Biofilm Formation by Pathogenic Bacteria: Applying a Staphylococcus aureus Model to Appraise Potential Targets for Therapeutic Intervention
Abstract
1. Introduction
2. Biofilm Formation
2.1. Microbial Surface Adhesion
2.2. Development to Mature Biofilm
2.3. Detachment
2.4. Quorum Sensing
3. Anti-Biofilm Treatments
3.1. Antibiotics in Single and Combination Therapy
3.2. Other Anti-Biofilm Agents
Anti-Biofilm Agent | Mechanism of Action | Level of Interruption | Advantages | Disadvantages | References |
---|---|---|---|---|---|
Rhamnolipids | Disrupt biofilm | Adhesion Maturation process | High surface activity Biodegradability Low toxicity | Limited production Increasing usage is a threat to synthetic surfactants | [162,211] |
Photodynamics | Affect bacterial LPS, endotoxin and cell differentiation | Mature biofilm | Synergic effect Strong treatment | Thermal damage Antibacterial resistance Surface modification | [167] |
Nanoparticles | Transport drugs | Adhesion and mature biofilm | Small size Higher surface area to volume ratio | Toxicity | [79] |
Bacteriophages | Disrupt biofilm | Mature biofilm | Specific for targets Effective against resistant strains | Further studies required Potential threat to human health | [26] |
Antimicrobial peptides | Increase permeability of cell membrane | All three phases | Less chance of resistance Strong antibacterial activity | Further in vivo verification required Synthesis and purification are challenging | [209] |
Antibodies | Help innate immune system | Adhesion and mature biofilm | Produce vaccine Prevention therapy | Further studies required | [78] |
Phytochemicals | Reduce cell adhesion and disperse biofilm | Mature biofilm and dispersal | Natural compounds Strong antimicrobial agents | Poor solubility in aqueous media Further in vivo verification required | [212,213] |
Chelators and Sulfhydryl Compounds | Decrease bacterial interaction and decrease PIA/PNAG | Adhesion | Potent antibiotic activity | Cytotoxic and genotoxic effects | [214] |
Laser Therapy | Oxidative stress and disrupt bacterial cell wall | Mature biofilm | Boost antibiotic efficacy | High temperature in host tissue Cellular damage Further studies required | [215,216] |
Enzymes | Target ECM and cell wall and increase chemical reaction | Adhesion and mature biofilm | Harmless to humans | Potential for activating immune system Further studies required | [154,217] |
4. Future Directions
5. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Jamal, M.; Ahmad, W.; Andleeb, S.; Jalil, F.; Imran, M.; Nawaz, M.A.; Hussain, T.; Ali, M.; Rafiq, M.; Kamil, M.A. Bacterial biofilm and associated infections. J. Chin. Med. Assoc. 2018, 81, 7–11. [Google Scholar] [CrossRef] [PubMed]
- Slavkin, H.C. Biofilms, microbial ecology and Antoni van Leeuwenhoek. J. Am. Dent. Assoc. 1997, 128, 492–495. [Google Scholar] [CrossRef] [PubMed]
- Sedarat, Z.; Taylor-Robinson, A.W. A Consideration of antibacterial agent efficacies in the treatment and prevention of formation of Staphylococcus aureus biofilm. J. Microbiol. Infect. Dis. 2019, 9, 167–172. [Google Scholar] [CrossRef]
- Hadjifrangiskou, M.; Hultgren, S.J. What does it take to stick around? Molecular insights into biofilm formation by uropathogenic Escherichia coli. Virulence 2012, 3, 231–233. [Google Scholar] [CrossRef]
- Jefferson, K.K. What drives bacteria to produce a biofilm? FEMS Microbiol. Lett. 2004, 236, 163–173. [Google Scholar] [CrossRef]
- McCarty, S.; Woods, E.; Percival, S.L. Biofilms: From concept to reality. In Biofilms in Infection Prevention and Control; Percival, S.L., Randle, J., Cooper, T., Williams, D.W., Eds.; Academic Press: Boston, MA, USA, 2014; pp. 143–163. [Google Scholar] [CrossRef]
- Jolivet-Gougeon, A.; Bonnaure-Mallet, M. Biofilms as a mechanism of bacterial resistance. Drug Discov. Today Technol. 2014, 11, 49–56. [Google Scholar] [CrossRef]
- Donlan, R.M. Biofilms: Microbial life on surfaces. Emerg. Infect. Dis. 2002, 8, 881–890. [Google Scholar] [CrossRef]
- Tong, S.Y.C.; Davis, J.S.; Eichenberger, E.; Holland, T.L.; Fowler, V.G., Jr. Staphylococcus aureus infections: Epidemiology, pathophysiology, clinical manifestations, and management. Clin. Microbiol. Rev. 2015, 28, 603–661. [Google Scholar] [CrossRef]
- Zecconi, A.; Scali, F. Staphylococcus aureus virulence factors in evasion from innate immune defenses in human and animal diseases. Immunol. Lett. 2013, 150, 12–22. [Google Scholar] [CrossRef]
- Lister, J.L.; Horswill, A.R. Staphylococcus aureus biofilms: Recent developments in biofilm dispersal. Front. Cell. Infect. Microbiol. 2014, 4, 178. [Google Scholar] [CrossRef]
- Mah, T.-F.C.; O’Toole, G.A. Mechanisms of biofilm resistance to antimicrobial agents. Trends Microbiol. 2001, 9, 34–39. [Google Scholar] [CrossRef]
- Manandhar, S.; Singh, A.; Varma, A.; Pandey, S.; Shrivastava, N. Biofilm producing clinical Staphylococcus aureus isolates augmented prevalence of antibiotic resistant cases in tertiary care hospitals of Nepal. Front. Microbiol. 2018, 9, 2749. [Google Scholar] [CrossRef] [PubMed]
- Høiby, N.; Ciofu, O.; Bjarnsholt, T. Pseudomonas aeruginosa biofilms in cystic fibrosis. Future Microbiol. 2010, 5, 1663–1674. [Google Scholar] [CrossRef] [PubMed]
- Foxman, B. The epidemiology of urinary tract infection. Nat. Rev. Urol. 2010, 7, 653–660. [Google Scholar] [CrossRef]
- MacKenzie, K.D.; Palmer, M.B.; Köster, W.L.; White, A.P. Examining the link between biofilm formation and the ability of pathogenic Salmonella strains to colonize multiple host species. Front. Vet. Sci. 2017, 4, 138. [Google Scholar] [CrossRef]
- Lemon, K.P.; Higgins, D.E.; Kolter, R. Flagellar motility is critical for Listeria monocytogenes biofilm formation. J. Bacteriol. 2007, 189, 4418–4424. [Google Scholar] [CrossRef]
- Vlamakis, H.; Chai, Y.; Beauregard, P.; Losick, R.; Kolter, R. Sticking together: Building a biofilm the Bacillus subtilis way. Nat. Rev. Microbiol. 2013, 11, 157–168. [Google Scholar] [CrossRef]
- Yonezawa, H.; Osaki, T.; Kurata, S.; Zaman, C.; Hanawa, T.; Kamiya, S. Assessment of in vitro biofilm formation by Helicobacter pylori. J. Gastroenterol. Hepatol. 2010, 25 (Suppl. S1), S90–S94. [Google Scholar] [CrossRef]
- Nicolas, G.G.; Lavoie, M.C. Streptococcus mutans and oral streptococci in dental plaque. Can. J. Microbiol. 2011, 57, 1–20. [Google Scholar] [CrossRef]
- Darouiche, R.O. Treatment of infections associated with surgical implants. N. Engl. J. Med. 2004, 350, 1422–1429. [Google Scholar] [CrossRef]
- Mulcahy, L.R.; Isabella, V.M.; Lewis, K. Pseudomonas aeruginosa biofilms in disease. Microb. Ecol. 2014, 68, 1–12. [Google Scholar] [CrossRef] [PubMed]
- Lewis, K. Persister cells. Annu. Rev. Microbiol. 2010, 64, 357–372. [Google Scholar] [CrossRef] [PubMed]
- Bridier, A.; Briandet, R. Microbial biofilms: Structural plasticity and emerging properties. Microorganisms 2022, 10, 138. [Google Scholar] [CrossRef] [PubMed]
- Qu, Y.; Daley, A.J.; Istivan, T.S.; Rouch, D.A.; Deighton, M.A. Densely adherent growth mode, rather than extracellular polymer substance matrix build-up ability, contributes to high resistance of Staphylococcus epidermidis biofilms to antibiotics. J. Antimicrob. Chemother. 2010, 65, 1405–1411. [Google Scholar] [CrossRef] [PubMed]
- Ferriol-González, C.; Domingo-Calap, P. Phages for biofilm removal. Antibiotics 2020, 9, 268. [Google Scholar] [CrossRef]
- Melter, O.; Radojevič, B. Small colony variants of Staphylococcus aureus—Review. Folia Microbiol. 2010, 55, 548–558. [Google Scholar] [CrossRef]
- Roca, I.; Akova, M.; Baquero, F.; Carlet, J.; Cavaleri, M.; Coenen, S.; Cohen, J.; Findlay, D.; Gyssens, I.; Heuer, O.E.; et al. The global threat of antimicrobial resistance: Science for intervention. New Microbes New Infect. 2015, 6, 22–29. [Google Scholar] [CrossRef]
- Serra, D.O.; Hengge, R. Stress responses go three dimensional—The spatial order of physiological differentiation in bacterial macrocolony biofilms. Environ. Microbiol. 2014, 16, 1455–1471. [Google Scholar] [CrossRef]
- Fletcher, M.; Loeb, G.I. Influence of substratum characteristics on the attachment of a marine pseudomonad to solid surfaces. Appl. Environ. Microbiol. 1979, 37, 67–72. [Google Scholar] [CrossRef]
- Bendinger, B.; Rijnaarts, H.H.; Altendorf, K.; Zehnder, A.J. Physicochemical cell surface and adhesive properties of coryneform bacteria related to the presence and chain length of mycolic acids. Appl. Environ. Microbiol. 1993, 59, 3973–3977. [Google Scholar] [CrossRef]
- Power, P.M.; Jennings, M.P. The genetics of glycosylation in Gram-negative bacteria. FEMS Microbiol. Lett. 2003, 218, 211–222. [Google Scholar] [CrossRef] [PubMed]
- Costerton, J.W.; Cheng, K.-J.; Geesey, G.G.; Ladd, T.I.; Nickel, J.C.; Dasgupta, M.; Marrie, T.J. Bacterial biofilms in nature and disease. Annu. Rev. Microbiol. 1987, 41, 435–464. [Google Scholar] [CrossRef] [PubMed]
- Hall-Stoodley, L.; Costerton, J.W.; Stoodley, P. Bacterial biofilms: From the natural environment to infectious diseases. Nat. Rev. Microbiol. 2004, 2, 95–108. [Google Scholar] [CrossRef] [PubMed]
- Flemming, H.C.; Wingender, J. The biofilm matrix. Nat. Rev. Microbiol. 2010, 8, 623–633. [Google Scholar] [CrossRef]
- Otto, M. Bacterial evasion of antimicrobial peptides by biofilm formation. Curr. Top. Microbiol. Immunol. 2006, 306, 251–258. [Google Scholar] [CrossRef]
- McConoughey, S.J.; Howlin, R.; Granger, J.F.; Manring, M.M.; Calhoun, J.H.; Shirtliff, M.; Kathju, S.; Stoodley, P. Biofilms in periprosthetic orthopedic infections. Future Microbiol. 2014, 9, 987–1007. [Google Scholar] [CrossRef]
- Taylor, P.K.; Yeung, A.T.Y.; Hancock, R.E.W. Antibiotic resistance in Pseudomonas aeruginosa biofilms: Towards the development of novel anti-biofilm therapies. J. Biotechnol. 2014, 191, 121–130. [Google Scholar] [CrossRef]
- Otto, M. Staphylococcal biofilms. Microbiol. Spectr. 2018, 6. [Google Scholar] [CrossRef]
- Gupta, P.; Sarkar, S.; Das, B.; Bhattacharjee, S.; Tribedi, P. Biofilm, pathogenesis and prevention—A journey to break the wall: A review. Arch. Microbiol. 2016, 198, 1–15. [Google Scholar] [CrossRef]
- Navarre, W.W.; Schneewind, O. Proteolytic cleavage and cell wall anchoring at the LPXTG motif of surface proteins in gram-positive bacteria. Mol. Microbiol. 1994, 14, 115–121. [Google Scholar] [CrossRef]
- O’Neill, E.; Pozzi, C.; Houston, P.; Smyth, D.; Humphreys, H.; Robinson, D.A.; O’Gara, J.P. Association between methicillin susceptibility and biofilm regulation in Staphylococcus aureus isolates from device-related infections. J. Clin. Microbiol. 2007, 45, 1379–1388. [Google Scholar] [CrossRef] [PubMed]
- Corrigan, R.M.; Miajlovic, H.; Foster, T.J. Surface proteins that promote adherence of Staphylococcus aureus to human desquamated nasal epithelial cells. BMC Microbiol. 2009, 9, 22. [Google Scholar] [CrossRef] [PubMed]
- McDevitt, D.; Francois, P.; Vaudaux, P.; Foster, T.J. Molecular characterization of the clumping factor (fibrinogen receptor) of Staphylococcus aureus. Mol. Microbiol. 1994, 11, 237–248. [Google Scholar] [CrossRef]
- Zong, Y.; Xu, Y.; Liang, X.; Keene, D.R.; Höök, A.; Gurusiddappa, S.; Höök, M.; Narayana, S.V. A ‘Collagen Hug’ model for Staphylococcus aureus CNA binding to collagen. EMBO J. 2005, 24, 4224–4236. [Google Scholar] [CrossRef] [PubMed]
- Nguyen, T.; Ghebrehiwet, B.; Peerschke, E.I. Staphylococcus aureus protein A recognizes platelet gC1qR/p33: A novel mechanism for staphylococcal interactions with platelets. Infect. Immun. 2000, 68, 2061–2068. [Google Scholar] [CrossRef]
- Cucarella, C.; Solano, C.; Valle, J.; Amorena, B.; Lasa, I.; Penadés, J.R. Bap, a Staphylococcus aureus surface protein involved in biofilm formation. J. Bacteriol. 2001, 183, 2888–2896. [Google Scholar] [CrossRef]
- Corrigan, R.M.; Rigby, D.; Handley, P.; Foster, T.J. The role of Staphylococcus aureus surface protein SasG in adherence and biofilm formation. Microbiology 2007, 153, 2435–2446. [Google Scholar] [CrossRef]
- Kennedy, C.A.; O’Gara, J.P. Contribution of culture media and chemical properties of polystyrene tissue culture plates to biofilm development by Staphylococcus aureus. J. Med. Microbiol. 2004, 53, 1171–1173. [Google Scholar] [CrossRef]
- Gross, M.; Cramton, S.E.; Götz, F.; Peschel, A. Key role of teichoic acid net charge in Staphylococcus aureus colonization of artificial surfaces. Infect. Immun. 2001, 69, 3423–3426. [Google Scholar] [CrossRef]
- Biswas, R.; Voggu, L.; Simon, U.K.; Hentschel, P.; Thumm, G.; Götz, F. Activity of the major staphylococcal autolysin Atl. FEMS Microbiol. Lett. 2006, 259, 260–268. [Google Scholar] [CrossRef]
- Speziale, P.; Pietrocola, G.; Foster, T.J.; Geoghegan, J.A. Protein-based biofilm matrices in staphylococci. Front. Cell. Infect. Microbiol. 2014, 4, 171. [Google Scholar] [CrossRef] [PubMed]
- Foster, T.J.; Geoghegan, J.A.; Ganesh, V.K.; Höök, M. Adhesion, invasion and evasion: The many functions of the surface proteins of Staphylococcus aureus. Nat. Rev. Microbiol. 2014, 12, 49–62. [Google Scholar] [CrossRef] [PubMed]
- Busscher, H.J.; van der Mei, H.C. How do bacteria know they are on a surface and regulate their response to an adhering state? PLoS Pathog. 2012, 8, e1002440. [Google Scholar] [CrossRef]
- Salgado-Pabón, W.; Schlievert, P.M. Models matter: The search for an effective Staphylococcus aureus vaccine. Nat. Rev. Microbiol. 2014, 12, 585–591. [Google Scholar] [CrossRef] [PubMed]
- Moormeier, D.E.; Bayles, K.W. Staphylococcus aureus biofilm: A complex developmental organism. Mol. Microbiol. 2017, 104, 365–376. [Google Scholar] [CrossRef] [PubMed]
- Mann, E.E.; Rice, K.C.; Boles, B.R.; Endres, J.L.; Ranjit, D.; Chandramohan, L.; Tsang, L.H.; Smeltzer, M.S.; Horswill, A.R.; Bayles, K.W. Modulation of eDNA release and degradation affects Staphylococcus aureus biofilm maturation. PLoS ONE 2009, 4, e5822. [Google Scholar] [CrossRef]
- Kiedrowski, M.R.; Kavanaugh, J.S.; Malone, C.L.; Mootz, J.M.; Voyich, J.M.; Smeltzer, M.S.; Bayles, K.W.; Horswill, A.R. Nuclease modulates biofilm formation in community-associated methicillin-resistant Staphylococcus aureus. PLoS ONE 2011, 6, e26714. [Google Scholar] [CrossRef]
- Ma, L.; Conover, M.; Lu, H.; Parsek, M.R.; Bayles, K.; Wozniak, D.J. Assembly and development of the Pseudomonas aeruginosa biofilm matrix. PLoS Pathog. 2009, 5, e1000354. [Google Scholar] [CrossRef]
- Rohde, H.; Burandt, E.C.; Siemssen, N.; Frommelt, L.; Burdelski, C.; Wurster, S.; Scherpe, S.; Davies, A.P.; Harris, L.G.; Horstkotte, M.A.; et al. Polysaccharide intercellular adhesin or protein factors in biofilm accumulation of Staphylococcus epidermidis and Staphylococcus aureus isolated from prosthetic hip and knee joint infections. Biomaterials 2007, 28, 1711–1720. [Google Scholar] [CrossRef]
- Pozzi, C.; Waters, E.M.; Rudkin, J.K.; Schaeffer, C.R.; Lohan, A.J.; Tong, P.; Loftus, B.J.; Pier, G.B.; Fey, P.D.; Massey, R.C.; et al. Methicillin resistance alters the biofilm phenotype and attenuates virulence in Staphylococcus aureus device-associated infections. PLoS Pathog. 2012, 8, e1002626. [Google Scholar] [CrossRef]
- Wireman, J.W.; Dworkin, M. Developmentally induced autolysis during fruiting body formation by Myxococcus xanthus. J. Bacteriol. 1977, 129, 798–802. [Google Scholar] [CrossRef] [PubMed]
- Sugimoto, S.; Sato, F.; Miyakawa, R.; Chiba, A.; Onodera, S.; Hori, S.; Mizunoe, Y. Broad impact of extracellular DNA on biofilm formation by clinically isolated methicillin-resistant and -sensitive strains of Staphylococcus aureus. Sci. Rep. 2018, 8, 2254. [Google Scholar] [CrossRef]
- Mack, D.; Fischer, W.; Krokotsch, A.; Leopold, K.; Hartmann, R.; Egge, H.; Laufs, R. The intercellular adhesin involved in biofilm accumulation of Staphylococcus epidermidis is a linear beta-1,6-linked glucosaminoglycan: Purification and structural analysis. J. Bacteriol. 1996, 178, 175–183. [Google Scholar] [CrossRef] [PubMed]
- Vuong, C.; Kocianova, S.; Voyich, J.M.; Yao, Y.; Fischer, E.R.; DeLeo, F.R.; Otto, M. A crucial role for exopolysaccharide modification in bacterial biofilm formation, immune evasion, and virulence. J. Biol. Chem. 2004, 279, 54881–54886. [Google Scholar] [CrossRef] [PubMed]
- Fitzpatrick, F.; Humphreys, H.; O’Gara, J.P. Evidence for icaADBC-independent biofilm development mechanism in methicillin-resistant Staphylococcus aureus clinical isolates. J. Clin. Microbiol. 2005, 43, 1973–1976. [Google Scholar] [CrossRef] [PubMed]
- Brooks, J.L.; Jefferson, K.K. Phase variation of poly-N-acetylglucosamine expression in Staphylococcus aureus. PLoS Pathog. 2014, 10, e1004292. [Google Scholar] [CrossRef] [PubMed]
- O’Toole, G.; Kaplan, H.B.; Kolter, R. Biofilm formation as microbial development. Annu. Rev. Microbiol. 2000, 54, 49–79. [Google Scholar] [CrossRef]
- Periasamy, S.; Joo, H.-S.; Duong, A.C.; Bach, T.-H.L.; Tan, V.Y.; Chatterjee, S.S.; Cheung, G.Y.; Otto, M. How Staphylococcus aureus biofilms develop their characteristic structure. Proc. Natl. Acad. Sci. USA 2012, 109, 1281–1286. [Google Scholar] [CrossRef]
- Moormeier, D.E.; Endres, J.L.; Mann, E.E.; Sadykov, M.R.; Horswill, A.R.; Rice, K.C.; Fey, P.D.; Bayles, K.W. Use of microfluidic technology to analyze gene expression during Staphylococcus aureus biofilm formation reveals distinct physiological niches. Appl. Environ. Microbiol. 2013, 79, 3413–3424. [Google Scholar] [CrossRef]
- Simões, M.; Simões, L.C.; Vieira, M.J. A review of current and emergent biofilm control strategies. LWT Food Sci. Technol. 2010, 43, 573–583. [Google Scholar] [CrossRef]
- Stewart, P.S.; Franklin, M.J. Physiological heterogeneity in biofilms. Nat. Rev. Microbiol. 2008, 6, 199–210. [Google Scholar] [CrossRef] [PubMed]
- Flemming, H.-C.; Neu, T.R.; Wozniak, D.J. The EPS matrix: The “house of biofilm cells”. J. Bacteriol. 2007, 189, 7945–7947. [Google Scholar] [CrossRef] [PubMed]
- Liaqat, I.; Liaqat, M.; Ali, S.; Ali, N.M.; Haneef, U.; Mirza, S.M.; Tahir, H.M. Biofilm formation, maturation and prevention: A review. J. Bacteriol. Mycol. 2019, 6, 1092. [Google Scholar] [CrossRef]
- Mulcahy, H.; Charron-Mazenod, L.; Lewenza, S. Extracellular DNA chelates cations and induces antibiotic resistance in Pseudomonas aeruginosa biofilms. PLoS Pathog. 2008, 4, e1000213. [Google Scholar] [CrossRef]
- Van Gennip, M.; Christensen, L.D.; Alhede, M.; Phipps, R.; Jensen, P.Ø.; Christophersen, L.; Pamp, S.J.; Moser, C.; Mikkelsen, P.J.; Koh, A.Y.; et al. Inactivation of the rhlA gene in Pseudomonas aeruginosa prevents rhamnolipid production, disabling the protection against polymorphonuclear leukocytes. APMIS 2009, 117, 537–546. [Google Scholar] [CrossRef]
- Harro, J.M.; Peters, B.M.; O’May, G.A.; Archer, N.; Kerns, P.; Prabhakara, R.; Shirtliff, M.E. Vaccine development in Staphylococcus aureus: Taking the biofilm phenotype into consideration. FEMS Immunol. Med. Microbiol. 2010, 59, 306–323. [Google Scholar] [CrossRef]
- Raafat, D.; Otto, M.; Reppschläger, K.; Iqbal, J.; Holtfreter, S. Fighting Staphylococcus aureus biofilms with monoclonal antibodies. Trends Microbiol. 2019, 27, 303–322. [Google Scholar] [CrossRef]
- Hong, Q.; Huo, S.; Tang, H.; Qu, X.; Yue, B. Smart nanomaterials for treatment of biofilm in orthopedic implants. Front. Bioeng. Biotechnol. 2021, 9, 694635. [Google Scholar] [CrossRef]
- Kathju, S.; Nistico, L.; Tower, I.; Lasko, L.A.; Stoodley, P. Bacterial biofilms on implanted suture material are a cause of surgical site infection. Surg. Infect. 2014, 15, 592–600. [Google Scholar] [CrossRef]
- Post, V.; Wahl, P.; Richards, R.G.; Moriarty, T.F. Vancomycin displays time-dependent eradication of mature Staphylococcus aureus biofilms. J. Orthop. Res. 2017, 35, 381–388. [Google Scholar] [CrossRef]
- Ibberson, C.B.; Parlet, C.P.; Kwiecinski, J.; Crosby, H.A.; Meyerholz, D.K.; Horswill, A.R. Hyaluronan modulation impacts Staphylococcus aureus biofilm infection. Infect. Immun. 2016, 84, 1917–1929. [Google Scholar] [CrossRef] [PubMed]
- Urish, K.L.; DeMuth, P.W.; Craft, D.W.; Haider, H.; Davis, C.M., 3rd. Pulse lavage is inadequate at removal of biofilm from the surface of total knee arthroplasty materials. J. Arthroplast. 2014, 29, 1128–1132. [Google Scholar] [CrossRef] [PubMed]
- Donelli, G.; Francolini, I.; Romoli, D.; Guaglianone, E.; Piozzi, A.; Ragunath, C.; Kaplan, J.B. Synergistic activity of dispersin B and cefamandole nafate in inhibition of staphylococcal biofilm growth on polyurethanes. Antimicrob. Agents Chemother. 2007, 51, 2733–2740. [Google Scholar] [CrossRef] [PubMed]
- Gilbert, P.; Evans, D.J.; Brown, M.R. Formation and dispersal of bacterial biofilms in vivo and in situ. J. Appl. Bacteriol. 1993, 74 (Suppl. S22), 67S–78S. [Google Scholar] [CrossRef]
- Otto, M. Staphylococcus Epidermidis—The ‘Accidental’ Pathogen. Nat. Rev. Microbiol. 2009, 7, 555–567. [Google Scholar] [CrossRef]
- Whitchurch, C.B.; Tolker-Nielsen, T.; Ragas, P.C.; Mattick, J.S. Extracellular DNA required for bacterial biofilm formation. Science 2002, 295, 1487. [Google Scholar] [CrossRef]
- Tang, J.; Zhou, R.; Shi, X.; Kang, M.; Wang, H.; Chen, H. Two thermostable nucleases coexisted in Staphylococcus aureus: Evidence from mutagenesis and in vitro expression. FEMS Microbiol. Lett. 2008, 284, 176–183. [Google Scholar] [CrossRef]
- Boles, B.R.; Horswill, A.R. Agr-mediated dispersal of Staphylococcus aureus biofilms. PLoS Pathog. 2008, 4, e1000052. [Google Scholar] [CrossRef]
- Yarwood, J.M.; Bartels, D.J.; Volper, E.M.; Greenberg, E.P. Quorum sensing in Staphylococcus aureus biofilms. J. Bacteriol. 2004, 186, 1838–1850. [Google Scholar] [CrossRef]
- Thoendel, M.; Kavanaugh, J.S.; Flack, C.E.; Horswill, A.R. Peptide signaling in the staphylococci. Chem. Rev. 2011, 111, 117–151. [Google Scholar] [CrossRef]
- Peschel, A.; Otto, M. Phenol-soluble modulins and staphylococcal infection. Nat. Rev. Microbiol. 2013, 11, 667–673. [Google Scholar] [CrossRef] [PubMed]
- Xie, H.; Cook, G.S.; Costerton, J.W.; Bruce, G.; Rose, T.M.; Lamont, R.J. Intergeneric communication in dental plaque biofilms. J. Bacteriol. 2000, 182, 7067–7069. [Google Scholar] [CrossRef] [PubMed]
- Vuong, C.; Saenz, H.L.; Götz, F.; Otto, M. Impact of the agr quorum-sensing system on adherence to polystyrene in Staphylococcus aureus. J. Infect. Dis. 2000, 182, 1688–1693. [Google Scholar] [CrossRef] [PubMed]
- Kavanaugh, J.S.; Horswill, A.R. Impact of environmental cues on staphylococcal quorum sensing and biofilm development. J. Biol. Chem. 2016, 291, 12556–12564. [Google Scholar] [CrossRef] [PubMed]
- Reen, F.J.; Gutiérrez-Barranquero, J.A.; Parages, M.L.; O’Garra, F. Coumarin: A novel player in microbial quorum sensing and biofilm formation inhibition. Appl. Microbiol. Biotechnol. 2018, 102, 2063–2073. [Google Scholar] [CrossRef]
- Miller, M.B.; Bassler, B.L. Quorum sensing in bacteria. Annu. Rev. Microbiol. 2001, 55, 165–199. [Google Scholar] [CrossRef]
- Reading, N.C.; Sperandio, V. Quorum sensing: The many languages of bacteria. FEMS Microbiol. Lett. 2006, 254, 1–11. [Google Scholar] [CrossRef]
- Banat, I.M.; De Rienzo, M.A.; Quinn, G.A. Microbial biofilms: Biosurfactants as antibiofilm agents. Appl. Microbiol. Biotechnol. 2014, 98, 9915–9929. [Google Scholar] [CrossRef]
- Queck, S.Y.; Jameson-Lee, M.; Villaruz, A.E.; Bach, T.H.; Khan, B.A.; Sturdevant, D.E.; Ricklefs, S.M.; Li, M.; Otto, M. RNAIII-independent target gene control by the agr quorum-sensing system: Insight into the evolution of virulence regulation in Staphylococcus aureus. Mol. Cell 2008, 32, 150–158. [Google Scholar] [CrossRef]
- Suresh, M.K.; Biswas, R.; Biswas, L. An update on recent developments in the prevention and treatment of Staphylococcus aureus biofilms. Int. J. Med. Microbiol. 2019, 309, 1–12. [Google Scholar] [CrossRef]
- Hengzhuang, W.; Wu, H.; Ciofu, O.; Song, Z.; Høiby, N. Pharmacokinetics/pharmacodynamics of colistin and imipenem on mucoid and nonmucoid Pseudomonas aeruginosa biofilms. Antimicrob. Agents Chemother. 2011, 55, 4469–4474. [Google Scholar] [CrossRef] [PubMed]
- Høiby, N.; Ciofu, O.; Johansen, H.K.; Song, Z.-J.; Moser, C.; Jensen, P.Ø.; Molin, S.; Givskov, M.; Tolker-Nielsen, T.; Bjarnsholt, T. The clinical impact of bacterial biofilms. Int. J. Oral Sci. 2011, 3, 55–65. [Google Scholar] [CrossRef] [PubMed]
- Ciofu, O.; Tolker-Nielsen, T. Tolerance and resistance of Pseudomonas aeruginosa biofilms to antimicrobial agents—How P. aeruginosa can escape antibiotics. Front. Microbiol. 2019, 10, 913. [Google Scholar] [CrossRef]
- Chen, M.; Yu, Q.; Sun, H. Novel strategies for the prevention and treatment of biofilm related infections. Int. J. Mol. Sci. 2013, 14, 18488–18501. [Google Scholar] [CrossRef]
- Wu, H.; Moser, C.; Wang, H.-Z.; Høiby, N.; Song, Z.-J. Strategies for combating bacterial biofilm infections. Int. J. Oral Sci. 2015, 7, 1–7. [Google Scholar] [CrossRef] [PubMed]
- Høiby, N. Recent advances in the treatment of Pseudomonas aeruginosa infections in cystic fibrosis. BMC Med. 2011, 9, 32. [Google Scholar] [CrossRef] [PubMed]
- Sun, F.; Qu, F.; Ling, Y.; Mao, P.; Xia, P.; Chen, H.; Zhou, D. Biofilm-associated infections: Antibiotic resistance and novel therapeutic strategies. Future Microbiol. 2013, 8, 877–886. [Google Scholar] [CrossRef] [PubMed]
- Ciofu, O.; Rojo-Molinero, E.; Macià, M.D.; Oliver, A. Antibiotic treatment of biofilm infections. APMIS 2017, 125, 304–319. [Google Scholar] [CrossRef]
- Flemming, H.C.; Wingender, J.; Szewzyk, U.; Steinberg, P.; Rice, S.A.; Kjelleberg, S. Biofilms: An emergent form of bacterial life. Nat. Rev. Microbiol. 2016, 14, 563–575. [Google Scholar] [CrossRef]
- Ranall, M.V.; Butler, M.S.; Blaskovich, M.A.; Cooper, M.A. Resolving biofilm infections: Current therapy and drug discovery strategies. Curr. Drug Targets 2012, 13, 1375–1385. [Google Scholar] [CrossRef]
- Lebeaux, D.; Ghigo, J.M.; Beloin, C. Biofilm-related infections: Bridging the gap between clinical management and fundamental aspects of recalcitrance toward antibiotics. Microbiol. Mol. Biol. Rev. 2014, 78, 510–543. [Google Scholar] [CrossRef] [PubMed]
- Stewart, P.S.; Costerton, J.W. Antibiotic resistance of bacteria in biofilms. Lancet 2001, 358, 135–138. [Google Scholar] [CrossRef]
- Williams, I.; Venables, W.A.; Lloyd, D.; Paul, F.; Critchley, I. The effects of adherence to silicone surfaces on antibiotic susceptibility in Staphylococcus aureus. Microbiology 1997, 143, 2407–2413. [Google Scholar] [CrossRef]
- Andrews, J.M. Determination of minimum inhibitory concentrations. J. Antimicrob. Chemother. 2001, 48 (Suppl. S1), 5–16. [Google Scholar] [CrossRef] [PubMed]
- Thieme, L.; Hartung, A.; Tramm, K.; Klinger-Strobel, M.; Jandt, K.D.; Makarewicz, O.; Pletz, M.W. MBEC versus MBIC: The lack of differentiation between biofilm reducing and inhibitory effects as a current problem in biofilm methodology. Biol. Proced. Online 2019, 21, 18. [Google Scholar] [CrossRef]
- Howlin, R.P.; Brayford, M.J.; Webb, J.S.; Cooper, J.J.; Aiken, S.S.; Stoodley, P. Antibiotic-loaded synthetic calcium sulfate beads for prevention of bacterial colonization and biofilm formation in periprosthetic infections. Antimicrob. Agents Chemother. 2015, 59, 111–120. [Google Scholar] [CrossRef] [PubMed]
- Okeke, I.N.; Lamikanra, A. Export of antimicrobial drugs by West African travelers. J. Travel Med. 2003, 10, 133–135. [Google Scholar] [CrossRef] [PubMed][Green Version]
- Kot, B.; Wierzchowska, K.; Piechota, M.; Grużewska, A. Antimicrobial resistance patterns in methicillin-resistant Staphylococcus aureus from patients hospitalized during 2015–2017 in hospitals in Poland. Med. Princ. Pract. 2020, 29, 61–68. [Google Scholar] [CrossRef]
- Brady, A.J.; Laverty, G.; Gilpin, D.F.; Kearney, P.; Tunney, M. Antibiotic susceptibility of planktonic- and biofilm-grown staphylococci isolated from implant-associated infections: Should MBEC and nature of biofilm formation replace MIC? J. Med. Microbiol. 2017, 66, 461–469. [Google Scholar] [CrossRef]
- Antunes, A.L.; Trentin, D.S.; Bonfanti, J.W.; Pinto, C.C.; Perez, L.R.; Macedo, A.J.; Barth, A.L. Application of a feasible method for determination of biofilm antimicrobial susceptibility in staphylococci. APMIS 2010, 118, 873–877. [Google Scholar] [CrossRef]
- Coenye, T.; Goeres, D.; Van Bambeke, F.; Bjarnsholt, T. Should standardized susceptibility testing for microbial biofilms be introduced in clinical practice? Clin. Microbiol. Infect. 2018, 24, 570–572. [Google Scholar] [CrossRef]
- Ghasemian, A.; Najar Peerayeh, S.; Bakhshi, B.; Mirzaee, M. Comparison of biofilm formation between methicillin-resistant and methicillin-susceptible isolates of Staphylococcus aureus. Iran. Biomed. J. 2016, 20, 175–181. [Google Scholar] [CrossRef] [PubMed]
- Stapleton, P.D.; Taylor, P.W. Methicillin resistance in Staphylococcus aureus: Mechanisms and modulation. Sci. Prog. 2002, 85, 57–72. [Google Scholar] [CrossRef] [PubMed]
- Rezaei, M.; Moniri, R.; Mousavi, S.; Jabbari Shiadeh, S.M. Prevalence of biofilm formation among methicillin resistance Staphylococcus aureus isolated from nasal carriers. Jundishapur J. Microbiol. 2013, 6, e9601. [Google Scholar] [CrossRef]
- Watanakunakorn, C. Mode of action and in-vitro activity of vancomycin. J. Antimicrob. Chemother. 1984, 14 (Suppl. D), 7–18. [Google Scholar] [CrossRef]
- Reynolds, P.E. Structure, biochemistry and mechanism of action of glycopeptide antibiotics. Eur. J. Clin. Microbiol. Infect. Dis. 1989, 8, 943–950. [Google Scholar] [CrossRef]
- Costerton, J.W.; Stewart, P.S.; Greenberg, E.P. Bacterial biofilms: A common cause of persistent infections. Science 1999, 284, 1318–1322. [Google Scholar] [CrossRef]
- Howden, B.P.; Davies, J.K.; Johnson, P.D.; Stinear, T.P.; Grayson, M.L. Reduced vancomycin susceptibility in Staphylococcus aureus, including vancomycin-intermediate and heterogeneous vancomycin-intermediate strains: Resistance mechanisms, laboratory detection, and clinical implications. Clin. Microbiol. Rev. 2010, 23, 99–139. [Google Scholar] [CrossRef]
- Vergidis, P.; Rouse, M.S.; Euba, G.; Karau, M.J.; Schmidt, S.M.; Mandrekar, J.N.; Steckelberg, J.M.; Patel, R. Treatment with linezolid or vancomycin in combination with rifampin is effective in an animal model of methicillin-resistant Staphylococcus aureus foreign body osteomyelitis. Antimicrob. Agents Chemother. 2011, 55, 1182–1186. [Google Scholar] [CrossRef]
- Peixoto, P.B.; Massinhani, F.H.; Netto Dos Santos, K.R.; Chamon, R.C.; Silva, R.B.; Lopes Correa, F.E.; Barata Oliveira, C.; Oliveira, A.G. Methicillin-resistant Staphylococcus epidermidis isolates with reduced vancomycin susceptibility from bloodstream infections in a neonatal intensive care unit. J. Med. Microbiol. 2020, 69, 41–45. [Google Scholar] [CrossRef]
- Antunes, A.L.; Bonfanti, J.W.; Perez, L.R.; Pinto, C.C.; Freitas, A.L.; Macedo, A.J.; Barth, A.L. High vancomycin resistance among biofilms produced by Staphylococcus species isolated from central venous catheters. Mem. Inst. Oswaldo Cruz 2011, 106, 51–55. [Google Scholar] [CrossRef] [PubMed]
- Burmølle, M.; Thomsen, T.R.; Fazli, M.; Dige, I.; Christensen, L.; Homøe, P.; Tvede, M.; Nyvad, B.; Tolker-Nielsen, T.; Givskov, M.; et al. Biofilms in chronic infections—A matter of opportunity—Monospecies biofilms in multispecies infections. FEMS Immunol. Med. Microbiol. 2010, 59, 324–336. [Google Scholar] [CrossRef] [PubMed]
- Hamad, T.; Hellmark, B.; Nilsdotter-Augustinsson, Å.; Söderquist, B. Antibiotic susceptibility among Staphylococcus epidermidis isolated from prosthetic joint infections, with focus on doxycycline. APMIS 2015, 123, 1055–1060. [Google Scholar] [CrossRef] [PubMed]
- Kotulová, D.; Slobodníková, L. Susceptibility of Staphylococcus aureus biofilms to vancomycin, gemtamicin and rifampin. J. Epidemiol. Mikrobiol. Imunol. 2010, 59, 80–87. [Google Scholar]
- Saginur, R.; Stdenis, M.; Ferris, W.; Aaron, S.D.; Chan, F.; Lee, C.; Ramotar, K. Multiple combination bactericidal testing of staphylococcal biofilms from implant-associated infections. Antimicrob. Agents Chemother. 2006, 50, 55–61. [Google Scholar] [CrossRef]
- Raad, I.; Hanna, H.; Jiang, Y.; Dvorak, T.; Reitzel, R.; Chaiban, G.; Sherertz, R.; Hachem, R. Comparative activities of daptomycin, linezolid, and tigecycline against catheter-related methicillin-resistant Staphylococcus bacteremic isolates embedded in biofilm. Antimicrob. Agents Chemother. 2007, 51, 1656–1660. [Google Scholar] [CrossRef]
- Smith, K.; Perez, A.; Ramage, G.; Gemmell, C.G.; Lang, S. Comparison of biofilm-associated cell survival following in vitro exposure of meticillin-resistant Staphylococcus aureus biofilms to the antibiotics clindamycin, daptomycin, linezolid, tigecycline and vancomycin. Int. J. Antimicrob. Agents 2009, 33, 374–378. [Google Scholar] [CrossRef]
- Cafiso, V.; Bertuccio, T.; Spina, D.; Purrello, S.; Stefani, S. Tigecycline inhibition of a mature biofilm in clinical isolates of Staphylococcus aureus: Comparison with other drugs. FEMS Immunol. Med. Microbiol. 2010, 59, 466–469. [Google Scholar] [CrossRef]
- Wang, L.-S.; Gupta, A.; Rotello, V.M. Nanomaterials for the treatment of bacterial biofilms. ACS Infect. Dis. 2016, 2, 3–4. [Google Scholar] [CrossRef]
- Gupta, A.; Landis, R.F.; Rotello, V.M. Nanoparticle-based antimicrobials: Surface functionality is critical. F1000Research 2016, 5, 364. [Google Scholar] [CrossRef]
- Kirui, D.K.; Weber, G.; Talackine, J.; Millenbaugh, N.J. Targeted laser therapy synergistically enhances efficacy of antibiotics against multi-drug resistant Staphylococcus aureus and Pseudomonas aeruginosa biofilms. Nanomedicine 2019, 20, 102018. [Google Scholar] [CrossRef] [PubMed]
- Saggu, S.K.; Jha, G.; Mishra, P.C. Enzymatic degradation of biofilm by metalloprotease from Microbacterium sp. SKS10. Front. Bioeng. Biotechnol. 2019, 7, 192. [Google Scholar] [CrossRef] [PubMed]
- Hrvatin, V. Combating antibiotic resistance: New drugs or alternative therapies? Can. Med. Assoc. J. 2017, 189, E1199. [Google Scholar] [CrossRef][Green Version]
- Khan, J.; Tarar, S.M.; Gul, I.; Nawaz, U.; Arshad, M. Challenges of antibiotic resistance biofilms and potential combating strategies: A review. 3 Biotech 2021, 11, 169. [Google Scholar] [CrossRef] [PubMed]
- Høiby, N.; Bjarnsholt, T.; Moser, C.; Bassi, G.L.; Coenye, T.; Donelli, G.; Hall-Stoodley, L.; Holá, V.; Imbert, C.; Kirketerp-Møller, K.; et al. ESCMID Study Group for Biofilms and Consulting External Expert Werner Zimmerli. ESCMID guideline for the diagnosis and treatment of biofilm infections 2014. Clin. Microbiol. Infect. 2015, 21 (Suppl. S1), S1–S25. [Google Scholar] [CrossRef] [PubMed]
- Fowler, V.G.; Allen, K.B.; Moreira, E.D.; Moustafa, M.; Isgro, F.; Boucher, H.W.; Corey, G.R.; Carmeli, Y.; Betts, R.; Hartzel, J.S.; et al. Effect of an investigational vaccine for preventing Staphylococcus aureus infections after cardiothoracic surgery: A randomized trial. JAMA 2013, 309, 1368–1378. [Google Scholar] [CrossRef]
- Spellberg, B.; Daum, R. Development of a vaccine against Staphylococcus aureus. Semin. Immunopathol. 2012, 34, 335–348. [Google Scholar] [CrossRef]
- Xiong, Y.Q.; Estellés, A.; Li, L.; Abdelhady, W.; Gonzales, R.; Bayer, A.S.; Tenorio, E.; Leighton, A.; Ryser, S.; Kauvar, L.M. A human biofilm-disrupting monoclonal antibody potentiates antibiotic efficacy in rodent models of both Staphylococcus aureus and Acinetobacter baumannii infections. Antimicrob. Agents Chemother. 2017, 61, e00904-17. [Google Scholar] [CrossRef]
- Lam, H.; Kesselly, A.; Stegalkina, S.; Kleanthous, H.; Yethon, J.A. Antibodies to PhnD inhibit staphylococcal biofilms. Infect. Immun. 2014, 82, 3764–3774. [Google Scholar] [CrossRef][Green Version]
- Otto, M. Novel targeted immunotherapy approaches for staphylococcal infection. Expert Opin. Biol. Ther. 2010, 10, 1049–1059. [Google Scholar] [CrossRef]
- Fowler, V.G.; Proctor, R.A. Where does a Staphylococcus aureus vaccine stand? Clin. Microbiol. Infect. 2014, 20, 66–75. [Google Scholar] [CrossRef] [PubMed]
- Kaplan, J.B.; LoVetri, K.; Cardona, S.T.; Madhyastha, S.; Sadovskaya, I.; Jabbouri, S.; Izano, E.A. Recombinant human DNase I decreases biofilm and increases antimicrobial susceptibility in staphylococci. J. Antibiot. 2012, 65, 73–77. [Google Scholar] [CrossRef] [PubMed]
- Chaignon, P.; Sadovskaya, I.; Ragunah, C.; Ramasubbu, N.; Kaplan, J.B.; Jabbouri, S. Susceptibility of staphylococcal biofilms to enzymatic treatments depends on their chemical composition. Appl. Microbiol. Biotechnol. 2007, 75, 125–132. [Google Scholar] [CrossRef] [PubMed]
- Barzegari, A.; Kheyrolahzadeh, K.; Hosseiniyan Khatibi, S.M.; Sharifi, S.; Memar, M.Y.; Zununi Vahed, S. The battle of probiotics and their derivatives against biofilms. Infect. Drug Resist. 2020, 13, 659–672. [Google Scholar] [CrossRef]
- Sánchez, B.; Delgado, S.; Blanco-Míguez, A.; Lourenço, A.; Gueimonde, M.; Margolles, A. Probiotics, gut microbiota, and their influence on host health and disease. Mol. Nutr. Food Res. 2017, 6, 1600240. [Google Scholar] [CrossRef]
- Iannitti, T.; Palmieri, B. Therapeutical use of probiotic formulations in clinical practice. Clin. Nutr. 2010, 29, 701–725. [Google Scholar] [CrossRef]
- Speranza, B.; Liso, A.; Corbo, M.R. Use of design of experiments to optimize the production of microbial probiotic biofilms. PeerJ 2018, 6, e4826. [Google Scholar] [CrossRef]
- Becirovic, A.; Abdi-Dezfuli, J.F.; Hansen, M.F.; Lie, S.A.; Vasstrand, E.N.; Bolstad, A.I. The effects of a probiotic milk drink on bacterial composition in the supra- and subgingival biofilm: A pilot study. Benef. Microbes 2018, 9, 865–874. [Google Scholar] [CrossRef]
- Wallis, J.K.; Krömker, V.; Paduch, J.-H. Biofilm challenge: Lactic acid bacteria isolated from bovine udders versus staphylococci. Foods 2019, 8, 79. [Google Scholar] [CrossRef]
- Aoudia, N.; Rieu, A.; Briandet, R.; Deschamps, J.; Chluba, J.; Jego, G.; Garrido, C.; Guzzo, J. Biofilms of Lactobacillus plantarum and Lactobacillus fermentum: Effect on stress responses, antagonistic effects on pathogen growth and immunomodulatory properties. Food Microbiol. 2016, 53, 51–59. [Google Scholar] [CrossRef]
- e Silva, S.S.; Carvalho, J.W.P.; Aires, C.P.; Nitschke, M. Disruption of Staphylococcus aureus biofilms using rhamnolipid biosurfactants. J. Dairy Sci. 2017, 100, 7864–7873. [Google Scholar] [CrossRef] [PubMed]
- De Rienzo, M.A.; Martin, P.J. Effect of mono and di-rhamnolipids on biofilms pre-formed by Bacillus subtilis BBK006. Curr. Microbiol. 2016, 73, 183–189. [Google Scholar] [CrossRef] [PubMed]
- Yi, G.; Son, J.; Yoo, J.; Park, C.; Koo, H. Rhamnolipid nanoparticles for in vivo drug delivery and photodynamic therapy. Nanomedicine 2019, 19, 12–21. [Google Scholar] [CrossRef] [PubMed]
- Diaz De Rienzo, M.A.; Stevenson, P.S.; Marchant, R.; Banat, I.M. Effect of biosurfactants on Pseudomonas aeruginosa and Staphylococcus aureus biofilms in a BioFlux channel. Appl. Microbiol. Biotechnol. 2016, 100, 5773–5779. [Google Scholar] [CrossRef] [PubMed]
- Diaz De Rienzo, M.A.; Stevenson, P.S.; Marchant, R.; Banat, I.M. Pseudomonas aeruginosa biofilm disruption using microbial surfactants. J. Appl. Microbiol. 2016, 120, 868–876. [Google Scholar] [CrossRef]
- Warrier, A.; Mazumder, N.; Prabhu, S.; Satyamoorthy, K.; Murali, T.S. Photodynamic therapy to control microbial biofilms. Photodiagnosis Photodyn. Ther. 2021, 33, 102090. [Google Scholar] [CrossRef]
- Bombeccari, G.P.; Guzzi, G.; Gualini, F.; Gualini, S.; Santoro, F.; Spadari, F. Photodynamic therapy to treat periimplantitis. Implant Dent. 2013, 22, 631–638. [Google Scholar] [CrossRef]
- Wainwright, M. Photodynamic antimicrobial chemotherapy (PACT). J. Antimicrob. Chemother. 1998, 42, 13–28. [Google Scholar] [CrossRef]
- Widodo, A.; Spratt, D.; Sousa, V.; Petrie, A.; Donos, N. An in vitro study on disinfection of titanium surfaces. Clin. Oral Implants Res. 2016, 27, 1227–1232. [Google Scholar] [CrossRef]
- Biel, M.A.; Sievert, C.; Usacheva, M.; Teichert, M.; Balcom, J. Antimicrobial photodynamic therapy treatment of chronic recurrent sinusitis biofilms. Int. Forum Allergy Rhinol. 2011, 1, 329–334. [Google Scholar] [CrossRef]
- Zoccolillo, M.L.; Rogers, S.C.; Mang, T.S. Antimicrobial photodynamic therapy of S. mutans biofilms attached to relevant dental materials. Lasers Surg. Med. 2016, 48, 995–1005. [Google Scholar] [CrossRef] [PubMed]
- Li, X.; Huang, W.; Zheng, X.; Chang, S.; Liu, C.; Cheng, Q.; Zhu, S. Synergistic in vitro effects of indocyanine green and ethylenediamine tetraacetate-mediated antimicrobial photodynamic therapy combined with antibiotics for resistant bacterial biofilms in diabetic foot infection. Photodiagnosis Photodyn. Ther. 2019, 25, 300–308. [Google Scholar] [CrossRef] [PubMed]
- Li, L.; Zhao, C.; Zhang, Y.; Yao, J.; Yang, W.; Hu, Q.; Wang, C.; Cao, C. Effect of stable antimicrobial nano-silver packaging on inhibiting mildew and in storage of rice. Food Chem. 2017, 215, 477–482. [Google Scholar] [CrossRef] [PubMed]
- Wang, F.; Hu, Q.; Mugambi Mariga, A.; Cao, C.; Yang, W. Effect of nano packaging on preservation quality of Nanjing 9108 rice variety at high temperature and humidity. Food Chem. 2018, 239, 23–31. [Google Scholar] [CrossRef]
- Elbourne, A.; Cheeseman, S.; Atkin, P.; Truong, N.P.; Syed, N.; Zavabeti, A.; Mohiuddin, M.; Esrafilzadeh, D.; Cozzolino, D.; McConville, C.F.; et al. Antibacterial liquid metals: Biofilm treatment via. magnetic activation. ACS Nano 2020, 14, 802–817. [Google Scholar] [CrossRef]
- Li, S.; Chang, R.; Chen, J.; Mi, G.; Xie, Z.; Webster, T. Novel magnetic nanocomposites combining selenium and iron oxide with excellent anti-biofilm properties. J. Mater. Sci. 2020, 55, 1012–1022. [Google Scholar] [CrossRef]
- Xie, X.; Sun, T.; Xue, J.; Miao, Z.; Yan, X.; Fang, W.; Li, Q.; Tang, R.; Lu, Y.; Tang, L.; et al. Ag nanoparticles cluster with pH-triggered reassembly in targeting antimicrobial applications. Adv. Funct. Mater. 2020, 30, 2000511. [Google Scholar] [CrossRef]
- Rabea, E.I.; Badawy, M.E.; Stevens, C.V.; Smagghe, G.; Steurbaut, W. Chitosan as antimicrobial agent: Applications and mode of action. Biomacromolecules 2003, 4, 1457–1465. [Google Scholar] [CrossRef]
- Albayaty, Y.N.; Thomas, N.; Jambhrunkar, M.; Al-Hawwas, M.; Kral, A.; Thorn, C.R.; Prestidge, C.A. Enzyme responsive copolymer micelles enhance the anti-biofilm efficacy of the antiseptic chlorhexidine. Int. J. Pharm. 2019, 566, 329–341. [Google Scholar] [CrossRef]
- Raju, P.; Arivalagan, P.; Natarajan, S. One-pot fabrication of multifunctional catechin@ZIF-L nanocomposite: Assessment of antibiofilm, larvicidal and photocatalytic activities. J. Photochem. Photobiol. B Biol. 2020, 203, 111774. [Google Scholar] [CrossRef]
- Xie, Y.; Zheng, W.; Jiang, X. Near-infrared light-activated phototherapy by gold nanoclusters for dispersing biofilms. ACS Appl. Mater. Interfaces 2020, 12, 9041–9049. [Google Scholar] [CrossRef] [PubMed]
- Sun, H.; Jiang, C.; Wu, L.; Bai, X.; Zhai, S. Cytotoxicity-related bioeffects induced by nanoparticles: The role of surface chemistry. Front. Bioeng. Biotechnol. 2019, 7, 414. [Google Scholar] [CrossRef] [PubMed]
- Sohaebuddin, S.K.; Thevenot, P.T.; Baker, D.; Eaton, J.W.; Tang, L. Nanomaterial cytotoxicity is composition, size, and cell type dependent. Part. Fibre Toxicol. 2010, 7, 22. [Google Scholar] [CrossRef] [PubMed]
- Harper, D.R.; Parracho, H.M.R.T.; Walker, J.; Sharp, R.; Hughes, G.; Werthén, M.; Lehman, S.; Morales, S. Bacteriophages and biofilms. Antibiotics 2014, 3, 270–284. [Google Scholar] [CrossRef]
- Bai, J.; Kim, Y.-T.; Ryu, S.; Lee, J.-H. Biocontrol and rapid detection of food-borne pathogens using bacteriophages and endolysins. Front. Microbiol. 2016, 7, 474. [Google Scholar] [CrossRef]
- Hu, J.; Miyanaga, K.; Tanji, Y. Diffusion properties of bacteriophages through agarose gel membrane. Biotechnol. Prog. 2010, 26, 1213–1221. [Google Scholar] [CrossRef]
- Cha, Y.; Son, B.; Ryu, S. Effective removal of staphylococcal biofilms on various food contact surfaces by Staphylococcus aureus phage endolysin LysCSA13. Food Microbiol. 2019, 84, 103245. [Google Scholar] [CrossRef]
- Fenton, M.; Ross, R.P.; McAuliffe, O.; O’Mahony, J.; Coffey, A. Characterization of the staphylococcal bacteriophage lysin CHAP(K). J. Appl. Microbiol. 2011, 111, 1025–1035. [Google Scholar] [CrossRef]
- Gutiérrez, D.; Ruas-Madiedo, P.; Martinez, B.; Rodríguez, A.; Garcia, P. Effective removal of staphylococcal biofilms by the endolysin LysH5. PLoS ONE 2014, 9, e107307. [Google Scholar] [CrossRef]
- Sass, P.; Bierbaum, G. Lytic activity of recombinant bacteriophage phi11 and phi12 endolysins on whole cells and biofilms of Staphylococcus aureus. Appl. Environ. Microbiol. 2007, 73, 347–352. [Google Scholar] [CrossRef]
- Vázquez, R.; Domenech, M.; Iglesias-Bexiga, M.; Menéndez, M.; García, P. Csl2, a novel chimeric bacteriophage lysin to fight infections caused by Streptococcus suis, an emerging zoonotic pathogen. Sci. Rep. 2017, 7, 16506. [Google Scholar] [CrossRef] [PubMed]
- Gutiérrez, D.; Briers, Y.; Rodríguez-Rubio, L.; Martínez, B.; Rodríguez, A.; Lavigne, R.; García, P. Role of the pre-neck appendage protein (Dpo7) from phage vB_SepiS-phiIPLA7 as an anti-biofilm agent in staphylococcal species. Front. Microbiol. 2015, 6, 1315. [Google Scholar] [CrossRef] [PubMed]
- Guo, Z.; Huang, J.; Yan, G.; Lei, L.; Wang, S.; Yu, L.; Zhou, L.; Gao, A.; Feng, X.; Han, W.; et al. Identification and characterization of Dpo42, a novel depolymerase derived from the Escherichia coli phage vB_EcoM_ECOO78. Front. Microbiol. 2017, 8, 1460. [Google Scholar] [CrossRef] [PubMed]
- Lu, T.K.; Collins, J.J. Dispersing biofilms with engineered enzymatic bacteriophage. Proc. Natl. Acad. Sci. USA 2007, 104, 11197–11202. [Google Scholar] [CrossRef]
- Tinoco, J.M.; Buttaro, B.; Zhang, H.; Liss, N.; Sassone, L.; Stevens, R. Effect of a genetically engineered bacteriophage on Enterococcus faecalis biofilms. Arch. Oral Biol. 2016, 71, 80–86. [Google Scholar] [CrossRef]
- Motlagh, A.M.; Bhattacharjee, A.S.; Goel, R. Biofilm control with natural and genetically-modified phages. World J. Microbiol. Biotechnol. 2016, 32, 67. [Google Scholar] [CrossRef]
- Torres-Barceló, C.; Hochberg, M.E. Evolutionary rationale for phages as complements of antibiotics. Trends Microbiol. 2016, 24, 249–256. [Google Scholar] [CrossRef]
- Tkhilaishvili, T.; Lombardi, L.; Klatt, A.-B.; Trampuz, A.; Di Luca, M. Bacteriophage Sb-1 enhances antibiotic activity against biofilm, degrades exopolysaccharide matrix and targets persisters of Staphylococcus aureus. Int. J. Antimicrob. Agents 2018, 52, 842–853. [Google Scholar] [CrossRef]
- Coulter, L.B.; McLean, R.J.; Rohde, R.E.; Aron, G.M. Effect of bacteriophage infection in combination with tobramycin on the emergence of resistance in Escherichia coli and Pseudomonas aeruginosa biofilms. Viruses 2014, 6, 3778–3786. [Google Scholar] [CrossRef]
- Yasir, M.; Willcox, M.D.P.; Dutta, D. Action of antimicrobial peptides against bacterial biofilms. Materials 2018, 11, 2468. [Google Scholar] [CrossRef]
- Bahar, A.A.; Ren, D. Antimicrobial peptides. Pharmaceuticals 2013, 6, 1543–1575. [Google Scholar] [CrossRef] [PubMed]
- de la Fuente-Núñez, C.; Korolik, V.; Bains, M.; Nguyen, U.; Breidenstein, E.B.; Horsman, S.; Lewenza, S.; Burrows, L.; Hancock, R.E. Inhibition of bacterial biofilm formation and swarming motility by a small synthetic cationic peptide. Antimicrob. Agents Chemother. 2012, 56, 2696–2704. [Google Scholar] [CrossRef] [PubMed]
- Nagant, C.; Pitts, B.; Nazmi, K.; Vandenbranden, M.; Bolscher, J.G.; Stewart, P.S.; Dehaye, J.P. Identification of peptides derived from the human antimicrobial peptide LL-37 active against biofilms formed by Pseudomonas aeruginosa using a library of truncated fragments. Antimicrob. Agents Chemother. 2012, 56, 5698–5708. [Google Scholar] [CrossRef] [PubMed]
- Haisma, E.M.; de Breij, A.; Chan, H.; van Dissel, J.T.; Drijfhout, J.W.; Hiemstra, P.S.; El Ghalbzouri, A.; Nibbering, P.H. LL-37-derived peptides eradicate multidrug-resistant Staphylococcus aureus from thermally wounded human skin equivalents. Antimicrob. Agents Chemother. 2014, 58, 4411–4419. [Google Scholar] [CrossRef]
- Dean, S.N.; Bishop, B.M.; van Hoek, M.L. Susceptibility of Pseudomonas aeruginosa biofilm to alpha-helical peptides: D-enantiomer of LL-37. Front. Microbiol. 2011, 2, 128. [Google Scholar] [CrossRef]
- Mataraci, E.; Dosler, S. In vitro activities of antibiotics and antimicrobial cationic peptides alone and in combination against methicillin-resistant Staphylococcus aureus biofilms. Antimicrob. Agents Chemother. 2012, 56, 6366–6371. [Google Scholar] [CrossRef]
- Bacalum, M.; Radu, M. Cationic antimicrobial peptides cytotoxicity on mammalian cells: An analysis using therapeutic index integrative concept. Int. J. Pept. Res. Ther. 2015, 21, 47–55. [Google Scholar] [CrossRef]
- de la Fuente-Núñez, C.; Cardoso, M.H.; de Souza Cândido, E.; Franco, O.L.; Hancock, R.E.W. Synthetic antibiofilm peptides. Biochim. Biophys. Acta 2016, 1858, 1061–1069. [Google Scholar] [CrossRef]
- Kong, C.; Chee, C.-F.; Richter, K.; Thomas, N.; Rahman, N.; Nathan, S. Suppression of Staphylococcus aureus biofilm formation and virulence by a benzimidazole derivative, UM-C162. Sci. Rep. 2018, 8, 2758. [Google Scholar] [CrossRef]
- Yamasaki, R.; Kawano, A.; Yoshioka, Y.; Ariyoshi, W. Rhamnolipids and surfactin inhibit the growth or formation of oral bacterial biofilm. BMC Microbiol. 2020, 20, 358. [Google Scholar] [CrossRef]
- Monte, J.; Abreu, A.C.; Borges, A.; Simões, L.C.; Simões, M. Antimicrobial activity of selected phytochemicals against Escherichia coli and Staphylococcus aureus and their Biofilms. Pathogens 2014, 3, 473–498. [Google Scholar] [CrossRef] [PubMed]
- Borges, A.; Abreu, A.C.; Dias, C.; Saavedra, M.J.; Borges, F.; Simões, M. New perspectives on the use of phytochemicals as an emergent strategy to control bacterial infections including biofilms. Molecules 2016, 21, 877. [Google Scholar] [CrossRef] [PubMed]
- Wu, X.; Wang, Y.; Tao, L. Sulfhydryl compounds reduce Staphylococcus aureus biofilm formation by inhibiting PIA biosynthesis. FEMS Microbiol. Lett. 2011, 316, 44–50. [Google Scholar] [CrossRef] [PubMed]
- Francis, N.C.; Yao, W.; Grundfest, W.S.; Taylor, Z.D. Laser-generated shockwaves as a treatment to reduce bacterial load and disrupt biofilm. IEEE Trans. Biomed. Eng. 2017, 64, 882–889. [Google Scholar] [CrossRef]
- Percival, S.L.; Francolini, I.; Donelli, G. Low-level laser therapy as an antimicrobial and antibiofilm technology and its relevance to wound healing. Fut. Microbiol. 2015, 10, 255–272. [Google Scholar] [CrossRef]
- Fanaei Pirlar, R.; Emaneini, M.; Beigverdi, R.; Banar, M.B.; van Leeuwen, W.; Jabalameli, F. Combinatorial effects of antibiotics and enzymes against dual-species Staphylococcus aureus and Pseudomonas aeruginosa biofilms in the wound-like medium. PLoS ONE 2020, 15, e0235093. [Google Scholar] [CrossRef]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Sedarat, Z.; Taylor-Robinson, A.W. Biofilm Formation by Pathogenic Bacteria: Applying a Staphylococcus aureus Model to Appraise Potential Targets for Therapeutic Intervention. Pathogens 2022, 11, 388. https://doi.org/10.3390/pathogens11040388
Sedarat Z, Taylor-Robinson AW. Biofilm Formation by Pathogenic Bacteria: Applying a Staphylococcus aureus Model to Appraise Potential Targets for Therapeutic Intervention. Pathogens. 2022; 11(4):388. https://doi.org/10.3390/pathogens11040388
Chicago/Turabian StyleSedarat, Zahra, and Andrew W. Taylor-Robinson. 2022. "Biofilm Formation by Pathogenic Bacteria: Applying a Staphylococcus aureus Model to Appraise Potential Targets for Therapeutic Intervention" Pathogens 11, no. 4: 388. https://doi.org/10.3390/pathogens11040388
APA StyleSedarat, Z., & Taylor-Robinson, A. W. (2022). Biofilm Formation by Pathogenic Bacteria: Applying a Staphylococcus aureus Model to Appraise Potential Targets for Therapeutic Intervention. Pathogens, 11(4), 388. https://doi.org/10.3390/pathogens11040388