Skip to main content
  • Research Note
  • Open access
  • Published:

Anti-biofilm activity of Pseudomonas fluorescens culture supernatants on biofilm formation of Staphylococcus epidermidis 1457

Abstract

Objective

Staphylococcus epidermidis is a skin colonizer and a major cause of nosocomial infections that can lead to sepsis. It causes opportunistic infections by forming biofilms on medical devices, which are hard to control with conventional antibiotics. In an attempt to develop its biofilm inhibitors, the culture supernatant (CS) of Pseudomonas fluorescens was assessed. This study examined the effect of the CS on S. epidermidis 1457 biofilm formation, the characteristics of inhibitors in the CS, and the differential gene expression of S. epidermidis when treated with the CS.

Results

P. fluorescens CS specifically targeted the maturation stage of S. epidermidis biofilm formation while not affecting planktonic growth. RT-qPCR analysis revealed that P. fluorescens CS significantly downregulated S. epidermidis ica genes and upregulated an ica repressor, tcaR. This indicates that the CS reduced polysaccharide intercellular adhesin synthesis, a major component of the S. epidermidis 1457 biofilm matrix. Further studies are required to elucidate the specific inhibitory components in the CS and their mechanism of action. Our results indicate that inhibitors in the P. fluorescens CS may have a significant value for inhibiting S. epidermidis biofilm. Combinations of specific inhibitors from the CS and antibiotics against staphylococci may provide an effective measure to control S. epidermidis biofilm formation while avoiding antibiotic resistance and compensating the attenuated effectiveness of antibiotics on biofilms.

Introduction

Staphylococcus epidermidis is a commensal bacterium found on human skin and mucous membranes. Along with Staphylococcus aureus, S. epidermidis-mediated infections are becoming increasingly prevalent as indwelling medical devices are more commonly used. They are often associated with hip implants and catheters, causing systemic infections [1].

S. epidermidis does not express as many virulence factors as S. aureus [2]. One important molecular mechanism that triggers the transition of commensal S. epidermidis to an opportunistic pathogen is the ability to form biofilms [3, 4]. Polysaccharide intercellular adhesin (PIA) composed of ß-1,6-N-acetyl-D-glucosamine polymers (PNAG) is a major component of S. epidermidis biofilm. PIA, proteins, and extracellular DNA are adhesion molecules and make up the extracellular polymeric substances (EPS) in the biofilm matrix [5]. With enhanced PIA production, the established biofilms clog medical devices, attenuating antibiotic penetration and host immune response [3, 6].

Taking into consideration the increasing prevalence of biofilm-mediated infections, it is critical to understand the mechanisms of S. epidermidis biofilm formation and develop methods of inhibition. Previously, extracellular products of Pseudomonas aeruginosa and the CS of some Actinomycete and Enterobacter strains were demonstrated to inhibit S. epidermidis biofilm formation [7,8,9].

P. fluorescens, soil-associated bacteria, suppress plant diseases by producing antimicrobial substances [10]. Recently, the quorum sensing (QS) molecules of P. fluorescens has been shown to inhibit biofilm formation of Shewanella baltica [11]. In this study, the effects of P. fluorescens CS were assessed to determine whether P. fluorescens produce antibiofilm agents against S. epidermidis in addition to previously known bioactive agents. In vitro biofilm assays and RT-qPCR revealed that P. fluorescens CS functions against S. epidermidis biofilm formation and influences genes involved in PIA production, indicating the presence of anti-biofilm agents in the CS.

Methods

Bacterial strains and preparation of bacterial CS for biofilm assay

S. epidermidis 1457, kindly donated by Dr. Paul Fey (University of Nebraska Medical Center), was cultured in tryptic soy broth (TSB) at 30 °C. P. fluorescens Pf01 strain was a gift from Dr. George O’Toole (Dartmouth University) and was maintained in Luria-Bertani broth (LB) at 30 °C. To prepare the CS, an overnight P. fluorescens culture was used to inoculate 200 ml LB (starting OD660 = 0.05). The culture was grown at 30 °C with agitation to the early-stationary phase (OD660 = 0.8), centrifuged at 4 °C, 12,000 g for 15 min, passed through a 0.22 µm filter system, and then heat-treated (100 °C) for 5 min for all biofilm assays.

Characterization of inhibitory factors in P. fluorescens CS

The filtered CS was subject to one of the following. 1. Heating at 100 °C for 1–5 min. 2. Proteinase K (400 µg/mL) treatment at 55 °C, 30 min. 3. Trypsin (2.5 µg/mL) treatment at 37 °C for 4 h [12]. 4. Cellulase (5 mg/ml, MP Biomedicals) treatment at 37 °C for 1 h [7]. 5. Lipase acrylic resin (2 mg/mL, Millipore Sigma, L4777) treatment at 37 °C for 48 h [13]. 6. Heating at 100 °C for 5 min and centrifuging in a Centricon tube (Millipore) with a 3 K molecular weight (MW) limit (7500X g, 40 min at 4 °C, [13]). Enzyme treatments of CS were performed with gentle shaking, followed by heat inactivation at 95 °C for 20 min.

Growth kinetics and biofilm assay of S. epidermidis

Overnight cultures of S. epidermidis were prepared to reach the exponential growth phase (OD600 = 0.8, ≈1X 109 CFU/mL), and diluted with TSB (1:5000). The diluted cultures (200 µl) were incubated with or without 50% (v/v) P. fluorescens CS in a 48-well plate (Costar, polystyrene) and monitored using a microtiter plate reader (BioTek) for 72 h to analyze growth kinetics. For the biofilm assays, after 24 h incubation without agitation, the plate was washed, stained with 0.1% crystal violet, and read at optical density 600 nm [7].

Biofilm disruption assay

Biofilm disruption was performed with NaIO4, Proteinase K, or DNase I. S. epidermidis was allowed to form biofilm with or without 25% (v/v) P. fluorescens CS at 30 °C for 16 h. After washing, the preformed biofilm was treated with either NaIO4 (40 mM in dH2O) for 2 h at 37 °C, Proteinase K (0.1 mg/mL, in 20 mM Tris-HCl, 1 mM CaCl2, pH 7.5), or DNase I (0.5 mg/mL in 5 mM MgCl2, DNA 25, Sigma) for 24 h at 37 °C [14,15,16]. After incubation with each disruptive agent, the remaining biofilms were quantified as above after crystal violet staining.

Reverse transcription-quantitative PCR

S. epidermidis biofilm was lysed with a sonicator (Fisher Scientific) at amplitude 4.0 for 20 s and treated with lysozyme (50 mg/mL, 2 h, 37 °C). Total RNA was isolated using the RiboPure Bacteria and treated with DNase I (Invitrogen). cDNA was generated from total RNA (1 µg) using the SuperScript III first-strand synthesis system (Invitrogen). qPCR was performed using cDNA (diluted 1:20), Advanced Universal SYBR green Supermix (Bio-Rad), Bio-Rad CFX96 Touch real-time PCR detection systems, and primers (Additional file 1: Table S1). All data were analyzed with Bio-Rad CFX manager software 3.1 using Cq values. 16S rRNA was used for normalization. The mean fold change between study groups was calculated following the 2−ΔΔCt method [17].

Statistical analysis

Three independent experiments were performed using 3 biological samples in triplicate. One representative data (n = 9) were analyzed by unpaired T-test or analysis of variance (ANOVA) using Graph Pad Prism 9 (P-value < 0.05 is considered statistically significant).

Results

P. fluorescens CS inhibits S. epidermidis biofilm maturation but not its growth

Various concentrations of the P. fluorescens CS were tested on S. epidermidis biofilm. Addition of 25% and 50% CS resulted in approximately 30% and 95% biofilm reduction, respectively (Fig. 1A). This result indicates that P. fluorescens CS effectively inhibited S. epidermidis biofilm formation in a concentration-dependent manner. Growth kinetics revealed that boiled P. fluorescens CS did not affect bacterial growth of S. epidermidis, although untreated CS resulted in slight but noticeable growth retardation after 40 h (Fig. 1B). These results suggest anti-biofilm activity of P. fluorescens CS on S. epidermidis independent of growth inhibition. Previous studies indicate there are four stages in forming S. epidermidis biofilm, including adherence, accumulation, maturation, and detachment [4]. Treatment of S. epidermidis with P. fluorescens CS for 8 h showed no significant change in biofilm formation, whereas treatments for 12, 18, and 24 h showed 70%, 63%, and 54% biofilm inhibition, respectively (Fig. 1C). This indicates that the CS specifically attenuates biofilm maturation stages rather than initial attachment (< 8 h).

Fig. 1
figure 1

P. fluorescens CS specifically inhibits S. epidermidis biofilm maturation but not its growth. A S. epidermidis biofilm assay was performed with 0%, 25%, or 50% addition of P. fluorescens CS. B The growth kinetics of S. epidermidis were measured with or without 50% P. fluorescens CS. One representative result from three biological samples is shown as means with standard deviations (n = 3). C S. epidermidis biofilm with 50% P. fluorescens CS were removed at time intervals of 8, 12, 18, or 24 h. The bars indicate standard deviation (n = 9). **, *** indicate statistical significance at P < 0.01 and P < 0.001, respectively

The inhibitory compounds of P. fluorescens CS are heat-resistant and multifactorial

To further characterize the active compound(s) essential for biofilm inhibition, P. fluorescens CS was treated with heat or various enzymes. The 5 min-boiled CS showed comparable inhibition with those heated for less time or not at all (< 5 min) (Fig. 2A). Biofilm inhibition also remained the same after treatment of CS with either trypsin, cellulase, or lipase (Additional file 1: Fig. S1A, B, C). However, Proteinase K treatment attenuated the inhibitory function of the CS, showing 15% less biofilm inhibition than untreated CS (Fig. 2B). These results indicate that the inhibitory components in the CS are heat-resistant, and some are proteinaceous. The CS fraction  > 3 k MW showed similar inhibition to whole CS, although the fraction < 3 k MW also inhibited biofilm but at a significantly lower rate than components > 3 k (Fig. 2C). This indicates that there are inhibitory components in the CS that are both larger and smaller than 3 k MW.

Fig. 2
figure 2

The inhibitory components of P. fluorescens CS are heat-resistant and not small molecules. A P. fluorescens CS was boiled for various times up to 5 min before treating S. epidermidis. B P. fluorescens CS was treated with proteinase K and used for S. epidermidis biofilm. C Boiled P. fluorescens CS was filtered in a Millipore Centricon tube with 3 K MW limit (> 3 K MW: retentate; < 3 K MW: filtrate). D S. epidermidis 1457 biofilm assay was performed with or without 25% CS. The preformed biofilms were treated with NaIO4 for disruption. Data shown as means from three separate assays. The bars indicate standard deviation (n = 9). **, *** indicate statistical significance at P < 0.01 and P < 0.001, respectively, ns: not significant

P. fluorescens CS targets polysaccharide intercellular adhesin

Preformed biofilms (16 h) were treated with biofilm-degrading agents such as NaIO4, Proteinase K, or DNase I to investigate which component of S. epidermidis biofilm was specifically influenced by P. fluorescens CS [14,15,16]. It is reasonable to assume that if a particular macromolecule of the biofilm matrix was targeted by the CS, there would be none left to be further disrupted by a chemical treatment. NaIO4 is an oxidizing agent that targets glucose-containing polysaccharides in biofilm [14]. The mature S. epidermidis biofilm without the CS was treated with NaIO4 and showed approximately 20% biofilm disruption (Fig. 2D). Mature biofilm formed in the presence of 25% CS was not significantly affected by NaIO4, implying inhibitors in the P. fluorescens CS already removed most of the polysaccharides in the biofilm matrix. Similar experiments using Proteinase K showed no noticeable disruption of S. epidermidis biofilm, suggesting that proteins are not major components of S. epidermidis 1457 biofilm, as previously reported (Additional file 1: Fig. S2A, [18]). The preformed biofilms with or without CS were further disrupted by DNase I, indicating P. fluorescens CS did not target nucleic acids in biofilms (Additional file 1: Fig. S2B).

P. fluorescens CS downregulates S. epidermidis PIA biosynthesis

RT-qPCR analysis revealed that CS treatment significantly downregulated the icaADBC operon, which is essential for PIA synthesis, and upregulated tcaR, a main ica repressor in S. epidermidis 1457 (Fig. 3A, B, [19]). No other biofilm-related regulatory genes tested showed different expression with CS treatment (Additional file 1: Fig. S3, [15, 20,21,22,23,24]). These results indicate that one of the main inhibitory mechanisms of P. fluorescens CS is to regulate icaADBC gene expression at the transcriptional level, resulting in significant reduction in PIA, a major component of the S. epidermidis 1457 biofilm matrix [18]. Biofilm assay was performed with additional glucose since PIA production and ica gene expression have been shown to be affected by glucose addition (Additional file 1: Fig. S4, [25]). Furthermore, S. epidermidis biofilm with CS treatment in our biofilm assay has fewer nutrients available than S. epidermidis without the CS. Supplementation with different concentrations of glucose did not affect the CS inhibition level, indicating that glucose did not modulate the ica operon in our system and the biofilm inhibition observed is not due to a lack of energy sources for PIA synthesis or planktonic growth of S. epidermidis with the spent media of P. fluorescens.

Fig. 3
figure 3

P. fluorescens CS differentially regulates genes involved in S. epidermidis polysaccharide biosynthesis. A RT-qPCR was performed on icaADBC from S. epidermidis with or without CS. B RT-qPCR was performed on ica repressors from S. epidermidis with or without CS. *, *** indicate statistical significance at P < 0.05 and P < 0.001, respectively

Discussion

Biofilm formation is a main mode of S. epidermidis pathogenicity which causes opportunistic infections in hospitalized patients, imposing heavy physical and economic burdens. The prevalence and structure of S. epidermidis biofilms vary greatly depending on environmental factors such as nutrients, pH, temperature, and multispecies presence, and the seed time of S. epidermidis on medical devices [26]. In our study, the CS of P. fluorescens strain Pf01 showed specific inhibition of the maturation stages of S. epidermidis biofilm formation while not affecting planktonic growth. This is partly due to gene regulation at the transcriptional level, affecting the ica operon and tcaR, resulting in significant reduction in PIA/PNAG synthesis and almost complete biofilm inhibition. Pf01 CS was shown to inhibit Shewanella baltica by producing various QS molecules such as acyl-l-homoserine lactones (AHLs) and the autoinducer-2 (AI-2) [9]. Although our liquid chromatography-mass spectrometry (LC-MS) analysis did not reveal the presence of known QS molecules in the CS (data not shown), it is possible that Pf01 CS contains specific QS inhibitors (QSI) against S. epidermidis, affecting the expression levels of stress-response genes, including the ica operon. The majority of inhibitory components were heat-stable and enzyme-resistant macromolecules (> 3 K MW), which may work together with small-molecule inhibitors like QSIs, displaying robust biofilm inhibition. Importantly, direct cell lysis is not an inhibitory action mode of the CS, since S. epidermidis was viable and showed no growth retardation with the boiled CS. This is different from P. aeruginosa exopolysaccharides, which have been shown to inhibit both the planktonic growth and biofilm formation of S. epidermidis [7]. Targeting only biofilm formation without interfering with bacterial growth is an important property in developing biofilm inhibitors since those interfering with cell growth often induce selective pressure on drug-resistant strains, resulting in survival of highly virulent pathogens [27]. To our knowledge, Pf01 is one of few CSs reported to specifically inhibit staphylococcal biofilms without affecting bacterial growth [8]. Other aspects of the CS’s functions are under investigation including removing existing S. epidermidis biofilms, regulating biofilm-related accessory genes at different stages of biofilm formation, and inhibiting the formation of other bacterial biofilms. The CS should also be tested on other S. epidermidis strains that produce a protein-dependent biofilm matrix. Taken together, the P. fluorescens CS is promising as an alternative and complementary measure to minimize staphylococcal biofilm EPS within medical devices, providing better control against S. epidermidis-mediated nosocomial infection.

Limitations

It is not certain whether S. epidermidis biofilm inhibition was due to specific inhibitors or nonspecific degradation by metabolic wastes in the CS. Ethyl acetate extraction and LC-MS analysis of P. fluorescens CS did not reveal any known QS molecules in Pf01. Assays using biosensor strains for detecting AHLs and AI-2 in the CS will be necessary. Generating mutant strains of P. fluorescens and S. epidermidis will be performed to determine the precise inhibitory mechanism of P. fluorescens CS on S. epidermidis biofilm.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Abbreviations

AHL:

Acyl-L-homoserine lactone

ANOVA:

Analysis of variance

AI-2:

Autoinducer-2

CFU:

Colony forming units

CS:

Culture supernatant

EPS:

Extracellular polymeric substances

LB:

Luria-Bertani

LC-MS:

Liquid chromatography-mass spectrometry

MW:

Molecular weight

PIA:

Polysaccharide intercellular adhesin

PNAG:

ß-1,6-N-acetyl-D-glucosamine polymers

P. fluorescens :

Pseudomonas fluorescens

QS:

Quorum-sensing

QSI:

Quorum-sensing inhibitor

RT-qPCR:

Reverse transcription-quantitative polymerase chain reaction

S. epidermidis :

Staphylococcus epidermidis

TSB:

Tryptic soy broth

References

  1. Oliveira WF, Silva PMS, Silva RCS, Silva GMM, Machado G, Coelho L, Correia MTS. Staphylococcus aureus and Staphylococcus epidermidis infections on implants. J Hosp Infect. 2018;98(2):111–7.

    Article  CAS  Google Scholar 

  2. Gill SR, Fouts DE, Archer GL, Mongodin EF, Deboy RT, Ravel J, Paulsen IT, Kolonay JF, Brinkac L, Beanan M, et al. Insights on evolution of virulence and resistance from the complete genome analysis of an early methicillin-resistant Staphylococcus aureus strain and a biofilm-producing methicillin-resistant Staphylococcus epidermidis strain. J Bacteriol. 2005;187(7):2426–38.

    Article  CAS  Google Scholar 

  3. 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):S14-20.

    Article  CAS  Google Scholar 

  4. Fey PD, Olson ME. Current concepts in biofilm formation of Staphylococcus epidermidis. Future Microbiol. 2010;5(6):917–33.

    Article  CAS  Google Scholar 

  5. Arciola CR, Campoccia D, Ravaioli S, Montanaro L. Polysaccharide intercellular adhesin in biofilm: structural and regulatory aspects. Front Cell Infect Microbiol. 2015;5:7.

    Article  Google Scholar 

  6. Vuong C, Voyich JM, Fischer ER, Braughton KR, Whitney AR, DeLeo FR, Otto M. Polysaccharide intercellular adhesin (PIA) protects Staphylococcus epidermidis against major components of the human innate immune system. Cell Microbiol. 2004;6(3):269–75.

    Article  CAS  Google Scholar 

  7. Qin Z, Yang L, Qu D, Molin S, Tolker-Nielsen T. Pseudomonas aeruginosa extracellular products inhibit staphylococcal growth, and disrupt established biofilms produced by Staphylococcus epidermidis. Microbiology (Reading). 2009;155(Pt 7):2148–56.

    Article  CAS  Google Scholar 

  8. Xie TT, Zeng H, Ren XP, Wang N, Chen ZJ, Zhang Y, Chen W. Antibiofilm activity of three Actinomycete strains against Staphylococcus epidermidis. Lett Appl Microbiol. 2019;68(1):73–80.

    Article  CAS  Google Scholar 

  9. De NunesOliveira S, da RosaSilva H, Canellas ALB, Romanos MTV, dos Santos KRN, Muricy G, Oelemanan WMR, Laport MS. High reduction of staphylococcal biofilm by aqueous extract from marine sponge-isolated Enterobacter sp. Res Microbiol. 2021. https://0-doi-org.brum.beds.ac.uk/10.1016/j.resmic.2020.10.002.

    Article  Google Scholar 

  10. Scales BS, Dickson RP, LiPuma JJ, Huffnagle GB. Microbiology, genomics, and clinical significance of the Pseudomonas fluorescens species complex, an unappreciated colonizer of humans. Clin Microbiol Rev. 2014;27(4):927–48.

    Article  Google Scholar 

  11. Zhao A, Zhu J, Ye X, Ge Y, Li J. Inhibition of biofilm development and spoilage potential of Shewanella baltica by quorum sensing signal in cell-free supernatant from Pseudomonas fluorescens. Int J Food Microbiol. 2016;230:73–80.

    Article  CAS  Google Scholar 

  12. Limoli DH, Warren EA, Yarrington KD, Donegan NP, Cheung AL, O’Toole GA. Interspecies interactions induce exploratory motility in Pseudomonas aeruginosa. Elife. 2019. https://0-doi-org.brum.beds.ac.uk/10.7554/eLife.47365.

    Article  Google Scholar 

  13. Doghri I, Portier E, Desriac F, Zhao JM, Bazire A, Dufour A, Rochette V, Sable S, Lanneluc I. Anti-biofilm activity of a low weight proteinaceous molecule from the marine bacterium Pseudoalteromonas sp. IIIA004 against marine bacteria and human pathogen biofilms. Microorganisms. 2020. https://0-doi-org.brum.beds.ac.uk/10.3390/microorganisms8091295.

    Article  Google Scholar 

  14. Kogan G, Sadovskaya I, Chaignon P, Chokr A, Jabbouri S. Biofilms of clinical strains of Staphylococcus that do not contain polysaccharide intercellular adhesin. FEMS Microbiol Lett. 2006;255(1):11–6.

    Article  CAS  Google Scholar 

  15. Freitas AI, Lopes N, Oliveira F, Bras S, Franca A, Vasconcelos C, Vilanova M, Cerca N. Comparative analysis between biofilm formation and gene expression in Staphylococcus epidermidis isolates. Future Microbiol. 2018;13:415–27.

    Article  CAS  Google Scholar 

  16. Loza-Correa M, Ayala JA, Perelman I, Hubbard K, Kalab M, Yi QL, Taha M, de Pedro MA, Ramirez-Arcos S. The peptidoglycan and biofilm matrix of Staphylococcus epidermidis undergo structural changes when exposed to human platelets. PLoS ONE. 2019;14(1):e0211132.

    Article  CAS  Google Scholar 

  17. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 2001;25(4):402–8.

    Article  CAS  Google Scholar 

  18. Schaeffer CR, Hoang TN, Sudbeck CM, Alawi M, Tolo IE, Robinson DA, Horswill AR, Rohde H, Fey PD. Versatility of biofilm matrix molecules in Staphylococcus epidermidis clinical isolates and importance of polysaccharide intercellular adhesin expression during high shear stress. mSphere. 2016. https://0-doi-org.brum.beds.ac.uk/10.1128/mSphere.00165-16.

    Article  Google Scholar 

  19. Hoang TM, Zhou C, Lindgren JK, Galac MR, Corey B, Endres JE, Olson ME, Fey PD. Transcriptional regulation of icaADBC by both IcaR and TcaR in Staphylococcus epidermidis. J Bacteriol. 2019. https://0-doi-org.brum.beds.ac.uk/10.1128/JB.00524-18.

    Article  Google Scholar 

  20. Huang Q, Fei J, Yu HJ, Gou YB, Huang XK. Effects of human beta-defensin-3 on biofilm formation regulating genes dltB and icaA in Staphylococcus aureus. Mol Med Rep. 2014;10(2):825–31.

    Article  CAS  Google Scholar 

  21. Schaeffer CR, Woods KM, Longo GM, Kiedrowski MR, Paharik AE, Buttner H, Christner M, Boissy RJ, Horswill AR, Rohde H, et al. Accumulation-associated protein enhances Staphylococcus epidermidis biofilm formation under dynamic conditions and is required for infection in a rat catheter model. Infect Immun. 2015;83(1):214–26.

    Article  Google Scholar 

  22. Wang C, Li M, Dong D, Wang J, Ren J, Otto M, Gao Q. Role of ClpP in biofilm formation and virulence of Staphylococcus epidermidis. Microbes Infect. 2007;9(11):1376–83.

    Article  CAS  Google Scholar 

  23. Li M, Lai Y, Villaruz AE, Cha DJ, Sturdevant DE, Otto M. Gram-positive three-component antimicrobial peptide-sensing system. Proc Natl Acad Sci U S A. 2007;104(22):9469–74.

    Article  CAS  Google Scholar 

  24. Vuong C, Gerke C, Somerville GA, Fischer ER, Otto M. Quorum-sensing control of biofilm factors in Staphylococcus epidermidis. J Infect Dis. 2003;188(5):706–18.

    Article  CAS  Google Scholar 

  25. Dobinsky S, Kiel K, Rohde H, Bartscht K, Knobloch JK, Horstkotte MA, Mack D. Glucose-related dissociation between icaADBC transcription and biofilm expression by Staphylococcus epidermidis: evidence for an additional factor required for polysaccharide intercellular adhesin synthesis. J Bacteriol. 2003;185(9):2879–86.

    Article  CAS  Google Scholar 

  26. Stewart EJ, Payne DE, Ma TM, VanEpps JS, Boles BR, Younger JG, Solomon MJ. Effect of antimucrobial and physical treatments on growth of multi species staphylococcal biofilms. Appl Environ Microbiol. 2017. https://0-doi-org.brum.beds.ac.uk/10.1128/AEM.03483-16.

    Article  Google Scholar 

  27. Barzegari A, Kheyrolahzadeh K, Hosseiniyan Khatibi SM, Sharifi S, Memar MY, Zununi Vahed S. The battle of probiotics and their derivatives against biofilms. Infect Drug Resist. 2020;13:659–72.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors would like to acknowledge Dr. Mark Bolyard (Union University) for his valuable comments in editing the manuscript.

Funding

This research was funded by the Union University Biology Department (E.C.) and supported by the Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (S.C.) (2016R1D1A1B01009752).

Author information

Authors and Affiliations

Authors

Contributions

EC conceived, designed, performed experiments, and analyzed the data; BW, GM, EA, and SC performed experiments and collected the data; EC, BW, GM wrote the manuscript. SC edited the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Euna Choi.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

competing interests

The authors declare no conflict of interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1: Figure S1.

S. epidermidis biofilm with enzyme-treated P. fluorescens CS. (A) P. fluorescens CS was pre-treated with trypsin, (B) cellulase, or (C) lipase and used for S. epidermidis biofilm assay. Data shown as means from 3 separate assays. The bars indicate standard deviation (n=9). *** indicate statistical significance at P<0.001, ns: not significant. Figure S2. Effects of Proteinase K and DNase on preformed S. epidermidis biofilm with or without CS. (A) S. epidermidis 1457 biofilm assay was performed with or without 25% CS. The preformed biofilms were treated with Proteinase K or (B) DNase for disruption. *** indicate statistical significance at P<0.001, ns: not significant. Figure S3. RT-qPCR was performed on multiple biofilm-associated genes from S. epidermidis with or without CS. ns: not significant. Figure S4. P. fluorescens CS was supplemented with different concentrations of glucose and used for biofilm assay. *** indicate statistical significance at P<0.001, ns: not significant. Table S1. List of primer sequences used for RT-qPCR.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Choi, E., Wells, B., Mirabella, G. et al. Anti-biofilm activity of Pseudomonas fluorescens culture supernatants on biofilm formation of Staphylococcus epidermidis 1457. BMC Res Notes 15, 370 (2022). https://0-doi-org.brum.beds.ac.uk/10.1186/s13104-022-06257-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://0-doi-org.brum.beds.ac.uk/10.1186/s13104-022-06257-z

Keywords