1
|
The Influence of Patterned Surface Features on the Accumulation of Bovine Synovial Fluid-Induced Aggregates of Staphylococcus aureus. Appl Environ Microbiol 2022; 88:e0121722. [PMID: 36286507 PMCID: PMC9680626 DOI: 10.1128/aem.01217-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Periprosthetic joint infections occurring after joint replacement are a major clinical problem requiring repeated surgeries and antibiotic interventions.
Staphylococcus aureus
is the most prominent bacterium causing most implant-related infections.
S. aureus
can form a biofilm, which is defined as a group of attached bacteria with the formation of an envelope that is resistant to antibiotics. The attachment and retention of these bacteria on implant surfaces are not clearly understood.
Collapse
|
2
|
Woitschach F, Kloss M, Grabow N, Reisinger EC, Sombetzki M. Mimicking critical environment factors for a static in vitro biofilm formation model on blood-contact implant materials. CURRENT RESEARCH IN MICROBIAL SCIENCES 2022; 3:100156. [DOI: 10.1016/j.crmicr.2022.100156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 11/30/2022] Open
|
3
|
Jiang Z, Nero T, Mukherjee S, Olson R, Yan J. Searching for the Secret of Stickiness: How Biofilms Adhere to Surfaces. Front Microbiol 2021; 12:686793. [PMID: 34305846 PMCID: PMC8295476 DOI: 10.3389/fmicb.2021.686793] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/28/2021] [Indexed: 01/01/2023] Open
Abstract
Bacterial biofilms are communities of cells enclosed in an extracellular polymeric matrix in which cells adhere to each other and to foreign surfaces. The development of a biofilm is a dynamic process that involves multiple steps, including cell-surface attachment, matrix production, and population expansion. Increasing evidence indicates that biofilm adhesion is one of the main factors contributing to biofilm-associated infections in clinics and biofouling in industrial settings. This review focuses on describing biofilm adhesion strategies among different bacteria, including Vibrio cholerae, Pseudomonas aeruginosa, and Staphylococcus aureus. Techniques used to characterize biofilm adhesion are also reviewed. An understanding of biofilm adhesion strategies can guide the development of novel approaches to inhibit or manipulate biofilm adhesion and growth.
Collapse
Affiliation(s)
- Zhaowei Jiang
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, United States
| | - Thomas Nero
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, United States
| | - Sampriti Mukherjee
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL, United States
| | - Rich Olson
- Department of Molecular Biology and Biochemistry, Molecular Biophysics Program, Wesleyan University, Middletown, CT, United States
| | - Jing Yan
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, United States.,Quantitative Biology Institute, Yale University, New Haven, CT, United States
| |
Collapse
|
4
|
Niek WK, Teh CSJ, Idris N, Thong KL, Ngoi ST, Ponnampalavanar SSLS. Investigation of biofilm formation in methicillin-resistant Staphylococcus aureus associated with bacteraemia in a tertiary hospital. Folia Microbiol (Praha) 2021; 66:741-749. [PMID: 34089493 DOI: 10.1007/s12223-021-00877-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 05/24/2021] [Indexed: 11/27/2022]
Abstract
Biofilm formation is an important physiological process in Staphylococcus aureus (S. aureus) that can cause infections in humans. In this study, the ability of 36 methicillin-resistant S. aureus (MRSA) clinical isolates to form biofilm was studied based on genotypic and phenotypic approaches. These isolates were genotyped based on the microbial surface components recognizing adhesive matrix molecules (MSCRAMMs) and biofilm-associated genes (icaAD) via polymerase chain reactions. Phenotyping was performed based on the determination of the strength of biofilm formation of MRSA isolates in vitro. The most prevalent MSCRAMMs and biofilm-associated genes were clfA, eno, and icaD, followed by clfB. The fnbB (38.9%) and ebpS (11.1%) occurred less frequently among the MRSA isolates, while bbp and fnbA genes were absent from all isolates. The MRSA isolates were mostly moderate to strong biofilm formers, despite the heterogeneity of the MSCRAMM profiles. MRSA isolates from different infection sources (primary, catheter-related bloodstream, or secondary infections) were capable of forming strong biofilms. However, persistent bacteraemia was observed only in 19.4% of the MRSA-infected individuals. This study suggested that persistent MRSA bacteraemia in patients might not be associated with the biofilm-forming ability of the isolates.
Collapse
Affiliation(s)
- Wen Kiong Niek
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Cindy Shuan Ju Teh
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Nuryana Idris
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Kwai Lin Thong
- Nanotechnology & Catalysis Research Centre (NANOCAT), Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Soo Tein Ngoi
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | | |
Collapse
|
5
|
Yanagisawa N, Li DQ, Ljungh Å. Protein Adsorption onEX VIVOCatheters and Polymers Exposed to Peritoneal Dialysis Effluent. Perit Dial Int 2020. [DOI: 10.1177/089686080402400309] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
BackgroundDeposition of proteins on surfaces of medical devices has been recognized to putatively relate to the process of regulation of biomaterial-associated complications by attachment of fibrin clots, eukaryotic cells, and microbes. The molecules adsorb to a varying extent, depending not only on the physicochemical properties of the biomaterial, but also on the composition of the host fluid.ObjectiveAdsorption of proteins on catheters exposed both ex vivo and in vitro to dialysate of patients on peritoneal dialysis (PD) was studied.MethodsPeritoneal dialysis effluent was collected from 5 patients with end-stage renal disease on continuous ambulatory PD. Tenckhoff catheters were obtained from 16 patients. Deposition of proteins on excised Tenckhoff catheters and tubing of different materials exposed to PD effluent in vitro was studied using125iodine-labeled antibodies. Adhesion of Staphylococcus aureus and Staphylococcus epidermidis strains was quantified on tubing exposed to PD effluent in vitro.ResultsThe presence of albumin, transferrin, immunoglobulin G, fibrinogen, fibronectin, von Willebrand factor, vitronectin, and thrombospondin was determined at various concentrations in PD effluent. All proteins analyzed were detected on PD catheters removed from patients. The extent of protein deposition on Tenckhoff catheters exposed to PD effluent, in vitro, rapidly reached a plateau and remained constant, as it did on polyvinyl chloride and polyethylene tubing. Adhesion of staphylococci was enhanced on Tenckhoff catheters exposed to PD effluent compared to unused PD solution.ConclusionsThe data identify surface exposed proteins that may serve as adhesion sites for microbes on peritoneal catheters indwelled in patients undergoing PD.
Collapse
Affiliation(s)
- Naoko Yanagisawa
- Medical Microbiology, Dermatology and Infection, Lund University, Lund, Sweden
| | - Dai-Qing Li
- Medical Microbiology, Dermatology and Infection, Lund University, Lund, Sweden
| | - Åsa Ljungh
- Medical Microbiology, Dermatology and Infection, Lund University, Lund, Sweden
| |
Collapse
|
6
|
Mancy A, Abutaleb NS, Elsebaei MM, Saad AY, Kotb A, Ali AO, Abdel-Aleem JA, Mohammad H, Seleem MN, Mayhoub AS. Balancing Physicochemical Properties of Phenylthiazole Compounds with Antibacterial Potency by Modifying the Lipophilic Side Chain. ACS Infect Dis 2020; 6:80-90. [PMID: 31718144 DOI: 10.1021/acsinfecdis.9b00211] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Bacterial resistance to antibiotics is presently one of the most pressing healthcare challenges and necessitates the discovery of new antibacterials with unique chemical scaffolds. However, the determination of the optimal balance between structural requirements for pharmacological action and pharmacokinetic properties of novel antibacterial compounds is a significant challenge in drug development. The incorporation of lipophilic moieties within a compound's core structure can enhance biological activity but have a deleterious effect on drug-like properties. In this Article, the lipophilicity of alkynylphenylthiazoles, previously identified as novel antibacterial agents, was reduced by introducing cyclic amines to the lipophilic side chain. In this regard, substitution with methylpiperidine (compounds 14-16) and thiomorpholine (compound 19) substituents significantly enhanced the aqueous solubility profile of the new compounds more than 150-fold compared to the first-generation lead compound 1b. Consequently, the pharmacokinetic profile of compound 15 was significantly enhanced with a notable improvement in both half-life and the time the compound's plasma concentration remained above its minimum inhibitory concentration (MIC) against methicillin-resistant Staphylococcus aureus (MRSA). In addition, compounds 14-16 and 19 were found to exert a bactericidal mode of action against MRSA and were not susceptible to resistance formation after 14 serial passages. Moreover, these compounds (at 2× MIC) were superior to the antibiotic vancomycin in the disruption of the mature MRSA biofilm. The modifications to the alkynylphenylthiazoles reported herein successfully improved the pharmacokinetic profile of this new series while maintaining the compounds' biological activity against MRSA.
Collapse
Affiliation(s)
- Ahmed Mancy
- Department of Pharmaceutical Organic Chemistry, College of Pharmacy, Al-Azhar University, 1-Elmokhayem Eldaem Street, Cairo 11884, Egypt
| | - Nader S. Abutaleb
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, 725 Harrison Street, West Lafayette, Indiana 47907, United States
| | - Mohamed M. Elsebaei
- Department of Pharmaceutical Organic Chemistry, College of Pharmacy, Al-Azhar University, 1-Elmokhayem Eldaem Street, Cairo 11884, Egypt
| | - Abdullah Y. Saad
- Department of Pharmaceutical Organic Chemistry, College of Pharmacy, Al-Azhar University, 1-Elmokhayem Eldaem Street, Cairo 11884, Egypt
| | - Ahmed Kotb
- Department of Pharmaceutical Organic Chemistry, College of Pharmacy, Al-Azhar University, 1-Elmokhayem Eldaem Street, Cairo 11884, Egypt
| | - Alsagher O. Ali
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, 725 Harrison Street, West Lafayette, Indiana 47907, United States
- Division of Infectious Diseases, Animal Medicine Department, Faculty of Veterinary Medicine, South Valley University, Qena 83523, Egypt
| | - Jelan A. Abdel-Aleem
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, 725 Harrison Street, West Lafayette, Indiana 47907, United States
- Department of Industrial Pharmacy, Faculty of Pharmacy, Assiut University, Assiut 71515, Egypt
| | - Haroon Mohammad
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, 725 Harrison Street, West Lafayette, Indiana 47907, United States
| | - Mohamed N. Seleem
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, 725 Harrison Street, West Lafayette, Indiana 47907, United States
- Purdue Institute of Inflammation, Immunology, and Infectious Disease, West Lafayette, Indiana 47907, United States
| | - Abdelrahman S. Mayhoub
- Department of Pharmaceutical Organic Chemistry, College of Pharmacy, Al-Azhar University, 1-Elmokhayem Eldaem Street, Cairo 11884, Egypt
- University of Science and Technology, Nanoscience Program, Zewail City of Science and Technology, Ahmed Zewail Street, October Gardens, 6th of October, Giza 12578, Egypt
| |
Collapse
|
7
|
Valliammai A, Sethupathy S, Priya A, Selvaraj A, Bhaskar JP, Krishnan V, Pandian SK. 5-Dodecanolide interferes with biofilm formation and reduces the virulence of Methicillin-resistant Staphylococcus aureus (MRSA) through up regulation of agr system. Sci Rep 2019; 9:13744. [PMID: 31551455 PMCID: PMC6760239 DOI: 10.1038/s41598-019-50207-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 08/30/2019] [Indexed: 12/20/2022] Open
Abstract
Methicillin resistant Staphylococcus aureus (MRSA) is a predominant human pathogen with high morbidity that is listed in the WHO high priority pathogen list. Being a primary cause of persistent human infections, biofilm forming ability of S. aureus plays a pivotal role in the development of antibiotic resistance. Hence, targeting biofilm is an alternative strategy to fight bacterial infections. The present study for the first time demonstrates the non-antibacterial biofilm inhibitory efficacy of 5-Dodecanolide (DD) against ATCC strain and clinical isolates of S. aureus. In addition, DD is able to inhibit adherence of MRSA on human plasma coated Titanium surface. Further, treatment with DD significantly reduced the eDNA synthesis, autoaggregation, staphyloxanthin biosynthesis and ring biofilm formation. Reduction in staphyloxanthin in turn increased the susceptibility of MRSA to healthy human blood and H2O2 exposure. Quantitative PCR analysis revealed the induced expression of agrA and agrC upon DD treatment. This resulted down regulation of genes involved in biofilm formation such as fnbA and fnbB and up regulation of RNAIII, hld, psmα and genes involved in biofilm matrix degradation such as aur and nuc. Inefficacy of DD on the biofilm formation of agr mutant further validated the agr mediated antibiofilm potential of DD. Notably, DD was efficient in reducing the in vivo colonization of MRSA in Caenorhabditis elegans. Results of gene expression studies and physiological assays unveiled the agr mediated antibiofilm efficacy of DD.
Collapse
Affiliation(s)
- Alaguvel Valliammai
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, 630003, Tamil Nadu, India
| | - Sivasamy Sethupathy
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, 630003, Tamil Nadu, India
- School of Chemical Engineering, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Arumugam Priya
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, 630003, Tamil Nadu, India
| | - Anthonymuthu Selvaraj
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, 630003, Tamil Nadu, India
| | | | | | | |
Collapse
|
8
|
Elsebaei MM, Abutaleb NS, Mahgoub AA, Li D, Hagras M, Mohammad H, Seleem MN, Mayhoub AS. Phenylthiazoles with nitrogenous side chain: An approach to overcome molecular obesity. Eur J Med Chem 2019; 182:111593. [PMID: 31446245 DOI: 10.1016/j.ejmech.2019.111593] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 07/11/2019] [Accepted: 08/05/2019] [Indexed: 02/01/2023]
Abstract
A novel series of phenylthiazoles bearing cyclic amines at the phenyl-4 position was prepared with the objective of decreasing lipophilicity and improving the overall physicochemical properties and pharmacokinetic profile of the compounds. Briefly, the piperidine ring (compounds 10 and 12) provided the best ring size in terms of antibacterial activity when tested against 16 multidrug-resistant clinical isolates. Both compounds were superior to vancomycin in the ability to eliminate methicillin-resistant Staphylococcus aureus (MRSA), residing within infected macrophages and to disrupt mature MRSA biofilm. Additionally, compounds 10 and 12 exhibited a fast-bactericidal mode of action in vitro. Furthermore, the new derivatives were 160-times more soluble in water than the previous lead compound 1b. Consequently, compound 10 was orally bioavailable with a highly-acceptable pharmacokinetic profile in vivo that exhibited a half-life of 4 h and achieved a maximum plasma concentration that exceeded the minimum inhibitory concentration (MIC) values against all tested bacterial isolates.
Collapse
Affiliation(s)
- Mohamed M Elsebaei
- Department of Pharmaceutical Organic Chemistry, College of Pharmacy, Al-Azhar University, Cairo, 11884, Egypt
| | - Nader S Abutaleb
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, 47907, USA
| | - Abdulrahman A Mahgoub
- Department of Pharmaceutical Organic Chemistry, College of Pharmacy, Al-Azhar University, Cairo, 11884, Egypt
| | - Daoyi Li
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, 47907, USA
| | - Mohamed Hagras
- Department of Pharmaceutical Organic Chemistry, College of Pharmacy, Al-Azhar University, Cairo, 11884, Egypt
| | - Haroon Mohammad
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, 47907, USA
| | - Mohamed N Seleem
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, 47907, USA; Purdue Institute of Inflammation, Immunology, and Infectious Disease, West Lafayette, IN, 47907, USA.
| | - Abdelrahman S Mayhoub
- Department of Pharmaceutical Organic Chemistry, College of Pharmacy, Al-Azhar University, Cairo, 11884, Egypt; University of Science and Technology, Nanoscience Program, Zewail City of Science and Technology, October Gardens, 6th of October, Giza, 12578, Egypt.
| |
Collapse
|
9
|
Fernandes MM, Ivanova K, Francesko A, Mendoza E, Tzanov T. Immobilization of antimicrobial core-shell nanospheres onto silicone for prevention of Escherichia coli biofilm formation. Process Biochem 2017. [DOI: 10.1016/j.procbio.2016.09.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
10
|
Zapotoczna M, Boksmati N, Donohue S, Bahtiar B, Boland A, Somali HA, Cox A, Humphreys H, O'Gara JP, Brennan M, O'Neill E. Novel anti-staphylococcal and anti-biofilm properties of two anti-malarial compounds: MMV665953 {1-(3-chloro-4-fluorophenyl)-3-(3,4-dichlorophenyl)urea} and MMV665807 {5-chloro-2-hydroxy-N-[3-(trifluoromethyl)phenyl]benzamide}. J Med Microbiol 2017; 66:377-387. [PMID: 28327271 DOI: 10.1099/jmm.0.000446] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
PURPOSE The treatment of device-related infections is challenging and current anti-microbial compounds have poor anti-biofilm activity. We aimed to identify and characterize novel compounds effective in the eradication of Staphylococcus aureus biofilms. METHODOLOGY Two novel compounds, MMV665953 {1-(3-chloro-4-fluorophenyl)-3-(3,4-dichlorophenyl)urea} and MMV665807{5-chloro-2-hydroxy-N-[3-(trifluoromethyl)phenyl]benzamide}, effective in killing S. aureus biofilms, were identified by screening of the open access 'malaria box' chemical library. The minimum bactericidal concentrations, half-maximal inhibition concentration (IC50) values and minimal biofilm killing concentrations effective in the killing of biofilm were determined against meticillin-resistant S. aureus and meticillin-sensitive S. aureus. Fibrin-embedded biofilms were grown under in vivo-relevant conditions, and viability was measured using a resazurin-conversion assay and confocal microscopy. The potential for the development of resistance and cytotoxicity was also assessed. RESULTS MMV665953 and MMV665807 were bactericidal against S. aureus isolates. The IC50 against S. aureus biofilms was at 0.15-0.58 mg l-1 after 24 h treatment, whereas the concentration required to eradicate all tested biofilms was 4 mg l-1, making the compounds more bactericidal than conventional antibiotics. The cytotoxicity against human keratinocytes and primary endothelial cells was determined as IC50 7.47 and 0.18 mg l-1 for MMV665953, and as 1.895 and 0.076 mg l-1 for MMV665807. Neither compound was haemolytic nor caused platelet activation. MMV665953 and MMV665807 derivatives with reduced cytotoxicity exhibited a concomitant loss in anti-staphylococcal activity. CONCLUSION MMV665953 and MMV665807 are more bactericidal against S. aureus biofilms than currently used anti-staphylococcal antibiotics and represent a valuable structural basis for further investigation in the treatment of staphylococcal biofilm-related infections.
Collapse
Affiliation(s)
- Marta Zapotoczna
- Department of Clinical Microbiology, Education and Research Centre, Beaumont Hospital, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Nabila Boksmati
- Molecular and Cellular Therapeutics, Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Sinead Donohue
- Department of Clinical Microbiology, Education and Research Centre, Beaumont Hospital, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Baizurina Bahtiar
- Molecular and Cellular Therapeutics, Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Ahmad Boland
- Molecular and Cellular Therapeutics, Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Hamzah Al Somali
- Molecular and Cellular Therapeutics, Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Alysia Cox
- Molecular and Cellular Therapeutics, Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Hilary Humphreys
- Department of Clinical Microbiology, Education and Research Centre, Beaumont Hospital, Royal College of Surgeons in Ireland, Dublin, Ireland.,Department of Microbiology, Beaumont Hospital, Dublin, Ireland
| | - James P O'Gara
- Department of Microbiology, School of Natural Sciences, National University of Ireland, Galway, Ireland
| | - Marian Brennan
- Molecular and Cellular Therapeutics, Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Eoghan O'Neill
- Department of Microbiology, Connolly Hospital, Dublin, Ireland.,Department of Clinical Microbiology, Education and Research Centre, Beaumont Hospital, Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
11
|
Crosby HA, Kwiecinski J, Horswill AR. Staphylococcus aureus Aggregation and Coagulation Mechanisms, and Their Function in Host-Pathogen Interactions. ADVANCES IN APPLIED MICROBIOLOGY 2016; 96:1-41. [PMID: 27565579 DOI: 10.1016/bs.aambs.2016.07.018] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The human commensal bacterium Staphylococcus aureus can cause a wide range of infections ranging from skin and soft tissue infections to invasive diseases like septicemia, endocarditis, and pneumonia. Muticellular organization almost certainly contributes to S. aureus pathogenesis mechanisms. While there has been considerable focus on biofilm formation and its role in colonizing prosthetic joints and indwelling devices, less attention has been paid to nonsurface-attached group behavior like aggregation and clumping. S. aureus is unique in its ability to coagulate blood, and it also produces multiple fibrinogen-binding proteins that facilitate clumping. Formation of clumps, which are large, tightly packed groups of cells held together by fibrin(ogen), has been demonstrated to be important for S. aureus virulence and immune evasion. Clumps of cells are able to avoid detection by the host's immune system due to a fibrin(ogen) coat that acts as a shield, and the size of the clumps facilitates evasion of phagocytosis. In addition, clumping could be an important early step in establishing infections that involve tight clusters of cells embedded in host matrix proteins, such as soft tissue abscesses and endocarditis. In this review, we discuss clumping mechanisms and regulation, as well as what is known about how clumping contributes to immune evasion.
Collapse
Affiliation(s)
- H A Crosby
- University of Iowa, Iowa City, IA, United States
| | - J Kwiecinski
- University of Iowa, Iowa City, IA, United States
| | - A R Horswill
- University of Iowa, Iowa City, IA, United States
| |
Collapse
|
12
|
Abstract
During infection, fungi frequently transition to a biofilm lifestyle, proliferating as communities of surface-adherent aggregates of cells. Phenotypically, cells in a biofilm are distinct from free-floating cells. Their high tolerance of antifungals and ability to withstand host defenses are two characteristics that foster resilience. Biofilm infections are particularly difficult to eradicate, and most available antifungals have minimal activity. Therefore, the discovery of novel compounds and innovative strategies to treat fungal biofilms is of great interest. Although many fungi have been observed to form biofilms, the most well-studied is Candida albicans. Animal models have been developed to simulate common Candida device-associated infections, including those involving vascular catheters, dentures, urinary catheters, and subcutaneous implants. Models have also reproduced the most common mucosal biofilm infections: oropharyngeal and vaginal candidiasis. These models incorporate the anatomical site, immune components, and fluid dynamics of clinical niches and have been instrumental in the study of drug resistance and investigation of novel therapies. This chapter describes the significance of fungal biofilm infections, the animal models developed for biofilm study, and how these models have contributed to the development of new strategies for the eradication of fungal biofilm infections.
Collapse
|
13
|
Nett JE. The Host's Reply to Candida Biofilm. Pathogens 2016; 5:pathogens5010033. [PMID: 26999221 PMCID: PMC4810154 DOI: 10.3390/pathogens5010033] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 03/09/2016] [Accepted: 03/10/2016] [Indexed: 12/22/2022] Open
Abstract
Candida spp. are among the most common nosocomial fungal pathogens and are notorious for their propensity toward biofilm formation. When growing on a medical device or mucosal surface, these organisms reside as communities embedded in a protective matrix, resisting host defenses. The host responds to Candida biofilm by depositing a variety of proteins that become incorporated into the biofilm matrix. Compared to free-floating Candida, leukocytes are less effective against Candida within a biofilm. This review highlights recent advances describing the host's response to Candida biofilms using ex vivo and in vivo models of mucosal and device-associated biofilm infections.
Collapse
Affiliation(s)
- Jeniel E Nett
- University of Wisconsin-Madison, Departments of Medicine, Medical Microbiology and Immunology, 5203 Microbial Sciences Building, 1550 Linden Drive, Madison, WI 53706, USA.
| |
Collapse
|
14
|
Host contributions to construction of three device-associated Candida albicans biofilms. Infect Immun 2015; 83:4630-8. [PMID: 26371129 DOI: 10.1128/iai.00931-15] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 09/09/2015] [Indexed: 01/05/2023] Open
Abstract
Among the most fascinating virulence attributes of Candida is the ability to transition to a biofilm lifestyle. As a biofilm, Candida cells adhere to a surface, such as a vascular catheter, and become encased in an extracellular matrix. During this mode of growth, Candida resists the normal immune response, often causing devastating disease. Based on scanning electron microscopy images, we hypothesized that host cells and proteins become incorporated into clinical biofilms. As a means to gain an understanding of these host-biofilm interactions, we explored biofilm-associated host components by using microscopy and liquid chromatography-mass spectrometry. Here we characterize the host proteins associated with several in vivo rat Candida albicans biofilms, including those from vascular catheter, denture, and urinary catheter models as well as uninfected devices. A conserved group of 14 host proteins were found to be more abundant during infection at each of the niches. The host proteins were leukocyte and erythrocyte associated and included proteins involved in inflammation, such as C-reactive protein, myeloperoxidase, and alarmin S100-A9. A group of 59 proteins were associated with both infected and uninfected devices, and these included matricellular and inflammatory proteins. In addition, site-specific proteins were identified, such as amylase in association with the denture device. Cellular analysis revealed neutrophils as the predominant leukocytes associating with biofilms. These experiments demonstrate that host cells and proteins are key components of in vivo Candida biofilms, likely with one subset associating with the device and another being recruited by the proliferating biofilm.
Collapse
|
15
|
Tolker-Nielsen T. Pseudomonas aeruginosa biofilm infections: from molecular biofilm biology to new treatment possibilities. APMIS 2015:1-51. [PMID: 25399808 DOI: 10.1111/apm.12335] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Bacteria in natural, industrial and clinical settings predominantly live in biofilms, i.e., sessile structured microbial communities encased in self-produced extracellular matrix material. One of the most important characteristics of microbial biofilms is that the resident bacteria display a remarkable increased tolerance toward antimicrobial attack. Biofilms formed by opportunistic pathogenic bacteria are involved in devastating persistent medical device-associated infections, and chronic infections in individuals who are immune-compromised or otherwise impaired in the host defense. Because the use of conventional antimicrobial compounds in many cases cannot eradicate biofilms, there is an urgent need to develop alternative measures to combat biofilm infections. The present review is focussed on the important opportunistic pathogen and biofilm model organism Pseudomonas aeruginosa. Initially, biofilm infections where P. aeruginosa plays an important role are described. Subsequently, current insights into the molecular mechanisms involved in P. aeruginosa biofilm formation and the associated antimicrobial tolerance are reviewed. And finally, based on our knowledge about molecular biofilm biology, a number of therapeutic strategies for combat of P. aeruginosa biofilm infections are presented.
Collapse
Affiliation(s)
- Tim Tolker-Nielsen
- Costerton Biofilm Center, Department of International Health, Immunology and Microbiology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
16
|
Biofilm formation by clinical isolates and its relevance to clinical infections. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 830:1-28. [PMID: 25366218 DOI: 10.1007/978-3-319-11038-7_1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Reports of biofilms have increased exponentially in the scientific literature over the past two decades, yet the vast majority of these are basic science investigations with limited clinical relevance. Biofilm studies involving clinical isolates are most often surveys of isolate collections, but suffer from lack of standardization in methodologies for producing and assessing biofilms. In contrast, more informative clinical studies correlating biofilm formation to patient data have infrequently been reported. In this chapter, biofilm surveys of clinical isolates of aerobic and anaerobic bacteria, mycobacteria, and Candida are reviewed, as well as those pertaining to the unique situation of cystic fibrosis. In addition, the influence of host components on in vitro biofilm formation, as well as published studies documenting the clinical impact of biofilms in human infections, are presented.
Collapse
|
17
|
Migonney V. Bioactive Polymers and Surfaces: A Solution for Implant Devices. Biomaterials 2014. [DOI: 10.1002/9781119043553.ch5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
18
|
Dinjaski N, Suri S, Valle J, Lehman SM, Lasa I, Prieto MA, García AJ. Near-infrared fluorescence imaging as an alternative to bioluminescent bacteria to monitor biomaterial-associated infections. Acta Biomater 2014; 10:2935-44. [PMID: 24632360 DOI: 10.1016/j.actbio.2014.03.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2013] [Revised: 02/13/2014] [Accepted: 03/04/2014] [Indexed: 11/28/2022]
Abstract
Biomaterial-associated infection is one of the most common complications related to the implantation of any biomedical device. Several in vivo imaging platforms have emerged as powerful diagnostic tools to longitudinally monitor biomaterial-associated infections in small animal models. In this study, we directly compared two imaging approaches: bacteria engineered to produce luciferase to generate bioluminescence and reactive oxygen species (ROS) imaging of the inflammatory response associated with the infected implant. We performed longitudinal imaging of bioluminescence associated with bacteria strains expressing plasmid-integrated luciferase driven by different promoters or a strain with the luciferase gene integrated into the chromosome. These luminescent strains provided an adequate signal for acute (0-4 days) monitoring of the infection, but the bioluminescence signal decreased over time and leveled off at 7 days post-implantation. This loss in the bioluminescence signal was attributed to changes in the metabolic activity of the bacteria. In contrast, near-infrared fluorescence imaging of ROS associated with inflammation to the implant provided sensitive and dose-dependent signals of biomaterial-associated bacteria. ROS imaging exhibited higher sensitivity than the bioluminescence imaging and was independent of the bacteria strain. Near-infrared fluorescence imaging of inflammatory responses represents a powerful alternative to bioluminescence imaging for monitoring biomaterial-associated bacterial infections.
Collapse
Affiliation(s)
- Nina Dinjaski
- Centro de Investigaciones Biológicas, CSIC, Madrid 28040, Spain
| | - Shalu Suri
- Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332-0363, USA
| | - Jaione Valle
- Instituto de Agrobiotecnología, UPNA-CSIC-Gobierno de Navarra, 31006 Pamplona, Spain
| | - Susan M Lehman
- Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332-0363, USA
| | - Iñigo Lasa
- Instituto de Agrobiotecnología, UPNA-CSIC-Gobierno de Navarra, 31006 Pamplona, Spain
| | | | - Andrés J García
- Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332-0363, USA.
| |
Collapse
|
19
|
Kim MK, Drescher K, Pak OS, Bassler BL, Stone HA. Filaments in curved streamlines: Rapid formation of Staphylococcus aureus biofilm streamers. NEW JOURNAL OF PHYSICS 2014; 16:065024. [PMID: 25484614 PMCID: PMC4255984 DOI: 10.1088/1367-2630/16/6/065024] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Biofilms are surface-associated conglomerates of bacteria that are highly resistant to antibiotics. These bacterial communities can cause chronic infections in humans by colonizing, for example, medical implants, heart valves, or lungs. Staphylococcus aureus, a notorious human pathogen, causes some of the most common biofilm-related infections. Despite the clinical importance of S. aureus biofilms, it remains mostly unknown how physical effects, in particular flow, and surface structure influence biofilm dynamics. Here we use model microfluidic systems to investigate how environmental factors, such as surface geometry, surface chemistry, and fluid flow affect biofilm development in S. aureus. We discovered that S. aureus rapidly forms flow-induced, filamentous biofilm streamers, and furthermore if surfaces are coated with human blood plasma, streamers appear within minutes and clog the channels more rapidly than if the channels are uncoated. To understand how biofilm streamer filaments reorient in flows with curved streamlines to bridge the distances between corners, we developed a mathematical model based on resistive force theory of slender filaments. Understanding physical aspects of biofilm formation in S. aureus may lead to new approaches for interrupting biofilm formation of this pathogen.
Collapse
Affiliation(s)
| | - Knut Drescher
- Department of Mechanical and Aerospace Engineering, Princeton University, Princeton, NJ 08544
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| | - On Shun Pak
- Department of Mechanical and Aerospace Engineering, Princeton University, Princeton, NJ 08544
| | - Bonnie L. Bassler
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
- Howard Hughes Medical Institute, Chevy Chase, MD 20815
| | - Howard A. Stone
- Department of Mechanical and Aerospace Engineering, Princeton University, Princeton, NJ 08544
- Corresponding author:
| |
Collapse
|
20
|
Chung PY, Toh YS. Anti-biofilm agents: recent breakthrough against multi-drug resistantStaphylococcus aureus. Pathog Dis 2014; 70:231-9. [DOI: 10.1111/2049-632x.12141] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 01/12/2014] [Accepted: 01/13/2014] [Indexed: 01/22/2023] Open
Affiliation(s)
- Pooi Y. Chung
- Department of Pathology; School of Medicine; International Medical University; Kuala Lumpur Malaysia
| | - Yien S. Toh
- Biomedical Science Program; School of Medicine; International Medical University; Kuala Lumpur Malaysia
| |
Collapse
|
21
|
Jahed Z, Lin P, Seo BB, Verma MS, Gu FX, Tsui TY, Mofrad MRK. Responses of Staphylococcus aureus bacterial cells to nanocrystalline nickel nanostructures. Biomaterials 2014; 35:4249-54. [PMID: 24576805 DOI: 10.1016/j.biomaterials.2014.01.080] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Accepted: 01/31/2014] [Indexed: 12/19/2022]
Abstract
A broad range of human diseases are associated with bacterial infections, often initiated by specific adhesion of a bacterium to the target environment. Despite the significant role of bacterial adhesion in human infectious diseases, details and mechanisms of bacterial adhesion have remained elusive. Herein, we study the physical interactions between Staphylococcus aureus, a type of micro-organism relevant to infections associated with medical implants, and nanocrystalline (nc) nickel nanostructures with various columnar features, including solid core, hollow, x-shaped and c-shaped pillars. Scanning electron microscopy results show the tendency of these bacterial cells to attach to the nickel nanostructures. Moreover, unique single bacterium attachment characteristics were observed on nickel nanostructures with dimensions comparable to the size of a single bacterium.
Collapse
Affiliation(s)
- Zeinab Jahed
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California Berkeley, 208A Stanley Hall, Berkeley, CA 94720-1762, USA; Physical Biosciences Division, Lawrence Berkeley National Lab, Berkeley, CA 94720, USA
| | - Peter Lin
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON N2L 3G1, Canada
| | - Brandon B Seo
- Department of Mechanical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON N2L 3G1, Canada
| | - Mohit S Verma
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON N2L 3G1, Canada
| | - Frank X Gu
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON N2L 3G1, Canada
| | - Ting Y Tsui
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON N2L 3G1, Canada; Department of Mechanical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON N2L 3G1, Canada
| | - Mohammad R K Mofrad
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California Berkeley, 208A Stanley Hall, Berkeley, CA 94720-1762, USA; Physical Biosciences Division, Lawrence Berkeley National Lab, Berkeley, CA 94720, USA.
| |
Collapse
|
22
|
Rochford E, Poulsson A, Salavarrieta Varela J, Lezuo P, Richards R, Moriarty T. Bacterial adhesion to orthopaedic implant materials and a novel oxygen plasma modified PEEK surface. Colloids Surf B Biointerfaces 2014; 113:213-22. [DOI: 10.1016/j.colsurfb.2013.09.012] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Revised: 08/23/2013] [Accepted: 09/05/2013] [Indexed: 10/26/2022]
|
23
|
Abstract
Staphylococcus aureus is a known cause of chronic biofilm infections that can reside on medical implants or host tissue. Recent studies have demonstrated an important role for proteinaceous material in the biofilm structure. The S. aureus genome encodes many secreted proteases, and there is growing evidence that these enzymes have self-cleavage properties that alter biofilm integrity. However, the specific contribution of each protease and mechanism of biofilm modulation is not clear. To address this issue, we utilized a sigma factor B (ΔsigB) mutant where protease activity results in a biofilm-negative phenotype, thereby creating a condition where the protease(s) responsible for the phenotype could be identified. Using a plasma-coated microtiter assay, biofilm formation was restored to the ΔsigB mutant through the addition of the cysteine protease inhibitor E-64 or by using Staphostatin inhibitors that specifically target the extracellular cysteine proteases SspB and ScpA (called Staphopains). Through construction of gene deletion mutants, we determined that an sspB scpA double mutant restored ΔsigB biofilm formation, and this recovery could be replicated in plasma-coated flow cell biofilms. Staphopain levels were also found to be decreased under biofilm-forming conditions, possibly allowing biofilm establishment. The treatment of S. aureus biofilms with purified SspB or ScpA enzyme inhibited their formation, and ScpA was also able to disperse an established biofilm. The antibiofilm properties of ScpA were conserved across S. aureus strain lineages. These findings suggest an underappreciated role of the SspB and ScpA cysteine proteases in modulating S. aureus biofilm architecture.
Collapse
|
24
|
Abstract
Bacterial pathogens have specific virulence factors (e.g., toxins) that contribute significantly to the virulence and infectivity of microorganisms within the human hosts. Virulence factors are molecules expressed by pathogens that enable colonization, immunoevasion, and immunosuppression, obtaining nutrients from the host or gaining entry into host cells. They can cause pathogenesis by inhibiting or stimulating certain host functions. For example, in systemic Staphylococcus aureus infections, virulence factors such as toxic shock syndrome toxin 1 (TSST-1), staphylococcal enterotoxin A (SEA), and staphylococcal enterotoxin B (SEB) cause sepsis or toxic shock by uncontrolled stimulation of T lymphocytes and by triggering a cytokine storm. In vitro, these superantigens stimulate the proliferation of human peripheral blood mononuclear cells (PBMC) and the release of many cytokines. NVC-422 (N,N-dichloro-2,2-dimethyltaurine) is a broad-spectrum, fast-acting topical anti-infective agent against microbial pathogens, including antibiotic-resistant microbes. Using mass spectrometry, we demonstrate here that NVC-422 oxidizes methionine residues of TSST-1, SEA, SEB, and exfoliative toxin A (ETA). Exposure of virulence factors to 0.1% NVC-422 for 1 h prevented TSST-1-, SEA-, SEB-, and ETA-induced cell proliferation and cytokine release. Moreover, NVC-422 also delayed and reduced the protein A- and clumping factor-associated agglutination of S. aureus cultures. These results show that, in addition to its well-described direct microbicidal activity, NVC-422 can inactivate S. aureus virulence factors through rapid oxidation of methionines.
Collapse
|
25
|
Walker JN, Horswill AR. A coverslip-based technique for evaluating Staphylococcus aureus biofilm formation on human plasma. Front Cell Infect Microbiol 2012; 2:39. [PMID: 22919630 PMCID: PMC3417647 DOI: 10.3389/fcimb.2012.00039] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2012] [Accepted: 03/12/2012] [Indexed: 01/12/2023] Open
Abstract
The ability of the opportunistic pathogen, Staphylococcus aureus, to form biofilms is increasingly being viewed as an important contributor to chronic infections. In vitro methods for analyzing S. aureus biofilm formation have focused on bacterial attachment and accumulation on abiotic surfaces, such as in microtiter plate and flow cell assays. Microtiter plates provide a rapid measure of relative biomass levels, while flow cells have limited experimental throughput but are superior for confocal microscopy biofilm visualization. Although these assays have proven effective at identifying mechanisms involved in cell attachment and biofilm accumulation, the significance of these assays in vivo remains unclear. Studies have shown that when medical devices are implanted they are coated with host factors, such as matrix proteins, that facilitate S. aureus attachment and biofilm formation. To address the challenge of integrating existing biofilm assay features with a biotic surface, we have established an in vitro biofilm technique utilizing UV-sterilized coverslips coated with human plasma. The substratum more closely resembles the in vivo state and provides a platform for S. aureus to establish a robust biofilm. Importantly, these coverslips are amenable to confocal microscopy imaging to provide a visual reference of the biofilm growth stage, effectively merging the benefits of the microtiter and flow cell assays. We confirmed the approach using clinical S. aureus isolates and mutants with known biofilm phenotypes. Altogether, this new biofilm assay can be used to assess the function of S. aureus virulence factors associated with biofilm formation and for monitoring the efficacy of biofilm treatment modalities.
Collapse
Affiliation(s)
- Jennifer N Walker
- Department of Microbiology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | | |
Collapse
|
26
|
Abraham NM, Jefferson KK. Staphylococcus aureus clumping factor B mediates biofilm formation in the absence of calcium. MICROBIOLOGY-SGM 2012; 158:1504-1512. [PMID: 22442307 DOI: 10.1099/mic.0.057018-0] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Staphylococcus aureus is the leading cause of nosocomial infections and a major cause of community-acquired infections. Biofilm formation is a key virulence determinant in certain types of S. aureus infection, especially those involving inserted medical devices. We found in a previous study that the calcium chelators sodium citrate and EGTA inhibit biofilm formation in certain strains of S. aureus but actually augment biofilm formation in other strains. Even two closely related strains, Newman and 10833, exhibited strikingly different biofilm phenotypes in the presence of calcium chelators, in that biofilm formation was inhibited in Newman but augmented in 10833. We also found that the surface protein clumping factor B (ClfB) plays a role in this phenomenon. In this study, we confirm that ClfB is required for biofilm formation under calcium-depleted conditions. We investigated the post-translational regulation of ClfB-mediated biofilm formation and found evidence that both calcium and the protease aureolysin disrupt established ClfB-dependent biofilms. Finally, we investigated the genetic basis for the biofilm-negative phenotype in strain Newman versus the biofilm-positive phenotype in strain 10833 under calcium-depleted conditions and found that strain 10833 contains a deletion that results in a stop codon within the aureolysin gene (aur). When 10833 expressed Newman aur, surface-associated ClfB and the ability to form a biofilm in chelating conditions was lost. Thus, the positive effect of chelating agents on biofilm formation in certain strains can be explained by increased ClfB activity in the absence of calcium and the discrepancy in the response of strains 10833 and Newman can be explained by point mutations in aur. This study reveals a previously unknown, to our knowledge, role for ClfB in biofilm formation and underscores the potential for striking phenotypic variability resulting from minor differences in strain background.
Collapse
Affiliation(s)
- Nabil M Abraham
- Virginia Commonwealth University, Department of Microbiology and Immunology, PO Box 980678, Richmond, VA 23928, USA
| | - Kimberly K Jefferson
- Virginia Commonwealth University, Department of Microbiology and Immunology, PO Box 980678, Richmond, VA 23928, USA
| |
Collapse
|
27
|
Abstract
Bacteria of the genus Staphylococcus are a prominent cause of acute and chronic infections. The ability of the staphylococci to establish biofilms has been linked to the persistence of chronic infections, which has drawn considerable interest from researchers over the past decade. Biofilms can be defined as sessile communities of surface-attached cells encased in an extracellular matrix, and treatment of bacteria in this mode of growth is challenging due to the resistance of biofilm structures to both antimicrobials and host defenses. In this review of the literature, we introduce Staphylococcus aureus and Staphylococcus epidermidis biofilms and summarize current antibiotic treatment approaches for staphylococcal biofilm infections. We also review recent studies on alternative strategies for preventing biofilm formation and dispersing established biofilms, including matrix-degrading enzymes, small-molecule approaches, and manipulation of natural staphylococcal disassembly mechanisms. While research on staphylococcal biofilm development is still in its early stages, new discoveries in the field hold promise for improved therapies that target staphylococcal biofilm infections.
Collapse
Affiliation(s)
- Megan R Kiedrowski
- Department of Microbiology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, 52242, USA
| | | |
Collapse
|
28
|
Kiedrowski MR, Kavanaugh JS, Malone CL, Mootz JM, Voyich JM, Smeltzer MS, Bayles KW, Horswill AR. Nuclease modulates biofilm formation in community-associated methicillin-resistant Staphylococcus aureus. PLoS One 2011; 6:e26714. [PMID: 22096493 PMCID: PMC3214024 DOI: 10.1371/journal.pone.0026714] [Citation(s) in RCA: 197] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2011] [Accepted: 10/03/2011] [Indexed: 12/13/2022] Open
Abstract
Community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA) is an emerging contributor to biofilm-related infections. We recently reported that strains lacking sigma factor B (sigB) in the USA300 lineage of CA-MRSA are unable to develop a biofilm. Interestingly, when spent media from a USA300 sigB mutant was incubated with other S. aureus strains, biofilm formation was inhibited. Following fractionation and mass spectrometry analysis, the major anti-biofilm factor identified in the spent media was secreted thermonuclease (Nuc). Considering reports that extracellular DNA (eDNA) is an important component of the biofilm matrix, we investigated the regulation and role of Nuc in USA300. The expression of the nuc gene was increased in a sigB mutant, repressed by glucose supplementation, and was unaffected by the agr quorum-sensing system. A FRET assay for Nuc activity was developed and confirmed the regulatory results. A USA300 nuc mutant was constructed and displayed an enhanced biofilm-forming capacity, and the nuc mutant also accumulated more high molecular weight eDNA than the WT and regulatory mutant strains. Inactivation of nuc in the USA300 sigB mutant background partially repaired the sigB biofilm-negative phenotype, suggesting that nuc expression contributes to the inability of the mutant to form biofilm. To test the generality of the nuc mutant biofilm phenotypes, the mutation was introduced into other S. aureus genetic backgrounds and similar increases in biofilm formation were observed. Finally, using multiple S. aureus strains and regulatory mutants, an inverse correlation between Nuc activity and biofilm formation was demonstrated. Altogether, our findings confirm the important role for eDNA in the S. aureus biofilm matrix and indicates Nuc is a regulator of biofilm formation.
Collapse
Affiliation(s)
- Megan R. Kiedrowski
- Department of Microbiology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Jeffrey S. Kavanaugh
- Department of Microbiology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Cheryl L. Malone
- Department of Microbiology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Joe M. Mootz
- Department of Microbiology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Jovanka M. Voyich
- Department of Veterinary Microbiology, Montana State University, Bozeman, Montana, United States of America
| | - Mark S. Smeltzer
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Kenneth W. Bayles
- Department of Pathology, Nebraska Medical Center, University of Nebraska, Omaha, Nebraska, United States of America
| | - Alexander R. Horswill
- Department of Microbiology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
- * E-mail:
| |
Collapse
|
29
|
Rot and Agr system modulate fibrinogen-binding ability mainly by regulating clfB expression in Staphylococcus aureus NCTC8325. Med Microbiol Immunol 2011; 201:81-92. [DOI: 10.1007/s00430-011-0208-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Indexed: 01/19/2023]
|
30
|
Beenken KE, Mrak LN, Griffin LM, Zielinska AK, Shaw LN, Rice KC, Horswill AR, Bayles KW, Smeltzer MS. Epistatic relationships between sarA and agr in Staphylococcus aureus biofilm formation. PLoS One 2010; 5:e10790. [PMID: 20520723 PMCID: PMC2875390 DOI: 10.1371/journal.pone.0010790] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2010] [Accepted: 04/30/2010] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The accessory gene regulator (agr) and staphylococcal accessory regulator (sarA) play opposing roles in Staphylococcus aureus biofilm formation. There is mounting evidence to suggest that these opposing roles are therapeutically relevant in that mutation of agr results in increased biofilm formation and decreased antibiotic susceptibility while mutation of sarA has the opposite effect. To the extent that induction of agr or inhibition of sarA could potentially be used to limit biofilm formation, this makes it important to understand the epistatic relationships between these two loci. METHODOLOGY/PRINCIPAL FINDINGS We generated isogenic sarA and agr mutants in clinical isolates of S. aureus and assessed the relative impact on biofilm formation. Mutation of agr resulted in an increased capacity to form a biofilm in the 8325-4 laboratory strain RN6390 but had little impact in clinical isolates S. aureus. In contrast, mutation of sarA resulted in a reduced capacity to form a biofilm in all clinical isolates irrespective of the functional status of agr. This suggests that the regulatory role of sarA in biofilm formation is independent of the interaction between sarA and agr and that sarA is epistatic to agr in this context. This was confirmed by demonstrating that restoration of sarA function restored the ability to form a biofilm even in the corresponding agr mutants. Mutation of sarA in clinical isolates also resulted in increased production of extracellular proteases and extracellular nucleases, both of which contributed to the biofilm-deficient phenotype of sarA mutants. However, studies comparing different strains with and without proteases inhibitors and/or mutation of the nuclease genes demonstrated that the agr-independent, sarA-mediated repression of extracellular proteases plays a primary role in this regard. CONCLUSIONS AND SIGNIFICANCE The results we report suggest that inhibitors of sarA-mediated regulation could be used to limit biofilm formation in S. aureus and that the efficacy of such inhibitors would not be limited by spontaneous mutation of agr in the human host.
Collapse
Affiliation(s)
- Karen E. Beenken
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Lara N. Mrak
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Linda M. Griffin
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Agnieszka K. Zielinska
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Lindsey N. Shaw
- Department of Biology, University of South Florida, Tampa, Florida, United States of America
| | - Kelly C. Rice
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, United States of America
| | - Alexander R. Horswill
- Department of Microbiology, University of Iowa, Iowa City, Iowa, United States of America
| | - Kenneth W. Bayles
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Mark S. Smeltzer
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- * E-mail:
| |
Collapse
|
31
|
Edgeworth J. Intravascular catheter infections. J Hosp Infect 2009; 73:323-30. [DOI: 10.1016/j.jhin.2009.05.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2009] [Accepted: 05/14/2009] [Indexed: 11/26/2022]
|
32
|
Kalasin S, Santore MM. Non-specific adhesion on biomaterial surfaces driven by small amounts of protein adsorption. Colloids Surf B Biointerfaces 2009; 73:229-36. [DOI: 10.1016/j.colsurfb.2009.05.028] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2009] [Revised: 05/05/2009] [Accepted: 05/25/2009] [Indexed: 01/07/2023]
|
33
|
Li D, Renzoni A, Estoppey T, Bisognano C, Francois P, Kelley WL, Lew DP, Schrenzel J, Vaudaux P. Induction of fibronectin adhesins in quinolone-resistant Staphylococcus aureus by subinhibitory levels of ciprofloxacin or by sigma B transcription factor activity is mediated by two separate pathways. Antimicrob Agents Chemother 2005; 49:916-24. [PMID: 15728884 PMCID: PMC549254 DOI: 10.1128/aac.49.3.916-924.2005] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
We recently reported on the involvement of a RecA-LexA-dependent pathway in the ciprofloxacin-triggered upregulation of fibronectin-binding proteins (FnBPs) by fluoroquinolone-resistant Staphylococcus aureus. The potential additional contribution of the transcription factor sigma B (SigB) to the ciprofloxacin-triggered upregulation of FnBPs was studied in isogenic mutants of fluoroquinolone-resistant strain RA1 (a topoisomerase IV gyrase double mutant of S. aureus NCTC strain 8325), which exhibited widely different levels of SigB activity, as assessed by quantitative reverse transcription-PCR of their respective sigB and SigB-dependent asp23 transcript levels. These mutants were Tn551 insertion sigB strain TE1 and rsbU(+) complemented strain TE2, which exhibited a wild-type SigB operon. Levels of FnBP surface display and fibronectin-mediated adhesion were lower in sigB mutant TE1 or higher in the rsbU(+)-restored strain TE2 compared to their sigB(+) but rsbU parent, strain RA1, exhibiting low levels of SigB activity. Steady-state fnbA and fnbB transcripts levels were similar in strains TE1 and RA1 but increased by 4- and 12-fold, respectively, in strain TE2 compared to those in strain RA1. In contrast, fibronectin-mediated adhesion of strains TE1, RA1, and TE2 was similarly enhanced by growth in the presence of one-eighth the MIC of ciprofloxacin, which led to a significantly higher increase in their fnbB transcript levels compared to the increase in their fnbA transcript levels. Increased SigB levels led to a significant reduction in agr RNAIII; in contrast, it led to a slight increase in sarA transcript levels. In conclusion, upregulation of FnBPs by increased SigB levels and ciprofloxacin exposure in fluoroquinolone-resistant S. aureus occurs via independent pathways whose concerted actions may significantly promote bacterial adhesion and colonization.
Collapse
Affiliation(s)
- Dongmei Li
- Service of Infectious Diseases, University Hospitals of Geneva, Geneva, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Grundmeier M, Hussain M, Becker P, Heilmann C, Peters G, Sinha B. Truncation of fibronectin-binding proteins in Staphylococcus aureus strain Newman leads to deficient adherence and host cell invasion due to loss of the cell wall anchor function. Infect Immun 2004; 72:7155-63. [PMID: 15557640 PMCID: PMC529102 DOI: 10.1128/iai.72.12.7155-7163.2004] [Citation(s) in RCA: 124] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Staphylococcus aureus fibronectin-binding proteins (FnBPs) play a critical role in S. aureus pathogenesis. FnBPs mediate adhesion to fibronectin and invasion of mammalian cells, including epithelial, endothelial, and fibroblastic cells, by fibronectin bridging to the host cell fibronectin receptor integrin (alpha(5))beta(1). Strain Newman is a laboratory strain frequently used for genetic, functional, and in vivo studies. However, despite pronounced production of FnBPs, strain Newman is only weakly adherent to immobilized Fn and weakly invasive. We examined whether these effects are due to a structural difference of FnBPs. Here, we show that both fnbA(Newman) and fnbB(Newman) contain a centrally located point mutation resulting in a stop codon. This leads to a truncation of both FnBPs at the end of the C domain at identical positions. Most likely, the stop codon occurred first in fnbB(Newman) and was subsequently transferred to fnbA(Newman) by replacement of the entire region encompassing the C, D, and W domains with the respective sequence of fnbB(Newman). Using heterologous expression in Staphylococcus carnosus, we found that truncated FnBPs were completely secreted into the culture medium and not anchored to the cell wall, since they lack the sortase motif (LPETG). Consequently, this led to a loss of FnBP-dependent functions, such as strong adhesion to immobilized fibronectin, binding of fibrinogen, and host cell invasion. This mutation may explain some of the earlier reported conflicting data with strain Newman. Thus, care should be taken when drawing negative conclusions about the role of FnBPs as a virulence factor in a given model.
Collapse
Affiliation(s)
- Matthias Grundmeier
- Institute of Medical Microbiology, University of Münster, Domagkstrasse 10, D-48149 Münster, Germany
| | | | | | | | | | | |
Collapse
|
35
|
Bjerketorp J, Rosander A, Nilsson M, Jacobsson K, Frykberg L. Sorting a Staphylococcus aureus phage display library against ex vivo biomaterial. J Med Microbiol 2004; 53:945-951. [PMID: 15358815 DOI: 10.1099/jmm.0.45638-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
A phage display library made from Staphylococcus aureus DNA was sorted against a central venous catheter (CVC) that had been removed from a patient 2 days after insertion. After the first panning, approximately 50% of the clones encoded proteins known to interact with mammalian proteins. After the second and third pannings, fibrinogen-binding and beta2-glycoprotein I (beta2-GPI)-binding phage particles were clearly dominating. Proteins adsorbed to different CVCs were investigated using specific antibodies. Among the proteins probed for, fibrinogen was most abundant, but, interestingly, beta2-GPI was also detected on all tested CVCs.
Collapse
Affiliation(s)
- Joakim Bjerketorp
- Department of Microbiology, Swedish University of Agricultural Sciences, SE-750 07 Uppsala, Sweden
| | - Anna Rosander
- Department of Microbiology, Swedish University of Agricultural Sciences, SE-750 07 Uppsala, Sweden
| | - Martin Nilsson
- Department of Microbiology, Swedish University of Agricultural Sciences, SE-750 07 Uppsala, Sweden
| | - Karin Jacobsson
- Department of Microbiology, Swedish University of Agricultural Sciences, SE-750 07 Uppsala, Sweden
| | - Lars Frykberg
- Department of Microbiology, Swedish University of Agricultural Sciences, SE-750 07 Uppsala, Sweden
| |
Collapse
|
36
|
Renzoni A, Francois P, Li D, Kelley WL, Lew DP, Vaudaux P, Schrenzel J. Modulation of fibronectin adhesins and other virulence factors in a teicoplanin-resistant derivative of methicillin-resistant Staphylococcus aureus. Antimicrob Agents Chemother 2004; 48:2958-65. [PMID: 15273106 PMCID: PMC478536 DOI: 10.1128/aac.48.8.2958-2965.2004] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2003] [Revised: 02/05/2004] [Accepted: 04/06/2004] [Indexed: 11/20/2022] Open
Abstract
The impact of glycopeptide resistance on the molecular regulation of Staphylococcus aureus virulence and attachment to host tissues is poorly documented. We compared stable teicoplanin-resistant methicillin-resistant S. aureus (MRSA) strain 14-4 with its teicoplanin-susceptible MRSA parent, strain MRGR3, which exhibits a high degree of virulence in a rat model of chronic foreign body MRSA infection. The levels of fibronectin-mediated adhesion and surface display of fibronectin-binding proteins were higher in teicoplanin-resistant strain 14-4 than in its teicoplanin-susceptible parent or a teicoplanin-susceptible revertant (strain 14-4rev) that spontaneously emerged during tissue cage infection. Quantitative reverse transcription-PCR (qRT-PCR) showed four- and twofold higher steady-state levels of fnbA and fnbB transcripts, respectively, in strain 14-4 than in its teicoplanin-susceptible counterparts. Analysis of global regulatory activities by qRT-PCR revealed a strong reduction in the steady-state levels of RNAIII and RNAII in the teicoplanin-resistant strain compared to in its teicoplanin-susceptible counterparts. In contrast, sarA mRNA levels were more than fivefold higher in strain 14-4 than in MRGR3 and 14-4rev. Furthermore, the alternative transcription factor sigma B had a higher level of functional activity in the teicoplanin-resistant strain than in its teicoplanin-susceptible counterparts, as evidenced by significant increases in both the sigma B-dependent asp23 mRNA levels and the sarA P3 promoter-derived transcript levels, as assayed by qRT-PCR and Northern blotting, respectively. These data provide further evidence that the emergence of glycopeptide resistance is linked by still poorly understood molecular pathways with significant pleiotropic changes in the expression and regulation of some major virulence genes. These molecular and phenotypic changes may have a profound impact on the bacterial adhesion and colonization properties of such multiresistant organisms.
Collapse
Affiliation(s)
- Adriana Renzoni
- Division of Infectious Diseases, University Hospitals of Geneva, CH-1211 Geneva 14, Switzerland
| | | | | | | | | | | | | |
Collapse
|
37
|
Rennermalm A, Nilsson M, Flock JI. The fibrinogen binding protein of Staphylococcus epidermidis is a target for opsonic antibodies. Infect Immun 2004; 72:3081-3. [PMID: 15102827 PMCID: PMC387914 DOI: 10.1128/iai.72.5.3081-3083.2004] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Antibodies against the fibrinogen binding protein (Fbe) of Staphylococcus epidermidis significantly increased macrophage phagocytosis. Antibodies against autolysin E were opsonic but to a lesser extent. Antibodies against a novel, putatively surface-located antigen were unable to enhance phagocytosis. The severity of systemic infection of mice with S. epidermidis was reduced if the bacteria were preopsonized with anti-Fbe prior to administration. Fbe is thus a strong candidate for protein vaccination against S. epidermidis infection, and antibodies against Fbe can be used to prevent or treat infections caused by S. epidermidis.
Collapse
Affiliation(s)
- Anna Rennermalm
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | | |
Collapse
|
38
|
Pei L, Arvholm IL, Lonnies L, Flock JI. GST-Fbe can recognize beta-chains of fibrin(ogen) on explanted materials. J Chromatogr B Analyt Technol Biomed Life Sci 2003; 786:319-25. [PMID: 12651029 DOI: 10.1016/s1570-0232(02)00744-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Staphylococcus epidermidis, a coagulase-negative staphylococcus (CoNS), is one of the leading pathogens of nosocomial infections, particularly associated with foreign body infections. Adherence of S. epidermidis to fibrinogen deposited on the surfaces of implants is important for the development of foreign body infections. A gene (fbe) encoding a fibrinogen-binding protein from S. epidermidis (Fbe) was identified by shotgun phage display. A portion of fbe was cloned into a GST-fusion vector. Affinity to glutathione-Sepharose by the GST-tag and affinity to fibrinogen-Sepharose by the Fbe part were applied to purify the recombinant Fbe. The purity and efficacy of the methods used in protein purification was compared. Furthermore, the potential physiological role of Fbe was studied by the interaction between GST-Fbe and components extracted from explanted materials in vitro.
Collapse
Affiliation(s)
- Lei Pei
- Division of Clinical Bacteriology, Department of Microbiology, Pathology and Immunology, Huddinge University Hospital, Karolinska Institute, SE-141 86, Stockholm, Sweden.
| | | | | | | |
Collapse
|
39
|
Vaudaux P, Francois P, Bisognano C, Kelley WL, Lew DP, Schrenzel J, Proctor RA, McNamara PJ, Peters G, Von Eiff C. Increased expression of clumping factor and fibronectin-binding proteins by hemB mutants of Staphylococcus aureus expressing small colony variant phenotypes. Infect Immun 2002; 70:5428-37. [PMID: 12228267 PMCID: PMC128368 DOI: 10.1128/iai.70.10.5428-5437.2002] [Citation(s) in RCA: 145] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Small colony variants (SCVs) of Staphylococcus aureus are slow-growing subpopulations that cause persistent and relapsing infections. The altered phenotype of SCV can arise from defects in menadione or hemin biosynthesis, which disrupt the electron transport chain and decrease ATP concentrations. With SCVs, virulence is altered by a decrease in exotoxin production and susceptibility to various antibiotics, allowing their intracellular survival. The expression of bacterial adhesins by SCVs is poorly documented. We tested fibrinogen- and fibronectin-mediated adhesion of a hemB mutant of S. aureus 8325-4 that is defective for hemin biosynthesis and exhibits a complete SCV phenotype. In this strain, adhesion to fibrinogen and fibronectin was significantly higher than that of its isogenic, normally growing parent and correlated with the increased surface display of these adhesins as assessed by flow cytometry. Real-time quantitative reverse transcription-PCR demonstrated increased expression of clfA and fnb genes by the hemB mutant compared to its isogenic parent. The influence of the hemB mutation on altered adhesin expression was confirmed by showing complete restoration of the wild-type adhesive phenotype in the hemB mutant, either by complementing with intact hemB or by supplementing the growth medium with hemin. Increased surface display of fibrinogen and fibronectin adhesins by the hemB mutation occurred independently from agr, a major regulatory locus of virulence factors in S. aureus. Both agr-positive and agr-lacking hemB mutants were also more efficiently internalized by human embryonic kidney cells than were their isogenic controls, presumably because of increased surface display of their fibronectin adhesins.
Collapse
Affiliation(s)
- Pierre Vaudaux
- Division of Infectious Diseases, University Hospital, CH-1211 Geneva 14, Switzerland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
Biofilm bacterial infections are implicated in most human bacterial infections and are also common in patients undergoing treatment with hemodialysis and peritoneal dialysis. Skin bacteria, which grow into microcolonies with biofilm formation in dialysis environments, are implicated in most of these infections. Dissemination of bacterial biofilms in hemodialysis patients induces bacteremia and endotoxemia. In peritoneal dialysis patients, biofilm causes peritonitis and catheter-related infections with consequent loss of catheters and technique failure. Effective strategies for the diagnosis, intervention, and prevention of biofilm-related infections in dialysis patients are described in this review.
Collapse
Affiliation(s)
- Mrinal K Dasgupta
- Division of Nephrology and Immunology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
41
|
Abstract
Bacteria frequently attach to medical devices such as intravascular catheters by forming sessile multicellular communities known as biofilms, which can be the source of persistent infections that are recalcitrant to systemic antibiotic therapy. As a result of this persistence, a number of technologies have been developed to prevent catheter-associated biofilm formation. Whereas the most straightforward approaches focus on impregnating catheter material with classical antimicrobial agents, these approaches are not universally effective, thereby underscoring the need for more potent and more sophisticated approaches to the prevention of catheter-related biofilm infections.
Collapse
Affiliation(s)
- Paul N Danese
- Microbia, Inc., One Kendall Square, Building 1400W, Cambridge, MA 02139, USA.
| |
Collapse
|
42
|
Brouillette E, Lacasse P, Shkreta L, Bélanger J, Grondin G, Diarra MS, Fournier S, Talbot BG. DNA immunization against the clumping factor A (ClfA) of Staphylococcus aureus. Vaccine 2002; 20:2348-57. [PMID: 12009291 DOI: 10.1016/s0264-410x(02)00100-7] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Adhesins are considered the most important virulence factors during early phases Staphylococcus aureus infection. Antibodies induced by vaccination toward an adhesin should reduce the adherence of the pathogen and augment its phagocytosis. The present report describes the immune response of mice to a DNA vaccine directed against one of these adhesins, clumping factor A (ClfA). Injection of plasmids expressing the fibrinogen-binding region A of ClfA induced a strong and specific antibody response to ClfA in mice. In addition, splenocyte proliferation was provoked by in vitro stimulation with recombinant ClfA, thus, indicating direct implication of these cells in the immune response. Pre-incubation of S. aureus with sera of vaccinated mice reduced the pathogen's ability to bind fibrinogen by up to 92%. These pre-incubated bacteria were phagocytosed by macrophages at an increased level in vitro and were less virulent in vivo in a mouse mastitis model. However, DNA-immunized mice were not protected against an intraperitoneal challenge. Overall, the results suggest that DNA immunization against adhesins represents a new and valuable approach to combat S. aureus infections.
Collapse
Affiliation(s)
- Eric Brouillette
- Département de Biologie, Faculté des Sciences, Université de Sherbrooke, Sherbrooke, Canada
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Berlot S, Aissaoui Z, Pavon-Djavid G, Belleney J, Jozefowicz M, Hélary G, Migonney V. Biomimetic poly(methyl methacrylate)-based terpolymers: modulation of bacterial adhesion effect. Biomacromolecules 2002; 3:63-8. [PMID: 11866557 DOI: 10.1021/bm015580m] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Adherence of Staphylococcus aureus, responsible for major foreign body infections, was assessed onto functionalized poly(methyl methacrylate)-based terpolymers bearing sulfonate and carboxylate groups and onto poly(methyl methacrylate) as control. These terpolymers, have been synthesized by radical copolymerization of methyl methacrylate, methacrylic acid, and sodium styrene sulfonate by varying the ratio R = [COO(-)]/[COO(-) + SO(3)(-)] from 0 to 1 and keeping ionic monomer content between 7 and 18%. Adsorption of fibronectin onto poly(methyl methacrylate) was shown to dramatically promote bacterial adherence, whereas a strong inhibition of bacteria adherence was observed onto functionalized terpolymers containing both carboxylate and sulfonate groups. When terpolymers were predominantly functionalized by carboxylate groups, bacteria adherence was favored and reached values close to those obtained for poly(methyl methacrylate). These results have been related to the distribution of the anionic groups along the macromolecular chains, creating active sites responsible for specific interactions with fibronectin and inducing modifications of its conformation. The conformation of the adsorbed adhesive protein was then suggested to have an influence on the availability of its interaction sites to bacteria adhesins and therefore on modulation of bacteria adherence. Inhibition of Staphylococcus aureus adherence by functionalized poly(methyl methacrylate)-based terpolymers is of great interest in the field of biomedical implants and especially in the case of ophthalmic applications.
Collapse
Affiliation(s)
- Sandrine Berlot
- Laboratoire de Recherches sur les Macromolécules, FRE 2314, Université Paris 13, 99 Avenue J-B Clément, 93430, Villetaneuse, France
| | | | | | | | | | | | | |
Collapse
|
44
|
Que YA, François P, Haefliger JA, Entenza JM, Vaudaux P, Moreillon P. Reassessing the role of Staphylococcus aureus clumping factor and fibronectin-binding protein by expression in Lactococcus lactis. Infect Immun 2001; 69:6296-302. [PMID: 11553573 PMCID: PMC98764 DOI: 10.1128/iai.69.10.6296-6302.2001] [Citation(s) in RCA: 114] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Since Staphylococcus aureus expresses multiple pathogenic factors, studying their individual roles in single-gene-knockout mutants is difficult. To circumvent this problem, S. aureus clumping factor A (clfA) and fibronectin-binding protein A (fnbA) genes were constitutively expressed in poorly pathogenic Lactococcus lactis using the recently described pOri23 vector. The recombinant organisms were tested in vitro for their adherence to immobilized fibrinogen and fibronectin and in vivo for their ability to infect rats with catheter-induced aortic vegetations. In vitro, both clfA and fnbA increased the adherence of lactococci to their specific ligands to a similar extent as the S. aureus gene donor. In vivo, the minimum inoculum size producing endocarditis in > or =80% of the rats (80% infective dose [ID80]) with the parent lactococcus was > or =10(7) CFU. In contrast, clfA-expressing and fnbA-expressing lactococci required only 10(5) CFU to infect the majority of the animals (P < 0.00005). This was comparable to the infectivities of classical endocarditis pathogens such as S. aureus and streptococci (ID80 = 10(4) to 10(5) CFU) in this model. The results confirmed the role of clfA in endovascular infection, but with a much higher degree of confidence than with single-gene-inactivated staphylococci. Moreover, they identified fnbA as a critical virulence factor of equivalent importance. This was in contrast to previous studies that produced controversial results regarding this very determinant. Taken together, the present observations suggest that if antiadhesin therapy were to be developed, at least both of the clfA and fnbA products should be blocked for the therapy to be effective.
Collapse
Affiliation(s)
- Y A Que
- Division of Infectious Diseases, Department of Internal Medicine, Centre Hospitalier Universitaire Vaudois, 1011 Lausanne, Switzerland
| | | | | | | | | | | |
Collapse
|
45
|
Pei L, Flock JI. Lack of fbe, the gene for a fibrinogen-binding protein from Staphylococcus epidermidis, reduces its adherence to fibrinogen coated surfaces. Microb Pathog 2001; 31:185-93. [PMID: 11562171 DOI: 10.1006/mpat.2001.0462] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The significance of Fbe, a fibrinogen-binding protein in Staphylococcus epidermidis, was investigated. A fbe mutant was constructed by allelic replacement, where a Gentamicin resistance gene replaced a portion of the A region of fbe. Adherence assay to immobilized fibrinogen on polyethylene surfaces and peripheral venous catheters from patients showed that the fibrinogen binding ability of the mutant was reduced compared to its parental strain. This shows that Fbe is a major factor involved in adherence of S. epidermidis to fibrinogen. No difference was found between the wild-type and mutant in their affinity to immobilized fibronectin.
Collapse
Affiliation(s)
- L Pei
- Division of Clinical Bacteriology, Department of Immunology, Pathology and Microbiology, Stockholm, S-141 86, Sweden
| | | |
Collapse
|
46
|
Hartford O, O'Brien L, Schofield K, Wells J, Foster TJ. The Fbe (SdrG) protein of Staphylococcus epidermidis HB promotes bacterial adherence to fibrinogen. MICROBIOLOGY (READING, ENGLAND) 2001; 147:2545-2552. [PMID: 11535794 DOI: 10.1099/00221287-147-9-2545] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Staphylococcus epidermidis strains HB and K28 express surface proteins called Fbe or SdrG, respectively, that have sequence similarity to the clumping factors ClfA and ClfB of Staphylococcus aureus. A mutation in the fbe gene of strain HB was isolated by directed plasmid integration using the broad-host-range temperature-sensitive plasmid pG(+)Host9 (pVE6155). An internal fragment of fbe was cloned into pG(+)Host9 and the chimaeric plasmid was mobilized from S. aureus RN4220 to S. epidermidis 9142 by conjugation promoted by plasmid pGO1. The plasmid was then transferred to S. epidermidis strain HB by phage-48-mediated transduction. The plasmid integrated into the chromosomal fbe gene at a frequency of 2.8 x 10(-4). All the survivors tested had a copy of pG(+)Host9'fbe' integrated into the chromosomal fbe gene either as a single copy or as a tandem array. Western immunoblotting showed that the wall-associated Fbe protein was absent in the mutant. Wild-type S. epidermidis HB adhered to immobilized fibrinogen in a dose-dependent and saturable fashion whereas the mutant did not bind. The Fbe proteins of HB and K28 were expressed at a high level in Lactococcus lactis MG1363 using the expression vector pKS80. These strains adhered strongly to immobilized fibrinogen. These results confirm that Fbe is a fibrinogen-binding adhesin.
Collapse
Affiliation(s)
- Orla Hartford
- Department of Microbiology, Moyne Institute of Preventive Medicine, Trinity College, Dublin 2, Ireland1
| | - Louise O'Brien
- Department of Microbiology, Moyne Institute of Preventive Medicine, Trinity College, Dublin 2, Ireland1
| | - Karin Schofield
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK2
| | - Jerry Wells
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK2
| | - Timothy J Foster
- Department of Microbiology, Moyne Institute of Preventive Medicine, Trinity College, Dublin 2, Ireland1
| |
Collapse
|
47
|
McAleese FM, Walsh EJ, Sieprawska M, Potempa J, Foster TJ. Loss of clumping factor B fibrinogen binding activity by Staphylococcus aureus involves cessation of transcription, shedding and cleavage by metalloprotease. J Biol Chem 2001; 276:29969-78. [PMID: 11399757 DOI: 10.1074/jbc.m102389200] [Citation(s) in RCA: 107] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The fibrinogen-binding protein clumping factor B (ClfB) of Staphylococcus aureus is present on the surface of cells from the early exponential phase of growth in greater amounts than on cells from late exponential phase and is barely detectable on cells from stationary phase. Expression of a clfB-lacZ fusion indicated that transcription stopped before the end of exponential phase. Mutations in the global regulators agr and sar had no effect on clfB transcription. The loss of ClfB protein from cells in stationary phase was due to expression ending before cells stopped growing, combined with shedding of some of the protein into the growth medium and dilution of those molecules remaining on the cell surface during the two to three cell division events leading to stationary phase. Two forms of the protein occurred on the cell surface, the smaller of which was generated by loss of a domain from the N terminus. The proportion of the smaller form increased as the cultures grew. The metalloprotease aureolysin was shown to be responsible for cleavage of ClfB. Cleavage was inhibited by EDTA and o-phenanthroline and did not occur in an aureolysin-deficient mutant. Purified aureolysin promoted cleavage of cell surface-located ClfB as well as the recombinant A domain of ClfB. Cleavage was detected at two sites, one located between residues Ser(197) and Leu(198) and the other between Ala(199) and Val(200). The truncated form of ClfB did not bind fibrinogen.
Collapse
MESH Headings
- Adhesins, Bacterial/metabolism
- Adhesins, Bacterial/physiology
- Alanine/chemistry
- Binding Sites
- Blotting, Southern
- Blotting, Western
- Coagulase/metabolism
- Coagulase/physiology
- Dose-Response Relationship, Drug
- Edetic Acid/pharmacology
- Electrophoresis, Polyacrylamide Gel
- Fibrinogen/metabolism
- Genes, Reporter
- Genotype
- Leucine/chemistry
- Metalloendopeptidases/metabolism
- Mutagenesis, Site-Directed
- Mutation
- Phenanthrolines/pharmacology
- Promoter Regions, Genetic
- Protein Binding
- Protein Structure, Tertiary
- Recombinant Fusion Proteins/metabolism
- Recombinant Proteins/metabolism
- Serine/chemistry
- Staphylococcus aureus/metabolism
- Time Factors
- Transcription, Genetic
- Valine/chemistry
- beta-Galactosidase/metabolism
Collapse
Affiliation(s)
- F M McAleese
- Microbiology Department, Moyne Institute for Preventive Medicine, Trinity College, Dublin 2, Ireland
| | | | | | | | | |
Collapse
|
48
|
Sinha B, Francois P, Que YA, Hussain M, Heilmann C, Moreillon P, Lew D, Krause KH, Peters G, Herrmann M. Heterologously expressed Staphylococcus aureus fibronectin-binding proteins are sufficient for invasion of host cells. Infect Immun 2000; 68:6871-8. [PMID: 11083807 PMCID: PMC97792 DOI: 10.1128/iai.68.12.6871-6878.2000] [Citation(s) in RCA: 178] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Staphylococcus aureus invasion of mammalian cells, including epithelial, endothelial, and fibroblastic cells, critically depends on fibronectin bridging between S. aureus fibronectin-binding proteins (FnBPs) and the host fibronectin receptor integrin alpha(5)beta(1) (B. Sinha et al., Cell. Microbiol. 1:101-117, 1999). However, it is unknown whether this mechanism is sufficient for S. aureus invasion. To address this question, various S. aureus adhesins (FnBPA, FnBPB, and clumping factor [ClfA]) were expressed in Staphylococcus carnosus and Lactococcus lactis subsp. cremoris. Both noninvasive gram-positive microorganisms are genetically distinct from S. aureus, lack any known S. aureus surface protein, and do not bind fibronectin. Transformants of S. carnosus and L. lactis harboring plasmids coding for various S. aureus surface proteins (FnBPA, FnBPB, and ClfA) functionally expressed adhesins (as determined by bacterial clumping in plasma, specific latex agglutination, Western ligand blotting, and binding to immobilized and soluble fibronectin). FnBPA or FnBPB but not of ClfA conferred invasiveness to S. carnosus and L. lactis. Invasion of 293 cells by transformants was comparable to that of strongly invasive S. aureus strain Cowan 1. Binding of soluble and immobilized fibronectin paralleled invasiveness, demonstrating that the amount of accessible surface FnBPs is rate limiting. Thus, S. aureus FnBPs confer invasiveness to noninvasive, apathogenic gram-positive cocci. Furthermore, FnBP-coated polystyrene beads were internalized by 293 cells, demonstrating that FnBPs are sufficient for invasion of host cells without the need for (S. aureus-specific) coreceptors.
Collapse
Affiliation(s)
- B Sinha
- Institute of Medical Microbiology, University of Münster, D-48129 Münster, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
Polymers are widely used for a large range of medical devices used as biomaterials on a temporary, intermittent, and long-term basis. It is now well accepted that the initial rapid adsorption of proteins to polymeric surfaces affects the performance of these biomaterials. However, protein adsorption to a polymer surface can be modulated by an appropriate design of the interface. Extensive study has shown that these interactions can be minimized by coating with a highly hydrated layer (hydrogel), by grafting on the surface different biomolecules, or by creating domains with chemical functions (charges, hydrophilic groups). Our laboratory has investigated the latter approach over the past 2 decades, in particular the synthesis and the biological activities of polymers to improve the biocompatibility of blood-contacting devices. These soluble and insoluble polymers were obtained by chemical substitution of macromolecular chains with suitable groups able to develop specific interactions with biological components. Applied to compatibility with the blood and the immune systems, this concept has been extended to interactions of polymeric biomaterials with eukaryotic and prokaryotic cells. The design of new biomaterials with low bacterial attachment is thus under intensive study. After a brief overview of current trends in the surface modifications of biocompatible materials, we will describe how biospecific polymers can be obtained and review our recent results on the inhibition of bacterial adhesion using one type of functionalized polymer obtained by random substitution. This strategy, applied to existing or new materials, seems promising for the limitation of biomaterial-associated infections.
Collapse
Affiliation(s)
- B Montdargent
- Laboratoire de Recherches sur les Macromolécules, Institut Galilée, Villetaneuse, France
| | | |
Collapse
|
50
|
Proctor RA. Toward an understanding of biomaterial infections: a complex interplay between the host and bacteria. THE JOURNAL OF LABORATORY AND CLINICAL MEDICINE 2000; 135:14-5. [PMID: 10638689 DOI: 10.1016/s0022-2143(00)70015-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|