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
|
Xia Y, Hu Z, Jin Q, Chen Q, Zhao C, Qiang R, Xie Z, Li L, Zhang H. Structural characteristics, functions, and counteracting strategies of biofilms in Staphylococcus aureus. Comput Struct Biotechnol J 2025; 27:488-500. [PMID: 39916696 PMCID: PMC11799891 DOI: 10.1016/j.csbj.2025.01.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 02/09/2025] Open
Abstract
Background Staphylococcus aureus (S. aureus) is a prevalent pathogen associated with a wide range of infections, exhibiting significant antibiotic resistance and posing therapeutic challenges in clinical settings. The formation of biofilms contributes to the emergence of resistant strains, further diminishing the efficacy of antibiotics. This, in turn, leads to chronic and recurrent infections, ultimately increasing the healthcare burden. Consequently, preventing and eliminating biofilms has become a critical focus in clinical management and research. Aim of review This review systematically examines the mechanisms underlying biofilm formation in S. aureus and its contribution to antibiotic resistance, emphasizing the essential roles biofilms play in maintaining structural integrity and enhancing resistance. It also analyses the protective mechanisms that fortify S. aureus biofilms against antimicrobial treatments. Furthermore, the review provides a comprehensive overview of recent therapeutic innovations, including enzymatic therapy, nanotechnology, gene editing, and phage therapy. Key scientific concepts of review Emerging therapeutic strategies present novel approaches to combat S. aureus biofilm-associated infections through various mechanisms. This review discusses recent advancements in these therapies, their practical challenges in clinical application, and provides an in-depth analysis of each strategy's mechanisms and therapeutic potential. By mapping future research directions, this review aims to refine anti-biofilm strategies to control infection progression and effectively mitigate recurrence.
Collapse
Affiliation(s)
- Yanze Xia
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhenghui Hu
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Qiyuan Jin
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qi Chen
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Chenhao Zhao
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Rui Qiang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Zonggang Xie
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Liubing Li
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Haifang Zhang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, China
| |
Collapse
|
3
|
Al-Momani H, Aolymat I, Ibrahim L, Albalawi H, Al Balawi D, Albiss BA, Almasri M, Alghweiri S. Low-dose zinc oxide nanoparticles trigger the growth and biofilm formation of Pseudomonas aeruginosa: a hormetic response. BMC Microbiol 2024; 24:290. [PMID: 39095741 PMCID: PMC11297655 DOI: 10.1186/s12866-024-03441-y] [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: 02/22/2024] [Accepted: 07/25/2024] [Indexed: 08/04/2024] Open
Abstract
INTRODUCTION Hormesis describes an inverse dose-response relationship, whereby a high dose of a toxic compound is inhibitory, and a low dose is stimulatory. This study explores the hormetic response of low concentrations of zinc oxide nanoparticles (ZnO NPs) toward Pseudomonas aeruginosa. METHOD Samples of P. aeruginosa, i.e. the reference strain, ATCC 27,853, together with six strains recovered from patients with cystic fibrosis, were exposed to ten decreasing ZnO NPs doses (0.78-400 µg/mL). The ZnO NPs were manufactured from Peganum harmala using a chemical green synthesis approach, and their properties were verified utilizing X-ray diffraction and scanning electron microscopy. A microtiter plate technique was employed to investigate the impact of ZnO NPs on the growth, biofilm formation and metabolic activity of P. aeruginosa. Real-time polymerase chain reactions were performed to determine the effect of ZnO NPs on the expression of seven biofilm-encoding genes. RESULT The ZnO NPs demonstrated concentration-dependent bactericidal and antibiofilm efficiency at concentrations of 100-400 µg/mL. However, growth was significantly stimulated at ZnO NPs concentration of 25 µg/mL (ATCC 27853, Pa 3 and Pa 4) and at 12.5 µg/mL and 6.25 µg/mL (ATCC 27853, Pa 2, Pa 4 and Pa 5). No significant positive growth was detected at dilutions < 6.25 µg/mL. similarly, biofilm formation was stimulated at concentration of 12.5 µg/mL (ATCC 27853 and Pa 1) and at 6.25 µg/mL (Pa 4). At concentration of 12.5 µg/mL, ZnO NPs upregulated the expression of LasB ( ATCC 27853, Pa 1 and Pa 4) and LasR and LasI (ATCC 27853 and Pa 1) as well as RhII expression (ATCC 27853, Pa 2 and Pa 4). CONCLUSION When exposed to low ZnO NPs concentrations, P. aeruginosa behaves in a hormetic manner, undergoing positive growth and biofilm formation. These results highlight the importance of understanding the response of P. aeruginosa following exposure to low ZnO NPs concentrations.
Collapse
Affiliation(s)
- Hafez Al-Momani
- Department of Microbiology, Pathology and Forensic Medicine, Faculty of Medicine, The Hashemite University, P.O box 330127, Zarqa, 13133, Jordan.
| | - Iman Aolymat
- Department of Anatomy, Physiology and Biochemistry, Faculty of Medicine, The Hashemite University, P.O box 330127, Zarqa, 13133, Jordan
| | - Lujain Ibrahim
- Faculty of Applied Medical Sciences, The Hashemite University, Zarqa, 13133, Jordan
| | - Hadeel Albalawi
- Faculty of Applied Medical Sciences, The Hashemite University, Zarqa, 13133, Jordan
| | - Dua'a Al Balawi
- Faculty of Applied Medical Sciences, The Hashemite University, Zarqa, 13133, Jordan
| | - Borhan Aldeen Albiss
- Nanotechnology Institute, Jordan University of Science & Technology, P.O. Box 3030, Irbid, 22110, Jordan
| | - Muna Almasri
- Faculty of Applied Medical Sciences, The Hashemite University, Zarqa, 13133, Jordan
| | - Sahar Alghweiri
- Medical Laboratory Department, Prince Hashem Military Hospital, Zarqa, 13133, Jordan
| |
Collapse
|
4
|
Palkar PM, Palghadmal SB, Mane SS, Darne PA, Vidhate SV, Mehta NA, Padmanabhan S. Insights from degradation studies of alpha mangostin from Garcinia mangostana: key findings. Nat Prod Res 2024:1-7. [PMID: 39034458 DOI: 10.1080/14786419.2024.2378201] [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: 11/28/2023] [Accepted: 07/05/2024] [Indexed: 07/23/2024]
Abstract
The present study emphasises the necessity of substantiating the stability of plant-derived bioactive compounds for their therapeutic effectiveness in pharmaceutical production. The limelight is on alpha-mangostin (AM), a xanthone from Garcinia mangostana L., renowned for its diverse biological properties. Acid exposure during a forced degradation study on AM resulted in degraded alpha-mangostin (DAM) formation, with structural modifications of the two prenyl groups at C2 and C8 positions as determined by NMR and HRMS analysis. Other conditions (temperature, humidity, photolytic, oxidative, and alkaline) showed a minimal impact on AM. DAM, although showed antibacterial activity at concentration higher than AM (MIC values for AM: 0.39-1.56 µg/mL; DAM: >25 µg/mL), it exhibited potential for binding with Glucosyltransferase-SI from Streptococcus mutans and human Acetylcholinesterase in molecular docking simulations, comparable to AM. This suggests, the importance of prenyl group at C2 and C8 positions for AM's potent antibacterial activity and the decreased activity of DAM is due to lack of the prenyl groups.
Collapse
Affiliation(s)
- Pratixha M Palkar
- Innovation and Drug Discovery, SAVA Healthcare Limited, Research Centre, MIDC, Pune, India
| | - Shital B Palghadmal
- Innovation and Drug Discovery, SAVA Healthcare Limited, Research Centre, MIDC, Pune, India
| | - Shankar S Mane
- Microbiology Department, Innovation and Drug Discovery, SAVA Healthcare Limited, Research Centre, MIDC, Pune, India
| | - Priti A Darne
- Microbiology Department, Innovation and Drug Discovery, SAVA Healthcare Limited, Research Centre, MIDC, Pune, India
| | - Shankar V Vidhate
- Innovation and Drug Discovery, SAVA Healthcare Limited, Research Centre, MIDC, Pune, India
| | - Nisha A Mehta
- Microbiology Department, Innovation and Drug Discovery, SAVA Healthcare Limited, Research Centre, MIDC, Pune, India
| | - Sriram Padmanabhan
- Innovation and Drug Discovery, SAVA Healthcare Limited, Research Centre, MIDC, Pune, India
| |
Collapse
|
5
|
Singh CK, Sodhi KK, Shree P, Nitin V. Heavy Metals as Catalysts in the Evolution of Antimicrobial Resistance and the Mechanisms Underpinning Co-selection. Curr Microbiol 2024; 81:148. [PMID: 38642082 DOI: 10.1007/s00284-024-03648-2] [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: 09/08/2023] [Accepted: 02/22/2024] [Indexed: 04/22/2024]
Abstract
The menace caused by antibiotic resistance in bacteria is acknowledged on a global scale. Concerns over the same are increasing because of the selection pressure exerted by a huge number of different antimicrobial agents, including heavy metals. Heavy metals are non-metabolizable and recalcitrant to degradation, therefore the bacteria can expel the pollutants out of the system and make it less harmful via different mechanisms. The selection of antibiotic-resistant bacteria may be influenced by heavy metals present in environmental reservoirs. Through co-resistance and cross-resistance processes, the presence of heavy metals in the environment can act as co-selecting agents, hence increasing resistance to both heavy metals and antibiotics. The horizontal gene transfer or mutation assists in the selection of mutant bacteria resistant to the polluted environment. Hence, bioremediation and biodegradation are sustainable methods for the natural clean-up of pollutants. This review sheds light on the occurrence of metal and antibiotic resistance in the environment via the co-resistance and cross-resistance mechanisms underpinning co-selection emphasizing the dearth of studies that specifically examine the method of co-selection in clinical settings. Furthermore, it is advised that future research incorporate both culture- and molecular-based methodologies to further our comprehension of the mechanisms underlying bacterial co- and cross-resistance to antibiotics and heavy metals.
Collapse
Affiliation(s)
| | - Kushneet Kaur Sodhi
- Department of Zoology, Sri Guru Tegh Bahadur Khalsa College, University of Delhi, Delhi, 110007, India.
| | - Pallee Shree
- Department of Zoology, Lady Irwin College, University of Delhi, Delhi, 110001, India
| | - V Nitin
- Bhaskaracharya College of Applied Sciences, University of Delhi, Delhi, 110075, India
| |
Collapse
|
6
|
Liu J, Huang T, Xu Z, Mao Y, Soteyome T, Liu G, Qu C, Yuan L, Ma Q, Zhou F, Seneviratne G. Sub-MIC streptomycin and tetracycline enhanced Staphylococcus aureus Guangzhou-SAU749 biofilm formation, an in-depth study on transcriptomics. Biofilm 2023; 6:100156. [PMID: 37779859 PMCID: PMC10539642 DOI: 10.1016/j.bioflm.2023.100156] [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: 05/19/2023] [Revised: 09/16/2023] [Accepted: 09/18/2023] [Indexed: 10/03/2023] Open
Abstract
Staphylococcus aureus is a major human pathogen, a potential "Super-bug" and a typical biofilm forming bacteria. With usage of large amount of antibiotics, the residual antibiotics in clinical settings further complicate the colonization, pathogenesis and resistance of S. aureus. This study aimed at investigating the phenotypical and global gene expression changes on biofilm formation of a clinical S. aureus isolate treated under different types of antibiotics. Firstly, an isolate Guangzhou-SAU749 was selected from a large sale of previously identified S. aureus isolates, which exhibited weak biofilm formation in terms of biomass and viability. Secondly, 9 commonly prescribed antibiotics for S. aureus infections treatment, together with 10 concentrations ranging from 1/128 to 4 minimum inhibitory concentration (MIC) with 2-fold serial dilution, were used as different antibiotic stress conditions. Then, biofilm formation of S. aureus Guangzhou-SAU749 at different stages including 8 h, 16 h, 24 h, and 48 h, was tested by crystal violet and MTS assays. Thirdly, the whole genome of S. aureus Guangzhou-SAU749 was investigated by genome sequencing on PacBio platform. Fourthly, since enhancement of biofilm formation occurred when treated with 1/2 MIC tetracycline (TCY) and 1/4 MIC streptomycin (STR) since 5 h, the relevant biofilm samples were selected and subjected to RNA-seq and bioinformatics analysis. Last, expression of two component system (TCS) and biofilm associated genes in 4 h, 8 h, 16 h, 24 h, and 48 h sub-MIC TCY and STR treated biofilm samples were performed by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Although most antibiotics lowered the biomass and cell viability of Guangzhou-SAU749 biofilm at concentrations higher than MIC, certain antibiotics including TCY and STR promoted biofilm formation at sub-MICs. Additionally, upon genome sequencing, RNA-seq and RT-qPCR on biofilm samples treated with sub-MIC of TCY and STR at key time points, genes lytR, arlR, hssR, tagA, clfB, atlA and cidA related to TCS and biofilm formation were identified to contribute to the enhanced biofilm formation, providing a theoretical basis for further controlling on S. aureus biofilm formation.
Collapse
Affiliation(s)
- Junyan Liu
- College of Light Industry and Food Science, Guangdong Provincial Key Laboratory of Lingnan Specialty Food Science and Technology, Academy of Contemporary Agricultural Engineering Innovations, Zhongkai University of Agriculture and Engineering, Guangzhou, 510225, China
- Key Laboratory of Green Processing and Intelligent Manufacturing of Lingnan Specialty Food, Ministry of Agriculture, Guangzhou, 510225, China
| | - Tengyi Huang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Zhenbo Xu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Yuzhu Mao
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, South China University of Technology, Guangzhou, 510640, China
| | - Thanapop Soteyome
- Home Economics Technology, Rajamangala University of Technology Phra Nakhon, Bangkok, Thailand
| | - Gongliang Liu
- College of Light Industry and Food Science, Guangdong Provincial Key Laboratory of Lingnan Specialty Food Science and Technology, Academy of Contemporary Agricultural Engineering Innovations, Zhongkai University of Agriculture and Engineering, Guangzhou, 510225, China
- Key Laboratory of Green Processing and Intelligent Manufacturing of Lingnan Specialty Food, Ministry of Agriculture, Guangzhou, 510225, China
| | - Chunyun Qu
- College of Light Industry and Food Science, Guangdong Provincial Key Laboratory of Lingnan Specialty Food Science and Technology, Academy of Contemporary Agricultural Engineering Innovations, Zhongkai University of Agriculture and Engineering, Guangzhou, 510225, China
- Key Laboratory of Green Processing and Intelligent Manufacturing of Lingnan Specialty Food, Ministry of Agriculture, Guangzhou, 510225, China
| | - Lei Yuan
- School of Food Science and Engineering, Yangzhou University, Yangzhou, Jiangsu, 225127, PR China
| | - Qin Ma
- Sericultural & Agri-Food Research Institute Guangdong Academy of Agricultural Sciences/Key Laboratory of Functional Foods, Ministry of Agriculture /Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou, 510610, China
| | - Fang Zhou
- The First Affiliated Hospital, Sun Yan-Sen University, Guangzhou, 510080, China
| | - Gamini Seneviratne
- National Institute of Fundamental Studies, Hantana road, Kandy, Sri Lanka
| |
Collapse
|
7
|
Hernández-Cuellar E, Tsuchiya K, Valle-Ríos R, Medina-Contreras O. Differences in Biofilm Formation by Methicillin-Resistant and Methicillin-Susceptible Staphylococcus aureus Strains. Diseases 2023; 11:160. [PMID: 37987271 PMCID: PMC10660471 DOI: 10.3390/diseases11040160] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 10/27/2023] [Accepted: 11/02/2023] [Indexed: 11/22/2023] Open
Abstract
Staphylococcus aureus (S. aureus) is a common pathogen involved in community- and hospital-acquired infections. Its biofilm formation ability predisposes it to device-related infections. Methicillin-resistant S. aureus (MRSA) strains are associated with more serious infections and higher mortality rates and are more complex in terms of antibiotic resistance. It is still controversial whether MRSA are indeed more virulent than methicillin-susceptible S. aureus (MSSA) strains. A difference in biofilm formation by both types of bacteria has been suggested, but how only the presence of the SCCmec cassette or mecA influences this phenotype remains unclear. In this review, we have searched for literature studying the difference in biofilm formation by MRSA and MSSA. We highlighted the relevance of the icaADBC operon in the PIA-dependent biofilms generated by MSSA under osmotic stress conditions, and the role of extracellular DNA and surface proteins in the PIA-independent biofilms generated by MRSA. We described the prominent role of surface proteins with the LPXTG motif and hydrolases for the release of extracellular DNA in the MRSA biofilm formation. Finally, we explained the main regulatory systems in S. aureus involved in virulence and biofilm formation, such as the SarA and Agr systems. As most of the studies were in vitro using inert surfaces, it will be necessary in the future to focus on biofilm formation on extracellular matrix components and its relevance in the pathogenesis of infection by both types of strains using in vivo animal models.
Collapse
Affiliation(s)
- Eduardo Hernández-Cuellar
- Laboratorio de Biología Celular y Tisular, Departamento de Morfología, Universidad Autónoma de Aguascalientes, Aguascalientes 20100, C.P., México
| | - Kohsuke Tsuchiya
- Division of Immunology and Molecular Biology, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan;
| | - Ricardo Valle-Ríos
- Research Division, Faculty of Medicine, Universidad Nacional Autónoma de México (UNAM), Mexico City 04360, C.P., México;
- Laboratory of Research in Immunology and Proteomics, Federico Gómez Children’s Hospital of Mexico, Mexico City 06720, C.P., México
| | - Oscar Medina-Contreras
- Epidemiology, Endocrinology & Nutrition Research Unit, Mexico Children’s Hospital (HIMFG), Mexico City 06720, C.P., México;
| |
Collapse
|
8
|
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
|
9
|
Francis D, Veeramanickathadathil Hari G, Koonthanmala Subash A, Bhairaddy A, Joy A. The biofilm proteome of Staphylococcus aureus and its implications for therapeutic interventions to biofilm-associated infections. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 138:327-400. [PMID: 38220430 DOI: 10.1016/bs.apcsb.2023.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
Staphylococcus aureus is a major healthcare concern due to its ability to inflict life-threatening infections and evolve antibiotic resistance at an alarming pace. It is frequently associated with hospital-acquired infections, especially device-associated infections. Systemic infections due to S. aureus are difficult to treat and are associated with significant mortality and morbidity. The situation is worsened by the ability of S. aureus to form social associations called biofilms. Biofilms embed a community of cells with the ability to communicate with each other and share resources within a polysaccharide or protein matrix. S. aureus establish biofilms on tissues and conditioned abiotic surfaces. Biofilms are hyper-tolerant to antibiotics and help evade host immune responses. Biofilms exacerbate the severity and recalcitrance of device-associated infections. The development of a biofilm involves various biomolecules, such as polysaccharides, proteins and nucleic acids, contributing to different structural and functional roles. Interconnected signaling pathways and regulatory molecules modulate the expression of these molecules. A comprehensive understanding of the molecular biology of biofilm development would help to devise effective anti-biofilm therapeutics. Although bactericidal agents, antimicrobial peptides, bacteriophages and nano-conjugated anti-biofilm agents have been employed with varying levels of success, there is still a requirement for effective and clinically viable anti-biofilm therapeutics. Proteins that are expressed and utilized during biofilm formation, constituting the biofilm proteome, are a particularly attractive target for anti-biofilm strategies. The proteome can be explored to identify potential anti-biofilm drug targets and utilized for rational drug discovery. With the aim of uncovering the biofilm proteome, this chapter explores the mechanism of biofilm formation and its regulation. Furthermore, it explores the antibiofilm therapeutics targeted against the biofilm proteome.
Collapse
Affiliation(s)
- Dileep Francis
- Department of Life Sciences, Kristu Jayanti College (Autonomous), Bengaluru, India.
| | | | | | - Anusha Bhairaddy
- Department of Life Sciences, Kristu Jayanti College (Autonomous), Bengaluru, India
| | - Atheene Joy
- Department of Life Sciences, Kristu Jayanti College (Autonomous), Bengaluru, India
| |
Collapse
|
10
|
Fathil MAM, Katas H. Antibacterial, Anti-Biofilm and Pro-Migratory Effects of Double Layered Hydrogels Packaged with Lactoferrin-DsiRNA-Silver Nanoparticles for Chronic Wound Therapy. Pharmaceutics 2023; 15:pharmaceutics15030991. [PMID: 36986852 PMCID: PMC10054788 DOI: 10.3390/pharmaceutics15030991] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/15/2023] [Accepted: 03/15/2023] [Indexed: 03/22/2023] Open
Abstract
Antimicrobial resistance and biofilm formation in diabetic foot infections worsened during the COVID-19 pandemic, resulting in more severe infections and increased amputations. Therefore, this study aimed to develop a dressing that could effectively aid in the wound healing process and prevent bacterial infections by exerting both antibacterial and anti-biofilm effects. Silver nanoparticles (AgNPs) and lactoferrin (LTF) have been investigated as alternative antimicrobial and anti-biofilm agents, respectively, while dicer-substrate short interfering RNA (DsiRNA) has also been studied for its wound healing effect in diabetic wounds. In this study, AgNPs were complexed with LTF and DsiRNA via simple complexation before packaging in gelatin hydrogels. The formed hydrogels exhibited 1668% maximum swellability, with a 46.67 ± 10.33 µm average pore size. The hydrogels demonstrated positive antibacterial and anti-biofilm effects toward the selected Gram-positive and Gram-negative bacteria. The hydrogel containing AgLTF at 125 µg/mL was also non-cytotoxic on HaCaT cells for up to 72 h of incubation. The hydrogels containing DsiRNA and LTF demonstrated superior pro-migratory effects compared to the control group. In conclusion, the AgLTF-DsiRNA-loaded hydrogel possessed antibacterial, anti-biofilm, and pro-migratory activities. These findings provide a further understanding and knowledge on forming multipronged AgNPs consisting of DsiRNA and LTF for chronic wound therapy.
Collapse
|
11
|
França A. The Role of Coagulase-Negative Staphylococci Biofilms on Late-Onset Sepsis: Current Challenges and Emerging Diagnostics and Therapies. Antibiotics (Basel) 2023; 12:antibiotics12030554. [PMID: 36978421 PMCID: PMC10044083 DOI: 10.3390/antibiotics12030554] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/24/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023] Open
Abstract
Infections are one of the most significant complications of neonates, especially those born preterm, with sepsis as one of the principal causes of mortality. Coagulase-negative staphylococci (CoNS), a group of staphylococcal species that naturally inhabit healthy human skin and mucosa, are the most common cause of late-onset sepsis, especially in preterms. One of the risk factors for the development of CoNS infections is the presence of implanted biomedical devices, which are frequently used for medications and/or nutrient delivery, as they serve as a scaffold for biofilm formation. The major concerns related to CoNS infections have to do with the increasing resistance to multiple antibiotics observed among this bacterial group and biofilm cells’ increased tolerance to antibiotics. As such, the treatment of CoNS biofilm-associated infections with antibiotics is increasingly challenging and considering that antibiotics remain the primary form of treatment, this issue will likely persist in upcoming years. For that reason, the development of innovative and efficient therapeutic measures is of utmost importance. This narrative review assesses the current challenges and emerging diagnostic tools and therapies for the treatment of CoNS biofilm-associated infections, with a special focus on late-onset sepsis.
Collapse
Affiliation(s)
- Angela França
- Centre of Biological Engineering, LIBRO—Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal;
- LABBELS—Associate Laboratory in Biotechnology and Bioengineering and Microelectromechanical Systems, Braga and Guimarães, Portugal
| |
Collapse
|
12
|
Biology and Regulation of Staphylococcal Biofilm. Int J Mol Sci 2023; 24:ijms24065218. [PMID: 36982293 PMCID: PMC10049468 DOI: 10.3390/ijms24065218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/15/2023] [Accepted: 03/03/2023] [Indexed: 03/11/2023] Open
Abstract
Despite continuing progress in medical and surgical procedures, staphylococci remain the major Gram-positive bacterial pathogens that cause a wide spectrum of diseases, especially in patients requiring the utilization of indwelling catheters and prosthetic devices implanted temporarily or for prolonged periods of time. Within the genus, if Staphylococcus aureus and S. epidermidis are prevalent species responsible for infections, several coagulase-negative species which are normal components of our microflora also constitute opportunistic pathogens that are able to infect patients. In such a clinical context, staphylococci producing biofilms show an increased resistance to antimicrobials and host immune defenses. Although the biochemical composition of the biofilm matrix has been extensively studied, the regulation of biofilm formation and the factors contributing to its stability and release are currently still being discovered. This review presents and discusses the composition and some regulation elements of biofilm development and describes its clinical importance. Finally, we summarize the numerous and various recent studies that address attempts to destroy an already-formed biofilm within the clinical context as a potential therapeutic strategy to avoid the removal of infected implant material, a critical event for patient convenience and health care costs.
Collapse
|
13
|
Banu Raza F, Vijayaragavalu S, Kandasamy R, Krishnaswami V, Kumar V A. Microbiome and the inflammatory pathway in peri-implant health and disease with an updated review on treatment strategies. J Oral Biol Craniofac Res 2023; 13:84-91. [PMID: 36504486 PMCID: PMC9730223 DOI: 10.1016/j.jobcr.2022.11.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 08/30/2022] [Accepted: 11/25/2022] [Indexed: 12/04/2022] Open
Abstract
Crestal bone preservation around the dental implant for aesthetic and functional success is widely researched and documented over a decade. Several etiological factors were put forth for crestal bone loss; of which biofilm plays a major role. Biofilm is formed by the colonization of wide spectra of bacteria inhabited around dental implants. Bacterial adherence affects the regulators of bone growth and an early intervention preserves the peri-implant bone. Primary modes of therapy stated in early literature were either prevention or treatment of infection caused by biofilm. This narrative review overviews the microbiome during different stages of peri-implant health, the mechanism of bone destruction, and the expression of the biomarkers at each stage. Microbial contamination and the associated biomarkers varied depending on the stage of peri-implant infection. The comprehensive review helps in formulating a research plan, both in diagnostics and treatment aspects in improving peri-implant health.
Collapse
Key Words
- Antibiotics
- Biomarkers
- CD14, Cluster of Differentiation 14
- CSF, Colony-Stimulating Factor
- Gene expression
- IL, Interleukins
- MMP 8, Matrix MetalloProteinase 8
- Microbiota
- OPG, Osteoprotegerin
- PSMB 2, Proteasome subunit beta type-2
- Peri-implant
- RANK, Receptor Activator of Nuclear factor Kappa-Β
- RANKL, Receptor Activator of Nuclear factor Kappa-ΒLigand
- TIMP, Tissue inhibitor of Metalloproteinase
- TNF, Tumor Necrosis Factor
- TWEAK, TNF-related weak inducer of apoptosis
- VEGF, Vascular Endothelial Growth Factor
- sRANKL, soluble Receptor Activator of Nuclear Factor-κB Ligand
Collapse
Affiliation(s)
- Fathima Banu Raza
- Department of Prosthodontics, Faculty of Dental Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | | | - Ruckmani Kandasamy
- Centre for Excellence in Nanobio Translational REsearch (CENTRE), Department of Pharmaceutical Technology, University College of Engineering, Anna University, BIT Campus, Tiruchirappalli, Tamil Nadu, India
| | - Venkateshwaran Krishnaswami
- Centre for Excellence in Nanobio Translational REsearch (CENTRE), Department of Pharmaceutical Technology, University College of Engineering, Anna University, BIT Campus, Tiruchirappalli, Tamil Nadu, India
| | - Anand Kumar V
- Department of Prosthodontics, Faculty of Dental Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, India
- Corresponding author. Department of Prosthodontics, Faculty of Dental Sciences, SRIHER (DU), Porur, Chennai, Tamil Nadu, India.
| |
Collapse
|
14
|
Liang J, Huang TY, Mao Y, Li X. Biofilm formation of two genetically diverse Staphylococcus aureus isolates under beta-lactam antibiotics. Front Microbiol 2023; 14:1139753. [PMID: 36950159 PMCID: PMC10025342 DOI: 10.3389/fmicb.2023.1139753] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 01/30/2023] [Indexed: 03/08/2023] Open
Abstract
PURPOSE Our aim was to evaluate the biofilm formation of 2 genetically diverse Staphylococcus aureus isolates, 10379 and 121940, under different concentrations of beta-lactam antibiotics on biomass content and biofilm viability. METHODS Biofilm formation and methicillin resistance genes were tested using PCR and multiplex PCR. PCR was combined with bioinformatics analysis to detect multilocal sequence typing (MLST) and SCCmec types, to study the genetical correlation between the tested strains. Then, the crystal violet (CV) test and XTT were used to detect biomass content and biofilm activity. Antibiotic susceptibility was tested using a broth dilution method. According to their specific MIC, different concentrations of beta-lactam antibiotics were used to study its effect on biomass content and biofilm viability. RESULTS Strain 10379 carried the icaD, icaBC, and MRSA genes, not the icaA, atl, app, and agr genes, and MLST and SCCmec typing was ST45 and IV, respectively. Strain 121940 carried the icaA, icaD, icaBC, atl, and agr genes, not the aap gene, and MLST and SCCmec typed as ST546 and IV, respectively. This suggested that strains 10379 and 121940 were genotypically very different. Two S. aureus isolates, 10379 and 121940, showed resistance to beta-lactam antibiotics, penicillin, ampicillin, meropenem, streptomycin and kanamycin, some of which promoted the formation of biofilm and biofilm viability at low concentrations. CONCLUSION Despite the large differences in the genetic background of S. aureus 10379 and 121940, some sub-inhibitory concentrations of beta-lactam antibiotics are able to promote biomass and biofilm viability of both two isolates.
Collapse
Affiliation(s)
- Jinglong Liang
- College of Light Industry and Food Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China
- Guangdong Provincial Key Laboratory of Lingnan Specialty Food Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China
- Key Laboratory of Green Processing and Intelligent Manufacturing of Lingnan Specialty Food, Ministry of Agriculture, Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Teng Yi Huang
- Department of Diagnostics, Second Affiliated Hospital of Shantou University Medical College, Shantou, China
- Teng Yi Huang,
| | - Yuzhu Mao
- Department of Civil and Environmental Engineering, University of Maryland, College Park, MD, United States
| | - Xuejie Li
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, South China University of Technology, Guangzhou, China
- Research Institute for Food Nutrition and Human Health, Guangzhou, China
- *Correspondence: Xuejie Li,
| |
Collapse
|
15
|
He X, Zhang W, Cao Q, Li Y, Bao G, Lin T, Bao J, Chang C, Yang C, Yin Y, Xu J, Ren Z, Jin Y, Lu F. Global Downregulation of Penicillin Resistance and Biofilm Formation by MRSA Is Associated with the Interaction between Kaempferol Rhamnosides and Quercetin. Microbiol Spectr 2022; 10:e0278222. [PMID: 36354319 PMCID: PMC9769653 DOI: 10.1128/spectrum.02782-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 10/24/2022] [Indexed: 11/12/2022] Open
Abstract
The rapid development of methicillin-resistant Staphylococcus aureus (MRSA) drug resistance and the formation of biofilms seriously challenge the clinical application of classic antibiotics. Extracts of the traditional herb Chenopodium ambrosioides L. were found to have strong antibiofilm activity against MRSA, but their mechanism of action remains poorly understood. This study was designed to investigate the antibacterial and antibiofilm activities against MRSA of flavonoids identified from C. ambrosioides L. in combination with classic antibiotics, including ceftazidime, erythromycin, levofloxacin, penicillin G, and vancomycin. Liquid chromatography-mass spectrometry (LC-MS) was used to analyze the nonvolatile chemical compositions. Reverse transcription (RT)-PCR was used to investigate potential multitargets of flavonoids based on global transcriptional responses of virulence and antibiotic resistance. A synergistic antibacterial and biofilm-inhibiting activity of the alcoholic extract of the ear of C. ambrosioides L. in combination with penicillin G was observed against MRSA, which proved to be closely related to the interaction of the main components of kaempferol rhamnosides with quercetin. In regard to the mechanism, the increased sensitivity of MRSA to penicillin G was shown to be related to the downregulation of penicillinase with SarA as a potential drug target, while the antibiofilm activity was mainly related to downregulation of various virulence factors involved in the initial and mature stages of biofilm development, with SarA and/or σB as drug targets. This study provides a theoretical basis for further exploration of the medicinal activity of kaempferol rhamnosides and quercetin and their application in combination with penicillin G against MRSA biofilm infection. IMPORTANCE In this study, the synergistic antibacterial and antibiofilm effects of the traditional herb C. ambrosioides L. and the classic antibiotic penicillin G on MRSA provide a potential strategy to deal with the rapid development of MRSA antibiotic resistance. This study also provides a theoretical basis for further optimizing the combined effect of kaempferol rhamnosides, quercetin, and penicillin G and exploring anti-MRSA biofilm infection research with SarA and σB as drug targets.
Collapse
Affiliation(s)
- Xinlong He
- Department of Pathogenic Biology, School of Medicine, Yangzhou University, Yangzhou, People’s Republic of China
- Affiliated Hospital of Yangzhou University, Yangzhou, People’s Republic of China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, People’s Republic of China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, School of Medicine, Yangzhou University, Yangzhou, People’s Republic of China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, People’s Republic of China
| | - Wenwen Zhang
- Department of Pathogenic Biology, School of Medicine, Yangzhou University, Yangzhou, People’s Republic of China
| | - Qingchao Cao
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou, People’s Republic of China
| | - Yinyue Li
- Department of Pathogenic Biology, School of Medicine, Yangzhou University, Yangzhou, People’s Republic of China
| | - Guangyu Bao
- Affiliated Hospital of Yangzhou University, Yangzhou, People’s Republic of China
| | - Tao Lin
- Affiliated Hospital of Yangzhou University, Yangzhou, People’s Republic of China
| | - Jiaojiao Bao
- Affiliated Hospital of Yangzhou University, Yangzhou, People’s Republic of China
| | - Caiwang Chang
- Affiliated Hospital of Yangzhou University, Yangzhou, People’s Republic of China
| | - Changshui Yang
- Department of Pharmacy, School of Medicine, Yangzhou University, Yangzhou, People’s Republic of China
| | - Yi Yin
- Department of Pathogenic Biology, School of Medicine, Yangzhou University, Yangzhou, People’s Republic of China
| | - Jiahui Xu
- Department of Pathogenic Biology, School of Medicine, Yangzhou University, Yangzhou, People’s Republic of China
| | - Zhenyu Ren
- Department of Pathogenic Biology, School of Medicine, Yangzhou University, Yangzhou, People’s Republic of China
| | - Yingshan Jin
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou, People’s Republic of China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, People’s Republic of China
| | - Feng Lu
- Department of Pathogenic Biology, School of Medicine, Yangzhou University, Yangzhou, People’s Republic of China
- Affiliated Hospital of Yangzhou University, Yangzhou, People’s Republic of China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, People’s Republic of China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, School of Medicine, Yangzhou University, Yangzhou, People’s Republic of China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, People’s Republic of China
| |
Collapse
|
16
|
Mirzaei R, Yousefimashouf R, Arabestani MR, Sedighi I, Alikhani MY. The issue beyond resistance: Methicillin-resistant Staphylococcus epidermidis biofilm formation is induced by subinhibitory concentrations of cloxacillin, cefazolin, and clindamycin. PLoS One 2022; 17:e0277287. [PMID: 36350834 PMCID: PMC9645612 DOI: 10.1371/journal.pone.0277287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 10/25/2022] [Indexed: 11/11/2022] Open
Abstract
Staphylococcus epidermis is one of the most frequent causes of device-associated infections due to biofilm formation. Current reports noted that subinhibitory concentrations of antibiotics induce biofilm production in some bacteria. Accordingly, we evaluated the effect of exposure of different subinhibitory concentrations of cloxacillin, cefazolin, clindamycin, and vancomycin on the biofilm formation of methicillin-resistant S. epidermidis (MRSE). Antimicrobial susceptibility testing and minimum inhibitory/bactericidal concentration of antimicrobial agents were determined. MRSE isolates were selected, and their biofilm formation ability was evaluated. The effect of subinhibitory concentrations of cloxacillin, cefazolin, clindamycin, and vancomycin, antibiotics selected among common choices in the clinic, on MRSE biofilm formation was determined by the microtitre method. Besides, the effect of subinhibitory concentrations of cloxacillin, cefazolin, clindamycin, and vancomycin on the expression of the biofilm-associated genes icaA and atlE was evaluated by Reverse-transcription quantitative real-time polymerase chain reaction (RT-qPCR). Antimicrobial susceptibility patterns of MRSE strains showed a high level of resistance as follows: 80%, 53.3%, 33.3%, 33.3%, and 26.6%, for erythromycin, trimethoprim-sulfamethoxazole, tetracycline, clindamycin, and gentamicin, respectively. Besides, 73.3% of S. epidermidis strains were Multidrug-resistant (MDR). Minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) values were in the range of 0.5 to512 μg/mL and 1 to1024 μg/mL for cloxacillin, 0.125 to256 μg/mL and 1 to512 μg/mL for cefazolin, 0.125 to64 μg/mL and 4 to>1024 μg/mL for clindamycin, and 2 to32 μg/mL and 4 to32 μg/mL for vancomycin, respectively. The findings showed that subinhibitory concentrations of cloxacillin, cefazolin, and clindamycin induce biofilm production in MRSE strains. In particular, the OD values of strains were in the range of 0.09-0.95, 0.05-0.86, and 0.06-1 toward cloxacillin, cefazolin, and clindamycin, respectively. On the other hand, exposure to subinhibitory vancomycin concentrations did not increase the biofilm formation in MRSE strains. The findings also demonstrated that sub-MIC of antibiotics up-regulated biofilm-associated genes. In particular, atlE and icaA were up-regulated 0.062 to 1.16 and 0.078 to 1.48 folds, respectively, for cloxacillin, 0.11 to 0.8, and 0.1 to 1.3 folds for cefazolin, 0.18 to 0.98, and 0.19 to 1.4 folds, respectively, for clindamycin. In contrast, the results showed that sub-MIC of vancomycin did not increase the biofilm-associated genes. These findings overall show that exposure to sub-MIC of traditional antibiotics can cause biofilm induction in MRSE, thereby increasing the survival and persistence on various surfaces that worsen the condition of comorbid infections.
Collapse
Affiliation(s)
- Rasoul Mirzaei
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rasoul Yousefimashouf
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Reza Arabestani
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Iraj Sedighi
- Department of Pediatrics, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Yousef Alikhani
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Brucellosis Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
17
|
Yang Y, Liu J, Xue T, Hanamoto S, Wang H, Sun P, Zhao L. Complex behavior between microplastic and antibiotic and their effect on phosphorus-removing Shewanella strain during wastewater treatment. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 845:157260. [PMID: 35820524 DOI: 10.1016/j.scitotenv.2022.157260] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/26/2022] [Accepted: 07/06/2022] [Indexed: 06/15/2023]
Abstract
Owing to their widespread application and use, microplastics (MPs) and antibiotics coexist in the sewage treatment systems. In this study, the effects and mechanisms of the combined stress of MPs and ciprofloxacin (CIP) on phosphorus removal by phosphorus-accumulating organisms (PAOs) were investigated. This study found that the four types of MPs and CIP exhibited different antagonistic effects on the inhibition of phosphorus removal by PAO. MPs reduced the effective concentration of CIP through adsorption and thus reduced its toxicity, which was affected by the biofilms on MPs. In addition, CIP may cause PAO to produce more extracellular polymeric substances, which reduces the physical and oxidative stress of MPs on PAO. Our results are helpful as they increase the understanding of the effects of complex emerging pollutants in sewage systems and propose measures to strengthen the biological phosphorus removal in sewage treatment processes.
Collapse
Affiliation(s)
- Yongkui Yang
- School of Environmental Science and Engineering, Tianjin University, Tianjin 300072, China.
| | - Jinyi Liu
- School of Environmental Science and Engineering, Tianjin University, Tianjin 300072, China
| | - Tongyu Xue
- School of Environmental Science and Engineering, Tianjin University, Tianjin 300072, China
| | - Seiya Hanamoto
- Environment Preservation Center, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
| | - Hongyang Wang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China
| | - Peizhe Sun
- School of Environmental Science and Engineering, Tianjin University, Tianjin 300072, China
| | - Lin Zhao
- School of Environmental Science and Engineering, Tianjin University, Tianjin 300072, China
| |
Collapse
|
18
|
Celebi D, Taghizadehghalehjoughi A, Baser S, Genc S, Yilmaz A, Yeni Y, Yesilyurt F, Yildirim S, Bolat I, Kordali S, Yilmaz F, Hacimuftuoglu A, Celebi O, Margina D, Nitulescu GM, Spandidos DA, Tsatsakis A. Effects of boric acid and potassium metaborate on cytokine levels and redox stress parameters in a wound model infected with methicillin‑resistant Staphylococcus aureus. Mol Med Rep 2022; 26:294. [PMID: 35920188 PMCID: PMC9366158 DOI: 10.3892/mmr.2022.12809] [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: 02/12/2022] [Accepted: 06/23/2022] [Indexed: 11/14/2022] Open
Abstract
Methicillin‑resistant Staphylococcus aureus (MRSA) infections are usually found in hospital settings and, frequently, in patients with open wounds. One of the most critical virulence factors affecting the severity and recurrence of infections is the biofilm; increasing antibiotic resistance due to biofilm formation has led to the search for alternative compounds to antibiotics. The present study aimed to use boric acid and potassium metaborate against MRSA infection in a fibroblast wound model. For this purpose, a two‑part experiment was designed: First, MRSA strains were used for the test, and both boric acid and potassium metaborate were prepared in microdilution. In the second step, an MRSA wound model was prepared using a fibroblast culture, and treatments with boric acid and potassium metaborate were applied for 24 h. For the evaluation of the effects of treatment, cell viability assay (MTT assay), analysis of redox stress parameters, including total oxidant status and total antioxidant capacity analyses, lactate dehydrogenase analysis and immunohistochemical staining were performed. In addition, IL‑1β and IL‑10 gene expression levels were assayed. According to the results, potassium metaborate was more effective and exhibited a lower toxicity to fibroblast cells compared to boric acid; moreover, potassium metaborate decreased the level of prooxidant species and increased the antioxidant status more effectively than boric acid. The IL‑1β level in the bacteria group was high; however, boric acid and potassium metaborate significantly decreased the expression levels of inflammatory markers, exhibiting the potential to improve the resolution of the lesion. On the whole, the findings of the present study suggest that boric acid and potassium metaborate may be effective on the tested microorganisms.
Collapse
Affiliation(s)
- Demet Celebi
- Department of Microbiology, Faculty of Veterinary Medicine, Ataturk University, 25240 Erzurum, Turkey
| | | | - Sumeyye Baser
- Department of Medical Microbiology, Faculty of Medicine, Ataturk University, 25240 Erzurum, Turkey
| | - Sidika Genc
- Department of Medical Pharmacology, Faculty of Medicine, Seyh Edebali University, 11000 Bilecik, Turkey
| | - Aysegul Yilmaz
- Department of Medical Pharmacology, Faculty of Medicine, Ataturk University, 25240 Erzurum, Turkey
| | - Yesim Yeni
- Department of Medical Pharmacology, Faculty of Medicine, Ataturk University, 25240 Erzurum, Turkey
| | - Fatma Yesilyurt
- Department of Medical Pharmacology, Faculty of Medicine, Ataturk University, 25240 Erzurum, Turkey
| | - Serkan Yildirim
- Department of Pathology, Faculty of Veterinary Medicine, Ataturk University, 25240 Erzurum, Turkey
| | - Ismail Bolat
- Department of Pathology, Faculty of Veterinary Medicine, Ataturk University, 25240 Erzurum, Turkey
| | - Saban Kordali
- Department of Plant Protection, Fethiye Faculty of Agriculture, Mugla Sitki Kocman University, 48000 Mugla, Turkey
| | - Ferah Yilmaz
- Department of Plant Protection, Fethiye Faculty of Agriculture, Mugla Sitki Kocman University, 48000 Mugla, Turkey
| | - Ahmet Hacimuftuoglu
- Department of Medical Pharmacology, Faculty of Medicine, Ataturk University, 25240 Erzurum, Turkey
| | - Ozgur Celebi
- Department of Medical Microbiology, Faculty of Medicine, Ataturk University, 25240 Erzurum, Turkey
| | - Denisa Margina
- Department of Pharmacy, Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 020956 Bucharest, Romania
| | - George Mihai Nitulescu
- Department of Pharmacy, Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 020956 Bucharest, Romania
| | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Aristidis Tsatsakis
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece
| |
Collapse
|
19
|
Umar NK, Kono M, Sakatani H, Murakami D, Onishi Y, Kamiyama T, Iyo T, Hijiya M, Shiga T, Kinoshita T, Tamagawa S, Hiraoka M, Ohtani M, Hotomi M. Respiratory quinolones can eradicate amoxicillin-induced mature biofilms and nontypeable Haemophilus influenzae in biofilms. J Infect Chemother 2022; 28:1595-1604. [PMID: 36002133 DOI: 10.1016/j.jiac.2022.07.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 06/23/2022] [Accepted: 07/28/2022] [Indexed: 10/15/2022]
Abstract
OBJECTIVES Biofilm is thought to be involved in the persistent bacterial infections caused by nontypeable Haemophilus influenzae (NTHi). This study aims to evaluate the efficacy of antibiotics against NTHi biofilms. METHODS A 96-wells pin replicator assay was applied for evaluation of antimicrobial efficacies against NTHi biofilms. The NTHi IH-202 strain for the standard and 10 clinical strains were evaluated, as well as the viability of NTHi in biofilms after antimicrobial exposures. RESULTS Biofilms formed by IH-202 strain accumulated during incubation. AMPC if not high concentrations, neither reduce or inhibit biofilm formation, nor eradicate matured NTHi biofilms. The NTHi in matured biofilm were alive after exposure to amoxicillin (AMPC). Even high concentration of AMPC produced live NTHi after suspension of exposure, while tosufloxacin and garenoxacin inhibited biofilm formation of NTHi and eradicated matured biofilms. The respiratory quinolones, but not AMPC, killed NTHi in biofilms even at sub-MIC. CONCLUSIONS NTHi persists in biofilms, even after exposure to AMPC. These findings may eventually lead to a better understanding of effective use of antibiotics to eradicate NTHi growing as biofilms, or even to the development of novel therapeutic agents for treating patients with mucosal NTHi biofilm infections. Meanwhile, respiratory quinolones are attractive agents in reducing NTHi biofilm formation and destroying established biofilm.
Collapse
Affiliation(s)
- Nafisa Khamis Umar
- Department of Otorhinolaryngology Head and Neck Surgery, Wakayama Medical University, 811-1 Kimiidera, Wakayama-shi, Wakayama, 641-5810, Japan
| | - Masamitsu Kono
- Department of Otorhinolaryngology Head and Neck Surgery, Wakayama Medical University, 811-1 Kimiidera, Wakayama-shi, Wakayama, 641-5810, Japan
| | - Hideki Sakatani
- Department of Otorhinolaryngology Head and Neck Surgery, Wakayama Medical University, 811-1 Kimiidera, Wakayama-shi, Wakayama, 641-5810, Japan
| | - Daichi Murakami
- Department of Otorhinolaryngology Head and Neck Surgery, Wakayama Medical University, 811-1 Kimiidera, Wakayama-shi, Wakayama, 641-5810, Japan; Department of Otorhinolaryngology Head and Neck Surgery, Kinan Hospital, 46-70 Shinjo-cho, Tanabe-shi, Wakayama, 646-8588, Japan
| | - Yoshimi Onishi
- FUJIFILM Toyama Chemical Co., Ltd., 14-1, Kyobashi 2-Chome, Chuo-Ku, Tokyo, 104-0031, Japan
| | - Tomoko Kamiyama
- FUJIFILM Toyama Chemical Co., Ltd., 14-1, Kyobashi 2-Chome, Chuo-Ku, Tokyo, 104-0031, Japan
| | - Takuro Iyo
- Department of Otorhinolaryngology Head and Neck Surgery, Wakayama Medical University, 811-1 Kimiidera, Wakayama-shi, Wakayama, 641-5810, Japan
| | - Masayoshi Hijiya
- Department of Otorhinolaryngology Head and Neck Surgery, Wakayama Medical University, 811-1 Kimiidera, Wakayama-shi, Wakayama, 641-5810, Japan
| | - Tatsuya Shiga
- Department of Otorhinolaryngology Head and Neck Surgery, Wakayama Medical University, 811-1 Kimiidera, Wakayama-shi, Wakayama, 641-5810, Japan
| | - Tetsuya Kinoshita
- Department of Otorhinolaryngology Head and Neck Surgery, Wakayama Medical University, 811-1 Kimiidera, Wakayama-shi, Wakayama, 641-5810, Japan
| | - Shunji Tamagawa
- Department of Otorhinolaryngology Head and Neck Surgery, Wakayama Medical University, 811-1 Kimiidera, Wakayama-shi, Wakayama, 641-5810, Japan
| | - Masanobu Hiraoka
- Department of Otorhinolaryngology Head and Neck Surgery, Wakayama Medical University, 811-1 Kimiidera, Wakayama-shi, Wakayama, 641-5810, Japan
| | - Makiko Ohtani
- Department of Otorhinolaryngology Head and Neck Surgery, Wakayama Medical University, 811-1 Kimiidera, Wakayama-shi, Wakayama, 641-5810, Japan
| | - Muneki Hotomi
- Department of Otorhinolaryngology Head and Neck Surgery, Wakayama Medical University, 811-1 Kimiidera, Wakayama-shi, Wakayama, 641-5810, Japan.
| |
Collapse
|
20
|
M Fathil MA, Faris Taufeq FY, Suleman Ismail Abdalla S, Katas H. Roles of chitosan in synthesis, antibacterial and anti-biofilm properties of bionano silver and gold. RSC Adv 2022; 12:19297-19312. [PMID: 35865585 PMCID: PMC9248368 DOI: 10.1039/d2ra01734b] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 06/24/2022] [Indexed: 11/21/2022] Open
Abstract
Antibiotic-resistance and bacterial bioburden on wound surfaces are the significant challenges to wound healing. Silver and gold nanoparticles (are termed as AgNPs and AuNPs) have been investigated as alternative antimicrobial agents to combat antibiotic-resistant bacterial infections owing to their antibacterial and anti-biofilm activities. Chitosan (CS) has largely been used in nanoparticle synthesis as a stabilizing or capping agent. In this study, AgNPs and AuNPs were synthesized using different concentrations of aqueous extract of tiger milk mushroom (Lignosus rhinocerotis) (WETMM) and CS as reducing and stabilizing agent, respectively. Particle size and morphology of both were determined by dynamic light scattering (DLS) method and transmission electron microscopic analysis (TEM). FTIR analysis was conducted to determine the interactions between nanoparticle precursors. The observed peaks at 450 nm and 534-565 nm using a spectrophotometer were corresponded to the surface Plasmon resonance of AgNPs and AuNPs respectively, indicating the formation of respective nanoparticles. FTIR analysis confirmed the role of WETMM as a reducing agent and CS as a stabilizer of AgNPs and AuNPs. Faster formation of nanoparticles was observed besides an increase in particle size when higher CS concentrations were used. TEM micrographs revealed the spherical shape of most nanoparticles with particle sizes in the range of 4 to 58 nm and 18 to 28 nm for AgNPs and AuNPs, respectively. Both nanoparticles exhibited antimicrobial activity against Gram-positive and -negative bacteria, with AgNPs showing a superior antibacterial efficacy than AuNPs. Both microbroth dilution and agar well diffusion assays indicated that CS was an important component to facilitate antibacterial activity for AuNPs. Contrarily, CS stabilization did not enhance the antibacterial efficacy of AgNPs. CS-stabilized AgNPs and AuNPs achieved biofilm inhibition of 53.21% and 79.39% for Pseudomonas aeruginosa and 48.71% and 48.16% for Staphylococcus aureus, respectively. Similarly, CS stabilization enhanced the anti-biofilm activity of AuNPs but no such effect was seen for AgNPs. In conclusion, CS-stabilized AgNPs and AuNPs possess both antimicrobial and anti-biofilm activities. However, CS acted differently when combined with AgNPs and AuNPs, needing further investigation and optimization to improve the antimicrobial activity of both nanoparticles.
Collapse
Affiliation(s)
- Mohammad Aqil M Fathil
- Centre for Drug Delivery Technology, Faculty of Pharmacy, Universiti Kebangsaan Malaysia Jalan Raja Muda Abdul Aziz Kuala Lumpur 50300 Malaysia +60-3-26983271 +60-3-92897971
| | - Farha Yasmin Faris Taufeq
- Centre for Drug Delivery Technology, Faculty of Pharmacy, Universiti Kebangsaan Malaysia Jalan Raja Muda Abdul Aziz Kuala Lumpur 50300 Malaysia +60-3-26983271 +60-3-92897971
| | - Sundos Suleman Ismail Abdalla
- Centre for Drug Delivery Technology, Faculty of Pharmacy, Universiti Kebangsaan Malaysia Jalan Raja Muda Abdul Aziz Kuala Lumpur 50300 Malaysia +60-3-26983271 +60-3-92897971
| | - Haliza Katas
- Centre for Drug Delivery Technology, Faculty of Pharmacy, Universiti Kebangsaan Malaysia Jalan Raja Muda Abdul Aziz Kuala Lumpur 50300 Malaysia +60-3-26983271 +60-3-92897971
| |
Collapse
|
21
|
Sionov RV, Steinberg D. Targeting the Holy Triangle of Quorum Sensing, Biofilm Formation, and Antibiotic Resistance in Pathogenic Bacteria. Microorganisms 2022; 10:1239. [PMID: 35744757 PMCID: PMC9228545 DOI: 10.3390/microorganisms10061239] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 06/12/2022] [Accepted: 06/14/2022] [Indexed: 12/12/2022] Open
Abstract
Chronic and recurrent bacterial infections are frequently associated with the formation of biofilms on biotic or abiotic materials that are composed of mono- or multi-species cultures of bacteria/fungi embedded in an extracellular matrix produced by the microorganisms. Biofilm formation is, among others, regulated by quorum sensing (QS) which is an interbacterial communication system usually composed of two-component systems (TCSs) of secreted autoinducer compounds that activate signal transduction pathways through interaction with their respective receptors. Embedded in the biofilms, the bacteria are protected from environmental stress stimuli, and they often show reduced responses to antibiotics, making it difficult to eradicate the bacterial infection. Besides reduced penetration of antibiotics through the intricate structure of the biofilms, the sessile biofilm-embedded bacteria show reduced metabolic activity making them intrinsically less sensitive to antibiotics. Moreover, they frequently express elevated levels of efflux pumps that extrude antibiotics, thereby reducing their intracellular levels. Some efflux pumps are involved in the secretion of QS compounds and biofilm-related materials, besides being important for removing toxic substances from the bacteria. Some efflux pump inhibitors (EPIs) have been shown to both prevent biofilm formation and sensitize the bacteria to antibiotics, suggesting a relationship between these processes. Additionally, QS inhibitors or quenchers may affect antibiotic susceptibility. Thus, targeting elements that regulate QS and biofilm formation might be a promising approach to combat antibiotic-resistant biofilm-related bacterial infections.
Collapse
Affiliation(s)
- Ronit Vogt Sionov
- The Biofilm Research Laboratory, The Institute of Biomedical and Oral Research, The Faculty of Dental Medicine, Hadassah Medical School, The Hebrew University, Jerusalem 9112102, Israel;
| | | |
Collapse
|
22
|
Low Concentration of the Neutrophil Proteases Cathepsin G, Cathepsin B, Proteinase-3 and Metalloproteinase-9 Induce Biofilm Formation in Non-Biofilm-Forming Staphylococcus epidermidis Isolates. Int J Mol Sci 2022; 23:ijms23094992. [PMID: 35563384 PMCID: PMC9102557 DOI: 10.3390/ijms23094992] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/26/2022] [Accepted: 04/28/2022] [Indexed: 12/04/2022] Open
Abstract
Neutrophils play a crucial role in eliminating bacteria that invade the human body; however, cathepsin G can induce biofilm formation in a non-biofilm-forming Staphylococcus epidermidis 1457 strain, suggesting that neutrophil proteases may be involved in biofilm formation. Cathepsin G, cathepsin B, proteinase-3, and metalloproteinase-9 (MMP-9) from neutrophils were tested on the biofilm induction in commensal (skin isolated) and clinical non-biofilm-forming S. epidermidis isolates. From 81 isolates, 53 (74%) were aap+, icaA−, icaD− genotype, and without the capacity of biofilm formation under conditions of 1% glucose, 4% ethanol or 4% NaCl, but these 53 non-biofilm-forming isolates induced biofilm by the use of different neutrophil proteases. Of these, 62.3% induced biofilm with proteinase-3, 15% with cathepsin G, 10% with cathepsin B and 5% with MMP -9, where most of the protease-induced biofilm isolates were commensal strains (skin). In the biofilm formation kinetics analysis, the addition of phenylmethylsulfonyl fluoride (PMSF; a proteinase-3 inhibitor) showed that proteinase-3 participates in the cell aggregation stage of biofilm formation. A biofilm induced with proteinase-3 and DNAse-treated significantly reduced biofilm formation at an early time (initial adhesion stage of biofilm formation) compared to untreated proteinase-3-induced biofilm (p < 0.05). A catheter inoculated with a commensal (skin) non-biofilm-forming S. epidermidis isolate treated with proteinase-3 and another one without the enzyme were inserted into the back of a mouse. After 7 days of incubation period, the catheters were recovered and the number of grown bacteria was quantified, finding a higher amount of adhered proteinase-3-treated bacteria in the catheter than non-proteinase-3-treated bacteria (p < 0.05). Commensal non-biofilm-forming S. epidermidis in the presence of neutrophil cells significantly induced the biofilm formation when multiplicity of infection (MOI) 1:0.01 (neutrophil:bacteria) was used, but the addition of a cocktail of protease inhibitors impeded biofilm formation. A neutrophil:bacteria assay did not induce neutrophil extracellular traps (NETs). Our results suggest that neutrophils, in the presence of commensal non-biofilm-forming S. epidermidis, do not generate NETs formation. The effect of neutrophils is the production of proteases, and proteinase-3 releases bacterial DNA at the initial adhesion, favoring cell aggregation and subsequently leading to biofilm formation.
Collapse
|
23
|
Donadu MG, Ferrari M, Mazzarello V, Zanetti S, Kushkevych I, Rittmann SKMR, Stájer A, Baráth Z, Szabó D, Urbán E, Gajdács M. No Correlation between Biofilm-Forming Capacity and Antibiotic Resistance in Environmental Staphylococcus spp.: In Vitro Results. Pathogens 2022; 11:pathogens11040471. [PMID: 35456146 PMCID: PMC9031815 DOI: 10.3390/pathogens11040471] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/08/2022] [Accepted: 04/13/2022] [Indexed: 01/18/2023] Open
Abstract
The production of biofilms is a critical factor in facilitating the survival of Staphylococcus spp. in vivo and in protecting against various environmental noxa. The possible relationship between the antibiotic-resistant phenotype and biofilm-forming capacity has raised considerable interest. The purpose of the study was to assess the interdependence between biofilm-forming capacity and the antibiotic-resistant phenotype in 299 Staphylococcus spp. (S. aureus n = 143, non-aureus staphylococci [NAS] n = 156) of environmental origin. Antimicrobial susceptibility testing and detection of methicillin resistance (MR) was performed. The capacity of isolates to produce biofilms was assessed using Congo red agar (CRA) plates and a crystal violet microtiter-plate-based (CV-MTP) method. MR was identified in 46.9% of S. aureus and 53.8% of NAS isolates (p > 0.05), with resistance to most commonly used drugs being significantly higher in MR isolates compared to methicillin-susceptible isolates. Resistance rates were highest for clindamycin (57.9%), erythromycin (52.2%) and trimethoprim-sulfamethoxazole (51.1%), while susceptibility was retained for most last-resort drugs. Based on the CRA plates, biofilm was produced by 30.8% of S. aureus and 44.9% of NAS (p = 0.014), while based on the CV-MTP method, 51.7% of S. aureus and 62.8% of NAS were identified as strong biofilm producers, respectively (mean OD570 values: S. aureus: 0.779±0.471 vs. NAS: 1.053±0.551; p < 0.001). No significant differences in biofilm formation were observed based on MR (susceptible: 0.824 ± 0.325 vs. resistant: 0.896 ± 0.367; p = 0.101). However, pronounced differences in biofilm formation were identified based on rifampicin susceptibility (S: 0.784 ± 0.281 vs. R: 1.239 ± 0.286; p = 0.011). The mechanistic understanding of the mechanisms Staphylococcus spp. use to withstand harsh environmental and in vivo conditions is crucial to appropriately address the therapy and eradication of these pathogens.
Collapse
Affiliation(s)
- Matthew Gavino Donadu
- Hospital Pharmacy, Azienda Ospedaliero Universitaria di Sassari, 07100 Sassari, Italy;
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (V.M.); (S.Z.)
| | - Marco Ferrari
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (V.M.); (S.Z.)
- Correspondence:
| | - Vittorio Mazzarello
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (V.M.); (S.Z.)
| | - Stefania Zanetti
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (V.M.); (S.Z.)
| | - Ivan Kushkevych
- Department of Experimental Biology, Faculty of Science, Masaryk University, 62500 Brno, Czech Republic;
| | - Simon K.-M. R. Rittmann
- Archaea Physiology & Biotechnology Group, Department of Functional and Evolutionary Ecology, Universität Wien, 1090 Wien, Austria;
| | - Anette Stájer
- Department of Periodontology, Faculty of Dentistry, University of Szeged, Tisza Lajos körút 62-64, 6720 Szeged, Hungary;
| | - Zoltán Baráth
- Department of Prosthodontics, Faculty of Dentistry, University of Szeged, Tisza Lajos körút 62–64, 6720 Szeged, Hungary;
| | - Dóra Szabó
- Institute of Medical Microbiology, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, 1089 Budapest, Hungary;
| | - Edit Urbán
- Department of Medical Microbiology and Immunology, University of Pécs Medical School, Szigeti út 12, 7624 Pécs, Hungary;
| | - Márió Gajdács
- Department of Oral Biology and Experimental Dental Research, Faculty of Dentistry, University of Szeged, Tisza Lajos krt. 63, 6720 Szeged, Hungary;
| |
Collapse
|
24
|
Vats P, Kaur UJ, Rishi P. Heavy metal-induced selection and proliferation of antibiotic resistance: A review. J Appl Microbiol 2022; 132:4058-4076. [PMID: 35170159 DOI: 10.1111/jam.15492] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/28/2021] [Accepted: 02/11/2022] [Indexed: 11/28/2022]
Abstract
Antibiotic resistance is recognized as a global threat to public health. The selection and evolution of antibiotic resistance in clinical pathogens was believed to be majorly driven by the imprudent use of antibiotics. However, concerns regarding the same, through selection pressure by a multitude of other antimicrobial agents, such as heavy metals, are also growing. Heavy metal contamination co-selects antibiotic and metal resistance through numerous mechanisms, such as co-resistance and cross-resistance. Here, we have reviewed the role of heavy metals as antimicrobial resistance driving agents and the underlying concept and mechanisms of co-selection, while also highlighting the scarcity in studies explicitly inspecting the process of co-selection in clinical settings. Prospective strategies to manage heavy metal-induced antibiotic resistance have also been deliberated, underlining the need to find specific inhibitors so that alternate medicinal combinations can be added to the existing therapeutic armamentarium.
Collapse
Affiliation(s)
- Prakriti Vats
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Ujjwal Jit Kaur
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Praveen Rishi
- Department of Microbiology, Panjab University, Chandigarh, India
| |
Collapse
|
25
|
Targeting Staphylococcus aureus and its biofilms with novel antibacterial compounds produced by Lactiplantibacillus plantarum SJ33. Arch Microbiol 2021; 204:20. [DOI: 10.1007/s00203-021-02630-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/30/2021] [Accepted: 10/01/2021] [Indexed: 11/26/2022]
|
26
|
Phenotypic PIA-Dependent Biofilm Production by Clinical Non-Typeable Staphylococcus aureus Is Not Associated with the Intensity of Inflammation in Mammary Gland: A Pilot Study Using Mouse Mastitis Model. Animals (Basel) 2021; 11:ani11113047. [PMID: 34827779 PMCID: PMC8614318 DOI: 10.3390/ani11113047] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/24/2021] [Accepted: 10/15/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Staphylococcus aureus-associated human clinical infections are predominantly caused by the encapsulated strains, with non-typeable strains representing less than 25%. In contrast, 80% of the S. aureus from bovine mastitis cases are non-typeable as they do not possess the Capsular Types 1, 2, 5, and 8. In our previous studies, it was demonstrated that the extent of mammary tissue damage was associated with the strength of biofilms formed by encapsulated S. aureus strains. This study assesses the impact of biofilm formation, as a virulence factor of non-typeable Staphylococcus aureus, causing mammary tissue damage in a mouse mastitis model. The study demonstrates no association between the strength of biofilm production by non-typeable S. aureus and the mammary tissue damage. However, the mice infected with strong biofilm producing non-typeable S. aureus died 6h earlier than those infected with weak biofilm producing non-typeable S. aureus suggesting the role of biofilm in the advancement of the time of mice mortality. Abstract Non-typeable (NT) Staphylococcus aureus strains are associated with chronic bovine mastitis. This study investigates the impact of biofilm formation by clinical NT S. aureus on cytokine production and mammary tissue damage by using a mouse mastitis model. Mice infected with two different NT S. aureus strains with strong and weak biofilm forming potential demonstrated identical clinical symptoms (moderate), minimal inflammatory infiltrates, and tissue damage (level 1 histopathological changes) in the mammary glands. However, the S. aureus load in the mammary glands of mice and the level of pro-inflammatory cytokines (IL-1β, IL-6, IL-12, IL-17 and IFN-γ) in serum were significantly higher (p ≤ 0.05) in those infected with the strong biofilm forming NT S. aureus strain. The level of IL-6 in sera samples of these mice was extremely high (15,479.9 ± 532 Pg/mL). Furthermore, these mice died in 24h of post infection compared to 30 h in the weak biofilm forming NT S. aureus infected group. The study demonstrates no association between the strength of PIA (polysaccharide intercellular adhesion)-dependent biofilm production by clinical NT S. aureus and mammary gland pathology in a mouse mastitis model. However, the role of biofilm in the virulence of S. aureus advancing the time of mortality in mice warrants further investigation.
Collapse
|
27
|
Luca A, Gallazzi E, De Vecchi E, Brayda-Bruno M, Lovi A, Babbi L, Peretti GM, Bidossi A. Bacterial adhesion on spinal implants: An in vitro study of "hot spots". J Orthop Res 2021; 39:2209-2216. [PMID: 33331674 DOI: 10.1002/jor.24960] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 12/06/2020] [Accepted: 12/14/2020] [Indexed: 02/04/2023]
Abstract
Few studies evaluated bacterial colonization of spinal implants from a "topographic" point of view. This lack of knowledge could hinder the development of more effective strategies in the prevention and treatment of postoperative spinal infections. The aim of this in vitro study was the analysis of the adhesion pattern of sessile cells on conventional spinal implants, to identify "hot spots" on implants where bacterial adhesion could be favored. Clinically relevant Staphylococcus aureus, Staphylococcus epidermidis, and Pseudomonas aeruginosa isolates were grown on commercially available end product spinal implants. To identify sessile cells attached to implant surfaces, confocal laser scan microscopy was used. Different areas from the spinal instrumentations (both Ti and CoCr) were selected for biofilm quantification. Bacterial biofilm was markedly increased in the cut of the rods, both Ti and CoCr, as the uneven surface deriving from the cut might foster cell adhesion. Though not statistically significant, a difference was observed between the rod and the area of the notch, possibly as a consequence of the smoothening effect deriving from the bending of the rod. Finally, the amount of biofilm produced on cobalt-chromium surfaces was always more significant than that formed on titanium surfaces. This study highlights how bacterial adhesion through biofilm formation is favored on the surfaces of higher irregularity and that staphylococci are able to increase sessile biomass on CoCr surfaces. These preliminary results show how surface modifications on the implants may play a key role in bacterial adhesion, opening an exciting field for future research.
Collapse
Affiliation(s)
- Andrea Luca
- Spine Unit III, IRCCS Orthopedic Institute Galeazzi, Milan, Italy
| | - Enrico Gallazzi
- Spine Unit III, IRCCS Orthopedic Institute Galeazzi, Milan, Italy
| | - Elena De Vecchi
- Laboratory of Clinica Chemistry and Microbiology, IRCCS Orthopedic Institute Galeazzi, Milan, Italy
| | | | - Alessio Lovi
- Spine Unit III, IRCCS Orthopedic Institute Galeazzi, Milan, Italy
| | - Lisa Babbi
- Spine Unit III, IRCCS Orthopedic Institute Galeazzi, Milan, Italy
| | - Giuseppe Michele Peretti
- IRCCS Orthopedic Insitutute Galeazzi, Milan, Italy.,Department of Biomedical Science, University of Milan, Milan, Italy
| | - Alessandro Bidossi
- Laboratory of Clinica Chemistry and Microbiology, IRCCS Orthopedic Institute Galeazzi, Milan, Italy
| |
Collapse
|
28
|
Characterization and Cytotoxic Evaluation of Bacteriocins Possessing Antibiofilm Activity Produced by Lactobacillus plantarum SJ33. Int J Pept Res Ther 2021. [DOI: 10.1007/s10989-021-10210-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
29
|
Biofilm Formation of Multidrug-Resistant MRSA Strains Isolated from Different Types of Human Infections. Pathogens 2021; 10:pathogens10080970. [PMID: 34451434 PMCID: PMC8400568 DOI: 10.3390/pathogens10080970] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 12/14/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is one of the main pathogens causing chronic infections, mainly due to its capacity to form biofilms. However, the mechanisms underlying the biofilm formation of MRSA strains from different types of human infections are not fully understood. MRSA strains isolated from distinct human infections were characterized aiming to determine their biofilm-forming capacity, the biofilm resistance to conventional antibiotics and the prevalence of biofilm-related genes, including, icaA, icaB, icaC, icaD, fnbA, fnbB, clfA, clfB, cna, eno, ebpS, fib and bbp. Eighty-three clinical MRSA strains recovered from bacteremia episodes, osteomyelitis and diabetic foot ulcers were used. The biofilm-forming capacity was evaluated by the microtiter biofilm assay and the biofilm structure was analyzed via confocal scanning laser microscopy. The antimicrobial susceptibility of 24-h-old biofilms was assessed against three antibiotics and the biomass reduction was measured. The metabolic activity of biofilms was evaluated by the XTT assay. The presence of biofilm-related genes was investigated by whole-genome sequencing and by PCR. Despite different intensities, all strains showed the capacity to form biofilms. Most strains had also a large number of biofilm-related genes. However, strains isolated from osteomyelitis showed a lower capacity to form biofilms and also a lower prevalence of biofilm-associated genes. There was a significant reduction in the biofilm biomass of some strains tested against antibiotics. Our results provide important information on the biofilm-forming capacity of clinical MRSA strains, which may be essential to understand the influence of different types of infections on biofilm production and chronic infections.
Collapse
|
30
|
Lawal OU, Barata M, Fraqueza MJ, Worning P, Bartels MD, Goncalves L, Paixão P, Goncalves E, Toscano C, Empel J, Urbaś M, Domiìnguez MA, Westh H, de Lencastre H, Miragaia M. Staphylococcus saprophyticus From Clinical and Environmental Origins Have Distinct Biofilm Composition. Front Microbiol 2021; 12:663768. [PMID: 34163443 PMCID: PMC8216562 DOI: 10.3389/fmicb.2021.663768] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 04/27/2021] [Indexed: 11/13/2022] Open
Abstract
Biofilm formation has been shown to be critical to the success of uropathogens. Although Staphylococcus saprophyticus is a common cause of urinary tract infections, its biofilm production capacity, composition, genetic basis, and origin are poorly understood. We investigated biofilm formation in a large and diverse collection of S. saprophyticus (n = 422). Biofilm matrix composition was assessed in representative strains (n = 63) belonging to two main S. saprophyticus lineages (G and S) recovered from human infection, colonization, and food-related environment using biofilm detachment approach. To identify factors that could be associated with biofilm formation and structure variation, we used a pangenome-wide association study approach. Almost all the isolates (91%; n = 384/422) produced biofilm. Among the 63 representative strains, we identified eight biofilm matrix phenotypes, but the most common were composed of protein or protein-extracellular DNA (eDNA)-polysaccharides (38%, 24/63 each). Biofilms containing protein-eDNA-polysaccharides were linked to lineage G and environmental isolates, whereas protein-based biofilms were produced by lineage S and infection isolates (p < 0.05). Putative biofilm-associated genes, namely, aas, atl, ebpS, uafA, sasF, sasD, sdrH, splE, sdrE, sdrC, sraP, and ica genes, were found with different frequencies (3-100%), but there was no correlation between their presence and biofilm production or matrix types. Notably, icaC_1 was ubiquitous in the collection, while icaR was lineage G-associated, and only four strains carried a complete ica gene cluster (icaADBCR) except one that was without icaR. We provided evidence, using a comparative genomic approach, that the complete icaADBCR cluster was acquired multiple times by S. saprophyticus and originated from other coagulase-negative staphylococci. Overall, the composition of S. saprophyticus biofilms was distinct in environmental and clinical isolates, suggesting that modulation of biofilm structure could be a key step in the pathogenicity of these bacteria. Moreover, biofilm production in S. saprophyticus is ica-independent, and the complete icaADBCR was acquired from other staphylococci.
Collapse
Affiliation(s)
- Opeyemi U. Lawal
- Laboratory of Bacterial Evolution and Molecular Epidemiology, Instituto de Tecnologia Química e Biológica António Xavier, Universidade NOVA de Lisboa, Oeiras, Portugal
- Laboratory of Molecular Genetics, Instituto de Tecnologia Química e Biológica António Xavier, Universidade NOVA de Lisboa, Oeiras, Portugal
| | - Marta Barata
- Laboratory of Bacterial Evolution and Molecular Epidemiology, Instituto de Tecnologia Química e Biológica António Xavier, Universidade NOVA de Lisboa, Oeiras, Portugal
| | - Maria J. Fraqueza
- Centre for Interdisciplinary Research in Animal Health, Faculdade de Medicina Veterinária, Universidade de Lisboa, Lisbon, Portugal
| | - Peder Worning
- Department of Clinical Microbiology, Hvidovre University Hospital, Hvidovre, Denmark
| | - Mette D. Bartels
- Department of Clinical Microbiology, Hvidovre University Hospital, Hvidovre, Denmark
| | | | | | | | | | - Joanna Empel
- Department of Epidemiology and Clinical Microbiology, Narodowy Instytut Leków, Warszawa, Poland
| | - Malgorzata Urbaś
- Department of Epidemiology and Clinical Microbiology, Narodowy Instytut Leków, Warszawa, Poland
| | | | - Henrik Westh
- Department of Clinical Microbiology, Hvidovre University Hospital, Hvidovre, Denmark
- Institute of Clinical Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hermínia de Lencastre
- Laboratory of Molecular Genetics, Instituto de Tecnologia Química e Biológica António Xavier, Universidade NOVA de Lisboa, Oeiras, Portugal
- The Laboratory of Microbiology and Infectious Diseases, The Rockefeller University, New York, NY, United States
| | - Maria Miragaia
- Laboratory of Bacterial Evolution and Molecular Epidemiology, Instituto de Tecnologia Química e Biológica António Xavier, Universidade NOVA de Lisboa, Oeiras, Portugal
| |
Collapse
|
31
|
Di Bonaventura G, Pompilio A. In Vitro Antimicrobial Susceptibility Testing of Biofilm-Growing Bacteria: Current and Emerging Methods. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1369:33-51. [PMID: 33963526 DOI: 10.1007/5584_2021_641] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The antibiotic susceptibility of bacterial pathogens is typically determined based on planktonic cells, as recommended by several international guidelines. However, most of chronic infections - such as those established in wounds, cystic fibrosis lung, and onto indwelling devices - are associated to the formation of biofilms, communities of clustered bacteria attached onto a surface, abiotic or biotic, and embedded in an extracellular matrix produced by the bacteria and complexed with molecules from the host. Sessile microorganisms show significantly increased tolerance/resistance to antibiotics compared with planktonic counterparts. Consequently, antibiotic concentrations used in standard antimicrobial susceptibility tests, although effective against planktonic bacteria in vitro, are not predictive of the concentrations required to eradicate biofilm-related infections, thus leading to treatment failure, chronicization and removal of material in patients with indwelling medical devices.Meeting the need for the in vitro evaluation of biofilm susceptibility to antibiotics, here we reviewed several methods proposed in literature highlighting their advantages and limitations to guide scientists towards an appropriate choice.
Collapse
Affiliation(s)
- Giovanni Di Bonaventura
- Department of Medical, Oral and Biotechnological Sciences, and Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy. .,Laboratory of Clinical Microbiology, Chieti, Italy.
| | - Arianna Pompilio
- Department of Medical, Oral and Biotechnological Sciences, and Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy.,Laboratory of Clinical Microbiology, Chieti, Italy
| |
Collapse
|
32
|
Regulatory mechanisms of sub-inhibitory levels antibiotics agent in bacterial virulence. Appl Microbiol Biotechnol 2021; 105:3495-3505. [PMID: 33893838 DOI: 10.1007/s00253-021-11291-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/01/2021] [Accepted: 04/11/2021] [Indexed: 01/13/2023]
Abstract
Antibiotics play a key role in the prevention and treatment of bacterial diseases for human and animals. The widespread use of antibiotics results in bacterial exposure to the concentrations that are lower than the MIC (that is, sub-inhibitory concentration (sub-MIC)) in the environment, humans, and livestock, which can lead to antibiotic resistance. In this review, we focus on the impact of sub-MIC antibiotics in bacterial virulence. This paper summarized the known relationships between sub-MIC antibiotics in the environment and bacterial virulence. Together, considering the impact of sub-MIC antibiotics and their alternative products in the virulence of bacteria, it is helpful to the rational use of antibiotics and the development of antibiotic alternative products to provide new insights.Key points• Sub-MIC level antibiotics exist in the environment, humans, and livestock.• The review includes mechanisms of sub-MIC antibiotics in bacterial virulence.• New antibacterial strategies and agents are being a new way to weaken virulence. Graphical Abstract.
Collapse
|
33
|
Senobar Tahaei SA, Stájer A, Barrak I, Ostorházi E, Szabó D, Gajdács M. Correlation Between Biofilm-Formation and the Antibiotic Resistant Phenotype in Staphylococcus aureus Isolates: A Laboratory-Based Study in Hungary and a Review of the Literature. Infect Drug Resist 2021. [PMID: 33790586 DOI: 10.2147/idrs303992] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023] Open
Abstract
INTRODUCTION Staphylococcus aureus (S. aureus) is an important causative pathogen in human infections. The production of biofilms by bacteria is an important factor, leading to treatment failures. There has been significant interest in assessing the possible relationship between the multidrug-resistant (MDR) status and the biofilm-producer phenotype in bacteria. The aim of our present study was to assess the biofilm-production rates in clinical methicillin-susceptible S. aureus [MSSA] and methicillin-resistant S. aureus [MRSA] isolates from Hungarian hospitals and the correlation between resistance characteristics and their biofilm-forming capacity. METHODS A total of three hundred (n=300) S. aureus isolates (corresponding to MSSA and MRSA isolates in equal measure) were included in this study. Identification of the isolates was carried out using the VITEK 2 ID/AST system and matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS). Antimicrobial susceptibility testing was performed using the Kirby-Bauer disk diffusion method and E-tests, confirmation of MRSA status was carried out using PBP2a agglutination assay. Biofilm-production was assessed using the crystal violet (CV) tube-adherence method and the Congo red agar (CRA) plate method. RESULTS There were significant differences among MSSA and MRSA isolates regarding susceptibility-levels to commonly used antibiotics (in case of erythromycin, clindamycin and ciprofloxacin: p<0.001, gentamicin: p=0.023, sulfamethoxazole/trimethoprim: p=0.027, rifampin: p=0.037). In the CV tube adherence-assay, 37% (n=56) of MSSA and 39% (n=58) of MRSA isolates were positive for biofilm-production, while during the use of CRA plates, 41% (n=61) of MSSA and 44% (n=66) of MRSA were positive; no associations were found between methicillin-resistance and biofilm-production. On the other hand, erythromycin, clindamycin and rifampin resistance was associated with biofilm-positivity (p=0.004, p<0.001 and p<0.001, respectively). Biofilm-positive isolates were most common from catheter-associated infections. DISCUSSION Our study emphasizes the need for additional experiments to assess the role biofilms have in the pathogenesis of implant-associated and chronic S. aureus infections.
Collapse
Affiliation(s)
- Seyyed Askhan Senobar Tahaei
- Department of Pharmacodynamics and Biopharmacy, Faculty of Pharmacy, University of Szeged, Szeged, 6720, Hungary
| | - Anette Stájer
- Department of Periodontology, Faculty of Dentistry, University of Szeged, Szeged, 6720, Hungary
| | - Ibrahim Barrak
- Department of Periodontology, Faculty of Dentistry, University of Szeged, Szeged, 6720, Hungary
| | - Eszter Ostorházi
- Institute of Medical Microbiology, Faculty of Medicine, Semmelweis University, Budapest, 1089, Hungary
| | - Dóra Szabó
- Institute of Medical Microbiology, Faculty of Medicine, Semmelweis University, Budapest, 1089, Hungary
| | - Márió Gajdács
- Department of Pharmacodynamics and Biopharmacy, Faculty of Pharmacy, University of Szeged, Szeged, 6720, Hungary.,Institute of Medical Microbiology, Faculty of Medicine, Semmelweis University, Budapest, 1089, Hungary
| |
Collapse
|
34
|
Senobar Tahaei SA, Stájer A, Barrak I, Ostorházi E, Szabó D, Gajdács M. Correlation Between Biofilm-Formation and the Antibiotic Resistant Phenotype in Staphylococcus aureus Isolates: A Laboratory-Based Study in Hungary and a Review of the Literature. Infect Drug Resist 2021; 14:1155-1168. [PMID: 33790586 PMCID: PMC8001189 DOI: 10.2147/idr.s303992] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 03/11/2021] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION Staphylococcus aureus (S. aureus) is an important causative pathogen in human infections. The production of biofilms by bacteria is an important factor, leading to treatment failures. There has been significant interest in assessing the possible relationship between the multidrug-resistant (MDR) status and the biofilm-producer phenotype in bacteria. The aim of our present study was to assess the biofilm-production rates in clinical methicillin-susceptible S. aureus [MSSA] and methicillin-resistant S. aureus [MRSA] isolates from Hungarian hospitals and the correlation between resistance characteristics and their biofilm-forming capacity. METHODS A total of three hundred (n=300) S. aureus isolates (corresponding to MSSA and MRSA isolates in equal measure) were included in this study. Identification of the isolates was carried out using the VITEK 2 ID/AST system and matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS). Antimicrobial susceptibility testing was performed using the Kirby-Bauer disk diffusion method and E-tests, confirmation of MRSA status was carried out using PBP2a agglutination assay. Biofilm-production was assessed using the crystal violet (CV) tube-adherence method and the Congo red agar (CRA) plate method. RESULTS There were significant differences among MSSA and MRSA isolates regarding susceptibility-levels to commonly used antibiotics (in case of erythromycin, clindamycin and ciprofloxacin: p<0.001, gentamicin: p=0.023, sulfamethoxazole/trimethoprim: p=0.027, rifampin: p=0.037). In the CV tube adherence-assay, 37% (n=56) of MSSA and 39% (n=58) of MRSA isolates were positive for biofilm-production, while during the use of CRA plates, 41% (n=61) of MSSA and 44% (n=66) of MRSA were positive; no associations were found between methicillin-resistance and biofilm-production. On the other hand, erythromycin, clindamycin and rifampin resistance was associated with biofilm-positivity (p=0.004, p<0.001 and p<0.001, respectively). Biofilm-positive isolates were most common from catheter-associated infections. DISCUSSION Our study emphasizes the need for additional experiments to assess the role biofilms have in the pathogenesis of implant-associated and chronic S. aureus infections.
Collapse
Affiliation(s)
- Seyyed Askhan Senobar Tahaei
- Department of Pharmacodynamics and Biopharmacy, Faculty of Pharmacy, University of Szeged, Szeged, 6720, Hungary
| | - Anette Stájer
- Department of Periodontology, Faculty of Dentistry, University of Szeged, Szeged, 6720, Hungary
| | - Ibrahim Barrak
- Department of Periodontology, Faculty of Dentistry, University of Szeged, Szeged, 6720, Hungary
| | - Eszter Ostorházi
- Institute of Medical Microbiology, Faculty of Medicine, Semmelweis University, Budapest, 1089, Hungary
| | - Dóra Szabó
- Institute of Medical Microbiology, Faculty of Medicine, Semmelweis University, Budapest, 1089, Hungary
| | - Márió Gajdács
- Department of Pharmacodynamics and Biopharmacy, Faculty of Pharmacy, University of Szeged, Szeged, 6720, Hungary
- Institute of Medical Microbiology, Faculty of Medicine, Semmelweis University, Budapest, 1089, Hungary
| |
Collapse
|
35
|
Mazhar SH, Li X, Rashid A, Su J, Xu J, Brejnrod AD, Su JQ, Wu Y, Zhu YG, Zhou SG, Feng R, Rensing C. Co-selection of antibiotic resistance genes, and mobile genetic elements in the presence of heavy metals in poultry farm environments. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 755:142702. [PMID: 33049532 DOI: 10.1016/j.scitotenv.2020.142702] [Citation(s) in RCA: 141] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/26/2020] [Accepted: 09/26/2020] [Indexed: 06/11/2023]
Abstract
Environmental selection of antibiotic resistance genes (ARGs) is considered to be caused by antibiotic or metal residues, frequently used in livestock. In this study we examined three commercial poultry farms to correlate the co-occurrence patterns of antibiotic and metal residues to the presence of ARGs. We quantified 283 ARGs, 12 mobile genetic elements (MGEs), 49 targeted antibiotics, 7 heavy metals and sequenced 16S rRNA genes. The abundance and type of ARG were significantly enriched in manure while soil harbored the most diverse bacterial community. Procrustes analysis displayed significant correlations between ARGs/MGEs and the microbiome. Cadmium (Cd), arsenic (As), zinc (Zn), copper (Cu) and lead (Pb) were responsible for a majority of positive correlations to ARGs when compared to antibiotics. Integrons and transposons co-occurred with ARGs corresponding to 9 classes of antibiotics, especially Class1 integrase intI-1LC. Redundancy analysis (RDA) and Variance partitioning analysis (VPA) showed that antibiotics, metals, MGEs and bacteria explain solely 0.7%, 5.7%, 12.4%, and 21.9% of variances of ARGs in the microbial community, respectively. These results suggested that bacterial composition and horizontal gene transfer were the major factors shaping the composition of ARGs; Metals had a bigger effect on ARG profile than detected antibiotics in this study.
Collapse
Affiliation(s)
- Sohaib H Mazhar
- Institute of Environmental Microbiology, College of Agricultural Resources and Environment, Fujian Agriculture and Forestry University, Fuzhou, China; Fujian Provincial Key Laboratory of Soil and Environmental Health and Regulation, College of Resources and Environment, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Xuanji Li
- Section of Microbiology, University of Copenhagen, Denmark
| | - Azhar Rashid
- Nuclear Institute for Food and Agriculture (PAEC), Tarnab, Peshawar 25000, Pakistan
| | - JunMing Su
- Institute of Environmental Microbiology, College of Agricultural Resources and Environment, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Junqiang Xu
- Institute of Environmental Microbiology, College of Agricultural Resources and Environment, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Asker Daniel Brejnrod
- Skaggs School of Pharmacy, University of California San Diego, La Jolla, United States
| | - Jian-Qiang Su
- Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, 361021 Xiamen, China
| | - Yijian Wu
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Yong-Guan Zhu
- Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, 361021 Xiamen, China; State Key Laboratory of Urban and Regional Ecology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Shun Gui Zhou
- Fujian Provincial Key Laboratory of Soil and Environmental Health and Regulation, College of Resources and Environment, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Renwei Feng
- Institute of Environmental Microbiology, College of Agricultural Resources and Environment, Fujian Agriculture and Forestry University, Fuzhou, China.
| | - Christopher Rensing
- Institute of Environmental Microbiology, College of Agricultural Resources and Environment, Fujian Agriculture and Forestry University, Fuzhou, China; Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, 361021 Xiamen, China; Fujian Provincial Key Laboratory of Soil and Environmental Health and Regulation, College of Resources and Environment, Fujian Agriculture and Forestry University, Fuzhou, China.
| |
Collapse
|
36
|
Rachmawati D, Kuntaman K, Alimsardjono L. The Correlation between icaA and icaD Genes with Biofilm Formation Staphylococcus epidermidis In Vitro. FOLIA MEDICA INDONESIANA 2021. [DOI: 10.20473/fmi.v55i4.24388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
This study was conducted to identify the presence of icaA and icaD genes in S. epidermidis and to analyze the relationship between the presence of icaA and icaD genes with the ability of in vitro biofilm formation in S. epidermidis. S. epidermidis isolates from patients and healthy people were collected and PCR was examined to detect icaA and icaD genes. which then continued to examine the ability of biofilm formation by the method of Congo Red Agar. The results of this genotypic and phenotypic examination were then tested for correlation with statistical tests using SPSS 23.0. A total of 40 S. epidermidis isolates were collected, consisting of 20 clinical isolates and 20 isolates of normal flora. The icaA gene was positive in 5 isolates (12.5%), and 8 isolates (20%) were positive for the icaD gene, 3 isolates with icaA and icaD were both positive. One hundred percent of isolates with icaA or icaD positively formed biofilms, but there were 15 isolates (42.9%) who did not have the icaA gene but showed the ability to form biofilms, while 12 isolates (37.5%) who did not have the icaD gene also formed biofilms. Fifty percent of S. epidermidis isolates showed the ability to form biofilms at CRA. The Fisher Exact test showed a significant relationship between the icaA gene and the ability of biofilm formation (p=0.047 (p<0.05)) as well as the icaD gene (p=0.03 (p<0.05)). The icaA and icaD genes have a significant relationship to biofilm formation in S. epidermidis. There was another mechanism in the formation of biofilms that are not dependent on the ica gene.
Collapse
|
37
|
Britton SJ, Neven H, Maskell DL. Microbial Small-Talk: Does Quorum Sensing Play a Role in Beer Fermentation? JOURNAL OF THE AMERICAN SOCIETY OF BREWING CHEMISTS 2020. [DOI: 10.1080/03610470.2020.1843928] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Scott J. Britton
- Research & Development, Duvel Moortgat, Puurs-Sint-Amands, Belgium
- International Centre for Brewing and Distilling, Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, UK
| | - Hedwig Neven
- Research & Development, Duvel Moortgat, Puurs-Sint-Amands, Belgium
- Centre for Food and Microbial Technology (CLMT), Department M2S, KU Leuven, Leuven, Belgium
| | - Dawn L. Maskell
- International Centre for Brewing and Distilling, Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, UK
| |
Collapse
|
38
|
The staphylococcal exopolysaccharide PIA - Biosynthesis and role in biofilm formation, colonization, and infection. Comput Struct Biotechnol J 2020. [PMID: 33240473 DOI: 10.1016/jcsbj202010027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023] Open
Abstract
Exopolysaccharide is a key part of the extracellular matrix that contributes to important mechanisms of bacterial pathogenicity, most notably biofilm formation and immune evasion. In the human pathogens Staphylococcus aureus and S. epidermidis, as well as in many other staphylococcal species, the only exopolysaccharide is polysaccharide intercellular adhesin (PIA), a cationic, partially deacetylated homopolymer of N-acetylglucosamine, whose biosynthetic machinery is encoded in the ica locus. PIA production is strongly dependent on environmental conditions and controlled by many regulatory systems. PIA contributes significantly to staphylococcal biofilm formation and immune evasion mechanisms, such as resistance to antimicrobial peptides and ingestion and killing by phagocytes, and presence of the ica genes is associated with infectivity. Due to its role in pathogenesis, PIA has raised considerable interest as a potential vaccine component or target.
Collapse
|
39
|
Pishchany G, Kolter R. On the possible ecological roles of antimicrobials. Mol Microbiol 2020; 113:580-587. [PMID: 31975454 DOI: 10.1111/mmi.14471] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 01/13/2020] [Accepted: 01/17/2020] [Indexed: 12/29/2022]
Abstract
The Introduction of antibiotics into the clinical use in the middle of the 20th century had a profound impact on modern medicine and human wellbeing. The contribution of these wonder molecules to public health and science is hard to overestimate. Much research has informed our understanding of antibiotic mechanisms of action and resistance at inhibitory concentrations in the lab and in the clinic. Antibiotics, however, are not a human invention as most of them are either natural products produced by soil microorganisms or semisynthetic derivatives of natural products. Because we use antibiotics to inhibit the bacterial growth, it is generally assumed that growth inhibition is also their primary ecological function in the environment. Nevertheless, multiple studies point to diverse nonlethal effects that are exhibited at lower levels of antibiotics. Here we review accumulating evidence of antibiosis and of alternative functions of antibiotics exhibited at subinhibitory concentrations. We also speculate on how these effects might alter phenotypes, fitness, and community composition of microbes in the context of the environment and suggest directions for future research.
Collapse
Affiliation(s)
- Gleb Pishchany
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Roberto Kolter
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
40
|
Nguyen HTT, Nguyen TH, Otto M. The staphylococcal exopolysaccharide PIA - Biosynthesis and role in biofilm formation, colonization, and infection. Comput Struct Biotechnol J 2020; 18:3324-3334. [PMID: 33240473 PMCID: PMC7674160 DOI: 10.1016/j.csbj.2020.10.027] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/23/2020] [Accepted: 10/25/2020] [Indexed: 12/20/2022] Open
Abstract
PIA is a key extracellular matrix component in staphylococci and other bacteria. PIA is a cationic, partially deacetylated N-acetylglucosamine polymer. PIA has a major role in bacterial biofilms and biofilm-associated infection.
Exopolysaccharide is a key part of the extracellular matrix that contributes to important mechanisms of bacterial pathogenicity, most notably biofilm formation and immune evasion. In the human pathogens Staphylococcus aureus and S. epidermidis, as well as in many other staphylococcal species, the only exopolysaccharide is polysaccharide intercellular adhesin (PIA), a cationic, partially deacetylated homopolymer of N-acetylglucosamine, whose biosynthetic machinery is encoded in the ica locus. PIA production is strongly dependent on environmental conditions and controlled by many regulatory systems. PIA contributes significantly to staphylococcal biofilm formation and immune evasion mechanisms, such as resistance to antimicrobial peptides and ingestion and killing by phagocytes, and presence of the ica genes is associated with infectivity. Due to its role in pathogenesis, PIA has raised considerable interest as a potential vaccine component or target.
Collapse
Affiliation(s)
- Hoai T T Nguyen
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, 50 South Drive, Bethesda 20814, MD, USA.,School of Biotechnology, International University, Vietnam National University of Ho Chi Minh City, Khu Pho 6, Thu Duc, Ho Chi Minh City, Viet Nam
| | - Thuan H Nguyen
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, 50 South Drive, Bethesda 20814, MD, USA
| | - Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, 50 South Drive, Bethesda 20814, MD, USA
| |
Collapse
|
41
|
Salzer A, Keinhörster D, Kästle C, Kästle B, Wolz C. Small Alarmone Synthetases RelP and RelQ of Staphylococcus aureus Are Involved in Biofilm Formation and Maintenance Under Cell Wall Stress Conditions. Front Microbiol 2020; 11:575882. [PMID: 33072039 PMCID: PMC7533549 DOI: 10.3389/fmicb.2020.575882] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 08/25/2020] [Indexed: 12/15/2022] Open
Abstract
The stringent response is characterized by the synthesis of the alarmone (p)ppGpp. The phenotypic consequences resulting from (p)ppGpp accumulation vary among species, and for several pathogenic bacteria, it has been shown that the activation of the stringent response strongly affects biofilm formation and maintenance. In Staphylococcus aureus, (p)ppGpp can be synthesized by the RelA/SpoT homolog Rel upon amino acid deprivation or by the two small alarmone synthetases RelP and RelQ under cell wall stress. We found that relP and relQ increase biofilm formation under cell wall stress conditions induced by a subinhibitory vancomycin concentration. However, the effect of (p)ppGpp on biofilm formation is independent of the regulators CodY and Agr. Biofilms formed by the strain HG001 or its (p)ppGpp-defective mutants are mainly composed of extracellular DNA and proteins. Furthermore, the induction of the RelPQ-mediated stringent response contributes to biofilm-related antibiotic tolerance. The proposed (p)ppGpp-inhibiting peptide DJK-5 shows bactericidal and biofilm-inhibitory activity. However, a non-(p)ppGpp-producing strain is even more vulnerable to DJK-5. This strongly argues against the assumption that DJK-5 acts via (p)ppGpp inhibition. In summary, RelP and RelQ play a major role in biofilm formation and maintenance under cell wall stress conditions.
Collapse
Affiliation(s)
| | | | | | | | - Christiane Wolz
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| |
Collapse
|
42
|
Antibiotic resistance related to biofilm formation in Streptococcus suis. Appl Microbiol Biotechnol 2020; 104:8649-8660. [PMID: 32897417 DOI: 10.1007/s00253-020-10873-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 08/23/2020] [Accepted: 08/31/2020] [Indexed: 12/13/2022]
Abstract
Streptococcus suis (S. suis) is an important zoonotic agent, which seriously impacts the pig industry and human health in various countries. Biofilm formation is likely contributing to the virulence and drug resistance in S. suis. A better knowledge of biofilm formation as well as to biofilm-dependent drug resistance mechanisms in S. suis can be of great significance for the prevention and treatment of S. suis infections. This literature review updates the latest scientific data related to biofilm formation in S. suis and its impact on drug tolerance and resistance.Key points• Biofilm formation is the important reasons for drug resistance of SS infections.• The review includes the regulatory mechanism of SS biofilm formation.• The review includes the drug resistance mechanisms of SS biofilm.
Collapse
|
43
|
Gudda FO, Waigi MG, Odinga ES, Yang B, Carter L, Gao Y. Antibiotic-contaminated wastewater irrigated vegetables pose resistance selection risks to the gut microbiome. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 264:114752. [PMID: 32417582 DOI: 10.1016/j.envpol.2020.114752] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 05/03/2020] [Accepted: 05/05/2020] [Indexed: 06/11/2023]
Abstract
Wastewater reuse in food crop irrigation has led to agroecosystem pollution concerns and human health risks. However, there is limited attention on the relationship of sub-lethal antibiotic levels in vegetables and resistance selection. Most risk assessment studies show non-significant toxicity, but overlook the link between antibiotics in crops and propagation of gut microbiome resistance selection. The review highlights the risk of antibiotics in treated water used for irrigation, uptake, and accumulation in edible vegetable parts. Moreover, it elucidates the risks to the adaptive resistance selection of the gut microbiome from sub-lethal antibiotic levels, as a result of dietary contaminated vegetables. Experiments have reported that bacterial resistance selection is possible at concentrations that are several hundred-folds lower than lethal effect levels on susceptible cells. Consequently, mutants selected at low antibiotic levels, such as those from vegetables, are fitter and more resistant compared to those selected at high concentrations. Necessary standardization, such as the development of minimum acceptable antibiotic limits allowable in food crop irrigation water, with a focus on minimum selection concentration, and not only toxicity, has been proposed. Wastewater irrigation offers environmental benefits and can contribute to food security, but it has non-addressed risks. Research gaps, future perspectives, and frameworks of mitigating the potential risks are discussed.
Collapse
Affiliation(s)
- Fredrick Owino Gudda
- Institute of Organic Contaminant Control and Soil Remediation, College of Resources and Environmental Sciences, Nanjing Agricultural University, Nanjing, 210095, China; Faculty of Environment and Resource Development, Department of Environmental Sciences, Egerton University, Box 536, Egerton, 20115, Kenya
| | - Michael Gatheru Waigi
- Institute of Organic Contaminant Control and Soil Remediation, College of Resources and Environmental Sciences, Nanjing Agricultural University, Nanjing, 210095, China
| | - Emmanuel Stephen Odinga
- Institute of Organic Contaminant Control and Soil Remediation, College of Resources and Environmental Sciences, Nanjing Agricultural University, Nanjing, 210095, China
| | - Bing Yang
- Institute of Organic Contaminant Control and Soil Remediation, College of Resources and Environmental Sciences, Nanjing Agricultural University, Nanjing, 210095, China
| | - Laura Carter
- School of Geography, University of Leeds, Leeds, LS2 9JT, UK
| | - Yanzheng Gao
- Institute of Organic Contaminant Control and Soil Remediation, College of Resources and Environmental Sciences, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
44
|
Goneau LW, Delport J, Langlois L, Poutanen SM, Razvi H, Reid G, Burton JP. Issues beyond resistance: inadequate antibiotic therapy and bacterial hypervirulence. FEMS MICROBES 2020; 1:xtaa004. [PMID: 37333955 PMCID: PMC10117437 DOI: 10.1093/femsmc/xtaa004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 10/15/2020] [Indexed: 10/15/2023] Open
Abstract
The administration of antibiotics while critical for treatment, can be accompanied by potentially severe complications. These include toxicities associated with the drugs themselves, the selection of resistant organisms and depletion of endogenous host microbiota. In addition, antibiotics may be associated with less well-recognized complications arising through changes in the pathogens themselves. Growing evidence suggests that organisms exposed to antibiotics can respond by altering the expression of toxins, invasins and adhesins, as well as biofilm, resistance and persistence factors. The clinical significance of these changes continues to be explored; however, it is possible that treatment with antibiotics may inadvertently precipitate a worsening of the clinical course of disease. Efforts are needed to adjust or augment antibiotic therapy to prevent the transition of pathogens to hypervirulent states. Better understanding the role of antibiotic-microbe interactions and how these can influence disease course is critical given the implications on prescription guidelines and antimicrobial stewardship policies.
Collapse
Affiliation(s)
- Lee W Goneau
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
- Lawson Health Research Institute, 268 Grosvenor St, London, Ontario, N6A 4V2 Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto,1 King's College Cir, Toronto, ON M5S 1A8 Ontario, Canada
| | - Johannes Delport
- Department of Pathology, London Health Sciences Center - Victoria Hospital, 800 Commissioners Rd E, London, Ontario, Canada N6A 5W9
| | - Luana Langlois
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Susan M Poutanen
- Department of Laboratory Medicine and Pathobiology, University of Toronto,1 King's College Cir, Toronto, ON M5S 1A8 Ontario, Canada
- Department of Medicine, University of Toronto, 1 King's College Cir, Toronto, ON M5S 1A8 Toronto, Ontario, Canada
- Department of Microbiology, University Health Network and Sinai Health, 190 Elizabeth St. Toronto, ON M5G 2C4, Ontario, Canada
| | - Hassan Razvi
- Lawson Health Research Institute, 268 Grosvenor St, London, Ontario, N6A 4V2 Canada
- Division of Urology, Department of Surgery, Western University, 1151 Richmond St, London, Ontario, N6A 3K7 Canada
| | - Gregor Reid
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
- Lawson Health Research Institute, 268 Grosvenor St, London, Ontario, N6A 4V2 Canada
- Division of Urology, Department of Surgery, Western University, 1151 Richmond St, London, Ontario, N6A 3K7 Canada
| | - Jeremy P Burton
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
- Lawson Health Research Institute, 268 Grosvenor St, London, Ontario, N6A 4V2 Canada
- Division of Urology, Department of Surgery, Western University, 1151 Richmond St, London, Ontario, N6A 3K7 Canada
| |
Collapse
|
45
|
Schilcher K, Horswill AR. Staphylococcal Biofilm Development: Structure, Regulation, and Treatment Strategies. Microbiol Mol Biol Rev 2020; 84:e00026-19. [PMID: 32792334 PMCID: PMC7430342 DOI: 10.1128/mmbr.00026-19] [Citation(s) in RCA: 385] [Impact Index Per Article: 77.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In many natural and clinical settings, bacteria are associated with some type of biotic or abiotic surface that enables them to form biofilms, a multicellular lifestyle with bacteria embedded in an extracellular matrix. Staphylococcus aureus and Staphylococcus epidermidis, the most frequent causes of biofilm-associated infections on indwelling medical devices, can switch between an existence as single free-floating cells and multicellular biofilms. During biofilm formation, cells first attach to a surface and then multiply to form microcolonies. They subsequently produce the extracellular matrix, a hallmark of biofilm formation, which consists of polysaccharides, proteins, and extracellular DNA. After biofilm maturation into three-dimensional structures, the biofilm community undergoes a disassembly process that leads to the dissemination of staphylococcal cells. As biofilms are dynamic and complex biological systems, staphylococci have evolved a vast network of regulatory mechanisms to modify and fine-tune biofilm development upon changes in environmental conditions. Thus, biofilm formation is used as a strategy for survival and persistence in the human host and can serve as a reservoir for spreading to new infection sites. Moreover, staphylococcal biofilms provide enhanced resilience toward antibiotics and the immune response and impose remarkable therapeutic challenges in clinics worldwide. This review provides an overview and an updated perspective on staphylococcal biofilms, describing the characteristic features of biofilm formation, the structural and functional properties of the biofilm matrix, and the most important mechanisms involved in the regulation of staphylococcal biofilm formation. Finally, we highlight promising strategies and technologies, including multitargeted or combinational therapies, to eradicate staphylococcal biofilms.
Collapse
Affiliation(s)
- Katrin Schilcher
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Alexander R Horswill
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Veterans Affairs Eastern Colorado Health Care System, Denver, Colorado, USA
| |
Collapse
|
46
|
Schilcher K, Horswill AR. Staphylococcal Biofilm Development: Structure, Regulation, and Treatment Strategies. Microbiol Mol Biol Rev 2020. [PMID: 32792334 DOI: 10.1128/mmbr.00026-19/asset/e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2023] Open
Abstract
In many natural and clinical settings, bacteria are associated with some type of biotic or abiotic surface that enables them to form biofilms, a multicellular lifestyle with bacteria embedded in an extracellular matrix. Staphylococcus aureus and Staphylococcus epidermidis, the most frequent causes of biofilm-associated infections on indwelling medical devices, can switch between an existence as single free-floating cells and multicellular biofilms. During biofilm formation, cells first attach to a surface and then multiply to form microcolonies. They subsequently produce the extracellular matrix, a hallmark of biofilm formation, which consists of polysaccharides, proteins, and extracellular DNA. After biofilm maturation into three-dimensional structures, the biofilm community undergoes a disassembly process that leads to the dissemination of staphylococcal cells. As biofilms are dynamic and complex biological systems, staphylococci have evolved a vast network of regulatory mechanisms to modify and fine-tune biofilm development upon changes in environmental conditions. Thus, biofilm formation is used as a strategy for survival and persistence in the human host and can serve as a reservoir for spreading to new infection sites. Moreover, staphylococcal biofilms provide enhanced resilience toward antibiotics and the immune response and impose remarkable therapeutic challenges in clinics worldwide. This review provides an overview and an updated perspective on staphylococcal biofilms, describing the characteristic features of biofilm formation, the structural and functional properties of the biofilm matrix, and the most important mechanisms involved in the regulation of staphylococcal biofilm formation. Finally, we highlight promising strategies and technologies, including multitargeted or combinational therapies, to eradicate staphylococcal biofilms.
Collapse
Affiliation(s)
- Katrin Schilcher
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Alexander R Horswill
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Veterans Affairs Eastern Colorado Health Care System, Denver, Colorado, USA
| |
Collapse
|
47
|
Anti-staphylococcal activity of quaternized mannan from the yeast Candida albicans. Carbohydr Polym 2020; 240:116288. [PMID: 32475569 DOI: 10.1016/j.carbpol.2020.116288] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/24/2020] [Accepted: 04/10/2020] [Indexed: 12/26/2022]
Abstract
Global increase of antibiotic-resistant pathogens as well as elevated content of drug residues in the foodstuffs and the environment urgently calls for new biocompatible antimicrobial biomaterials. Yeast mannans represent readily available source of biodegradable materials for tailor-made derivatives that could be effective in biomedical applications. Here, antimicrobial properties of quaternized mannans (DSQ 0.12, 0.24, 0.30, 0.62) from Candida albicans against clinical multi-resistant strains of Staphylococcus aureus are confronted with possible cytotoxicity against human cells. As expected, both effects increase with increasing degree of quaternization. However, it is possible to define the "window", at quaternized mannan with DSQ 0.30 with good anti-microbial effectiveness and low cytotoxicity. This derivative exhibit minimum inhibitory (MIC) and minimum bactericidal (MBC) concentration from 62.5 to 250 μg/mL and demonstrate good biofilm inhibition effect. Also acceptable values were obtained in hemagglutination and hemolytic activity assays and also in cytotoxicity tests on human fibroblasts.
Collapse
|
48
|
Winters M, Arneborg N, Appels R, Howell K. Can community-based signalling behaviour in Saccharomyces cerevisiae be called quorum sensing? A critical review of the literature. FEMS Yeast Res 2020; 19:5528315. [PMID: 31271429 DOI: 10.1093/femsyr/foz046] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 07/02/2019] [Indexed: 12/15/2022] Open
Abstract
Quorum sensing is a well-described mechanism of intercellular signalling among bacteria, which involves cell-density-dependent chemical signal molecules. The concentration of these quorum-sensing molecules increases in proportion to cell density until a threshold value is exceeded, which triggers a community-wide response. In this review, we propose that intercellular signalling mechanisms can be associated with a corresponding ecological interaction type based on similarities between how the interaction affects the signal receiver and producer. Thus, we do not confine quorum sensing, a specific form of intercellular signalling, to only cooperative behaviours. Instead, we define it as cell-density-dependent responses that occur at a critical concentration of signal molecules and through a specific signalling pathway. For fungal species, the medically important yeast Candida albicans has a well-described quorum sensing system, while this system is not well described in Saccharomyces cerevisiae, which is involved in food and beverage fermentations. The more precise definition for quorum sensing proposed in this review is based on the studies suggesting that S. cerevisiae may undergo intercellular signalling through quorum sensing. Through this lens, we conclude that there is a lack of evidence to support a specific signalling mechanism and a critical signal concentration of these behaviours in S. cerevisiae, and, thus, these features require further investigation.
Collapse
Affiliation(s)
- Michela Winters
- School of Agriculture and Food, Faculty of Veterinary and Agricultural Science, University of Melbourne, Parkville 3010, Australia
| | - Nils Arneborg
- Department of Food Science, University of Copenhagen, Frederiksberg 1958, Denmark
| | - Rudi Appels
- School of Agriculture and Food, Faculty of Veterinary and Agricultural Science, University of Melbourne, Parkville 3010, Australia
| | - Kate Howell
- School of Agriculture and Food, Faculty of Veterinary and Agricultural Science, University of Melbourne, Parkville 3010, Australia
| |
Collapse
|
49
|
Effect of Achillea santolina essential oil on bacterial biofilm and its mode of action. CURRENT ISSUES IN PHARMACY AND MEDICAL SCIENCES 2020. [DOI: 10.2478/cipms-2020-0016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Abstract
Increased multidrug resistance prompted researchers to search for a new drug that has the ability to overcome antibiotic resistant pathogens. Essential oils have been used in folk medicine for centuries, therefore, they could be employed as an effective alternative to antibiotics without having secondary side effects.
The aim of the present study was to test the antibacterial and antibiofilm activity of the essential oil of Achillea santolina and to ascertain its mode of action.
Minimum Biofilm Inhibitory Concentration (MBIC) susceptibility assays were performed using a biofilm inoculator with a 96-well plate with peg led. Minimum Inhibitory Concentration (MIC) was performed in normal microtitre plates using a twofold dilution series.
Achillea santolina essential oil (ASEO) was able to overcome the resistance of all tested bacteria. The MIC values were in the range of 250-1000 µg/ml, while the MBC values were in the range of 500-2000 µg/ml. ASEO increased leakage of potassium ions from the cell membrane and increased release of cellular materials – suggesting that the cell membrane is the target and site of action of ASEO. Moreover, ASEO was able to inhibit initial adherence of methicillin-resistant Staphylococcus aureus (MRSA) (ATCC 43300) at sub-inhibitory concentrations through alterations to cell membrane.
Collapse
|
50
|
Fernández-Calderón M, Romero-Guzmán D, Ferrández-Montero A, Pérez-Giraldo C, González-Carrasco JL, Lieblich M, Benavente R, Ferrari B, González-Martín M, Gallardo-Moreno AM. Impact of PLA/Mg films degradation on surface physical properties and biofilm survival. Colloids Surf B Biointerfaces 2020; 185:110617. [DOI: 10.1016/j.colsurfb.2019.110617] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 10/25/2019] [Accepted: 10/28/2019] [Indexed: 10/25/2022]
|