Skip to main content

Confocal laser scanning microscopy analysis of S. epidermidis biofilms exposed to farnesol, vancomycin and rifampicin

Abstract

Background

Staphylococcus epidermidis is the major bacterial species found in biofilm-related infections on indwelling medical devices. Microbial biofilms are communities of bacteria adhered to a surface and surrounded by an extracellular polymeric matrix. Biofilms have been associated with increased antibiotic tolerance to the immune system. This increased resistance to conventional antibiotic therapy has lead to the search for new antimicrobial therapeutical agents. Farnesol, a quorum-sensing molecule in Candida albicans, has been described as impairing growth of several different microorganisms and we have previously shown its potential as an adjuvant in antimicrobial therapy against S. epidermidis. However, its mechanism of action in S. epidermidis is not fully known. In this work we better elucidate the role of farnesol against S: epidermidis biofilms using confocal laser scanning microscopy (CLSM).

Findings

24 h biofilms were exposed to farnesol, vancomycin or rifampicin and were analysed by CLSM, after stained with a Live/Dead stain, a known indicator of cell viability, related with cell membrane integrity. Biofilms were also disrupted by sonication and viable and cultivable cells were quantified by colony forming units (CFU) plating. Farnesol showed a similar effect as vancomycin, both causing little reduction of cell viability but at the same time inducing significant changes in the biofilm structure. On the other hand, rifampicin showed a distinct action in S. epidermidis biofilms, by killing a significant proportion of biofilm bacteria.

Conclusions

While farnesol is not very efficient at killing biofilm bacteria, it damages cell membrane, as determined by the live/dead staining, in a similar way as vancomycin. Furthermore, farnesol might induce biofilm detachment, as determined by the reduced biofilm biomass, which can partially explain the previous findings regarding its role as a possible chemotherapy adjuvant.

Findings

Background

Staphylococcus epidermidis, a normal inhabitant of human skin and mucosa, has recently emerged as a leading cause of biofilm-related infections, particularly, in patients with indwelling medical devices[1, 2] due to its ability to adhere to abiotic surfaces and to form biofilms[2, 3]. Biofilms are often defined as three-dimensional communities of microorganisms that are attached to a surface and encased in an extracellular matrix composed mainly of polysaccharides, proteins and extracellular DNA[4, 5]. The major virulence factor of S. epidermidis is biofilm formation[6] and cells in biofilms are normally more tolerant to antibiotics than planktonic cells[79], making drug resistance in a S. epidermidis biofilm-related infection a serious problem, especially in nosocomial infections[10]. S. epidermidis are also inherently resistant to host defense mechanisms[11, 12]. The increased resistance to conventional antibiotic therapy led to the search for new therapeutical agents.

Despite concerted efforts to treat biofilm-related infections with antibiotic therapy, the physical removal of the infected medical device is often necessary[13], which carries an additional economic and health cost. Therefore, novel strategies are necessary to combat biofilm-related infections to improve clinical outcomes.

Farnesol is a sesquiterpene alcohol and is the first quorum-sensing molecule described in eukaryotes[14] and produced by most Candida species[15]. Farnesol has been shown to have antimicrobial activity in many species[16], including the coagulase positive Staphylococcus aureus[17, 18] and the coagulase negative Staphyloccous epidermidis[19]. Recently, we showed that farnesol could act as an adjuvant against S. epidermidis infections, when combined with common antibiotics such as vancomycin, tetracycline and rifampicin. However, this effect was more proeminent in planktonic cells and was significantly reduced in biofilms[20]. Nevertheless, Pammi et al. had shown that combinations of farnesol with other antibiotics were also effective in reducing S. epidermidis biofilm biomass[21].

Despite the promising role of farnesol as an adjuvant in antimicrobial activity against S. epidermidis, little is known regarding its mechanism of action. We have previously shown that the biofilm matrix composition and distribution of S. epidermidis biofilms was affected by farnesol[22]. In this study we present a more in-depth analysis of biofilm structure, viability and biomass changes when in contact with farnesol, and compare it with biofilms exposed to vancomycin or rifampicin.

Material and methods

Bacterial strains and growth conditions

The biofilm-producing strain used in this study, S. epidermidis 9142,was previously described[23]. This strain was originally isolated from blood cultures from an infected central venous catheter by Mack et al.[24], Strains were grown as previously described, in Tryptic Soy Broth (TSB) and Tryptic Soy Agar (TSA) (Oxoid, Cambridge, UK)[25]. Briefly the strain was inoculated into 15 mL of TSB from TSA plates not older than 2 days and grown for 18 (± 2) h at 37°C in an orbital shaker at 130 rpm. Cells were harvested by centrifugation (for 10 min at 9500 × g and 4°C), and resuspended in TSB supplemented with 0.25% glucose (TSBG) (Fisher Scientific, Waltham, MA, US) adjusted to an optical density (640 nm) equivalent to 1 × 109 cells mL-1 and then used in the subsequent assays.

Biofilm challenge with farnesol, vancomycin or rifampicin

Biofilm challenge was done as described before[26], with some modifications. Briefly, biofilms were formed during 24 h at 37°C and 120 rpm on 96 (for killing assays) or 6-well tissue culture (for confocal analysis) plates by dispensing 4 mL of a 1 × 109 cells mL-1 cell suspension in TSBG. Then, the growth medium was removed and replaced with fresh TSBG supplemented with either 300 μM of farnesol (Sigma, St Louis, MO, US)[19], or the antibiotics at the respective peak serum concentration (vancomycin: 40 mg/L and rifampicin: 10 mg/L) (Sigma, St Louis, MO, US)[9]. Biofilms were exposed to the respective antimicrobial agents for 2, 4, 6, 8 or 24 h. After exposure, biofilms were sonicated for 10 seconds at 10 W (Ultrasonic Processor, Cole-Parmer, USA). This procedure did not reduced cell viability, as showed before[9]. The cell suspension was washed twice to prevent antibiotic carry over and resuspended in 0.9% NaCl. Viable bacteria were determined by standard colony-forming-units (CFU) plating in TSA plates. This experiment was repeated three times with duplicates.

Confocal scanning laser microscopy (CLSM) analysis of the biofilms

CSLM was performed as described before[22]. Briefly, 24 h after the biofilms were exposed to farnesol, vancomycin or rifampicin, they were washed twice with 0.9% NaCl. Biofilm cell viability was determined with live/dead staining (Molecular Probes, USA) following the manufacturer´s instructions. A negative control was used to determine the baseline threshold for dead cells, by killing the biofilm with 96% ethanol for 4 h. The plates were incubated for 20 min at room temperature in the dark. After staining, the biofilms were gently rinsed with 0.9% NaCl. The biofilm images were acquired in an OlympusTM FluoView FV1000 (Olympus, Lisboa, Portugal) confocal scanning laser microscope. Biofilms were observed using a 60x water-immersion objective (60x/1.2 W). For each condition, three independent biofilms were used. Images were acquired with 512 x 512 resolutions in at least four different regions of each surface analyzed. For biofilm maximum thickness determination, at least twenty different regions per surface were analyzed, by determining the the top and bottom layer of the biofilm, and calculating the maximum thickness of each region.

Statistical analysis

Quantitative assays were compared using unpaired t-test or one-way analysis of variance (ANOVA) by applying Levene’s test of homogeneity of variances and the Tukey multiple comparisons test. All tests were performed with a confidence level of 95%.

Results and discussion

Antibiotic resistance is a serious problem in S. epidermidis since many clinical isolates of this organism are resistant up to eight different antibiotics[10]. Farnesol is one promising candidate to be used as an adjuvant in antimicrobial chemotherapy against biofilms. Some studies have indicated a possible interaction of farnesol with cell membranes of S. aureus, resulting in a reduced cell membrane integrity[18]. We have recently shown that farnesol increased cell death by the action of tetracycline or rifampicin[20], possibly due to the permeabilization of the cell wall[22]. The same was true in combination with N-acetylcysteine[27]. However, the exact mechanism of farnesol action in S. epidermidis is not fully known. A recent study of Zhu et al.[28] revealed that farnesol induced Candida albicans apoptosis by conjugating with intracellular glutathione, inducing a strong oxidative stress and eventual cell death. However, as C. albicans biology is fundamentally different from S. epidermidis, it is not possible to infer a similar role of farnesol in S. epidermidis biofilms.

Based on our previous findings[19, 20, 22, 27], we selected two antibiotics with distinct mechanisms of action: vancomycin, that inhibits the cell wall synthesis and shows low activity against S. epidermidis biofilms, and rifampicin, a RNA synthesis inhibitor, that shows high activity against S. epidermidis biofilms and compared their effects with farnesol action on biofilm structure and cell viability. Figure1 shows a time-course study on viable cells after exposure to the three antimicrobial agents. As expected, vancomycin was not very effective in killing biofilm bacteria. In fact, as compared with the control, the only significant difference found was at 24 h of exposure. On the other hand, rifampicin was able to kill more than 1 log of bacteria in the first two hours of exposure, and more than 2.5 log after 8 h. Farnesol showed similar results as vancomycin in all time periods studied.

Figure 1
figure 1

Effect of farnesol, vancomycin, and rifampicin on viability of S. epidermidis biofilm cells versus time of treatment.* statistically different as compared with the control, at the respective time-point (p < 0.05).

CLSM has been widely used to study biofilm structure, composition and metabolism in several different microorganisms[2931]. One of the big advantages of CLSM is that it allows in-depth analysis of biological structures, without killing or damaging the biological structure[32]. Live/dead staining has been used as indicator of cell viability, as determined by the integrity of the cell wall membrane in many bacterial populations, including biofilms[19, 33]. When we analyzed the biofilm structure and viability by CLSM an interesting result was found: despite the fact that vancomycin and farnesol were not able to effectively kill biofilm bacteria, biofilm biomass was strongly reduced, as can be seen qualitatively in Figure2 or quantitatively in Figure3. In fact, the biofilm maximum depth, an indirect measure of the amount of biofilm biomass was equally reduced when treated with any of the three different antimicrobial compounds used (Figure3). For better comparison of the antimicrobials action, regions of biofilms with similar deepness were chosen (Figure2C, D, E). More images can be found in supplementary information (Additional file1, Additional file2, Additional file3 and Additional file4). It is well known that bacterial biofilms are heterogeneous communities, regarding both the three dimensional structure but also the physiology of in vitro biofilm bacteria[4]. It was recently described that S. epidermidis biofilms have a variable sub-population of cells presenting a partially damaged cell membrane. This sub-population was able to incorporate propidium iodide at low levels, but were still viable bacteria[34]. In the present study, biofilms treated with vancomycin or farnesol showed clusters of live (green) and death bacteria (red) but also a sub-population of bacteria with somewhat damaged cell membrane (yellow). Despite the fact that biofilms exposed to vancomycin or farnesol had smaller amounts of biomass, the typical S. epidermidis biofilm high density cell clusters were still detected[3538]. On the other hand, rifampicin-treated biofilms hardly presented regions of live or somewhat damage cell clusters. While live and viable bacteria were mainly dispersed in small microcolonies, the few cluster found were of dead bacteria (Figure2E).

Figure 2
figure 2

Effect of farnesol and antibiotics on biofilm structure and viability. (A) Biofilm without antimicrobial agent; (B) Negative control for live/dead staining, treated with 96% ethanol for 4 h; (C) Biofilm exposed to 300 μM farnesol for 24 h; (D) Biofilm exposed to 40 mg/L of vancomycin for 24 h; (E) Biofilm exposed to 10 mg/L of rifampicin for 24 h.

Figure 3
figure 3

Effect of farnesol and antibiotics on biofilms maximum depth. * statistically different as compared with the control (p < 0.05).

Since it is well known that biofilm cells are metabolically less active than planktonic ones[39] and some cells are in a dormant state[40], antibiotics that require active cell division, such as vancomycin, are highly hindered in such conditions. Nevertheless, the reduced biomass present suggested that both agents targeting the cell wall were able to, somehow, induce cell detachment. While S. epidermidis biofilms have been proposed to partially detach fragments of the biofilm[6] in order to potentially colonize other regions of the host, cell detachment is not yet fully understood, however some genetic regulation might be involved[41]. Recently Kaneko et al. showed that farnesol interfered with the S. aureus mevalonate pathway, which is related with cell membrane maintenance and protein anchoring, among other important cell functions[42]. S. aureus biofilm production and metabolism is similar to S. epidermidis[43]. It has been shown before that the lack of proper protein anchoring results in reduced biofilm formation in S. aureus and S. epidermidis, related with regulation of poly-N-acetyl-β-(1–6)-glucosamine deacetylation[11, 44].

Conclusions

Treatment of S. epidermidis biofilms with farnesol and vancomycin did not significantly decrease cell viability. However, they both appeared to reduce the biomass in these biofilms. Our previous reports regarding the role of farnesol as an adjuvant in antimicrobial chemotherapy can now be better explained, since released cells from the biofilms would potentially be more prone to antibiotic attack, taking in consideration that no diffusion barrier would be present in such cases[45]. Kaneko et al.[42] results and ours further provide more evidence that farnesol mechanism of action in S. epidermidis is related with the cell wall membrane integrity. We are currently exploring the role of farnesol in S. epidermidis cell membrane integrity as well as its impact on biofilm physiology.

References

  1. Ziebuhr W, Hennig S, Eckart M, Kranzler H, Batzilla C, Kozitskaya S: Nosocomial infections by Staphylococcus epidermidis: how a commensal bacterium turns into a pathogen. Int J Antimicrob Agents. 2006, 28 Suppl 1: S14-S20.

    Article  PubMed  Google Scholar 

  2. Vuong C, Otto M:Staphylococcus epidermidis infections. Microbes Infect. 2002, 4: 481-489. 10.1016/S1286-4579(02)01563-0.

    Article  PubMed  Google Scholar 

  3. Gotz F: Staphylococcus and biofilms. Mol Microbiol. 2002, 43: 1367-1378. 10.1046/j.1365-2958.2002.02827.x.

    Article  PubMed  CAS  Google Scholar 

  4. Costerton J, Lewandowski Z, Caldwell DE, Korber D, Lappin-Scott H: Microbial biofilms. Annu Rev Microbiol. 1995, 49: 711-745. 10.1146/annurev.mi.49.100195.003431.

    Article  PubMed  CAS  Google Scholar 

  5. Otto M: Staphylococcal biofilms. Curr Top Microbiol Immunol. 2008, 322: 207-228. 10.1007/978-3-540-75418-3_10.

    PubMed  CAS  PubMed Central  Google Scholar 

  6. Otto M: Staphylococcus epidermidis–the 'accidental' pathogen. Nat Rev Microbiol. 2009, 7: 555-567. 10.1038/nrmicro2182.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  7. Arciola C, Campoccia D, Montanaro L: Effects on antibiotic resistance of Staphylococcus epidermidis following adhesion to polymethylmethacrylate and to silicone surfaces. Biomaterials. 2002, 23: 1495-1502. 10.1016/S0142-9612(01)00275-7.

    Article  PubMed  CAS  Google Scholar 

  8. Szomolay B, Klapper I, Dockery J, Stewart PS: Adaptive responses to antimicrobial agents in biofilms. Environ Microbiol. 2005, 7: 1186-1191. 10.1111/j.1462-2920.2005.00797.x.

    Article  PubMed  CAS  Google Scholar 

  9. Cerca N, Martins S, Cerca F, Jefferson KK, Pier GB, Oliveira R, Azeredo J: Comparative assessment of antibiotic susceptibility of coagulase-negative staphylococci in biofilm versus planktonic culture as assessed by bacterial enumeration or rapid XTT colorimetry. J Antimicrob Chemother. 2005, 56: 331-336. 10.1093/jac/dki217.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  10. Fluit A, Schmitz F, Verhoef J: Multi-resistance to antimicrobial agents for the ten most frequently isolated bacterial pathogens. Int J Antimicrob Agents. 2001, 18: 147-160. 10.1016/S0924-8579(01)00357-0.

    Article  PubMed  CAS  Google Scholar 

  11. Vuong C, Kocianova S, Voyich JM, Yao Y, Fischer ER, DeLeo FR, Otto M: A crucial role for exopolysaccharide modification in bacterial biofilm formation, immune evasion, and virulence. J Biol Chem. 2004, 279: 54881-54886. 10.1074/jbc.M411374200.

    Article  PubMed  CAS  Google Scholar 

  12. Cerca N, Jefferson K, Oliveira R, Pier GB, Azeredo J: Comparative antibody-mediated phagocytosis of Staphylococcus epidermidis cells grown in a biofilm or in the planktonic state. Infect Immun. 2006, 74: 4849-4855. 10.1128/IAI.00230-06.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  13. Jansen B, Kristinsson K, Jansen S, Peters G, Pulverer G: In-vitro efficacy of a central venous catheter complexed with iodine to prevent bacterial colonization. J Antimicrob Chemother. 1992, 30: 135-139. 10.1093/jac/30.2.135.

    Article  PubMed  CAS  Google Scholar 

  14. Hornby J, Jensen E, Lisec A, Tasto J, Jahnke B, Shoemaker R, Dussault P, Nickerson K: Quorum sensing in the dimorphic fungus Candida albicans is mediated by farnesol. Appl Environ Microbiol. 2001, 67: 2982-2992. 10.1128/AEM.67.7.2982-2992.2001.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  15. Weber K, Sohr R, Schulz B, Fleischhacker M, Ruhnke M: Secretion of E, E-farnesol and biofilm formation in eight different Candida species. Antimicrob Agents Chemother. 2008, 52: 1859-1861. 10.1128/AAC.01646-07.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  16. Brehm-Stecher BF, Johnson EA: Sensitization of Staphylococcus aureus and Escherichia coli to antibiotics by the sesquiterpenoids nerolidol, farnesol, bisabolol, and apritone. Antimicrob Agents Chemother. 2003, 47: 3357-3360. 10.1128/AAC.47.10.3357-3360.2003.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  17. Inoue Y, Shiraishi A, Hada T, Hirose K, Hamashima H, Shimada J: The antibacterial effects of terpene alcohols on Staphylococcus aureus and their mode of action. FEMS Microbiol Lett. 2004, 237: 325-331.

    PubMed  CAS  Google Scholar 

  18. Jabra-Rizk MA, Meiller TF, James CE, Shirtliff ME: Effect of farnesol on Staphylococcus aureus biofilm formation and antimicrobial susceptibility. Antimicrob Agents Chemother. 2006, 50: 1463-1469. 10.1128/AAC.50.4.1463-1469.2006.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  19. Gomes FIA, Teixeira P, Azeredo J, Oliveira R: Effect of Farnesol on Planktonic and Biofilm Cells of Staphylococcus epidermidis. Curr Microbiol. 2009, 59: 118-122. 10.1007/s00284-009-9408-9.

    Article  PubMed  CAS  Google Scholar 

  20. Gomes FIA, Leite B, Teixeira P, Cerca N, Azeredo J, Oliveira R: Farnesol as antibiotics adjuvant in Staphylococcus epidermidis control in vitro. Am J Med Sci. 2011, 341: 191-195. 10.1097/MAJ.0b013e3181fcf138.

    Article  PubMed  Google Scholar 

  21. Pammi M, Liang R, Hicks J, Barrish J, Versalovic J: Farnesol decreases biofilms of Staphylococcus epidermidis and exhibits synergy with nafcillin and vancomycin. Pediatr Res. 2011, 70: 578-583.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  22. Gomes FIA, Teixeira P, Cerca N, Azeredo J, Oliveira R: Effect of farnesol on structure and composition of Staphylococcus epidermidis biofilm matrix. Curr Microbiol. 2011, 63: 354-359. 10.1007/s00284-011-9984-3.

    Article  PubMed  CAS  Google Scholar 

  23. Cerca N, Pier GB, Vilanova M, Oliveira R, Azeredo J: Quantitative analysis of adhesion and biofilm formation on hydrophilic and hydrophobic surfaces of clinical isolates of Staphylococcus epidermidis. Res Microbiol. 2005, 156: 506-514. 10.1016/j.resmic.2005.01.007.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  24. Mack D, Siemssen N, Laufs R: Parallel induction by glucose of adherence and a polysaccharide antigen specific for plastic-adherent Staphylococcus epidermidis: evidence for functional relation to intercellular adhesion. Infect Immun. 1992, 60: 2048-2057.

    PubMed  CAS  PubMed Central  Google Scholar 

  25. Cerca N, Pier GB, Vilanova M, Oliveira R, Azeredo J: Influence of batch or fed-batch growth on Staphylococcus epidermidis biofilm formation. Lett Appl Microbiol. 2004, 39: 420-424. 10.1111/j.1472-765X.2004.01601.x.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  26. Cerca N, Martins S, Sillankorva S, Jefferson KK, Pier GB, Oliveira R, Azeredo J: Effects of growth in the presence of subinhibitory concentrations of dicloxacillin on Staphylococcus epidermidis and Staphylococcus haemolyticus biofilms. Appl Environ Microbiol. 2005, 71 (12): 8677-8682. 10.1128/AEM.71.12.8677-8682.2005.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  27. Gomes F, Leite B, Teixeira P, Azeredo J, Oliveira R: Effect of farnesol in combination with N-acetylcysteine against Staphylococcus epidermidis planktonic and biofilm cells. Braz J Microbiol. 2012, in press

    Google Scholar 

  28. Zhu J, Krom BP, Sanglard D, Intapa C, Dawson CC, Peters BM, Shirliff ME, Jabra-Rizk MA: Farnesol-Induced Apoptosis in Candida albicans Is Mediated by Cdr1-p Extrusion and Depletion of Intracellular Glutathione. PLoS One. 2011, 6: e28830-10.1371/journal.pone.0028830.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  29. Almeida C, Azevedo NF, Santos S, Keevil CW, Vieira MJ: Discriminating multi-species populations in biofilms with peptide nucleic acid fluorescence in situ hybridization (PNA FISH). PLoS One. 2011, 6: e14786-10.1371/journal.pone.0014786.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  30. Ma Q, Zhang G, Wood TK: Escherichia coli BdcA controls biofilm dispersal in Pseudomonas aeruginosa and Rhizobium meliloti. BMC Res Notes. 2011, 4: 447-10.1186/1756-0500-4-447.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  31. Bodor AM, Jansch L, Wissing J, Wagner-Dobler I: The luxS mutation causes loosely-bound biofilms in Shewanella oneidensis. BMC Res Notes. 2011, 4: 180-10.1186/1756-0500-4-180.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  32. Neu TR, Lawrence JR: Lectin-binding analysis in biofilm systems. Methods Enzymol. 1999, 310: 145-152.

    Article  PubMed  CAS  Google Scholar 

  33. Flemming K, Klingenberg C, Cavanagh JP, Sletteng M, Stensen W, Svendsen JS, Flaegstad T: High in vitro antimicrobial activity of synthetic antimicrobial peptidomimetics against staphylococcal biofilms. J Antimicrob Chemother. 2009, 63: 136-45.

    Article  PubMed  CAS  Google Scholar 

  34. Cerca F, Franca A, Guimaraes R, Hinzmann M, Cerca N, Lobo dC, Azeredo J: Vilanova M. Modulation of poly-N-acetylglucosamine accumulation within mature Staphylococcus epidermidis biofilms grown in excess glucose. Microbiol Immunol. 2011, 55: 673-682. 10.1111/j.1348-0421.2011.00368.x.

    Article  PubMed  CAS  Google Scholar 

  35. Lawrence JR, Swerhone GD, Leppard GG, Araki T, Zhang X, West MM, Hitchcock AP: Scanning transmission X-ray, laser scanning, and transmission electron microscopy mapping of the exopolymeric matrix of microbial biofilms. Appl Environ Microbiol. 2003, 69: 5543-5554. 10.1128/AEM.69.9.5543-5554.2003.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  36. Shapiro JA, Nguyen VL, Chamberlain NR: Evidence for persisters in Staphylococcus epidermidis RP62a planktonic cultures and biofilms. J Med Microbiol. 2011, 60: 950-960. 10.1099/jmm.0.026013-0.

    Article  PubMed  CAS  Google Scholar 

  37. Pintens V, Massonet C, Merckx R, Vandecasteele S, Peetermans WE, Knobloch JK, Van Eldere J: The role of sigmaB in persistence of Staphylococcus epidermidis foreign body infection. Microbiology. 2008, 154: 2827-2836. 10.1099/mic.0.2007/015768-0.

    Article  PubMed  CAS  Google Scholar 

  38. Shapiro JA, Nguyen VL, Chamberlain NR: Evidence for persisters in Staphylococcus epidermidis RP62a planktonic cultures and biofilms. J Med Microbiol. 2011, 60: 950-960. 10.1099/jmm.0.026013-0.

    Article  PubMed  CAS  Google Scholar 

  39. Cerca F, Trigo G, Correia A, Cerca N, Azeredo J, Vilanova M: SYBR green as a fluorescent probe to evaluate the biofilm physiological state of Staphylococcus epidermidis, using flow cytometry. Can J Microbiol. 2011, 57: 850-856. 10.1139/w11-078.

    Article  PubMed  CAS  Google Scholar 

  40. Cerca F, Andrade F, Franca A, Andrade EB, Ribeiro A, Almeida AA, Cerca N, Pier G, Azeredo J, Vilanova M: Staphylococcus epidermidis biofilms with higher proportions of dormant bacteria induce a lower activation of murine macrophages. J Med Microbiol. 2011, 60: 1717-1724. 10.1099/jmm.0.031922-0.

    Article  PubMed  Google Scholar 

  41. Wang R, Khan BA, Cheung GY, Bach TH, Jameson-Lee M, Kong KF, Queck SY, Otto M: Staphylococcus epidermidis surfactant peptides promote biofilm maturation and dissemination of biofilm-associated infection in mice. J Clin Invest. 2011, 121: 238-248. 10.1172/JCI42520.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  42. Kaneko M, Togashi N, Hamashima H, Hirohara M, Inoue Y: Effect of farnesol on mevalonate pathway of Staphylococcus aureus. J Antibiot. 2011, 64: 547-549. 10.1038/ja.2011.49.

    Article  PubMed  CAS  Google Scholar 

  43. Cerca N, Brooks JL, Jefferson KK: Regulation of the intercellular adhesin locus regulator (icaR) by SarA, sigmaB, and IcaR in Staphylococcus aureus. J Bacteriol. 2008, 190: 6530-6533. 10.1128/JB.00482-08.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  44. Cerca N, Jefferson K, Pier D, Kelly-Quintos C, Maira-Litrán T, Goldmann D, Azeredo J, Pier G: Molecular basis for preferential protective efficacy of antibodies directed to the poorly-acetylated form of staphylococcal poly-N-acetyl-(1–6)-glucosamine (PNAG). Infect Immun. 2007, 75: 3406-3413. 10.1128/IAI.00078-07.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  45. Stewart PS: Diffusion in biofilms. J Bacteriol. 2003, 185: 1485-1491. 10.1128/JB.185.5.1485-1491.2003.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

Download references

Acknowledgements

FG and PT were financial supported by Fundação para a Ciência e a Tecnologia (FCT) through the grants SFRH/BD/32126/2006 and SFRH/BPD/26803/2006, respectively. SP had a BII grant from FCT attributed to CEB-IBB.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Nuno Cerca.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors contributions

NC did the confocal microscopy analysis and writing of the manuscript. FG and PT did the timelapse studies. SP and FG prepared the samples for confocal analysis. NC and RO planned the experiments. All authors read and approved the final manuscript.

Electronic supplementary material

Authors’ original submitted files for images

Below are the links to the authors’ original submitted files for images.

Authors’ original file for figure 1

Authors’ original file for figure 2

Authors’ original file for figure 3

Rights and permissions

This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Cerca, N., Gomes, F., Pereira, S. et al. Confocal laser scanning microscopy analysis of S. epidermidis biofilms exposed to farnesol, vancomycin and rifampicin. BMC Res Notes 5, 244 (2012). https://doi.org/10.1186/1756-0500-5-244

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1756-0500-5-244

Keywords