1
|
Mao R, Zhao Q, Lu H, Yang N, Li Y, Teng D, Hao Y, Gu X, Wang J. The Marine Antimicrobial Peptide AOD with Intact Disulfide Bonds Has Remarkable Antibacterial and Anti-Biofilm Activity. Mar Drugs 2024; 22:463. [PMID: 39452871 PMCID: PMC11509771 DOI: 10.3390/md22100463] [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: 08/30/2024] [Revised: 10/02/2024] [Accepted: 10/05/2024] [Indexed: 10/26/2024] Open
Abstract
American Oyster Defensin (AOD) is a marine peptide that is derived from North American mussels. It has been demonstrated to exhibit potent antimicrobial activity and high safety in both in vitro and in vivo models. In this study, to facilitate synthesis, mutants of AOD with fewer disulfide bonds were designed and subjected to structural, antimicrobial, and anti-biofilm analysis. The antimicrobial activity of AOD-derived peptides decreased after reduction in the disulfide bond, and among its three derivatives, only AOD-1 inhibited very few bacteria with a MIC value of 64 μg/mL, whereas the others had no inhibitory effect on pathogenic bacteria. The findings demonstrated that full disulfide bonds are indispensable for bactericidal activity, with the α-helix playing a pivotal role in inhibiting bacterial membranes. Furthermore, the results of the ATP, ROS, membrane potential, and membrane fluidity assays demonstrated that intracellular ATP, reactive oxygen species, and membrane fluidity were all increased, while membrane potential was reduced. This indicated that AOD resulted in the impairment of membrane fluidity and induced metabolic disorders, ultimately leading to bacterial death. The inhibitory effect of AOD on the biofilm of S. epidermidis G-81 was determined through the crystal violet and confocal microscopy. The results demonstrated that AOD exhibited a notable inhibitory impact on the biofilm of S. epidermidis G-81. The minimum biofilm inhibitory concentration of AOD on S. epidermidis G-81 was 16 μg/mL, and the minimum biofilm scavenging concentration was 32 μg/mL, which exhibited superior efficacy compared to that of lincomycin. The inhibitory effect on the primary biofilm was 90.3%, and that on the mature biofilm was 82.85%, with a dose-dependent inhibition effect. Concurrently, AOD cleared intra-biofilm organisms and reduced the number of biofilm-holding bacteria by six orders of magnitude. These data indicate that disulfide bonds are essential to the structure and activity of AOD, and AOD may potentially become an effective dual-action antimicrobial and anti-biofilm agent.
Collapse
Affiliation(s)
- Ruoyu Mao
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Qingyi Zhao
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Haiqiang Lu
- Enzyme Engineering Laboratory, College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, China
| | - Na Yang
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Yuanyuan Li
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Da Teng
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Ya Hao
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Xinxi Gu
- Enzyme Engineering Laboratory, College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, China
| | - Jianhua Wang
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| |
Collapse
|
2
|
Williams J, Burton N, Dhanoa G, Sagona AP. Host-phage interactions and modeling for therapy. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 200:127-158. [PMID: 37739552 DOI: 10.1016/bs.pmbts.2023.03.010] [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: 09/24/2023]
Abstract
Phage are drivers of numerous ecological processes on the planet and have the potential to be developed into a therapy alternative to antibiotics. Phage at all points of their life cycle, from initiation of infection to their release, interact with their host in some manner. More importantly, to harness their antimicrobial potential it is vital to understand how phage interact with the eukaryotic environment in the context of applying phage for therapy. In this chapter, the various mechanisms of phage interplay with their hosts as part of their natural life cycle are discussed in depth for Gram-positive and negative bacteria. Further, the literature surrounding the various models utilized to develop phage as a therapeutic are examined, and how these models may improve our understanding of phage-host interactions and current progress in utilizing phage for therapy in the clinical environment.
Collapse
Affiliation(s)
- Joshua Williams
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - Nathan Burton
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - Gurneet Dhanoa
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - Antonia P Sagona
- School of Life Sciences, University of Warwick, Coventry, United Kingdom.
| |
Collapse
|
3
|
Lan S, Chen X, Yin C, Xie S, Wang S, Deng R, Shen Z. Antibacterial and anti-biofilm activities of Disaspidin BB against Staphylococcus epidermidis. Front Microbiol 2023; 14:999449. [PMID: 36744091 PMCID: PMC9892941 DOI: 10.3389/fmicb.2023.999449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 01/03/2023] [Indexed: 01/20/2023] Open
Abstract
Introduction Staphylococcus epidermidis infections are an important concern in worldwide, especially when associated with biofilms, and resistance of this agent to many drugs makes the situation even worse. We investigated the inhibitory effect of Disaspidin BB obtained from plant extracts and purifications on clinical S. epidermidis strains and their biofilms, and preliminarily investigated its mechanism of of its anti-biofilm activity. Methods and Results The broth dilution method was used to determine the minimum inhibitory concentrations (MIC) of Disaspidin BB on 11 clinical S. epidermidis strains (MIC value of 0.63 ~ 2.5 μg/ml). SEP-05 was found to be erythromycin-resistant (MIC value>8 μg/ml) and Disaspidin BB sensitive with an MIC value of 0.63 μg/ml. The time-kill curve assay indicated that the antibacterial activity of Disaspidin BB against SEP-05 with concentration dependence. The metabolic activity and total biomass of the drug-treated SEP-05 biofilm in each stage were significantly inhibited by the crystalline violet and XTT assay, and the scavenging effect of Disaspidin BB on SEP-05 biofilm was also confirmed by SEM observation. The results of real-time quantitative PCR showed that subinhibitory concentrations Disaspidin BB can inhibit biofilm formation by affecting the expression level of key genes (aap, atlE, icaA, luxS, recA) in SEP-05 biofilm formation. In addition, the content of polysaccharides, proteins and extracellular DNA in biofilm matrix after the intervention of Disaspidin BB was significantly reduced, and it was tentatively determined that the ability of SEP-05 biofilm formation and its stability were thus disturbed. Discussion The results show that Disaspidin BB has promising antibacterial effect on erythromycin-resistant S. epidermidis and significant scavenging effect on its biofilm, which provides a theoretical basis for the further development of BB as a new drug for the treatment of skin infections caused by S. epidermidis.
Collapse
Affiliation(s)
- Shihua Lan
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiaofeng Chen
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Chuanping Yin
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Shengjun Xie
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Shuaishuai Wang
- Guangzhou Hipower Pharmaceutical R&D Co., Ltd., Guangzhou, China
| | - Rongrong Deng
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China,*Correspondence: Rongrong Deng, ✉
| | - Zhibin Shen
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China,Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, China,Guangdong Cosmetics Engineering and Technology Research Center, Guangdong Pharmaceutical University, Guangzhou, China,Zhibin Shen, ✉
| |
Collapse
|
4
|
Skovdal SM, Jørgensen NP, Meyer RL. JMM Profile: Staphylococcus epidermidis. J Med Microbiol 2022; 71. [DOI: 10.1099/jmm.0.001597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Staphylococcus epidermidis
is the most abundant commensal bacterium of human skin. Despite protecting us from foreign invaders,
S. epidermidis
itself exploits human vulnerability when given the opportunity. Such opportunities arise when patients are immunocompromised or when biomedical implants present an opportunity to colonize the surface and form biofilms.
S. epidermidis
is one of the most frequently isolated organisms from implanted devices and from bloodstream infections. However,
S. epidermidis
infections are often recalcitrant to antibiotics because of biofilm-associated antibiotic tolerance. Furthermore, the emergence and spread of nearly pan-resistant strains is a considerable health concern. Symptoms can be subclinical, making diagnosis challenging, and treatment with antibiotics is inefficient. For now, infection prevention remains the best strategy available.
Collapse
Affiliation(s)
- Sandra Maria Skovdal
- Department of Ophthalmology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | | | - Rikke Louise Meyer
- Interdisciplinary Nanoscience Center (iNANO), Faculty of Natural Sciences, Aarhus University, Aarhus, Denmark
- Department of Biology, Faculty of Natural Sciences, Aarhus University, Aarhus, Denmark
| |
Collapse
|
5
|
Colonization and Infection of Indwelling Medical Devices by Staphylococcus aureus with an Emphasis on Orthopedic Implants. Int J Mol Sci 2022; 23:ijms23115958. [PMID: 35682632 PMCID: PMC9180976 DOI: 10.3390/ijms23115958] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 02/08/2023] Open
Abstract
The use of indwelling medical devices has constantly increased in recent years and has revolutionized the quality of life of patients affected by different diseases. However, despite the improvement of hygiene conditions in hospitals, implant-associated infections remain a common and serious complication in prosthetic surgery, mainly in the orthopedic field, where infection often leads to implant failure. Staphylococcus aureus is the most common cause of biomaterial-centered infection. Upon binding to the medical devices, these bacteria proliferate and develop dense communities encased in a protective matrix called biofilm. Biofilm formation has been proposed as occurring in several stages-(1) attachment; (2) proliferation; (3) dispersal-and involves a variety of host and staphylococcal proteinaceous and non-proteinaceous factors. Moreover, biofilm formation is strictly regulated by several control systems. Biofilms enable staphylococci to avoid antimicrobial activity and host immune response and are a source of persistent bacteremia as well as of localized tissue destruction. While considerable information is available on staphylococcal biofilm formation on medical implants and important results have been achieved on the treatment of biofilms, preclinical and clinical applications need to be further investigated. Thus, the purpose of this review is to gather current studies about the mechanism of infection of indwelling medical devices by S. aureus with a special focus on the biochemical factors involved in biofilm formation and regulation. We also provide a summary of the current therapeutic strategies to combat biomaterial-associated infections and highlight the need to further explore biofilm physiology and conduct research for innovative anti-biofilm approaches.
Collapse
|
6
|
Gomes Von Borowski R, Chat S, Schneider R, Nonin-Lecomte S, Bouaziz S, Giudice E, Rigon Zimmer A, Baggio Gnoatto SC, Macedo AJ, Gillet R. Capsicumicine, a New Bioinspired Peptide from Red Peppers Prevents Staphylococcal Biofilm In Vitro and In Vivo via a Matrix Anti-Assembly Mechanism of Action. Microbiol Spectr 2021; 9:e0047121. [PMID: 34704807 PMCID: PMC8549733 DOI: 10.1128/spectrum.00471-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 09/09/2021] [Indexed: 11/20/2022] Open
Abstract
Staphylococci are pathogenic biofilm-forming bacteria and a source of multidrug resistance and/or tolerance causing a broad spectrum of infections. These bacteria are enclosed in a matrix that allows them to colonize medical devices, such as catheters and tissues, and that protects against antibiotics and immune systems. Advances in antibiofilm strategies for targeting this matrix are therefore extremely relevant. Here, we describe the development of the Capsicum pepper bioinspired peptide "capsicumicine." By using microbiological, microscopic, and nuclear magnetic resonance (NMR) approaches, we demonstrate that capsicumicine strongly prevents methicillin-resistant Staphylococcus epidermidis biofilm via an extracellular "matrix anti-assembly" mechanism of action. The results were confirmed in vivo in a translational preclinical model that mimics medical device-related infection. Since capsicumicine is not cytotoxic, it is a promising candidate for complementary treatment of infectious diseases. IMPORTANCE Pathogenic biofilms are a global health care concern, as they can cause extensive antibiotic resistance, morbidity, mortality, and thereby substantial economic loss. So far, no effective treatments targeting the bacteria in biofilms have been developed. Plants are constantly attacked by a wide range of pathogens and have protective factors, such as peptides, to defend themselves. These peptides are common components in Capsicum baccatum (red pepper). Here, we provide insights into an antibiofilm strategy based on the development of capsicumicine, a natural peptide that strongly controls biofilm formation by Staphylococcus epidermidis, the most prevalent pathogen in device-related infections.
Collapse
Affiliation(s)
- Rafael Gomes Von Borowski
- Université de Rennes, CNRS, Institut de Génétique et de Développement de Rennes (IGDR), UMR6290, Rennes, France
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Sophie Chat
- Université de Rennes, CNRS, Institut de Génétique et de Développement de Rennes (IGDR), UMR6290, Rennes, France
| | - Rafael Schneider
- Université de Rennes, CNRS, Institut de Génétique et de Développement de Rennes (IGDR), UMR6290, Rennes, France
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Sylvie Nonin-Lecomte
- Université de Paris, CNRS, CiTCoM (Cibles Thérapeutiques et Conception de Médicaments) UMR 8038, Faculté de Pharmacie, Paris, France
| | - Serge Bouaziz
- Université de Paris, CNRS, CiTCoM (Cibles Thérapeutiques et Conception de Médicaments) UMR 8038, Faculté de Pharmacie, Paris, France
| | - Emmanuel Giudice
- Université de Rennes, CNRS, Institut de Génétique et de Développement de Rennes (IGDR), UMR6290, Rennes, France
| | - Aline Rigon Zimmer
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Simone Cristina Baggio Gnoatto
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Alexandre José Macedo
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Centro de Biotecnologia da Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Reynald Gillet
- Université de Rennes, CNRS, Institut de Génétique et de Développement de Rennes (IGDR), UMR6290, Rennes, France
| |
Collapse
|
7
|
Li T, Lu Y, Zhang H, Wang L, Beier RC, Jin Y, Wang W, Li H, Hou X. Antibacterial Activity and Membrane-Targeting Mechanism of Aloe-Emodin Against Staphylococcus epidermidis. Front Microbiol 2021; 12:621866. [PMID: 34484130 PMCID: PMC8415635 DOI: 10.3389/fmicb.2021.621866] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 07/26/2021] [Indexed: 11/17/2022] Open
Abstract
The emergence of multidrug-resistant Staphylococcus epidermidis (S. epidermidis) dwarfs the current antibiotic development and calls for the discovery of new antibacterial agents. Aloe-emodin is a plant-derived compound that holds promise to battle against these strains. This work reports the antimicrobial activity of aloe-emodin against S. epidermidis and other Gram-positive pathogenic species, manifesting minimum inhibitory concentrations (MICs) and minimum bactericidal concentration (MBCs) around 4-32 and 32-128 μg/mL, respectively. For Gram-negative bacteria tested, the MICs and MBCs of aloe-emodin were 128-256 and above 1024 μg/mL, respectively. Aloe-emodin at the MBC for 4 h eradicated 96.9% of S. epidermidis cells. Aloe-emodin treatment led to deformities in the morphology of S. epidermidis cells and the destroy of the selective permeability of the cell membranes. Analysis of the transcriptional profiles of aloe-emodin-treated cells revealed changes of genes involved in sulfur metabolism, L-lysine and peptidoglycan biosynthesis, and biofilm formation. Aloe-emodin therefore can safely control Gram-positive bacterial infections and proves to target the bacterial outer membrane.
Collapse
Affiliation(s)
- Tao Li
- Shanghai Veterinary Research Institute, CAAS, Shanghai, China
| | - Yan Lu
- Beijing Key Laboratory of Chinese Veterinary Medicine, Department of Veterinary Medicine, National Demonstration Center for Experimental Animal Education, Beijing University of Agriculture, Beijing, China
| | - Hua Zhang
- Beijing Key Laboratory of Chinese Veterinary Medicine, Department of Veterinary Medicine, National Demonstration Center for Experimental Animal Education, Beijing University of Agriculture, Beijing, China
| | - Lei Wang
- Beijing Huafukang Bioscience Co., Ltd., Beijing, China
| | - Ross C. Beier
- Food and Feed Safety Research Unit, Southern Plains Agricultural Research Center, Agricultural Research Service, United States Department of Agriculture, College Station, TX, United States
| | - Yajie Jin
- Shanghai Veterinary Research Institute, CAAS, Shanghai, China
| | - Wenjing Wang
- Shanghai Veterinary Research Institute, CAAS, Shanghai, China
| | - Huanrong Li
- Beijing Key Laboratory of Chinese Veterinary Medicine, Department of Veterinary Medicine, National Demonstration Center for Experimental Animal Education, Beijing University of Agriculture, Beijing, China
| | - Xiaolin Hou
- Beijing Key Laboratory of Chinese Veterinary Medicine, Department of Veterinary Medicine, National Demonstration Center for Experimental Animal Education, Beijing University of Agriculture, Beijing, China
| |
Collapse
|
8
|
Boero E, Mnich ME, Manetti AGO, Soldaini E, Grimaldi L, Bagnoli F. Human Three-Dimensional Models for Studying Skin Pathogens. Curr Top Microbiol Immunol 2021; 430:3-27. [PMID: 32601967 DOI: 10.1007/82_2020_219] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Skin is the most exposed surface of the human body, separating the microbe-rich external environment, from the sterile inner part. When skin is breached or its homeostasis is perturbed, bacterial, fungal and viral pathogens can cause local infections or use the skin as an entry site to spread to other organs. In the last decades, it has become clear that skin provides niches for permanent microbial colonization, and it actively interacts with microorganisms. This crosstalk promotes skin homeostasis and immune maturation, preventing expansion of harmful organisms. Skin commensals, however, are often found to be skin most prevalent and dangerous pathogens. Despite the medical interest, mechanisms of colonization and invasion for most skin pathogens are poorly understood. This limitation is due to the lack of reliable skin models. Indeed, animal models do not adequately mimic neither the anatomy nor the immune response of human skin. Human 3D skin models overcome these limitations and can provide new insights into the molecular mechanisms of microbial pathogenesis. Herein, we address the strengths and weaknesses of different types of human skin models and we review the main findings obtained using these models to study skin pathogens.
Collapse
Affiliation(s)
| | | | | | | | - Luca Grimaldi
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | | |
Collapse
|
9
|
Vashchenko AO, Voronkova YS, Kulyk EE, Snisar OS, Sidashenko OI, Voronkova OS. Influence of sugars on biofilm formation of Staphylococcus epidermidis. REGULATORY MECHANISMS IN BIOSYSTEMS 2021. [DOI: 10.15421/022143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
The problem of biofilm formation by clinical strains of opportunistic bacteria is one of the most significant for medicine, because in a state of biofilm bacteria become more resistant to environmental factors, including antibiotics, a situation that can cause failure of treatment. Among opportunistic pathogens staphylococci are of special interest. Knowledge about the peculiarities of biofilm formation of these strains, in particular the polysaccharide biosynthesis, can be used for creation of a strategy of prophylaxis of different lesions that bind with staphylococci. The effect of different concentrations of the most widespread sugars (glucose, sucrose, lactose, galactose) on the activity of biofilm formation by strains of Staphylococcus epidermidis was investigated. Strains of S. epidermidis (n = 7) were isolated from the reproductive tract of women with dysbiosis. The cultures were grown in universal synthetic media with concentration of one of the listed sugars (0.5–3.0%) during 72 h. Results were obtained colorimetrically. We studied the number of cells in biofilm and the index of biofilm formation. The largest number of cells in the biofilm was observed when the culture incubated in a medium with 2.0% of glucose (increase of 25.3 times compared to control). The amount of CFU in the control biofilm was 9.96 lg CFU/mL. The glucose concentration of 3.0% inhibited the biofilm formation: the number of cells in the biofilm was 569 times less compared to the control. The highest value of biofilm formation index was 7.2, which was 1.3 times higher than the control (5.4). In the presence of lactose and galactose in nutrient medium in concentrations from 1.0% a decrease in the number of cells and biofilm formation index were observed. The received data show that process of biofilm formation is significantly dependent on external sources of sugars, which can indicate the possibility of their use as antibiofilm drug compounds, which inhibit membrane transport of sugars in bacteria.
Collapse
|
10
|
França A, Gaio V, Lopes N, Melo LDR. Virulence Factors in Coagulase-Negative Staphylococci. Pathogens 2021; 10:170. [PMID: 33557202 PMCID: PMC7913919 DOI: 10.3390/pathogens10020170] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/29/2021] [Accepted: 01/29/2021] [Indexed: 12/13/2022] Open
Abstract
Coagulase-negative staphylococci (CoNS) have emerged as major pathogens in healthcare-associated facilities, being S. epidermidis, S. haemolyticus and, more recently, S. lugdunensis, the most clinically relevant species. Despite being less virulent than the well-studied pathogen S. aureus, the number of CoNS strains sequenced is constantly increasing and, with that, the number of virulence factors identified in those strains. In this regard, biofilm formation is considered the most important. Besides virulence factors, the presence of several antibiotic-resistance genes identified in CoNS is worrisome and makes treatment very challenging. In this review, we analyzed the different aspects involved in CoNS virulence and their impact on health and food.
Collapse
Affiliation(s)
- Angela França
- Laboratory of Research in Biofilms Rosário Oliveira, Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal; (V.G.); (N.L.)
| | | | | | - Luís D. R. Melo
- Laboratory of Research in Biofilms Rosário Oliveira, Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal; (V.G.); (N.L.)
| |
Collapse
|
11
|
Ge Z, Li X, Cao X, Wang R, Hu W, Gen L, Han S, Shang Y, Liu Y, Zhou JH. Viral adaption of staphylococcal phage: A genome-based analysis of the selective preference based on codon usage Bias. Genomics 2020; 112:4657-4665. [PMID: 32818632 DOI: 10.1016/j.ygeno.2020.08.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 07/19/2020] [Accepted: 08/11/2020] [Indexed: 12/09/2022]
Abstract
Given the high therapeutic value of the staphylococcal phage, the genome co-evolution of the phage and the host has gained great attention. Though the genome-wide AT richness in staphylococcal phages has been well-studied with nucleotide usage bias, here we proved that host factor, lifestyle and taxonomy are also important factors in understanding the phage nucleotide usages bias using information entropy formula. Such correlation is especially prominent when it comes to the synonymous codon usages of staphylococcal phages, despite the overall scattered codon usage pattern represented by principal component analysis. This strong relationship is explained by nucleotide skew which testified that the usage biases of nucleotide at different codon positions are acting on synonymous codons. Therefore, our study reveals a hidden relationship of genome evolution with host limitation and phagic phenotype, providing new insight into phage genome evolution at genetic level.
Collapse
Affiliation(s)
- Zhiyi Ge
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, Gansu, PR China
| | - Xuerui Li
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, Gansu, PR China
| | - Xiaoan Cao
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, Gansu, PR China
| | - Rui Wang
- Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, United States of America
| | - Wen Hu
- Gansu Police Vocational College, Lanzhou 730046, Gansu, PR China
| | - Ling Gen
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, Gansu, PR China
| | - Shengyi Han
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, Gansu, PR China; The College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, Gansu Province, PR China
| | - Youjun Shang
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, Gansu, PR China
| | - Yongsheng Liu
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, Gansu, PR China
| | - Jian-Hua Zhou
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, Gansu, PR China.
| |
Collapse
|
12
|
Lyu Z, Shang Y, Wang X, Wu Y, Zheng J, Liu H, Gong T, Ye L, Qu D. Monoclonal Antibodies Specific to the Extracellular Domain of Histidine Kinase YycG of Staphylococcus epidermidis Inhibit Biofilm Formation. Front Microbiol 2020; 11:1839. [PMID: 32849437 PMCID: PMC7426370 DOI: 10.3389/fmicb.2020.01839] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/14/2020] [Indexed: 01/12/2023] Open
Abstract
Staphylococcus epidermidis is frequently associated with biofilm-related infections. Biofilms drastically reduce the efficacy of conventional antibiotics and the host immune system. In S. epidermidis biofilm formation, a major role is played by the YycG/YycF two-component system, and previous findings have indicated that inhibitors targeting the cytoplasmic HATPase_c domain of YycG kinase in S. epidermidis exhibit bactericidal and biofilm-killing activities. Therefore, we hypothesized that monoclonal antibodies (mAbs) against YycG extracellular (YycGex) domain would block the signal transduction and influence the biofilm formation of S. epidermidis. In this study, we screened out two YycGex-specific mAbs showing the highest affinity for the target, mAbs 2F3 and 1H1. These mAbs inhibited S. epidermidis biofilm formation in a dose-dependent manner, and at a concentration of 160 μg/mL, mAbs 2F3 and 1H1 caused 78.3 and 93.1% biofilm reduction, respectively, relative to normal mouse IgG control. When co-cultivated with YycGex mAbs, S. epidermidis cells showed diminished initial-adherence capacity, and the antibody treatment further led to a marked decrease in the synthesis of polysaccharide intercellular adhesin and in the transcriptional level of genes encoding proteins involved in biofilm formation. Lastly, we determined that the epitopes recognized by the two YycGex mAbs are located within aa 59–70 of the YycGex domain. It indicates that the YycGex domain may be a potential candidate as a vaccine for the prevention of S. epidermidis biofilm infections.
Collapse
Affiliation(s)
- Zhihui Lyu
- Key Laboratory of Medical Molecular Virology of MOE and MOH, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yongpeng Shang
- Key Laboratory of Medical Molecular Virology of MOE and MOH, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xiaofei Wang
- Key Laboratory of Medical Molecular Virology of MOE and MOH, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yang Wu
- Key Laboratory of Medical Molecular Virology of MOE and MOH, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jinxin Zheng
- Department of Infectious Diseases and Shenzhen Key Lab for Endogenous Infection, Shenzhen Nanshan Hospital, Shenzhen University, Shenzhen, China
| | - Huayong Liu
- Key Laboratory of Medical Molecular Virology of MOE and MOH, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ting Gong
- Key Laboratory of Medical Molecular Virology of MOE and MOH, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Lina Ye
- Key Laboratory of Medical Molecular Virology of MOE and MOH, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Di Qu
- Key Laboratory of Medical Molecular Virology of MOE and MOH, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
13
|
Gravina G, Svedin P, Ardalan M, Levy O, Ek CJ, Mallard C, Lai JCY. Staphylococcus epidermidis Sensitizes Perinatal Hypoxic-Ischemic Brain Injury in Male but Not Female Mice. Front Immunol 2020; 11:516. [PMID: 32373108 PMCID: PMC7186320 DOI: 10.3389/fimmu.2020.00516] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 03/06/2020] [Indexed: 12/21/2022] Open
Abstract
Background: Staphylococcus epidermidis is the most common nosocomial infection and the predominant pathogen in late-onset sepsis in preterm infants. Infection and inflammation are linked to neurological and developmental sequelae and bacterial infections increase the vulnerability of the brain to hypoxia-ischemia (HI). We thus tested the hypothesis that S. epidermidis exacerbates HI neuropathology in neonatal mice. Methods: Male and female C57Bl/6 mice were injected intraperitoneally with sterile saline or 3.5 × 107 colony-forming units of S. epidermidis on postnatal day (PND) 4 and then subjected to HI on PND5 (24 h after injection) or PND9 (5 d after injection) by left carotid artery ligation and exposure to 10% O2. White and gray matter injury was assessed on PND14-16. In an additional group of animals, the plasma, brain, and liver were collected on PND5 or PND9 after infection to evaluate cytokine and chemokine profiles, C5a levels and C5 signaling. Results: HI induced 24 h after injection of S. epidermidis resulted in greater gray and white matter injury compared to saline injected controls in males, but not in females. Specifically, males demonstrated increased gray matter injury in the cortex and striatum, and white matter loss in the subcortical region, hippocampal fimbria and striatum. In contrast, there was no potentiation of brain injury when HI occurred 5 d after infection in either sex. In the plasma, S. epidermidis-injected mice demonstrated increased levels of pro- and anti-inflammatory cytokines and chemokines and a reduction of C5a at 24 h, but not 5 d after infection. Brain CCL2 levels were increased in both sexes 24 h after infection, but increased only in males at 5 d post infection. Conclusion: Ongoing S. epidermidis infection combined with neonatal HI increases the vulnerability of the developing brain in male but not in female mice. These sex-dependent effects were to a large extent independent of expression of systemic cytokines or brain CCL2 expression. Overall, we provide new insights into how systemic S. epidermidis infection affects the developing brain and show that the time interval between infection and HI is a critical sensitizing factor in males.
Collapse
Affiliation(s)
- Giacomo Gravina
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Pernilla Svedin
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Maryam Ardalan
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ofer Levy
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, United States.,Department of Pediatrics, Harvard Medical School, Boston, MA, United States.,Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - C Joakim Ek
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Carina Mallard
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jacqueline C Y Lai
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
14
|
Vitsos A, Tsagarousianos C, Vergos O, Stithos D, Mathioudakis D, Vitsos I, Zouni P, Kakolyri A, Meimeti E, Kyriazi M, Antoniadou I, Tentolouris N, Dallas P, Roussis V, Rallis M. Efficacy of a Ceratothoa oestroides Olive Oil Extract in Patients With Chronic Ulcers: A Pilot Study. INT J LOW EXTR WOUND 2019; 18:309-316. [DOI: 10.1177/1534734619856143] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Chronic wounds unresponsive to existing treatments constitute a serious disease burden. Factors that contribute to the pathogenesis of chronic ulcers include oxidative stress, comorbid microbial infections, and the type of immune system response. Preclinically, and in a case study, a formulation containing a Ceratothoa oestroides olive oil extract promoted wound healing. Patients with chronic venous and pressure ulcers, clinically assessed as being unresponsive to healing agents, were treated for 3 months with an ointment containing the C oestroides extract combined with antibiotic and/or antiseptic agents chosen according to the type of bacterial infection. Treatment evaluation was performed using the Bates-Jensen criteria with +WoundDesk and MOWA cell phone applications. After 3 months of treatment, C oestroides resulted in an average decrease of 36% in the Bates-Jensen score of ulcers ( P < .000), with the decrease being significant from the first month ( P < .007). The combined use of topically applied antibiotics and antiseptics efficiently controlled microbial ulcer infection and facilitated wound healing. In relation to other factors such as initial wound size, chronicity appeared to be an important prognostic factor regarding the extent of wound healing. Future clinical investigations assessing the wound healing efficacy of the C oestroides olive oil extract are warranted.
Collapse
Affiliation(s)
- Andreas Vitsos
- National and Kapodistrian University of Athens, Athens, Greece
| | | | - Orestis Vergos
- Zakynthos General Hospital Agios Dionysios, Zakynthos, Greece
| | | | | | - Ioannis Vitsos
- Zakynthos General Hospital Agios Dionysios, Zakynthos, Greece
| | - Panayota Zouni
- Zakynthos General Hospital Agios Dionysios, Zakynthos, Greece
| | - Athina Kakolyri
- Zakynthos General Hospital Agios Dionysios, Zakynthos, Greece
| | | | - Maria Kyriazi
- National and Kapodistrian University of Athens, Athens, Greece
| | - Ivi Antoniadou
- National and Kapodistrian University of Athens, Athens, Greece
- European University Cyprus, Nicosia, Cyprus
| | - Nikolaos Tentolouris
- National and Kapodistrian University of Athens, Athens, Greece
- Laiko General Hospital, Athens, Greece
| | | | | | - Michail Rallis
- National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
15
|
Cabal B, Sevillano D, Fernández-García E, Alou L, Suárez M, González N, Moya JS, Torrecillas R. Bactericidal ZnO glass-filled thermoplastic polyurethane and polydimethyl siloxane composites to inhibit biofilm-associated infections. Sci Rep 2019; 9:2762. [PMID: 30808968 PMCID: PMC6391378 DOI: 10.1038/s41598-019-39324-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 01/21/2019] [Indexed: 01/16/2023] Open
Abstract
This study investigates a novel approach to controlling biofilms of the most frequent pathogens implicated in the etiology of biomaterials-associated infections. New bactericidal filler based on a non-toxic glass, belonging to B2O3-SiO2-Al2O3-Na2O-ZnO system, was used to formulate composites of the most widely used polymers in biomedical applications [i.e. thermoplastic polyurethane (TPU) and polydimethyl siloxane (PDMS)], with varying percentage by weight of the bactericidal glass (5, 15, 25, 35, 50%). Glass-filled polymer composites show dramatically restricted bacterial colonisation and biofilm formation. They exhibit time- and dose-dependent killing, with maximal action at 5 days. The highest activity was found against S.epidermidis biofilm (99% of reduction), one of the most common cause of nosocomial infections. The tensile properties of the obtained glass-filled composites are comparable with the literature data concerning polymeric biomaterials for medical implants and devices. In addition, all the materials presented in this research, revealed an excellent biocompatibility. This was disclosed by cell viability values above 70%, none alteration on erythrocyte membrane or cell functionality in contact with materials (haemolytic index 0-2%), and absence of interferences in blood coagulation (intrinsic, extrinsic and final pathways).
Collapse
Affiliation(s)
- Belén Cabal
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Universidad de Oviedo (UO), Principado de Asturias, Avda de la Vega 4-6, 33940, El Entrego, Spain.
- Nanoker Research, Pol. Ind. Olloniego, Parcela 22A, Nave 5, 33660, Oviedo, Spain.
| | - David Sevillano
- Microbiology Unit, Medicine Department, School of Medicine, Universidad Complutense, Avda. Complutense s/n, 28040, Madrid, Spain
| | - Elisa Fernández-García
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Universidad de Oviedo (UO), Principado de Asturias, Avda de la Vega 4-6, 33940, El Entrego, Spain
| | - Luis Alou
- Microbiology Unit, Medicine Department, School of Medicine, Universidad Complutense, Avda. Complutense s/n, 28040, Madrid, Spain
| | - Marta Suárez
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Universidad de Oviedo (UO), Principado de Asturias, Avda de la Vega 4-6, 33940, El Entrego, Spain
- Nanoker Research, Pol. Ind. Olloniego, Parcela 22A, Nave 5, 33660, Oviedo, Spain
| | - Natalia González
- Microbiology Unit, Medicine Department, School of Medicine, Universidad Complutense, Avda. Complutense s/n, 28040, Madrid, Spain
| | - José S Moya
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Universidad de Oviedo (UO), Principado de Asturias, Avda de la Vega 4-6, 33940, El Entrego, Spain
| | - Ramón Torrecillas
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Universidad de Oviedo (UO), Principado de Asturias, Avda de la Vega 4-6, 33940, El Entrego, Spain
| |
Collapse
|
16
|
Abstract
Staphylococci, with the leading species Staphylococcus aureus and Staphylococcus epidermidis, are the most frequent causes of infections on indwelling medical devices. The biofilm phenotype that those bacteria adopt during device-associated infection facilitates increased resistance to antibiotics and host immune defenses. This review presents and discusses the molecular mechanisms contributing to staphylococcal biofilm development and their in-vivo importance. Furthermore, it summarizes current strategies for the development of therapeutics against staphylococcal biofilm-associated infection.
Collapse
|
17
|
Tong C, Wu Z, Zhao X, Xue H. Arginine Catabolic Mobile Elements in Livestock-Associated Methicillin-Resistant Staphylococcal Isolates From Bovine Mastitic Milk in China. Front Microbiol 2018; 9:1031. [PMID: 29867908 PMCID: PMC5964201 DOI: 10.3389/fmicb.2018.01031] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Accepted: 05/01/2018] [Indexed: 12/19/2022] Open
Abstract
The arginine catabolic mobile element (ACME) facilitates colonization of staphylococci on skin and mucous membranes by improving their tolerances to polyamines and acidic conditions. ACME is inserted in tandem with the SCCmec element and Staphylococcus epidermidis has been proposed to be a reservoir of ACME for other staphylococci. In this study, we investigated the existence of ACME in 146 staphylococcal isolates from mastitic milk and found 21 of them carried ACME. Almost half of the investigated S. epidermidis isolates contained the element. The whole genome of a S. epidermidis strain Y24 with ACME was further sequenced and the ACME-SCCmec composite island was assembled. This composite island is 81.3 kb long and consisted of 77 ORFs including a methicillin resistance gene mecA, a type II’ ACME gene cluster, a virulence gene pls and eight heavy metal tolerance genes. Wide existence of ACME in livestock-associated staphylococci from this study and a potential risk of spreading ACME among different staphylococcal species warrant close monitoring and further studies.
Collapse
Affiliation(s)
- Chao Tong
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Zhaowei Wu
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Xin Zhao
- College of Animal Science and Technology, Northwest A&F University, Yangling, China.,Department of Animal Science, McGill University, Montreal, QC, Canada
| | - Huping Xue
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| |
Collapse
|
18
|
Saporito P, Vang Mouritzen M, Løbner-Olesen A, Jenssen H. LL-37 fragments have antimicrobial activity against Staphylococcus epidermidis biofilms and wound healing potential in HaCaT cell line. J Pept Sci 2018; 24:e3080. [PMID: 29737589 DOI: 10.1002/psc.3080] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 02/16/2018] [Accepted: 03/13/2018] [Indexed: 11/07/2022]
Abstract
Staphylococcus epidermidis is a common nosocomial pathogen able to form biofilms in indwelling devices, resulting in chronic infections, which are refractory to antibiotics treatment. Staphylococcal biofilms are also associated with the delayed reepithelization and healing of chronic wounds. The human cathelicidin peptide LL-37 has been proven active against S. epidermidis biofilms in vitro and to promote wound healing. As previous studies have demonstrated that fragments of LL-37 could possess an equal antibacterial activity as the parent peptide, we tested whether shorter (12-mer) synthetic fragments of LL-37 maintained the antibiofilm and/or immune modulating activity, aiming at the identification of essential regions within the LL-37 parent sequence. Three fragments of LL-37 displayed improved activity against S. epidermidis in terms of biofilm inhibition and eradication, a reduced cytotoxicity to human keratinocytes and erythrocytes. In addition, KR-12 and VQ-12V26 enhanced wound healing potential, relative to LL37. FK-12 and KR-12 are truncated version of the cathelicidin, previously reported as valid antimicrobials, whereas VQ-12V26 is a single substituted LL-37 fragment. Remarkably, the single substitution aspartic acid to valine in position 26 caused gain of antimicrobial function in the inactive VQ-12 fragment. The combination of antibiofilm, wound healing potential, and low cytotoxicity makes KR-12 and VQ-12V26 promising therapeutic agents and lead compounds for further improvement and understanding of antibiofilm and wound healing properties.
Collapse
Affiliation(s)
- Paola Saporito
- Section for Functional Genomics and Center for Bacterial Stress Response and Persistence, Department of Biology, University of Copenhagen, Copenhagen, Denmark.,Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | | | - Anders Løbner-Olesen
- Section for Functional Genomics and Center for Bacterial Stress Response and Persistence, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Håvard Jenssen
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
| |
Collapse
|
19
|
Zheng Y, He L, Asiamah TK, Otto M. Colonization of medical devices by staphylococci. Environ Microbiol 2018; 20:3141-3153. [PMID: 29633455 DOI: 10.1111/1462-2920.14129] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/16/2018] [Accepted: 04/03/2018] [Indexed: 12/19/2022]
Abstract
The use of medical devices in modern medicine is constantly increasing. Despite the multiple precautionary strategies that are being employed in hospitals, which include increased hygiene and sterilization measures, bacterial infections on these devices still happen frequently. Staphylococci are among the major causes of medical device infection. This is mostly due to the strong capacity of those bacteria to form device-associated biofilms, which provide resistance to chemical and physical treatments as well as attacks by the host's immune system. Biofilm development is a multistep process with specific factors participating in each step. It is tightly regulated to provide a balance between biofilm expansion and detachment. Detachment from a biofilm on a medical device can lead to severe systemic infection, such as bacteremia and sepsis. While our understanding of staphylococcal biofilm formation has increased significantly and staphylococcal biofilm formation on medical devices is among the best understood biofilm-associated infections, the extensive effort put in preclinical studies with the goal to find novel therapies against staphylococcal device-associated infections has not yet resulted in efficient, applicable therapeutic options for that difficult-to-treat type of disease.
Collapse
Affiliation(s)
- Yue Zheng
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, Bethesda, MD, USA
| | - Lei He
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, Bethesda, MD, USA
| | - Titus K Asiamah
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, Bethesda, MD, USA
| | - Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
20
|
Blank JJ, Rothstein AE, Lee CJ, Malinowski MJ, Lewis BD, Ridolfi TJ, Otterson MF. Aortic Graft Infection Secondary to Iatrogenic Transcolonic Graft Malposition. Vasc Endovascular Surg 2018; 52:386-390. [PMID: 29554857 DOI: 10.1177/1538574418764037] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Aortic graft infections are a rare but devastating complication of aortic revascularization. Often infections occur due to contamination at the time of surgery. Iatrogenic misplacement of the limbs of an aortobifemoral graft is exceedingly rare, and principles of evaluation and treatment are not well defined. We report 2 cases of aortobifemoral bypass graft malposition through the colon. CASE REPORT Case 1 is a 54-year-old male who underwent aortobifemoral bypass grafting for acute limb ischemia. He had previously undergone a partial sigmoid colectomy for diverticulitis. Approximately 6 months after vascular surgery, he presented with an occult graft infection. Preoperative imaging and intraoperative findings were consistent with graft placement through the sigmoid colon. Case 2 is a 60-year-old male who underwent aortobifemoral bypass grafting due to a nonhealing wound after toe amputation. His postoperative course was complicated by pneumonia, bacteremia thought to be secondary to the pneumonia, general malaise, and persistent fevers. Approximately 10 weeks after the vascular surgery, he presented with imaging and intraoperative findings of graft malposition through the cecum. CONCLUSIONS Aortic graft infection is usually caused by surgical contamination and presents as an indolent infection. Case 1 presented as such; Case 2 presented more acutely. Both grafts were iatrogenically misplaced through the colon at the index operation. The patients underwent extra-anatomic bypass and graft explantation and subsequently recovered.
Collapse
Affiliation(s)
- Jacqueline J Blank
- 1 Division of Colorectal Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Abby E Rothstein
- 2 Division of Vascular Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Cheong Jun Lee
- 2 Division of Vascular Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Michael J Malinowski
- 2 Division of Vascular Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Brian D Lewis
- 2 Division of Vascular Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Timothy J Ridolfi
- 1 Division of Colorectal Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Mary F Otterson
- 1 Division of Colorectal Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
21
|
Complete Genome Sequence of Staphylococcus epidermidis ATCC 12228 Chromosome and Plasmids, Generated by Long-Read Sequencing. GENOME ANNOUNCEMENTS 2017; 5:5/36/e00954-17. [PMID: 28883148 PMCID: PMC5589542 DOI: 10.1128/genomea.00954-17] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Staphylococcus epidermidis ATCC 12228 was sequenced using a long-read method to generate a complete genome sequence, including some plasmid sequences. Some differences from the previously generated short-read sequence of this nonpathogenic and non-biofilm-forming strain were noted. The assembly size was 2,570,371 bp with a total G+C% content of 32.08%.
Collapse
|
22
|
Paharik AE, Kotasinska M, Both A, Hoang TMN, Büttner H, Roy P, Fey PD, Horswill AR, Rohde H. The metalloprotease SepA governs processing of accumulation-associated protein and shapes intercellular adhesive surface properties in Staphylococcus epidermidis. Mol Microbiol 2017; 103:860-874. [PMID: 27997732 DOI: 10.1111/mmi.13594] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2016] [Indexed: 12/11/2022]
Abstract
The otherwise harmless skin inhabitant Staphylococcus epidermidis is a major cause of healthcare-associated medical device infections. The species' selective pathogenic potential depends on its production of surface adherent biofilms. The Cell wall-anchored protein Aap promotes biofilm formation in S. epidermidis, independently from the polysaccharide intercellular adhesin PIA. Aap requires proteolytic cleavage to act as an intercellular adhesin. Whether and which staphylococcal proteases account for Aap processing is yet unknown. Here, evidence is provided that in PIA-negative S. epidermidis 1457Δica, the metalloprotease SepA is required for Aap-dependent S. epidermidis biofilm formation in static and dynamic biofilm models. qRT-PCR and protease activity assays demonstrated that under standard growth conditions, sepA is repressed by the global regulator SarA. Inactivation of sarA increased SepA production, and in turn augmented biofilm formation. Genetic and biochemical analyses demonstrated that SepA-related induction of biofilm accumulation resulted from enhanced Aap processing. Studies using recombinant proteins demonstrated that SepA is able to cleave the A domain of Aap at residue 335 and between the A and B domains at residue 601. This study identifies the mechanism behind Aap-mediated biofilm maturation, and also demonstrates a novel role for a secreted staphylococcal protease as a requirement for the development of a biofilm.
Collapse
Affiliation(s)
- Alexandra E Paharik
- Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Marta Kotasinska
- Institut für Medizinische Mikrobiologie, Virologie und Hygiene, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Anna Both
- Institut für Medizinische Mikrobiologie, Virologie und Hygiene, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Tra-My N Hoang
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Henning Büttner
- Institut für Medizinische Mikrobiologie, Virologie und Hygiene, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Paroma Roy
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Paul D Fey
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Alexander R Horswill
- Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Holger Rohde
- Institut für Medizinische Mikrobiologie, Virologie und Hygiene, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
23
|
Antibacterial and Antibiofilm Effect of Low Viscosity Chitosan against Staphylococcus epidermidis. Int J Microbiol 2016; 2016:9159761. [PMID: 27635144 PMCID: PMC5007360 DOI: 10.1155/2016/9159761] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 07/20/2016] [Indexed: 11/23/2022] Open
Abstract
Aim. The aim of this study was to investigate the antibacterial and antibiofilm properties of low viscosity chitosan on S. epidermidis growth and biofilm formation. Methods and Results. The antibacterial and antibiofilm properties were investigated, during both planktonic growth and biofilm formation. This was performed using different concentrations in media and by coating on polystyrene surfaces. In addition, the bactericidal effect was investigated using a modified direct contact test. The results showed that low viscosity chitosan in media had both a bacteriostatic and bactericidal effect on planktonic growth and biofilm formation of S. epidermidis in a concentration dependent manner. Polystyrene discs coated with chitosan reduced both early biofilm formation (6 h) and late biofilm formation (18 h), as confirmed by scanning electron microscopy. The modified direct contact test showed a bactericidal effect. Conclusion. This study demonstrated that low viscosity chitosan has a bacteriostatic and bactericidal activity against S. epidermidis and that the activity is dependent on the amount of chitosan added. In addition, low viscosity chitosan reduced biofilm formation both when added to media and when coated on polystyrene surfaces. Significance and Impact of Study. Low viscosity chitosan could be a contribution to new treatment approaches of biofilm-related infections of S. epidermidis.
Collapse
|
24
|
Effects of Total Alkaloids of Sophora alopecuroides on Biofilm Formation in Staphylococcus epidermidis. BIOMED RESEARCH INTERNATIONAL 2016; 2016:4020715. [PMID: 27413745 PMCID: PMC4930803 DOI: 10.1155/2016/4020715] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Revised: 05/08/2016] [Accepted: 05/17/2016] [Indexed: 11/18/2022]
Abstract
Staphylococcus epidermidis (S. epidermidis) is an opportunistic pathogen with low pathogenicity and a cause of the repeated outbreak of bovine mastitis in veterinary clinical settings. In this report, a biofilm model of S. epidermidis was generated and the minimal inhibitory concentration (MIC) and sub-MIC (SMIC) on bacterial cultures were assessed for the following agents: total alkaloids of Sophora alopecuroides (TASA), ciprofloxacin (CIP), and erythromycin (ERY). The formation and characteristic parameters of biofilm were analyzed in terms of XTT assay, silver staining, and confocal laser scanning microscope (CLSM). Results showed that a sub-MIC of TASA could inhibit 50% biofilm of bacterial activity, while 250-fold MIC of CIP and ERY MICs only inhibited 50% and 47% of biofilm formation, respectively. All three agents could inhibit the biofilm formation at an early stage, but TASA showed a better inhibitory effect on the late stage of biofilm thickening. A morphological analysis using CLSM further confirmed the destruction of biofilm by these agents. These results thus suggest that TASA has an inhibitory effect on biofilm formation of clinic S. epidermidis, which may be a potential agent warranted for further study on the treatment prevention of infection related to S. epidermidis in veterinary clinic.
Collapse
|
25
|
Gutiérrez D, Briers Y, Rodríguez-Rubio L, Martínez B, Rodríguez A, Lavigne R, García P. Role of the Pre-neck Appendage Protein (Dpo7) from Phage vB_SepiS-phiIPLA7 as an Anti-biofilm Agent in Staphylococcal Species. Front Microbiol 2015; 6:1315. [PMID: 26635776 PMCID: PMC4658415 DOI: 10.3389/fmicb.2015.01315] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 11/09/2015] [Indexed: 01/10/2023] Open
Abstract
Staphylococcus epidermidis and Staphylococcus aureus are important causative agents of hospital-acquired infections and bacteremia, likely due to their ability to form biofilms. The production of a dense exopolysaccharide (EPS) matrix enclosing the cells slows the penetration of antibiotic down, resulting in therapy failure. The EPS depolymerase (Dpo7) derived from bacteriophage vB_SepiS-phiIPLA7, was overexpressed in Escherichia coli and characterized. A dose dependent but time independent response was observed after treatment of staphylococcal 24 h-biofilms with Dpo7. Maximum removal (>90%) of biofilm-attached cells was obtained with 0.15 μM of Dpo7 in all polysaccharide producer strains but Dpo7 failed to eliminate polysaccharide-independent biofilm formed by S. aureus V329. Moreover, the pre-treatment of polystyrene surfaces with Dpo7 reduced the biofilm biomass by 53–85% in the 67% of the tested strains. This study supports the use of phage-encoded EPS depolymerases to prevent and disperse staphylococcal biofilms, thereby making bacteria more susceptible to the action of antimicrobials.
Collapse
Affiliation(s)
- Diana Gutiérrez
- Consejo Superior de Investigaciones Científicas - Instituto de Productos Lácteos de Asturias Villaviciosa, Spain
| | - Yves Briers
- Laboratory of Gene Technology, KU Leuven Heverlee, Belgium ; Laboratory of Applied Biotechnology, Ghent University Ghent, Belgium
| | - Lorena Rodríguez-Rubio
- Consejo Superior de Investigaciones Científicas - Instituto de Productos Lácteos de Asturias Villaviciosa, Spain ; Laboratory of Gene Technology, KU Leuven Heverlee, Belgium
| | - Beatriz Martínez
- Consejo Superior de Investigaciones Científicas - Instituto de Productos Lácteos de Asturias Villaviciosa, Spain
| | - Ana Rodríguez
- Consejo Superior de Investigaciones Científicas - Instituto de Productos Lácteos de Asturias Villaviciosa, Spain
| | - Rob Lavigne
- Laboratory of Gene Technology, KU Leuven Heverlee, Belgium
| | - Pilar García
- Consejo Superior de Investigaciones Científicas - Instituto de Productos Lácteos de Asturias Villaviciosa, Spain
| |
Collapse
|
26
|
Kanai H, Sato H, Takei Y. Community-acquired methicillin-resistant Staphylococcus epidermidis pyelonephritis in a child: a case report. J Med Case Rep 2014; 8:415. [PMID: 25488491 PMCID: PMC4308012 DOI: 10.1186/1752-1947-8-415] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 10/30/2014] [Indexed: 11/19/2022] Open
Abstract
Introduction Staphylococcus epidermidis is currently the most frequent pathogen of opportunistic and nosocomial infections worldwide. Most cases of Staphylococcus epidermidis infections are associated with indwelling medical devices and/or immunocompromised conditions. Community-acquired urinary tract infections are rare, particularly among pediatric populations, and clinicians often do not consider Staphylococcus epidermidis as a uropathogen. Case presentation A previously healthy Japanese boy developed pyelonephritis caused by Enterococcus faecalis at 10 months of age. Subsequently, he was diagnosed with severe bilateral vesicoureteral reflux (right side grade V, left side grade III), and was administered trimethoprim/sulfamethoxazole as the prophylaxis. At 18 months of age, he presented with fever. Gram staining of urine obtained through catheterization revealed gram-positive cocci. We suspected pyelonephritis caused by enterococci, and administered oral fluoroquinolone empirically. The fever promptly resolved, and eventually, methicillin-resistant Staphylococcus epidermidis was detected at significant levels in the urine. Thus, our final diagnosis was pyelonephritis caused by community-acquired methicillin-resistant Staphylococcus epidermidis. Conclusions Our case indicated that even immunocompetent children without a urinary catheter can develop Staphylococcus epidermidis pyelonephritis. Staphylococcus epidermidis can be underdiagnosed or misdiagnosed as sample contamination in community-acquired urinary tract infections. Therefore, when Gram staining of appropriately obtained urine samples reveals gram-positive cocci, clinicians should take into consideration not only the possibility of enterococci but also staphylococci, including Staphylococcus epidermidis, particularly in children with urinary abnormalities and/or those receiving continuous antibiotic prophylaxis.
Collapse
Affiliation(s)
- Hiroaki Kanai
- Department of Pediatrics, Suwa Central Hospital, Tamagawa 4300, Chino-city, Nagano 391-8503, Japan.
| | | | | |
Collapse
|
27
|
Accumulation-associated protein enhances Staphylococcus epidermidis biofilm formation under dynamic conditions and is required for infection in a rat catheter model. Infect Immun 2014; 83:214-26. [PMID: 25332125 DOI: 10.1128/iai.02177-14] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Biofilm formation is the primary virulence factor of Staphylococcus epidermidis. S. epidermidis biofilms preferentially form on abiotic surfaces and may contain multiple matrix components, including proteins such as accumulation-associated protein (Aap). Following proteolytic cleavage of the A domain, which has been shown to enhance binding to host cells, B domain homotypic interactions support cell accumulation and biofilm formation. To further define the contribution of Aap to biofilm formation and infection, we constructed an aap allelic replacement mutant and an icaADBC aap double mutant. When subjected to fluid shear, strains deficient in Aap production produced significantly less biofilm than Aap-positive strains. To examine the in vivo relevance of our findings, we modified our previously described rat jugular catheter model and validated the importance of immunosuppression and the presence of a foreign body to the establishment of infection. The use of our allelic replacement mutants in the model revealed a significant decrease in bacterial recovery from the catheter and the blood in the absence of Aap, regardless of the production of polysaccharide intercellular adhesin (PIA), a well-characterized, robust matrix molecule. Complementation of the aap mutant with full-length Aap (containing the A domain), but not the B domain alone, increased initial attachment to microtiter plates, as did in trans expression of the A domain in adhesion-deficient Staphylococcus carnosus. These results demonstrate Aap contributes to S. epidermidis infection, which may in part be due to A domain-mediated attachment to abiotic surfaces.
Collapse
|
28
|
Azevedo MM, Ramalho P, Silva AP, Teixeira-Santos R, Pina-Vaz C, Rodrigues AG. Polyethyleneimine and polyethyleneimine-based nanoparticles: novel bacterial and yeast biofilm inhibitors. J Med Microbiol 2014; 63:1167-1173. [DOI: 10.1099/jmm.0.069609-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Biofilms are commonly involved in medical device-related infections. The purpose of this study was to determine the antimicrobial and anti-biofilm activity of polyethyleneimine (PEI) and PEI-based nanoparticles (nanoPEI) against Staphylococcus aureus, Staphylococcus epidermidis, Acinetobacter baumannii and Candida albicans (clinical and ATCC strains), and to evaluate their effect upon biofilm formation on polyurethane (PUR)-like catheters. MICs and minimal lethal concentrations of PEI and nanoPEI were determined according to CLSI microdilution reference protocols. For PEI, the MIC value was 195.31 mg l−1 for all the bacteria and 48.83 mg l−1 for the yeast strains. For nanoPEI, the MIC value was 1250 mg l−1 for all the strains except A. baumannii, for which it was 2500 mg l−1. Biofilm formation was assessed with PUR-like catheter segments and biofilm metabolic activity was quantified by colorimetry with a tetrazolium reduction assay. Plasma membrane integrity and membrane potential were assessed by flow cytometry after staining microbial cells with a membrane-impermeable dye, propidium iodide, and a membrane-potential marker, DiBAC4(3). PEI inhibited growth of all microbial species; higher concentrations of nanoPEI were needed to inhibit growth of all species. Biofilm formation in the presence of anti-bacterial PEI activity was dose-dependent (except for S. epidermidis) and species-related. NanoPEI at 0.5×MIC and MIC significantly reduced the metabolic activity of biofilms of S. aureus, S. epidermidis and A. baumannii, whereas 2×MIC was required in order to inhibit biofilm metabolic activity.
Collapse
Affiliation(s)
- M. M. Azevedo
- Center for Research in Health Technologies and Information Systems, Faculty of Medicine, University of Porto, Portugal
- School D. Maria II, Rua da Alegria, V.N. Famalicão, Portugal
- Department of Microbiology, Faculty of Medicine, University of Porto, Portugal
| | - P. Ramalho
- Department of Microbiology, Faculty of Medicine, University of Porto, Portugal
| | - A. P. Silva
- Center for Research in Health Technologies and Information Systems, Faculty of Medicine, University of Porto, Portugal
- Department of Microbiology, Faculty of Medicine, University of Porto, Portugal
| | - R. Teixeira-Santos
- Department of Microbiology, Faculty of Medicine, University of Porto, Portugal
| | - C. Pina-Vaz
- Department of Microbiology, Hospital S. João, Porto, Portugal
- Center for Research in Health Technologies and Information Systems, Faculty of Medicine, University of Porto, Portugal
- Department of Microbiology, Faculty of Medicine, University of Porto, Portugal
| | - A. G. Rodrigues
- Burn Unit, Department of Plastic and Reconstructive Surgery, Hospital S. João, Porto
- Center for Research in Health Technologies and Information Systems, Faculty of Medicine, University of Porto, Portugal
- Department of Microbiology, Faculty of Medicine, University of Porto, Portugal
| |
Collapse
|