1
|
BAL H, ALTANLAR N, YILDIZ S. The Effect of Sub-Minimal Inhibitory Concentrations of Daptomycin and Linezolid on Biofilm Formation of Methicillin Resistant Staphylococcus aureus Isolated from Clinical Samples. Turk J Pharm Sci 2025; 22:131-139. [PMID: 40366226 PMCID: PMC12080290 DOI: 10.4274/tjps.galenos.2025.26723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 03/28/2025] [Indexed: 05/15/2025]
Abstract
Objectives The aim of this study was to determine the development of in vitro resistance and changes in biofilm forming abilities in methicillin-resistant Staphylococcus aureus (MRSA) isolates exposed to sub-minimal inhibitory concentrations (sub-MICs) of daptomycin and linezolid; and to investigate the presence of the methicillin resistance gene (mecA) and the biofilm-associated genes (icaA, icaD) by polymerase chain reaction. Materials and Methods This study was carried out with thirty-two MRSA isolates. The susceptibility of the isolates to daptomycin and linezolid was investigated by the broth microdilution method, and MIC values were determined (1st MIC). After serial passages, the 2nd MIC and the 3rd MIC values were similarly detected. Before and after serial passages, the biofilm-forming abilities of MRSA isolates were examined using the microtiter plate (MTP) method. Results When the daptomycin and linezolid 1st MIC and 3rd MIC values of the isolates were compared, there was a 2-8 fold increase in linezolid (p<0.05) and a 4-32 fold increase in daptomycin (p<0.05). According to the MTP method, 20 (62.5%) of the 32 isolates formed biofilm at various levels, while 12 (37.5%) did not form biofilm. After the second series of passages, biofilm formation was observed in 19 (59.4%) isolates with daptomycin (p>0.05) and in 16 (50%) isolates with linezolid (p>0.05). The mecA gene was found in all isolates. Also, icaA and icaD genes were detected in 31 (96.9%) of 32 MRSA isolates. Conclusion MRSA isolates exposed to sub-MICs of the antibiotics daptomycin and linezolid were observed to form biofilms at varying levels or to lose their ability to form biofilms. The induction, reduction or eradication of biofilm depended on the type of antibiotic and the MRSA isolate.
Collapse
Affiliation(s)
- Halil BAL
- Sivas Cumhuriyet University Faculty of Pharmacy, Department of Pharmaceutical Microbiology, Sivas, Türkiye
| | - Nurten ALTANLAR
- Ankara University Faculty of Pharmacy, Department of Pharmaceutical Microbiology, Ankara, Türkiye
| | - Sulhiye YILDIZ
- Lokman Hekim University Faculty of Pharmacy, Department of Pharmaceutical Microbiology, Ankara, Türkiye
| |
Collapse
|
2
|
Meenatchi R, Priya S, Shreya C, Gopi S, Rajagopal R, Kaliraj S, Kumaradoss KM, Arockiaraj J. Exploring the Anti-Adherence Potential of Skt35 to Combat Catheter-Associated Staphylococcus aureus Infections: Efficacy, Toxicity and Mechanism of Action. Chem Biodivers 2025; 22:e202402087. [PMID: 39832262 DOI: 10.1002/cbdv.202402087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 12/23/2024] [Accepted: 12/27/2024] [Indexed: 01/22/2025]
Abstract
Catheter-associated urinary tract infections (CAUTIs), often caused by biofilm-forming Staphylococcus aureus, present significant clinical challenges. Skt35, a dioxopiperidinamide derivative of cinnamic acid, was investigated for its potential antibacterial and antibiofilm activities against S. aureus biofilms. The antibacterial effect of Skt35 was assessed using the zone of inhibition and microdilution methods, revealing a minimum inhibitory concentration (MIC) of 250 µM. Antibiofilm properties were confirmed through crystal violet assays, scanning electron microscopy and confocal laser scanning microscopy, showing significant biofilm inhibition at the Sub-MIC. In an in vitro bladder model, Skt35-coated silicone catheter tubes exhibited significant antiadhesive effects. Zebrafish embryo tests indicated no toxicity at concentrations up to 125 µM. Molecular docking and simulation analysis revealed strong binding affinities of Skt35 to Accessory Gene Regulator A (-7.9 kcal/mol) and Lux Small protein (-4.96 kcal/mol), suggesting potential disruption of quorum sensing and gene expression in S. aureus, making it a promising candidate for catheter coatings to prevent CAUTIs.
Collapse
Affiliation(s)
- Ramu Meenatchi
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulatur, India
| | - Snega Priya
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulatur, India
| | - Chakraborty Shreya
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulatur, India
| | - Sanjay Gopi
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulatur, India
| | - Rajakrishnan Rajagopal
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - S Kaliraj
- Department of Chemistry, Faculty of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, India
| | - Kathiravan Muthu Kumaradoss
- Department of Pharmaceutical Chemistry, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, India
| | - Jesu Arockiaraj
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulatur, India
| |
Collapse
|
3
|
Li J, Xie L, Lin F, Ling B. Indole derivatives display antimicrobial and antibiofilm effects against extensively drug-resistant Acinetobacter baumannii. Microbiol Spectr 2025; 13:e0338824. [PMID: 40231681 PMCID: PMC12073863 DOI: 10.1128/spectrum.03388-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 03/24/2025] [Indexed: 04/16/2025] Open
Abstract
Acinetobacter baumannii is a critical priority gram-negative bacterial species featured with multidrug resistance and biofilm formation. This study screened 46 indole derivative agents for their antimicrobial activities against clinical isolates of extensively drug-resistant A. baumannii (XDRAB) with various degrees of biofilm production. Three selected indole agents-5-iodoindole, 3-methylindole, and 7-hydroxyindole-were revealed to display potent antimicrobial and antibiofilm activity, including synergistic interplay with anti-A. baumannii antimicrobial drugs against XDRAB. Sub-inhibitory concentrations of these agents (particularly 7-hydroxyindole at 1/64 of MIC) not only inhibited XDRAB biofilm formation but also eradicated the mature biofilm. The survival rate of XDRAB-infected Galleria mellonella was improved with the treatment of 7-hydroxyindole. Mechanistically, 7-hydroxyindole was found to reduce the expression of quorum sensing/biofilm-implicated genes abaI and abaR. Together, the findings highlight the potential of indole derivatives against A. baumannii infections. IMPORTANCE Extensively drug-resistant Acinetobacter baumannii (XDRAB) isolates pose a major public health threat to antimicrobial therapy and are highly prevalent in hospital settings. This study identified and characterized indole derivative agents for their antimicrobial and antibiofilm activities against XDRAB. Sub-inhibitory indole agents such as 7-hydroxyindole can both inhibit XDRAB biofilm formation and eradicate the mature biofilm. Indole agents warrant further investigation against hard-to-treat antimicrobial-resistant pathogens.
Collapse
Affiliation(s)
- Junwei Li
- Key Laboratory of
Structure-Specific Small Molecule Drugs at Chengdu Medical College of
Sichuan Province, School of Pharmacy, Chengdu Medical
College,
Chengdu, China
| | - Lulin Xie
- Key Laboratory of
Structure-Specific Small Molecule Drugs at Chengdu Medical College of
Sichuan Province, School of Pharmacy, Chengdu Medical
College,
Chengdu, China
| | - Fei Lin
- Department of
Pharmacy, Clinical Medical College and The First Affiliated Hospital of
Chengdu Medical College,
Chengdu, China
| | - Baodong Ling
- Key Laboratory of
Structure-Specific Small Molecule Drugs at Chengdu Medical College of
Sichuan Province, School of Pharmacy, Chengdu Medical
College,
Chengdu, China
| |
Collapse
|
4
|
Subbarayudu S, Snega Priya P, Rajagopal R, Alfarhan A, Guru A, Arockiaraj J. Impact of acidic and alkaline conditions on Staphylococcus aureus and Acinetobacter baumannii interactions and their biofilms. Arch Microbiol 2024; 206:426. [PMID: 39375235 DOI: 10.1007/s00203-024-04142-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/10/2024] [Accepted: 09/14/2024] [Indexed: 10/09/2024]
Abstract
Bacterial biofilms pose significant challenges due to their association with antibiotic resistance, metabolic adaptation, and survival under harsh conditions. Among notable pathogens forming biofilms, Staphylococcus aureus and Acinetobacter baumannii are concerning pathogens in nosocomial settings. However, their behaviour under acidic (pH 4.5) and alkaline (pH10.5) conditions, especially in co-culture setups, remains insufficiently understood. This study investigates these aspects, by examining growth rates, biofilm formation, pH shifts, phenotypic analysis, and gene expression profiles. The results showed A. baumannii exhibited reduced growth and biofilm formation at pH 4.5, while S. aureus showed slow growth and low biofilm formation at pH10.5 in mono-cultures. S. aureus leaned towards an acidic pH (6-6.5), whereas A. baumannii shifted towards an alkaline pH (8-9). In co-culture environments, growth rates and biofilm formation increased across all pH conditions, converging towards a neutral pH over time. Phenotypic motility assays indicated that A. baumannii exhibited greater motility in alkaline conditions, while S. aureus showed increased staphyloxanthin production under acidic conditions. Gene expression analyses revealed that the fibronectin-binding protein A (FnbA) and N-acetylglucosaminyl-transferase (icaA) genes, responsible for initial attachment during biofilm formation, were highly expressed in acidic co-culture condition but poorly expressed in alkaline condition. In A. baumannii, the outer membrane protein A (OmpA) gene associated with adhesion and virulence, was upregulated in co-culture. The LuxR gene involved in quorum sensing was upregulated in acidic conditions and poorly expressed at pH 10.5. This study elucidates the metabolic adaptability and biofilm formation tendencies of S. aureus towards acidic conditions and A. baumannii towards alkaline conditions, providing insights for better management of biofilm-related infections.
Collapse
Affiliation(s)
- Suthi Subbarayudu
- Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Chennai, Tamil Nadu, 603203, India
| | - P Snega Priya
- Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Chennai, Tamil Nadu, 603203, India
| | - Rajakrishnan Rajagopal
- Department of Botany and Microbiology, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Ahmed Alfarhan
- Department of Botany and Microbiology, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Ajay Guru
- Department of Cariology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
| | - Jesu Arockiaraj
- Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Chennai, Tamil Nadu, 603203, India.
| |
Collapse
|
5
|
Omar A, El-Banna TE, Sonbol FI, El-Bouseary MM. Potential antivirulence and antibiofilm activities of sub-MIC of oxacillin against MDR S. aureus isolates: an in-vitro and in-vivo study. BMC Microbiol 2024; 24:295. [PMID: 39123138 PMCID: PMC11312681 DOI: 10.1186/s12866-024-03429-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 07/16/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Multi-drug resistant Staphylococcus aureus is one of the most common causes of nosocomial and community-acquired infections, with high morbidity and mortality. Treatment of such infections is particularly problematic; hence, it is complicated by antibiotic resistance, and there is currently no reliable vaccine. Furthermore, it is well known that S. aureus produces an exceptionally large number of virulence factors that worsen infection. Consequently, the urgent need for anti-virulent agents that inhibit biofilm formation and virulence factors has gained momentum. Therefore, we focused our attention on an already-approved antibiotic and explored whether changing the dosage would still result in the intended anti-virulence effect. METHODS In the present study, we determined the antibiotic resistance patterns and the MICs of oxacillin against 70 MDR S. aureus isolates. We also investigated the effect of sub-MICs of oxacillin (at 1/4 and 1/8 MICs) on biofilm formation using the crystal violet assay, the phenol-sulphuric acid method, and confocal laser scanning microscopy (CLSM). We examined the effect of sub-MICs on virulence factors and bacterial morphology using quantitative reverse transcription polymerase chain reaction (qRT-PCR) and electron microscopy, respectively. Moreover, we studied the effect of sub-MICs of oxacillin (OX) in-vivo using a wound infection model. RESULTS Oxacillin at 1/2 MIC showed a significant decrease in bacterial viability, while 1/4 and 1/8 MICs had negligible effects on treated bacterial isolates. Treatment of MDR isolates with 1/4 or 1/8 MICs of oxacillin significantly reduced biofilm formation (64% and 40%, respectively). The treated MDR S. aureus with sub-MICs of OX exhibited a dramatic reduction in several virulence factors, including protease, hemolysin, coagulase, and toxic shock syndrome toxin-1 (TSST-1) production. The sub-MICs of OX significantly decreased (P < 0.05) the gene expression of biofilm and virulence-associated genes such as agrA, icaA, coa, and tst. Furthermore, oxacillin at sub-MICs dramatically accelerated wound healing, according to the recorded scoring of histological parameters. CONCLUSION The treatment of MDR S. aureus with sub-MICs of oxacillin can help in combating the bacterial resistance and may be considered a promising approach to attenuating the severity of S. aureus infections due to the unique anti-biofilm and anti-virulence activities.
Collapse
Affiliation(s)
- Amira Omar
- Department of Microbiology and Immunology, Faculty of Pharmacy, Tanta University, Tanta, Egypt.
| | - Tarek E El-Banna
- Department of Microbiology and Immunology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Fatma I Sonbol
- Department of Microbiology and Immunology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Maisra M El-Bouseary
- Department of Microbiology and Immunology, Faculty of Pharmacy, Tanta University, Tanta, Egypt.
| |
Collapse
|
6
|
van Groesen E, Mons E, Kotsogianni I, Arts M, Tehrani KHME, Wade N, Lysenko V, Stel FM, Zwerus JT, De Benedetti S, Bakker A, Chakraborty P, van der Stelt M, Scheffers DJ, Gooskens J, Smits WK, Holden K, Gilmour PS, Willemse J, Hitchcock CA, van Hasselt JGC, Schneider T, Martin NI. Semisynthetic guanidino lipoglycopeptides with potent in vitro and in vivo antibacterial activity. Sci Transl Med 2024; 16:eabo4736. [PMID: 39110780 DOI: 10.1126/scitranslmed.abo4736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 02/23/2024] [Accepted: 07/16/2024] [Indexed: 08/13/2024]
Abstract
Gram-positive bacterial infections present a major clinical challenge, with methicillin- and vancomycin-resistant strains continuing to be a cause for concern. In recent years, semisynthetic vancomycin derivatives have been developed to overcome this problem as exemplified by the clinically used telavancin, which exhibits increased antibacterial potency but has also raised toxicity concerns. Thus, glycopeptide antibiotics with enhanced antibacterial activities and improved safety profiles are still necessary. We describe the development of a class of highly potent semisynthetic glycopeptide antibiotics, the guanidino lipoglycopeptides, which contain a positively charged guanidino moiety bearing a variable lipid group. These glycopeptides exhibited enhanced in vitro activity against a panel of Gram-positive bacteria including clinically relevant methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant strains, showed minimal toxicity toward eukaryotic cells, and had a low propensity for resistance selection. Mechanistically, guanidino lipoglycopeptides engaged with bacterial cell wall precursor lipid II with a higher binding affinity than vancomycin. Binding to both wild-type d-Ala-d-Ala lipid II and the vancomycin-resistant d-Ala-d-Lac variant was confirmed, providing insight into the enhanced activity of guanidino lipoglycopeptides against vancomycin-resistant isolates. The in vivo efficacy of guanidino lipoglycopeptide EVG7 was evaluated in a S. aureus murine thigh infection model and a 7-day sepsis survival study, both of which demonstrated superiority to vancomycin. Moreover, the minimal to mild kidney effects at supratherapeutic doses of EVG7 indicate an improved therapeutic safety profile compared with vancomycin. These findings position guanidino lipoglycopeptides as candidates for further development as antibacterial agents for the treatment of clinically relevant multidrug-resistant Gram-positive infections.
Collapse
Affiliation(s)
- Emma van Groesen
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2300 RA Leiden, Netherlands
| | - Elma Mons
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2300 RA Leiden, Netherlands
| | - Ioli Kotsogianni
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2300 RA Leiden, Netherlands
| | - Melina Arts
- Institute for Pharmaceutical Microbiology, University Hospital Bonn, University of Bonn, 53113 Bonn, Germany
| | - Kamaleddin H M E Tehrani
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2300 RA Leiden, Netherlands
| | - Nicola Wade
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2300 RA Leiden, Netherlands
| | - Vladyslav Lysenko
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2300 RA Leiden, Netherlands
| | - Florence M Stel
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2300 RA Leiden, Netherlands
| | - Jordy T Zwerus
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2300 RA Leiden, Netherlands
| | - Stefania De Benedetti
- Institute for Pharmaceutical Microbiology, University Hospital Bonn, University of Bonn, 53113 Bonn, Germany
| | - Alexander Bakker
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, 2300 RA Leiden, Netherlands
| | - Parichita Chakraborty
- Department of Molecular Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9700 AB Groningen, Netherlands
| | - Mario van der Stelt
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, 2300 RA Leiden, Netherlands
| | - Dirk-Jan Scheffers
- Department of Molecular Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9700 AB Groningen, Netherlands
| | - Jairo Gooskens
- Department of Medical Microbiology, Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, 2333 ZA Leiden, Netherlands
| | - Wiep Klaas Smits
- Experimental Bacteriology, Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, 2333 ZA Leiden, Netherlands
| | - Kirsty Holden
- Evotec (U.K.) Ltd., Alderley Park, Macclesfield, Cheshire, SK10 4TG UK
| | | | - Joost Willemse
- Institute of Biology Leiden, Leiden University, 2300 RA Leiden, Netherlands
| | | | - J G Coen van Hasselt
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, 2300 RA Leiden, Netherlands
| | - Tanja Schneider
- Institute for Pharmaceutical Microbiology, University Hospital Bonn, University of Bonn, 53113 Bonn, Germany
| | - Nathaniel I Martin
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2300 RA Leiden, Netherlands
| |
Collapse
|
7
|
Amparo TR, Sousa LRD, Xavier VF, Seibert JB, Paiva DL, da Silva DDS, Teixeira LFDM, dos Santos ODH, Vieira PMDA, de Souza GHB, Brandão GC. Protium spruceanum Extract Enhances Mupirocin Activity When Combined with Nanoemulsion-Based Hydrogel: A Multi-Target Strategy for Treating Skin and Soft Tissue Infections. Pharmaceutics 2024; 16:700. [PMID: 38931824 PMCID: PMC11207036 DOI: 10.3390/pharmaceutics16060700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/07/2024] [Accepted: 05/13/2024] [Indexed: 06/28/2024] Open
Abstract
The treatment of skin and soft tissue infections (SSTIs) can be challenging due to bacterial resistance, particularly from strains like MRSA and biofilm formation. However, combining conventional antibiotics with natural products shows promise in treating SSTIs. The objective of this study is to develop a nanoemulsion-based hydrogel containing Protium spruceanum extract and mupirocin and evaluate its potential for the treatment of SSTIs. The nanoemulsion was obtained by phase inversion and subsequently characterized. The antibacterial activity was evaluated in vitro against S. aureus MRSA, including the synergism of the combination, changes in membrane permeability using flow cytometry, and the anti-biofilm effect. In addition, the irritative potential was evaluated by the HET-CAM assay. The combination exhibited synergistic antibacterial activity against S. aureus and MRSA due to the extract enhancing membrane permeability. The hydrogel demonstrated suitable physicochemical properties, inhibited biofilm formation, and exhibited low irritation. The formulation was nanometric (176.0 ± 1.656 nm) and monodisperse (polydispersity index 0.286 ± 0.011). It exhibited a controlled release profile at 48 h and high encapsulation efficacy (94.29 ± 4.54% for quercitrin and 94.20 ± 5.44% for mupirocin). Therefore, these findings suggest that the hydrogel developed could be a safe and effective option for treating SSTIs.
Collapse
Affiliation(s)
- Tatiane Roquete Amparo
- Department of Pharmacy, Federal University of Ouro Preto, Ouro Preto 35400-000, Brazil; (L.R.D.S.); (V.F.X.); (D.L.P.); (D.d.S.d.S.); (O.D.H.d.S.); (G.H.B.d.S.); (G.C.B.)
| | - Lucas Resende Dutra Sousa
- Department of Pharmacy, Federal University of Ouro Preto, Ouro Preto 35400-000, Brazil; (L.R.D.S.); (V.F.X.); (D.L.P.); (D.d.S.d.S.); (O.D.H.d.S.); (G.H.B.d.S.); (G.C.B.)
| | - Viviane Flores Xavier
- Department of Pharmacy, Federal University of Ouro Preto, Ouro Preto 35400-000, Brazil; (L.R.D.S.); (V.F.X.); (D.L.P.); (D.d.S.d.S.); (O.D.H.d.S.); (G.H.B.d.S.); (G.C.B.)
| | - Janaína Brandão Seibert
- Laboratory of Pathology and Microbial Control, University of São Paulo (USP-ESALQ), Piracicaba 13418-900, Brazil;
| | - Débora Luiza Paiva
- Department of Pharmacy, Federal University of Ouro Preto, Ouro Preto 35400-000, Brazil; (L.R.D.S.); (V.F.X.); (D.L.P.); (D.d.S.d.S.); (O.D.H.d.S.); (G.H.B.d.S.); (G.C.B.)
| | - Débora dos Santos da Silva
- Department of Pharmacy, Federal University of Ouro Preto, Ouro Preto 35400-000, Brazil; (L.R.D.S.); (V.F.X.); (D.L.P.); (D.d.S.d.S.); (O.D.H.d.S.); (G.H.B.d.S.); (G.C.B.)
| | | | - Orlando David Henrique dos Santos
- Department of Pharmacy, Federal University of Ouro Preto, Ouro Preto 35400-000, Brazil; (L.R.D.S.); (V.F.X.); (D.L.P.); (D.d.S.d.S.); (O.D.H.d.S.); (G.H.B.d.S.); (G.C.B.)
| | | | - Gustavo Henrique Bianco de Souza
- Department of Pharmacy, Federal University of Ouro Preto, Ouro Preto 35400-000, Brazil; (L.R.D.S.); (V.F.X.); (D.L.P.); (D.d.S.d.S.); (O.D.H.d.S.); (G.H.B.d.S.); (G.C.B.)
| | - Geraldo Célio Brandão
- Department of Pharmacy, Federal University of Ouro Preto, Ouro Preto 35400-000, Brazil; (L.R.D.S.); (V.F.X.); (D.L.P.); (D.d.S.d.S.); (O.D.H.d.S.); (G.H.B.d.S.); (G.C.B.)
| |
Collapse
|
8
|
Reffuveille F, Dghoughi Y, Colin M, Torres MDT, de la Fuente-Nunez C. Antibiofilm approaches as a new paradigm for treating infections. PROGRESS IN BIOMEDICAL ENGINEERING (BRISTOL, ENGLAND) 2024; 6:023001. [PMID: 39506977 PMCID: PMC11540418 DOI: 10.1088/2516-1091/ad1cd6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/08/2024]
Abstract
The lack of effective antibiotics for drug-resistant infections has led the World Health Organization to declare antibiotic resistance a global priority. Most bacterial infections are caused by microbes growing in structured communities called biofilms. Bacteria growing in biofilms are less susceptible to antibiotics than their planktonic counterparts. Despite their significant clinical implications, bacterial biofilms have not received the attention they warrant, with no approved antibiotics specifically designed for their eradication. In this paper, we aim to shed light on recent advancements in antibiofilm strategies that offer compelling alternatives to traditional antibiotics. Additionally, we will briefly explore the potential synergy between computational approaches, including the emerging field of artificial intelligence, and the accelerated design and discovery of novel antibiofilm molecules in the years ahead.
Collapse
Affiliation(s)
- Fany Reffuveille
- Université de Reims Champagne-Ardenne, Biomatériaux et Inflammation en Site Osseux, BIOS EA 4691, SFR Cap Santé, 51097 Reims, France
- Université de Reims Champagne-Ardenne, UFR Pharmacie, Service de Microbiologie, 51097 Reims, France
| | - Yasser Dghoughi
- Université de Reims Champagne-Ardenne, Biomatériaux et Inflammation en Site Osseux, BIOS EA 4691, SFR Cap Santé, 51097 Reims, France
| | - Marius Colin
- Université de Reims Champagne-Ardenne, Biomatériaux et Inflammation en Site Osseux, BIOS EA 4691, SFR Cap Santé, 51097 Reims, France
- Université de Reims Champagne-Ardenne, UFR Pharmacie, Service de Microbiologie, 51097 Reims, France
| | - Marcelo D T Torres
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
- Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States of America
- Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, United States of America
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Cesar de la Fuente-Nunez
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
- Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States of America
- Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, United States of America
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA, United States of America
| |
Collapse
|
9
|
Eckmann C, Sunderkötter C, Becker K, Grabein B, Hagel S, Hanses F, Wichmann D, Thalhammer F. Left ventricular assist device-associated driveline infections as a specific form of complicated skin and soft tissue infection/acute bacterial skin and skin structure infection - issues and therapeutic options. Curr Opin Infect Dis 2024; 37:95-104. [PMID: 38085707 PMCID: PMC10911258 DOI: 10.1097/qco.0000000000000999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
PURPOSE OF REVIEW This review comments on the current guidelines for the treatment of wound infections under definition of acute bacterial skin and skin structure infections (ABSSSI). However, wound infections around a catheter, such as driveline infections of a left ventricular assist device (LVAD) are not specifically listed under this definition in any of the existing guidelines. RECENT FINDINGS Definitions and classification of LVAD infections may vary across countries, and the existing guidelines and recommendations may not be equally interpreted among physicians, making it unclear if these infections can be considered as ABSSSI. Consequently, the use of certain antibiotics that are approved for ABSSSI may be considered as 'off-label' for LVAD infections, leading to rejection of reimbursement applications in some countries, affecting treatment strategies, and hence, patients' outcomes. However, we believe driveline exit site infections related to LVAD can be included within the ABSSSI definition. SUMMARY We argue that driveline infections meet the criteria for ABSSSI which would enlarge the 'on-label' antibiotic armamentarium for treating these severe infections, thereby improving the patients' quality of life.
Collapse
Affiliation(s)
- Christian Eckmann
- Academic Hospital of Goettingen University, Department of General, Visceral and Thoracic Surgery, Klinikum Hannoversch-Muenden, Hannoversch-Muenden
| | - Cord Sunderkötter
- Martin-Luther-University Halle-Wittenberg, University and University Hospital of Halle, Department of Dermatology and Venerology, Halle
| | - Karsten Becker
- University Medicine Greifswald, Friedrich Loeffler-Institute of Medical Microbiology, Greifswald
| | - Béatrice Grabein
- LMU Hospital, Clinical Microbiology and Hospital Hygiene, Munich
| | - Stefan Hagel
- Jena University Hospital-Friedrich Schiller University Jena, Institute for Infectious Diseases and Infection Control, Jena
| | - Frank Hanses
- University Hospital Regensburg, Department of Infection Prevention and Infectious Diseases
- University Hospital Regensburg, Emergency Department, Regensburg
| | - Dominic Wichmann
- University Medical Center Hamburg-Eppendorf, Department of Intensive Care Medicine, Hamburg
| | | |
Collapse
|
10
|
Azzam A, Shawky RM, El-Mahdy TS. Sub-inhibitory concentrations of ceftriaxone induce morphological alterations and PIA-independent biofilm formation in Staphylococcus aureus. Braz J Microbiol 2024; 55:297-308. [PMID: 37979131 PMCID: PMC10920565 DOI: 10.1007/s42770-023-01177-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 11/07/2023] [Indexed: 11/19/2023] Open
Abstract
The exposure of bacteria to sub-inhibitory concentrations of antibiotics is of biological significance since it can occur in vivo under many circumstances, including low-dose treatment, poor adherence to a regimen, poor drug penetration, drug-drug interactions, and antibiotic resistance of the pathogen. In this study, we investigated the effects of subinhibitory concentrations of four antibiotics: ampicillin, ceftriaxone, gentamicin, and norfloxacin, which are commonly used in clinical settings and on cell morphology and biofilm formation in Staphylococcus aureus as one of the leading causes of nosocomial and biofilm-associated infections. Nine clinical S. aureus biofilm-producing isolates and two known biofilm-producing reference strains, S. aureus ATCC 29213 and S. aureus ATCC 6538, were used in this study. Sub-MICs of beta-lactam antibiotics (ampicillin and ceftriaxone) significantly induced biofilm formation in S. aureus ATCC 29213 and S. aureus ATCC 6538 and in six clinical isolates out of the nine selected isolates when compared with the antibiotic-free control group (P < 0.05), with an approximately 2- to 2.5-fold increase. Gentamicin and norfloxacin induced biofilms in S. aureus ATCC 29213 and S. aureus ATCC 6538, while gentamicin and norfloxacin induced biofilms only in three and two of the nine tested isolates, respectively (P < 0.05). The chemical nature of the biofilm matrix produced by half the MIC of ceftriaxone in the six isolates that showed increased biofilm was all non-polysaccharide in composition (PIA-independent). Gene expression of biofilm-encoding genes atl and sarA in biofilms of the two tested strains (S. aureus ATCC 6538) and clinical strain (S. aureus 16) showed a significant upregulation after exposure to half MIC of ceftriaxone. Additionally, the bacterial cell morphological changes in planktonic cells caused by half MIC of ceftriaxone were evaluated by scanning electron microscopy, which demonstrated a significant cell enlargement when compared with the antibiotic-free control (P < 0.05), and some deformed cells were also noticed. In S. aureus clinical isolates, sub-MICs of ampicillin, ceftriaxone, gentamicin, and norfloxacin may stimulate substantial production of biofilm, which could have important clinical significance and make infection treatment challenges. Further, in vivo research is needed to fully comprehend how sub-MIC of antibiotics can affect biofilm formation in clinical settings. Additionally, more research is required to reveal the clinical implications of the morphological alterations in S. aureus brought on by exposure to ceftriaxone at concentrations below its MIC.
Collapse
Affiliation(s)
- Ahmed Azzam
- Department of Microbiology and Immunology, Faculty of Pharmacy, Helwan University, Ain Helwan, Cairo, Egypt
| | - Riham M Shawky
- Department of Microbiology and Immunology, Faculty of Pharmacy, Helwan University, Ain Helwan, Cairo, Egypt
| | - Taghrid S El-Mahdy
- Department of Microbiology and Immunology, Faculty of Pharmacy, Helwan University, Ain Helwan, Cairo, Egypt.
- Department of Microbiology and Immunology, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo, Egypt.
| |
Collapse
|
11
|
Constantinescu S, Niculescu AG, Hudiță A, Grumezescu V, Rădulescu D, Bîrcă AC, Dorcioman G, Gherasim O, Holban AM, Gălățeanu B, Vasile BȘ, Grumezescu AM, Bolocan A, Rădulescu R. Nanostructured Coatings Based on Graphene Oxide for the Management of Periprosthetic Infections. Int J Mol Sci 2024; 25:2389. [PMID: 38397066 PMCID: PMC10889398 DOI: 10.3390/ijms25042389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 02/12/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
To modulate the bioactivity and boost the therapeutic outcome of implantable metallic devices, biodegradable coatings based on polylactide (PLA) and graphene oxide nanosheets (nGOs) loaded with Zinforo™ (Zin) have been proposed in this study as innovative alternatives for the local management of biofilm-associated periprosthetic infections. Using a modified Hummers protocol, high-purity and ultra-thin nGOs have been obtained, as evidenced by X-ray diffraction (XRD) and transmission electron microscopy (TEM) investigations. The matrix-assisted pulsed laser evaporation (MAPLE) technique has been successfully employed to obtain the PLA-nGO-Zin coatings. The stoichiometric and uniform transfer was revealed by infrared microscopy (IRM) and scanning electron microscopy (SEM) studies. In vitro evaluation, performed on fresh blood samples, has shown the excellent hemocompatibility of PLA-nGO-Zin-coated samples (with a hemolytic index of 1.15%), together with their anti-inflammatory ability. Moreover, the PLA-nGO-Zin coatings significantly inhibited the development of mature bacterial biofilms, inducing important anti-biofilm efficiency in the as-coated samples. The herein-reported results evidence the promising potential of PLA-nGO-Zin coatings to be used for the biocompatible and antimicrobial surface modification of metallic implants.
Collapse
Affiliation(s)
- Sorin Constantinescu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Street, 050474 Bucharest, Romania; (S.C.); (D.R.); (A.B.); (R.R.)
| | - Adelina-Gabriela Niculescu
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 90-92 Panduri, 050663 Bucharest, Romania; (A.-G.N.); (A.H.); (A.M.H.)
- Department of Science and Engineering of Oxide Materials and Nanomaterials, University Politehnica of Bucharest, 1-7 Gh. Polizu Street, 060042 Bucharest, Romania; (A.C.B.); (B.Ș.V.)
| | - Ariana Hudiță
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 90-92 Panduri, 050663 Bucharest, Romania; (A.-G.N.); (A.H.); (A.M.H.)
- Department of Biochemistry and Molecular Biology, University of Bucharest, 91-95 Splaiul Independentei Street, 050095 Bucharest, Romania;
| | - Valentina Grumezescu
- Lasers Department, National Institute for Lasers, Plasma and Radiation Physics, 409 Atomistilor Street, 077125 Magurele, Romania; (V.G.); (G.D.); (O.G.)
| | - Dragoș Rădulescu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Street, 050474 Bucharest, Romania; (S.C.); (D.R.); (A.B.); (R.R.)
| | - Alexandra Cătălina Bîrcă
- Department of Science and Engineering of Oxide Materials and Nanomaterials, University Politehnica of Bucharest, 1-7 Gh. Polizu Street, 060042 Bucharest, Romania; (A.C.B.); (B.Ș.V.)
| | - Gabriela Dorcioman
- Lasers Department, National Institute for Lasers, Plasma and Radiation Physics, 409 Atomistilor Street, 077125 Magurele, Romania; (V.G.); (G.D.); (O.G.)
| | - Oana Gherasim
- Lasers Department, National Institute for Lasers, Plasma and Radiation Physics, 409 Atomistilor Street, 077125 Magurele, Romania; (V.G.); (G.D.); (O.G.)
| | - Alina Maria Holban
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 90-92 Panduri, 050663 Bucharest, Romania; (A.-G.N.); (A.H.); (A.M.H.)
- Microbiology and Immunology Department, Faculty of Biology, University of Bucharest, 1-3 Portocalelor Lane, 77206 Bucharest, Romania
| | - Bianca Gălățeanu
- Department of Biochemistry and Molecular Biology, University of Bucharest, 91-95 Splaiul Independentei Street, 050095 Bucharest, Romania;
| | - Bogdan Ștefan Vasile
- Department of Science and Engineering of Oxide Materials and Nanomaterials, University Politehnica of Bucharest, 1-7 Gh. Polizu Street, 060042 Bucharest, Romania; (A.C.B.); (B.Ș.V.)
| | - Alexandru Mihai Grumezescu
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 90-92 Panduri, 050663 Bucharest, Romania; (A.-G.N.); (A.H.); (A.M.H.)
- Department of Science and Engineering of Oxide Materials and Nanomaterials, University Politehnica of Bucharest, 1-7 Gh. Polizu Street, 060042 Bucharest, Romania; (A.C.B.); (B.Ș.V.)
| | - Alexandra Bolocan
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Street, 050474 Bucharest, Romania; (S.C.); (D.R.); (A.B.); (R.R.)
| | - Radu Rădulescu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Street, 050474 Bucharest, Romania; (S.C.); (D.R.); (A.B.); (R.R.)
| |
Collapse
|
12
|
Luo ZX, Li Y, Liu MF, Zhao R. Ciprofloxacin enhances the biofilm formation of Staphylococcus aureus via an agrC-dependent mechanism. Front Microbiol 2023; 14:1328947. [PMID: 38179460 PMCID: PMC10764545 DOI: 10.3389/fmicb.2023.1328947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 12/04/2023] [Indexed: 01/06/2024] Open
Abstract
Staphylococcus aureus readily forms biofilms on host tissues and medical devices, enabling its persistence in chronic infections and resistance to antibiotic therapy. The accessory gene regulator (Agr) quorum sensing system plays a key role in regulating S. aureus biofilm formation. This study reveals the widely used fluoroquinolone antibiotic, ciprofloxacin, strongly stimulates biofilm formation in methicillin-resistant S. aureus, methicillin-sensitive S. aureus, and clinical isolates with diverse genetic backgrounds. Crystal violet staining indicated that ciprofloxacin induced a remarkable 12.46- to 15.19-fold increase in biofilm biomass. Confocal laser scanning microscopy revealed that ciprofloxacin induced denser biofilms. Phenotypic assays suggest that ciprofloxacin may enhance polysaccharide intercellular adhesin production, inhibit autolysis, and reduce proteolysis during the biofilm development, thus promoting initial adhesion and enhancing biofilm stability. Mechanistically, ciprofloxacin significantly alters the expression of various biofilm-related genes (icaA, icaD, fnbA, fnbB, eap, emp) and regulators (agrA, saeR). Gene knockout experiments revealed that deletion of agrC, rather than saeRS, abolishes the ciprofloxacin-induced enhancement of biofilm formation, underscoring the key role of agrC. Thermal shift assays showed ciprofloxacin binds purified AgrC protein, thereby inhibiting the Agr system. Molecular docking results further support the potential interaction between ciprofloxacin and AgrC. In summary, subinhibitory concentrations of ciprofloxacin stimulate S. aureus biofilm formation via an agrC-dependent pathway. This inductive effect may facilitate local infection establishment and bacterial persistence, ultimately leading to therapeutic failure.
Collapse
Affiliation(s)
- Zhao-xia Luo
- Department of Clinical Laboratory, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, Nanchang, China
- School of Public Health, Nanchang University, Nanchang, China
| | - Yuting Li
- Department of Clinical Laboratory, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, Nanchang, China
- School of Public Health, Nanchang University, Nanchang, China
| | - Mei-fang Liu
- Department of Clinical Laboratory, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Rui Zhao
- Department of Clinical Laboratory, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
13
|
Wang B, Zhan Q, Xiao Y, Xu Y, Zhao H, Rao L, Wang X, Zhang J, Shen L, Zhou Y, Guo Y, Wu X, Yu J, Yu F. Mupirocin enhances the biofilm formation of Staphylococcus epidermidis in an atlE-dependent manner. Int J Antimicrob Agents 2023; 62:106904. [PMID: 37385560 DOI: 10.1016/j.ijantimicag.2023.106904] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 05/27/2023] [Accepted: 06/25/2023] [Indexed: 07/01/2023]
Abstract
The pathogenicity of Staphylococcus epidermidis is largely attributed to its exceptional ability to form biofilms. Here, we report that mupirocin, an antimicrobial agent widely used for staphylococcal decolonization and anti-infection, strongly stimulates the biofilm formation of S. epidermidis. Although the polysaccharide intercellular adhesin (PIA) production was unaffected, mupirocin significantly facilitated extracellular DNA (eDNA) release by accelerating autolysis, thereby positively triggering cell surface attachment and intercellular agglomeration during biofilm development. Mechanistically, mupirocin regulated the expression of genes encoding for the autolysin AtlE as well as the programmed cell death system CidA-LrgAB. Critically, through gene knockout, we found out that deletion of atlE, but not cidA or lrgA, abolished the enhancement of biofilm formation and eDNA release in response to mupirocin treatment, indicating that atlE is required for this effect. In Triton X-100 induced autolysis assay, mupirocin treated atlE mutant displayed a slower autolysis rate compared with the wild-type strain and complementary strain. Therefore, we concluded that subinhibitory concentrations of mupirocin enhance the biofilm formation of S. epidermidis in an atlE dependent manner. This induction effect could conceivably be responsible for some of the more unfavourable outcomes of infectious diseases.
Collapse
Affiliation(s)
- Bingjie Wang
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qing Zhan
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Infection Control Department, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanghua Xiao
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yanlei Xu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Huilin Zhao
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lulin Rao
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xinyi Wang
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiao Zhang
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Li Shen
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ying Zhou
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yinjuan Guo
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiaocui Wu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jingyi Yu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Fangyou Yu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
14
|
Morais MLGDS, Santos MGC, Costa CL, Martins CS, Leitão RFDC, de Melo Pacífico D, Quesada-Gómez C, Castelo Branco D, Ferreira EDO, Brito GADC. Comparative biofilm-forming ability between Clostridioides difficile strains isolated in Latin America and the epidemic NAP1/027 strain. Front Cell Infect Microbiol 2022; 12:1033698. [PMID: 36619751 PMCID: PMC9815708 DOI: 10.3389/fcimb.2022.1033698] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 11/07/2022] [Indexed: 12/03/2022] Open
Abstract
Introduction One of the challenges in treating Clostridioides difficile infection (CDI) is that the bacterium forms biofilms, a critical virulence mechanism known to promote antibiotic resistance and, as a result, consequently, a higher recurrence of the disease. The goal of this study was to compare the ability of three MLST Clade 2 strains to form a biofilm in vitro: ICC-45 (ribotype SLO231/UK[CE]821), a ST41 toxinotype IXb isolated in Brazil; and two epidemic NAP1/027/ST01 strains: NAP1/027/ST01 (LIBA5756), isolated during a 2010 outbreak in Costa Rica and the reference epidemic strain NAP1/027/ST01 (R20291); and ATCC700057, a non-toxigenic strain. Methods The ability of strains to form biofilm was evaluated using crystal violet staining. In addition, samples were stained with the Film Tracer biofilm matrix (Invitrogen®) and the biofilm matrix thickness was measured using confocal microscopy. The matrix architecture was determined using Scanning electron microscop. Confocal microscopy was used to detect the presence of toxin A (tcdA) using an anti-Clostridioides difficile TcdA antibody. The expression of virulence genes (tcdA, tcdB, tcdC, cdtB, spo0A, slpA, cwp66 and cwp84) was examined, as well as the effect of antibiotics metronidazole (MTZ) and vancomycin (VAN) on biofilm growth. Results All of the strains tested formed a moderate biofilm with 1.1 <DO570nm>3.5. After 72h, biofilm biomass of the NAP1/027/ST01 epidemic strains (LIBA5756 and R20291) was significantly higher than ICC-45 and ATCC 700057 biofilms, as confirmed by electron and confocal microscopy. At 120h, the LIBA5756 biofilm biomass decreased compared to other strains. The toxigenic strains R20291 or LIBA 5756 had higher expression of genes tcdA, tcdB, tcdC, cdtA, slpA and spo0A than ICC-45, but there were no significant differences in the expression levels of cdtB, cwp66 and cwp84. In epidemic strains, VAN and MTZ inhibited biofilm formation; however, in the ICC-45 strain, MIC concentrations of VAN and MIC and 4MIC of MTZ did not inhibit biofilm formation. Conclusion The three MLST Clade 2 isolated from different rybotipes, two of which were isolated from Latin America, are competent biofilm-forming bacteria, indicating their ability to induce C. difficile infection recurrence, making treatment difficult.
Collapse
Affiliation(s)
- Maria Luana Gaudencio dos Santos Morais
- Department of Morphology, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil,Laboratory of Bacteriology, Department of Pathology, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Mayara Gilde Castro Santos
- Laboratório de Biologia de Anaeróbios, Instituto de Microbiologia Paulo de Góes Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cecília Leite Costa
- Department of Morphology, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil,Laboratory of Bacteriology, Department of Pathology, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Conceição Silva Martins
- Department of Morphology, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | | | - Dvison de Melo Pacífico
- Department of Morphology, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Carlos Quesada-Gómez
- Facultad de Microbiología and Centro de Investigación en Enfermedades Tropicales, Universidad de Costa Rica, San José, Costa Rica
| | - Débora Castelo Branco
- Laboratory of Bacteriology, Department of Pathology, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Eliane de Oliveira Ferreira
- Laboratory of Bacteriology, Department of Pathology, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Gerly Anne de Castro Brito
- Department of Morphology, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil,*Correspondence: Gerly Anne de Castro Brito,
| |
Collapse
|
15
|
Role of RNAIII in Resistance to Antibiotics and Antimicrobial Agents in Staphylococcus epidermidis Biofilms. Int J Mol Sci 2022; 23:ijms231911094. [PMID: 36232391 PMCID: PMC9569910 DOI: 10.3390/ijms231911094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/29/2022] [Accepted: 09/16/2022] [Indexed: 11/17/2022] Open
Abstract
Staphylococcus epidermidis is a known opportunistic pathogen and is one of the leading causes of chronic biofilm-associated infections. Biofilm formation is considered as a main strategy to resist antibiotic treatment and help bacteria escape from the human immune system. Understanding the complex mechanisms in biofilm formation can help find new ways to treat resistant strains and lower the prevalence of nosocomial infections. In order to examine the role of RNAIII regulated by the agr quorum sensing system and to what extent it influences biofilm resistance to antimicrobial agents, deletion mutant S. epidermidis RP62a-ΔRNAIII deficient in repressor domains with a re-maining functional hld gene was created. A deletion strain was used to examine the influence of oxacillin in combination with vanillin on biofilm resistance and cell survival was determined. Utilizing real-time qPCR, confocal laser scanning microscopy (CLSM), and crystal violet staining analyses, we found that the RNAIII-independent controlled phenol soluble modulins (PSMs) and RNAIII effector molecule have a significant role in biofilm resistance to antibiotics and phenolic compounds, and it protects the integrity of biofilms. Moreover, a combination of antibiotic and antimicrobial agents can induce methicillin-resistant S. epidermidis biofilm formation and can lead to exceedingly difficult medical treatment.
Collapse
|
16
|
Raj DS, Dhamodharan D, Thanigaivel S, Vickram AS, Byun HS. Nanoemulsion as an Effective Inhibitor of Biofilm-forming Bacterial Associated Drug Resistance: An Insight into COVID Based Nosocomial Infections. BIOTECHNOL BIOPROC E 2022; 27:543-555. [PMID: 36092682 PMCID: PMC9449957 DOI: 10.1007/s12257-022-0055-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/30/2022] [Accepted: 04/06/2022] [Indexed: 11/26/2022]
Abstract
Antibiotic overuse has resulted in the microevolution of drug-tolerant bacteria. Understandably it has become one of the most significant obstacles of the current century for scientists and researchers to overcome. Bacteria have a tendency to form biofilm as a survival mechanism. Biofilm producing microorganism become far more resistant to antimicrobial agents and their tolerance to drugs also increases. Prevention of biofilm development and curbing the virulency factors of these multi drug resistant or tolerant bacterial pathogens is a newly recognised tactic for overcoming the challenges associated with such bacterial infections and has become a niche to be addressed. In order to inhibit virulence and biofilm from planktonic bacteria such as, Pseudomonas aeruginosa, Acinetobacter baumannii, and others, stable nanoemulsions (NEs) of essential oils (EOs) and their bioactive compounds prove to be an interesting solution. These NEs demonstrated significantly greater anti-biofilm and anti-virulence activity than commercial antibiotics. The EO reduces disease-causing gene expression, which is required for pathogenicity, biofilm formation and attachment to the surfaces. Essential NE and NE-loaded hydrogel surface coatings demonstrates superior antibiofilm activity which can be employed in healthcare-related equipments like glass, plastic, and metal chairs, hospital beds, ventilators, catheters, and tools used in intensive care units. Thus, anti-virulence and anti-biofilm forming strategies based on NEs-loaded hydrogel may be used as coatings to combat biofilm-mediated infection on solid surfaces.
Collapse
Affiliation(s)
- Deena Santhana Raj
- Department of Biotechnology, Saveetha School of Engineering, SIMATS, Saveetha University, Thandalam, Chennai, Tamil Nadu 602105 India
| | - Duraisami Dhamodharan
- Department of Chemical and Biomolecular Engineering, Chonnam National University, Yeosu, 59626 Korea
| | - S. Thanigaivel
- Department of Biotechnology, Saveetha School of Engineering, SIMATS, Saveetha University, Thandalam, Chennai, Tamil Nadu 602105 India
| | - A. S. Vickram
- Department of Biotechnology, Saveetha School of Engineering, SIMATS, Saveetha University, Thandalam, Chennai, Tamil Nadu 602105 India
| | - Hun-Soo Byun
- Department of Chemical and Biomolecular Engineering, Chonnam National University, Yeosu, 59626 Korea
| |
Collapse
|
17
|
Daptomycin exerts differential immunomodulatory effects on host responses against methicillin-resistant Staphylococcus aureus biofilms. Int J Antimicrob Agents 2022; 60:106666. [PMID: 36038095 DOI: 10.1016/j.ijantimicag.2022.106666] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 08/10/2022] [Accepted: 08/22/2022] [Indexed: 11/23/2022]
Abstract
BACKGROUND Daptomycin (DAP) is indicated for difficult-to-treat Gram-positive infections, especially those caused by methicillin-resistant S. aureus (MRSA). Exposure of S. aureus to sub-inhibitory concentrations (sub-MICs) of antibiotics have been shown to alter cell morphology or biofilm formation. OBJECTIVES To investigate the influence of DAP biofilm sub-MICs on the damage caused by human polymorphonuclear neutrophils (PMNs) against MRSA biofilms and the potential immunomodulatory activity of DAP on human monocytes (MNCs) exposed to MRSA biofilms. METHODS DAP activity against biofilms and the impact of DAP on the PMNs-induced biofilm damage were evaluated by the XTT reduction assay, whereas pathogen recognition, signal transduction and cytokine modulation of DAP on MNCs in response to MRSA biofilms were assessed by RT-PCR and ELISA methodology. RESULTS The MIC50 of DAP to MRSA biofilms was 16 to 32 mg/L. Pre-treatment of MRSA to 1, 2 or 4 mg/L DAP caused a synergistic effect on PMN-mediated biofilm damage, being dependent on the effector-to-target ratio. MNCs responded to MRSA biofilms and DAP through Toll like receptor 2 (TLR2) upregulation and increased NLRP3 inflammasome production. DAP caused 2.5-fold greater TLR2 mRNA levels than those caused by MRSA biofilms. A predominantly inflammatory response was induced by either component, causing the release of significantly increased IFN-γ, TNF-α, IL-8 and IL-6 levels by MNCs exposed to the combination treatment. MRSA biofilms alone or combined with DAP caused low amounts of IL-10 production, but increased IL-1β levels. CONCLUSIONS DAP may condition MNCs towards an inflammatory response through TLR2 engagement and NLRP3 inflammasome activation, possibly controlling biofilm-associated pathogenicity.
Collapse
|
18
|
Zha L, Xie Y, Wu C, Lei M, Lu X, Tang W, Zhang J. Novel benzothiazole‒urea hybrids: Design, synthesis and biological activity as potent anti-bacterial agents against MRSA. Eur J Med Chem 2022; 236:114333. [PMID: 35397402 DOI: 10.1016/j.ejmech.2022.114333] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/19/2022] [Accepted: 03/28/2022] [Indexed: 11/26/2022]
Abstract
Novel benzothiazole‒urea hybrids were designed, synthesized and evaluated their anti-bacterial activity. They only exhibited anti-bacterial activity against Gram-positive bacteria, including clinical methicillin-resistant S. aureus (MRSA), compounds 5f, 5i, 8e, 8k and 8l exhibited potent activity (MIC = 0.39 and 0.39/0.78 μM against SA and MRSA, respectively). Crystal violet assay showed that compounds 5f, 8e and 8l not only inhibited the formation of biofilms but also eradicated preformed biofilms. Compound 8l had membrane disruption, little propensity to induce resistance, benign safety and in vivo anti-MRSA efficacy in a mouse model of abdominal infection. Therefore, our data demonstrated the potential to advance benzothiazole‒urea hybrids as a new class of antibiotics.
Collapse
Affiliation(s)
- Liang Zha
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Yunfeng Xie
- School of Medicine, Anhui University of Science and Technology, Huainan, 232001, China
| | - Chengyao Wu
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Ming Lei
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Xueer Lu
- School of Medicine, Anhui University of Science and Technology, Huainan, 232001, China
| | - Wenjian Tang
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China.
| | - Jing Zhang
- Anhui Prevention and Treatment Center for Occupational Disease, Anhui No. 2 Provincial People's Hospital, Hefei, 230022, China.
| |
Collapse
|
19
|
Hagras SAA, Hosny AEDMS, Helmy OM, Salem-Bekhit MM, Shakeel F, Farrag HA. Effect of sub-inhibitory concentrations of cefepime on biofilm formation by Pseudomonas aeruginosa. Can J Microbiol 2021; 67:894-901. [PMID: 34731576 DOI: 10.1139/cjm-2021-0229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This study investigated the effect of cefepime at sub-minimum inhibitory concentrations (sub-MICs) on in vitro biofilm formation (BF) by clinical isolates of Pseudomonas aeruginosa. The effect of cefepime at sub-MIC levels (½-1/256 MIC) on in vitro BF by six clinical isolates of P. aeruginosa was phenotypically assessed following 24 and 48 h of challenge using the tissue culture plate (TCP) assay. Quantitative real-time polymerase chain reaction (qRT-PCR) was employed to observe the change in expression of three biofilm-related genes, namely, a protease-encoding gene (lasA), fimbrial protein-encoding gene (cupA1), and alginate-encoding gene (algC), in a weak biofilm-producing strain of P. aeruginosa following 24 and 48 h of challenge with sub-MICs of cefepime. The BF morphology in response to cefepime was imaged using scanning electron microscopy (SEM). The TCP assay showed strain-, time-, and concentration-dependent changes in in vitro BF in P. aeruginosa following challenge with sub-MICs of cefepime, with a profound increase in strains with inherently no or weak biofilm-producing ability. RT-PCR revealed time-dependent upregulation in the expression of the investigated genes following challenge with ½ and ¼ MIC levels, as confirmed by SEM. Cefepime at sub-MICs could upregulate the expression of BF-related genes and enhance BF by P. aeruginosa clinical isolates.
Collapse
Affiliation(s)
- Soheir A A Hagras
- Department of Drug Radiation Research, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt
- Inaya Medical Colleges, Riyadh, Saudi Arabia
| | - Alaa El-Dien M S Hosny
- Department of Microbiology & Immunology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Omneya M Helmy
- Department of Microbiology & Immunology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Mounir M Salem-Bekhit
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Department of Microbiology & Immunology, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Faiyaz Shakeel
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Hala A Farrag
- Department of Drug Radiation Research, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt
| |
Collapse
|
20
|
Tao J, Yan S, Zhou C, Liu Q, Zhu H, Wen Z. Total flavonoids from Potentilla kleiniana Wight et Arn inhibits biofilm formation and virulence factors production in methicillin-resistant Staphylococcus aureus (MRSA). JOURNAL OF ETHNOPHARMACOLOGY 2021; 279:114383. [PMID: 34214645 DOI: 10.1016/j.jep.2021.114383] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 06/22/2021] [Accepted: 06/28/2021] [Indexed: 06/13/2023]
Abstract
ETHANOPHARMACOLOGICAL RELEVANCE Potentilla kleiniana Wight et Arn is a wide-spread wild plant in the mountainous areas in southern China. The whole herb has been used as a traditional herbal medicine to treat fever, arthritis, malaria, insect and snake bites, hepatitis, and traumatic injury. In vitro studies have reported the antibacterial activity use of the plant in traditional medicinal systems. AIM OF THE STUDY The aim of this study was to investigate the inhibitory activity of total flavonoid from Potentilla kleiniana Wight et Arn (TFP) on methicillin-resistant Staphylococcus aureus (MRSA) in planktonic state and biofilm state. MATERIALS AND METHODS Antibacterial activities of TFP on planktonic MRSA were determined by agar diffusion method, microtiter plate assay and time-kill curve assay. Electrical conductivity, membrane permeability, membrane potential and autoaggregation were analyzed to study TFP effects on planktonic MRSA growth. Crystal violet (CV) staining and confocal laser scanning microscopy (CLSM) were analyzed to study TFP effects on aggregation and maturation of MRSA biofilm. After TFP treatment, extracellular polymeric substances (EPS) production were examined. Morphological changes in planktonic and MRSA biofilm following TFP treatment were determined with scanning electron microscopy (SEM) and transmission electron microscopy (TEM). Moreover, α-Toxin protein expression and adhesion-related gene expression were also determined. RESULTS The minimum inhibitory concentration (MIC) of TFP against MRSA was 20 μg/mL. The agar diffusion method and time-kill curve assay results indicated that TFP inhibited planktonic MRSA growth. TFP treatment significantly inhibited planktonic MRSA growth by inhibiting autoaggregation, α-hemolysin activity, α-Toxin protein expression, but increasing electrolyte leakage, membrane permeability and membrane potential and impacting cell structure. Moreover, TFP treatment significantly inhibited aggregation and maturation on MRSA biofilm by decreasing surface hydrophobicity, EPS production and adhesion-related gene expression. CONCLUSION The results of this trial provide scientific experimental data on the traditional use of Potentilla Kleiniana Wight et Arn for traumatic injury treatment and further demonstrate the potential of TFP to be developed as a novel anti-biofilm drug.
Collapse
Affiliation(s)
- Junyu Tao
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, 518000, China; Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, 518000, China; School of Food Engineering and Biotechnology, Hanshan Normal University, Chaozhou, 521041, China
| | - Shilun Yan
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, 518000, China; Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, 518000, China; School of Food Engineering and Biotechnology, Hanshan Normal University, Chaozhou, 521041, China
| | - Chuyue Zhou
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, 518000, China
| | - Qiong Liu
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518000, China
| | - Hui Zhu
- School of Food Engineering and Biotechnology, Hanshan Normal University, Chaozhou, 521041, China
| | - Zhen Wen
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, 518000, China; School of Food Engineering and Biotechnology, Hanshan Normal University, Chaozhou, 521041, China.
| |
Collapse
|
21
|
Oliva A, Stefani S, Venditti M, Di Domenico EG. Biofilm-Related Infections in Gram-Positive Bacteria and the Potential Role of the Long-Acting Agent Dalbavancin. Front Microbiol 2021; 12:749685. [PMID: 34745053 PMCID: PMC8569946 DOI: 10.3389/fmicb.2021.749685] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 10/04/2021] [Indexed: 01/08/2023] Open
Abstract
Infections caused by Gram-positive bacteria are a major public health problem due to their increasing resistance to antibiotics. Staphylococcus and Enterococcus species' resistance and pathogenicity are enhanced by their ability to form biofilm. The biofilm lifestyle represents a significant obstacle to treatment because bacterial cells become highly tolerant to a wide range of antimicrobial compounds normally effective against their planktonic forms. Thus, novel therapeutic strategies targeting biofilms are urgently needed. The lipoglycopeptide dalbavancin is a long-acting agent for treating acute bacterial skin and skin structure infections caused by a broad range of Gram-positive pathogens. Recent studies have shown promising activity of dalbavancin against Gram-positive biofilms, including methicillin-resistant S. aureus (MRSA), methicillin-resistant S. epidermidis (MRSE), and vancomycin-susceptible enterococci. This review outlines the mechanisms regulating biofilm development in Staphylococcus and Enterococcus species and the clinical impact of biofilm-related infections. In addition, it discusses the clinical implications and potential therapeutic perspectives of the long-acting drug dalbavancin against biofilm-forming Gram-positive pathogens.
Collapse
Affiliation(s)
- Alessandra Oliva
- Department of Public Health and Infectious Diseases, “La Sapienza” University of Rome, Rome, Italy
| | - Stefania Stefani
- Laboratory of Molecular Medical Microbiology and Antimicrobial Resistance Research (Mmarl), Department of Biomedical and Biotechnological Sciences (Biometec), University of Catania, Catania, Italy
| | - Mario Venditti
- Department of Public Health and Infectious Diseases, “La Sapienza” University of Rome, Rome, Italy
| | | |
Collapse
|
22
|
Potentiating effects of leaderless enterocin DD14 in combination with methicillin on clinical methicillin-resistant Staphylococcus aureus S1 strain. Microbiol Res 2021; 252:126864. [PMID: 34521050 DOI: 10.1016/j.micres.2021.126864] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/27/2021] [Accepted: 09/06/2021] [Indexed: 11/22/2022]
Abstract
Biofilm formation by pathogenic bacteria as well as their resilience to antibiotic treatments are a major health problem. Here, we sequenced and analyzed the genome of the clinical methicillin-resistant Staphylococcus aureus S1 (MRSA-S1) strain and established its sensitivity to the combination of methicillin and the leaderless two peptides enterocin DD14 (EntDD14). Such sensitivity was assessed in vitro based on the MIC/FIC values as well as on killing curves experiments. Moreover, combination of EntDD14 and methicillin was able to reduce the biofilm formation of Staphylococcus aureus S1 of about ∼30 %. Interestingly, genes thought to be involved in the virulence of MRSA-S1, like nuc and pvl which code, respectively, for nuclease and Panton-Valentine leucocidin, were shown to be downregulated following treatment with EntDD14 and methicillin. Similar effects were registered for other genes such as cflA, cflB and icaB, coding for bacterial ligands clumping factors A, B and intercellular adhesion factor respectively. All these data, suggest that combinations of bacteriocins and antibiotics are useful as a backup for treatment of bacterial infections.
Collapse
|
23
|
Virulence alterations in staphylococcus aureus upon treatment with the sub-inhibitory concentrations of antibiotics. J Adv Res 2021; 31:165-175. [PMID: 34194840 PMCID: PMC8240104 DOI: 10.1016/j.jare.2021.01.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 01/09/2021] [Accepted: 01/12/2021] [Indexed: 12/24/2022] Open
Abstract
Background The treatment of patients with Staphylococcus aureus infections mainly relies on antistaphylococcal regimens that are established with effective antibiotics. In antibiotic therapy or while living in nature, pathogens often face the sub-inhibitory concentrations (sub-MICs) of antibiotics due to drug pharmacokinetics, diffusion barriers, waste emission, resistant organism formation, and farming application. Different categories of antibiotics at sub-MICs have diverse effects on the physiological and chemical properties of microorganisms. These effects can result in virulence alterations. However, the mechanisms underlying the actions of antibiotics at sub-MICs on S. aureus virulence are obscure. Aim of review In this review, we focus on the effects of sub-MICs of antibiotics on S. aureus virulence from the aspects of cell morphological change, virulence factor expression, bacterial adherence and invasion, staphylococcal biofilm formation, and small-colony variant (SCV) production. The possible mechanisms of antibiotic-induced S. aureus virulence alterations are also addressed. Key scientific concepts of review Five main aspects of bacterial virulence can be changed in S. aureus exposure to the sub-MIC levels of antibiotics, resulting in deformed bacterial cells to stimulate abnormal host immune responses, abnormally expressed virulence factors to alter disease development, changed bacterial adhesion and invasion abilities to affect colonization and diffusion, altered biofilm formation to potentate material-related infections, and increased SCV formation to achieve persistent infection and recurrence. These advanced findings expand our knowledge to rethink the molecular signaling roles of antibiotics beyond their actions as antimicrobial agents.
Collapse
|
24
|
Cataneli Pereira V, Pinheiro-Hubinger L, de Oliveira A, Moraes Riboli DF, Benini Martins K, Calixto Romero L, Ribeiro de Souza da Cunha MDL. Detection of the agr System and Resistance to Antimicrobials in Biofilm-Producing S. epidermidis. Molecules 2020; 25:molecules25235715. [PMID: 33287389 PMCID: PMC7729762 DOI: 10.3390/molecules25235715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/18/2020] [Accepted: 11/24/2020] [Indexed: 11/19/2022] Open
Abstract
The ability of Staphylococcus epidermidis to produce virulence factors, such as biofilm, added to its increased resistance to antimicrobials can cause infections that are difficult to treat. Many staphylococcal virulence factors are under the control of the accessory gene regulator (agr). The objective of this study was to establish the agr locus and susceptibility of biofilm-producing S. epidermidis specimens to antimicrobial agents, through PCR reactions, reverse transcription polymerase chain reaction (RT-PCR), and the determination of minimum inhibitory concentration (MIC), and to analyze the clonal profile of 300 strains isolated from blood culture specimens from inpatients at a University Hospital in Brazil, over a 20-year period by pulsed-field gel electrophoresis (PFGE) and multilocus sequence typing (MLST) techniques. The ica operon expression was shown in 83.6% strains, bhp gene in 11.5%, and aap gene in 32.8%. Oxacillin resistance was detected in 90.1%, while 4.9% showed tigecycline resistance, and intermediate resistance to quinupristin/dalfopristin was identified in 0.4%. Clonal profile determination showed 11 clusters, with the ST2 type determined as the major cluster. The S. epidermidis biofilm producer demonstrated a predominance of agr I locus, oxacillin resistance, and SCCmec III as well as the potential dissemination of pathogenic clones in hospital settings over long periods.
Collapse
Affiliation(s)
- Valéria Cataneli Pereira
- Sector of Microbiology and Immunology, Department of Chemical and Biological Sciences, Institute of Biosciences, UNESP—University Estadual Paulista, Botucatu CEP 18618-689, São Paulo, Brazil; (V.C.P.); (L.P.-H.); (A.d.O.); (D.F.M.R.); (K.B.M.); (L.C.R.)
- Sector of Microbiology and Immunology, UNOESTE—University of West Paulista, Presidente Prudente CEP 19050-920, São Paulo, Brazil
| | - Luiza Pinheiro-Hubinger
- Sector of Microbiology and Immunology, Department of Chemical and Biological Sciences, Institute of Biosciences, UNESP—University Estadual Paulista, Botucatu CEP 18618-689, São Paulo, Brazil; (V.C.P.); (L.P.-H.); (A.d.O.); (D.F.M.R.); (K.B.M.); (L.C.R.)
| | - Adilson de Oliveira
- Sector of Microbiology and Immunology, Department of Chemical and Biological Sciences, Institute of Biosciences, UNESP—University Estadual Paulista, Botucatu CEP 18618-689, São Paulo, Brazil; (V.C.P.); (L.P.-H.); (A.d.O.); (D.F.M.R.); (K.B.M.); (L.C.R.)
| | - Danilo Flávio Moraes Riboli
- Sector of Microbiology and Immunology, Department of Chemical and Biological Sciences, Institute of Biosciences, UNESP—University Estadual Paulista, Botucatu CEP 18618-689, São Paulo, Brazil; (V.C.P.); (L.P.-H.); (A.d.O.); (D.F.M.R.); (K.B.M.); (L.C.R.)
| | - Katheryne Benini Martins
- Sector of Microbiology and Immunology, Department of Chemical and Biological Sciences, Institute of Biosciences, UNESP—University Estadual Paulista, Botucatu CEP 18618-689, São Paulo, Brazil; (V.C.P.); (L.P.-H.); (A.d.O.); (D.F.M.R.); (K.B.M.); (L.C.R.)
| | - Letícia Calixto Romero
- Sector of Microbiology and Immunology, Department of Chemical and Biological Sciences, Institute of Biosciences, UNESP—University Estadual Paulista, Botucatu CEP 18618-689, São Paulo, Brazil; (V.C.P.); (L.P.-H.); (A.d.O.); (D.F.M.R.); (K.B.M.); (L.C.R.)
| | - Maria de Lourdes Ribeiro de Souza da Cunha
- Sector of Microbiology and Immunology, Department of Chemical and Biological Sciences, Institute of Biosciences, UNESP—University Estadual Paulista, Botucatu CEP 18618-689, São Paulo, Brazil; (V.C.P.); (L.P.-H.); (A.d.O.); (D.F.M.R.); (K.B.M.); (L.C.R.)
- Correspondence: ; Tel.: +55-14-3880-0428
| |
Collapse
|
25
|
Algorri M, Wong-Beringer A. Differential effects of antibiotics on neutrophils exposed to lipoteichoic acid derived from Staphylococcus aureus. Ann Clin Microbiol Antimicrob 2020; 19:50. [PMID: 33143710 PMCID: PMC7641855 DOI: 10.1186/s12941-020-00392-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 10/20/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Persistent bacteremia occurs in at least 30% of patients with Staphylococcus aureus bloodstream infection (SAB) and may be attributable to a dysregulated host immune response. Neutrophils interact with a variety of S. aureus microbial factors, including lipoteichoic acid (LTA), to activate phagocytic function in a concentration-dependent manner. Antibiotics have been shown to exert both direct antimicrobial action as well as immunomodulatory effects. In this study, we compared the effects of different anti-staphylococcal antibiotics on LTA-mediated immune activation of neutrophils. METHODS Neutrophils obtained from healthy volunteers were exposed to two levels of LTA (1 and 10 μg/ml) with or without addition of antibiotics from different pharmacologic classes (vancomycin, daptomycin, ceftaroline). Neutrophil function was assessed by examining phagocytic response, activation (CD11b, CD62L expression), Toll-like receptor-2 expression, cell survival and apoptosis, and CXCL8 release. RESULTS Differential LTA-mediated antibiotic effects on neutrophil function were observed primarily at the high LTA exposure level. Ceftaroline in the presence of 10 μg/ml LTA had the most prominent effects on phagocytosis and CD11b and CD62L expression, with trends towards increased neutrophil survival and preservation of CXCL8 release when compared to daptomycin and vancomycin with the latter significantly dampening PMN CXCL8 release. CONCLUSIONS Select antimicrobial agents, such as ceftaroline, exert immunostimulatory effects on neutrophils exposed to S. aureus LTA, which when confirmed in vivo, could be leveraged for its dual immunomodulatory and antibacterial actions for the treatment of persistent SAB mediated by a dysregulated host response.
Collapse
Affiliation(s)
- Marquerita Algorri
- University of Southern California, School of Pharmacy, 1985 Zonal Avenue, Los Angeles, CA, 90089, USA
| | - Annie Wong-Beringer
- University of Southern California, School of Pharmacy, 1985 Zonal Avenue, Los Angeles, CA, 90089, USA.
| |
Collapse
|
26
|
Detection of Heavy Metal Tolerance among different MLSB Resistance Phenotypes of Methicillin-Resistant S. aureus (MRSA). JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2020. [DOI: 10.22207/jpam.14.3.29] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) strains are widespread globally. Besides their virulence factors, the co-occurrence of antimicrobial and metal resistance has been reported. This study was designed to evaluate the antibiotic resistance and resistance phenotypes, investigate the occurrence of virulence factors, and detect heavy metal tolerance among MRSA strains. Antibiogram profiling was done as recommended by CLSI instructions. Resistance phenotypes were detected by D test, followed by characterization of enzymatic activities and biofilm formation assay. Antibacterial activity of different heavy metals was tested, and predictable synergistic assay was performed. Among MRSA strains collected, high resistance to ampicillin and amoxicillin/clavulanate (100%) and high susceptibility to clindamycin (70%) were obtained. Resistance phenotypes were detected as S, constitutive MLSB, inducible MLSB, and MS by percentages of 10%, 30%, 30% and 30% respectively. Virulence factors like lipolytic (50%) and hemolytic (70%) activity, and biofilm formation ability (100%) were detected. High resistance towards potassium and magnesium was observed. MTC of 500 ppm was detected for all isolates in case of cobalt and iron. In case of zinc and copper, MTC was detected as 500 ppm except for one isolate which was highly resistant, and 500 ppm for all isolates except for two isolates which were highly sensitive respectively. Magnesium in different concentrations (500 and 2000 ppm) showed synergistic activity with erythromycin and clindamycin. Results reveal high heavy metal tolerance among antibiotic resistant MRSA strains, in addition to the presence of virulence factors. Upcoming studies must be focused on the combination of sub-inhibitory concentration of different heavy metals with the available antibiotics.
Collapse
|
27
|
Muñoz-Gallego I, Viedma E, Esteban J, Mancheño-Losa M, García-Cañete J, Blanco-García A, Rico A, García-Perea A, Ruiz Garbajosa P, Escudero-Sánchez R, Sánchez Somolinos M, Marín Arriaza M, Romanyk J, Barbero JM, Arribi Vilela A, González Romo F, Pérez-Jorge C, M. Arana D, Monereo A, Domingo D, Cordero J, Sánchez Romero MI, García Viejo MÁ, Lora-Tamayo J, Chaves F. Genotypic and Phenotypic Characteristics of Staphylococcus aureus Prosthetic Joint Infections: Insight on the Pathogenesis and Prognosis of a Multicenter Prospective Cohort. Open Forum Infect Dis 2020; 7:ofaa344. [PMID: 33005695 PMCID: PMC7519778 DOI: 10.1093/ofid/ofaa344] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 08/08/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Staphylococcus aureus is the leading cause of prosthetic joint infection (PJI). Beyond the antibiogram, little attention has been paid to the influence of deep microbiological characteristics on patient prognosis. Our aim was to investigate whether microbiological genotypic and phenotypic features have a significant influence on infection pathogenesis and patient outcome. METHODS A prospective multicenter study was performed, including all S. aureus PJIs (2016-2017). Clinical data and phenotypic (agr functionality, β-hemolysis, biofilm formation) and genotypic characteristics of the strains were collected. Biofilm susceptibility to antimicrobials was investigated (minimal biofilm eradication concentration [MBEC] assay). RESULTS Eighty-eight patients (39.8% men, age 74.7 ± 14.1 years) were included. Forty-five had early postoperative infections (EPIs), 21 had chronic infections (CPIs), and 19 had hematogenous infections (HIs). Twenty (22.7%) were caused by methicillin-resistant S. aureus. High genotypic diversity was observed, including 16 clonal complexes (CCs), with CC5 being the most frequent (30.7%). agr activity was greater in EPI than CPI (55.6% vs 28.6%; P = .041). Strains causing EPI were phenotypically and genotypically similar, regardless of symptom duration. Treatment failure (36.5%) occurred less frequently among cases treated with implant removal. In cases treated with debridement and implant retention, there were fewer failures among those who received combination therapy with rifampin. No genotypic or phenotypic characteristics predicted failure, except vancomycin minimal inhibitory concentration ≥1.5 mg/L (23.1% failure vs 3.4%; P = .044). MBEC50 was >128 mg/L for all antibiotics tested and showed no association with prognosis. CONCLUSIONS S. aureus with different genotypic backgrounds is capable of causing PJI, showing slight differences in clinical presentation and pathogenesis. No major microbiological characteristics were observed to influence the outcome, including MBEC.
Collapse
Affiliation(s)
- Irene Muñoz-Gallego
- Servicio de Microbiología, Hospital Universitario 12 de Octubre, Universidad Complutense, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Esther Viedma
- Servicio de Microbiología, Hospital Universitario 12 de Octubre, Universidad Complutense, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- Red Española de Investigación en Patología Infecciosa (REIPI), Instituto de Salud Carlos III, Ministerio de Ciencia e Innovación, Sevilla, Spain
| | - Jaime Esteban
- Servicio de Microbiología, IIS-Fundación Jiménez Díaz, Madrid, Spain
| | - Mikel Mancheño-Losa
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- Servicio de Medicina Interna, Hospital Universitario 12 de Octubre, Universidad Complutense, Madrid, Spain
| | - Joaquín García-Cañete
- Servicio de Medicina Interna-Urgencias, Hospital Universitario Fundación Jiménez Díaz, Madrid, Spain
| | - Antonio Blanco-García
- Servicio de Medicina Interna-Urgencias, Hospital Universitario Fundación Jiménez Díaz, Madrid, Spain
| | - Alicia Rico
- Servicio de Medicina Interna, Hospital Universitario La Paz, Madrid, Spain
| | | | - Patricia Ruiz Garbajosa
- Red Española de Investigación en Patología Infecciosa (REIPI), Instituto de Salud Carlos III, Ministerio de Ciencia e Innovación, Sevilla, Spain
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Rosa Escudero-Sánchez
- Red Española de Investigación en Patología Infecciosa (REIPI), Instituto de Salud Carlos III, Ministerio de Ciencia e Innovación, Sevilla, Spain
- Servicio de Enfermedades Infecciosas, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Mar Sánchez Somolinos
- Servicio de Microbiología y Enfermedades Infecciosas, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Mercedes Marín Arriaza
- Servicio de Microbiología y Enfermedades Infecciosas, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- CIBER de Enfermedades Respiratorias (CIBERES CB06/06/0058), Madrid, Spain
| | - Juan Romanyk
- Servicio de Microbiología, Hospital Universitario Príncipe de Asturias, Alcalá de Henares, Madrid, Spain
| | - José María Barbero
- Servicio de Medicina Interna, Hospital Universitario Príncipe de Asturias, Alcalá de Henares, Madrid, Spain
| | - Ana Arribi Vilela
- Servicio de Microbiología, Hospital Clínico San Carlos, Madrid, Spain
| | | | - Conchita Pérez-Jorge
- Servicio de Microbiología, Hospital Universitario Rey Juan Carlos, Móstoles, Madrid, Spain
| | - David M. Arana
- Servicio de Microbiología, Hospital Universitario de Getafe, Getafe, Madrid, Spain
| | - Alfonso Monereo
- Servicio de Medicina Interna, Hospital Universitario de Getafe, Getafe, Madrid, Spain
| | - Diego Domingo
- Servicio de Microbiología, Hospital Universitario de La Princesa, Madrid, Spain
| | - José Cordero
- Servicio de Traumatología, Hospital Universitario de La Princesa, Madrid, Spain
| | | | | | - Jaime Lora-Tamayo
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- Red Española de Investigación en Patología Infecciosa (REIPI), Instituto de Salud Carlos III, Ministerio de Ciencia e Innovación, Sevilla, Spain
- Servicio de Medicina Interna, Hospital Universitario 12 de Octubre, Universidad Complutense, Madrid, Spain
| | - Fernando Chaves
- Servicio de Microbiología, Hospital Universitario 12 de Octubre, Universidad Complutense, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- Red Española de Investigación en Patología Infecciosa (REIPI), Instituto de Salud Carlos III, Ministerio de Ciencia e Innovación, Sevilla, Spain
| |
Collapse
|
28
|
Muñoz-Gallego I, Mancheño M, Pérez-Montarelo D, Viedma E, Chaves F, Lora-Tamayo J. Staphylococcus aureus native arthritis over 10 years. Med Mal Infect 2020; 50:257-262. [PMID: 32057526 DOI: 10.1016/j.medmal.2020.01.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 09/25/2018] [Accepted: 01/28/2020] [Indexed: 12/11/2022]
|
29
|
Subinhibitory Concentrations of Mupirocin Stimulate Staphylococcus aureus Biofilm Formation by Upregulating cidA. Antimicrob Agents Chemother 2020; 64:AAC.01912-19. [PMID: 31932378 DOI: 10.1128/aac.01912-19] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 12/04/2019] [Indexed: 01/02/2023] Open
Abstract
Previous studies have shown that the administration of antibiotics at subinhibitory concentrations stimulates biofilm formation by the majority of multidrug-resistant Staphylococcus aureus (MRSA) strains. Here, we investigated the effect of subinhibitory concentrations of mupirocin on biofilm formation by the community-associated (CA) mupirocin-sensitive MRSA strain USA300 and the highly mupirocin-resistant clinical S. aureus SA01 to SA05 isolates. We found that mupirocin increased the ability of MRSA cells to attach to surfaces and form biofilms. Confocal laser scanning microscopy (CLSM) demonstrated that mupirocin treatment promoted thicker biofilm formation, which also correlated with the production of extracellular DNA (eDNA). Furthermore, quantitative real-time PCR (RT-qPCR) results revealed that this effect was largely due to the involvement of holin-like and antiholin-like proteins (encoded by the cidA gene), which are responsible for modulating cell death and lysis during biofilm development. We found that cidA expression levels significantly increased by 6.05- to 35.52-fold (P < 0.01) after mupirocin administration. We generated a cidA-deficient mutant of the USA300 S. aureus strain. Exposure of the ΔcidA mutant to mupirocin did not result in thicker biofilm formation than that in the parent strain. We therefore hypothesize that the mupirocin-induced stimulation of S. aureus biofilm formation may involve the upregulation of cidA.
Collapse
|
30
|
Marquès C, Collin V, Franceschi C, Charbonnel N, Chatellier S, Forestier C. Fosfomycin and Staphylococcus aureus: transcriptomic approach to assess effect on biofilm, and fate of unattached cells. J Antibiot (Tokyo) 2019; 73:91-100. [PMID: 31705133 DOI: 10.1038/s41429-019-0256-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 08/08/2019] [Accepted: 10/14/2019] [Indexed: 11/09/2022]
Abstract
Interest has been rekindled in the old antibiotic fosfomycin, partly because of its ability to penetrate biofilm. Using a transcriptomic approach, we investigated the modifications induced by fosfomycin in sessile cells of a clinical Staphylococcus aureus isolated from a device-associated infection. Cells still able to form biofilm after 4 h of incubation in the presence of subinhibitory concentrations of fosfomycin and cells from 24-h-old biofilm later submitted to fosfomycin had 6.77% and 9.41%, respectively, of differentially expressed genes compared with their antibiotic-free control. Fosfomycin induced mostly downregulation of genes assigned to nucleotide, amino acid and carbohydrate transport, and metabolism. Adhesins and capsular biosynthesis proteins encoding genes were downregulated in fosfomycin-grown biofilm, whereas the murein hydrolase regulator lgrA and a D-lactate dehydrogenase-encoding gene were upregulated. In fosfomycin-treated biofilm, the expression of genes encoding adhesins, the cell wall biosynthesis protein ScdA, and to a lesser extent the fosfomycin target MurA was also decreased. Unattached cells surrounding fosfomycin-grown biofilm showed greater ability to form aggregates than their counterparts obtained without fosfomycin. Reducing their global metabolism and lowering cell wall turnover would allow some S. aureus cells to grow in biofilm despite fosfomycin stress while promoting hyperadherent phenotype in the vicinity of the fosfomycin-treated biofilm.
Collapse
Affiliation(s)
- Claire Marquès
- bioMérieux SA, 38390, La Balme les Grottes, France.,Laboratoire des Microorganismes, Génome et Environnement, UMR CNRS 6023-Université Clermont Auvergne, 63000, Clermont-Ferrand, France
| | | | | | - Nicolas Charbonnel
- Laboratoire des Microorganismes, Génome et Environnement, UMR CNRS 6023-Université Clermont Auvergne, 63000, Clermont-Ferrand, France
| | | | - Christiane Forestier
- Laboratoire des Microorganismes, Génome et Environnement, UMR CNRS 6023-Université Clermont Auvergne, 63000, Clermont-Ferrand, France.
| |
Collapse
|
31
|
Wang J, Wang J, Wang Y, Sun P, Zou X, Ren L, Zhang C, Liu E. Protein expression profiles in methicillin-resistant Staphylococcus aureus (MRSA) under effects of subminimal inhibitory concentrations of imipenem. FEMS Microbiol Lett 2019; 366:5570583. [PMID: 31529016 DOI: 10.1093/femsle/fnz195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 09/12/2019] [Indexed: 12/25/2022] Open
Abstract
Imipenem is a beta-lactam antibiotic mainly active against gram-negative bacterial pathogens and also could cause cell wall impairment in methicillin-resistant Staphylococcus aureus(MRSA). However, related antibacterial mechanisms of imipenem on MRSA and mixed infections of MRSA and gram-negative bacteria are relatively poorly revealed. This study was to identify proteins in the MRSA response to subminimal inhibitory concentrations (sub-MICs) of imipenem treatment. Our results showed that 240 and 58 different expression proteins (DEPs) in sub-MICs imipenem-treated S3 (a standard MRSA strain) and S23 (a clinical MRSA strain) strains were identified through the isobaric tag for relative and absolute quantitation method when compared with untreated S3 and S23 strains, respectively, which was further confirmed by multiple reactions monitoring. Our result also demonstrated that expressions of multiple DEPs involved in cellular proliferation, metabolism and virulence were significantly changed in S3 and S23 strains, which was proved by gene ontology annotations and qPCR analysis. Further, transmission electron microscopy and scanning electron microscopy analysis showed cell wall deficiency, cell lysis and abnormal nuclear mitosis on S23 strain. Our study provides important information for understanding the antibacterial mechanisms of imipenem on MRSA and for better usage of imipenem on patients co-infected with MRSA and other multidrug-resistant gram-negative bacteria.
Collapse
Affiliation(s)
- Jichun Wang
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, No. 136, Zhongshan 2nd Road, Yuzhong District, Chongqing 400014, China.,Department of Pediatrics, Affiliated Hospital of Inner Mongolia Medical University, No. 1, Tongdao North Street, Huimin District, Hohhot, Inner Mongolia 010050, China
| | - Junrui Wang
- Clinical Laboratory, Affiliated Hospital of Inner Mongolia Medical University, No. 1, Tongdao North Street, Huimin District, Hohhot, Inner Mongolia 010050, China
| | - Yanyan Wang
- Clinical Laboratory, Affiliated Hospital of Inner Mongolia Medical University, No. 1, Tongdao North Street, Huimin District, Hohhot, Inner Mongolia 010050, China
| | - Peng Sun
- Pathogen and Immunity Research Center, College of Basic Medicine, Inner Mongolia Medical University, Jinshan Avenue, Hohhot, Inner Mongolia 010110, China
| | - Xiaohui Zou
- Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention; China CDC, Key Laboratory for Medical Virology, Ministry of Health, Beijing 102206, China
| | - Luo Ren
- Pediatrics Institute, Children's Hospital Chongqing Medical University, No. 136, Zhong Shan 2nd Road, Yuzhong District, Chongqing 400014, China
| | - Chunxia Zhang
- Department of Pediatrics, Affiliated Hospital of Inner Mongolia Medical University, No. 1, Tongdao North Street, Huimin District, Hohhot, Inner Mongolia 010050, China
| | - Enmei Liu
- Pediatrics Institute, Children's Hospital Chongqing Medical University, No. 136, Zhong Shan 2nd Road, Yuzhong District, Chongqing 400014, China
| |
Collapse
|
32
|
Potential efficacy of garlic lock therapy in combating biofilm and catheter-associated infections; experimental studies on an animal model with focus on toxicological aspects. Saudi Pharm J 2019; 27:830-840. [PMID: 31516325 PMCID: PMC6734154 DOI: 10.1016/j.jsps.2019.05.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 05/14/2019] [Indexed: 12/29/2022] Open
Abstract
Background Life-threatening central venous catheter-related infections are primarily initiated by biofilm formation on the catheter surface. Antibiotic lock therapy is recommended for eradicating intraluminal biofilm. In the era of antibiotic resistance, antibiotics of natural origins provide an effective and cheap option for combating resistant strains. Garlic especially stole the spotlight because of its impressive antimicrobial effectiveness against such superbugs. Aim Is to estimate the potential use of fresh garlic extract (FGE) as a lock agent against multi-drug resistant (MDR) bacteria. Methods The agar well diffusion and broth microdilution techniques were employed to test the antimicrobial activities of FGE against five MDR strains; E. coli, Pseudomonas aeruginosa (P. aeruginosa), Klebsiella pneumoniae (K. pneumoniae), Serratia marscens (S. marscens) and Methicillin-resistant Staphylococcus aureus (MRSA). Then the protective and therapeutic efficiencies of FGE against bacterial biofilms were in-vitro evaluated; at concentrations of 100, 75, 50 and 25%; in tissue culture plate (TCP) and on the polyurethane (PU) sheets using the crystal violet (CV) assay and colony-forming unit (CFU), respectively. Scanning electron microscopy (SEM) was also used to confirm eradication of biofilms on PU sheets. Finally, systemic and deep tissue infections by P. aeruginosa and MRSA were induced in mice that were then treated by FGE at either 100 or 200 mg/kg for seven days. Where the antibacterial activity was assessed by tissue and blood culturing at the end of the treatment period. Biochemical, hematological and histological parameters were also investigated. Results FGE exhibited potent in-vitro and in-vivo antibacterial and antibiofilm activities against MDR strains. It not only didn’t exhibit toxicological effects at the hematological and the histological levels but also provided protective effects as demonstrated by the significant drop in the biochemical parameters. Conclusion FGE has the potential to be used as a prophylactic and/or therapeutic lock agent against biofilm-associated infections caused by MDR bacteria.
Collapse
|
33
|
Preliminary study on the effect of brazilin on biofilms of Staphylococcus aureus. Exp Ther Med 2018; 16:2108-2118. [PMID: 30186447 PMCID: PMC6122259 DOI: 10.3892/etm.2018.6403] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 06/01/2018] [Indexed: 12/13/2022] Open
Abstract
Biofilms significantly enhance antibiotic resistance by inhibiting penetration of antibiotics and are shielded from the immune system via the formation of an extracellular polymeric matrix. Innovative and novel approaches are required for the inhibition of biofilm formation and treatment of biofilm-associated infectious diseases. In the current study, a biofilm model of Staphylococcus aureus was established in vitro to explore inhibitory effects of brazilin (BN) on biofilm formation and to evaluate damaging effects of BN in the presence and absence of vancomycin (VCM) on the biofilm. Antibiofilm-infection mechanisms of BN were observed. In these experiments, the clinical strain of S. aureus C-4-4 was isolated for biofilm formation. Crystal violet staining and fluorescence microscopy revealed that BN inhibited biofilm formation in vitro and the best effect was observed with two times the minimum inhibitory concentration of BN following 48 h incubation. Additionally, the results demonstrated that BN in combination with VCM enhanced the damage to biofilms, whereas VCM alone did not. The results of the reverse transcription-quantitative polymerase chain reaction analyses demonstrated that BN downregulated gene expression of intercellular adhesion (ica)A and upregulated icaR and the quorum-sensing (QS) system regulator accessory gene regulator A. In summary, BN inhibited S. aureus biofilm formation and destroyed biofilms, while simultaneously increasing permeability to VCM. BN was able to reduce production of the extracellular polymeric matrix and inhibited the QS system. These results support the use of BN as a novel drug and treatment strategy for S. aureus biofilm-associated infections.
Collapse
|
34
|
Influence of subinhibitory concentrations of NH125 on biofilm formation & virulence factors of Staphylococcus aureus. Future Med Chem 2018; 10:1319-1331. [PMID: 29846088 DOI: 10.4155/fmc-2017-0286] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
AIM l-benzyl-3-cetyl-2-methylimidazolium iodide (NH125) can inhibit Staphylococcus aureus growth. We investigated the effects of sub-MIC concentrations of NH125 on S. aureus biofilm and virulence. Methodology & results: Three strains of S. aureus were tested. Sub-lethal concentrations of NH125 repressed biofilm formation. At partial sub-MICs, NH125 downregulated the expression of most virulence, while strain-dependent effects were found in the production of α-hemolysin, δ-hemolysin, coagulase and nuclease. In Galleria mellonella model, methicillin-resistant S. aureus pre-exposed to NH125 demonstrated significantly lower killing (p = 0.032 for 1/16 and 1/8 MICs; 0.008 for 1/4 MIC; and 0.001 for 1/2 MIC). CONCLUSION Sub-MIC concentrations of NH125 inhibited biofilm formation and virulence of S. aureus. These findings provide further support for evaluating the clinical efficacy of NH125 in staphylococcal infection.
Collapse
|
35
|
Ackerman DL, Craft KM, Doster RS, Weitkamp JH, Aronoff DM, Gaddy JA, Townsend SD. Antimicrobial and Antibiofilm Activity of Human Milk Oligosaccharides against Streptococcus agalactiae, Staphylococcus aureus, and Acinetobacter baumannii. ACS Infect Dis 2018; 4:315-324. [PMID: 29198102 DOI: 10.1021/acsinfecdis.7b00183] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
In a previous study, we reported that human milk oligosaccharides (HMOs) isolated from five donor milk samples possessed antimicrobial and antibiofilm activity against Streptococcus agalactiae, also known as Group B Streptococcus or GBS. Herein, we present a broader evaluation of the antimicrobial and antibiofilm activity by screening HMOs from 14 new donors against three strains of GBS and two of the ESKAPE pathogens of particular interest to child health, Staphylococcus aureus and Acinetobacter baumannii. Growth and biofilm assays showed that HMOs from these new donors possessed antimicrobial and antibiofilm activity against all three strains of GBS, antibiofilm activity against methicillin-resistant S. aureus strain USA300, and antimicrobial activity against A. baumannii strain ATCC 19606.
Collapse
Affiliation(s)
- Dorothy L. Ackerman
- Department of Chemistry, Vanderbilt University, 7330 Stevenson Center, Nashville, Tennessee 37235, United States
| | - Kelly M. Craft
- Department of Chemistry, Vanderbilt University, 7330 Stevenson Center, Nashville, Tennessee 37235, United States
| | - Ryan S. Doster
- Department of Medicine, Vanderbilt University Medical Center, 1161 21st Avenue South, D-3100 Medical Center North, Nashville, Tennessee 37232, United States
| | - Jörn-Hendrik Weitkamp
- Department of Medicine, Vanderbilt University Medical Center, 1161 21st Avenue South, D-3100 Medical Center North, Nashville, Tennessee 37232, United States
- Department of Pediatrics, Monroe Carell Jr. Children’s Hospital at Vanderbilt, 2200 Children’s Way, Suite 2404, Nashville, Tennessee 37232, United States
| | - David M. Aronoff
- Department of Medicine, Vanderbilt University Medical Center, 1161 21st Avenue South, D-3100 Medical Center North, Nashville, Tennessee 37232, United States
| | - Jennifer A. Gaddy
- Department of Medicine, Vanderbilt University Medical Center, 1161 21st Avenue South, D-3100 Medical Center North, Nashville, Tennessee 37232, United States
- Tennessee Valley Healthcare Systems, Department of Veterans Affairs, 1310 24th Avenue South, Nashville, Tennessee 37212, United States
| | - Steven D. Townsend
- Department of Chemistry, Vanderbilt University, 7330 Stevenson Center, Nashville, Tennessee 37235, United States
- Institute of Chemical Biology, Vanderbilt University, 896 Preston Research Building, Nashville, Tennessee 37232-6304, United States
| |
Collapse
|
36
|
Abstract
PURPOSE OF REVIEW Staphylococcus aureus (S. aureus) is well known for its ability to cause life-threatening infections. On the other hand, this bacterium can thrive as a commensal on and in human tissues without causing much problems. How big a threat is S. aureus actually? Furthermore, commensalism is associated with biofilms, where can we find them, and which natural and artificial components activate biofilm formation? RECENT FINDINGS Recent findings on S. aureus carriage on skin, mucosa, and in wounds indicate the presence of large numbers of S. aureus, yet its abundance can be without major implications for the host. S. aureus is often present in biofilms, together with other microorganisms, which can stimulate biofilm formation of S. aureus, in addition medicine including antibiotics can do the same. SUMMARY S. aureus can cause devastating infections, but when we take into consideration the ubiquitous presence of S. aureus, the risk seems to be relatively low. S. aureus forms biofilms in response to the 'hazards' on the human body, and signal to do so can come from various sources. All this has to be taken into consideration when we treat a patient as this might have enormous impact on the outcome.
Collapse
|
37
|
Abstract
Staphylococcus aureus is often involved in severe infections, in which the effects of bacterial virulence factors have great importance. Antistaphylococcal regimens should take into account the different effects of antibacterial agents on the expression of virulence factors and on the host's immune response. A PubMed literature search was performed to select relevant articles on the effects of antibiotics on staphylococcal toxin production and on the host immune response. Information was sorted according to the methods used for data acquisition (bacterial strains, growth models, and antibiotic concentrations) and the assays used for readout generation. The reported mechanisms underlying S. aureus virulence modulation by antibiotics were reviewed. The relevance of in vitro observations is discussed in relation to animal model data and to clinical evidence extracted from case reports and recommendations on the management of toxin-related staphylococcal diseases. Most in vitro data point to a decreased level of virulence expression upon treatment with ribosomally active antibiotics (linezolid and clindamycin), while cell wall-active antibiotics (beta-lactams) mainly increase exotoxin production. In vivo studies confirmed the suppressive effect of clindamycin and linezolid on virulence expression, supporting their utilization as a valuable management strategy to improve patient outcomes in cases of toxin-associated staphylococcal disease.
Collapse
|
38
|
Yan X, Gu S, Shi Y, Cui X, Wen S, Ge J. The effect of emodin on Staphylococcus aureus strains in planktonic form and biofilm formation in vitro. Arch Microbiol 2017; 199:1267-1275. [PMID: 28616631 DOI: 10.1007/s00203-017-1396-8] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 05/06/2017] [Accepted: 06/05/2017] [Indexed: 10/19/2022]
Abstract
Staphylococcus aureus (S. aureus) is a Gram-positive pathogen and forms biofilm easily. Bacteria inside biofilms display an increased resistance to antibiotics and disinfectants. The objective of the current study was to assess the antimicrobial activities of emodin, 1,2,8-trihydroxy-6-methylanthraquinone, an anthraquinone derivative isolated from Polygonum cuspidatum and Rheum palmatum, against S. aureus CMCC26003 grown in planktonic and biofilm cultures in vitro. In addition, a possible synergistic effect between emodin and berberine chloride was evaluated. As quantified by crystal violet method, emodin significantly decreased S. aureus biofilm growth in a dose-dependent manner. The above findings were further supported by scanning electron microscopy. Moreover, the present study demonstrated that sub-MICs emodin obviously intervened the release of extracellular DNA and inhibited expression of the biofilm-related genes (cidA, icaA, dltB, agrA, sortaseA and sarA) by real-time RT-PCR. These results revealed a promising application for emodin as a therapeutic agent and an effective strategy to prevent S. aureus biofilm-related infections.
Collapse
Affiliation(s)
- Xin Yan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Shanshan Gu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Yunjia Shi
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Xingyang Cui
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Shanshan Wen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Junwei Ge
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China.
| |
Collapse
|
39
|
Neoh KG, Li M, Kang ET, Chiong E, Tambyah PA. Surface modification strategies for combating catheter-related complications: recent advances and challenges. J Mater Chem B 2017; 5:2045-2067. [DOI: 10.1039/c6tb03280j] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This review summarizes the progress made in addressing bacterial colonization and other surface-related complications arising from catheter use.
Collapse
Affiliation(s)
- Koon Gee Neoh
- Department of Chemical and Biomolecular Engineering
- National University of Singapore
- Singapore 119077
| | - Min Li
- Department of Chemical and Biomolecular Engineering
- National University of Singapore
- Singapore 119077
| | - En-Tang Kang
- Department of Chemical and Biomolecular Engineering
- National University of Singapore
- Singapore 119077
| | - Edmund Chiong
- Department of Surgery
- National University of Singapore
- Singapore 119077
| | | |
Collapse
|
40
|
Dakheel KH, Abdul Rahim R, Neela VK, Al-Obaidi JR, Hun TG, Yusoff K. Methicillin-Resistant Staphylococcus aureus Biofilms and Their Influence on Bacterial Adhesion and Cohesion. BIOMED RESEARCH INTERNATIONAL 2016; 2016:4708425. [PMID: 28078291 PMCID: PMC5203895 DOI: 10.1155/2016/4708425] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 11/08/2016] [Accepted: 11/13/2016] [Indexed: 01/30/2023]
Abstract
Twenty-five methicillin-resistant Staphylococcus aureus (MRSA) isolates were characterized by staphylococcal protein A gene typing and the ability to form biofilms. The presence of exopolysaccharides, proteins, and extracellular DNA and RNA in biofilms was assessed by a dispersal assay. In addition, cell adhesion to surfaces and cell cohesion were evaluated using the packed-bead method and mechanical disruption, respectively. The predominant genotype was spa type t127 (22 out of 25 isolates); the majority of isolates were categorized as moderate biofilm producers. Twelve isolates displayed PIA-independent biofilm formation, while the remaining 13 isolates were PIA-dependent. Both groups showed strong dispersal in response to RNase and DNase digestion followed by proteinase K treatment. PIA-dependent biofilms showed variable dispersal after sodium metaperiodate treatment, whereas PIA-independent biofilms showed enhanced biofilm formation. There was no correlation between the extent of biofilm formation or biofilm components and the adhesion or cohesion abilities of the bacteria, but the efficiency of adherence to glass beads increased after biofilm depletion. In conclusion, nucleic acids and proteins formed the main components of the MRSA clone t127 biofilm matrix, and there seems to be an association between adhesion and cohesion in the biofilms tested.
Collapse
Affiliation(s)
- Khulood Hamid Dakheel
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor Darul Ehsan, Malaysia
| | - Raha Abdul Rahim
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor Darul Ehsan, Malaysia
- Institute of Bioscience, Universiti Putra Malaysia, 43400 Serdang, Selangor Darul Ehsan, Malaysia
| | - Vasantha Kumari Neela
- Department of Medical Microbiology and Parasitology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor Darul Ehsan, Malaysia
| | - Jameel R. Al-Obaidi
- Agro-Biotechnology Institute Malaysia (ABI), c/o MARDI Headquarters, 43400 Serdang, Selangor, Malaysia
| | - Tan Geok Hun
- Department of Agriculture Technology, Faculty of Agriculture, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Khatijah Yusoff
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor Darul Ehsan, Malaysia
- Institute of Bioscience, Universiti Putra Malaysia, 43400 Serdang, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
41
|
Miquel S, Lagrafeuille R, Souweine B, Forestier C. Anti-biofilm Activity as a Health Issue. Front Microbiol 2016; 7:592. [PMID: 27199924 PMCID: PMC4845594 DOI: 10.3389/fmicb.2016.00592] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 04/11/2016] [Indexed: 12/13/2022] Open
Abstract
The formation and persistence of surface-attached microbial communities, known as biofilms, are responsible for 75% of human microbial infections (National Institutes of Health). Biofilm lifestyle confers several advantages to the pathogens, notably during the colonization process of medical devices and/or patients’ organs. In addition, sessile bacteria have a high tolerance to exogenous stress including anti-infectious agents. Biofilms are highly competitive communities and some microorganisms exhibit anti-biofilm capacities such as bacterial growth inhibition, exclusion or competition, which enable them to acquire advantages and become dominant. The deciphering and control of anti-biofilm properties represent future challenges in human infection control. The aim of this review is to compare and discuss the mechanisms of natural bacterial anti-biofilm strategies/mechanisms recently identified in pathogenic, commensal and probiotic bacteria and the main synthetic strategies used in clinical practice, particularly for catheter-related infections.
Collapse
Affiliation(s)
- Sylvie Miquel
- Laboratoire Microorganismes : Génome et Environnement - UMR, CNRS 6023, Université Clermont Auvergne Clermont-Ferrand, France
| | - Rosyne Lagrafeuille
- Laboratoire Microorganismes : Génome et Environnement - UMR, CNRS 6023, Université Clermont Auvergne Clermont-Ferrand, France
| | - Bertrand Souweine
- Laboratoire Microorganismes : Génome et Environnement - UMR, CNRS 6023, Université Clermont AuvergneClermont-Ferrand, France; Service de Réanimation Médicale Polyvalente, CHU de Clermont-Ferrand, Clermont-FerrandFrance
| | - Christiane Forestier
- Laboratoire Microorganismes : Génome et Environnement - UMR, CNRS 6023, Université Clermont Auvergne Clermont-Ferrand, France
| |
Collapse
|