1151
|
Brockhurst MA, Hochberg ME, Bell T, Buckling A. Character displacement promotes cooperation in bacterial biofilms. Curr Biol 2006; 16:2030-4. [PMID: 17055982 DOI: 10.1016/j.cub.2006.08.068] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2006] [Revised: 08/09/2006] [Accepted: 08/21/2006] [Indexed: 11/18/2022]
Abstract
Resource competition within a group of cooperators is expected to decrease selection for cooperative behavior but can also result in diversifying selection for the use of different resources, which in turn could retard the breakdown of cooperation. Diverse groups are likely to be less susceptible to invasion by noncooperating social cheats: First, competition repression resulting from character displacement may provide less of a selective advantage to cheating; second, cheats may trade off the ability to exploit cooperators that specialize in one type of resource against cooperators that specialize in another ; third, diverse communities of any kind may have higher invasion resistance because there are fewer resources available for an invader to use . Furthermore, diverse groups are likely to be more productive than clonal groups if a wider range of total resources are being used . We addressed these issues by using the cooperative trait of biofilm formation in Pseudomonas fluorescens. Character displacement through resource competition evolved within biofilms; productivity increased with increasing character displacement, and diverse biofilms were less susceptible to invasion by cheats. These results demonstrate that diversification into different ecological niches can minimize selection against cooperation in the face of local resource competition.
Collapse
Affiliation(s)
- Michael A Brockhurst
- School of Biological Sciences, Biosciences Building, University of Liverpool, Crown Street, Liverpool L69 7ZB, United Kingdom.
| | | | | | | |
Collapse
|
1152
|
Tu Quoc PH, Genevaux P, Pajunen M, Savilahti H, Georgopoulos C, Schrenzel J, Kelley WL. Isolation and characterization of biofilm formation-defective mutants of Staphylococcus aureus. Infect Immun 2006; 75:1079-88. [PMID: 17158901 PMCID: PMC1828571 DOI: 10.1128/iai.01143-06] [Citation(s) in RCA: 121] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Staphylococcus aureus produces biofilm and this mode of colonization facilitates infections that are often difficult to treat and engender high morbidity and mortality. We have exploited bacteriophage Mu transposition methods to create an insertional mutant library in a highly biofilm-forming S. aureus clinical isolate. Our screen identified 38 insertions in 23 distinct genes together with one intergenic region that significantly reduced biofilm formation. Nineteen insertions were mapped in loci not previously known to affect biofilm in this organism. These include insertions in codY, srrA, mgrA, and fmtA, a putative DEAD-box helicase, two members of the zinc-metallo-beta lactamase/beta-CASP family, and a hypothetical protein with a GGDEF motif. Fifteen insertions occurred in the icaADBC operon, which produces intercellular adhesion antigen (PIA) and is important for biofilm formation in many strains of S. aureus and Staphylococcus epidermidis. Obtaining a high proportion of independent Em-Mu disruptions in icaADBC demonstrated both the importance of PIA for biofilm formation in this clinical strain and the strong validation of the screening procedure that concomitantly uncovered additional mutants. All non-ica mutants were further analyzed by immunoblotting and biochemical fractionation for perturbation of PIA and wall teichoic acid. PIA levels were diminished in the majority of non-ica insertional mutants. Three mutant strains were chosen and were functionally complemented for restored biofilm formation by transformation with plasmids carrying the cloned wild-type gene under the control of a xylose-inducible promoter. This is a comprehensive collection of biofilm-defective mutants that underscores the multifactorial genetic program underlying the establishment of biofilm in this insidious pathogen.
Collapse
Affiliation(s)
- Patrick H Tu Quoc
- Division of Infectious Diseases, University Hospital of Geneva, 24 rue Micheli-du-Crest, CH-1211 Geneva 14, Switzerland
| | | | | | | | | | | | | |
Collapse
|
1153
|
Romeo T. When the party is over: a signal for dispersal of Pseudomonas aeruginosa biofilms. J Bacteriol 2006; 188:7325-7. [PMID: 17050919 PMCID: PMC1636271 DOI: 10.1128/jb.01317-06] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Affiliation(s)
- Tony Romeo
- Department of Microbiology and Immunology, Emory University School of Medicine, 3105 Rollins Research Center, 1510 Clifton Road N.E., Atlanta, GA 30322, USA.
| |
Collapse
|
1154
|
Kim YH, Lee Y, Kim S, Yeom J, Yeom S, Seok Kim B, Oh S, Park S, Jeon CO, Park W. The role of periplasmic antioxidant enzymes (superoxide dismutase and thiol peroxidase) of the Shiga toxin-producingEscherichia coli O157:H7 in the formation of biofilms. Proteomics 2006; 6:6181-93. [PMID: 17133368 DOI: 10.1002/pmic.200600320] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
This study examined the role of the periplasmic oxidative defense proteins, copper, zinc superoxide dismutase (SodC), and thiol peroxidase (Tpx), from the Shiga toxin-producing Escherichia coli O157:H7 (STEC) in the formation of biofilms. Proteomic analyses have shown significantly higher expression levels of both periplasmic antioxidant systems (SodC and Tpx) in STEC cells grown under biofilm conditions than under planktonic conditions. An analysis of their growth phase-dependent gene expression indicated that a high level of the sodC expression occurred during the stationary phase and that the expression of the tpx gene was strongly induced only during the exponential growth phase. Exogenous hydrogen peroxide reduced the aerobic growth of the STEC sodC and tpx mutants by more than that of their parental strain. The two mutants also displayed significant reductions in their attachment to both biotic (HT-29 epithelial cell) and abiotic surfaces (polystyrene and polyvinyl chloride microplates) during static aerobic growth. However, the growth rates of both wild-type and mutants were similar under aerobic growth conditions. The formation of an STEC biofilm was only observed with the wild-type STEC cells in glass capillary tubes under continuous flow-culture conditions compared with the STEC sodC and tpx mutants. To the best of our knowledge, this is the first mutational study to show the contribution of sodC and tpx gene products to the formation of an E. coli O157:H7 biofilm. These results also suggest that these biofilms are physiologically heterogeneous and that oxidative stress defenses in both the exponential and stationary growth stages play important roles in the formation of STEC biofilms.
Collapse
Affiliation(s)
- Young Hoon Kim
- Division of Food Science, Korea University, Seoul, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1155
|
Van Wamel WJB, Hendrickx APA, Bonten MJM, Top J, Posthuma G, Willems RJL. Growth condition-dependent Esp expression by Enterococcus faecium affects initial adherence and biofilm formation. Infect Immun 2006; 75:924-31. [PMID: 17118984 PMCID: PMC1828491 DOI: 10.1128/iai.00941-06] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
A genetic subpopulation of Enterococcus faecium, called clonal complex 17 (CC-17), is strongly associated with hospital outbreaks and invasive infections. Most CC-17 strains contain a putative pathogenicity island encoding the E. faecium variant of enterococcal surface protein (Esp). Western blotting, flow cytometric analyses, and electron microscopy showed that Esp is expressed and exposed on the surface of E. faecium, though Esp expression and surface exposure are highly varied among different strains. Furthermore, Esp expression depends on growth conditions like temperature and anaerobioses. When grown at 37 degrees C, five of six esp-positive E. faecium strains showed significantly increased levels of surface-exposed Esp compared to bacteria grown at 21 degrees C, which was confirmed at the transcriptional level by real-time PCR. In addition, a significant increase in surface-exposed Esp was found in half of these strains when grown at 37 degrees C under anaerobic conditions compared to the level in bacteria grown under aerobic conditions. Finally, amounts of surface-exposed Esp correlated with initial adherence to polystyrene (R(2) = 0.7146) and biofilm formation (R(2) = 0.7535). Polystyrene adherence was competitively inhibited by soluble recombinant N-terminal Esp. This study demonstrates that Esp expression on the surface of E. faecium (i) varies consistently between strains, (ii) is growth condition dependent, and (iii) is quantitatively correlated with initial adherence and biofilm formation. These data indicate that E. faecium senses and responds to changing environmental conditions, which might play a role in the early stages of infection when bacteria transit from oxygen-rich conditions at room temperature to anaerobic conditions at body temperature. In addition, variation of surface exposure may explain the contrasting findings reported on the role of Esp in biofilm formation.
Collapse
Affiliation(s)
- Willem J B Van Wamel
- Eijkman-Winkler Institute, Room G04-614, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands.
| | | | | | | | | | | |
Collapse
|
1156
|
Parise G, Mishra M, Itoh Y, Romeo T, Deora R. Role of a putative polysaccharide locus in Bordetella biofilm development. J Bacteriol 2006; 189:750-60. [PMID: 17114249 PMCID: PMC1797297 DOI: 10.1128/jb.00953-06] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Bordetellae are gram-negative bacteria that colonize the respiratory tracts of animals and humans. We and others have recently shown that these bacteria are capable of living as sessile communities known as biofilms on a number of abiotic surfaces. During the biofilm mode of existence, bacteria produce one or more extracellular polymeric substances that function, in part, to hold the cells together and to a surface. There is little information on either the constituents of the biofilm matrix or the genetic basis of biofilm development by Bordetella spp. By utilizing immunoblot assays and by enzymatic hydrolysis using dispersin B (DspB), a glycosyl hydrolase that specifically cleaves the polysaccharide poly-beta-1,6-N-acetyl-D-glucosamine (poly-beta-1,6-GlcNAc), we provide evidence for the production of poly-beta-1,6-GlcNAc by various Bordetella species (Bordetella bronchiseptica, B. pertussis, and B. parapertussis) and its role in their biofilm development. We have investigated the role of a Bordetella locus, here designated bpsABCD, in biofilm formation. The bps (Bordetella polysaccharide) locus is homologous to several bacterial loci that are required for the production of poly-beta-1,6-GlcNAc and have been implicated in bacterial biofilm formation. By utilizing multiple microscopic techniques to analyze biofilm formation under both static and hydrodynamic conditions, we demonstrate that the bps locus, although not essential at the initial stages of biofilm formation, contributes to the stability and the maintenance of the complex architecture of Bordetella biofilms.
Collapse
Affiliation(s)
- Gina Parise
- Program in Molecular Genetics, Department of Microbiology and Immunology, Wake Forest University Health Sciences, Medical Center Blvd., Gray 5086, Winston-Salem, NC 27157, USA
| | | | | | | | | |
Collapse
|
1157
|
Dong C, Beis K, Nesper J, Brunkan AL, Clarke BR, Whitfield C, Naismith JH. Wza the translocon for E. coli capsular polysaccharides defines a new class of membrane protein. Nature 2006; 444:226-9. [PMID: 17086202 PMCID: PMC3315050 DOI: 10.1038/nature05267] [Citation(s) in RCA: 281] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2006] [Accepted: 09/21/2006] [Indexed: 11/09/2022]
Abstract
Many types of bacteria produce extracellular polysaccharides (EPSs). Some are secreted polymers and show only limited association with the cell surface, whereas others are firmly attached to the cell surface and form a discrete structural layer, the capsule, which envelopes the cell and allows the bacteria to evade or counteract the host immune system. EPSs have critical roles in bacterial colonization of surfaces, such as epithelia and medical implants; in addition some EPSs have important industrial and biomedical applications in their own right. Here we describe the 2.26 A resolution structure of the 340 kDa octamer of Wza, an integral outer membrane lipoprotein, which is essential for group 1 capsule export in Escherichia coli. The transmembrane region is a novel alpha-helical barrel. The bulk of the Wza structure is located in the periplasm and comprises three novel domains forming a large central cavity. Wza is open to the extracellular environment but closed to the periplasm. We propose a route and mechanism for translocation of the capsular polysaccharide. This work may provide insight into the export of other large polar molecules such as DNA and proteins.
Collapse
Affiliation(s)
- Changjiang Dong
- Centre for Biomolecular Sciences, EaStChem, The University, St Andrews, KY16 9RH
| | - Konstantinos Beis
- Centre for Biomolecular Sciences, EaStChem, The University, St Andrews, KY16 9RH
| | - Jutta Nesper
- Department of Molecular and Cellular Biology, The University of Guelph, Guelph, Ontario, Canada N1G 2W1
| | - Anne L. Brunkan
- Department of Molecular and Cellular Biology, The University of Guelph, Guelph, Ontario, Canada N1G 2W1
| | - Bradley R. Clarke
- Department of Molecular and Cellular Biology, The University of Guelph, Guelph, Ontario, Canada N1G 2W1
| | - Chris Whitfield
- Department of Molecular and Cellular Biology, The University of Guelph, Guelph, Ontario, Canada N1G 2W1
| | - James H. Naismith
- Centre for Biomolecular Sciences, EaStChem, The University, St Andrews, KY16 9RH
| |
Collapse
|
1158
|
Ngo Thi NA, Naumann D. Investigating the heterogeneity of cell growth in microbial colonies by FTIR microspectroscopy. Anal Bioanal Chem 2006; 387:1769-77. [PMID: 17066287 DOI: 10.1007/s00216-006-0829-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2006] [Revised: 08/30/2006] [Accepted: 09/04/2006] [Indexed: 11/29/2022]
Abstract
Microorganisms rarely occur as individual cells in nature and are, instead, organized in complex multicellular communities such as colonies, fruiting bodies, or biofilms. Interest in the natural microbial life-style has increased during the last decade and a whole plethora of techniques has been used to gain insight into the development, structure and composition of diverse microbial communities. We have developed a technique for investigating the spatial heterogeneity of microbial growth in macro-colonies which essentially entails excision of the colonies with the underlying agar, freezing and subsequent cryotoming of the colonies, then FTIR microspectroscopic mapping of the cryosections. Colonies from Legionella, Bacillus, and Candida strains were chosen as model systems of multi-cellular communities to evaluate the technique. The results obtained indicate pronounced cell population heterogeneity even in relatively young colonies cultivated under laboratory conditions. Spectral data obtained from different positions within, e.g., a colony of Legionella bozemanii 120 h old indicated that levels of the storage material poly-beta-hydroxybutyric acid were significantly higher in cells at the surface of the colonies than in those growing at the bottom next to the agar surface. Similarly, in a 24-h-old macro-colony of Bacillus megaterium significantly more of the capsular compound polyglutamic acid was detected in upper layers than in deeper layers of the colony. Results demonstrate that FTIR microspectroscopy can be an useful tool for investigation of the spatial heterogeneity of cell growth within microbial macro-colonies. It is suggested that the method also can be adapted to the analysis of more complex multicellular communities, for example fruiting bodies, biofilms, or colonies growing under natural conditions.
Collapse
Affiliation(s)
- N A Ngo Thi
- Robert Koch-Institute, Nordufer 20, 13353 Berlin, Germany
| | | |
Collapse
|
1159
|
Boddey JA, Flegg CP, Day CJ, Beacham IR, Peak IR. Temperature-regulated microcolony formation by Burkholderia pseudomallei requires pilA and enhances association with cultured human cells. Infect Immun 2006; 74:5374-81. [PMID: 16926432 PMCID: PMC1594825 DOI: 10.1128/iai.00569-06] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Burkholderia pseudomallei is the causative agent of melioidosis, a potentially fatal disease that is endemic to Northern Australia and Southeast Asia and is acquired from soil or water. Adherence of B. pseudomallei 08 to cultured cells increases dramatically following prior growth at 30 degrees C or less compared to that following prior growth at 37 degrees C. Here, we show that this occurs almost entirely as the result of microcolony formation (bacterium-bacterium interactions) following growth at 27 degrees C but not at 37 degrees C, which considerably enhances bacterial association with eukaryotic cells. Further, we demonstrate that the type IVA pilin-encoding gene, pilA, is essential for microcolony development by B. pseudomallei 08, and thus optimum association with eukaryotic cells, but is not required for direct adherence (bacterium-cell interactions). In contrast, although the B. pseudomallei genome sequence strain, K96243, also contains transcriptionally active pilA, microcolony formation rarely occurs following growth at either 27 degrees C or 37 degrees C and cell association occurs significantly less than with strain 08. Analysis of pilA transcription in 08 identified that pilA is dramatically upregulated under microcolony-forming conditions, viz., growth at low temperature, and association with eukaryotic cells; the pattern of transcription of pilA in K96243 differed from that in 08. Our study also suggests that biofilm formation by B. pseudomallei 08 and K96243 on polyvinylchloride is not mediated by pilA. Adherence and microcolony formation, and pilA transcription, vary between strains, consistent with known genomic variation in B. pseudomallei, and these phenotypes may be relevant to colonization from the environment.
Collapse
Affiliation(s)
- Justin A Boddey
- Institute for Glycomics, Griffith University, Gold Coast Campus, Gold Coast Mail Centre, Queensland 9726, Australia
| | | | | | | | | |
Collapse
|
1160
|
Orme R, Douglas CWI, Rimmer S, Webb M. Proteomic analysis of Escherichia coli biofilms reveals the overexpression of the outer membrane protein OmpA. Proteomics 2006; 6:4269-77. [PMID: 16888722 DOI: 10.1002/pmic.200600193] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Bacterial colonisation and biofilm formation on the surface of urinary catheters is a common cause of nosocomial infection, and as such is a major impediment to their long-term use. Understanding the mechanisms of biofilm formation on urinary catheters is critical to their control and will aid the future development of materials used in their manufacture. In this report we have used proteomic analysis coupled with immunoassays to show that the major outer membrane protein (OmpA) of Escherichia coli is overexpressed during biofilm formation. A series of synthetic hydrogels being developed for potential use as catheter coatings were used as the substrata and OmpA expression was increased in biofilms on all these surfaces, as well as being a feature of both a laboratory and a clinical strain of E. coli. Up-regulation of OmpA may, therefore, be a common feature of E. coli biofilms. These findings present OmpA as a potential target for biofilm inhibition and may contribute to the rational design of biofilm inhibiting hydrogel coatings for urinary catheters.
Collapse
Affiliation(s)
- Rowan Orme
- University of Manchester, Faculty of Medicine and Human Health, Centre for Molecular Medicine, Department of Medical Genetics, Manchester, UK
| | | | | | | |
Collapse
|
1161
|
Lynch SV, Mukundakrishnan K, Benoit MR, Ayyaswamy PS, Matin A. Escherichia coli biofilms formed under low-shear modeled microgravity in a ground-based system. Appl Environ Microbiol 2006; 72:7701-10. [PMID: 17028231 PMCID: PMC1694224 DOI: 10.1128/aem.01294-06] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Bacterial biofilms cause chronic diseases that are difficult to control. Since biofilm formation in space is well documented and planktonic cells become more resistant and virulent under modeled microgravity, it is important to determine the effect of this gravity condition on biofilms. Inclusion of glass microcarrier beads of appropriate dimensions and density with medium and inoculum, in vessels specially designed to permit ground-based investigations into aspects of low-shear modeled microgravity (LSMMG), facilitated these studies. Mathematical modeling of microcarrier behavior based on experimental conditions demonstrated that they satisfied the criteria for LSMMG conditions. Experimental observations confirmed that the microcarrier trajectory in the LSMMG vessel concurred with the predicted model. At 24 h, the LSMMG Escherichia coli biofilms were thicker than their normal-gravity counterparts and exhibited increased resistance to the general stressors salt and ethanol and to two antibiotics (penicillin and chloramphenicol). Biofilms of a mutant of E. coli, deficient in sigma(s), were impaired in developing LSMMG-conferred resistance to the general stressors but not to the antibiotics, indicating two separate pathways of LSMMG-conferred resistance.
Collapse
Affiliation(s)
- S V Lynch
- Department of Microbiology and Immunology, Sherman Fairchild Science Building, Stanford University School of Medicine, 299 Campus Drive, Stanford, CA 94305, USA
| | | | | | | | | |
Collapse
|
1162
|
Boes N, Schreiber K, Härtig E, Jaensch L, Schobert M. The Pseudomonas aeruginosa universal stress protein PA4352 is essential for surviving anaerobic energy stress. J Bacteriol 2006; 188:6529-38. [PMID: 16952944 PMCID: PMC1595484 DOI: 10.1128/jb.00308-06] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
During infection of the cystic fibrosis (CF) lung, Pseudomonas aeruginosa microcolonies are embedded in the anaerobic CF mucus. This anaerobic environment seems to contribute to the formation of more robust P. aeruginosa biofilms and to an increased antibiotic tolerance and therefore promotes persistent infection. This study characterizes the P. aeruginosa protein PA4352, which is important for survival under anaerobic energy stress conditions. PA4352 belongs to the universal stress protein (Usp) superfamily and harbors two Usp domains in tandem. In Escherichia coli, Usp-type stress proteins are involved in survival during aerobic growth arrest and under various other stresses. A P. aeruginosa PA4352 knockout mutant was tested for survival under several stress conditions. We found a decrease in viability of this mutant compared to the P. aeruginosa wild type during anaerobic energy starvation caused by the missing electron acceptors oxygen and nitrate. Consistent with this phenotype under anaerobic conditions, the PA4352 knockout mutant was also highly sensitive to carbonyl cyanide m-chlorophenylhydrazone, the chemical uncoupler of the electron transport chain. Primer extension experiments identified two promoters upstream of the PA4352 gene. One promoter is activated in response to oxygen limitation by the oxygen-sensing regulatory protein Anr. The center of a putative Anr binding site was identified 41.5 bp upstream of the transcriptional start site. The second promoter is active only in the stationary phase, however, independently of RpoS, RelA, or quorum sensing. This is the second P. aeruginosa Usp-type stress protein that we have identified as important for survival under anaerobic conditions, which resembles the environment during persistent infection.
Collapse
Affiliation(s)
- Nelli Boes
- Institute of Microbiology, Technical University Braunschweig, Spielmannstr. 7D-38106, Braunschweig, Germany
| | | | | | | | | |
Collapse
|
1163
|
Murillo O, Doménech A, Garcia A, Tubau F, Cabellos C, Gudiol F, Ariza J. Efficacy of high doses of levofloxacin in experimental foreign-body infection by methicillin-susceptible Staphylococcus aureus. Antimicrob Agents Chemother 2006; 50:4011-7. [PMID: 17015630 PMCID: PMC1693998 DOI: 10.1128/aac.00523-06] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Antimicrobial efficacy in orthopedic device infections is diminished because of bacterial biofilms which express tolerance to antibiotics. Recently, the use of high doses of levofloxacin with rifampin has been recommended for staphylococcal infections. In the present study, we evaluated the efficacy of levofloxacin at doses of 50 mg/kg/day and 100 mg/kg/day (mimicking the usual and high human doses of 500 mg/day and 750 to 1,000 mg/day, respectively) and compared it to that of to linezolid, cloxacillin, vancomycin, and rifampin in a rat tissue cage model of experimental foreign-body infection by Staphylococcus aureus. The antimicrobial efficacy in vitro (by MIC, minimum bactericidal concentration, and kill curves) for logarithmic- and stationary-phase bacteria was compared with the in vivo efficacy. In vitro bactericidal activity at clinically relevant concentrations was reached by all drugs except rifampin and linezolid in the log-phase studies but only by levofloxacin in the stationary-phase studies. The bacterial count decreases from in vivo tissue cage fluids (means) for levofloxacin at 50 and 100 mg/kg/day, rifampin, cloxacillin, vancomycin, linezolid, and controls, respectively, were: -1.24, -2.26, -2.1, -1.56, -1.47, -1.15, and 0.33 (all groups versus controls, P < 0.05). Levofloxacin at 100 mg/kg/day (area under the concentration-time curve/MIC ratio, 234) was the most active therapy (P = 0.03 versus linezolid). Overall, in vivo efficacy was better predicted by stationary-phase studies, in which it reached a high correlation coefficient even if the rifampin group was excluded (r = 0.96; P < 0.05). Our results, including in vitro studies with nongrowing bacteria, pharmacodynamic parameters, and antimicrobial efficacy in experimental infection, provide good evidence to support the use of levofloxacin at high doses (750 to 1,000 mg/day), as recently recommended for treating patients with orthopedic prosthesis infections.
Collapse
Affiliation(s)
- O Murillo
- Laboratory of Experimental Infection, Infectious Diseases Service, IDIBELL, Hospital Universitari de Bellvitge, Feixa Llarga s/n, 08907 Barcelona, Spain.
| | | | | | | | | | | | | |
Collapse
|
1164
|
Goller C, Wang X, Itoh Y, Romeo T. The cation-responsive protein NhaR of Escherichia coli activates pgaABCD transcription, required for production of the biofilm adhesin poly-beta-1,6-N-acetyl-D-glucosamine. J Bacteriol 2006; 188:8022-32. [PMID: 16997959 PMCID: PMC1698181 DOI: 10.1128/jb.01106-06] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The pgaABCD operon of Escherichia coli is required for production of the biofilm adhesin poly-beta-1,6-N-acetyl-d-glucosamine (PGA). We establish here that NhaR, a DNA-binding protein of the LysR family of transcriptional regulators, activates transcription of this operon. Disruption of the nhaR gene decreased biofilm formation without affecting planktonic growth. PGA production was undetectable in an nhaR mutant strain. Expression of a pgaA'-'lacZ translational fusion was induced by NaCl and alkaline pH, but not by CaCl(2) or sucrose, in an nhaR-dependent fashion. Primer extension and quantitative real-time reverse transcription-PCR analyses further revealed that NhaR affects the steady-state level of pga mRNA. A purified recombinant NhaR protein bound specifically and with high affinity within the pgaABCD promoter region; one apparent binding site overlaps the -35 element, and a second site lies immediately upstream of the first. This protein was necessary and sufficient for activation of in vitro transcription from the pgaA promoter. These results define a novel mechanism for regulation of biofilm formation in response to environmental conditions and suggest an expanded role for NhaR in promoting bacterial survival.
Collapse
MESH Headings
- Adaptation, Physiological
- Adhesins, Bacterial/genetics
- Adhesins, Bacterial/metabolism
- Antigens, Bacterial/genetics
- Base Sequence
- Biofilms/growth & development
- Cations
- DNA, Bacterial/genetics
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- DNA-Binding Proteins/physiology
- Escherichia coli/genetics
- Escherichia coli/physiology
- Escherichia coli Proteins/genetics
- Escherichia coli Proteins/metabolism
- Escherichia coli Proteins/physiology
- Gene Expression Regulation, Bacterial
- Hydrogen-Ion Concentration
- Molecular Sequence Data
- Operon/genetics
- Promoter Regions, Genetic/physiology
- Protein Binding
- RNA, Bacterial/genetics
- RNA, Messenger/genetics
- Sodium Chloride/pharmacology
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Transcription Factors/physiology
- Transcription, Genetic
Collapse
Affiliation(s)
- Carlos Goller
- Department of Microbiology and Immunology, Emory University School of Medicine, 3105 Rollins Research Center, 1510 Clifton Rd., N.E., Atlanta, GA 30322, USA
| | | | | | | |
Collapse
|
1165
|
Ma L, Jackson KD, Landry RM, Parsek MR, Wozniak DJ. Analysis of Pseudomonas aeruginosa conditional psl variants reveals roles for the psl polysaccharide in adhesion and maintaining biofilm structure postattachment. J Bacteriol 2006; 188:8213-21. [PMID: 16980452 PMCID: PMC1698210 DOI: 10.1128/jb.01202-06] [Citation(s) in RCA: 279] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The ability to form biofilms in the airways of people suffering from cystic fibrosis is a critical element of Pseudomonas aeruginosa pathogenesis. The 15-gene psl operon encodes a putative polysaccharide that plays an important role in biofilm initiation in nonmucoid P. aeruginosa strains. Biofilm initiation by a P. aeruginosa PAO1 strain with disruption of pslA and pslB (DeltapslAB) was severely compromised, indicating that psl has a role in cell-surface interactions. In this study, we investigated the adherence properties of this DeltapslAB mutant using biotic surfaces (epithelial cells and mucin-coated surfaces) and abiotic surfaces. Our results showed that psl is required for attachment to a variety of surfaces, independent of the carbon source. To study the potential roles of Psl apart from attachment, we generated a psl-inducible P. aeruginosa strain (Deltapsl/p(BAD)-psl) by replacing the psl promoter region with araC-p(BAD), so that expression of psl could be controlled by addition of arabinose. Analysis of biofilms formed by the Deltapsl/p(BAD)-psl strain indicated that expression of the psl operon is required to maintain the biofilm structure at steps postattachment. Overproduction of the Psl polysaccharide led to enhanced cell-surface and intercellular adhesion of P. aeruginosa. This translated into significant changes in the architecture of the biofilm. We propose that Psl has an important role in P. aeruginosa adhesion, which is critical for initiation and maintenance of the biofilm structure.
Collapse
Affiliation(s)
- Luyan Ma
- Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | | | | | | | | |
Collapse
|
1166
|
Singh R, Paul D, Jain RK. Biofilms: implications in bioremediation. Trends Microbiol 2006; 14:389-97. [PMID: 16857359 DOI: 10.1016/j.tim.2006.07.001] [Citation(s) in RCA: 395] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2005] [Revised: 06/12/2006] [Accepted: 07/05/2006] [Indexed: 11/16/2022]
Abstract
Biofilms are assemblages of single or multiple populations that are attached to abiotic or biotic surfaces through extracellular polymeric substances. Gene expression in biofilm cells differs from planktonic stage expression and these differentially expressed genes regulate biofilm formation and development. Biofilm systems are especially suitable for the treatment of recalcitrant compounds because of their high microbial biomass and ability to immobilize compounds. Bioremediation is also facilitated by enhanced gene transfer among biofilm organisms and by the increased bioavailability of pollutants for degradation as a result of bacterial chemotaxis. Strategies for improving bioremediation efficiency include genetic engineering to improve strains and chemotactic ability, the use of mixed population biofilms and optimization of physico-chemical conditions. Here, we review the formation and regulation of biofilms, the importance of gene transfer and discuss applications of biofilm-mediated bioremediation processes.
Collapse
Affiliation(s)
- Rajbir Singh
- Institute of Microbial Technology, Sector 39-A, Chandigarh-160036, India
| | | | | |
Collapse
|
1167
|
Cirioni O, Giacometti A, Ghiselli R, Bergnach C, Orlando F, Mocchegiani F, Silvestri C, Licci A, Skerlavaj B, Zanetti M, Saba V, Scalise G. Pre-treatment of central venous catheters with the cathelicidin BMAP-28 enhances the efficacy of antistaphylococcal agents in the treatment of experimental catheter-related infection. Peptides 2006; 27:2104-2110. [PMID: 16621147 DOI: 10.1016/j.peptides.2006.03.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2005] [Revised: 03/03/2006] [Accepted: 03/07/2006] [Indexed: 10/24/2022]
Abstract
An in vitro antibiotic susceptibility assay for Staphylococcus aureus biofilms developed on 96-well polystyrene tissue culture plates was performed to elucidate the activity of the 27 residues cathelicidin peptide BMAP-28, quinupristin/dalfopristin (Q/D), linezolid, and vancomycin. Efficacy studies were performed in a rat model of staphylococcal CVC infection. Silastic catheters were implanted into the superior cava. Twenty-four hours after implantation the catheters were filled with BMAP-28. Thirty minutes later rats were challenged via the CVC with 1.0x10(6) CFU of S. aureus strain Smith diffuse. Administration of antibiotics into the CVC at a concentration equal to the MBC observed using adherent cells, or at a much higher concentration (1024 microg/mL) began 24 h later. The inhibition activities of all antibiotics against adherent bacteria were at least two-four-fold lower that against freely growing cells. When antibiotics were used in BMAP-28 pre-treated wells, they showed higher activities. The in vivo studies showed that when CVCs were pre-treated with BMAP-28 or with a high dose of antibiotics, biofilm bacterial load was reduced from 10(7) to 10(3) CFU/mL and bacteremia reduced from 10(3) to 10(1) CFU/mL. When CVCs were treated with both BMAP-28 and antibiotics, biofilm bacterial load was further decreased to 10(1) CFU/mL and bacteremia was not detected. These results suggest that CVC pre-treated with BMAP-28 represents an attractive choice for the treatment of device-related infections caused by staphylococci.
Collapse
Affiliation(s)
- Oscar Cirioni
- Institute of Infectious Diseases and Public Health, Università Politecnica delle Marche, Ancona, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1168
|
Burmølle M, Webb JS, Rao D, Hansen LH, Sørensen SJ, Kjelleberg S. Enhanced biofilm formation and increased resistance to antimicrobial agents and bacterial invasion are caused by synergistic interactions in multispecies biofilms. Appl Environ Microbiol 2006; 72:3916-23. [PMID: 16751497 PMCID: PMC1489630 DOI: 10.1128/aem.03022-05] [Citation(s) in RCA: 452] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Most biofilms in their natural environments are likely to consist of consortia of species that influence each other in synergistic and antagonistic manners. However, few reports specifically address interactions within multispecies biofilms. In this study, 17 epiphytic bacterial strains, isolated from the surface of the marine alga Ulva australis, were screened for synergistic interactions within biofilms when present together in different combinations. Four isolates, Microbacterium phyllosphaerae, Shewanella japonica, Dokdonia donghaensis, and Acinetobacter lwoffii, were found to interact synergistically in biofilms formed in 96-well microtiter plates: biofilm biomass was observed to increase by >167% in biofilms formed by the four strains compared to biofilms composed of single strains. When exposed to the antibacterial agent hydrogen peroxide or tetracycline, the relative activity (exposed versus nonexposed biofilms) of the four-species biofilm was markedly higher than that in any of the single-species biofilms. Moreover, in biofilms established on glass surfaces in flow cells and subjected to invasion by the antibacterial protein-producing Pseudoalteromonas tunicata, the four-species biofilms resisted invasion to a greater extent than did the biofilms formed by the single species. Replacement of each strain by its cell-free culture supernatant suggested that synergy was dependent both on species-specific physical interactions between cells and on extracellular secreted factors or less specific interactions. In summary, our data strongly indicate that synergistic effects promote biofilm biomass and resistance of the biofilm to antimicrobial agents and bacterial invasion in multispecies biofilms.
Collapse
Affiliation(s)
- Mette Burmølle
- Department of Microbiology, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | |
Collapse
|
1169
|
Boddicker JD, Anderson RA, Jagnow J, Clegg S. Signature-tagged mutagenesis of Klebsiella pneumoniae to identify genes that influence biofilm formation on extracellular matrix material. Infect Immun 2006; 74:4590-7. [PMID: 16861646 PMCID: PMC1539622 DOI: 10.1128/iai.00129-06] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Klebsiella pneumoniae causes urinary tract infections, respiratory tract infections, and septicemia in susceptible individuals. Strains of Klebsiella frequently produce extended-spectrum beta-lactamases, and infections with these strains can lead to relatively high mortality rates (approximately 15%). Other virulence factors include production of an antiphagocytic capsule and outer membrane lipopolysaccharide (LPS), which mediates serum resistance, as well as fimbriae on the surface of the bacteria. Type 1 fimbriae mediate adherence to many types of epithelial cells and may facilitate adherence of the bacteria to the bladder epithelium. Type 3 fimbriae can bind in vitro to the extracellular matrix of urinary and respiratory tissues, suggesting that they mediate binding to damaged epithelial surfaces. In addition, type 3 fimbriae are required for biofilm formation by Klebsiella pneumoniae on plastics and human extracellular matrix; thus, they may facilitate the formation of treatment-resistant biofilm on indwelling plastic devices, such as catheters and endotracheal tubing. The presence of these devices may cause tissue damage, allowing Klebsiella to grow as a biofilm on exposed tissue basement membrane components. Though in vivo biofilm growth may be an important step in the infection process, little is known about the genetic factors required for biofilm formation by Klebsiella pneumoniae. Thus, we performed signature-tagged mutagenesis to identify factors produced by K. pneumoniae strain 43816 that are required for biofilm formation. We identified mutations in the cps capsule gene cluster, previously unidentified transcriptional regulators, fimbrial, and sugar phosphotransferase homologues, as well as genetic loci of unknown function, that affect biofilm formation.
Collapse
Affiliation(s)
- Jennifer D Boddicker
- Department of Microbiology, College of Medicine, University of Iowa, Iowa City, Iowa 52242, USA
| | | | | | | |
Collapse
|
1170
|
Melstrom KA, Smith JW, Gamelli RL, Shankar R. New perspectives for a new century: implications of pathogen responses for the future of antimicrobial therapy. J Burn Care Res 2006; 27:251-64. [PMID: 16679890 DOI: 10.1097/01.bcr.0000216291.68192.54] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Although the discovery of new classes of antibiotics has lagged behind in the last three decades, the incidence of life-threatening nosocomial infections that are resistant to multiple antibacterial agents has increased steadily. Recent advances in bacterial pathogenicity through the identification of a number of virulence factors and the bacterial genetics behind it have opened the way to a clearer understanding of the pathogen-host relationship. Bacteria communicate with each other through specific signaling chemicals to act as a community rather than individual cells to achieve a critical density or a "quorum." Establishment of quorum is the initiating signal for turning on a variety of virulence factors essential for the pathogenicity and dissemination of pathogens through the host. Pathogenic bacteria use a variety of biochemical mediators, collectively called "virulence factors," to invade and attack host tissues and to avoid detection and elimination by the host immune system. Delineating the specific responses the host immune system elicits in response to specific virulence factors and quorum-sensing molecules is essential to the development of new diagnostic methods for early detection of an infection and the prognosis to a given antibacterial therapy. Identification of inhibitors of virulence factors will represent new antimicrobial therapeutic modalities, and this can be used synergistically with current antibiotic therapy because they act through independent prokaryotic pathways to inhibit bacterial growth and survival.
Collapse
Affiliation(s)
- Kurt A Melstrom
- Department of Surgery and Burn & Shock Trauma Institute, Loyola University Medical Center, Maywood, Illinois 60153, USA
| | | | | | | |
Collapse
|
1171
|
Nobile CJ, Andes DR, Nett JE, Smith FJ, Yue F, Phan QT, Edwards JE, Filler SG, Mitchell AP. Critical role of Bcr1-dependent adhesins in C. albicans biofilm formation in vitro and in vivo. PLoS Pathog 2006; 2:e63. [PMID: 16839200 PMCID: PMC1487173 DOI: 10.1371/journal.ppat.0020063] [Citation(s) in RCA: 398] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2006] [Accepted: 05/12/2006] [Indexed: 11/27/2022] Open
Abstract
The fungal pathogen Candida albicans is frequently associated with catheter-based infections because of its ability to form resilient biofilms. Prior studies have shown that the transcription factor Bcr1 governs biofilm formation in an in vitro catheter model. However, the mechanistic role of the Bcr1 pathway and its relationship to biofilm formation in vivo are unknown. Our studies of biofilm formation in vitro indicate that the surface protein Als3, a known adhesin, is a key target under Bcr1 control. We show that an als3/als3 mutant is biofilm-defective in vitro, and that ALS3 overexpression rescues the biofilm defect of the bcr1/bcr1 mutant. We extend these findings with an in vivo venous catheter model. The bcr1/bcr1 mutant is unable to populate the catheter surface, though its virulence suggests that it has no growth defect in vivo. ALS3 overexpression rescues the bcr1/bcr1 biofilm defect in vivo, thus arguing that Als3 is a pivotal Bcr1 target in this setting. Surprisingly, the als3/als3 mutant forms a biofilm in vivo, and we suggest that additional Bcr1 targets compensate for the Als3 defect in vivo. Indeed, overexpression of Bcr1 targets ALS1, ECE1, and HWP1 partially restores biofilm formation in a bcr1/bcr1 mutant background in vitro, though these genes are not required for biofilm formation in vitro. Our findings demonstrate that the Bcr1 pathway functions in vivo to promote biofilm formation, and that Als3-mediated adherence is a fundamental property under Bcr1 control. Known adhesins Als1 and Hwp1 also contribute to biofilm formation, as does the novel protein Ece1. The formation of biofilms (surface-attached microbial communities) on implanted medical devices such as catheters is a major cause of fungal and bacterial infections. Prior studies of the fungal pathogen Candida albicans have shown that the regulator Bcr1 is required for biofilm formation in vitro, but the mechanism through which it promotes biofilm formation and its significance for biofilm formation in vivo was uncertain. The authors demonstrate that Bcr1 is required for biofilm formation in vivo in a rat model of catheter-based infection. Manipulation of Bcr1 target genes through mutation and gene overexpression shows that the known surface adhesin Als3 has a pivotal role in biofilm formation and that adhesins Als1 and Hwp1 also contribute to biofilm formation. The results thus indicate that adherence is the key property regulated by Bcr1 and highlight a group of adhesins as potential therapeutic targets.
Collapse
Affiliation(s)
- Clarissa J Nobile
- Department of Microbiology, Columbia University, New York, New York, United States of America
- Biological Sciences Program, Department of Biological Sciences, Columbia University, New York, New York, United States of America
| | - David R Andes
- Department of Medicine, Section of Infectious Diseases, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Jeniel E Nett
- Department of Medicine, Section of Infectious Diseases, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Frank J Smith
- Department of Microbiology, Columbia University, New York, New York, United States of America
| | - Fu Yue
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Quynh-Trang Phan
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - John E Edwards
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California, United States of America
- The David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Scott G Filler
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California, United States of America
- The David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Aaron P Mitchell
- Department of Microbiology, Columbia University, New York, New York, United States of America
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
1172
|
|
1173
|
Paddick JS, Brailsford SR, Rao S, Soares RF, Kidd EAM, Beighton D, Homer KA. Effect of biofilm growth on expression of surface proteins of Actinomyces naeslundii genospecies 2. Appl Environ Microbiol 2006; 72:3774-9. [PMID: 16672534 PMCID: PMC1472377 DOI: 10.1128/aem.72.5.3774-3779.2006] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The predominant surface proteins of biofilm and planktonic Actinomyces naeslundii, a primary colonizer of the tooth surface, were examined. Seventy-nine proteins (the products of 52 genes) were identified in biofilm cells, and 30 of these, including adhesins, chaperones, and stress-response proteins, were significantly up-regulated relative to planktonic cells.
Collapse
Affiliation(s)
- James S Paddick
- Department of Microbiology, King's College Dental Institute at Guy's, King's College and St. Thomas' Hospitals, Floor 17, Guy's Tower, Guy's Hospital, London SE1 9RT, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
1174
|
Gallaher TK, Wu S, Webster P, Aguilera R. Identification of biofilm proteins in non-typeable Haemophilus Influenzae. BMC Microbiol 2006; 6:65. [PMID: 16854240 PMCID: PMC1559630 DOI: 10.1186/1471-2180-6-65] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2006] [Accepted: 07/19/2006] [Indexed: 11/18/2022] Open
Abstract
Background Non-typeable Haemophilus influenzae biofilm formation is implicated in a number of chronic infections including otitis media, sinusitis and bronchitis. Biofilm structure includes cells and secreted extracellular matrix that is "slimy" and believed to contribute to the antibiotic resistant properties of biofilm bacteria. Components of biofilm extracellular matrix are largely unknown. In order to identify such biofilm proteins an ex-vivo biofilm of a non-typeable Haemophilus influenzae isolate, originally from an otitis media patent, was produced by on-filter growth. Extracellular matrix fraction was subjected to proteomic analysis via LC-MS/MS to identify proteins. Results 265 proteins were identified in the extracellular matrix sample. The identified proteins were analyzed for COG grouping and predicted cellular location via the TMHMM and SignalP predictive algorithms. The most over-represented COG groups identified compared to their frequency in the Haemophilus influenzae genome were cell motility and secretion (group N) followed by ribosomal proteins of group J. A number of hypothetical or un-characterized proteins were observed, as well as proteins previously implicated in biofilm function. Conclusion This study represents an initial approach to identifying and cataloguing numerous proteins associated with biofilm structure. The approach can be applied to biofilms of other bacteria to look for commonalities of expression and obtained information on biofilm protein expression can be used in multidisciplinary approaches to further understand biofilm structure and function.
Collapse
Affiliation(s)
- Timothy K Gallaher
- Proteomic Core Facility, School of Pharmacy, Health Sciences Campus, University of Southern California, Los Angeles, CA 90033, USA
| | - Siva Wu
- Ahmanson Advanced Electron Microscopy and Imaging Center, House Ear Institute, 2100 West Third Street, Los Angeles, CA 90057, USA
| | - Paul Webster
- Ahmanson Advanced Electron Microscopy and Imaging Center, House Ear Institute, 2100 West Third Street, Los Angeles, CA 90057, USA
| | - Rodrigo Aguilera
- Proteomic Core Facility, School of Pharmacy, Health Sciences Campus, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
1175
|
Kalmokoff M, Lanthier P, Tremblay TL, Foss M, Lau PC, Sanders G, Austin J, Kelly J, Szymanski CM. Proteomic analysis of Campylobacter jejuni 11168 biofilms reveals a role for the motility complex in biofilm formation. J Bacteriol 2006; 188:4312-20. [PMID: 16740937 PMCID: PMC1482957 DOI: 10.1128/jb.01975-05] [Citation(s) in RCA: 208] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Campylobacter jejuni remains the leading cause of bacterial gastroenteritis in developed countries, and yet little is known concerning the mechanisms by which this fastidious organism survives within its environment. We have demonstrated that C. jejuni 11168 can form biofilms on a variety of surfaces. Proteomic analyses of planktonic and biofilm-grown cells demonstrated differences in protein expression profiles between the two growth modes. Proteins involved in the motility complex, including the flagellins (FlaA, FlaB), the filament cap (FliD), the basal body (FlgG, FlgG2), and the chemotactic protein (CheA), all exhibited higher levels of expression in biofilms than found in stationary-phase planktonic cells. Additional proteins with enhanced expression included those involved in the general (GroEL, GroES) and oxidative (Tpx, Ahp) stress responses, two known adhesins (Peb1, FlaC), and proteins involved in biosynthesis, energy generation, and catabolic functions. An aflagellate flhA mutant not only lost the ability to attach to a solid matrix and form a biofilm but could no longer form a pellicle at the air-liquid interface of a liquid culture. Insertional inactivation of genes that affect the flagellar filament (fliA, flaA, flaB, flaG) or the expression of the cell adhesin (flaC) also resulted in a delay in pellicle formation. These findings demonstrate that the flagellar motility complex plays a crucial role in the initial attachment of C. jejuni 11168 to solid surfaces during biofilm formation as well as in the cell-to-cell interactions required for pellicle formation. Continued expression of the motility complex in mature biofilms is unusual and suggests a role for the flagellar apparatus in the biofilm phenotype.
Collapse
Affiliation(s)
- Martin Kalmokoff
- Health Canada Bureau of Microbial Hazards, Ottawa, Ontario K1A 0L2
| | | | | | | | | | | | | | | | | |
Collapse
|
1176
|
Johnson EM, Flannagan SE, Sedgley CM. Coaggregation interactions between oral and endodontic Enterococcus faecalis and bacterial species isolated from persistent apical periodontitis. J Endod 2006; 32:946-50. [PMID: 16982270 DOI: 10.1016/j.joen.2006.03.023] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2006] [Revised: 03/26/2006] [Accepted: 03/27/2006] [Indexed: 11/21/2022]
Abstract
Interactions between Enterococcus faecalis and other species found in root canal infections might be important for the development and persistence of periapical disease. The aim of this study was to investigate the coaggregation interactions between E. faecalis clinical isolates and species previously shown to survive and induce apical periodontitis in monkeys: Peptostreptococcus anaerobius, Prevotella oralis, Fusobacterium nucleatum, and Streptococcus anginosus. Intergeneric coaggregation assays were conducted in duplicate with observations scored immediately at 0 h, 1 h and 24 h after mixing of combinations of strains. All E. faecalis strains (n = 53) coaggregated with F. nucleatum; E. faecalis did not coaggregate with P. anaerobius or S. anginosus. One strain, E. faecalis E1, coaggregated with P. oralis, with aggregates visible at 1 h. Coaggregation interactions between E. faecalis and F. nucleatum observed in this study suggest a potential role for this combination in endodontic infections.
Collapse
Affiliation(s)
- Erika M Johnson
- Department of Cariology, Restorative Sciences and Endodontics, The University of Michigan, School of Dentistry, Ann Arbor, Michigan 48109-1078, USA
| | | | | |
Collapse
|
1177
|
Affiliation(s)
- Harold C Slavkin
- School of Dentistry, University of Southern California, Los Angeles, California, USA
| | | |
Collapse
|
1178
|
Slinger R, Chan F, Ferris W, Yeung SW, St Denis M, Gaboury I, Aaron SD. Multiple combination antibiotic susceptibility testing of nontypeable Haemophilus influenzae biofilms. Diagn Microbiol Infect Dis 2006; 56:247-53. [PMID: 16769194 DOI: 10.1016/j.diagmicrobio.2006.04.012] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2006] [Revised: 04/18/2006] [Accepted: 04/27/2006] [Indexed: 11/21/2022]
Abstract
Haemophilus influenzae is a cause of otitis media with effusion (OME). Animal models demonstrate growth of H. influenzae biofilms in OME, which may explain why OME does not respond well to conventional antibiotic therapy. Using a previously developed in vitro model, we performed H. influenzae susceptibility studies to see if H. influenzae biofilm cultures were more resistant to antibiotics than planktonic (broth) cultures, and to determine which antibiotics were most effective against H. influenzae biofilms. H. influenzae isolates were grown as biofilms on polystyrene pins. Biofilm and planktonic minimum inhibitory concentrations (MICs) were measured for 8 antibiotics, and multiple combination testing was performed with 66 groupings of 1, 2, or 3 antibiotics. We found that biofilm cultures were more resistant to antibiotics than planktonic ones. Antibiotic combinations containing rifampin and ciprofloxacin were most effective against biofilms. Biofilm testing reveals differences in effectiveness among antibiotics not apparent from conventional susceptibility testing, and suggests novel antibiotic regimens that could be studied for treatment of OME.
Collapse
Affiliation(s)
- Robert Slinger
- Department of Pediatrics, Ottawa, Ontario, Canada K1H 8L1.
| | | | | | | | | | | | | |
Collapse
|
1179
|
Peschel A, Sahl HG. The co-evolution of host cationic antimicrobial peptides and microbial resistance. Nat Rev Microbiol 2006; 4:529-36. [PMID: 16778838 DOI: 10.1038/nrmicro1441] [Citation(s) in RCA: 785] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Endogenous cationic antimicrobial peptides (CAMPs) are among the most ancient and efficient components of host defence. It is somewhat of an enigma that bacteria have not developed highly effective CAMP-resistance mechanisms, such as those that inhibit many therapeutic antibiotics. Here, we propose that CAMPs and CAMP-resistance mechanisms have co-evolved, leading to a transient host-pathogen balance that has shaped the existing CAMP repertoire. Elucidating the underlying principles of this process could help in the development of more sustainable antibiotics.
Collapse
Affiliation(s)
- Andreas Peschel
- Cellular and Molecular Microbiology Division, Medical Microbiology and Hygiene Department, University of Tübingen, 72076 Tübingen, Germany.
| | | |
Collapse
|
1180
|
Burton E, Gawande PV, Yakandawala N, LoVetri K, Zhanel GG, Romeo T, Friesen AD, Madhyastha S. Antibiofilm activity of GlmU enzyme inhibitors against catheter-associated uropathogens. Antimicrob Agents Chemother 2006; 50:1835-40. [PMID: 16641457 PMCID: PMC1472218 DOI: 10.1128/aac.50.5.1835-1840.2006] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The colonization of uropathogenic bacteria on urinary catheters resulting in biofilm formation frequently leads to the infection of surrounding tissue and often requires removal of the catheter. Infections associated with biofilms are difficult to treat since they may be more than 1,000 times more resistant to antibiotics than their planktonic counterparts. We have developed an antibiofilm composition comprising an N-acetyl-D-glucosamine-1-phosphate acetyltransferase (GlmU) inhibitor and protamine sulfate, a cationic polypeptide. The antibiofilm activity of GlmU inhibitors, such as iodoacetamide (IDA), N-ethyl maleimide (NEM), and NEM analogs, including N-phenyl maleimide, N,N'-(1,2-phenylene)dimaleimide (oPDM), and N-(1-pyrenyl)maleimide (PyrM), was tested against that of catheter-associated uropathogens. Both IDA and NEM inhibited biofilm formation in Escherichia coli. All NEM analogs showed antibiofilm activity against clinical isolates of E. coli, Pseudomonas aeruginosa, Klebsiella pneumoniae, Staphylococcus epidermidis, and Enterococcus faecalis. The combination of oPDM with protamine sulfate (PS) enhanced its antibiofilm activity and reduced its effective concentration to as low as 12.5 microM. In addition, we found that the in vitro inhibitory activity of oPDM-plus-PS-coated silicone catheters against P. aeruginosa and S. epidermidis colonization was superior to that of catheters coated with silver hydrogel. Confocal scanning laser microscopy further confirmed that the oPDM-plus-PS-coated silicone catheters were almost free from bacterial colonization. Thus, a broad-spectrum antibiofilm composition comprising a GlmU inhibitor and protamine sulfate shows promise for use in anti-infective coatings for medical devices, including urinary catheters.
Collapse
Affiliation(s)
- Euan Burton
- Kane Biotech Inc., Winnipeg, MB, Canada, Medical Microbiology, University of Manitoba, Health Science Centre, MS673-820 Sherbrook St., Winnipeg, MB, Canada R3A 1R9, Department of Microbiology and Immunology, Emory University School of Medicine, 3105 Rollins Research Centre, 1510 Clifton Rd. NE, Atlanta, Georgia 30322, Medicure Inc., 4-1200 Waverley Street, Winnipeg, MB, Canada, R3T 0P4
| | - Purushottam V. Gawande
- Kane Biotech Inc., Winnipeg, MB, Canada, Medical Microbiology, University of Manitoba, Health Science Centre, MS673-820 Sherbrook St., Winnipeg, MB, Canada R3A 1R9, Department of Microbiology and Immunology, Emory University School of Medicine, 3105 Rollins Research Centre, 1510 Clifton Rd. NE, Atlanta, Georgia 30322, Medicure Inc., 4-1200 Waverley Street, Winnipeg, MB, Canada, R3T 0P4
| | - Nandadeva Yakandawala
- Kane Biotech Inc., Winnipeg, MB, Canada, Medical Microbiology, University of Manitoba, Health Science Centre, MS673-820 Sherbrook St., Winnipeg, MB, Canada R3A 1R9, Department of Microbiology and Immunology, Emory University School of Medicine, 3105 Rollins Research Centre, 1510 Clifton Rd. NE, Atlanta, Georgia 30322, Medicure Inc., 4-1200 Waverley Street, Winnipeg, MB, Canada, R3T 0P4
| | - Karen LoVetri
- Kane Biotech Inc., Winnipeg, MB, Canada, Medical Microbiology, University of Manitoba, Health Science Centre, MS673-820 Sherbrook St., Winnipeg, MB, Canada R3A 1R9, Department of Microbiology and Immunology, Emory University School of Medicine, 3105 Rollins Research Centre, 1510 Clifton Rd. NE, Atlanta, Georgia 30322, Medicure Inc., 4-1200 Waverley Street, Winnipeg, MB, Canada, R3T 0P4
| | - George G. Zhanel
- Kane Biotech Inc., Winnipeg, MB, Canada, Medical Microbiology, University of Manitoba, Health Science Centre, MS673-820 Sherbrook St., Winnipeg, MB, Canada R3A 1R9, Department of Microbiology and Immunology, Emory University School of Medicine, 3105 Rollins Research Centre, 1510 Clifton Rd. NE, Atlanta, Georgia 30322, Medicure Inc., 4-1200 Waverley Street, Winnipeg, MB, Canada, R3T 0P4
| | - Tony Romeo
- Kane Biotech Inc., Winnipeg, MB, Canada, Medical Microbiology, University of Manitoba, Health Science Centre, MS673-820 Sherbrook St., Winnipeg, MB, Canada R3A 1R9, Department of Microbiology and Immunology, Emory University School of Medicine, 3105 Rollins Research Centre, 1510 Clifton Rd. NE, Atlanta, Georgia 30322, Medicure Inc., 4-1200 Waverley Street, Winnipeg, MB, Canada, R3T 0P4
| | - Albert D. Friesen
- Kane Biotech Inc., Winnipeg, MB, Canada, Medical Microbiology, University of Manitoba, Health Science Centre, MS673-820 Sherbrook St., Winnipeg, MB, Canada R3A 1R9, Department of Microbiology and Immunology, Emory University School of Medicine, 3105 Rollins Research Centre, 1510 Clifton Rd. NE, Atlanta, Georgia 30322, Medicure Inc., 4-1200 Waverley Street, Winnipeg, MB, Canada, R3T 0P4
| | - Srinivasa Madhyastha
- Kane Biotech Inc., Winnipeg, MB, Canada, Medical Microbiology, University of Manitoba, Health Science Centre, MS673-820 Sherbrook St., Winnipeg, MB, Canada R3A 1R9, Department of Microbiology and Immunology, Emory University School of Medicine, 3105 Rollins Research Centre, 1510 Clifton Rd. NE, Atlanta, Georgia 30322, Medicure Inc., 4-1200 Waverley Street, Winnipeg, MB, Canada, R3T 0P4
- Corresponding author. Mailing address: Kane Biotech Inc., 5-1250 Waverley Street, Winnipeg, MB, Canada R3T 6C6. Phone: (204) 453-1301, ext. 235. Fax: (204) 453-1314. E-mail:
| |
Collapse
|
1181
|
Cirioni O, Giacometti A, Ghiselli R, Kamysz W, Orlando F, Mocchegiani F, Silvestri C, Licci A, Chiodi L, Lukasiak J, Saba V, Scalise G. Citropin 1.1-treated central venous catheters improve the efficacy of hydrophobic antibiotics in the treatment of experimental staphylococcal catheter-related infection. Peptides 2006; 27:1210-1216. [PMID: 16289474 DOI: 10.1016/j.peptides.2005.10.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2005] [Revised: 10/05/2005] [Accepted: 10/05/2005] [Indexed: 11/30/2022]
Abstract
An in vitro antibiotic susceptibility assay for Staphylococcus aureus biofilms developed on 96-well polystyrene tissue culture plates was performed to elucidate the activity of citropin 1.1, rifampin and minocycline. Efficacy studies were performed in a rat model of staphylococcal CVC infection. Silastic catheters were implanted into the superior cava. Twenty-four hours after implantation the catheters were filled with citropin 1.1 (10 microg/mL). Thirty minutes later the rats were challenged via the CVC with 1.0 x 10(6) CFU of S. aureus strain Smith diffuse. Administration of antibiotics into the CVC (the antibiotic lock technique) began 24 h later. The study included: one control group (no CVC infection), one contaminated group that did not receive any antibiotic prophylaxis, one contaminated group that received citropin 1.1-treated CVC, two contaminated groups that received citropin 1.1-treated CVC plus rifampin and minocycline at concentrations equal to MBCs for adherent cells and 1024 microg/mL in a volume of 0.1 mL that filled the CVC and two contaminated groups that received rifampin or minocycline at the same concentrations. All catheters were explanted 7 days after implantation. Main outcome measures were: minimal inhibitory concentration (MIC), minimal bactericidal concentration (MBC), synergy studies, quantitative culture of the biofilm formed on the catheters and surrounding venous tissues, and quantitative peripheral blood cultures. MICs of conventional antibiotics against the bacteria in a biofilm were at least four-fold higher than against the freely growing planktonic cells. In contrast, when antibiotics were used on citropin 1.1 pre-treated cells they showed comparable activity against both biofilm and planktonic organisms. The in vivo studies show that when CVCs were pre-treated with citropin 1.1 or with a high dose of antibiotics, biofilm bacterial load was reduced from 10(7) to 10(3) CFU/mL and bacteremia reduced from 10(3) to 10(1) CFU/mL. When CVCs were treated both with citropin 1.1 and antibiotics, biofilm bacterial load was further reduced to 10(1) CFU/mL and bacteremia was not detected, suggesting 100% elimination of bacteremia and a log 6 reduction in biofilm load. Citropin 1.1 significantly reduces bacterial load and enhances the effect of hydrophobic antibiotics in the treatment of CVC-associated S. aureus infections.
Collapse
Affiliation(s)
- Oscar Cirioni
- Institute of Infectious Diseases and Public Health, Università Politecnica delle Marche, Ancona, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1182
|
Abstract
Antibiotic therapy for deep-seated staphylococcal infections, especially when they are associated with artificial devices used for orthopedic surgery is often associated with failure. Standard anti-staphylococcal bactericidal antibiotics, such as semi-synthetic penicillins, cephalosporins, or glycopeptides, are effective when given prophylactically in clinical conditions or experimental trials of implant-related infections. However, the efficacy of all anti-staphylococcal agents is seriously diminished on already established implant-related deep-seated infections, which then frequently require surgical implant removal to obtain a cure. The failure of antibiotic therapy to cure established staphylococcal foreign-body infections may arise in part from a broad-spectrum phenotypic tolerance expressed in vivo to different classes of antimicrobial agents. The molecular and physiological mechanisms of this in vivo tolerance remain poorly understood.
Collapse
Affiliation(s)
- Pierre Vaudaux
- Abteilung für Infektionskrankheiten, Universitätsklinikum Genf (HUG), Genf, Switzerland.
| | | |
Collapse
|
1183
|
Xi C, Marks D, Schlachter S, Luo W, Boppart SA. High-resolution three-dimensional imaging of biofilm development using optical coherence tomography. JOURNAL OF BIOMEDICAL OPTICS 2006; 11:34001. [PMID: 16822051 DOI: 10.1117/1.2209962] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
We describe the use of optical coherence tomography (OCT) for high-resolution, real-time imaging of three-dimensional structure and development of a Pseudomonas aeruginosa biofilm in a standard capillary flow-cell model. As the penetration depth of OCT can reach several millimeters in scattering samples, we are able to observe complete biofilm development on all surfaces of a 1 mm x 1 mm flow-cell. We find that biofilm growing at the bottom of the tube has more structural features including voids, outward projections, and microcolonies while the biofilm growing on the top of the tube is relatively flat and contains less structural features. Volume-rendered reconstructions of cross-sectional OCT images also reveal three-dimensional structural information. These three-dimensional OCT images are visually similar to biofilm images obtained with confocal laser scanning microscopy, but are obtained at greater depths. Based on the imaging capabilities of OCT and the biofilm imaging data obtained, OCT has potential to be used as a non-invasive, label-free, real-time, in-situ and/or in-vivo imaging modality for biofilm characterization.
Collapse
Affiliation(s)
- Chuanwu Xi
- University of Illinois at Urbana-Champaign, Department of Civil and Environmental Engineering, Biophotonics Imaging Laboratory, Beckman Institute for Advanced Science and Technology, 61801, USA
| | | | | | | | | |
Collapse
|
1184
|
Fontaine MC, Smith DGE. Microbial biofilms: Does breaking the microbes’ community spirit hold the key to beating persistent mastitis? Vet J 2006; 171:387-8. [PMID: 15908244 DOI: 10.1016/j.tvjl.2005.04.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
1185
|
Shanks RMQ, Sargent JL, Martinez RM, Graber ML, O'Toole GA. Catheter lock solutions influence staphylococcal biofilm formation on abiotic surfaces. Nephrol Dial Transplant 2006; 21:2247-55. [PMID: 16627606 DOI: 10.1093/ndt/gfl170] [Citation(s) in RCA: 151] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Microbial biofilms form on central venous catheters and may be associated with systemic infections as well as decreased dialysis efficiency due to catheter thrombosis. The most widely used anticoagulant catheter lock solution in the US is sodium heparin. We have previously shown that sodium heparin in clinically relevant concentrations enhances Staphylococcus aureus biofilm formation. In the present study, we examine the effect of several alternative catheter lock solutions on in vitro biofilm formation by laboratory and clinical isolates of S. aureus and coagulase-negative staphylococci (CNS). METHODS Lepirudin, low molecular weight heparin, tissue plasminogen activator, sodium citrate, sodium citrate with gentamicin and sodium ethylene diamine tetra-acetic acid (EDTA) were assessed for their effect on biofilm formation on polystyrene, polyurethane and silicon elastomer. RESULTS Sodium citrate at concentrations above 0.5% efficiently inhibits biofilm formation and cell growth of S. aureus and Staphylococcus epidermidis. Subinhibitory concentrations of sodium citrate significantly stimulate biofilm formation in most tested S. aureus strains, but not in CNS strains. Sodium EDTA was effective in prevention of biofilm formation as was a combination of sodium citrate and gentamicin. Low molecular weight heparin stimulated biofilm formation of S. aureus, while lepirudin and tissue plasminogen activator had little effect on S. aureus biofilm formation. CONCLUSIONS This in vitro study demonstrates that heparin alternatives, sodium citrate and sodium EDTA, can prevent the formation of S. aureus biofilms, suggesting that they may reduce the risk of biofilm-associated complications in indwelling catheters. This finding suggests a biological mechanism for the observed improvement in catheter-related outcomes in recent clinical comparisons of heparin and trisodium citrate as catheter locking solutions. A novel and potential clinically relevant finding of the present study is the observation that citrate at low levels strongly stimulates biofilm formation by S. aureus.
Collapse
Affiliation(s)
- Robert M Q Shanks
- Department of Microbiology and Immunology, Dartmouth Medical School, Hanover, NH 03755, USA
| | | | | | | | | |
Collapse
|
1186
|
Tendolkar PM, Baghdayan AS, Shankar N. Putative surface proteins encoded within a novel transferable locus confer a high-biofilm phenotype to Enterococcus faecalis. J Bacteriol 2006; 188:2063-72. [PMID: 16513736 PMCID: PMC1428127 DOI: 10.1128/jb.188.6.2063-2072.2006] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Enterococci are opportunistic pathogens and among the leading causes of nosocomial infections. Enterococcus faecalis, the dominant species among infection-derived isolates, has recently been recognized as capable of forming biofilms on abiotic surfaces in vitro as well as on indwelling medical devices. A few bacterial factors known to contribute to biofilm formation in E. faecalis have been characterized. To identify additional factors which may be important to this process, we utilized a Tn917-based insertional mutagenesis strategy to generate a mutant bank in a high-biofilm-forming E. faecalis strain, E99. The resulting mutant bank was screened for mutants exhibiting a significantly reduced ability to form biofilms. One mutant, P101D12, which showed greater than 70% reduction in its ability to form biofilms compared to the wild-type parent, was further characterized. The single Tn917 insertion in P101D12 was mapped to a gene, bee-2, encoding a probable cell wall-anchored protein. Sequence information for the region flanking bee-2 revealed that this gene was a member of a locus (termed the bee locus for biofilm enhancer in enterococcus) comprised of five genes encoding three putative cell wall-anchored proteins and two probable sortases. Contour-clamped homogeneous electric field gel and Southern hybridization analyses suggested that the bee locus is likely harbored on a large conjugative plasmid. Filter mating assays using wild-type E99 or mutant P101D12 as a donor confirmed that the bee locus could transfer conjugally at high frequency to recipient E. faecalis strains. This represents the first instance of the identification of a mobile genetic element conferring biofilm-forming property in E. faecalis.
Collapse
Affiliation(s)
- Preeti M Tendolkar
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, P.O. Box 26901, Oklahoma City, OK 73190, USA
| | | | | |
Collapse
|
1187
|
Ewald A, Glückermann SK, Thull R, Gbureck U. Antimicrobial titanium/silver PVD coatings on titanium. Biomed Eng Online 2006; 5:22. [PMID: 16556327 PMCID: PMC1435898 DOI: 10.1186/1475-925x-5-22] [Citation(s) in RCA: 132] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2005] [Accepted: 03/24/2006] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Biofilm formation and deep infection of endoprostheses is a recurrent complication in implant surgery. Post-operative infections may be overcome by adjusting antimicrobial properties of the implant surface prior to implantation. In this work we described the development of an antimicrobial titanium/silver hard coating via the physical vapor deposition (PVD) process. METHODS Coatings with a thickness of approximately 2 mum were deposited on titanium surfaces by simultaneous vaporisation of both metals in an inert argon atmosphere with a silver content of approximately 0.7-9% as indicated by energy dispersive X-ray analysis. On these surfaces microorganisms and eukaryotic culture cells were grown. RESULTS The coatings released sufficient silver ions (0.5-2.3 ppb) when immersed in PBS and showed significant antimicrobial potency against Staphylococcus epidermis and Klebsiella pneumoniae strains. At the same time, no cytotoxic effects of the coatings on osteoblast and epithelial cells were found. CONCLUSION Due to similar mechanical performance when compared to pure titanium, the TiAg coatings should be suitable to provide antimicrobial activity on load-bearing implant surfaces.
Collapse
Affiliation(s)
- Andrea Ewald
- Department for Functional Materials in Medicine and Dentistry, University of Würzburg, Pleicherwall 2, D-97070 Würzburg, Germany
| | - Susanne K Glückermann
- Department for Functional Materials in Medicine and Dentistry, University of Würzburg, Pleicherwall 2, D-97070 Würzburg, Germany
| | - Roger Thull
- Department for Functional Materials in Medicine and Dentistry, University of Würzburg, Pleicherwall 2, D-97070 Würzburg, Germany
| | - Uwe Gbureck
- Department for Functional Materials in Medicine and Dentistry, University of Würzburg, Pleicherwall 2, D-97070 Würzburg, Germany
| |
Collapse
|
1188
|
Verran J, Whitehead K. Factors affecting microbial adhesion to stainless steel and other materials used in medical devices. Int J Artif Organs 2006; 28:1138-45. [PMID: 16353120 DOI: 10.1177/039139880502801111] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The role of biofilm in medical device associated infections is well documented. Biofilms are more resistant to antibiotics than planktonic cells, these are extremely difficult to treat. Prevention strategies include efforts to insert implants under stringent aseptic conditions, and also encompass the development of novel materials which interfere with the initial attachment of microorganisms to the surface of the device. Microbial cells also attach onto hygienic surfaces in the hospital setting, and thereby pose a cross-infection problem. In this case, vigorous cleaning and sanitizing regimes may be employed in addition to any surface modifications. Many factors affect the initial attachment of organisms to inert substrata, and their subsequent retention or removal/detachment, including the physical and chemical nature and location of the substratum, the type of organic material and microorganisms potentially fouling the surface, and the nature of the interface (solid-liquid in the body; solid-air on environmental surfaces). Focusing on one factor, surface topography, it is apparent that many further variables need to be defined in order to fully understand the interactions occurring between the cell and surface. It is therefore important when modifying one substratum surface property in order to reduce adhesion, to also consider other potentially confounding factors.
Collapse
Affiliation(s)
- J Verran
- Department of Biological Sciences, Manchester Metropolitan University, Manchester, UK.
| | | |
Collapse
|
1189
|
Price-Whelan A, Dietrich LEP, Newman DK. Rethinking 'secondary' metabolism: physiological roles for phenazine antibiotics. Nat Chem Biol 2006; 2:71-8. [PMID: 16421586 DOI: 10.1038/nchembio764] [Citation(s) in RCA: 402] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Microorganisms exist in the environment as multicellular communities that face the challenge of surviving under nutrient-limited conditions. Chemical communication is an essential part of the way in which these populations coordinate their behavior, and there has been an explosion of understanding in recent years regarding how this is accomplished. Much less, however, is understood about the way these communities sustain their metabolism. Bacteria of the genus Pseudomonas are ubiquitous, and are distinguished by their production of colorful secondary metabolites called phenazines. In this article, we suggest that phenazines, which are produced under conditions of high cell density and nutrient limitation, may be important for the persistence of pseudomonads in the environment.
Collapse
Affiliation(s)
- Alexa Price-Whelan
- Division of Biology, California Institute of Technology, Pasadena, California 91125, USA
| | | | | |
Collapse
|
1190
|
ROTSTEIN ILAN, SIMON JAMESH. The endo-perio lesion: a critical appraisal of the disease condition. ACTA ACUST UNITED AC 2006. [DOI: 10.1111/j.1601-1546.2006.00211.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
1191
|
|
1192
|
Abstract
Microbes cohabit our planet and are engaged in a struggle for survival though on a microscopic scale. This endeavor allows them to develop and devise means for survival and proliferation. One such strategy is the formation of biofilms leading to establishment of a protected community. Such multi-communities may consist of harmful and pathogenic microbes, and they may cause economic problems and threats to human health. Biofilms are formed when microorganisms are typically attached to support surfaces. Biofilm-associated cells are sessile and differentiated from their suspended counterparts by generation of an extracellular polymeric substance matrix, reduced growth rates, and the up- and downregulation of specific genes. Biofilm formation is a complex process regulated by diverse characteristics of the growth medium, substratum, and cell surface. Development of strategies to control or prevent biofilms requires a thorough understanding of the biofilm development process. Biofilm research has witnessed exponential growth, and exciting findings have been reported. This has led us to visualize some previously un-thought-of and fascinating events. This article aims to provide an overview of biofilm research and associated challenges.
Collapse
Affiliation(s)
- V S Bhinu
- Center for Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Ibaraki, Japan.
| |
Collapse
|
1193
|
Mittal R, Sharma S, Chhibber S, Harjai K. Alteration in virulence characteristics of biofilm cells of Pseudomonas aeruginosa in presence of Tamm-Horsfall protein. World J Microbiol Biotechnol 2006. [DOI: 10.1007/s11274-006-9135-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
1194
|
Ojha A, Anand M, Bhatt A, Kremer L, Jacobs WR, Hatfull GF. GroEL1: a dedicated chaperone involved in mycolic acid biosynthesis during biofilm formation in mycobacteria. Cell 2006; 123:861-73. [PMID: 16325580 DOI: 10.1016/j.cell.2005.09.012] [Citation(s) in RCA: 297] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2005] [Revised: 07/27/2005] [Accepted: 09/07/2005] [Indexed: 11/20/2022]
Abstract
Mycobacteria are unusual in encoding two GroEL paralogs, GroEL1 and GroEL2. GroEL2 is essential--presumably providing the housekeeping chaperone functions--while groEL1 is nonessential, contains the attB site for phage Bxb1 integration, and encodes a putative chaperone with unusual structural features. Inactivation of the Mycobacterium smegmatis groEL1 gene by phage Bxb1 integration allows normal planktonic growth but prevents the formation of mature biofilms. GroEL1 modulates synthesis of mycolates--long-chain fatty acid components of the mycobacterial cell wall--specifically during biofilm formation and physically associates with KasA, a key component of the type II Fatty Acid Synthase involved in mycolic acid synthesis. Biofilm formation is associated with elevated synthesis of short-chain (C56-C68) fatty acids, and strains with altered mycolate profiles--including an InhA mutant resistant to the antituberculosis drug isoniazid and a strain overexpressing KasA--are defective in biofilm formation.
Collapse
Affiliation(s)
- Anil Ojha
- Pittsburgh Bacteriophage Institute, Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | | | | | | | | | | |
Collapse
|
1195
|
Garcia-Medina R, Dunne WM, Singh PK, Brody SL. Pseudomonas aeruginosa acquires biofilm-like properties within airway epithelial cells. Infect Immun 2006; 73:8298-305. [PMID: 16299327 PMCID: PMC1307054 DOI: 10.1128/iai.73.12.8298-8305.2005] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Pseudomonas aeruginosa can notably cause both acute and chronic infection. While several virulence factors are implicated in the acute phase of infection, advances in understanding bacterial pathogenesis suggest that chronic P. aeruginosa infection is related to biofilm formation. However, the relationship between these two forms of disease is not well understood. Accumulating evidence indicates that, during acute infection, P. aeruginosa enters epithelial cells, a process viewed as either a host-mediated defense response or a pathogenic mechanism to avoid host-mediated killing. We investigated the possibility that epithelial cell entry during early P. aeruginosa-epithelial cell contact favors bacterial survival and is linked to chronic infection. Using electron microscopy and confocal microscopy to analyze primary culture airway epithelial cells infected with P. aeruginosa, we found that epithelial cells developed pod-like clusters of intracellular bacteria with regional variation in protein expression. Extracellular gentamicin added to the medium after acute infection led to the persistence of intracellular P. aeruginosa for at least 3 days. Importantly, compared to bacterial culture under planktonic conditions, the intracellular bacteria were insensitive to growth inhibition or killing by antibiotics that were capable of intraepithelial cell penetration. These findings suggest that P. aeruginosa can use airway epithelial cells as a sanctuary for persistence and develop a reversible antibiotic resistance phenotype characteristic of biofilm physiology that can contribute to development of chronic infection.
Collapse
Affiliation(s)
- Raquel Garcia-Medina
- Department of Internal Medicine, Washington University School of Medicine, Box 8052, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | | | | | | |
Collapse
|
1196
|
Abstract
During the past two decades, the number of antibacterials that has reached the marketplace each year has declined, whilst resistance to existing antibacterials has increased. New antibacterials are needed to replace those that have become less effective as a result of the emergence of a high level of resistance amongst target bacteria. Antibacterials are developed by targeting live multiplying whole bacterial cells, or essential bacterial molecules such as enzymes. Using these targets, libraries of natural, recombinant or chemically synthesised compounds are screened. Most existing antibacterials have been developed by creating novel analogues of established antibacterials, which are themselves derivatives of natural compounds. Recently, live non-multiplying bacteria have been used as targets. Bacteria in such a phase are much more tolerant to antibacterials than logarithmic phase organisms. Targeting of non-multiplying bacteria has the potential to yield new antibacterials that would shorten the duration of therapy. This would be more convenient for the patient, could reduce the incidence of adverse effects of treatment, and might reduce the emergence of antibacterial resistance. However, there is much to learn about non-multiplying bacteria, particularly the mechanisms that lie behind their profound antibacterial tolerance. New terminology has been proposed for susceptibility tests for antibacterial agents against non-multiplying bacteria, namely: the minimum stationary-cidal concentration and the minimum dormicidal concentration, which are defined as the minimum concentrations of drug that will kill stationary and dormant bacteria, respectively. The relationship between the antibiotic susceptibility of stationary and logarithmic phase bacteria is the stationary/logarithmic ratio. This terminology is suitable for both planktonic and biofilm cultures. In the future, it is likely that most antibacterial drug design will be based on existing antibacterial structures, but an increasing number of new molecular antibacterial structures may emerge from screening against multiplying and perhaps non-multiplying bacteria. The genomic approach has been disappointing so far, but it is still hoped that this will produce novel antibacterial agents.
Collapse
Affiliation(s)
- Anthony R M Coates
- Department of Cellular and Molecular Medicine, Medical Microbiology, Centre for Infection, St George's University of London, London, UK.
| | | |
Collapse
|
1197
|
DeRyke CA, Lee SY, Kuti JL, Nicolau DP. Optimising Dosing Strategies of Antibacterials Utilising Pharmacodynamic Principles. Drugs 2006; 66:1-14. [PMID: 16398565 DOI: 10.2165/00003495-200666010-00001] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Evolving antimicrobial resistance is of global concern. The impact of decreased susceptibility to current antibacterials coupled with the decline in the marketing of new agents with novel mechanisms of action places a tremendous burden on clinicians to appropriately use available agents. Optimising antibacterial dose administration through the use of pharmacodynamic principles can aid clinicians in accomplishing this task more effectively. Methods to achieve this include: continuous or prolonged infusion, or the use of smaller doses administered more frequently for the time-dependent beta-lactam agents; or higher, less frequent dose administration of the concentration-dependent aminoglycosides and fluoroquinolones. Pharmacodynamic breakpoints, which are predictive of clinical and/or microbiological success in the treatment of infection, have been determined for many classes of antibacterials, including the fluoroquinolones, aminoglycosides and beta-lactams. Although surpassing these values may predict efficacy, it may not prevent the development of resistance. Recent studies seek to determine the pharmacodynamic breakpoints that prevent the development of resistance. Numerous studies to this point have determined these values in fluoroquinolones in both Gram-positive and Gram-negative bacteria. However, among the other antibacterial classes, there is a lack of sufficient data. Additionally, a new term, the mutant prevention concentration, has been based on the concentrations above which resistance is unlikely to occur. Future work is needed to fully characterise these target concentrations that prevent resistance.
Collapse
Affiliation(s)
- C Andrew DeRyke
- Center for Anti-Infective Research and Development, Hartford Hospital, Hartford, Connecticut 06102, USA
| | | | | | | |
Collapse
|
1198
|
Baron C. From bioremediation to biowarfare: On the impact and mechanism of type IV secretion systems. FEMS Microbiol Lett 2005; 253:163-70. [PMID: 16239080 DOI: 10.1016/j.femsle.2005.09.030] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2005] [Revised: 09/21/2005] [Accepted: 09/21/2005] [Indexed: 11/28/2022] Open
Abstract
Type IV secretion systems are employed by a wide variety of Gram-negative microorganisms for the translocation of macromolecules across the cell envelope. The translocated substrates (proteins, protein-DNA complexes and DNA) are as diverse as the organisms on the donor and recipient side of the translocation process. Over the course of evolution, these macromolecular transporters were adapted to many different purposes, but their basic mechanism was conserved. They impact human life in various ways, as there are driving forces of horizontal gene transfer, which spreads biodegradative capabilities of environmental bacteria as well as antibiotic resistance of pathogens in hospitals. Also, they translocate toxins and other effectors, which have an effect on host cell metabolism and are essential for the virulence of bacterial pathogens. We here present recent developments of research on the mechanism of type IV secretion focusing on the energetization of transport and assembly processes, formation of the translocation channel and of surface-exposed pili, which initiate host cell interactions.
Collapse
Affiliation(s)
- Christian Baron
- McMaster University, Department of Biology, 1280 Main Street, West Hamilton, ON, Canada.
| |
Collapse
|
1199
|
Matz C, McDougald D, Moreno AM, Yung PY, Yildiz FH, Kjelleberg S. Biofilm formation and phenotypic variation enhance predation-driven persistence of Vibrio cholerae. Proc Natl Acad Sci U S A 2005; 102:16819-24. [PMID: 16267135 PMCID: PMC1283802 DOI: 10.1073/pnas.0505350102] [Citation(s) in RCA: 226] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Persistence of the opportunistic bacterial pathogen Vibrio cholerae in aquatic environments is the principal cause for seasonal occurrence of cholera epidemics. This causality has been explained by postulating that V. cholerae forms biofilms in association with animate and inanimate surfaces. Alternatively, it has been proposed that bacterial pathogens are an integral part of the natural microbial food web and thus their survival is constrained by protozoan predation. Here, we report that both explanations are interrelated. Our data show that biofilms are the protective agent enabling V. cholerae to survive protozoan grazing while their planktonic counterparts are eliminated. Grazing on planktonic V. cholerae was found to select for the biofilm-enhancing rugose phase variant, which is adapted to the surface-associated niche by the production of exopolymers. Interestingly, grazing resistance in V. cholerae biofilms was not attained by exopolymer production alone but was accomplished by the secretion of an antiprotozoal factor that inhibits protozoan feeding activity. We identified that the cell density-dependent regulator hapR controls the production of this factor in biofilms. The inhibitory effect of V. cholerae biofilms was found to be widespread among toxigenic and nontoxigenic isolates. Our results provide a mechanistic explanation for the adaptive advantage of surface-associated growth in the environmental persistence of V. cholerae and suggest an important contribution of protozoan predation in the selective enrichment of biofilm-forming strains in the out-of-host environment.
Collapse
Affiliation(s)
- Carsten Matz
- School of Biotechnology and Biomolecular Sciences, Centre for Marine Biofouling and Bio-Innovation, University of New South Wales, Sydney, Australia.
| | | | | | | | | | | |
Collapse
|
1200
|
Abstract
BACKGROUND Most microorganisms in nature attach to surfaces and form matrix-embedded biofilms. Biofilms are highly structured and spatially organized, and are often composed of consortia of interacting microorganisms, termed microbial communities, the properties of which are more than the sum of the component species. Microbial gene expression alters markedly in biofilms; organisms communicate by gene transfer and by secretion of diffusible signalling molecules. Cells in biofilms are less susceptible to antimicrobial agents. AIM AND MATERIALS & METHODS To comprehensively review the literature to determine whether dental plaque displays properties consistent with those of a typical biofilm and microbial community. RESULTS Novel microscopic and molecular techniques have demonstrated that plaque has a structured architecture with an extracellular matrix, and a diverse composition (around 50% of cells are unculturable). The constituent species communicate by gene transfer, by secreted peptides (gram-positive bacteria) and autoinducer-2 (gram-positive and gram-negative bacteria). These organisms are functionally organized for increased metabolic efficiency, greater resistance to stress and for enhanced virulence. Plaque formation has direct and indirect effects on gene expression. CONCLUSION Dental plaque displays properties that are typical of biofilms and microbial communities in general, a clinical consequence of which is a reduced susceptibility to antimicrobial agents as well as pathogenic synergism.
Collapse
Affiliation(s)
- P D Marsh
- Leeds Dental Institute, and Health Protection Agency, Centre for Emergency Preparedness and Response, Porton Down, UK.
| |
Collapse
|