101
|
Xue T, Chen X, Shang F. Short communication: Effects of lactose and milk on the expression of biofilm-associated genes in Staphylococcus aureus strains isolated from a dairy cow with mastitis. J Dairy Sci 2014; 97:6129-34. [DOI: 10.3168/jds.2014-8344] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 07/09/2014] [Indexed: 11/19/2022]
|
102
|
Wu Y, Liu J, Jiang J, Hu J, Xu T, Wang J, Qu D. Role of the two-component regulatory system arlRS in ica operon and aap positive but non-biofilm-forming Staphylococcus epidermidis isolates from hospitalized patients. Microb Pathog 2014; 76:89-98. [PMID: 25263000 DOI: 10.1016/j.micpath.2014.09.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 09/21/2014] [Accepted: 09/24/2014] [Indexed: 10/24/2022]
Abstract
The ica operon and aap gene are important factors for Staphylococcus epidermidis biofilm formation. However, we found 15 out of 101 S. epidermidis strains isolated from patients had both the ica operon and the aap gene in the genome but could not form biofilms (ica(+)aap(+)/BF(-) isolates). Compared with standard strain RP62A, the 15 ica(+)aap(+)/BF(-) isolates had similar growth curves and initial attachment abilities, but had much lower apparent transcription levels of the icaA gene and significantly less production of polysaccharide intercellular adhesion (PIA). Furthermore, the expression of accumulation-associated protein in ica(+)aap(+)/BF(-) isolates was much weaker than in RP62A. The mRNA levels of icaADBC transcription-related regulatory genes, including icaR, sarA, rsbU, srrA, arlRS and luxS, were measured in the 15 ica(+)aap(+)/BF(-) clinical isolates. The mRNA levels of arlR and rsbU in all of the ica(+)aap(+)/BF(-) isolates were lower than in RP62A at 4 h. At 10 h, 14/15 of the isolates showed lower mRNA levels of arlR and rsbU than shown by RP62A. However, expression of sarA, luxS, srrA and icaR varied in different ica(+)aap(+)/BF(-) isolates. To further investigate the role of arlRS in biofilm formation, we analyzed icaA, sarA and rsbU transcription, PIA synthesis, Aap expression and biofilm formation in an arlRS deletion mutant of S. epidermidis strain 1457 and all were much less than in the wild type strain. This is consistent with the hypothesis that ArlRS may play an important role in regulating biofilm formation by the ica(+)aap(+)/BF(-)S. epidermidis clinical isolates and operate via both ica-dependent and Aap-dependent pathways.
Collapse
Affiliation(s)
- Yang Wu
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Institute of Medical Microbiology and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, 138 Yi Xue Yuan Road, Shanghai 200032, PR China
| | - Jingran Liu
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Institute of Medical Microbiology and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, 138 Yi Xue Yuan Road, Shanghai 200032, PR China
| | - Juan Jiang
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Institute of Medical Microbiology and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, 138 Yi Xue Yuan Road, Shanghai 200032, PR China
| | - Jian Hu
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Institute of Medical Microbiology and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, 138 Yi Xue Yuan Road, Shanghai 200032, PR China
| | - Tao Xu
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Institute of Medical Microbiology and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, 138 Yi Xue Yuan Road, Shanghai 200032, PR China
| | - Jiaxue Wang
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Institute of Medical Microbiology and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, 138 Yi Xue Yuan Road, Shanghai 200032, PR China
| | - Di Qu
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Institute of Medical Microbiology and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, 138 Yi Xue Yuan Road, Shanghai 200032, PR China.
| |
Collapse
|
103
|
Staphylococcus epidermidis agr quorum-sensing system: signal identification, cross talk, and importance in colonization. J Bacteriol 2014; 196:3482-93. [PMID: 25070736 DOI: 10.1128/jb.01882-14] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Staphylococcus epidermidis is an opportunistic pathogen that is one of the leading causes of medical device infections. Global regulators like the agr quorum-sensing system in this pathogen have received a limited amount of attention, leaving important questions unanswered. There are three agr types in S. epidermidis strains, but only one of the autoinducing peptide (AIP) signals has been identified (AIP-I), and cross talk between agr systems has not been tested. We structurally characterized all three AIP types using mass spectrometry and discovered that the AIP-II and AIP-III signals are 12 residues in length, making them the largest staphylococcal AIPs identified to date. S. epidermidis agr reporter strains were developed for each system, and we determined that cross-inhibitory interactions occur between the agr type I and II systems and between the agr type I and III systems. In contrast, no cross talk was observed between the type II and III systems. To further understand the outputs of the S. epidermidis agr system, an RNAIII mutant was constructed, and microarray studies revealed that exoenzymes (Ecp protease and Geh lipase) and low-molecular-weight toxins were downregulated in the mutant. Follow-up analysis of Ecp confirmed the RNAIII is required to induce protease activity and that agr cross talk modulates Ecp activity in a manner that mirrors the agr reporter results. Finally, we demonstrated that the agr system enhances skin colonization by S. epidermidis using a porcine model. This work expands our knowledge of S. epidermidis agr system function and will aid future studies on cell-cell communication in this important opportunistic pathogen.
Collapse
|
104
|
Taha M, Kalab M, Yi QL, Landry C, Greco-Stewart V, Brassinga AK, Sifri CD, Ramirez-Arcos S. Biofilm-forming skin microflora bacteria are resistant to the bactericidal action of disinfectants used during blood donation. Transfusion 2014; 54:2974-82. [DOI: 10.1111/trf.12728] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 04/04/2014] [Accepted: 04/04/2014] [Indexed: 12/21/2022]
Affiliation(s)
- Mariam Taha
- Canadian Blood Services; Ottawa Ontario Canada
| | | | - Qi-Long Yi
- Canadian Blood Services; Ottawa Ontario Canada
| | | | | | | | - Costi D. Sifri
- University of Virginia Health System; Charlottesville Virginia
| | | |
Collapse
|
105
|
You Y, Xue T, Cao L, Zhao L, Sun H, Sun B. Staphylococcus aureus glucose-induced biofilm accessory proteins, GbaAB, influence biofilm formation in a PIA-dependent manner. Int J Med Microbiol 2014; 304:603-12. [PMID: 24836943 DOI: 10.1016/j.ijmm.2014.04.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Revised: 03/28/2014] [Accepted: 04/19/2014] [Indexed: 10/25/2022] Open
Abstract
The Gram-positive bacteria Staphylococcus aureus and Staphylococcus epidermidis are capable of attaching to a biomaterial surface and forming resistant biofilms. The identification of biomolecular and regulatory factors involved in staphylococcal adhesion and biofilm formation is needed to understand biofilm-associated infection in humans. Here, we have identified a new operon, gbaAB (glucose induced biofilm accessory gene), that affects biofilm formation in S. aureus NCTC8325. Real-time reverse transcription PCR (RT-PCR) and electrophoretic mobility shift assay showed that GbaA and GbaB are transcribed from the same transcript, and GbaA directly inhibits the transcription of the gbaAB operon through self-repression. Our results indicated that the gbaA mutant displayed enhanced biofilm formation compared with the wild type. However, the gbaB and the gbaAB double mutant displayed reduced biofilm formation, suggesting that the gbaAB operon is involved in biofilm formation and that gbaB might be the key gene in biofilm regulation. Phenotypic analysis suggested that the gbaAB operon mediated biofilm formation of S. aureus at the multicellular aggregation stage rather than during initial attachment. In addition, real-time RT-PCR analysis showed that icaA was upregulated in the gbaA mutant and downregulated in the gbaB and gbaAB mutants compared with the wild type. In addition, the gbaA and the gbaB mutants affected the induction of biofilm formation by glucose. Our results suggest that the gbaAB operon is involved in the regulation of the multicellular aggregation step of S. aureus biofilm formation in response to glucose and that this regulation may be mediated through the ica operon.
Collapse
Affiliation(s)
- Yibo You
- Department of Microbiology and Immunology, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Ting Xue
- Department of Microbiology and Immunology, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Linyan Cao
- Department of Microbiology and Immunology, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Liping Zhao
- Department of Microbiology and Immunology, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Haipeng Sun
- Department of Microbiology and Immunology, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Baolin Sun
- Department of Microbiology and Immunology, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China.
| |
Collapse
|
106
|
Arginine deiminase in Staphylococcus epidermidis functions to augment biofilm maturation through pH homeostasis. J Bacteriol 2014; 196:2277-89. [PMID: 24727224 DOI: 10.1128/jb.00051-14] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Allelic replacement mutants were constructed within arginine deiminase (arcA1 and arcA2) to assess the function of the arginine deiminase (ADI) pathway in organic acid resistance and biofilm formation of Staphylococcus epidermidis 1457. A growth-dependent acidification assay (pH ∼5.0 to ∼5.2) determined that strain 1457 devoid of arginine deiminase activity (1457 ΔADI) was significantly less viable than the wild type following depletion of glucose and in the presence of arginine. However, no difference in viability was noted for individual 1457 ΔarcA1 (native) or ΔarcA2 (arginine catabolic mobile element [ACME]-derived) mutants, suggesting that the native and ACME-derived ADIs are compensatory in S. epidermidis. Furthermore, flow cytometry and electron paramagnetic resonance spectroscopy results suggested that organic acid stress resulted in oxidative stress that could be partially rescued by the iron chelator dipyridyl. Collectively, these results suggest that formation of hydroxyl radicals is partially responsible for cell death via organic acid stress and that ADI-derived ammonia functions to counteract this acid stress. Finally, static biofilm assays determined that viability, ammonia synthesis, and pH were reduced in strain 1457 ΔADI following 120 h of growth in comparison to strain 1457 and the arcA1 and arcA2 single mutants. It is hypothesized that ammonia synthesis via the ADI pathway is important to reduce pH stress in specific microniches that contain high concentrations of organic acids.
Collapse
|
107
|
Influence of iron and aeration on Staphylococcus aureus growth, metabolism, and transcription. J Bacteriol 2014; 196:2178-89. [PMID: 24706736 DOI: 10.1128/jb.01475-14] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Staphylococcus aureus is a prominent nosocomial pathogen and a major cause of biomaterial-associated infections. The success of S. aureus as a pathogen is due in part to its ability to adapt to stressful environments. As an example, the transition from residing in the nares to residing in the blood or deeper tissues is accompanied by changes in the availability of nutrients and elements such as oxygen and iron. As such, nutrients, oxygen, and iron are important determinants of virulence factor synthesis in S. aureus. In addition to influencing virulence factor synthesis, oxygen and iron are critical cofactors in enzymatic and electron transfer reactions; thus, a change in iron or oxygen availability alters the bacterial metabolome. Changes in metabolism create intracellular signals that alter the activity of metabolite-responsive regulators such as CodY, RpiRc, and CcpA. To assess the extent of metabolomic changes associated with oxygen and iron limitation, S. aureus cells were cultivated in iron-limited medium and/or with decreasing aeration, and the metabolomes were examined by nuclear magnetic resonance (NMR) spectroscopy. As expected, oxygen and iron limitation dramatically decreased tricarboxylic acid (TCA) cycle activity, creating a metabolic block and significantly altering the metabolome. These changes were most prominent during post-exponential-phase growth, when TCA cycle activity was maximal. Importantly, many of the effects of iron limitation were obscured by aeration limitation. Aeration limitation not only obscured the metabolic effects of iron limitation but also overrode the transcription of iron-regulated genes. Finally, in contrast to previous speculation, we confirmed that acidification of the culture medium occurs independent of the availability of iron.
Collapse
|
108
|
Alhede M, Er Ö, Eickhardt S, Kragh K, Alhede M, Christensen LD, Poulsen SS, Givskov M, Christensen LH, Høiby N, Tvede M, Bjarnsholt T. Bacterial biofilm formation and treatment in soft tissue fillers. Pathog Dis 2014; 70:339-46. [DOI: 10.1111/2049-632x.12139] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 01/07/2014] [Accepted: 01/07/2014] [Indexed: 12/01/2022] Open
Affiliation(s)
- Morten Alhede
- Department of International Health, Immunology and Microbiology; Faculty of Health Sciences; University of Copenhagen; Copenhagen Denmark
- Department of Clinical Microbiology 9301; Rigshospitalet; Copenhagen Denmark
| | - Özge Er
- Department of International Health, Immunology and Microbiology; Faculty of Health Sciences; University of Copenhagen; Copenhagen Denmark
| | - Steffen Eickhardt
- Department of International Health, Immunology and Microbiology; Faculty of Health Sciences; University of Copenhagen; Copenhagen Denmark
| | - Kasper Kragh
- Department of International Health, Immunology and Microbiology; Faculty of Health Sciences; University of Copenhagen; Copenhagen Denmark
| | - Maria Alhede
- Department of International Health, Immunology and Microbiology; Faculty of Health Sciences; University of Copenhagen; Copenhagen Denmark
| | - Louise Dahl Christensen
- Department of International Health, Immunology and Microbiology; Faculty of Health Sciences; University of Copenhagen; Copenhagen Denmark
| | - Steen Seier Poulsen
- Department of Biomedical Science; University of Copenhagen; Copenhagen Denmark
| | - Michael Givskov
- Department of International Health, Immunology and Microbiology; Faculty of Health Sciences; University of Copenhagen; Copenhagen Denmark
- Singapore Centre on Environmental Life Sciences Engineering; Nanyang Technological University; Nanyang Singapore
| | - Lise H. Christensen
- Bispebjerg Section; Department of Pathology; Rigshospitalet; University of Copenhagen; Copenhagen Denmark
| | - Niels Høiby
- Department of International Health, Immunology and Microbiology; Faculty of Health Sciences; University of Copenhagen; Copenhagen Denmark
- Department of Clinical Microbiology 9301; Rigshospitalet; Copenhagen Denmark
| | - Michael Tvede
- Department of Clinical Microbiology 9301; Rigshospitalet; Copenhagen Denmark
| | - Thomas Bjarnsholt
- Department of International Health, Immunology and Microbiology; Faculty of Health Sciences; University of Copenhagen; Copenhagen Denmark
- Department of Clinical Microbiology 9301; Rigshospitalet; Copenhagen Denmark
| |
Collapse
|
109
|
Combination prophylactic therapy with rifampin increases efficacy against an experimental Staphylococcus epidermidis subcutaneous implant-related infection. Antimicrob Agents Chemother 2014; 58:2377-86. [PMID: 24514089 DOI: 10.1128/aac.01943-13] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The incidence of infections related to cardiac devices (such as permanent pacemakers) has been increasing out of proportion to implantation rates. As management of device infections typically requires explantation of the device, optimal prophylactic strategies are needed. Cefazolin and vancomycin are widely used as single agents for surgical prophylaxis against cardiac device-related infections. However, combination antibiotic prophylaxis may further reduce infectious complications. To model a localized subcutaneous implant-related infection, a bioluminescent strain of Staphylococcus epidermidis was inoculated onto a medical-procedure-grade titanium disc, which was placed into a subcutaneous pocket in the backs of mice. In vivo bioluminescence imaging, quantification of ex vivo CFU from the capsules and implants, variable-pressure scanning electron microscopy (VP-SEM), and neutrophil enhanced green fluorescent protein (EGFP) fluorescence in LysEGFP mice were employed to monitor the infection. This model was used to evaluate the efficacies of low- and high-dose cefazolin (50 and 200 mg/kg of body weight) and vancomycin (10 and 110 mg/kg) intravenous prophylaxis with or without rifampin (25 mg/kg). High-dose cefazolin and high-dose vancomycin treatment resulted in almost complete bacterial clearance, whereas both low-dose cefazolin and low-dose vancomycin reduced the in vivo and ex vivo bacterial burden only moderately. The addition of rifampin to low-dose cefazolin and vancomycin was highly effective in further reducing the CFU harvested from the implants. However, vancomycin-rifampin was more effective than cefazolin-rifampin in further reducing the CFU harvested from the surrounding tissue capsules. Future studies in humans will be required to determine whether the addition of rifampin has improved efficacy in preventing device-related infections in clinical practice.
Collapse
|
110
|
Carvalhais V, França A, Cerca F, Vitorino R, Pier GB, Vilanova M, Cerca N. Dormancy within Staphylococcus epidermidis biofilms: a transcriptomic analysis by RNA-seq. Appl Microbiol Biotechnol 2014; 98:2585-96. [DOI: 10.1007/s00253-014-5548-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 01/13/2014] [Accepted: 01/14/2014] [Indexed: 12/17/2022]
|
111
|
Krismer B, Liebeke M, Janek D, Nega M, Rautenberg M, Hornig G, Unger C, Weidenmaier C, Lalk M, Peschel A. Nutrient limitation governs Staphylococcus aureus metabolism and niche adaptation in the human nose. PLoS Pathog 2014; 10:e1003862. [PMID: 24453967 PMCID: PMC3894218 DOI: 10.1371/journal.ppat.1003862] [Citation(s) in RCA: 137] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 11/18/2013] [Indexed: 01/23/2023] Open
Abstract
Colonization of the human nose by Staphylococcus aureus in one-third of the population represents a major risk factor for invasive infections. The basis for adaptation of S. aureus to this specific habitat and reasons for the human predisposition to become colonized have remained largely unknown. Human nasal secretions were analyzed by metabolomics and found to contain potential nutrients in rather low amounts. No significant differences were found between S. aureus carriers and non-carriers, indicating that carriage is not associated with individual differences in nutrient supply. A synthetic nasal medium (SNM3) was composed based on the metabolomics data that permits consistent growth of S. aureus isolates. Key genes were expressed in SNM3 in a similar way as in the human nose, indicating that SNM3 represents a suitable surrogate environment for in vitro simulation studies. While the majority of S. aureus strains grew well in SNM3, most of the tested coagulase-negative staphylococci (CoNS) had major problems to multiply in SNM3 supporting the notion that CoNS are less well adapted to the nose and colonize preferentially the human skin. Global gene expression analysis revealed that, during growth in SNM3, S. aureus depends heavily on de novo synthesis of methionine. Accordingly, the methionine-biosynthesis enzyme cysteine-γ-synthase (MetI) was indispensable for growth in SNM3, and the MetI inhibitor DL-propargylglycine inhibited S. aureus growth in SNM3 but not in the presence of methionine. Of note, metI was strongly up-regulated by S. aureus in human noses, and metI mutants were strongly abrogated in their capacity to colonize the noses of cotton rats. These findings indicate that the methionine biosynthetic pathway may include promising antimicrobial targets that have previously remained unrecognized. Hence, exploring the environmental conditions facultative pathogens are exposed to during colonization can be useful for understanding niche adaptation and identifying targets for new antimicrobial strategies.
Collapse
Affiliation(s)
- Bernhard Krismer
- Interfaculty Institute of Microbiology and Infection Medicine, Cellular and Molecular Microbiology, Eberhard-Karls-University Tübingen, Tübingen, Germany
- German Center for Infection Research (DZIF), partner site Tübingen, Tübingen, Germany
- * E-mail:
| | - Manuel Liebeke
- Institute of Pharmacy, Ernst-Moritz-Arndt University of Greifswald, Greifswald, Germany
| | - Daniela Janek
- Interfaculty Institute of Microbiology and Infection Medicine, Cellular and Molecular Microbiology, Eberhard-Karls-University Tübingen, Tübingen, Germany
- German Center for Infection Research (DZIF), partner site Tübingen, Tübingen, Germany
| | - Mulugeta Nega
- Interfaculty Institute of Microbiology and Infection Medicine, Microbial Genetics, Eberhard-Karls-University Tübingen, Tübingen, Germany
| | - Maren Rautenberg
- Interfaculty Institute of Microbiology and Infection Medicine, Cellular and Molecular Microbiology, Eberhard-Karls-University Tübingen, Tübingen, Germany
- German Center for Infection Research (DZIF), partner site Tübingen, Tübingen, Germany
| | - Gabriele Hornig
- Interfaculty Institute of Microbiology and Infection Medicine, Cellular and Molecular Microbiology, Eberhard-Karls-University Tübingen, Tübingen, Germany
- German Center for Infection Research (DZIF), partner site Tübingen, Tübingen, Germany
| | - Clemens Unger
- Institute for Tropical Medicine, University of Tübingen, Tübingen, Germany
| | - Christopher Weidenmaier
- Interfaculty Institute of Microbiology and Infection Medicine, Cellular and Molecular Microbiology, Eberhard-Karls-University Tübingen, Tübingen, Germany
- German Center for Infection Research (DZIF), partner site Tübingen, Tübingen, Germany
| | - Michael Lalk
- Institute of Pharmacy, Ernst-Moritz-Arndt University of Greifswald, Greifswald, Germany
| | - Andreas Peschel
- Interfaculty Institute of Microbiology and Infection Medicine, Cellular and Molecular Microbiology, Eberhard-Karls-University Tübingen, Tübingen, Germany
- German Center for Infection Research (DZIF), partner site Tübingen, Tübingen, Germany
| |
Collapse
|
112
|
Waters EM, McCarthy H, Hogan S, Zapotoczna M, O'Neill E, O'Gara JP. Rapid quantitative and qualitative analysis of biofilm production by Staphylococcus epidermidis under static growth conditions. Methods Mol Biol 2014; 1106:157-166. [PMID: 24222464 DOI: 10.1007/978-1-62703-736-5_14] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Rapid screening of biofilm forming capacity by Staphylococcus epidermidis is possible using in vitro assays with 96-well plates. This method first developed by Christensen et al. in 1985 is fast and does not require specialized instruments. Thus, laboratories with standard microbiology infrastructure and a 96-well plate reader can easily use this technique to generate data on the biofilm phenotypes of multiple S. epidermidis strains and clinical isolates. Furthermore, this method can be adapted to gain insights into biofilm regulation and the characteristics of biofilms produced by different S. epidermidis isolates. Although this assay is extremely useful for showing whether individual strains are biofilm-positive or biofilm-negative and distinguishing between form weak, moderate or strong biofilm, it is important to acknowledge that the absolute levels of biofilm produced by an individual strain can vary significantly between experiments meaning that strict adherence to the protocol used is of paramount importance. Furthermore, measuring biofilm under static conditions does not generally reflect in vivo conditions in which bacteria are often subjected to shear stresses under flow conditions. Hence, the biofilm characteristics of some strains are dramatically different under flow and static conditions. Nevertheless, rapid measurement of biofilm production under static conditions is a useful tool in the analysis of the S. epidermidis biofilm phenotype.
Collapse
Affiliation(s)
- Elaine M Waters
- Department of Pathology and Microbiology, Center for Staphylococcal Research, University of Nebraska Medical Center, Omaha, NE, USA
| | | | | | | | | | | |
Collapse
|
113
|
Somerville GA, Powers R. Growth and preparation of Staphylococcus epidermidis for NMR metabolomic analysis. Methods Mol Biol 2014; 1106:71-91. [PMID: 24222456 DOI: 10.1007/978-1-62703-736-5_6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The "omics" era began with transcriptomics and this progressed into proteomics. While useful, these approaches provide only circumstantial information about carbon flow, metabolic status, redox poise, etc. To more directly address these metabolic concerns, researchers have turned to the emerging field of metabolomics. In our laboratories, we frequently use NMR metabolomics to acquire a snapshot of bacterial metabolomes during stressful or transition events. Irrespective of the "omics" method of choice, the experimental outcome depends on the proper cultivation and preparation of bacterial samples. In addition, the integration of these large datasets requires that these cultivation conditions be clearly defined.
Collapse
Affiliation(s)
- Greg A Somerville
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA
| | | |
Collapse
|
114
|
Widhelm TJ, Yajjala VK, Endres JL, Fey PD, Bayles KW. Methods to generate a sequence-defined transposon mutant library in Staphylococcus epidermidis strain 1457. Methods Mol Biol 2014; 1106:135-42. [PMID: 24222462 DOI: 10.1007/978-1-62703-736-5_12] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Transposon mutant libraries are valuable resources to investigators studying bacterial species, including Staphylococcus epidermidis, which are difficult to genetically manipulate. Although sequence-defined transposon mutant libraries have been constructed in Staphylococcus aureus, no such library exists for S. epidermidis. Nevertheless, the study of Tn917-mediated mutations has been paramount in discovering unique aspects of S. epidermidis biology including initial adherence and accumulation during biofilm formation. Herein, we describe modifications to the methodology first described by Bae et al. to utilize the mariner-based transposon bursa aurealis to generate mutants in S. epidermidis strain 1457.
Collapse
Affiliation(s)
- Todd J Widhelm
- Department of Pathology and Microbiology, Center for Staphylococcal Research, University of Nebraska Medical Center, Omaha, NE, USA
| | | | | | | | | |
Collapse
|
115
|
Nicholson TL, Shore SM, Smith TC, Frana TS. Livestock-associated methicillin-resistant Staphylococcus aureus (LA-MRSA) isolates of swine origin form robust biofilms. PLoS One 2013; 8:e73376. [PMID: 23951352 PMCID: PMC3739819 DOI: 10.1371/journal.pone.0073376] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Accepted: 07/22/2013] [Indexed: 01/11/2023] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) colonization of livestock animals is common and prevalence rates for pigs have been reported to be as high as 49%. Mechanisms contributing to the persistent carriage and high prevalence rates of livestock-associated methicillin-resistant Staphylococcus aureus (LA-MRSA) strains in swine herds and production facilities have not been investigated. One explanation for the high prevalence of MRSA in swine herds is the ability of these organisms to exist as biofilms. In this report, the ability of swine LA-MRSA strains, including ST398, ST9, and ST5, to form biofilms was quantified and compared to several swine and human isolates. The contribution of known biofilm matrix components, polysaccharides, proteins and extracellular DNA (eDNA), was tested in all strains as well. All MRSA swine isolates formed robust biofilms similar to human clinical isolates. The addition of Dispersin B had no inhibitory effect on swine MRSA isolates when added at the initiation of biofilm growth or after pre-established mature biofilms formed. In contrast, the addition of proteinase K inhibited biofilm formation in all strains when added at the initiation of biofilm growth and was able to disperse pre-established mature biofilms. Of the LA-MRSA strains tested, we found ST398 strains to be the most sensitive to both inhibition of biofilm formation and dispersal of pre-formed biofilms by DNaseI. Collectively, these findings provide a critical first step in designing strategies to control or eliminate MRSA in swine herds.
Collapse
Affiliation(s)
- Tracy L Nicholson
- National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, Iowa, USA.
| | | | | | | |
Collapse
|
116
|
Freitas AI, Vasconcelos C, Vilanova M, Cerca N. Optimization of an automatic counting system for the quantification of Staphylococcus epidermidis cells in biofilms. J Basic Microbiol 2013; 54:750-7. [PMID: 23686681 DOI: 10.1002/jobm.201200603] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 01/19/2013] [Indexed: 11/05/2022]
Abstract
Biofilm formation is recognized as the main virulence factor in a variety of chronic infections. In vitro evaluation of biofilm formation is often achieved by quantification of viable or total cells. However, these methods depend on biofilm disruption, which is often achieved by vortexing or sonication. In this study, we investigated the effects of sonication on the elimination of Staphylococcus epidermidis cell clusters from biofilms grown over time, and quantification was performed by three distinct analytical techniques. Even when a higher number of sonication cycles was used, some stable cell clusters remained in the samples obtained from 48- and 72-h-old biofilms, interfering with the quantification of sessile bacteria by plate counting. On the other hand, the fluorescence microscopy automatic counting system allowed proper quantification of biofilm samples that had undergone any of the described sonication cycles, suggesting that this is a more accurate method for assessing the cell concentration in S. epidermidis biofilms, especially in mature biofilms.
Collapse
Affiliation(s)
- Ana Isabel Freitas
- IBB - Institute for Biotechnology and Bioengineering, University of Minho, Campus de Gualtar, Braga, Portugal; ICBAS-UP - Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | | | | | | |
Collapse
|
117
|
Abstract
Staphylococcus aureus is a ubiquitous bacterial pathogen that is the causative agent of numerous acute and chronic infections. S. aureus colonizes the anterior nares of a significant portion of the healthy adult population, but the mechanisms of colonization remain incompletely defined. Sialic acid (N-acetylneuraminic acid [Neu5Ac]) is a bioavailable carbon and nitrogen source that is abundant on mucosal surfaces and in secretions in the commensal environment. Our findings demonstrate that Neu5Ac can serve as an S. aureus carbon source, and we have identified a previously uncharacterized chromosomal locus (nan) that is required for Neu5Ac utilization. Molecular characterization of the nan locus indicates that it contains five genes, organized into four transcripts, and the genes were renamed nanE, nanR, nanK, nanA, and nanT. Initial studies with gene deletions indicate that nanT, predicted to encode the Neu5Ac transporter, and nanA and nanE, predicted to encode catabolic enzymes, are essential for growth on Neu5Ac. Furthermore, a nanE deletion mutant exhibits a growth inhibition phenotype in the presence of Neu5Ac. Transcriptional fusions and Northern blot analyses indicate that NanR represses the expression of both the nanAT and nanE transcripts, which can be relieved with Neu5Ac. Electrophoretic mobility studies demonstrate that NanR binds to the nanAT and nanE promoter regions, and the Neu5Ac catabolic intermediate N-acetylmannosamine-6-phosphate (ManNAc-6P) relieves NanR promoter binding. Taken together, these data indicate that the nan gene cluster is essential for Neu5Ac utilization and may perform an important function for S. aureus survival in the host.
Collapse
|
118
|
Giant extracellular matrix binding protein expression in Staphylococcus epidermidis is regulated by biofilm formation and osmotic pressure. Curr Microbiol 2013; 66:627-33. [PMID: 23380801 DOI: 10.1007/s00284-013-0316-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 01/10/2013] [Indexed: 10/27/2022]
Abstract
Staphylococcus epidermidis is an opportunistic bacterium that thrives as a commensal cutaneous organism and as a vascular pathogen. The S. epidermidis extracellular matrix binding protein (Embp) has been reported to be a virulence factor involved in colonization of medical device implants and subsequent biofilm formation. Here, we characterize the expression patterns of Embp in planktonic and biofilm cultures, as well as under high osmotic stresses that typify the commensal environment of the skin. Embp expression without osmotic stress was similar for planktonic and adherent cultures. Addition of osmotic stress via NaCl caused slight increases in embp expression in planktonic cultures. However, in adherent cultures a 100-fold increase in embp expression with NaCl versus controls occurred and coincided with altered biofilm morphology. Results suggest that the central role of Embp lies in commensal skin colonization, stabilizing the cell wall against osmotic stresses, rather than as a virulence factor promoting adhesion.
Collapse
|
119
|
Quaternized chitosan as an antimicrobial agent: antimicrobial activity, mechanism of action and biomedical applications in orthopedics. Int J Mol Sci 2013; 14:1854-69. [PMID: 23325051 PMCID: PMC3565352 DOI: 10.3390/ijms14011854] [Citation(s) in RCA: 198] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 01/08/2013] [Accepted: 01/09/2013] [Indexed: 12/13/2022] Open
Abstract
Chitosan (CS) is a linear polysaccharide with good biodegradability, biocompatibility and antimicrobial activity, which makes it potentially useful for biomedical applications, including an antimicrobial agent either alone or blended with other polymers. However, the poor solubility of CS in most solvents at neutral or high pH substantially limits its use. Quaternary ammonium CS, which was prepared by introducing a quaternary ammonium group on a dissociative hydroxyl group or amino group of the CS, exhibited improved water solubility and stronger antibacterial activity relative to CS over an entire range of pH values; thus, this quaternary modification increases the potential biomedical applications of CS in the field of anti-infection. This review discusses the current findings on the antimicrobial properties of quaternized CS synthesized using different methods and the mechanisms of its antimicrobial actions. The potential antimicrobial applications in the orthopedic field and perspectives regarding future studies in this field are also considered.
Collapse
|
120
|
Lucilia sericata chymotrypsin disrupts protein adhesin-mediated staphylococcal biofilm formation. Appl Environ Microbiol 2012; 79:1393-5. [PMID: 23220967 DOI: 10.1128/aem.03689-12] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Staphylococcus aureus and Staphylococcus epidermidis biofilms cause chronic infections due to their ability to form biofilms. The excretions/secretions of Lucilia sericata larvae (maggots) have effective activity for debridement and disruption of bacterial biofilms. In this paper, we demonstrate how chymotrypsin derived from maggot excretions/secretions disrupts protein-dependent bacterial biofilm formation mechanisms.
Collapse
|
121
|
Staphylococcus aureus autoinducer-2 quorum sensing decreases biofilm formation in an icaR-dependent manner. BMC Microbiol 2012; 12:288. [PMID: 23216979 PMCID: PMC3539994 DOI: 10.1186/1471-2180-12-288] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Accepted: 11/29/2012] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Staphylococcus aureus is an important pathogen that causes biofilm-associated infection in humans. Autoinducer 2 (AI-2), a quorum-sensing (QS) signal for interspecies communication, has a wide range of regulatory functions in both Gram-positive and Gram-negative bacteria, but its exact role in biofilm formation in S. aureus remains unclear. RESULTS Here we demonstrate that mutation of the AI-2 synthase gene luxS in S. aureus RN6390B results in increased biofilm formation compared with the wild-type (WT) strain under static, flowing and anaerobic conditions and in a mouse model. Addition of the chemically synthesized AI-2 precursor in the luxS mutation strain (ΔluxS) restored the WT phenotype. Real-time RT-PCR analysis showed that AI-2 activated the transcription of icaR, a repressor of the ica operon, and subsequently a decreased level of icaA transcription, which was presumably the main reason why luxS mutation influences biofilm formation. Furthermore, we compared the roles of the agr-mediated QS system and the LuxS/AI-2 QS system in the regulation of biofilm formation using the ΔluxS strain, RN6911 and the Δagr ΔluxS strain. Our data indicate a cumulative effect of the two QS systems on the regulation of biofilm formation in S. aureus. CONCLUSION These findings demonstrate that AI-2 can decrease biofilm formation in S. aureus via an icaR-activation pathway. This study may provide clues for therapy in S. aureus biofilm-associated infection.
Collapse
|
122
|
Pavlukhina SV, Kaplan JB, Xu L, Chang W, Yu X, Madhyastha S, Yakandawala N, Mentbayeva A, Khan B, Sukhishvili SA. Noneluting enzymatic antibiofilm coatings. ACS APPLIED MATERIALS & INTERFACES 2012; 4:4708-16. [PMID: 22909396 PMCID: PMC3459334 DOI: 10.1021/am3010847] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
We developed a highly efficient, biocompatible surface coating that disperses bacterial biofilms through enzymatic cleavage of the extracellular biofilm matrix. The coating was fabricated by binding the naturally existing enzyme dispersin B (DspB) to surface-attached polymer matrices constructed via a layer-by-layer (LbL) deposition technique. LbL matrices were assembled through electrostatic interactions of poly(allylamine hydrochloride) (PAH) and poly(methacrylic acid) (PMAA), followed by chemical cross-linking with glutaraldehyde and pH-triggered removal of PMAA, producing a stable PAH hydrogel matrix used for DspB loading. The amount of DspB loaded increased linearly with the number of PAH layers in surface hydrogels. DspB was retained within these coatings in the pH range from 4 to 7.5. DspB-loaded coatings inhibited biofilm formation by two clinical strains of Staphylococcus epidermidis. Biofilm inhibition was ≥98% compared to mock-loaded coatings as determined by CFU enumeration. In addition, DspB-loaded coatings did not inhibit attachment or growth of cultured human osteoblast cells. We suggest that the use of DspB-loaded multilayer coatings presents a promising method for creating biocompatible surfaces with high antibiofilm efficiency, especially when combined with conventional antimicrobial treatment of dispersed bacteria.
Collapse
Affiliation(s)
- Svetlana V. Pavlukhina
- Department of Chemistry, Chemical Biology and Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, USA
| | - Jeffrey B. Kaplan
- Department of Oral Biology, New Jersey Dental School, University of Medicine and Dentistry of New Jersey, Newark, NJ 07103, USA
| | - Li Xu
- Department of Chemistry, Chemical Biology and Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, USA
| | - Wei Chang
- Department of Chemistry, Chemical Biology and Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, USA
| | - Xiaojun Yu
- Department of Chemistry, Chemical Biology and Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, USA
| | - Srinivasa Madhyastha
- Kane Biotech Inc., 162–196 Innovation Drive, Winnipeg, MB, Canada R3T 2N2, Canada
| | | | - Almagul Mentbayeva
- Department of Chemistry, 71, Al-Farabi Ave, Kazakh National University, Almaty, 050038, Kazakhstan
| | - Babar Khan
- Department of Oral Biology, New Jersey Dental School, University of Medicine and Dentistry of New Jersey, Newark, NJ 07103, USA
| | - Svetlana A. Sukhishvili
- Department of Chemistry, Chemical Biology and Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, USA
| |
Collapse
|
123
|
Sibbald MJJB, Yang XM, Tsompanidou E, Qu D, Hecker M, Becher D, Buist G, van Dijl JM. Partially overlapping substrate specificities of staphylococcal group A sortases. Proteomics 2012; 12:3049-62. [PMID: 22930668 DOI: 10.1002/pmic.201200144] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Revised: 07/12/2012] [Accepted: 07/27/2012] [Indexed: 11/10/2022]
Abstract
Sortases catalyze the covalent attachment of proteins with a C-terminal LPxTG motif to the cell walls of Gram-positive bacteria. Here, we show that deletion of the srtA genes of Staphylococcus aureus and Staphylococcus epidermidis resulted in the dislocation of several LPxTG proteins from the cell wall to the growth medium. Nevertheless, proteomics and Western blotting analyses revealed that substantial amounts of the identified proteins remained cell wall bound through noncovalent interactions. The protein dislocation phenotypes of srtA mutants of S. aureus and S. epidermidis were reverted by ectopic expression of srtA genes of either species. Interestingly, S. epidermidis contains a second sortase A, which was previously annotated as ``SrtC.'' Ectopic expression of this SrtC in srtA mutant cells reverted the dislocation of some, but not all, cell wall associated proteins. Similarly, defects in biofilm formation were reverted by ectopic expression of SrtC in some, but not all, tested srtA mutant strains. Finally, overexpression of SrtA resulted in increased levels of biofilm formation in some tested strains. Taken together, these findings show that the substrate specificities of SrtA and SrtC overlap partially, and that sortase levels may be limiting for biofilm formation in some staphylococci.
Collapse
Affiliation(s)
- Mark J J B Sibbald
- Department of Medical Microbiology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
124
|
Christner M, Heinze C, Busch M, Franke G, Hentschke M, Bayard Dühring S, Büttner H, Kotasinska M, Wischnewski V, Kroll G, Buck F, Molin S, Otto M, Rohde H. sarAnegatively regulatesStaphylococcus epidermidisbiofilm formation by modulating expression of 1 MDa extracellular matrix binding protein and autolysis-dependent release of eDNA. Mol Microbiol 2012; 86:394-410. [DOI: 10.1111/j.1365-2958.2012.08203.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/03/2012] [Indexed: 12/13/2022]
Affiliation(s)
- Martin Christner
- Institute for Medical Microbiology, Virology and Hygiene; University Medical Centre Hamburg-Eppendorf; Martinistraße 52; 20246; Hamburg; Germany
| | - Constanze Heinze
- Institute for Medical Microbiology, Virology and Hygiene; University Medical Centre Hamburg-Eppendorf; Martinistraße 52; 20246; Hamburg; Germany
| | - Michael Busch
- Institute for Medical Microbiology, Virology and Hygiene; University Medical Centre Hamburg-Eppendorf; Martinistraße 52; 20246; Hamburg; Germany
| | - Gefion Franke
- Institute for Medical Microbiology, Virology and Hygiene; University Medical Centre Hamburg-Eppendorf; Martinistraße 52; 20246; Hamburg; Germany
| | - Moritz Hentschke
- Institute for Medical Microbiology, Virology and Hygiene; University Medical Centre Hamburg-Eppendorf; Martinistraße 52; 20246; Hamburg; Germany
| | - Sara Bayard Dühring
- Center for Systems Microbiology, Department of Systems Biology; Technical University of Denmark; 2800; Lyngby; Denmark
| | - Henning Büttner
- Institute for Medical Microbiology, Virology and Hygiene; University Medical Centre Hamburg-Eppendorf; Martinistraße 52; 20246; Hamburg; Germany
| | - Marta Kotasinska
- Institute for Medical Microbiology, Virology and Hygiene; University Medical Centre Hamburg-Eppendorf; Martinistraße 52; 20246; Hamburg; Germany
| | - Victoria Wischnewski
- Institute for Medical Microbiology, Virology and Hygiene; University Medical Centre Hamburg-Eppendorf; Martinistraße 52; 20246; Hamburg; Germany
| | - Gesche Kroll
- Institute for Medical Microbiology, Virology and Hygiene; University Medical Centre Hamburg-Eppendorf; Martinistraße 52; 20246; Hamburg; Germany
| | - Friedrich Buck
- Institute for Clinical Chemistry; University Medical Centre Hamburg-Eppendorf; Martinistraße 52; 20246; Hamburg; Germany
| | - Soeren Molin
- Center for Systems Microbiology, Department of Systems Biology; Technical University of Denmark; 2800; Lyngby; Denmark
| | - Michael Otto
- Laboratory of Human Bacterial Pathogenesis; National Institute of Allergy and Infectious Diseases; The National Institutes of Health; Bethesda; MD; 20892; USA
| | - Holger Rohde
- Institute for Medical Microbiology, Virology and Hygiene; University Medical Centre Hamburg-Eppendorf; Martinistraße 52; 20246; Hamburg; Germany
| |
Collapse
|
125
|
Linnes J, Mikhova K, Bryers J. Adhesion of Staphylococcus epidermidis to biomaterials is inhibited by fibronectin and albumin. J Biomed Mater Res A 2012; 100:1990-7. [PMID: 22566405 PMCID: PMC3381423 DOI: 10.1002/jbm.a.34036] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Revised: 11/14/2011] [Accepted: 11/29/2011] [Indexed: 11/07/2022]
Abstract
Decades of contradictory results have obscured the exact role of adsorbed fibronectin in the adhesion of the bacterium, Staphylococcus epidermidis, to biomaterials. Here, the ability of adsorbed fibronectin (FN) or bovine serum albumin (BSA) to modulate S. epidermidis adhesion to various biomaterials is reported. FN or BSA was adsorbed in increasing surface densities up to saturated monolayer coverage onto various common biomaterials, including poly(ethylene terephthalate), fluorinated ethylene propylene, poly(ether urethane), silicone, and borosilicate glass. Despite the wide range of surface characteristics represented, adsorption isotherms varied only subtly between materials for the two proteins considered. S. epidermidis adhesion to the various protein-coated biomaterials was quantified in a static-fluid batch adhesion assay. Although slight differences in overall adherent cell numbers were observed between the various protein-coated substrata, all materials exhibited significant dose-dependent decreases in S. epidermidis adhesion with increasing adsorption of either protein (FN, BSA) to all surfaces. Results here indicate that S. epidermidis adhesion to FN-coated surfaces is not a specific adhesion (i.e., receptor: ligand) mediated process, as no significant difference in adhesion was found between FN- and BSA-coated materials. Rather, results indicate that increasing surface density of either FN or BSA actually inhibited S. epidermidis adhesion to all biomaterials examined.
Collapse
Affiliation(s)
- J.C. Linnes
- Department of Bioengineering, University of Washington, Seattle, WA, USA 98195-5061
| | - K. Mikhova
- Department of Bioengineering, University of Washington, Seattle, WA, USA 98195-5061
| | - J.D. Bryers
- Department of Bioengineering, University of Washington, Seattle, WA, USA 98195-5061
| |
Collapse
|
126
|
Abstract
The increased use of medical implants has resulted in a concomitant rise in device-related infections. The majority of these infections are caused by Staphylococcus epidermidis biofilms. Immunoprophylaxis and immunotherapy targeting in vivo-expressed, biofilm-associated, bacterial cell surface-exposed proteins are promising new approaches to prevent and treat biofilm-related infections, respectively. Using an in silico procedure, we identified 64 proteins that are predicted to be S. epidermidis surface exposed (Ses), of which 36 were annotated as (conserved) hypothetical. Of these 36 proteins, 5 proteins-3 LPXTG motif-containing proteins (SesL, SesB, and SesC) and 2 of the largest ABC transporters (SesK and SesM)-were selected for evaluation as vaccine candidates. This choice was based on protein size, number of antigenic determinants, or the established role in S. epidermidis biofilm formation of the protein family to which the candidate protein belongs. Anti-SesC antibodies exhibited the greatest inhibitory effect on S. epidermidis biofilm formation in vitro and on colonization and infection in a mouse jugular vein catheter infection model that includes biofilms and organ infections. Active vaccination with a recombinant truncated SesC inhibited S. epidermidis biofilm formation in a rat model of subcutaneous foreign body infection. Antibodies to SesC were shown to be opsonic by an in vitro opsonophagocytosis assay. We conclude that SesC is a promising target for antibody mediated strategies against S. epidermidis biofilm formation.
Collapse
|
127
|
Strunk T, Prosser A, Levy O, Philbin V, Simmer K, Doherty D, Charles A, Richmond P, Burgner D, Currie A. Responsiveness of human monocytes to the commensal bacterium Staphylococcus epidermidis develops late in gestation. Pediatr Res 2012; 72:10-8. [PMID: 22460219 DOI: 10.1038/pr.2012.48] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Staphylococcus epidermidis (SE) rarely causes infection in term infants but is a leading cause of late-onset sepsis in preterm infants. We hypothesized that the innate immune responses to SE in preterm infants are impaired in a gestational age (GA)-dependent manner. METHODS Cord and peripheral blood mononuclear cells (MNCs) were stimulated with SE bacteria, and a range of innate immune responses were assessed, including phagocytosis, intracellular killing, Toll-like receptor (TLR) pathway transcriptional activation, cytokine production, TLR2 and TLR4 expression, and cell signaling. RESULTS Phagocytosis and intracellular killing of SE bacteria were similar in neonatal and adult monocytes. Cytokine gene expression and protein synthesis increased in a GA-dependent manner, which was confirmed at the single-cell level. These GA-related effects were not associated with differences in expression of TLR2 or TLR4, nor with downstream activation of nuclear factor-κB or mitogen-activated protein kinase pathways. DISCUSSION The expression of TLRs, phagocytic capacity, and intracellular killing by monocytes develops early in fetal development, whereas the ability to mount a bacteria-induced cytokine response requires further maturation. The functional immaturity of monocyte activation pathways in the preterm infant may underpin their particular susceptibility to sepsis with commensal bacteria.
Collapse
Affiliation(s)
- Tobias Strunk
- University of Western Australia Centre for Neonatal Research and Education, Perth, Western Australia, Australia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
128
|
Abstract
Infectious diseases can be difficult to cure, especially if the pathogen forms a biofilm. After decades of extensive research into the morphology, physiology and genomics of biofilm formation, attention has recently been directed toward the analysis of the cellular metabolome in order to understand the transformation of a planktonic cell to a biofilm. Metabolomics can play an invaluable role in enhancing our understanding of the underlying biological processes related to the structure, formation and antibiotic resistance of biofilms. A systematic view of metabolic pathways or processes responsible for regulating this 'social structure' of microorganisms may provide critical insights into biofilm-related drug resistance and lead to novel treatments. This review will discuss the development of NMR-based metabolomics as a technology to study medically relevant biofilms. Recent advancements from case studies reviewed in this manuscript have shown the potential of metabolomics to shed light on numerous biological problems related to biofilms.
Collapse
Affiliation(s)
- Bo Zhang
- Department of Chemistry, University of Nebraska-Lincoln, 722 Hamilton Hall, Lincoln, NE 68588-0304, USA
| | - Robert Powers
- Department of Chemistry, University of Nebraska-Lincoln, 722 Hamilton Hall, Lincoln, NE 68588-0304, USA
| |
Collapse
|
129
|
Patel JD, Colton E, Ebert M, Anderson JM. Gene expression duringS. epidermidisbiofilm formation on biomaterials. J Biomed Mater Res A 2012; 100:2863-9. [DOI: 10.1002/jbm.a.34221] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Accepted: 01/17/2012] [Indexed: 11/09/2022]
|
130
|
Spiliopoulou AI, Krevvata MI, Kolonitsiou F, Harris LG, Wilkinson TS, Davies AP, Dimitracopoulos GO, Karamanos NK, Mack D, Anastassiou ED. An extracellular Staphylococcus epidermidis polysaccharide: relation to Polysaccharide Intercellular Adhesin and its implication in phagocytosis. BMC Microbiol 2012; 12:76. [PMID: 22594478 PMCID: PMC3431232 DOI: 10.1186/1471-2180-12-76] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2011] [Accepted: 05/17/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The skin commensal and opportunistic pathogen Staphylococcus epidermidis is a leading cause of hospital-acquired and biomaterial-associated infections. The polysaccharide intercellular adhesin (PIA), a homoglycan composed of β-1,6-linked N-acetylglucosamine residues, synthesized by enzymes encoded in icaADBC is a major functional factor in biofilm accumulation, promoting virulence in experimental biomaterial-associated S. epidermidis infection. Extracellular mucous layer extracts of S. epidermidis contain another major polysaccharide, referred to as 20-kDa polysaccharide (20-kDaPS), composed mainly out of glucose, N-acetylglucosamine, and being partially sulfated. 20-kDaPS antiserum prevents adhesion of S. epidermidis on endothelial cells and development of experimental keratitis in rabbits. Here we provide experimental evidence that 20-kDaPS and PIA represent distinct molecules and that 20-kDaPS is implicated in endocytosis of S. epidermidis bacterial cells by human monocyte-derived macrophages. RESULTS Analysis of 75 clinical coagulase-negative staphylococci from blood-cultures and central venous catheter tips indicated that 20-kDaPS is expressed exclusively in S. epidermidis but not in other coagulase-negative staphylococcal species. Tn917-insertion in various locations in icaADBC in mutants M10, M22, M23, and M24 of S. epidermidis 1457 are abolished for PIA synthesis, while 20-kDaPS expression appears unaltered as compared to wild-type strains using specific anti-PIA and anti-20-kDaPS antisera. While periodate oxidation and dispersin B treatments abolish immuno-reactivity and intercellular adhesive properties of PIA, no abrogative activity is exerted towards 20-kDaPS immunochemical reactivity following these treatments. PIA polysaccharide I-containing fractions eluting from Q-Sepharose were devoid of detectable 20-kDaPS using specific ELISA. Preincubation of non-20-kDaPS-producing clinical strain with increasing amounts of 20-kDaPS inhibits endocytosis by human macrophages, whereas, preincubation of 20-kDaPS-producing strain ATCC35983 with 20-kDaPS antiserum enhances bacterial endocytosis by human macrophages. CONCLUSIONS In conclusion, icaADBC is not involved in 20-kDaPS synthesis, while the chemical and chromatographic properties of PIA and 20-kDaPS are distinct. 20-kDaPS exhibits anti-phagocytic properties, whereas, 20-kDaPS antiserum may have a beneficial effect on combating infection by 20-kDaPS-producing S. epidermidis.
Collapse
Affiliation(s)
- Anastasia I Spiliopoulou
- Department of Microbiology, School of Medicine, University of Patras, Patras, Greece
- Medical Microbiology and Infectious Diseases, Institute of Life Science, The College of Medicine, Swansea University, Swansea, UK
| | - Maria I Krevvata
- Department of Microbiology, School of Medicine, University of Patras, Patras, Greece
| | - Fevronia Kolonitsiou
- Department of Microbiology, School of Medicine, University of Patras, Patras, Greece
| | - Llinos G Harris
- Medical Microbiology and Infectious Diseases, Institute of Life Science, The College of Medicine, Swansea University, Swansea, UK
| | - Thomas S Wilkinson
- Medical Microbiology and Infectious Diseases, Institute of Life Science, The College of Medicine, Swansea University, Swansea, UK
| | - Angharad P Davies
- Medical Microbiology and Infectious Diseases, Institute of Life Science, The College of Medicine, Swansea University, Swansea, UK
| | | | - Nikos K Karamanos
- Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Dietrich Mack
- Medical Microbiology and Infectious Diseases, Institute of Life Science, The College of Medicine, Swansea University, Swansea, UK
| | | |
Collapse
|
131
|
Cerca N, Gomes F, Pereira S, Teixeira P, Oliveira R. Confocal laser scanning microscopy analysis of S. epidermidis biofilms exposed to farnesol, vancomycin and rifampicin. BMC Res Notes 2012; 5:244. [PMID: 22591918 PMCID: PMC3481475 DOI: 10.1186/1756-0500-5-244] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Accepted: 04/30/2012] [Indexed: 11/10/2022] Open
Abstract
Background Staphylococcus epidermidis is the major bacterial species found in biofilm-related infections on indwelling medical devices. Microbial biofilms are communities of bacteria adhered to a surface and surrounded by an extracellular polymeric matrix. Biofilms have been associated with increased antibiotic tolerance to the immune system. This increased resistance to conventional antibiotic therapy has lead to the search for new antimicrobial therapeutical agents. Farnesol, a quorum-sensing molecule in Candida albicans, has been described as impairing growth of several different microorganisms and we have previously shown its potential as an adjuvant in antimicrobial therapy against S. epidermidis. However, its mechanism of action in S. epidermidis is not fully known. In this work we better elucidate the role of farnesol against S: epidermidis biofilms using confocal laser scanning microscopy (CLSM). Findings 24 h biofilms were exposed to farnesol, vancomycin or rifampicin and were analysed by CLSM, after stained with a Live/Dead stain, a known indicator of cell viability, related with cell membrane integrity. Biofilms were also disrupted by sonication and viable and cultivable cells were quantified by colony forming units (CFU) plating. Farnesol showed a similar effect as vancomycin, both causing little reduction of cell viability but at the same time inducing significant changes in the biofilm structure. On the other hand, rifampicin showed a distinct action in S. epidermidis biofilms, by killing a significant proportion of biofilm bacteria. Conclusions While farnesol is not very efficient at killing biofilm bacteria, it damages cell membrane, as determined by the live/dead staining, in a similar way as vancomycin. Furthermore, farnesol might induce biofilm detachment, as determined by the reduced biofilm biomass, which can partially explain the previous findings regarding its role as a possible chemotherapy adjuvant.
Collapse
Affiliation(s)
- Nuno Cerca
- Institute for Biotechnology and Bioengineering, Centre of Biological Engineering, University of Minho, Campus de Gualtar, Braga, Portugal.
| | | | | | | | | |
Collapse
|
132
|
Pozzi C, Waters EM, Rudkin JK, Schaeffer CR, Lohan AJ, Tong P, Loftus BJ, Pier GB, Fey PD, Massey RC, O'Gara JP. Methicillin resistance alters the biofilm phenotype and attenuates virulence in Staphylococcus aureus device-associated infections. PLoS Pathog 2012; 8:e1002626. [PMID: 22496652 PMCID: PMC3320603 DOI: 10.1371/journal.ppat.1002626] [Citation(s) in RCA: 221] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Accepted: 02/23/2012] [Indexed: 01/15/2023] Open
Abstract
Clinical isolates of Staphylococcus aureus can express biofilm phenotypes promoted by the major cell wall autolysin and the fibronectin-binding proteins or the icaADBC-encoded polysaccharide intercellular adhesin/poly-N-acetylglucosamine (PIA/PNAG). Biofilm production in methicillin-susceptible S. aureus (MSSA) strains is typically dependent on PIA/PNAG whereas methicillin-resistant isolates express an Atl/FnBP-mediated biofilm phenotype suggesting a relationship between susceptibility to β-lactam antibiotics and biofilm. By introducing the methicillin resistance gene mecA into the PNAG-producing laboratory strain 8325-4 we generated a heterogeneously resistant (HeR) strain, from which a homogeneous, high-level resistant (HoR) derivative was isolated following exposure to oxacillin. The HoR phenotype was associated with a R602H substitution in the DHHA1 domain of GdpP, a recently identified c-di-AMP phosphodiesterase with roles in resistance/tolerance to β-lactam antibiotics and cell envelope stress. Transcription of icaADBC and PNAG production were impaired in the 8325-4 HoR derivative, which instead produced a proteinaceous biofilm that was significantly inhibited by antibodies against the mecA-encoded penicillin binding protein 2a (PBP2a). Conversely excision of the SCCmec element in the MRSA strain BH1CC resulted in oxacillin susceptibility and reduced biofilm production, both of which were complemented by mecA alone. Transcriptional activity of the accessory gene regulator locus was also repressed in the 8325-4 HoR strain, which in turn was accompanied by reduced protease production and significantly reduced virulence in a mouse model of device infection. Thus, homogeneous methicillin resistance has the potential to affect agr- and icaADBC-mediated phenotypes, including altered biofilm expression and virulence, which together are consistent with the adaptation of healthcare-associated MRSA strains to the antibiotic-rich hospital environment in which they are frequently responsible for device-related infections in immuno-compromised patients. The acquisition of mecA, which encodes penicillin binding protein 2a (PBP2a) and methicillin resistance, by Staphylococcus aureus has added to an already impressive array of virulence mechanisms including enzyme and toxin production, biofilm forming capacity and immune evasion. And yet clinical data does not indicate that healthcare-associated methicillin resistant S. aureus (MRSA) strains are more virulent than their methicillin-susceptible counterparts. Here our findings suggest that MRSA sacrifices virulence potential for antibiotic resistance and that expression of methicillin resistance alters the biofilm phenotype but does not interfere with the colonization of implanted medical devices in vivo. High level expression of PBP2a, which was associated with a mutation in the c-di-AMP phosphodiesterase gene gdpP, resulted in these pleiotrophic effects by blocking icaADBC-dependent polysaccharide type biofilm development and promoting an alternative PBP2a-mediated biofilm, repressing the accessory gene regulator and extracellular protease production, and attenuating virulence in a mouse device-infection model. Thus the adaptation of MRSA to the hospital environment has apparently focused on the acquisition of antibiotic resistance and retention of biofilm forming capacity, which are likely to be more advantageous than metabolically-expensive enzyme and toxin production in immunocompromised patients with implanted medical devices offering a route to infection.
Collapse
Affiliation(s)
- Clarissa Pozzi
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
- Channing Laboratory, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Elaine M. Waters
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Justine K. Rudkin
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Carolyn R. Schaeffer
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Amanda J. Lohan
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Pin Tong
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Brendan J. Loftus
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Gerald B. Pier
- Channing Laboratory, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Paul D. Fey
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Ruth C. Massey
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - James P. O'Gara
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
- * E-mail:
| |
Collapse
|
133
|
Kwasny SM, Opperman TJ. Static biofilm cultures of Gram-positive pathogens grown in a microtiter format used for anti-biofilm drug discovery. ACTA ACUST UNITED AC 2012; Chapter 13:Unit 13A.8. [PMID: 22294365 DOI: 10.1002/0471141755.ph13a08s50] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
An in vitro assay is presented for culturing staphylococcal biofilms and biofilms of nonmotile Gram-positive bacteria under static conditions in microtiter assay plates, and for the quantification of biofilm growth, using a simple staining procedure that measures amounts of bacterial cells and extracellular matrix. This basic assay can be adapted readily to study several aspects of biofilm formation, for high-throughput screening to identify small molecule inhibitors of biofilm formation or biofilm-defective mutants, and for quantifying the anti-biofilm activity of biofilm inhibitors.
Collapse
Affiliation(s)
- Steven M Kwasny
- Microbiotix, Anti-Infectives R&D, Worcester, Massachusetts, USA
| | | |
Collapse
|
134
|
Transforming the untransformable: application of direct transformation to manipulate genetically Staphylococcus aureus and Staphylococcus epidermidis. mBio 2012; 3:mBio.00277-11. [PMID: 22434850 PMCID: PMC3312211 DOI: 10.1128/mbio.00277-11] [Citation(s) in RCA: 415] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
UNLABELLED The strong restriction barrier present in Staphylococcus aureus and Staphylococcus epidermidis has limited functional genomic analysis to a small subset of strains that are amenable to genetic manipulation. Recently, a conserved type IV restriction system termed SauUSI (which specifically recognizes cytosine methylated DNA) was identified as the major barrier to transformation with foreign DNA. Here we have independently corroborated these findings in a widely used laboratory strain of S. aureus. Additionally, we have constructed a DNA cytosine methyltransferase mutant in the high-efficiency Escherichia coli cloning strain DH10B (called DC10B). Plasmids isolated from DC10B can be directly transformed into clinical isolates of S. aureus and S. epidermidis. We also show that the loss of restriction (both type I and IV) in an S. aureus USA300 strain does not have an impact on virulence. Circumventing the SauUSI restriction barrier, combined with an improved deletion and transformation protocol, has allowed the genetic manipulation of previously untransformable strains of these important opportunistic pathogens. IMPORTANCE Staphylococcal infections place a huge burden on the health care sector due both to their severity and also to the economic impact of treating the infections because of prolonged hospitalization. To improve the understanding of Staphylococcus aureus and Staphylococcus epidermidis infections, we have developed a series of improved techniques that allow the genetic manipulation of strains that were previously refractory to transformation. These developments will speed up the process of mutant construction and increase our understanding of these species as a whole, rather than just a small subset of strains that could previously be manipulated.
Collapse
|
135
|
Spiliopoulou AI, Kolonitsiou F, Krevvata MI, Leontsinidis M, Wilkinson TS, Mack D, Anastassiou ED. Bacterial adhesion, intracellular survival and cytokine induction upon stimulation of mononuclear cells with planktonic or biofilm phase Staphylococcus epidermidis. FEMS Microbiol Lett 2012; 330:56-65. [PMID: 22360699 DOI: 10.1111/j.1574-6968.2012.02533.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Revised: 02/11/2012] [Accepted: 02/20/2012] [Indexed: 11/28/2022] Open
Abstract
Staphylococcus epidermidis is a leading cause of hospital-acquired and biofilm-associated infections. Interactions of peripheral blood mononuclear cells (PBMCs) and monocyte-derived macrophages with planktonic or biofilm phase S. epidermidis cells were studied. Biofilm phase bacteria exhibited higher attachment, as well as, a 10-fold higher intracellular survival in monocyte-derived macrophages than their planktonic counterparts. Stimulation of PBMCs and monocyte-derived macrophages was performed with live or formalin-fixed bacterial cells. Supernatant concentration of selected cytokines was measured by Luminex(®) xMAP(™) technology at different time points. As compared to planktonic phase, biofilm phase bacteria elicited lower amounts of proinflammatory cytokines and Th1 response cytokines, such as TNFα, IL-12p40, IL-12p70 and IFN-γ, whereas they enhanced production of IL-8, GM-CSF and IL-13. This phenomenon was independent of formalin pretreatment. Taken together, these results may contribute to interpretation of observed silent course of biofilm-associated infections.
Collapse
|
136
|
Fredheim EGA, Granslo HN, Flægstad T, Figenschau Y, Rohde H, Sadovskaya I, Mollnes TE, Klingenberg C. Staphylococcus epidermidis polysaccharide intercellular adhesin activates complement. ACTA ACUST UNITED AC 2012; 63:269-80. [PMID: 22077230 DOI: 10.1111/j.1574-695x.2011.00854.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Staphylococcus epidermidis is a frequent cause of nosocomial infections. The central virulence factor of S. epidermidis is biofilm formation. Polysaccharide intercellular adhesin (PIA) constitutes the major biofilm matrix-component. PIA and biofilm have been implicated in S. epidermidis evasion of host immune defence. We examined the effects of S. epidermidis PIA on the inflammatory response with focus on complement activation. We used a human whole-blood ex vivo model of infection and compared the effects of a PIA-positive S. epidermidis strain (SE1457) and its PIA-negative isogenic mutant (M10). The independent effect of purified PIA on complement activation was investigated. In glucose-rich media, the mutant formed a proteinacious DNA-rich biofilm, whereas SE1457 formed a thick PIA-biofilm. In biofilm growth, SE1457 induced a stronger activation of the complement system compared with M10. We verified that purified PIA was independently responsible for a strong activation of the complement system. In contrast, M10 induced higher granulocyte activation by expression of CD11b and higher secretion of cytokines. We conclude that PIA has potent pro-inflammatory properties by activating the complement system. However, in a complex balance of the immune response, the decreased activation of granulocytes and cytokines by a PIA biofilm may limit host eradication of S. epidermidis.
Collapse
|
137
|
Hofmann CM, Bednar KJ, Anderson JM, Marchant RE. Disruption of Staphylococcus epidermidis biofilm formation using a targeted cationic peptide. J Biomed Mater Res A 2012; 100:1061-7. [DOI: 10.1002/jbm.a.33273] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Accepted: 04/18/2011] [Indexed: 11/11/2022]
|
138
|
Cerca F, Andrade F, França Â, Andrade EB, Ribeiro A, Almeida AA, Cerca N, Pier G, Azeredo J, Vilanova M. Staphylococcus epidermidis biofilms with higher proportions of dormant bacteria induce a lower activation of murine macrophages. J Med Microbiol 2011; 60:1717-1724. [PMID: 21799197 PMCID: PMC10727147 DOI: 10.1099/jmm.0.031922-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2011] [Accepted: 07/22/2011] [Indexed: 02/02/2023] Open
Abstract
Staphylococcus epidermidis is an opportunistic pathogen and, due to its ability to establish biofilms, is a leading causative agent of indwelling medical device-associated infection. The presence of high amounts of dormant bacteria is a hallmark of biofilms, making them more tolerant to antimicrobials and to the host immune response. We observed that S. epidermidis biofilms grown in excess glucose accumulated high amounts of viable but non-culturable (VBNC) bacteria, as assessed by their low ratio of culturable bacteria over the number of viable bacteria. This effect, which was a consequence of the accumulation of acidic compounds due to glucose metabolism, was counteracted by high extracellular levels of calcium and magnesium added to the culture medium allowing modulation of the proportions of VBNC bacteria within S. epidermidis biofilms. Using bacterial inocula obtained from biofilms with high and low proportions of VBNC bacteria, their stimulatory effect on murine macrophages was evaluated in vitro and in vivo. The inoculum enriched in VBNC bacteria induced in vitro a lower production of tumour necrosis factor alpha, interleukin-1 and interleukin-6 by bone-marrow-derived murine macrophages and, in vivo, a lower stimulatory effect on peritoneal macrophages, assessed by increased surface expression of Gr1 and major histocompatibility complex class II molecules. Overall, these results show that environmental conditions, such as pH and extracellular levels of calcium and magnesium, can induce dormancy in S. epidermidis biofilms. Moreover, they show that bacterial suspensions enriched in dormant cells are less inflammatory, suggesting that dormancy can contribute to the immune evasion of biofilms.
Collapse
Affiliation(s)
- Filipe Cerca
- ICBAS – Instituto de Ciências Biomédicas de Abel Salazar, Largo do
Professor Abel Salazar 2, 4099-003 Porto, Portugal
- IBMC – Instituto de Biologia Molecular e Celular, Rua do Campo Alegre
83, Porto, Portugal
| | - Filipa Andrade
- ICBAS – Instituto de Ciências Biomédicas de Abel Salazar, Largo do
Professor Abel Salazar 2, 4099-003 Porto, Portugal
| | - Ângela França
- ICBAS – Instituto de Ciências Biomédicas de Abel Salazar, Largo do
Professor Abel Salazar 2, 4099-003 Porto, Portugal
- CEB-IBB, Campus de Gualtar, Universidade do Minho, 4710-057 Braga,
Portugal
| | - Elva Bonifácio Andrade
- ICBAS – Instituto de Ciências Biomédicas de Abel Salazar, Largo do
Professor Abel Salazar 2, 4099-003 Porto, Portugal
- IBMC – Instituto de Biologia Molecular e Celular, Rua do Campo Alegre
83, Porto, Portugal
| | - Adília Ribeiro
- ICBAS – Instituto de Ciências Biomédicas de Abel Salazar, Largo do
Professor Abel Salazar 2, 4099-003 Porto, Portugal
| | - Agostinho A. Almeida
- REQUIMTE, Departamento de Química–Física, Faculdade de Farmácia,
Universidade do Porto, Rua Aníbal Cunha 164, 4099-030 Porto, Portugal
| | - Nuno Cerca
- CEB-IBB, Campus de Gualtar, Universidade do Minho, 4710-057 Braga,
Portugal
| | - Gerald Pier
- Channing Laboratory, Department of Medicine, Brigham and Women's
Hospital, Boston, MA 02115, USA
| | - Joana Azeredo
- CEB-IBB, Campus de Gualtar, Universidade do Minho, 4710-057 Braga,
Portugal
| | - Manuel Vilanova
- ICBAS – Instituto de Ciências Biomédicas de Abel Salazar, Largo do
Professor Abel Salazar 2, 4099-003 Porto, Portugal
- IBMC – Instituto de Biologia Molecular e Celular, Rua do Campo Alegre
83, Porto, Portugal
| |
Collapse
|
139
|
Sadykov MR, Hartmann T, Mattes TA, Hiatt M, Jann NJ, Zhu Y, Ledala N, Landmann R, Herrmann M, Rohde H, Bischoff M, Somerville GA. CcpA coordinates central metabolism and biofilm formation in Staphylococcus epidermidis. MICROBIOLOGY-SGM 2011; 157:3458-3468. [PMID: 21964732 DOI: 10.1099/mic.0.051243-0] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Staphylococcus epidermidis is an opportunistic bacterium whose infections often involve the formation of a biofilm on implanted biomaterials. In S. epidermidis, the exopolysaccharide facilitating bacterial adherence in a biofilm is polysaccharide intercellular adhesin (PIA), whose synthesis requires the enzymes encoded within the intercellular adhesin operon (icaADBC). In vitro, the formation of S. epidermidis biofilms is enhanced by conditions that repress tricarboxylic acid (TCA) cycle activity, such as growth in a medium containing glucose. In many Gram-positive bacteria, repression of TCA cycle genes in response to glucose is accomplished by catabolite control protein A (CcpA). CcpA is a member of the GalR-LacI repressor family that mediates carbon catabolite repression, leading us to hypothesize that catabolite control of S. epidermidis biofilm formation is indirectly regulated by CcpA-dependent repression of the TCA cycle. To test this hypothesis, ccpA deletion mutants were constructed in strain 1457 and 1457-acnA and the effects on TCA cycle activity, biofilm formation and virulence were assessed. As anticipated, deletion of ccpA derepressed TCA cycle activity and inhibited biofilm formation; however, ccpA deletion had only a modest effect on icaADBC transcription. Surprisingly, deletion of ccpA in strain 1457-acnA, a strain whose TCA cycle is inactive and where icaADBC transcription is derepressed, strongly inhibited icaADBC transcription. These observations demonstrate that CcpA is a positive effector of biofilm formation and icaADBC transcription and a repressor of TCA cycle activity.
Collapse
Affiliation(s)
- Marat R Sadykov
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska, Lincoln, NE, USA
| | - Torsten Hartmann
- Institute of Medical Microbiology and Hygiene, University of Saarland Hospital, Homburg/Saar, Germany
| | - Theodoric A Mattes
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska, Lincoln, NE, USA
| | - Megan Hiatt
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska, Lincoln, NE, USA
| | - Naja J Jann
- Division of Infection Biology, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Yefei Zhu
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska, Lincoln, NE, USA
| | - Nagender Ledala
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska, Lincoln, NE, USA
| | - Regine Landmann
- Division of Infection Biology, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Mathias Herrmann
- Institute of Medical Microbiology and Hygiene, University of Saarland Hospital, Homburg/Saar, Germany
| | - Holger Rohde
- Institute for Medical Microbiology, Virology and Hygiene, University Clinic Hamburg-Eppendorf, Hamburg, Germany
| | - Markus Bischoff
- Institute of Medical Microbiology and Hygiene, University of Saarland Hospital, Homburg/Saar, Germany
| | - Greg A Somerville
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska, Lincoln, NE, USA
| |
Collapse
|
140
|
Cerca F, França Â, Guimarães R, Hinzmann M, Cerca N, Lobo da Cunha A, Azeredo J, Vilanova M. Modulation of poly-N-acetylglucosamine accumulation within mature Staphylococcus epidermidis biofilms grown in excess glucose. Microbiol Immunol 2011; 55:673-82. [DOI: 10.1111/j.1348-0421.2011.00368.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
141
|
Cerca F, Trigo G, Correia A, Cerca N, Azeredo J, Vilanova M. SYBR green as a fluorescent probe to evaluate the biofilm physiological state of Staphylococcus epidermidis, using flow cytometry. Can J Microbiol 2011; 57:850-6. [PMID: 21950962 DOI: 10.1139/w11-078] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Staphylococcus epidermidis biofilms with different proportions of viable but nonculturable bacteria were used to show that SYBR green (SYBR) may be used as a probe to evaluate the bacterial physiological state using flow cytometry. Biofilms grown in excess glucose presented significantly higher proportions of dormant bacteria than biofilms grown in excess glucose with buffered pH conditions or with exponential-phase planktonic cultures. Bacteria obtained from biofilms with high or low proportions of viable but nonculturable cells were further cultured in broth medium and stained with SYBR at different time points. An association between bacterial growth and SYBR staining intensity was observed. In addition, bacteria presenting higher SYBR fluorescence intensity also stained more intensely with cyanoditolyl tetrazolium chloride, used as a probe to evaluate cellular metabolism. Accordingly, planktonic bacteria treated with rifampicin, an inhibitor of bacterial RNA transcription, presented lower SYBR and cyanoditolyl tetrazolium chloride staining intensity than nontreated bacteria. Overall, our results indicate that SYBR, in addition to being used as a component of LIVE/DEAD stain, may also be used as a probe to evaluate the physiological state of S. epidermidis cells.
Collapse
Affiliation(s)
- Filipe Cerca
- Laboratório de Imunologia, Departamento de Imunofisiologia e Farmacologia, Instituto de Ciências Biomédicas de Abel Salazar - Universidade do Porto, Portugal
| | | | | | | | | | | |
Collapse
|
142
|
Liu Q, Fan J, Niu C, Wang D, Wang J, Wang X, Villaruz AE, Li M, Otto M, Gao Q. The eukaryotic-type serine/threonine protein kinase Stk is required for biofilm formation and virulence in Staphylococcus epidermidis. PLoS One 2011; 6:e25380. [PMID: 21966513 PMCID: PMC3179523 DOI: 10.1371/journal.pone.0025380] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Accepted: 09/02/2011] [Indexed: 01/15/2023] Open
Abstract
Background Serine/threonine kinases are involved in gene regulation and signal transduction in prokaryotes and eukaryotes. Here, we investigated the role of the serine/threonine kinase Stk in the opportunistic pathogen Staphylococcus epidermidis. Methodology/Principal Findings We constructed an isogenic stk mutant of a biofilm-forming clinical S. epidermidis isolate. Presence of stk was important for biofilm formation in vitro and virulence in a murine subcutaneous foreign body infection model. Furthermore, the stk mutant exhibited phenotypes indicating an impact of stk on metabolic pathways. Using different constructs for the genetic complementation of the stk mutant strain with full-length Stk or specific Stk domains, we determined that the Stk intracellular kinase domain is important for biofilm formation and regulation of purine metabolism. Site-specific inactivation of the Stk kinase domain led to defective biofilm formation, in further support of the notion that the kinase activity of Stk regulates biofilm formation of S. epidermidis. According to immunological detection of the biofilm exopolysaccharide PIA and real-time PCR of the PIA biosynthesis genes, the impact of stk on biofilm formation is mediated, at least in part, by a strong influence on PIA expression. Conclusions Our study identifies Stk as an important regulator of biofilm formation and virulence of S. epidermidis, with additional involvement in purine metabolism and the bacterial stress response.
Collapse
Affiliation(s)
- Qian Liu
- Key Laboratory of Medical Molecular Virology, Institutes of Biomedical Sciences, and Institute of Medical Microbiology, Fudan University, Shanghai, People's Republic of China
| | - Jiajia Fan
- Key Laboratory of Medical Molecular Virology, Institutes of Biomedical Sciences, and Institute of Medical Microbiology, Fudan University, Shanghai, People's Republic of China
| | - Chen Niu
- Key Laboratory of Medical Molecular Virology, Institutes of Biomedical Sciences, and Institute of Medical Microbiology, Fudan University, Shanghai, People's Republic of China
| | - Decheng Wang
- Key Laboratory of Medical Molecular Virology, Institutes of Biomedical Sciences, and Institute of Medical Microbiology, Fudan University, Shanghai, People's Republic of China
| | - Jianping Wang
- Key Laboratory of Medical Molecular Virology, Institutes of Biomedical Sciences, and Institute of Medical Microbiology, Fudan University, Shanghai, People's Republic of China
| | - Xing Wang
- Key Laboratory of Medical Molecular Virology, Institutes of Biomedical Sciences, and Institute of Medical Microbiology, Fudan University, Shanghai, People's Republic of China
| | - Amer E. Villaruz
- National Institute of Allergy and Infectious Diseases, The National Institutes of Health, Bethesda, Maryland, United States of America
| | - Min Li
- Department of Laboratory Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Michael Otto
- National Institute of Allergy and Infectious Diseases, The National Institutes of Health, Bethesda, Maryland, United States of America
| | - Qian Gao
- Key Laboratory of Medical Molecular Virology, Institutes of Biomedical Sciences, and Institute of Medical Microbiology, Fudan University, Shanghai, People's Republic of China
- * E-mail:
| |
Collapse
|
143
|
Zhang B, Halouska S, Schiaffo CE, Sadykov MR, Somerville GA, Powers R. NMR analysis of a stress response metabolic signaling network. J Proteome Res 2011; 10:3743-54. [PMID: 21692534 PMCID: PMC3153081 DOI: 10.1021/pr200360w] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We previously hypothesized that Staphylococcus epidermidis senses a diverse set of environmental and nutritional factors associated with biofilm formation through a modulation in the activity of the tricarboxylic acid (TCA) cycle. Herein, we report our further investigation of the impact of additional environmental stress factors on TCA cycle activity and provide a detailed description of our NMR methodology. S. epidermidis wild-type strain 1457 was treated with stressors that are associated with biofilm formation, a sublethal dose of tetracycline, 5% NaCl, 2% glucose, and autoinducer-2 (AI-2). As controls and to integrate our current data with our previous study, 4% ethanol stress and iron-limitation were also used. Consistent with our prior observations, the effect of many environmental stress factors on the S. epidermidis metabolome was essentially identical to the effect of TCA cycle inactivation in the aconitase mutant strain 1457-acnA::tetM. A detailed quantitative analysis of metabolite concentration changes using 2D (1)H-(13)C HSQC and (1)H-(1)H TOCSY spectra identified a network of 37 metabolites uniformly affected by the stressors and TCA cycle inactivation. We postulate that the TCA cycle acts as the central pathway in a metabolic signaling network.
Collapse
Affiliation(s)
- Bo Zhang
- Department of Chemistry, University of Nebraska-Lincoln Lincoln, NE 68588-0304
| | - Steven Halouska
- Department of Chemistry, University of Nebraska-Lincoln Lincoln, NE 68588-0304
| | - Charles E. Schiaffo
- Department of Chemistry, University of Nebraska-Lincoln Lincoln, NE 68588-0304
| | - Marat R. Sadykov
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln Lincoln, NE 68588-0905
| | - Greg A. Somerville
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln Lincoln, NE 68588-0905
| | - Robert Powers
- Department of Chemistry, University of Nebraska-Lincoln Lincoln, NE 68588-0304
| |
Collapse
|
144
|
Monoclonal antibodies against accumulation-associated protein affect EPS biosynthesis and enhance bacterial accumulation of Staphylococcus epidermidis. PLoS One 2011; 6:e20918. [PMID: 21687690 PMCID: PMC3110253 DOI: 10.1371/journal.pone.0020918] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Accepted: 05/15/2011] [Indexed: 11/24/2022] Open
Abstract
Because there is no effective antibiotic to eradicate Staphylococcus epidermidis biofilm infections that lead to the failure of medical device implantations, the development of anti-biofilm vaccines is necessary. Biofilm formation by S. epidermidis requires accumulation-associated protein (Aap) that contains sequence repeats known as G5 domains, which are responsible for the Zn2+-dependent dimerization of Aap to mediate intercellular adhesion. Antibodies against Aap have been reported to inhibit biofilm accumulation. In the present study, three monoclonal antibodies (MAbs) against the Aap C-terminal single B-repeat construct followed by the 79-aa half repeat (AapBrpt1.5) were generated. MAb18B6 inhibited biofilm formation by S. epidermidis RP62A to 60% of the maximum, while MAb25C11 and MAb20B9 enhanced biofilm accumulation. All three MAbs aggregated the planktonic bacteria to form visible cell clusters. Epitope mapping revealed that the epitope of MAb18B6, which recognizes an identical area within AapBrpt constructs from S. epidermidis RP62A, was not shared by MAb25C11 and MAb20B9. Furthermore, all three MAbs were found to affect both Aap expression and extracellular polymeric substance (EPS, including extracellular DNA and PIA) biosynthesis in S. epidermidis and enhance the cell accumulation. These findings contribute to a better understanding of staphylococcal biofilm formation and will help to develop epitope-peptide vaccines against staphylococcal infections.
Collapse
|
145
|
Katzenmeyer KN, Bryers JD. Multivalent artificial opsonin for the recognition and phagocytosis of Gram-positive bacteria by human phagocytes. Biomaterials 2011; 32:4042-51. [PMID: 21388677 PMCID: PMC3069663 DOI: 10.1016/j.biomaterials.2011.02.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2011] [Accepted: 02/04/2011] [Indexed: 12/30/2022]
Abstract
Hospital-acquired infections (HAIs) remain a leading cause of death in the United States. Unfortunately, treatment of HAIs is complicated by the emergence of antibiotic-resistant bacterial strains. In an effort to enhance the body's natural immune response to infection, we have developed an artificial opsonin to promote the recognition, phagocytosis, and destruction of pathogenic bacteria by human phagocytes. The artificial opsonin is constructed from multivalent conjugates of poly(L-lysine)-graft-poly(ethylene glycol) with vancomycin and human IgG-Fc. Our approach utilizes vancomycin's inherent ability to bind to D-Ala-D-Ala terminated peptides present in the cell wall of Gram-positive bacteria. Here, we show that conjugation of vancomycin to PLL-g-PEG prevents its action as an antibiotic and allows vancomycin to function solely as a recognition molecule. Human IgG-Fc antibody fragment serves as a phagocyte recognition molecule and is recognized by the Fcγ cell surface receptors expressed on professional human phagocytes. Using flow cytometry, we found that a polysaccharide-encapsulated, methicillin-resistant strain of Staphylococcus epidermidis is efficiently recognized by the artificial opsonin (nearly 100% of cells were opsonized) and that opsonin binding is specific since it can be inhibited by the soluble cell wall peptide analog acetyl-Lys-D-Ala-D-Ala. Opsonization of S. epidermidis resulted in an approximate 2-fold increase in phagocytosis by a human neutrophil cell line. Notably, Enterococcus faecalis VanB, a bacterial strain with inducible vancomycin resistance, was used to show that the artificial opsonin does not unintentionally induce antibiotic resistance mechanisms.
Collapse
Affiliation(s)
- Kristy N. Katzenmeyer
- Department of Bioengineering, University of Washington, 3720 15 Ave NE, Seattle, WA 98195, USA
| | - James D. Bryers
- Department of Bioengineering, University of Washington, 3720 15 Ave NE, Seattle, WA 98195, USA
| |
Collapse
|
146
|
Kaplan JB, Jabbouri S, Sadovskaya I. Extracellular DNA-dependent biofilm formation by Staphylococcus epidermidis RP62A in response to subminimal inhibitory concentrations of antibiotics. Res Microbiol 2011; 162:535-41. [PMID: 21402153 PMCID: PMC3109171 DOI: 10.1016/j.resmic.2011.03.008] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Accepted: 02/25/2011] [Indexed: 10/18/2022]
Abstract
We measured the ability of Staphylococcus epidermidis to form biofilms in the presence of subminimal inhibitory concentrations (sub-MICs) of vancomycin, tigecycline, linezolid and novobiocin. Six strains that produce different amounts of biofilm were tested. The three strains that produced the highest amounts of biofilm exhibited steady-state or decreased biofilm formation in the presence of sub-MIC antibiotics, whereas the three strains that produced lower amounts of biofilm exhibited up to 10-fold-increased biofilm formation in the presence of sub-MIC antibiotics. In two of the inducible strains (9142 and 456a), antibiotic-induced biofilm formation was inhibited by dispersin B, an enzyme that degrades poly-N-acetylglucosamine (PNAG) biofilm polysaccharide. In the third inducible strain (RP62A), dispersin B inhibited biofilm formation in response to sub-MIC vancomycin, but not to sub-MIC tigecycline. In contrast, DNase I efficiently inhibited biofilm formation by strain RP62A in response to sub-MIC tigecycline and vancomycin. DNase I had no effect on antibiotic-induced biofilm formation in strains 9142 and 456a. Our findings indicate that antibiotic-induced biofilm formation in S. epidermidis is both strain- and antibiotic-dependent and that S. epidermidis RP62A utilizes an extracellular DNA-dependent mechanism to form biofilms in response to sub-MIC antibiotics.
Collapse
Affiliation(s)
| | - Saïd Jabbouri
- Institut de Recherche pour le Developpement, 46, Street 7, Maadi 11431 Cairo, Egypt
| | - Irina Sadovskaya
- Université Lille Nord de France, Université du Littoral-Côte d’Opale, UMT 08, 62327 Boulogne sur mer, France
| |
Collapse
|
147
|
Disinfection of meticillin-resistant Staphylococcus aureus and Staphylococcus epidermidis biofilms using a remote non-thermal gas plasma. J Hosp Infect 2011; 78:204-7. [PMID: 21601949 DOI: 10.1016/j.jhin.2011.03.019] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2010] [Accepted: 03/11/2011] [Indexed: 01/31/2023]
Abstract
The effective disinfection of hospital surfaces is recognised as an important factor in preventing hospital-acquired infections. The purpose of this study was to quantify the disinfection rate of a novel gas plasma system on clinically relevant biofilms. Clinical isolates of Staphylococcus epidermidis and meticillin-resistant Staphylococcus aureus (MRSA) were grown as biofilms on glass surfaces and tested in a disinfection container remote from the plasma source. The strains used in this study were known to produce substantial quantities of biofilm and average log₁₀ counts were 9.0 and 9.1 cfu/cm(2) for S. epidermidis and MRSA respectively. Counts were reduced by between 4 and 4.5 log₁₀ after 1h of exposure for MRSA and S. epidermidis respectively. More prolonged treatment in the case of MRSA biofilms resulted in a 5.5 log₁₀ reduction after 90 min. Biofilm samples were also placed in medical device packaging bags and similar rates of disinfection were observed.
Collapse
|
148
|
Screening of Escherichia coli species biodiversity reveals new biofilm-associated antiadhesion polysaccharides. mBio 2011; 2:e00043-11. [PMID: 21558434 PMCID: PMC3101779 DOI: 10.1128/mbio.00043-11] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Bacterial biofilms often form multispecies communities in which complex but ill-understood competition and cooperation interactions occur. In light of the profound physiological modifications associated with this lifestyle, we hypothesized that the biofilm environment might represent an untapped source of natural bioactive molecules interfering with bacterial adhesion or biofilm formation. We produced cell-free solutions extracted from in vitro mature biofilms formed by 122 natural Escherichia coli isolates, and we screened these biofilm extracts for antiadhesion molecules active on a panel of Gram-positive and Gram-negative bacteria. Using this approach, we showed that 20% of the tested biofilm extracts contained molecules that antagonize bacterial growth or adhesion. We characterized a compound, produced by a commensal animal E. coli strain, for which activity is detected only in biofilm extract. Biochemical and genetic analyses showed that this compound corresponds to a new type of released high-molecular-weight polysaccharide whose biofilm-associated production is regulated by the RfaH protein. We demonstrated that the antiadhesion activity of this polysaccharide was restricted to Gram-positive bacteria and that its production reduced susceptibility to invasion and provided rapid exclusion of Staphylococcus aureus from mixed E. coli and S. aureus biofilms. Our results therefore demonstrate that biofilms contain molecules that contribute to the dynamics of mixed bacterial communities and that are not or only poorly detected in unconcentrated planktonic supernatants. Systematic identification of these compounds could lead to strategies that limit pathogen surface colonization and reduce the burden associated with the development of bacterial biofilms on medical devices. We sought to demonstrate that bacterial biofilms are reservoirs for unknown molecules that antagonize bacterial adhesion. The use of natural strains representative of Escherichia coli species biodiversity showed that nonbiocidal antiadhesion polysaccharides are frequently found in mature biofilm extracts (bacterium-free suspensions which contain soluble molecules produced within the biofilm). Release of an antiadhesion polysaccharide confers a competitive advantage upon the producing strain against clinically relevant pathogens such as Staphylococcus aureus. Hence, exploring the biofilm environment provides a better understanding of bacterial interactions within complex communities and could lead to improved control of pathogen colonization.
Collapse
|
149
|
Staphylococcus epidermidis uses distinct mechanisms of biofilm formation to interfere with phagocytosis and activation of mouse macrophage-like cells 774A.1. Infect Immun 2011; 79:2267-76. [PMID: 21402760 DOI: 10.1128/iai.01142-10] [Citation(s) in RCA: 127] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Assembly of adherent biofilms is the key mechanism involved in Staphylococcus epidermidis virulence during device-associated infections. Aside from polysaccharide intercellular adhesin (PIA), the accumulation-associated protein Aap and the extracellular matrix binding protein Embp act as intercellular adhesins, mediating S. epidermidis cell aggregation and biofilm accumulation. The aim of this study was to investigate structural features of PIA-, Aap-, and Embp-mediated S. epidermidis biofilms in more detail and to evaluate their specific contributions to biofilm-related S. epidermidis immune escape. PIA-, Embp-, and Aap-mediated biofilms exhibited substantial morphological differences. Basically, PIA synthesis induced formation of macroscopically visible, rough cell clusters, whereas Aap- and Embp-dependent biofilms preferentially displayed a smooth layer of aggregated bacteria. On the microscopic level, PIA was found to form a string-like organized extracellular matrix connecting the bacteria, while Embp produced small deposits of intercellular matrix and Aap was strictly localized to the bacterial surface. Despite marked differences, S. epidermidis strains using PIA, Aap, or Embp for biofilm formation were protected from uptake by J774A.1 macrophages, with similarly efficiencies. In addition, compared to biofilm-negative S. epidermidis strains, isogenic biofilm-forming S. epidermidis induced only a diminished inflammatory J774A.1 macrophage response, leading to significantly (88.2 to 88.7%) reduced NF-κB activation and 68.8 to 83% reduced interleukin-1β (IL-1β) production. Mechanical biofilm dispersal partially restored induction of NF-κB activation, although bacterial cell surfaces remained decorated with the respective intercellular adhesins. Our results demonstrate that distinct S. epidermidis biofilm morphotypes are similarly effective at protecting S. epidermidis from phagocytic uptake and at counteracting macrophage activation, providing novel insights into mechanisms that could contribute to the chronic and persistent course of biofilm-related S. epidermidis foreign material infections.
Collapse
|
150
|
Rawlinson LAB, O'Gara JP, Jones DS, Brayden DJ. Resistance of Staphylococcus aureus to the cationic antimicrobial agent poly(2-(dimethylamino ethyl)methacrylate) (pDMAEMA) is influenced by cell-surface charge and hydrophobicity. J Med Microbiol 2011; 60:968-976. [PMID: 21393458 DOI: 10.1099/jmm.0.025619-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Cationic antimicrobial agents may prevent device-associated infections caused by Staphylococcus epidermidis and Staphylococcus aureus. This study reports that the cationic antimicrobial polymer poly(2-(dimethylamino ethyl)methacrylate) (pDMAEMA) was more effective at antagonizing growth of clinical isolates of S. epidermidis than of S. aureus. Importantly, mature S. epidermidis biofilms were significantly inactivated by pDMAEMA. The S. aureus isolates tested were generally more hydrophobic than the S. epidermidis isolates and had a less negative charge, although a number of individual S. aureus and S. epidermidis clinical isolates had similar surface hydrophobicity and charge values. Fluorescence spectroscopy and flow cytometry revealed that fluorescently labelled pDMAEMA interacted strongly with S. epidermidis compared with S. aureus. S. aureus ΔdltA and ΔmprF mutants were less hydrophobic and therefore more susceptible to pDMAEMA than wild-type S. aureus. Although the different susceptibility of S. epidermidis and S. aureus isolates to pDMAEMA is complex, influenced in part by surface hydrophobicity and charge, these findings nevertheless reveal the potential of pDMAEMA to treat S. epidermidis infections.
Collapse
Affiliation(s)
- Lee-Anne B Rawlinson
- Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland.,School of Agriculture, University College Dublin, Belfield, Dublin 4, Ireland
| | - James P O'Gara
- School of Biomolecular and Biomedical Science, University College Dublin, Belfield, Dublin 4, Ireland.,Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - David S Jones
- School of Pharmacy, Queen's University of Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - David J Brayden
- Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland.,School of Agriculture, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|