from preclinical models to actual clinical treatment strategies.
Collapse
Affiliation(s)
- Christian Johann Lerche
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Franziska Schwartz
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Marie Theut
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Emil Loldrup Fosbøl
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Kasper Iversen
- Department of Cardiology, Herlev and Gentofte Hospital, University of Copenhagen, Herlev, Denmark
- Department of Emergency Medicine, Herlev and Gentofte Hospital, University of Copenhagen, Herlev, Denmark
| | - Henning Bundgaard
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Niels Høiby
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Claus Moser
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
14
|
Jung CJ, Hsu CC, Chen JW, Cheng HW, Yuan CT, Kuo YM, Hsu RB, Chia JS. PspC domain-containing protein (PCP) determines Streptococcus mutans biofilm formation through bacterial extracellular DNA release and platelet adhesion in experimental endocarditis. PLoS Pathog 2021; 17:e1009289. [PMID: 33577624 PMCID: PMC7906467 DOI: 10.1371/journal.ppat.1009289] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 02/25/2021] [Accepted: 01/07/2021] [Indexed: 11/19/2022] Open
Abstract
Bacterial extracellular DNA (eDNA) and activated platelets have been found to contribute to biofilm formation by Streptococcus mutans on injured heart valves to induce infective endocarditis (IE), yet the bacterial component directly responsible for biofilm formation or platelet adhesion remains unclear. Using in vivo survival assays coupled with microarray analysis, the present study identified a LiaR-regulated PspC domain-containing protein (PCP) in S. mutans that mediates bacterial biofilm formation in vivo. Reverse transcriptase- and chromatin immunoprecipitation-polymerase chain reaction assays confirmed the regulation of pcp by LiaR, while PCP is well-preserved among streptococcal pathogens. Deficiency of pcp reduced in vitro and in vivo biofilm formation and released the eDNA inside bacteria floe along with reduced bacterial platelet adhesion capacity in a fibrinogen-dependent manner. Therefore, LiaR-regulated PCP alone could determine release of bacterial eDNA and binding to platelets, thus contributing to biofilm formation in S. mutans-induced IE.
Collapse
Affiliation(s)
- Chiau-Jing Jung
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chih-Chieh Hsu
- Graduate Institute of Oral Biology, School of Dentistry, National Taiwan University, Taipei, Taiwan
| | - Jeng-Wei Chen
- Division of Cardiovascular Surgery, Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hung-Wei Cheng
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chang-Tsu Yuan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Pathology, National Taiwan University Cancer Center, Taipei, Taiwan
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
| | - Yu-Min Kuo
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ron-Bin Hsu
- Division of Cardiovascular Surgery, Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Jean-San Chia
- Graduate Institute of Oral Biology, School of Dentistry, National Taiwan University, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
15
|
Schnurr E, Paqué PN, Attin T, Nanni P, Grossmann J, Holtfreter S, Bröker BM, Kohler C, Diep BA, Ribeiro ADA, Thurnheer T. Staphylococcus aureus Interferes with Streptococci Spatial Distribution and with Protein Expression of Species within a Polymicrobial Oral Biofilm. Antibiotics (Basel) 2021; 10:116. [PMID: 33530340 PMCID: PMC7911025 DOI: 10.3390/antibiotics10020116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 11/21/2022] Open
Abstract
We asked whether transient Staphylococcus aureus in the oral environment synergistically interacts with orally associated bacterial species such as Actinomyces oris, Candida albicans, Fusobacterium nucleatum, Streptococcus oralis, Streptococcus mutans, and Veillonella dispar (six-species control biofilm 6S). For this purpose, four modified biofilms with seven species that contain either the wild type strain of the S. aureus genotype (USA300-MRSA WT), its isogenic mutant with MSCRAMM deficiency (USA300-MRSA ΔMSCRAMM), a methicillin-sensitive S. aureus (ST72-MSSA-) or a methicillin-resistant S. aureus (USA800-MRSA) grown on hydroxyapatite disks were examined. Culture analyses, confocal-laser-scanning microscopy and proteome analyses were performed. S. aureus strains affected the amount of supragingival biofilm-associated species differently. The deletion of MSCRAMM genes disrupted the growth of S. aureus and the distribution of S. mutans and S. oralis within the biofilms. In addition, S. aureus caused shifts in the number of detectable proteins of other species in the 6S biofilm. S. aureus (USA300-MRSA WT), aggregated together with early colonizers such as Actinomyces and streptococci, influenced the number of secondary colonizers such as Fusobacterium nucleatum and was involved in structuring the biofilm architecture that triggered the change from a homeostatic biofilm to a dysbiotic biofilm to the development of oral diseases.
Collapse
Affiliation(s)
- Etyene Schnurr
- Instituto de Saúde de Nova Friburgo, Federal Fluminense University, 28625-650 Nova Friburgo, Brazil
| | - Pune N. Paqué
- Clinic of Conservative and Preventive Dentistry, Center of Dental Medicine, University of Zurich, 8032 Zurich, Switzerland; (P.N.P.); (T.A.); (T.T.)
| | - Thomas Attin
- Clinic of Conservative and Preventive Dentistry, Center of Dental Medicine, University of Zurich, 8032 Zurich, Switzerland; (P.N.P.); (T.A.); (T.T.)
| | - Paolo Nanni
- Functional Genomics Center, ETH Zürich and University of Zurich, 8057 Zurich, Switzerland; (P.N.); (J.G.)
| | - Jonas Grossmann
- Functional Genomics Center, ETH Zürich and University of Zurich, 8057 Zurich, Switzerland; (P.N.); (J.G.)
- SIB Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Silva Holtfreter
- Department of Immunology, University Medicine Greifswald, 17475 Greifswald, Germany; (S.H.); (B.M.B.)
| | - Barbara M. Bröker
- Department of Immunology, University Medicine Greifswald, 17475 Greifswald, Germany; (S.H.); (B.M.B.)
| | - Christian Kohler
- Friedrich-Loeffler Institute for Medical Microbiology, University Medicine Greifswald, 17475 Greifswald, Germany;
| | - Binh An Diep
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California San Francisco, San Francisco, CA 94143, USA;
| | | | - Thomas Thurnheer
- Clinic of Conservative and Preventive Dentistry, Center of Dental Medicine, University of Zurich, 8032 Zurich, Switzerland; (P.N.P.); (T.A.); (T.T.)
| |
Collapse
|
16
|
Staphylococcus lugdunensis: a Skin Commensal with Invasive Pathogenic Potential. Clin Microbiol Rev 2020; 34:34/2/e00205-20. [PMID: 33361142 DOI: 10.1128/cmr.00205-20] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Staphylococcus lugdunensis is a species of coagulase-negative staphylococcus (CoNS) that causes serious infections in humans akin to those of S. aureus It was often misidentified as S. aureus, but this has been rectified by recent routine use of matrix-assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF MS) in diagnostic laboratories. It encodes a diverse array of virulence factors for adhesion, cytotoxicity, and innate immune evasion, but these are less diverse than those encoded by S. aureus It expresses an iron-regulated surface determinant (Isd) system combined with a novel energy-coupling factor (ECF) mechanism for extracting heme from hemoproteins. Small cytolytic S. lugdunensis synergistic hemolysins (SLUSH), peptides related to phenol-soluble modulins of S. aureus, act synergistically with β-toxin to lyse erythrocytes. S. lugdunensis expresses a novel peptide antibiotic, lugdunin, that can influence the nasal and skin microbiota. Endovascular infections are initiated by bacterial adherence to fibrinogen promoted by a homologue of Staphylococcus aureus clumping factor A and to von Willebrand factor on damaged endothelium by an uncharacterized mechanism. S. lugdunensis survives within mature phagolysosomes of macrophages without growing and is released only following apoptosis. This differs fundamentally from S. aureus, which actively grows and expresses bicomponent leukotoxins that cause membrane damage and could contribute to survival in the infected host. S. lugdunensis is being investigated as a probiotic to eradicate S. aureus from the nares of carriers. However, this is contraindicated by its innate virulence. Studies to obtain a deeper understanding of S. lugdunensis colonization, virulence, and microbiome interactions are therefore warranted.
Collapse
|
17
|
Petrie LE, Leonard AC, Murphy J, Cox G. Development and validation of a high-throughput whole cell assay to investigate Staphylococcus aureus adhesion to host ligands. J Biol Chem 2020; 295:16700-16712. [PMID: 32978256 PMCID: PMC7864066 DOI: 10.1074/jbc.ra120.015360] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/22/2020] [Indexed: 12/20/2022] Open
Abstract
Staphylococcus aureus adhesion to the host's skin and mucosae enables asymptomatic colonization and the establishment of infection. This process is facilitated by cell wall-anchored adhesins that bind to host ligands. Therapeutics targeting this process could provide significant clinical benefits; however, the development of anti-adhesives requires an in-depth knowledge of adhesion-associated factors and an assay amenable to high-throughput applications. Here, we describe the development of a sensitive and robust whole cell assay to enable the large-scale profiling of S. aureus adhesion to host ligands. To validate the assay, and to gain insight into cellular factors contributing to adhesion, we profiled a sequence-defined S. aureus transposon mutant library, identifying mutants with attenuated adhesion to human-derived fibronectin, keratin, and fibrinogen. Our screening approach was validated by the identification of known adhesion-related proteins, such as the housekeeping sortase responsible for covalently linking adhesins to the cell wall. In addition, we also identified genetic loci that could represent undescribed anti-adhesive targets. To compare and contrast the genetic requirements of adhesion to each host ligand, we generated a S. aureus Genetic Adhesion Network, which identified a core gene set involved in adhesion to all three host ligands, and unique genetic signatures. In summary, this assay will enable high-throughput chemical screens to identify anti-adhesives and our findings provide insight into the target space of such an approach.
Collapse
Affiliation(s)
- Laurenne E Petrie
- College of Biological Sciences, Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Allison C Leonard
- College of Biological Sciences, Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Julia Murphy
- College of Biological Sciences, Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Georgina Cox
- College of Biological Sciences, Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada.
| |
Collapse
|
18
|
Maddur AA, Kroh HK, Aschenbrenner ME, Gibson BHY, Panizzi P, Sheehan JH, Meiler J, Bock PE, Verhamme IM. Specificity and affinity of the N-terminal residues in staphylocoagulase in binding to prothrombin. J Biol Chem 2020; 295:5614-5625. [PMID: 32156702 PMCID: PMC7186164 DOI: 10.1074/jbc.ra120.012588] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/09/2020] [Indexed: 11/06/2022] Open
Abstract
In Staphylococcus aureus-caused endocarditis, the pathogen secretes staphylocoagulase (SC), thereby activating human prothrombin (ProT) and evading immune clearance. A previous structural comparison of the SC(1-325) fragment bound to thrombin and its inactive precursor prethrombin 2 has indicated that SC activates ProT by inserting its N-terminal dipeptide Ile1-Val2 into the ProT Ile16 pocket, forming a salt bridge with ProT's Asp194, thereby stabilizing the active conformation. We hypothesized that these N-terminal SC residues modulate ProT binding and activation. Here, we generated labeled SC(1-246) as a probe for competitively defining the affinities of N-terminal SC(1-246) variants preselected by modeling. Using ProT(R155Q,R271Q,R284Q) (ProTQQQ), a variant refractory to prothrombinase- or thrombin-mediated cleavage, we observed variant affinities between ∼1 and 650 nm and activation potencies ranging from 1.8-fold that of WT SC(1-246) to complete loss of function. Substrate binding to ProTQQQ caused allosteric tightening of the affinity of most SC(1-246) variants, consistent with zymogen activation through occupation of the specificity pocket. Conservative changes at positions 1 and 2 were well-tolerated, with Val1-Val2, Ile1-Ala2, and Leu1-Val2 variants exhibiting ProTQQQ affinity and activation potency comparable with WT SC(1-246). Weaker binding variants typically had reduced activation rates, although at near-saturating ProTQQQ levels, several variants exhibited limiting rates similar to or higher than that of WT SC(1-246). The Ile16 pocket in ProTQQQ appears to favor nonpolar, nonaromatic residues at SC positions 1 and 2. Our results suggest that SC variants other than WT Ile1-Val2-Thr3 might emerge with similar ProT-activating efficiency.
Collapse
Affiliation(s)
- Ashoka A Maddur
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232-2561.
| | - Heather K Kroh
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232-2561
| | - Mary E Aschenbrenner
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232-2561
| | - Breanne H Y Gibson
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232-2561
| | - Peter Panizzi
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, Alabama 36849
| | - Jonathan H Sheehan
- Center for Structural Biology, Vanderbilt University, Nashville, Tennessee 37232
| | - Jens Meiler
- Center for Structural Biology, Vanderbilt University, Nashville, Tennessee 37232; Institute for Drug Discovery, Departments of Chemistry and Computer Science, Leipzig University Medical School, SAC 04103 Leipzig, Germany
| | - Paul E Bock
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232-2561
| | - Ingrid M Verhamme
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232-2561.
| |
Collapse
|
19
|
Farkas ÁZ, Farkas VJ, Szabó L, Wacha A, Bóta A, Csehi L, Kolev K, Thelwell C. Structure, Mechanical, and Lytic Stability of Fibrin and Plasma Coagulum Generated by Staphylocoagulase From Staphylococcus aureus. Front Immunol 2019; 10:2967. [PMID: 31921206 PMCID: PMC6933771 DOI: 10.3389/fimmu.2019.02967] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 12/03/2019] [Indexed: 12/18/2022] Open
Abstract
Staphylococcus aureus causes localized infections or invasive diseases (abscesses or endocarditis). One of its virulence factors is staphylocoagulase (SCG), which binds prothrombin to form a complex with thrombin-like proteolytic activity and leads to uncontrolled fibrin generation at sites of bacterial inoculation. The aim of this study was to characterize the formation, structure, mechanical properties and lysis of SCG-generated clots. Recombinant SCG was expressed in Escherichia coli, purified and the amidolytic activity of its complexes with human prothrombin (SCG-PT) and thrombin (SCG-T) was determined using human thrombin as a reference. Fibrin clots were prepared from purified fibrinogen and human plasma using thrombin, SCG-PT or SCG-T as a coagulase. The kinetics of clot formation and lysis by tissue-type plasminogen activator (tPA) were monitored with turbidimetric assays. Fibrin ultrastructure was examined with scanning electron microscopy and small-angle X-ray scattering (SAXS). Fibrin clot porosity was characterized with fluid permeation assays, whereas the viscoelastic properties and mechanical stability were evaluated with oscillation rheometry. Compared to thrombin, the amidolytic and clotting activity of SCG-PT was 1.6- to 2.5-fold lower on a molar basis. SCG-T had equivalent amidolytic, but reduced clotting activity both on pure fibrinogen (1.6-fold), and in plasma (1.3-fold). The SCG-PT and SCG-T generated fibrin with thicker fibers (10-60% increase in median diameter) than thrombin due to increased number of fibrin protofibrils per fiber cross-section. According to the fluid permeability of the clots SCG-PT and SCG-T promoted the formation of more porous structures. The shear stress resistance in the pure fibrin and plasma clots generated by SCG-PT was significantly lower than in the thrombin clots (243.8 ± 22.0 Pa shear stress was sufficient for disassembly of SCG-PT fibrin vs. 937.3 ± 65.6 Pa in thrombin clots). The tPA-mediated lysis of both pure fibrin and plasma clots produced by SCG-PT or SCG-T was accelerated compared to thrombin, resulting in up to a 2.1-fold increase in tPA potency. Our results indicate that SCG generates a thrombus scaffold with a structure characterized by impaired mechanical stability and increased lytic susceptibility. This proneness to clot disintegration could have implications in the septic embolism from endocardial bacterial vegetation.
Collapse
Affiliation(s)
- Ádám Z Farkas
- Department of Medical Biochemistry, Semmelweis University, Budapest, Hungary
| | - Veronika J Farkas
- Department of Medical Biochemistry, Semmelweis University, Budapest, Hungary
| | - László Szabó
- Department of Medical Biochemistry, Semmelweis University, Budapest, Hungary
| | - András Wacha
- Biological Nanochemistry Research Group, Research Centre for Natural Sciences, Budapest, Hungary
| | - Attila Bóta
- Biological Nanochemistry Research Group, Research Centre for Natural Sciences, Budapest, Hungary
| | - Lóránt Csehi
- Department of Medical Biochemistry, Semmelweis University, Budapest, Hungary
| | - Krasimir Kolev
- Department of Medical Biochemistry, Semmelweis University, Budapest, Hungary
| | - Craig Thelwell
- Haemostasis Section, Biotherapeutics Group, National Institute for Standards and Control, Potters Bar, United Kingdom
| |
Collapse
|
20
|
Horino T, Hori S. Metastatic infection during Staphylococcus aureus bacteremia. J Infect Chemother 2019; 26:162-169. [PMID: 31676266 DOI: 10.1016/j.jiac.2019.10.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 07/25/2019] [Accepted: 10/03/2019] [Indexed: 12/17/2022]
Abstract
Staphylococcus aureus causes various infections, including skin and soft tissue infections and pneumonia via both, community-associated and nosocomial infection. These infectious diseases can lead to bacteremia, and may subsequently result in metastatic infections in several cases. Metastatic infections are critical complications in patients with S. aureus bacteremia, since the optimal duration of the antimicrobial treatment differs in patients with and without metastatic infection. Notably, two weeks of antimicrobial treatment is recommended in case of uncomplicated S. aureus bacteremia, whereas in patients with S. aureus bacteremia-associated endocarditis or vertebral osteomyelitis, six weeks of antimicrobial administration is vital. In addition, misdiagnosis or insufficient treatment in metastatic infection is associated with poor prognosis, functional disability, and relapse. Although echocardiography is recommended to examine endocarditis in the patients with S. aureus bacteremia, it remains unclear which patients should undergo additional examinations, such as CT and MRI, to detect the presence of other metastatic infections. Clinical studies have revealed that permanent foreign body and persistent bacteremia are predictive factors for metastatic infections, and experimental studies have demonstrated that the virulence factors of S. aureus, such as fnbA and clfA, are associated with endocarditis; however, these factors are not proven to increase the risk of metastatic infections. In this review, we assessed the incidence, predictive factors, diagnosis, and treatment for metastatic infections during S. aureus bacteremia.
Collapse
Affiliation(s)
- Tetsuya Horino
- Department of Infectious Diseases and Infection Control, Jikei University School of Medicine, Japan.
| | - Seiji Hori
- Department of Infectious Diseases and Infection Control, Jikei University School of Medicine, Japan
| |
Collapse
|
21
|
The MSCRAMM Family of Cell-Wall-Anchored Surface Proteins of Gram-Positive Cocci. Trends Microbiol 2019; 27:927-941. [PMID: 31375310 DOI: 10.1016/j.tim.2019.06.007] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 06/10/2019] [Accepted: 06/19/2019] [Indexed: 01/21/2023]
Abstract
The microbial surface components recognizing adhesive matrix molecules (MSCRAMMs) are a family of proteins that are defined by the presence of two adjacent IgG-like folded subdomains. These promote binding to ligands by mechanisms that involve major conformational changes exemplified by the binding to fibrinogen by the 'dock-lock-latch' mechanism or to collagen by the 'collagen hug'. Clumping factors A and B are two such MSCRAMMs that have several important roles in the pathogenesis of Staphylococcus aureus infections. MSCRAMM architecture, ligand binding, and roles in infection and colonization are examined with a focus on recent developments with clumping factors.
Collapse
|
22
|
Foster TJ. Surface Proteins of Staphylococcus aureus. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0046-2018. [PMID: 31267926 PMCID: PMC10957221 DOI: 10.1128/microbiolspec.gpp3-0046-2018] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Indexed: 12/20/2022] Open
Abstract
The surface of Staphylococcus aureus is decorated with over 20 proteins that are covalently anchored to peptidoglycan by the action of sortase A. These cell wall-anchored (CWA) proteins can be classified into several structural and functional groups. The largest is the MSCRAMM family, which is characterized by tandemly repeated IgG-like folded domains that bind peptide ligands by the dock lock latch mechanism or the collagen triple helix by the collagen hug. Several CWA proteins comprise modules that have different functions, and some individual domains can bind different ligands, sometimes by different mechanisms. For example, the N-terminus of the fibronectin binding proteins comprises an MSCRAMM domain which binds several ligands, while the C-terminus is composed of tandem fibronectin binding repeats. Surface proteins promote adhesion to host cells and tissue, including components of the extracellular matrix, contribute to biofilm formation by stimulating attachment to the host or indwelling medical devices followed by cell-cell accumulation via homophilic interactions between proteins on neighboring cells, help bacteria evade host innate immune responses, participate in iron acquisition from host hemoglobin, and trigger invasion of bacteria into cells that are not normally phagocytic. The study of genetically manipulated strains using animal infection models has shown that many CWA proteins contribute to pathogenesis. Fragments of CWA proteins have the potential to be used in multicomponent vaccines to prevent S. aureus infections.
Collapse
|
23
|
Staphylococcus aureus aggregation in the plasma fraction of silkworm hemolymph. PLoS One 2019; 14:e0217517. [PMID: 31145754 PMCID: PMC6542516 DOI: 10.1371/journal.pone.0217517] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 05/13/2019] [Indexed: 11/19/2022] Open
Abstract
Staphylococcus aureus formed bacterial aggregates in the plasma fraction of the hemolymph of silkworm, the larva of Bombyx mori, in a growth-dependent manner. The addition of arabinose or galactose inhibited the formation of S. aureus aggregates in the silkworm plasma. Formation of the bacterial aggregates depended on S. aureus genes required for the synthesis of bacterial surface polysaccharides–ypfP and ltaA, which are involved in lipoteichoic acid synthesis, and the tagO gene, which is involved in wall teichoic acid synthesis. These findings suggest that S. aureus forms bacterial aggregates in the silkworm plasma via bacterial surface teichoic acids.
Collapse
|
24
|
Sheehan G, Dixon A, Kavanagh K. Utilization of Galleria mellonella larvae to characterize the development of Staphylococcus aureus infection. MICROBIOLOGY-SGM 2019; 165:863-875. [PMID: 31107207 DOI: 10.1099/mic.0.000813] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Staphylococcus aureus is a human opportunistic pathogen that causes a wide range of superficial and systemic infections in susceptible patients. Here we describe how an inoculum of S. aureus activates the cellular and humoral response of Galleria mellonella larvae while growing and disseminating throughout the host, forming nodules and ultimately killing the host. An inoculum of S. aureus (2×106 larva- 1 ) decreased larval viability at 24 (80±5.77 %), 48 (55.93±5.55 %) and 72 h (10.23±2.97 %) and was accompanied by significant proliferation and dissemination of S. aureus between 6 and 48 h and the formation of nodules in the host. The hemocyte (immune cell) densities increased between 4 and 24 h and hemocytes isolated from larvae after 24 h exposure to heat-killed S. aureus (2×106 larva- 1 ) showed altered killing kinetics as compared to those from control larvae. Alterations in the humoral immune response of larvae 6 and 24 h post-infection were also determined by quantitative shotgun proteomics. The proteome of 6 h-infected larvae was enriched for antimicrobial proteins, proteins of the prophenoloxidase cascade and a range of peptidoglycan recognition proteins. By 24 h there was a significant increase in the abundance of a range of antimicrobial peptides with anti-staphylococcal activity and proteins associated with nodule formation. The results presented here indicate how S. aureus interacts with the larval immune response, induces the expression of a variety of immune-related peptides and also forms nodules which are a hallmark of soft tissue infections during human infection.
Collapse
Affiliation(s)
- Gerard Sheehan
- Department of Biology, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - Amy Dixon
- Department of Biology, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - Kevin Kavanagh
- Department of Biology, Maynooth University, Maynooth, Co. Kildare, Ireland
| |
Collapse
|
25
|
Brignoli T, Manetti AGO, Rosini R, Haag AF, Scarlato V, Bagnoli F, Delany I. Absence of Protein A Expression Is Associated With Higher Capsule Production in Staphylococcal Isolates. Front Microbiol 2019; 10:863. [PMID: 31133995 PMCID: PMC6523524 DOI: 10.3389/fmicb.2019.00863] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 04/04/2019] [Indexed: 12/19/2022] Open
Abstract
Staphylococcus aureus is a major human pathogen, and a leading cause of soft tissue and blood stream infections. One of the causes of its success as a pathogen is the peculiar array of immune evasion factors through which the bacterium avoids host defenses, where the staphylococcal protein A (SpA) plays a major role thanks to its IgG binding activities. Moreover, SpA has recently been proposed as a promising vaccine antigen. In this study, we evaluated the expression of SpA in a collection of staphylococcal strains, about 7% of which did not express SpA (SpA- strains), despite the presence of the gene. By a comparative genomic analysis, we identified that a mutation in the spa 5′ UTR sequence affecting the RBS is responsible for the loss of SpA in a subset of SpA- strains. Using a high-throughput qRT-PCR approach on a selected panel of virulence-related genes, we identified that the SpA- phenotype is associated with lower spa transcript levels and increased expression and production of capsule as well as other changes in the transcription of several key virulence factors. Our data suggest that the SpA- phenotype has occurred in geographically distinct strains through different molecular mechanisms including both mutation, leading likely to translation alterations, and transcriptional deregulation. Furthermore, we provide evidence that SpA- strains are highly susceptible to phagocytic uptake mediated by anti-capsule antibodies. These data suggest that S. aureus may alter its virulence factor expression pattern as an adaptation to the host or environment. Vaccination strategies targeting both SpA and capsule could therefore result in broader coverage against staphylococcal isolates than SpA alone.
Collapse
Affiliation(s)
- Tarcisio Brignoli
- GSK Vaccines, Siena, Italy.,Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Bologna, Italy
| | | | | | - Andreas F Haag
- GSK Vaccines, Siena, Italy.,Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Vincenzo Scarlato
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Bologna, Italy
| | | | | |
Collapse
|
26
|
Laumay F, Corvaglia AR, Diene SM, Girard M, Oechslin F, van der Mee-Marquet N, Entenza JM, François P. Temperate Prophages Increase Bacterial Adhesin Expression and Virulence in an Experimental Model of Endocarditis Due to Staphylococcus aureus From the CC398 Lineage. Front Microbiol 2019; 10:742. [PMID: 31105650 PMCID: PMC6492496 DOI: 10.3389/fmicb.2019.00742] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 03/25/2019] [Indexed: 01/13/2023] Open
Abstract
Until 2007, Staphylococcus aureus from clonal complex 398 (CC398) was exclusively associated with livestock species and companion animals. Recently, several studies described the emergence of S. aureus CC398 as etiologies of severe infections in humans living in an animal-free environment. Recent sequencing efforts showed that the mobile genetic elements found in CC398 isolates were specific for each population and enabled differentiation of strains responsible for asymptomatic colonization from strains involved in bloodstream infections. We mobilized prophages from a human CC398 isolate and introduced them into two naïve ancestral isolates devoid of prophages that exclusively colonize animals. These lysogenized ancestral CC398 isolates acquired features related to virulence, such as an increased capacity to adhere to human extracellular matrix proteins and the ability to invade and survive within non-phagocytic cells. Pathogenicity of several clinical isolates from the CC398 lineage as well as ancestral and in vitro lysogenized ancestral counterparts was assessed in a model of infectious endocarditis in rats. Natural and artificial lysogens were not only more invasive than their prophage-free parent but also showed an increased capacity to multiply within aortic vegetations. This study identified prophages as mediators of bacterial virulence in a model of infectious endocarditis, probably through promotion of interaction with extracellular matrix components. Further studies are needed to identify mechanisms leading to promotion of intrinsic virulence.
Collapse
Affiliation(s)
- Floriane Laumay
- Genomic Research Laboratory, Service of Infectious Diseases, Medical University Center, Geneva University Hospitals, Geneva, Switzerland
| | - Anna-Rita Corvaglia
- Genomic Research Laboratory, Service of Infectious Diseases, Medical University Center, Geneva University Hospitals, Geneva, Switzerland
| | - Seydina M Diene
- Unité de Recherche sur les Maladies Infectieuses et Tropicales Émergentes, Faculté de Médecine et de Pharmacie, Aix-Marseille University, Marseille, France
| | - Myriam Girard
- Genomic Research Laboratory, Service of Infectious Diseases, Medical University Center, Geneva University Hospitals, Geneva, Switzerland
| | - Frank Oechslin
- Department of Fundamental Microbiology, University of Lausanne, Lausanne, Switzerland
| | | | - José Manuel Entenza
- Department of Fundamental Microbiology, University of Lausanne, Lausanne, Switzerland
| | - Patrice François
- Genomic Research Laboratory, Service of Infectious Diseases, Medical University Center, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
27
|
Mancini S, Oechslin F, Menzi C, Que YA, Claes J, Heying R, Veloso TR, Vanassche T, Missiakas D, Schneewind O, Moreillon P, Entenza JM. Marginal role of von Willebrand factor-binding protein and coagulase in the initiation of endocarditis in rats with catheter-induced aortic vegetations. Virulence 2019; 9:1615-1624. [PMID: 30280967 PMCID: PMC7000203 DOI: 10.1080/21505594.2018.1528845] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Staphylococcus aureus is the leading cause of infective endocarditis (IE). While the role of S. aureus cell-wall associated protein clumping factor A (ClfA) in promoting IE has been already demonstrated, that of the secreted plasma-clotting factors staphylocoagulase (Coa) and von Willebrand factor-binding protein (vWbp) has not yet been elucidated. We investigated the role of Coa and vWbp in IE initiation in rats with catheter-induced aortic vegetations, using Lactococcus lactis expressing coa, vWbp, clfA or vWbp/clfA, and S. aureus Newman Δcoa, ΔvWbp, ΔclfA or Δcoa/ΔvWbp/ΔclfA mutants. vWbp-expression increased L. lactis valve infection compared to parent and coa-expressing strains (incidence: 62%, versus 0% and 13%, respectively; P < 0.01). Likewise, expression of clfA increased L. lactis infectivity (incidence: 80%), which was not further affected by co-expression of vWbp. In symmetry, deletion of the coa or vWbp genes in S. aureus did not decrease infectivity (incidence: 68 and 64%, respectively) whereas deletion of clfA did decrease valve infection (incidence: 45%; P = 0.03 versus parent), which was not further affected by the triple deletion Δcoa/ΔvWbp/ΔclfA (incidence: 36%; P > 0.05 versus ΔclfA mutant). Coa does not support the initial colonization of IE (in L. lactis) without other key virulence factors and vWbp contributes to initiation of IE (in L. lactis) but is marginal in the present of ClfA.
Collapse
Affiliation(s)
- Stefano Mancini
- a Department of Fundamental Microbiology , University of Lausanne , Lausanne , Switzerland
| | - Frank Oechslin
- a Department of Fundamental Microbiology , University of Lausanne , Lausanne , Switzerland
| | - Carmen Menzi
- a Department of Fundamental Microbiology , University of Lausanne , Lausanne , Switzerland
| | - Yok Ai Que
- b Department of Intensive Care Medicine , Bern University Hospital , Bern , Switzerland
| | - Jorien Claes
- c Cardiovascular Developmental Biology, Department of Cardiovascular Sciences , KU Leuven , Leuven , Belgium.,d Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences , KU Leuven , Leuven , Belgium
| | - Ruth Heying
- c Cardiovascular Developmental Biology, Department of Cardiovascular Sciences , KU Leuven , Leuven , Belgium
| | - Tiago Rafael Veloso
- a Department of Fundamental Microbiology , University of Lausanne , Lausanne , Switzerland
| | - Thomas Vanassche
- d Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences , KU Leuven , Leuven , Belgium
| | | | - Olaf Schneewind
- e Department of Microbiology , University of Chicago , Chicago , IL , USA
| | - Philippe Moreillon
- a Department of Fundamental Microbiology , University of Lausanne , Lausanne , Switzerland
| | - José Manuel Entenza
- a Department of Fundamental Microbiology , University of Lausanne , Lausanne , Switzerland
| |
Collapse
|
28
|
Thomas S, Liu W, Arora S, Ganesh V, Ko YP, Höök M. The Complex Fibrinogen Interactions of the Staphylococcus aureus Coagulases. Front Cell Infect Microbiol 2019; 9:106. [PMID: 31041195 PMCID: PMC6476931 DOI: 10.3389/fcimb.2019.00106] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 03/27/2019] [Indexed: 11/22/2022] Open
Abstract
The two coagulases, von Willebrand factor binding protein (vWbp) and Coagulase (Coa), are critical virulence factors in several animal models of invasive Staphylococcus aureus (S. aureus) infections. These proteins are part of an intricate system of proteins that S. aureus uses to assemble a fibrinogen (Fg)/fibrin protective shield surrounding itself. This shield allows the microorganism to evade clearance by the host phagocytic cells. The coagulases can non-proteolytically activate the zymogen prothrombin to convert Fg to fibrin and promote the Fg/fibrin shield formation. The coagulases also bind directly to Fg and the interaction between Coa and Fg has been previously characterized in some detail. However, the mechanism(s) by which vWbp interacts with Fg remains unclear. Here, we show that vWbp and Coa have distinct interactions with Fg, despite being structurally similar. Coa binds with a significantly higher affinity to soluble Fg than to Fg coated on a plastic surface, whereas vWbp demonstrates no preference between the two forms of Fg. The two coagulases appear to target different sites on Fg, as they do not compete with each other in binding to Fg. Similar to Coa, both the N- and C-terminal halves of vWbp (vWbp-N, vWbp-C, respectively) harbor Fg-binding activities. The higher affinity Fg-binding activity resides in vWbp-N; whereas, the C-terminal region of Coa encompasses the major Fg-binding activity. Peptides constituting the previously identified Coa/Efb1 Fg-binding motif fail to inhibit vWbp-C from binding to Fg, indicating that vWbp-C lacks a functional homolog to this motif. Interestingly, the N-terminal prothrombin-binding domains of both coagulases recognize the Fg β-chain, but they appear to interact with different sequence motifs in the host protein. Collectively, our data provide insight into the complex interactions between Fg and the S. aureus coagulases.
Collapse
Affiliation(s)
- Sheila Thomas
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX, United States
| | - Wen Liu
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX, United States
| | - Srishtee Arora
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX, United States
| | - Vannakambodi Ganesh
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX, United States
| | - Ya-Ping Ko
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX, United States
| | - Magnus Höök
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX, United States
| |
Collapse
|
29
|
Zheng Y, Shang W, Peng H, Rao Y, Zhao X, Hu Z, Yang Y, Hu Q, Tan L, Xiong K, Li S, Zhu J, Hu X, Zhou R, Li M, Rao X. Virulence Determinants Are Required for Brain Abscess Formation Through Staphylococcus aureus Infection and Are Potential Targets of Antivirulence Factor Therapy. Front Microbiol 2019; 10:682. [PMID: 31024479 PMCID: PMC6460967 DOI: 10.3389/fmicb.2019.00682] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 03/19/2019] [Indexed: 11/24/2022] Open
Abstract
Bacterial brain abscesses (BAs) are difficult to treat with conventional antibiotics. Thus, the development of alternative therapeutic strategies for BAs is of high priority. Identifying the virulence determinants that contribute to BA formation induced by Staphylococcus aureus would improve the effectiveness of interventions for this disease. In this study, RT-qPCR was performed to compare the expression levels of 42 putative virulence determinants of S. aureus strains Newman and XQ during murine BA formation, ear colonization, and bacteremia. The alterations in the expression levels of 23 genes were further confirmed through specific TaqMan RT-qPCR. Eleven S. aureus genes that persistently upregulated expression levels during BA infection were identified, and their functions in BA formation were confirmed through isogenic mutant experiments. Bacterial loads and BA volumes in mice infected with isdA, isdC, lgt, hla, or spa deletion mutants and the hla/spa double mutant strain were lower than those in mice infected with the wild-type Newman strain. The therapeutic application of monoclonal antibodies against Hla and SpA decreased bacterial loads and BA volume in mice infected with Newman. This study provides insights into the virulence determinants that contribute to staphylococcal BA formation and a paradigm for antivirulence factor therapy against S. aureus infections.
Collapse
Affiliation(s)
- Ying Zheng
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, China
| | - Weilong Shang
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, China
| | - Huagang Peng
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, China
| | - Yifan Rao
- Institute of Modern Biopharmaceuticals, School of Life Sciences, Southwest University, Chongqing, China
| | - Xia Zhao
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, China
| | - Zhen Hu
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, China
| | - Yi Yang
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, China
| | - Qiwen Hu
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, China
| | - Li Tan
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, China
| | - Kun Xiong
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, China
| | - Shu Li
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, China
| | - Junmin Zhu
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, China
| | - Xiaomei Hu
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, China
| | - Renjie Zhou
- Department of Emergency, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Ming Li
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, China
| | - Xiancai Rao
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, China
| |
Collapse
|
30
|
Ejiofor OS, Ajunwa OM, Ezeudu CE, Emechebe GO, Okeke KN, Ifezulike CC, Ekejindu IM, Okoyeh JN, Osuala EO, Oli AN. The Bacteriology and Its Virulence Factors in Neonatal Infections: Threats to Child Survival Strategies. J Pathog 2018; 2018:4801247. [PMID: 30112215 PMCID: PMC6077539 DOI: 10.1155/2018/4801247] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 04/23/2018] [Accepted: 05/19/2018] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Neonatal infection refers to the infection of the newborn during the first twenty-eight days of life. It is one of the causes of infant morbidity and mortality worldwide. The aim of the study is to determine the relative contribution of the different pathogens to the overall disease burden. It will also determine the mechanisms of virulence of these pathogens that cause neonatal infections at Chukwuemeka Odumegwu Ojukwu University Teaching Hospital (COOUTH), Awka. METHODS Biological samples were collected from 30 neonates admitted at the special care baby unit (SCBU) of COOUTH and cultured using selective media and nutrient agar. The isolates were identified using microbiological and biochemical tests. The antibiogram study was determined using Kirby-Bauer disc diffusion method on Mueller Hinton Agar. Several methods previously reported in literature were used for the characterization of the virulence factors. RESULTS From the 30 blood samples collected, Pseudomonas spp. (19.7%), Escherichia coli (23%), Salmonella spp. (24.6%), and Staphylococcus aureus (32.8%) were isolated. Male to female ratio of study population was 1.5: 1. The isolates were 100 % resistant to ticarcillin, cephalothin, ceftazidime, and cefuroxime but appreciably susceptible to only levofloxacin (88.85%). They were moderately susceptible to ceftriaxone/sulbactam (39.05%) and azithromycin (26.46%). Common virulence factors identified among the isolates (up to 90 %) were hemolysin, biofilm formation, and acid resistance. Less common virulence factors were proteases (50 %), deoxyribonucleases (50 %), enterotoxins (63%), and lipopolysaccharide (70%). The virulence factors were found mostly among the S. aureus isolates. CONCLUSIONS Pseudomonas spp., Escherichia coli, Salmonella spp., and Staphylococcus aureus were implicated in neonatal infections in the center and most of them were resistant to conventional antibiotics. The organisms showed marked virulence and multidrug resistance properties. Levofloxacin, a fluoroquinolone, had superior activity on the isolates compared to other antibiotics used in the study.
Collapse
Affiliation(s)
- Obiora Shedrach Ejiofor
- Department of Pediatrics, Chukwuemeka Odumegwu Ojukwu University, Awka, Anambra State, Nigeria
| | - Onyinye Mercy Ajunwa
- Department of Pharmaceutical Microbiology and Biotechnology, Faculty of Pharmaceutical Sciences, Agulu, Nnamdi Azikiwe University, Anambra State, Nigeria
| | - Chijioke Elias Ezeudu
- Department of Pediatrics, College of Health Sciences, Faculty of Medicine, Nnamdi Azikiwe University, Nnewi Campus, Anambra State, Nigeria
| | - George Ogonna Emechebe
- Department of Pediatrics, Chukwuemeka Odumegwu Ojukwu University, Awka, Anambra State, Nigeria
| | - Kenneth Nchekwube Okeke
- Department of Pediatrics, Chukwuemeka Odumegwu Ojukwu University, Awka, Anambra State, Nigeria
| | | | - Ifeoma Mercy Ekejindu
- Department of Medical Laboratory Science, Faculty of Health Science and Technology, Nnamdi Azikiwe University, Nnewi Campus, Anambra State, Nigeria
| | - Jude Nnaemeka Okoyeh
- Department of Clinical Laboratory Science, School of Health Sciences, Winston-Salem State University, Winston-Salem, NC, USA
| | - Eunice Ogonna Osuala
- Department of Nursing Sciences, Faculty of Health Science and Technology, Nnamdi Azikiwe University, Nnewi Campus, Nigeria
| | - Angus Nnamdi Oli
- Department of Pharmaceutical Microbiology and Biotechnology, Faculty of Pharmaceutical Sciences, Agulu, Nnamdi Azikiwe University, Anambra State, Nigeria
| |
Collapse
|
31
|
Abstract
Thrombocytopenia is common in patients with invasive bacterial infections. Bacteria can activate platelets, but it is unclear if this affects platelet count. The aim of this study was to examine whether bacteraemia with Staphylococcus aureus, which readily activate human platelets, was more likely to be complicated by thrombocytopenia than bacteraemia with Escherichia coli or Streptococcus pneumoniae with different abilities to activate platelets.We compared information from 600 adult patients with community-acquired bacteraemia with S. aureus (n = 140), E. coli (n = 420) and S. pneumoniae (n = 40) in Southern Sweden, 2012, linking information on positive blood cultures from microbiological databases and medical charts. The proportion of patients with thrombocytopenia (platelet count <150 × 109/ml) was calculated. Logistic regression was used to estimate the odds ratios (OR) for thrombocytopenia according to bacterial species adjusted for confounders.The proportion of thrombocytopenia was 29% in S. aureus, 28% in E. coli and 20% in S. pneumonia bacteraemia (P = 0.50), corresponding to an OR of 1.2 (95% confidence interval 0.7-1.9) for thrombocytopenia for S. aureus as compared with E. coli or S. pneumoniae, adjusted for confounders.This study indicates that platelet activation by bacteria is not a major causative mechanism in sepsis-associated thrombocytopenia.
Collapse
|
32
|
Staphylococcus aureus clumping factor A is a force-sensitive molecular switch that activates bacterial adhesion. Proc Natl Acad Sci U S A 2018; 115:5564-5569. [PMID: 29735708 PMCID: PMC6003445 DOI: 10.1073/pnas.1718104115] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The Staphylococcus aureus surface protein clumping factor A (ClfA) binds to the blood plasma protein fibrinogen (Fg) via molecular interactions that are poorly understood. Here, we unravel the forces guiding the interaction between ClfA and immobilized Fg, showing that it is dramatically enhanced by tensile loading. Our findings favor a model whereby ClfA interacts with Fg via two distinct binding sites, the adhesive function of which is tightly regulated by mechanical force. Reminiscent of a catch bond mechanism, this force-enhanced adhesion explains the ability of ClfA to promote S. aureus colonization of host tissues and biomedical devices under physical stress. Clumping factor A (ClfA), a cell-wall–anchored protein from Staphylococcus aureus, is a virulence factor in various infections and facilitates the colonization of protein-coated biomaterials. ClfA promotes bacterial adhesion to the blood plasma protein fibrinogen (Fg) via molecular forces that have not been studied so far. A unique, yet poorly understood, feature of ClfA is its ability to favor adhesion to Fg at high shear stress. Unraveling the strength and dynamics of the ClfA–Fg interaction would help us better understand how S. aureus colonizes implanted devices and withstands physiological shear stress. By means of single-molecule experiments, we show that ClfA behaves as a force-sensitive molecular switch that potentiates staphylococcal adhesion under mechanical stress. The bond between ClfA and immobilized Fg is weak (∼0.1 nN) at low tensile force, but is dramatically enhanced (∼1.5 nN) by mechanical tension, as observed with catch bonds. Strong bonds, but not weak ones, are inhibited by a peptide mimicking the C-terminal segment of the Fg γ-chain. These results point to a model whereby ClfA interacts with Fg via two distinct binding sites, the adhesive function of which is regulated by mechanical tension. This force-activated mechanism is of biological significance because it explains at the molecular level the ability of ClfA to promote bacterial attachment under high physiological shear stress.
Collapse
|
33
|
Liesenborghs L, Verhamme P, Vanassche T. Staphylococcus aureus, master manipulator of the human hemostatic system. J Thromb Haemost 2018; 16:441-454. [PMID: 29251820 DOI: 10.1111/jth.13928] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Indexed: 12/15/2022]
Abstract
The coagulation system does not only offer protection against bleeding, but also aids in our defense against invading microorganisms. The hemostatic system and innate immunity are strongly entangled, which explains why so many infections are complicated by either bleeding or thrombosis. Staphylococcus aureus (S. aureus), currently the most deadly infectious agent in the developed world, causes devastating intravascular infections such as sepsis and infective endocarditis. During these infections S. aureus comes in close contact with the host hemostatic system and proves to be a master in manipulating coagulation. The coagulases of S. aureus directly induce coagulation by activating prothrombin. S. aureus also manipulates fibrinolysis by triggering plasminogen activation via staphylokinase. Furthermore, S. aureus binds and activates platelets and interacts with key coagulation proteins such as fibrin(ogen), fibronectin and von Willebrand factor. By manipulating the coagulation system S. aureus gains a significant advantage over the host defense mechanisms. Studying the interplay between S. aureus and the hemostatic system can therefore lead to new innovative therapies for battling S. aureus infections.
Collapse
Affiliation(s)
- L Liesenborghs
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KULeuven - University Hospitals Leuven, Leuven, Belgium
| | - P Verhamme
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KULeuven - University Hospitals Leuven, Leuven, Belgium
| | - T Vanassche
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KULeuven - University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
34
|
Lattwein KR, Shekhar H, van Wamel WJB, Gonzalez T, Herr AB, Holland CK, Kooiman K. An in vitro proof-of-principle study of sonobactericide. Sci Rep 2018; 8:3411. [PMID: 29467474 PMCID: PMC5821825 DOI: 10.1038/s41598-018-21648-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 02/06/2018] [Indexed: 12/24/2022] Open
Abstract
Infective endocarditis (IE) is associated with high morbidity and mortality rates. The predominant bacteria causing IE is Staphylococcus aureus (S. aureus), which can bind to existing thrombi on heart valves and generate vegetations (biofilms). In this in vitro flow study, we evaluated sonobactericide as a novel strategy to treat IE, using ultrasound and an ultrasound contrast agent with or without other therapeutics. We developed a model of IE biofilm using human whole-blood clots infected with patient-derived S. aureus (infected clots). Histology and live-cell imaging revealed a biofilm layer of fibrin-embedded living Staphylococci around a dense erythrocyte core. Infected clots were treated under flow for 30 minutes and degradation was assessed by time-lapse microscopy imaging. Treatments consisted of either continuous plasma flow alone or with different combinations of therapeutics: oxacillin (antibiotic), recombinant tissue plasminogen activator (rt-PA; thrombolytic), intermittent continuous-wave low-frequency ultrasound (120-kHz, 0.44 MPa peak-to-peak pressure), and an ultrasound contrast agent (Definity). Infected clots exposed to the combination of oxacillin, rt-PA, ultrasound, and Definity achieved 99.3 ± 1.7% loss, which was greater than the other treatment arms. Effluent size measurements suggested low likelihood of emboli formation. These results support the continued investigation of sonobactericide as a therapeutic strategy for IE.
Collapse
Affiliation(s)
- Kirby R Lattwein
- Department of Biomedical Engineering, Thoraxcenter, Erasmus MC, Room Ee2302, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands. .,Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, Cincinnati, Ohio, USA. .,Department of Medical Microbiology and Infectious Diseases, Erasmus MC, Rotterdam, The Netherlands.
| | - Himanshu Shekhar
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, Cincinnati, Ohio, USA
| | - Willem J B van Wamel
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, Rotterdam, The Netherlands
| | - Tammy Gonzalez
- Cincinnati Children's Hospital Medical Center, Division of Immunobiology, Center for Systems Immunology, and Division of Infectious Diseases, Cincinnati, Ohio, USA
| | - Andrew B Herr
- Cincinnati Children's Hospital Medical Center, Division of Immunobiology, Center for Systems Immunology, and Division of Infectious Diseases, Cincinnati, Ohio, USA
| | - Christy K Holland
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, Cincinnati, Ohio, USA
| | - Klazina Kooiman
- Department of Biomedical Engineering, Thoraxcenter, Erasmus MC, Room Ee2302, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| |
Collapse
|
35
|
Abstract
Nosocomial or hospital-acquired infections are associated with prolonged hospitalizations and increased healthcare costs. Infections associated with surgical implants are becoming more difficult and more costly to manage, as they require repeated surgical procedures and a longer period of time to treat patients. Continued advances in the use of medical devices, an increase in the number of immunocompromised patients, and a steady rise in the prevalence of antibiotic-resistant organisms has renewed interest in the development of novel therapies that can be used to prevent and treat nosocomial infections. This review provides an overview of bacterial adhesins and focuses on novel immunological therapies developed to treat staphylococcal infections.
Collapse
Affiliation(s)
- J M Patti
- Inhibitex, Inc., Alpharetta, Georgia, USA.
| |
Collapse
|
36
|
The Staphylococcus aureus Cell Wall-Anchored Protein Clumping Factor A Is an Important T Cell Antigen. Infect Immun 2017; 85:IAI.00549-17. [PMID: 28947645 PMCID: PMC5695125 DOI: 10.1128/iai.00549-17] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 09/18/2017] [Indexed: 02/06/2023] Open
Abstract
Staphylococcus aureus has become increasingly resistant to antibiotics, and vaccines offer a potential solution to this epidemic of antimicrobial resistance. Targeting of specific T cell subsets is now considered crucial for next-generation anti-S. aureus vaccines; however, there is a paucity of information regarding T cell antigens of S. aureus. This study highlights the importance of cell wall-anchored proteins as human CD4+ T cell activators capable of driving antigen-specific Th1 and Th17 cell activation. Clumping factor A (ClfA), which contains N1, N2, and N3 binding domains, was found to be a potent human T cell activator. We further investigated which subdomains of ClfA were involved in T cell activation and found that the full-length ClfA N123 and N23 were potent Th1 and Th17 activators. Interestingly, the N1 subdomain was capable of exclusively activating Th1 cells. Furthermore, when these subdomains were used in a model vaccine, N23 and N1 offered Th1- and Th17-mediated systemic protection in mice upon intraperitoneal challenge. Overall, however, full-length ClfA N123 is required for maximal protection both locally and systemically.
Collapse
|
37
|
Saraiva MM, De Leon CM, Santos SC, Stipp DT, Souza MM, Santos Filho L, Gebreyes WA, Oliveira CJ. Accuracy of PCR targeting different markers for Staphylococcus aureus identification: a comparative study using matrix-assisted laser desorption/ionization time-of-flight mass spectrometry as the gold standard. J Vet Diagn Invest 2017; 30:252-255. [PMID: 29105571 DOI: 10.1177/1040638717732370] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Staphylococcus aureus is considered a major pathogen in veterinary and human medicine, and the emergence of multidrug-resistant strains, such as livestock-associated methicillin-resistant S. aureus, means that reliable, inexpensive, and fast methods are required to identify S. aureus obtained from animal sources. We tested the accuracy of a PCR targeting the genes femA, nuc, and coa in identifying S. aureus from animals. A total of 157 Staphylococcus spp. isolates were examined by matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometry; 18 different Staphylococcus species were identified. Of 68 S. aureus isolates, the genes femA, nuc, and coa were found in 61, 53, and 32 isolates, respectively. Considering MALDI-TOF as the gold standard, the PCR assays targeting all 3 genes showed 100% specificity; the sensitivity values were 89.7, 77.9, and 47.0% for femA, nuc, and coa, respectively. Sensitivity was 100% when femA and nuc markers were targeted simultaneously. These results confirm PCR as an accurate method to identify S. aureus species from animal sources and strongly suggest the simultaneous use of primers targeting femA and nuc genes.
Collapse
Affiliation(s)
- Mauro M Saraiva
- Departments of Animal Sciences (Saraiva, De Leon, Santos, Oliveira), College of Agricultural Sciences, Federal University of Paraiba, Areia, Paraíba, Brazil.,Veterinary Sciences (Stipp), College of Agricultural Sciences, Federal University of Paraiba, Areia, Paraíba, Brazil.,Institute of Veterinary Medicine, Federal Rural University of Rio de Janeiro, Seropédica, Rio de Janeiro, Brazil (Souza).,Department of Pharmaceutical Sciences, College of Health Sciences, Federal University of Paraiba, João Pessoa, Paraíba, Brazil (Santos Filho).,Veterinary Public Health and Biotechnology Global Consortium, The Ohio State University, Columbus, OH (Gebreyes, Oliveira)
| | - Candice M De Leon
- Departments of Animal Sciences (Saraiva, De Leon, Santos, Oliveira), College of Agricultural Sciences, Federal University of Paraiba, Areia, Paraíba, Brazil.,Veterinary Sciences (Stipp), College of Agricultural Sciences, Federal University of Paraiba, Areia, Paraíba, Brazil.,Institute of Veterinary Medicine, Federal Rural University of Rio de Janeiro, Seropédica, Rio de Janeiro, Brazil (Souza).,Department of Pharmaceutical Sciences, College of Health Sciences, Federal University of Paraiba, João Pessoa, Paraíba, Brazil (Santos Filho).,Veterinary Public Health and Biotechnology Global Consortium, The Ohio State University, Columbus, OH (Gebreyes, Oliveira)
| | - Silvana C Santos
- Departments of Animal Sciences (Saraiva, De Leon, Santos, Oliveira), College of Agricultural Sciences, Federal University of Paraiba, Areia, Paraíba, Brazil.,Veterinary Sciences (Stipp), College of Agricultural Sciences, Federal University of Paraiba, Areia, Paraíba, Brazil.,Institute of Veterinary Medicine, Federal Rural University of Rio de Janeiro, Seropédica, Rio de Janeiro, Brazil (Souza).,Department of Pharmaceutical Sciences, College of Health Sciences, Federal University of Paraiba, João Pessoa, Paraíba, Brazil (Santos Filho).,Veterinary Public Health and Biotechnology Global Consortium, The Ohio State University, Columbus, OH (Gebreyes, Oliveira)
| | - Danilo T Stipp
- Departments of Animal Sciences (Saraiva, De Leon, Santos, Oliveira), College of Agricultural Sciences, Federal University of Paraiba, Areia, Paraíba, Brazil.,Veterinary Sciences (Stipp), College of Agricultural Sciences, Federal University of Paraiba, Areia, Paraíba, Brazil.,Institute of Veterinary Medicine, Federal Rural University of Rio de Janeiro, Seropédica, Rio de Janeiro, Brazil (Souza).,Department of Pharmaceutical Sciences, College of Health Sciences, Federal University of Paraiba, João Pessoa, Paraíba, Brazil (Santos Filho).,Veterinary Public Health and Biotechnology Global Consortium, The Ohio State University, Columbus, OH (Gebreyes, Oliveira)
| | - Miliane M Souza
- Departments of Animal Sciences (Saraiva, De Leon, Santos, Oliveira), College of Agricultural Sciences, Federal University of Paraiba, Areia, Paraíba, Brazil.,Veterinary Sciences (Stipp), College of Agricultural Sciences, Federal University of Paraiba, Areia, Paraíba, Brazil.,Institute of Veterinary Medicine, Federal Rural University of Rio de Janeiro, Seropédica, Rio de Janeiro, Brazil (Souza).,Department of Pharmaceutical Sciences, College of Health Sciences, Federal University of Paraiba, João Pessoa, Paraíba, Brazil (Santos Filho).,Veterinary Public Health and Biotechnology Global Consortium, The Ohio State University, Columbus, OH (Gebreyes, Oliveira)
| | - Lauro Santos Filho
- Departments of Animal Sciences (Saraiva, De Leon, Santos, Oliveira), College of Agricultural Sciences, Federal University of Paraiba, Areia, Paraíba, Brazil.,Veterinary Sciences (Stipp), College of Agricultural Sciences, Federal University of Paraiba, Areia, Paraíba, Brazil.,Institute of Veterinary Medicine, Federal Rural University of Rio de Janeiro, Seropédica, Rio de Janeiro, Brazil (Souza).,Department of Pharmaceutical Sciences, College of Health Sciences, Federal University of Paraiba, João Pessoa, Paraíba, Brazil (Santos Filho).,Veterinary Public Health and Biotechnology Global Consortium, The Ohio State University, Columbus, OH (Gebreyes, Oliveira)
| | - Wondwossen A Gebreyes
- Departments of Animal Sciences (Saraiva, De Leon, Santos, Oliveira), College of Agricultural Sciences, Federal University of Paraiba, Areia, Paraíba, Brazil.,Veterinary Sciences (Stipp), College of Agricultural Sciences, Federal University of Paraiba, Areia, Paraíba, Brazil.,Institute of Veterinary Medicine, Federal Rural University of Rio de Janeiro, Seropédica, Rio de Janeiro, Brazil (Souza).,Department of Pharmaceutical Sciences, College of Health Sciences, Federal University of Paraiba, João Pessoa, Paraíba, Brazil (Santos Filho).,Veterinary Public Health and Biotechnology Global Consortium, The Ohio State University, Columbus, OH (Gebreyes, Oliveira)
| | - Celso J Oliveira
- Departments of Animal Sciences (Saraiva, De Leon, Santos, Oliveira), College of Agricultural Sciences, Federal University of Paraiba, Areia, Paraíba, Brazil.,Veterinary Sciences (Stipp), College of Agricultural Sciences, Federal University of Paraiba, Areia, Paraíba, Brazil.,Institute of Veterinary Medicine, Federal Rural University of Rio de Janeiro, Seropédica, Rio de Janeiro, Brazil (Souza).,Department of Pharmaceutical Sciences, College of Health Sciences, Federal University of Paraiba, João Pessoa, Paraíba, Brazil (Santos Filho).,Veterinary Public Health and Biotechnology Global Consortium, The Ohio State University, Columbus, OH (Gebreyes, Oliveira)
| |
Collapse
|
38
|
Abstract
Staphylococcus aureus is often involved in severe infections, in which the effects of bacterial virulence factors have great importance. Antistaphylococcal regimens should take into account the different effects of antibacterial agents on the expression of virulence factors and on the host's immune response. A PubMed literature search was performed to select relevant articles on the effects of antibiotics on staphylococcal toxin production and on the host immune response. Information was sorted according to the methods used for data acquisition (bacterial strains, growth models, and antibiotic concentrations) and the assays used for readout generation. The reported mechanisms underlying S. aureus virulence modulation by antibiotics were reviewed. The relevance of in vitro observations is discussed in relation to animal model data and to clinical evidence extracted from case reports and recommendations on the management of toxin-related staphylococcal diseases. Most in vitro data point to a decreased level of virulence expression upon treatment with ribosomally active antibiotics (linezolid and clindamycin), while cell wall-active antibiotics (beta-lactams) mainly increase exotoxin production. In vivo studies confirmed the suppressive effect of clindamycin and linezolid on virulence expression, supporting their utilization as a valuable management strategy to improve patient outcomes in cases of toxin-associated staphylococcal disease.
Collapse
|
39
|
Tan Y, Leonhard M, Schneider-Stickler B. Evaluation of culture conditions for mixed biofilm formation with clinically isolated non- albicans Candida species and Staphylococcus epidermidis on silicone. Microb Pathog 2017; 112:215-220. [DOI: 10.1016/j.micpath.2017.10.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 10/03/2017] [Accepted: 10/04/2017] [Indexed: 12/24/2022]
|
40
|
Hyperbaric oxygen therapy augments tobramycin efficacy in experimental Staphylococcus aureus endocarditis. Int J Antimicrob Agents 2017; 50:406-412. [DOI: 10.1016/j.ijantimicag.2017.04.025] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 03/24/2017] [Accepted: 04/05/2017] [Indexed: 12/14/2022]
|
41
|
Abstract
The staphylococci comprise a diverse genus of Gram-positive, nonmotile commensal organisms that inhabit the skin and mucous membranes of humans and other mammals. In general, staphylococci are benign members of the natural flora, but many species have the capacity to be opportunistic pathogens, mainly infecting individuals who have medical device implants or are otherwise immunocompromised. Staphylococcus aureus and Staphylococcus epidermidis are major sources of hospital-acquired infections and are the most common causes of surgical site infections and medical device-associated bloodstream infections. The ability of staphylococci to form biofilms in vivo makes them highly resistant to chemotherapeutics and leads to chronic diseases. These biofilm infections include osteomyelitis, endocarditis, medical device infections, and persistence in the cystic fibrosis lung. Here, we provide a comprehensive analysis of our current understanding of staphylococcal biofilm formation, with an emphasis on adhesins and regulation, while also addressing how staphylococcal biofilms interact with the immune system. On the whole, this review will provide a thorough picture of biofilm formation of the staphylococcus genus and how this mode of growth impacts the host.
Collapse
|
42
|
Pumipuntu N, Kulpeanprasit S, Santajit S, Tunyong W, Kong-Ngoen T, Hinthong W, Indrawattana N. Screening method for Staphylococcus aureus identification in subclinical bovine mastitis from dairy farms. Vet World 2017; 10:721-726. [PMID: 28831211 PMCID: PMC5553136 DOI: 10.14202/vetworld.2017.721-726] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 05/12/2017] [Indexed: 11/16/2022] Open
Abstract
Background: Staphylococcus aureus is one of the most important contagious bacteria causing subclinical bovine mastitis. This bacterial infection is commonly identified by determine the pathogen in bovine milk samples through conventional technique including coagulase test. However, this test has several disadvantages as low sensitivity, risk of biohazard, cost expensive, and limited preparation especially in local area. Aim: Aim of this study was to compare and assess the screening method, Mannitol fermentation test (Mannitol salt agar [MSA]), and deoxyribonuclease (DNase) test, for S. aureus identification in milk samples. Materials and Methods: A total of 224 subclinical bovine mastitis milk samples were collected from four provinces of Thailand and determined S. aureus using conventional method and also subjected to the screening test, MSA and DNase test. The sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV) among both tests were analyzed and compared to the tube coagulase test (TCT), as reference method. Immunological test by latex agglutination and molecular assay by determined spa gene were also used to identify and differentiate S. aureus. Results: A total of 130 staphylococci were isolated by selective media, Gram-stain, and catalase test. The number of S. aureus which identified using TCT, MSA and DNase test were 32, 102, and 74 isolates, respectively. All TCT results were correlated to results of latex agglutination and spa gene which were 32 S. aureus. MSA showed 100% sensitivity, 28.57% specificity, 31.37% PPV, and 100% NPV, whereas DNase showed 53.13% sensitivity, 41.84% specificity, 22.97% PPV, and 73.21% NPV. DNase test showed higher specificity value than MSA but the test presented 26.79% false negative results whereas no false-negative result from MSA when comparing to TCT. Conclusion: MSA had a tendency to be a good preference for screening S. aureus because of its high sensitivity and NPV. The result from this study will improve a choice to use a screening test to diagnose S. aureus of veterinary field for prompt disease controlling and effective treatment.
Collapse
Affiliation(s)
- Natapol Pumipuntu
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Suphang Kulpeanprasit
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Sirijan Santajit
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Witawat Tunyong
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Thida Kong-Ngoen
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Woranich Hinthong
- Faculty of Medicine and Allied Health, HRH Princess Chulabhorn College of Medical Science Bangkok, Thailand
| | - Nitaya Indrawattana
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| |
Collapse
|
43
|
Guo H, Hall JW, Yang J, Ji Y. The SaeRS Two-Component System Controls Survival of Staphylococcus aureus in Human Blood through Regulation of Coagulase. Front Cell Infect Microbiol 2017; 7:204. [PMID: 28611950 PMCID: PMC5447086 DOI: 10.3389/fcimb.2017.00204] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Accepted: 05/08/2017] [Indexed: 12/03/2022] Open
Abstract
The SaeRS two-component system plays important roles in regulation of key virulence factors and pathogenicity. In this study, however, we found that the deletion mutation of saeRS enhanced bacterial survival in human blood, whereas complementation of the mutant with SaeRS returned survival to wild-type levels. Moreover, these phenomena were observed in different MRSA genetic background isolates, including HA-MRSA WCUH29, CA-MRSA 923, and MW2. To elucidate which gene(s) regulated by SaeRS contribute to the effect, we conducted a series of complementation studies with selected known SaeRS target genes in trans. We found coagulase complementation abolished the enhanced survival of the SaeRS mutant in human blood. The coa and saeRS deletion mutants exhibited a similar survival phenotype in blood. Intriguingly, heterologous expression of coagulase decreased survival of S. epidermidis in human blood. Further, the addition of recombinant coagulase to blood significantly decreased the survival of S. aureus. Further, analysis revealed staphylococcal resistance to killing by hydrogen peroxide was partially dependent on the presence or absence of coagulase. Furthermore, complementation with coagulase, but not SaeRS, returned saeRS/coa double mutant survival in blood to wild-type levels. These data indicate SaeRS modulates bacterial survival in blood in coagulase-dependent manner. Our results provide new insights into the role of staphylococcal SaeRS and coagulase on bacterial survival in human blood.
Collapse
Affiliation(s)
- Haiyong Guo
- Department of Biological Science, School of Life Science, Jilin Normal UniversitySiping, China.,Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of MinnesotaSt Paul, MN, United States
| | - Jeffrey W Hall
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of MinnesotaSt Paul, MN, United States
| | - Junshu Yang
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of MinnesotaSt Paul, MN, United States
| | - Yinduo Ji
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of MinnesotaSt Paul, MN, United States
| |
Collapse
|
44
|
Mohamed N, Wang MY, Le Huec JC, Liljenqvist U, Scully IL, Baber J, Begier E, Jansen KU, Gurtman A, Anderson AS. Vaccine development to prevent Staphylococcus aureus surgical-site infections. Br J Surg 2017; 104:e41-e54. [PMID: 28121039 DOI: 10.1002/bjs.10454] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 10/27/2016] [Accepted: 11/06/2016] [Indexed: 12/12/2022]
Abstract
BACKGROUND Staphylococcus aureus surgical-site infections (SSIs) are a major cause of poor health outcomes, including mortality, across surgical specialties. Despite current advances as a result of preventive interventions, the disease burden of S. aureus SSI remains high, and increasing antibiotic resistance continues to be a concern. Prophylactic S. aureus vaccines may represent an opportunity to prevent SSI. METHODS A review of SSI pathophysiology was undertaken in the context of evaluating new approaches to developing a prophylactic vaccine to prevent S. aureus SSI. RESULTS A prophylactic vaccine ideally would provide protective immunity at the time of the surgical incision to prevent initiation and progression of infection. Although the pathogenicity of S. aureus is attributed to many virulence factors, previous attempts to develop S. aureus vaccines targeted only a single virulence mechanism. The field has now moved towards multiple-antigen vaccine strategies, and promising results have been observed in early-phase clinical studies that supported the recent initiation of an efficacy trial to prevent SSI. CONCLUSION There is an unmet medical need for novel S. aureus SSI prevention measures. Advances in understanding of S. aureus SSI pathophysiology could lead to the development of effective and safe prophylactic multiple-antigen vaccines to prevent S. aureus SSI.
Collapse
Affiliation(s)
- N Mohamed
- Pfizer Vaccine Research and Development, Pearl River, New York, USA
| | - M Y Wang
- Departments of Neurological Surgery and Rehabilitation Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - J-C Le Huec
- Spine Unit 2, Surgical Research Laboratory, Bordeaux University Hospital, Bordeaux, France
| | - U Liljenqvist
- Department of Spine Surgery, St Franziskus Hospital Muenster, Münster, Germany
| | - I L Scully
- Pfizer Vaccine Research and Development, Pearl River, New York, USA
| | - J Baber
- Pfizer Vaccine Clinical Research and Development, Sydney, New South Wales, Australia
| | - E Begier
- Pfizer Vaccine Clinical Research and Development, Pearl River, New York, USA
| | - K U Jansen
- Pfizer Vaccine Research and Development, Pearl River, New York, USA
| | - A Gurtman
- Pfizer Vaccine Clinical Research and Development, Pearl River, New York, USA
| | - A S Anderson
- Pfizer Vaccine Research and Development, Pearl River, New York, USA
| |
Collapse
|
45
|
Lin J, Peng Y, Ou Q, Lin D, Li Y, Ye X, Zhou J, Yao Z. A molecular epidemiological study of methicillin-resistantStaphylococcienvironmental contamination in railway stations and coach stations in Guangzhou of China. Lett Appl Microbiol 2017; 64:131-137. [DOI: 10.1111/lam.12700] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 11/11/2016] [Accepted: 12/01/2016] [Indexed: 11/29/2022]
Affiliation(s)
- J.L. Lin
- Department of Epidemiology and Health Statistics; School of Public Health; Guangdong Pharmaceutical University; Guangzhou China
| | - Y. Peng
- Centre for Chronic Disease; University of Queensland; Brisbane QLD Australia
| | - Q.T. Ou
- Department of Epidemiology and Health Statistics; School of Public Health; Guangdong Pharmaceutical University; Guangzhou China
| | - D.X. Lin
- Department of Epidemiology and Health Statistics; School of Public Health; Guangdong Pharmaceutical University; Guangzhou China
| | - Y. Li
- Division of Environmental Health; Public Health Laboratory Center; Guangdong Pharmaceutical University; Guangzhou China
| | - X.H. Ye
- Department of Epidemiology and Health Statistics; School of Public Health; Guangdong Pharmaceutical University; Guangzhou China
| | - J.L. Zhou
- Department of Epidemiology and Health Statistics; School of Public Health; Guangdong Pharmaceutical University; Guangzhou China
| | - Z.J. Yao
- Department of Epidemiology and Health Statistics; School of Public Health; Guangdong Pharmaceutical University; Guangzhou China
| |
Collapse
|
46
|
Wang X, Li X, Liu W, Huang W, Fu Q, Li M. Molecular Characteristic and Virulence Gene Profiles of Community-Associated Methicillin-Resistant Staphylococcus aureus Isolates from Pediatric Patients in Shanghai, China. Front Microbiol 2016; 7:1818. [PMID: 27895635 PMCID: PMC5108810 DOI: 10.3389/fmicb.2016.01818] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 10/28/2016] [Indexed: 11/13/2022] Open
Abstract
Staphylococcus aureus is a globally important human pathogen, especially among children and immunocompromised patients. The emergence and spread of community-associated methicillin-resistant S. aureus (CA-MRSA) has become a serious public health problem worldwide. The aim of this study was to investigate the prevalence, molecular characteristics and virulence profiles of CA-MRSA infections from pediatric patients in a university hospital in Shanghai, China. A total of 80 CA-MRSA isolates were collected from July 2012 to December 2013 in Shanghai Children's Medical Center and analyzed by multilocus sequence typing, staphylococcus chromosomal cassette mec (SCCmec) typing, and spa typing. The detection of Panton-Valentine Leukocidin (pvl), superantigenic and exfoliative toxins, and adhesin genes was also performed. Overall, 16 distinct sequence types (STs) were identified among the 80 isolates. Among them, ST59 was found to be the most prevalent, followed by ST398 (11.3%, 9/80) and ST88 (8.8%, 7/80). SCCmec types IV and V were observed, at 60 and 40%, respectively. Thirty spa types were identified, spa t437 (23.8%) was the most predominant type. All 80 isolates exhibited carriage of at least four virulence genes. Thirty-four (42.5%, 34/80) isolates harbored ≥10 tested virulence genes. Adhesion genes were present in most of the MRSA isolates, including the following: icaA (100%), clfA (100%), sdrC (95%), and sdrE (63.8%). The prevalence of pvl gene was 20%, and multidrug resistance was observed in 36% of all strains. In addition, ST59-MRSA-IV with t437 accounted for 21.3% of occurrences, making it the most prevalent clone. Isolates that were carriers of toxin genes, and hla (100%) and hlg (87.5%) were the most frequent. In conclusion, simultaneous carriage of multiple virulence genes and genetically considerable diversity were very common among CA-MRSA from pediatric patients in Shanghai. ST59-MRSA-IV with t437 was still the most predominant type. The combination of virulence gene profiles and antibiotic resistance may help ST59 to be successfully spread among children.
Collapse
Affiliation(s)
- Xing Wang
- Department of Laboratory Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine Shanghai, China
| | - Xia Li
- Department of Medical Microbiology and Immunology, University of California, Davis Davis, CA, USA
| | - Wei Liu
- Department of Laboratory Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine Shanghai, China
| | - Weichun Huang
- Department of Laboratory Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine Shanghai, China
| | - Qihua Fu
- Department of Laboratory Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine Shanghai, China
| | - Min Li
- Department of Laboratory Medicine, Renji Hospital, Shanghai Jiaotong University School of Medicine Shanghai, China
| |
Collapse
|
47
|
Abstract
Infective endocarditis (IE) is a rare, life-threatening disease that has long-lasting effects even among patients who survive and are cured. IE disproportionately affects those with underlying structural heart disease and is increasingly associated with health care contact, particularly in patients who have intravascular prosthetic material. In the setting of bacteraemia with a pathogenic organism, an infected vegetation may form as the end result of complex interactions between invading microorganisms and the host immune system. Once established, IE can involve almost any organ system in the body. The diagnosis of IE may be difficult to establish and a strategy that combines clinical, microbiological and echocardiography results has been codified in the modified Duke criteria. In cases of blood culture-negative IE, the diagnosis may be especially challenging, and novel microbiological and imaging techniques have been developed to establish its presence. Once diagnosed, IE is best managed by a multidisciplinary team with expertise in infectious diseases, cardiology and cardiac surgery. Antibiotic prophylaxis for the prevention of IE remains controversial. Efforts to develop a vaccine that targets common bacterial causes of IE are ongoing, but have not yet yielded a commercially available product.
Collapse
Affiliation(s)
- Thomas L Holland
- Department of Medicine, Division of Infectious Diseases, Duke University Medical Center, Room 185 Hanes Building, 315 Trent Drive, Durham, North Carolina 27710, USA
- Duke Clinical Research Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Larry M Baddour
- Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Arnold S Bayer
- Department of Medicine, David Geffen School of Medicine at UCLA, Torrance, California, USA
| | - Bruno Hoen
- Department of Infectious Diseases, University Hospital of Pointe-Pitre, Pointe-Pitre, France
| | - Jose M Miro
- Infectious Diseases Service, Hospital Clinic - IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Vance G Fowler
- Department of Medicine, Division of Infectious Diseases, Duke University Medical Center, Room 185 Hanes Building, 315 Trent Drive, Durham, North Carolina 27710, USA
- Duke Clinical Research Institute, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
48
|
Crosby HA, Kwiecinski J, Horswill AR. Staphylococcus aureus Aggregation and Coagulation Mechanisms, and Their Function in Host-Pathogen Interactions. ADVANCES IN APPLIED MICROBIOLOGY 2016; 96:1-41. [PMID: 27565579 DOI: 10.1016/bs.aambs.2016.07.018] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The human commensal bacterium Staphylococcus aureus can cause a wide range of infections ranging from skin and soft tissue infections to invasive diseases like septicemia, endocarditis, and pneumonia. Muticellular organization almost certainly contributes to S. aureus pathogenesis mechanisms. While there has been considerable focus on biofilm formation and its role in colonizing prosthetic joints and indwelling devices, less attention has been paid to nonsurface-attached group behavior like aggregation and clumping. S. aureus is unique in its ability to coagulate blood, and it also produces multiple fibrinogen-binding proteins that facilitate clumping. Formation of clumps, which are large, tightly packed groups of cells held together by fibrin(ogen), has been demonstrated to be important for S. aureus virulence and immune evasion. Clumps of cells are able to avoid detection by the host's immune system due to a fibrin(ogen) coat that acts as a shield, and the size of the clumps facilitates evasion of phagocytosis. In addition, clumping could be an important early step in establishing infections that involve tight clusters of cells embedded in host matrix proteins, such as soft tissue abscesses and endocarditis. In this review, we discuss clumping mechanisms and regulation, as well as what is known about how clumping contributes to immune evasion.
Collapse
Affiliation(s)
- H A Crosby
- University of Iowa, Iowa City, IA, United States
| | - J Kwiecinski
- University of Iowa, Iowa City, IA, United States
| | - A R Horswill
- University of Iowa, Iowa City, IA, United States
| |
Collapse
|
49
|
Silva LN, Zimmer KR, Macedo AJ, Trentin DS. Plant Natural Products Targeting Bacterial Virulence Factors. Chem Rev 2016; 116:9162-236. [PMID: 27437994 DOI: 10.1021/acs.chemrev.6b00184] [Citation(s) in RCA: 263] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Decreased antimicrobial efficiency has become a global public health issue. The paucity of new antibacterial drugs is evident, and the arsenal against infectious diseases needs to be improved urgently. The selection of plants as a source of prototype compounds is appropriate, since plant species naturally produce a wide range of secondary metabolites that act as a chemical line of defense against microorganisms in the environment. Although traditional approaches to combat microbial infections remain effective, targeting microbial virulence rather than survival seems to be an exciting strategy, since the modulation of virulence factors might lead to a milder evolutionary pressure for the development of resistance. Additionally, anti-infective chemotherapies may be successfully achieved by combining antivirulence and conventional antimicrobials, extending the lifespan of these drugs. This review presents an updated discussion of natural compounds isolated from plants with chemically characterized structures and activity against the major bacterial virulence factors: quorum sensing, bacterial biofilms, bacterial motility, bacterial toxins, bacterial pigments, bacterial enzymes, and bacterial surfactants. Moreover, a critical analysis of the most promising virulence factors is presented, highlighting their potential as targets to attenuate bacterial virulence. The ongoing progress in the field of antivirulence therapy may therefore help to translate this promising concept into real intervention strategies in clinical areas.
Collapse
Affiliation(s)
- Laura Nunes Silva
- Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul , Porto Alegre, Rio Grande do Sul 90610-000, Brazil.,Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul , Porto Alegre, Rio Grande do Sul 91501-970, Brazil
| | - Karine Rigon Zimmer
- Departamento de Ciências Básicas da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre , Porto Alegre, Rio Grande do Sul 90050-170, Brazil
| | - Alexandre José Macedo
- Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul , Porto Alegre, Rio Grande do Sul 90610-000, Brazil.,Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul , Porto Alegre, Rio Grande do Sul 91501-970, Brazil.,Instituto Nacional do Semiárido , Campina Grande, Paraı́ba 58429-970, Brazil
| | - Danielle Silva Trentin
- Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul , Porto Alegre, Rio Grande do Sul 90610-000, Brazil.,Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul , Porto Alegre, Rio Grande do Sul 91501-970, Brazil
| |
Collapse
|
50
|
Ko YP, Flick MJ. Fibrinogen Is at the Interface of Host Defense and Pathogen Virulence in Staphylococcus aureus Infection. Semin Thromb Hemost 2016; 42:408-21. [PMID: 27056151 PMCID: PMC5514417 DOI: 10.1055/s-0036-1579635] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Fibrinogen not only plays a pivotal role in hemostasis but also serves key roles in antimicrobial host defense. As a rapidly assembled provisional matrix protein, fibrin(ogen) can function as an early line of host protection by limiting bacterial growth, suppressing dissemination of microbes to distant sites, and mediating host bacterial killing. Fibrinogen-mediated host antimicrobial activity occurs predominantly through two general mechanisms, namely, fibrin matrices functioning as a protective barrier and fibrin(ogen) directly or indirectly driving host protective immune function. The potential of fibrin to limit bacterial infection and disease has been countered by numerous bacterial species evolving and maintaining virulence factors that engage hemostatic system components within vertebrate hosts. Bacterial factors have been isolated that simply bind fibrinogen or fibrin, promote fibrin polymer formation, or promote fibrin dissolution. Staphylococcus aureus is an opportunistic gram-positive bacterium, the causative agent of a wide range of human infectious diseases, and a prime example of a pathogen exquisitely sensitive to host fibrinogen. Indeed, current data suggest fibrinogen serves as a context-dependent determinant of host defense or pathogen virulence in Staphylococcus infection whose ultimate contribution is dictated by the expression of S. aureus virulence factors, the path of infection, and the tissue microenvironment.
Collapse
Affiliation(s)
- Ya-Ping Ko
- Center for Infectious and Inflammatory Diseases, Institute for Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas
| | - Matthew J. Flick
- Division of Experimental Hematology and Cancer Biology, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|