301
|
Marom T, Habashi N, Cohen R, Tamir SO. Role of Biofilms in Post-Tympanostomy Tube Otorrhea. EAR, NOSE & THROAT JOURNAL 2020; 99:22S-29S. [PMID: 32204627 DOI: 10.1177/0145561320914437] [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] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVE Nearly half of children who undergo tympanostomy tube (TT) insertion may experience otorrhea following surgery. We sought to review the evidence for the role of bacterial biofilms in post-tympanostomy tube otorrhea (PTTO) and the accumulated experience regarding the preventive measures for biofilm formation/adhesion on TTs. METHODS English literature search for relevant MeSH keywords was conducted in the following databases: MEDLINE (via PubMed), Ovid Medline, Google Scholar, and Clinical Evidence (BMJ Publishing) between January 1, 1995, and December 31, 2019. Subsequently, articles were reviewed and included if biofilm was evident in PTTO. RESULTS There is an increased evidence supporting the role of biofilms in PTTO. Studies on TT design and material suggest that nitinol and/or silicone TTs had a lower risk for PTTO and that biofilms appeared in specific areas, such as the perpendicular junction of the T-tubes and the round rims of the Paparella-type tubes. Biofilm-component DNAB-II protein family was present in half of children with PTTO, and targeting this protein may lead to biofilm collapse and serve as a potential strategy for PTTO treatment. Novel approaches for the prevention of biofilm-associated PTTO include changing the inherent tube composition; tube coating with antibiotics, polymers, plant extracts, or other biofilm-resistant materials; impregnation with antimicrobial compounds; and surface alterations by ion-bombardment or surface ionization, which are still under laboratory investigation. CONCLUSIONS Currently, there is no type of TT on which bacteria will not adhere. The challenges of treating PTTO indicate the need for further research in optimization of TT design, composition, and coating.
Collapse
Affiliation(s)
- Tal Marom
- Department of Otolaryngology-Head and Neck Surgery, Samson 511918Assuta Ashdod University Hospital, Faculty of Health Sciences, Ben Gurion University, Ashdod, Israel
| | - Nadeem Habashi
- Department of Otolaryngology-Head and Neck Surgery, Samson 511918Assuta Ashdod University Hospital, Faculty of Health Sciences, Ben Gurion University, Ashdod, Israel
| | - Robert Cohen
- Association Clinique et Thérapeutique Infantile du Val-de-Marne, Saint-Maur des Fossés, France.,Paris Est University, IMRB-GRC GEMINI, Créteil, France
| | - Sharon Ovnat Tamir
- Department of Otolaryngology-Head and Neck Surgery, Samson 511918Assuta Ashdod University Hospital, Faculty of Health Sciences, Ben Gurion University, Ashdod, Israel
| |
Collapse
|
302
|
Rotering H, Al Shakaki M, Welp H, Dell'Aquila AM. Preliminary Results of a New Treatment Strategy for Relapsed Left Ventricular Assist Device-Specific Infections. Ann Thorac Surg 2020; 110:1302-1307. [PMID: 32169499 DOI: 10.1016/j.athoracsur.2020.02.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 12/27/2019] [Accepted: 02/04/2020] [Indexed: 11/18/2022]
Abstract
BACKGROUND Relapsed ventricular assist device-specific infections are associated with high morbidity, mortality, and hospital costs. A new combination of cold atmospheric plasma and special dressing technique with negative pressure wound therapy with an additional underlay of carbon cloth and hypochlorite rinsing solutions has been developed and reported in this study. METHODS Between January 2016 and January 2018, 9 patients with relapsed infected driveline or pump pocket infection were treated with this new combined strategy. The primary endpoint was complete wound healing without recurrence of infection, defined as the presence at the same site within the first year after treatment. The secondary endpoint was control of infection, defined as a marked reduction of the infected area. RESULTS After a median treatment time of 3 weeks, an immediate response was observed in all patients, and complete healing was achieved in 6 patients. Five patients met the primary endpoint, and infection did not recur after a median follow-up of 17.5 (range, 12.1 to 21.8) months. One patient underwent heart transplantation 6 months after successful wound treatment (complete wound healing). The remaining 3 patients were discharged with controlled infection. After a median follow-up of 5.7 months, 1 destination therapy patient died at home, and 2 patients underwent urgent heart transplantation because of recurrence of infection caused by Pseudomonas aeruginosa. Side effects were not observed. CONCLUSIONS The new combination treatment offers a promising option for patients with ventricular assist device-relapsed infection. Despite this, further studies are warranted to confirm those encouraging preliminary results.
Collapse
Affiliation(s)
- Heinrich Rotering
- Department of Cardiac Surgery, University Hospital Münster, Münster, Germany
| | - Mosab Al Shakaki
- Department of Cardiac Surgery, University Hospital Münster, Münster, Germany
| | - Henryk Welp
- Department of Cardiac Surgery, University Hospital Münster, Münster, Germany
| | | |
Collapse
|
303
|
Wannigama DL, Hurst C, Hongsing P, Pearson L, Saethang T, Chantaravisoot N, Singkham-In U, Luk-In S, Storer RJ, Chatsuwan T. A rapid and simple method for routine determination of antibiotic sensitivity to biofilm populations of Pseudomonas aeruginosa. Ann Clin Microbiol Antimicrob 2020; 19:8. [PMID: 32169075 PMCID: PMC7071750 DOI: 10.1186/s12941-020-00350-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 02/26/2020] [Indexed: 02/07/2023] Open
Abstract
Treatment of infections by Pseudomonas aeruginosa forming biofilms after antimicrobial testing on planktonic bacteria can result in substantial failure. Therefore, we offer a robust and simple experimental platform to test the impact of antimicrobials on biofilms. Antibiotic response patterns varied uniquely within biofilm formation capacity and minimal biofilm eradication concentrations (MBECs) has a significantly better discriminatory power than minimum inhibitory concentrations (MICs) to differentiate the overall efficiency of antibiotics to eradicate biofilm. Our resazurin-based 96-well-plate platform is able to emulate bacterial responses to antibiotics under biofilm conditions in a fast, simple, and cost-effective screening method adaptable to automation, and warrants trials in the clinic.
Collapse
Affiliation(s)
- Dhammika Leshan Wannigama
- Department of Microbiology, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Chulalongkorn University, Bangkok, Thailand
- School of Medicine, Faculty of Health and Medical Sciences, The University of Western Australia, Nedlands, WA, Australia
- Antimicrobial Resistance and Stewardship Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Cameron Hurst
- Department of Statistics, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Center of Excellence in Biostatistics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Parichart Hongsing
- School of Integrative Medicine, Mae Fah Luang University, Chiang Rai, Thailand
| | - Lachlan Pearson
- Center of Excellence in Systems Biology, Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Centre for Heart Research, Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Thammakorn Saethang
- Center of Excellence in Systems Biology, Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Computer Science, Faculty of Science, Kasetsart University, Bangkok, Thailand
| | - Naphat Chantaravisoot
- Center of Excellence in Systems Biology, Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Uthaibhorn Singkham-In
- Department of Microbiology, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Chulalongkorn University, Bangkok, Thailand
- Antimicrobial Resistance and Stewardship Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Sirirat Luk-In
- Department of Microbiology, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Chulalongkorn University, Bangkok, Thailand
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
- Antimicrobial Resistance and Stewardship Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Robin James Storer
- Office of Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Tanittha Chatsuwan
- Department of Microbiology, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Chulalongkorn University, Bangkok, Thailand.
- Antimicrobial Resistance and Stewardship Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
304
|
Bacterial lipase triggers the release of antibiotics from digestible liquid crystal nanoparticles. J Control Release 2020; 319:168-182. [DOI: 10.1016/j.jconrel.2019.12.037] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 12/18/2019] [Accepted: 12/19/2019] [Indexed: 01/01/2023]
|
305
|
Henriksen K, Rørbo N, Rybtke ML, Martinet MG, Tolker-Nielsen T, Høiby N, Middelboe M, Ciofu O. P. aeruginosa flow-cell biofilms are enhanced by repeated phage treatments but can be eradicated by phage-ciprofloxacin combination. Pathog Dis 2020; 77:5368070. [PMID: 30821815 DOI: 10.1093/femspd/ftz011] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 02/28/2019] [Indexed: 02/06/2023] Open
Abstract
Phage therapy has shown promising results in the treatment of Pseudomonas aeruginosa biofilm infections in animal studies and case reports. The aim of this study was to quantify effects of phage treatments on P. aeruginosa biofilm production and structure. Confocal scanning microscopy was used to follow the interaction between a cocktail of three virulent phages and P. aeruginosa flow-cell biofilms. The role of (i) biofilm age, (ii) repeated phage treatments, (iii) alginate production and (iv) the combination with sub-MIC levels of ciprofloxacin was investigated. Single phage treatment in the early biofilm stages significantly reduced P. aeruginosa PAO1 biovolume (85%-98% reduction). Repeated phage treatments increased the biovolume from 18.25 (untreated biofilm) to 22.24 and 31.07 µm3/µm2 for biofilms treated with phages twice and thrice, respectively. Alginate protected against the phage treatment as the live biovolume remained unaffected by the phage treatment in the mucoid biofilm (20.11 µm3/µm2 in untreated and 21.74 µm3/µm2 in phage-treated biofilm) but decreased in the PAO1 biofilm from 27.35 to 0.89 µm3/µm2. We show that the combination of phages with antibiotics at sub-MIC levels caused a ∼6 log units reduction in the abundance of P. aeruginosa cells in biofilms and that phage treatment increased the size of microcolonies in flow-cell system.
Collapse
Affiliation(s)
- Karoline Henriksen
- Department of Immunology and Microbiology, Costerton Biofilm Center, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 N, Copenhagen, Denmark
| | - Nanna Rørbo
- Department of Biology, Marine Biological Section, University of Copenhagen, Strandpromenaden 5, 3000 Helsingør, Denmark
| | - Morten Levin Rybtke
- Department of Immunology and Microbiology, Costerton Biofilm Center, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 N, Copenhagen, Denmark
| | - Mark Grevsen Martinet
- Department of Immunology and Microbiology, Costerton Biofilm Center, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 N, Copenhagen, Denmark
| | - Tim Tolker-Nielsen
- Department of Immunology and Microbiology, Costerton Biofilm Center, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 N, Copenhagen, Denmark
| | - Niels Høiby
- Department of Immunology and Microbiology, Costerton Biofilm Center, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 N, Copenhagen, Denmark.,Department of Clinical Microbiology, University Hospital, Rigshospitalet, Henrik Harpestrengs Vej 4A, 2100 , Copenhagen, Denmark
| | - Mathias Middelboe
- Department of Biology, Marine Biological Section, University of Copenhagen, Strandpromenaden 5, 3000 Helsingør, Denmark
| | - Oana Ciofu
- Department of Immunology and Microbiology, Costerton Biofilm Center, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 N, Copenhagen, Denmark
| |
Collapse
|
306
|
In vivo demonstration of Pseudomonas aeruginosa biofilms as independent pharmacological microcompartments. J Cyst Fibros 2020; 19:996-1003. [PMID: 32067957 DOI: 10.1016/j.jcf.2020.01.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 01/24/2020] [Accepted: 01/25/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Pseudomonas aeruginosa is difficult to eradicate from the lungs of cystic fibrosis (CF) patients due to biofilm formation. Organs and blood are independent pharmacokinetic (PK) compartments. Previously, we showed in vitro biofilms behave as independent compartments impacting the pharmacodynamics. The present study investigated this phenomenon in vivo. METHODS Seaweed alginate beads with P. aeruginosa resembling biofilms, either freshly produced (D0) or incubated for 5 days (D5) were installed s.c in BALB/c mice. Mice (n = 64) received tobramycin 40 mg/kg s.c. and were sacrificed at 0.5, 3, 6, 8, 16 or 24 h after treatment. Untreated controls (n = 14) were sacrificed, correspondingly. Tobramycin concentrations were determined in serum, muscle tissue, lung tissue and beads. Quantitative bacteriology was determined. RESULTS The tobramycin peak concentrations in serum was 58.3 (±9.2) mg/L, in lungs 7.1 mg/L (±2.3), muscle tissue 2.8 mg/L (±0.5) all after 0.5 h and in D0 beads 19.8 mg/L (±3.5) and in D5 beads 24.8 mg/L (±4.1) (both 3 h). A 1-log killing of P. aeruginosa in beads was obtained at 8h, after which the bacterial level remained stable at 16 h and even increased in D0 beads at 24 h. Using the established diffusion retardation model the free tobramycin concentration inside the beads showed a delayed buildup of 3 h but remained lower than the MIC throughout the 24 h. CONCLUSIONS The present in vivo study based on tobramycin exposure supports that biofilms behave as independent pharmacological microcompartments. The study indicates, reducing the biofilm matrix would increase free tobramycin concentrations and improve therapeutic effects.
Collapse
|
307
|
Lüthje FL, Jensen LK, Jensen HE, Skovgaard K. The inflammatory response to bone infection - a review based on animal models and human patients. APMIS 2020; 128:275-286. [PMID: 31976582 DOI: 10.1111/apm.13027] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 01/14/2020] [Indexed: 12/17/2022]
Abstract
Bone infections are difficult to diagnose and treat, especially when a prosthetic joint replacement or implant is involved. Bone loss is a major complication of osteomyelitis, but the mechanism behind has mainly been investigated in cell cultures and has not been confirmed in human settings. Inflammation is important in initiating an appropriate immune response to invading pathogens. However, many of the signaling molecules used by the immune system can also modulate bone remodeling and contribute to bone resorption during osteomyelitis. Our current knowledge of the inflammatory response relies heavily on animal models as research based on human samples is scarce. Staphylococcus aureus is one of the most common causes of bone infections and is the pathogen of choice in animal models. The regulation of inflammatory genes during prosthetic joint infections and implant-associated osteomyelitis has only been studied in rodent models. It is important to consider the validity of an animal model when results are extrapolated to humans, and both bone composition and the immune system of pigs has been shown to be more similar to humans, than to rodents. Here in vivo studies on the inflammatory response to prosthetic joint infections and implant-associated osteomyelitis are reviewed.
Collapse
Affiliation(s)
- Freja Lea Lüthje
- Department of Veterinary and Animal Science, University of Copenhagen, Frederiksberg C, Denmark.,Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Louise Kruse Jensen
- Department of Veterinary and Animal Science, University of Copenhagen, Frederiksberg C, Denmark
| | - Henrik Elvang Jensen
- Department of Veterinary and Animal Science, University of Copenhagen, Frederiksberg C, Denmark
| | - Kerstin Skovgaard
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| |
Collapse
|
308
|
Khalid V, Schønheyder HC, Larsen LH, Nielsen PT, Kappel A, Thomsen TR, Aleksyniene R, Lorenzen J, Ørsted I, Simonsen O, Jordal PL, Rasmussen S. Multidisciplinary Diagnostic Algorithm for Evaluation of Patients Presenting with a Prosthetic Problem in the Hip or Knee: A Prospective Study. Diagnostics (Basel) 2020; 10:E98. [PMID: 32053936 PMCID: PMC7168188 DOI: 10.3390/diagnostics10020098] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 01/28/2020] [Accepted: 02/04/2020] [Indexed: 12/17/2022] Open
Abstract
The predominant indications for revision surgery after total hip (THA) or knee arthroplasty (TKA) are an aseptic failure (AF) and prosthetic joint infection (PJI). Accurate diagnosis is crucial. Therefore, we evaluated prospectively a multidisciplinary diagnostic algorithm including multi-modal radionucleid imaging (RNI) and extended microbiological diagnostics. If the surgeon suspected PJI or AF, revision surgery was performed with multiple samples obtained in parallel for special culture procedures and later molecular analyses. Alternatively, if the underlying cause was not evident, RNI was scheduled comprising 99Tc - HDP SPECT/CT, 111In-labeled white blood cells combined with 99Tc-nanocoll bone marrow SPECT/CT, and 18F-FDG PET/CT. A multidisciplinary clinical team made a recommendation on the indication for a diagnostic procedure guided by RNI images or revision surgery. A total of 156 patients with 163 arthroplasties were included. Fifty-five patients underwent RNI. In all, 118 revision surgeries were performed in 112 patients: 71 on the indication of AF and 41 revision of PJI. Thirty-four patients were concluded with chronic pain, and revision surgery refrained. The effective median follow-up period was 13 months. A structured approach offered by the algorithm was useful for the clinician in the evaluation of patients with a failing TKA or THA. Surgical revision was possibly obviated in approximately 20% of patients where an explanation or cause of failure was not found. The algorithm served as an effective tool.
Collapse
Affiliation(s)
- Vesal Khalid
- Orthopaedic Research Unit, Aalborg University Hospital, 9000 Aalborg, Denmark (A.K.); (O.S.); (S.R.)
- Department of Orthopaedic Surgery, Aalborg University Hospital, 9000 Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, 9000 Aalborg, Denmark;
| | - Henrik Carl Schønheyder
- Department of Clinical Medicine, Aalborg University, 9000 Aalborg, Denmark;
- Department of Clinical Microbiology, Aalborg University Hospital, 9000 Aalborg, Denmark;
| | - Lone Heimann Larsen
- Department of Clinical Microbiology, Aalborg University Hospital, 9000 Aalborg, Denmark;
- Center for Microbial Communities, Department of Biotechnology, Chemistry and Environmental Engineering, Aalborg University, 9000 Aalborg, Denmark;
| | - Poul Torben Nielsen
- Orthopaedic Research Unit, Aalborg University Hospital, 9000 Aalborg, Denmark (A.K.); (O.S.); (S.R.)
- Department of Orthopaedic Surgery, Aalborg University Hospital, 9000 Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, 9000 Aalborg, Denmark;
| | - Andreas Kappel
- Orthopaedic Research Unit, Aalborg University Hospital, 9000 Aalborg, Denmark (A.K.); (O.S.); (S.R.)
- Department of Orthopaedic Surgery, Aalborg University Hospital, 9000 Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, 9000 Aalborg, Denmark;
| | - Trine Rolighed Thomsen
- Center for Microbial Communities, Department of Biotechnology, Chemistry and Environmental Engineering, Aalborg University, 9000 Aalborg, Denmark;
- Danish Technological Institute, Medical Biotechnology, 8000 Aarhus C, Denmark; (J.L.); (P.L.J.)
| | - Ramune Aleksyniene
- Department of Nuclear Medicine, Aalborg University Hospital, 9000 Aalborg, Denmark;
| | - Jan Lorenzen
- Danish Technological Institute, Medical Biotechnology, 8000 Aarhus C, Denmark; (J.L.); (P.L.J.)
| | - Iben Ørsted
- Department of Infectious Disease, Aalborg University Hospital, 9000 Aalborg, Denmark;
| | - Ole Simonsen
- Orthopaedic Research Unit, Aalborg University Hospital, 9000 Aalborg, Denmark (A.K.); (O.S.); (S.R.)
- Department of Orthopaedic Surgery, Aalborg University Hospital, 9000 Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, 9000 Aalborg, Denmark;
| | - Peter Lüttge Jordal
- Danish Technological Institute, Medical Biotechnology, 8000 Aarhus C, Denmark; (J.L.); (P.L.J.)
| | - Sten Rasmussen
- Orthopaedic Research Unit, Aalborg University Hospital, 9000 Aalborg, Denmark (A.K.); (O.S.); (S.R.)
- Department of Orthopaedic Surgery, Aalborg University Hospital, 9000 Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, 9000 Aalborg, Denmark;
| |
Collapse
|
309
|
Vestby LK, Grønseth T, Simm R, Nesse LL. Bacterial Biofilm and its Role in the Pathogenesis of Disease. Antibiotics (Basel) 2020; 9:E59. [PMID: 32028684 PMCID: PMC7167820 DOI: 10.3390/antibiotics9020059] [Citation(s) in RCA: 523] [Impact Index Per Article: 104.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 01/28/2020] [Accepted: 01/29/2020] [Indexed: 12/31/2022] Open
Abstract
Recognition of the fact that bacterial biofilm may play a role in the pathogenesis of disease has led to an increased focus on identifying diseases that may be biofilm-related. Biofilm infections are typically chronic in nature, as biofilm-residing bacteria can be resilient to both the immune system, antibiotics, and other treatments. This is a comprehensive review describing biofilm diseases in the auditory, the cardiovascular, the digestive, the integumentary, the reproductive, the respiratory, and the urinary system. In most cases reviewed, the biofilms were identified through various imaging technics, in addition to other study approaches. The current knowledge on how biofilm may contribute to the pathogenesis of disease indicates a number of different mechanisms. This spans from biofilm being a mere reservoir of pathogenic bacteria, to playing a more active role, e.g., by contributing to inflammation. Observations also indicate that biofilm does not exclusively occur extracellularly, but may also be formed inside living cells. Furthermore, the presence of biofilm may contribute to development of cancer. In conclusion, this review shows that biofilm is part of many, probably most chronic infections. This is important knowledge for development of effective treatment strategies for such infections.
Collapse
Affiliation(s)
- Lene K. Vestby
- Department of Immunology and Virology, Norwegian Veterinary Institute, P.O. Box 750 Sentrum, N-0106 Oslo, Norway;
| | - Torstein Grønseth
- Department of Otolaryngology, Head and Neck Surgery, Oslo University Hospital HF, Postboks 4950 Nydalen, 0424 Oslo, Norway;
| | - Roger Simm
- Institute of Oral Biology, University of Oslo, P.O. Box 1052, Blindern, 0316 Oslo, Norway;
| | - Live L. Nesse
- Department of Food Safety and Animal Health Research, Norwegian Veterinary Institute, P.O. Box 750 Sentrum, N-0106 Oslo, Norway
| |
Collapse
|
310
|
Stryja J, Sandy-Hodgetts K, Collier M, Moser C, Ousey K, Probst S, Wilson J, Xuereb D. PREVENTION AND MANAGEMENT ACROSS HEALTH-CARE SECTORS. J Wound Care 2020; 29:S1-S72. [DOI: 10.12968/jowc.2020.29.sup2b.s1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Jan Stryja
- Vascular Surgeon, Centre of vascular and miniinvasive surgery, Hospital Podlesi, Trinec, The Czech Republic. Salvatella Ltd., Centre of non-healing wounds treatment, Podiatric outpatients’ department, Trinec, The Czech Republic
| | - Kylie Sandy-Hodgetts
- Senior Research Fellow – Senior Lecturer, Faculty of Medicine, School of Biomedical Sciences, University of Western Australia, Director, Skin Integrity Clinical Trials Unit, University of Western Australia
| | - Mark Collier
- Nurse Consultant and Associate Lecturer – Tissue Viability, Independent – formerly at the United Lincolnshire Hospitals NHS Trust, c/o Pilgrim Hospital, Sibsey Road, Boston, Lincolnshire, PE21 9Q
| | - Claus Moser
- Clinical microbiologist, Rigshospitalet, Department of Clinical Microbiology, Copenhagen, Denmark
| | - Karen Ousey
- Professor of Skin Integrity, University of Huddersfield. Institute of Skin Integrity and Infection Prevention, Huddersfield, UK
| | - Sebastian Probst
- Professor of wound care, HES-SO University of Applied Sciences and Arts Western Switzerland, Geneva, Switzerland
| | - Jennie Wilson
- Professor of Healthcare Epidemiology, University of West London, College of Nursing, Midwifery and Healthcare, London, UK
| | - Deborah Xuereb
- Senior Infection Prevention & infection Control Nurse, Mater Dei Hospital, Msida, Malta
| |
Collapse
|
311
|
Biofilms of Mycobacterium abscessus Complex Can Be Sensitized to Antibiotics by Disaggregation and Oxygenation. Antimicrob Agents Chemother 2020; 64:AAC.01212-19. [PMID: 31740557 DOI: 10.1128/aac.01212-19] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 11/03/2019] [Indexed: 12/25/2022] Open
Abstract
Pulmonary infection with the multidrug-resistant Mycobacterium abscessus complex (MABSC) is difficult to treat in individuals with cystic fibrosis (CF). MABSC grows as biofilm aggregates in CF patient lungs, which are known to have anaerobic niches. How aggregation and anoxic conditions affect antibiotic tolerance is not well understood. We sought to determine whether disaggregation and oxygen availability sensitize MABSC isolates to recommended antibiotics. We tested the susceptibilities of 33 isolates from 22 CF patients with MABSC infection and a reference strain to the following antibiotics: amikacin, azithromycin, cefoxitin, ciprofloxacin, clarithromycin, imipenem, kanamycin, linezolid, moxifloxacin, rifampin, tigecycline, and sulfamethoxazole-trimethoprim. Isolates were grown in Mueller-Hinton broth with and without the disaggregating detergent Tween 80 (5%). Time-kill curves at days 1 and 3 were generated for oxic and anoxic amikacin treatment in 4-fold dilutions ranging from 2 to 512 mg liter-1 Scanning electron microscopy was used to visualize the aggregation patterns, while confocal laser scanning microscopy and microrespirometry were used to visualize biofilm growth patterns. Disruption of MABSC aggregates increased susceptibility to amikacin, tigecycline, kanamycin, azithromycin, imipenem, cefoxitin, and clarithromycin (P < 0.05, n = 29 to 31). Oxygenation enhanced the killing of disaggregated MABSC isolates by amikacin (P < 0.05) by 1 to 6 log units when 2 to 512 mg liter-1 of amikacin was used. This study explains why current drug susceptibility testing results correlate poorly with treatment outcomes. The conditions achieved by oxic culturing of planktonic isolates in vitro do not resemble the hypoxic conditions in CF patient lungs. Biofilm disruption and increased O2 availability during antibiotic therapy may be new therapeutic strategies for chronic MABSC infection.
Collapse
|
312
|
Francolini I, Hall-Stoodley L, Stoodley P. Biofilms, Biomaterials, and Device-Related Infections. Biomater Sci 2020. [DOI: 10.1016/b978-0-12-816137-1.00054-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
313
|
Lachowicz JI, Dalla Torre G, Cappai R, Randaccio E, Nurchi VM, Bachor R, Szewczuk Z, Jaremko L, Jaremko M, Pisano MB, Cosentino S, Orrù G, Ibba A, Mujika J, Lopez X. Metal self-assembly mimosine peptides with enhanced antimicrobial activity: towards a new generation of multitasking chelating agents. Dalton Trans 2020; 49:2862-2879. [DOI: 10.1039/c9dt04545g] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Mimosine is a non-protein amino acid that can be used as a building block in peptides with metal coordination ability.
Collapse
|
314
|
Schwarzer S, James GA, Goeres D, Bjarnsholt T, Vickery K, Percival SL, Stoodley P, Schultz G, Jensen SO, Malone M. The efficacy of topical agents used in wounds for managing chronic biofilm infections: A systematic review. J Infect 2019; 80:261-270. [PMID: 31899281 DOI: 10.1016/j.jinf.2019.12.017] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 12/18/2019] [Accepted: 12/24/2019] [Indexed: 12/19/2022]
Abstract
OBJECTIVES Clinicians have increasingly adopted the widespread use of topical agents to manage chronic wound infections, despite limited data on their effectiveness in vivo. This study sought to evaluate the evidence for commonly employed topical agents used in wounds for the purpose of treating chronic infections caused by biofilm. METHOD We included in vitro, animal and human in vivo studies where topical agents were tested for their efficacy against biofilms, for use in wound care. For human studies, we only included those which utilised appropriate identification techniques for visualising and confirming the presence of biofilms. RESULT A total of 640 articles were identified, with 43 included after meeting eligibility. In vitro testing accounted for 90% (n = 39) of all included studies, five studies using animal models and three human in vivo studies. Sixteen different laboratory models were utilised, with the most frequent being the minimum biofilm eradication concentration (MBEC™) / well plate assay (38%, n = 15 of 39). A total of 44 commercially available topical agents were grouped into twelve categories with the most commonly tested agents being silver, iodine and polyhexamethylene biguanide (PHMB). In vitro results on efficacy demonstrated iodine as having the highest mean log10 reductions of all agents (4.81, ±3.14). CONCLUSION There is large disparity in the translation of laboratory studies to researchers undertaking human trials relating to the effectiveness of commercially available topical agents. There is insufficient human in vivo evidence to definitively recommend any commercially available topical agent over another for the treatment of chronic wound biofilms. The heterogeneity identified between study designs (in vitro to in vivo) further limits the generalisability of results.
Collapse
Affiliation(s)
- S Schwarzer
- South West Sydney Limb Preservation and Wound Research, South West Sydney Local Health District, Sydney, Australia.
| | - G A James
- Centre for Biofilm Engineering, Montana State University, Bozeman, MT, United States
| | - D Goeres
- Centre for Biofilm Engineering, Montana State University, Bozeman, MT, United States
| | - T Bjarnsholt
- Department of Immunology and Microbiology, Costerton Biofilm Centre, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Microbiology, Copenhagen University Hospital, Copenhagen, Denmark
| | - K Vickery
- Surgical Infection Research Group, Faculty of Medicine and Health Sciences, Macquarie University, Sydney Australia
| | - S L Percival
- 5D Health Protection Group Ltd, Centre of Excellence in Biofilm Science (CEBS), Liverpool Bio-Innovation Hub, Liverpool UK
| | - P Stoodley
- Departments of Microbial Infection and Immunity, and Orthopaedics, Ohio State University, Columbus, OH, United States
| | - G Schultz
- Department of Obstetrics & Gynecology, Institute for Wound Research, University of Florida, Gainesville, FL, United States
| | - S O Jensen
- South West Sydney Limb Preservation and Wound Research, South West Sydney Local Health District, Sydney, Australia; Infectious Diseases and Microbiology, School of Medicine, Ingham Institute for Applied Medical Research, Western Sydney University, United States
| | - M Malone
- South West Sydney Limb Preservation and Wound Research, South West Sydney Local Health District, Sydney, Australia; Infectious Diseases and Microbiology, School of Medicine, Ingham Institute for Applied Medical Research, Western Sydney University, United States
| |
Collapse
|
315
|
Jalili M, Amraei M, Sadeghifard N, Ghafourian S. Evaluation of Biofilm Formation and Anti-biofilm Properties of Peganum Harmala and Crocus Sativus in Shigella Flexneri Clinical Isolates. Open Microbiol J 2019. [DOI: 10.2174/1874285801913010297] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Background:
Biofilm formation causes many serious problems in the treatment of bacterial infections. In addition, chronic infections due to biofilm formation can pose a huge burden to the health care systems. Also, many bacteria are biofilm producers as an important strategy for pathogenicity. Furthermore, the traditional use of herbal medicines such as Peganum harmala and Crocus sativus in Iran is interesting.
Objective:
The purpose of the current study was to investigate the biofilm formation in Shigella flexneri clinical isolates and to evaluate the anti-biofilm properties of P. harmala and C. sativus on Shigella flexneri clinical isolates.
Methods:
For the study purpose, Thirty S.flexneri clinical isolates were collected from Ahvaz, Iran. Then, the collected bacteria were subjected to biofilm formation assay. Afterward, P. harmala and C. sativus were applied as an anti-biofilm formation in S. flexneri.
Results & Conclusion:
Our results demonstrated that a significant number of samples were identified as strong biofilm producers. Then, P. harmala and C . sativus in a concentration of 30μg/ml and 60μg/ml were able to eradicate a strong biofilm formation in S. flexneri, respectively. In addition, it seems that more extensive studies and in vivo research should be done to confirm their properties.
Collapse
|
316
|
Slade EA, Thorn RMS, Young A, Reynolds DM. An in vitro collagen perfusion wound biofilm model; with applications for antimicrobial studies and microbial metabolomics. BMC Microbiol 2019; 19:310. [PMID: 31888471 PMCID: PMC6937849 DOI: 10.1186/s12866-019-1682-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 12/11/2019] [Indexed: 12/12/2022] Open
Abstract
Background The majority of in vitro studies of medically relevant biofilms involve the development of biofilm on an inanimate solid surface. However, infection in vivo consists of biofilm growth on, or suspended within, the semi-solid matrix of the tissue, whereby current models do not effectively simulate the nature of the in vivo environment. This paper describes development of an in vitro method for culturing wound associated microorganisms in a system that combines a semi-solid collagen gel matrix with continuous flow of simulated wound fluid. This enables culture of wound associated reproducible steady state biofilms under conditions that more closely simulate the dynamic wound environment. To demonstrate the use of this model the antimicrobial kinetics of ceftazidime, against both mature and developing Pseudomonas aeruginosa biofilms, was assessed. In addition, we have shown the potential application of this model system for investigating microbial metabolomics by employing selected ion flow tube mass spectrometry (SIFT-MS) to monitor ammonia and hydrogen cyanide production by Pseudomonas aeruginosa biofilms in real-time. Results The collagen wound biofilm model facilitates growth of steady-state reproducible Pseudomonas aeruginosa biofilms under wound like conditions. A maximum biofilm density of 1010 cfu slide− 1 was achieved by 30 h of continuous culture and maintained throughout the remainder of the experiment. Treatment with ceftazidime at a clinically relevant dose resulted in a 1.2–1.6 log reduction in biofilm density at 72 h compared to untreated controls. Treatment resulted in loss of complex biofilm architecture and morphological changes to bacterial cells, visualised using confocal microscopy. When monitoring the biofilms using SIFT-MS, ammonia and hydrogen cyanide levels peaked at 12 h at 2273 ppb (±826.4) and 138 ppb (±49.1) respectively and were detectable throughout experimentation. Conclusions The collagen wound biofilm model has been developed to facilitate growth of reproducible biofilms under wound-like conditions. We have successfully used this method to: (1) evaluate antimicrobial efficacy and kinetics, clearly demonstrating the development of antimicrobial tolerance in biofilm cultures; (2) characterise volatile metabolite production by P. aeruginosa biofilms, demonstrating the potential use of this method in metabolomics studies.
Collapse
Affiliation(s)
- Elisabeth A Slade
- Centre for Research in Biosciences, University of the West of England, Bristol, UK
| | - Robin M S Thorn
- Centre for Research in Biosciences, University of the West of England, Bristol, UK
| | - Amber Young
- Scar Free Foundation Centre for Children's Burns Research, Bristol Royal Hospital for Children, Bristol, UK
| | - Darren M Reynolds
- Centre for Research in Biosciences, University of the West of England, Bristol, UK. .,University of the West of England, Frenchay Campus, Coldharbour Lane, Bristol, BS16 1QY, England.
| |
Collapse
|
317
|
Kirsner RS, Amaya R, Bass K, Boyar V, Ciprandi G, Glat PM, Percival SL, Romanelli M, Pittinger TP. Effects of a surfactant-based gel on acute and chronic paediatric wounds: a panel discussion and case series. J Wound Care 2019; 28:398-408. [PMID: 31166855 DOI: 10.12968/jowc.2019.28.6.398] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
On 20 November 2018, following the International Society for Paediatric Wound Care conference, a closed panel meeting took place in which the use of a surfactant-based gel (PluroGel (PMM), Medline Industries, Illinois, US) in paediatric wound care was discussed. The authors shared their experiences, thoughts, experimental data and clinical results. The panel identified the need for a product that can gently cleanse paediatric wounds and remove devitalised tissue without causing discomfort or skin reactions, as well as potentially promote healing. In adults, PMM has been shown to assist healing by hydrating the wound, controlling exudate and debriding non-viable tissue. Islands of neo-epithelium have also been reported to appear rapidly in different parts of the wound bed. No adverse effects on these proliferating cells have been observed. In vitro data suggest that PMM can remove biofilm, as well as potentially promote healing through cell salvage. The panel, therefore, set out to discuss their experiences of using PMM in the paediatric patients and to establish a consensus on the indications for its use and application in this population. This article will describe the main outcomes of that discussion and present case studies from paediatric patients with a variety of wound types, who were treated with PMM by members of the panel.
Collapse
Affiliation(s)
- Robert S Kirsner
- Chairman and Harvey Blank Professor, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, US
| | - Rene Amaya
- Director, Pediatric Wound Care and Laser Specialist, Houston, Texas, US
| | - Kathryn Bass
- Associate Professor of Surgery, Department of Pediatric Surgery, Women and Children's Hospital of Buffalo, New York, US
| | - Vita Boyar
- Director of Neonatal Wound Services, Neonatal-Perinatal Medicine, Alexandra and Steven Cohen Children's Medical Center of New York, Northwell Health, Zucker School of Medicine at Hofstra, Northwell, US
| | - Guido Ciprandi
- Paediatric and Plastic Surgeon, Bambino Gesù Children's Hospital, Department of Surgery, Division of Plastic and Maxillofacial Surgery, Head Wound Care Surgical Unit, Rome, Italy
| | - Paul M Glat
- Professor of Surgery and Pediatrics, Drexel University College of Medicine and Chief of Plastic Surgery, St. Christopher's Hospital for Children, Philadelphia, US
| | - Steven L Percival
- CEO of 5D Heath Protection Group Ltd, 5D Health Protection Group Ltd, Liverpool, UK
| | - Marco Romanelli
- Professor and Chairman, Department of Dermatology, University of Pisa, Italy
| | - Timothy P Pittinger
- Paediatric Surgeon, Department of Surgery & Regional Burn Center, Akron Children's Hospital, Akron, Ohio, US
| |
Collapse
|
318
|
Verderosa AD, Dhouib R, Fairfull-Smith KE, Totsika M. Nitroxide Functionalized Antibiotics Are Promising Eradication Agents against Staphylococcus aureus Biofilms. Antimicrob Agents Chemother 2019; 64:e01685-19. [PMID: 31636066 PMCID: PMC7187575 DOI: 10.1128/aac.01685-19] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 10/09/2019] [Indexed: 01/10/2023] Open
Abstract
Treatment of biofilm-related Staphylococcus aureus infections represents an important medical challenge worldwide, as biofilms, even those involving drug-susceptible S. aureus strains, are highly refractory to conventional antibiotic therapy. Nitroxides were recently shown to induce the dispersal of Gram-negative biofilms in vitro, but their action against Gram-positive bacterial biofilms remains unknown. Here, we demonstrate that the biofilm dispersal activity of nitroxides extends to S. aureus, a clinically important Gram-positive pathogen. Coadministration of the nitroxide CTEMPO (4-carboxy-2,2,6,6-tetramethylpiperidin-1-yloxyl) with ciprofloxacin significantly improved the biofilm eradication activity of the antibiotic against S. aureus Moreover, covalently linking the nitroxide to the antibiotic moiety further reduced the ciprofloxacin minimal biofilm eradication concentration. Microscopy analysis revealed that fluorescent nitroxide-antibiotic hybrids could penetrate S. aureus biofilms and enter cells localized at the surface and base of the biofilm structure. No toxicity to human cells was observed for the nitroxide CTEMPO or the nitroxide-antibiotic hybrids. Taken together, our results show that nitroxides can mediate the dispersal of Gram-positive biofilms and that dual-acting biofilm eradication antibiotics may provide broad-spectrum therapies for the treatment of biofilm-related infections.
Collapse
Affiliation(s)
- Anthony D Verderosa
- School of Chemistry, Physics, and Mechanical Engineering, Queensland University of Technology, Brisbane, Queensland, Australia
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Rabeb Dhouib
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Kathryn E Fairfull-Smith
- School of Chemistry, Physics, and Mechanical Engineering, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Makrina Totsika
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| |
Collapse
|
319
|
Al-Obaidi H, Kowalczyk RM, Kalgudi R, Zariwala MG. Griseofulvin solvate solid dispersions with synergistic effect against fungal biofilms. Colloids Surf B Biointerfaces 2019; 184:110540. [DOI: 10.1016/j.colsurfb.2019.110540] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/27/2019] [Accepted: 09/29/2019] [Indexed: 01/22/2023]
|
320
|
Xu Y, Dhaouadi Y, Stoodley P, Ren D. Sensing the unreachable: challenges and opportunities in biofilm detection. Curr Opin Biotechnol 2019; 64:79-84. [PMID: 31766008 DOI: 10.1016/j.copbio.2019.10.009] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 10/21/2019] [Accepted: 10/24/2019] [Indexed: 12/20/2022]
Abstract
Bacteria can attach to essentially all materials and form multicellular biofilms with high-level tolerance to antimicrobials. Detrimental biofilms are responsible for a variety of problems ranging from food and water contamination, bio-corrosion, to drug resistant infections. Besides the challenges in control, biofilms are also difficult to detect due to the lack of biofilm-specific biomarkers and methods for non-destructive imaging. In this article, we present a concise review of recent advancements in this field, with a focus on medical device-associated infections. We also discuss the technologies that have potential for non-destructive detection of bacterial biofilms.
Collapse
Affiliation(s)
- Yikang Xu
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY 13244, United States; Syracuse Biomaterials Institute, Syracuse University, Syracuse, NY 13244, United States
| | - Yousr Dhaouadi
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY 13244, United States; Syracuse Biomaterials Institute, Syracuse University, Syracuse, NY 13244, United States
| | - Paul Stoodley
- Departments of Microbial Infection and Immunity and Orthopaedics and the Infectious Diseases Institute, Ohio State University, Columbus, OH 43210, United States; National Centre for Advanced Tribology at Southampton (nCATS), and National Biofilm Innovation Centre, Mechanical Engineering, University of Southampton, United Kingdom
| | - Dacheng Ren
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY 13244, United States; Syracuse Biomaterials Institute, Syracuse University, Syracuse, NY 13244, United States; Department of Civil and Environmental Engineering, Syracuse University, Syracuse, NY 13244, United States; Department of Biology, Syracuse University, Syracuse, NY 13244, United States.
| |
Collapse
|
321
|
Structural and Functional Dynamics of Staphylococcus aureus Biofilms and Biofilm Matrix Proteins on Different Clinical Materials. Microorganisms 2019; 7:microorganisms7120584. [PMID: 31756969 PMCID: PMC6955704 DOI: 10.3390/microorganisms7120584] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 11/14/2019] [Accepted: 11/18/2019] [Indexed: 12/27/2022] Open
Abstract
Medical device-associated staphylococcal infections are a common and challenging problem. However, detailed knowledge of staphylococcal biofilm dynamics on clinically relevant surfaces is still limited. In the present study, biofilm formation of the Staphylococcus aureus ATCC 25923 strain was studied on clinically relevant materials-borosilicate glass, plexiglass, hydroxyapatite, titanium and polystyrene-at 18, 42 and 66 h. Materials with the highest surface roughness and porosity (hydroxyapatite and plexiglass) did not promote biofilm formation as efficiently as some other selected materials. Matrix-associated poly-N-acetyl-β-(1-6)-glucosamine (PNAG) was considered important in young (18 h) biofilms, whereas proteins appeared to play a more important role at later stages of biofilm development. A total of 460 proteins were identified from biofilm matrices formed on the indicated materials and time points-from which, 66 proteins were proposed to form the core surfaceome. At 18 h, the appearance of several r-proteins and glycolytic adhesive moonlighters, possibly via an autolysin (AtlA)-mediated release, was demonstrated in all materials, whereas classical surface adhesins, resistance- and virulence-associated proteins displayed greater variation in their abundances depending on the used material. Hydroxyapatite-associated biofilms were more susceptible to antibiotics than biofilms formed on titanium, but no clear correlation between the tolerance and biofilm age was observed. Thus, other factors, possibly the adhesive moonlighters, could have contributed to the observed chemotolerant phenotype. In addition, a protein-dependent matrix network was observed to be already well-established at the 18 h time point. To the best of our knowledge, this is among the first studies shedding light into matrix-associated surfaceomes of S. aureus biofilms grown on different clinically relevant materials and at different time points.
Collapse
|
322
|
Yu M, Chua SL. Demolishing the great wall of biofilms in Gram‐negative bacteria: To disrupt or disperse? Med Res Rev 2019; 40:1103-1116. [DOI: 10.1002/med.21647] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 11/03/2019] [Accepted: 11/07/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Miao Yu
- Department of Applied Biology and Chemical TechnologyThe Hong Kong Polytechnic University, KowloonHong Kong SAR China
- State Key Laboratory of Chemical Biology and Drug DiscoveryThe Hong Kong Polytechnic University, KowloonHong Kong SAR China
| | - Song Lin Chua
- Department of Applied Biology and Chemical TechnologyThe Hong Kong Polytechnic University, KowloonHong Kong SAR China
- State Key Laboratory of Chemical Biology and Drug DiscoveryThe Hong Kong Polytechnic University, KowloonHong Kong SAR China
| |
Collapse
|
323
|
Chronic wound biofilms. Chin Med J (Engl) 2019. [DOI: 10.1097/cm9.0000000000000523 and 21=21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2022] Open
|
324
|
Soares A, Roussel V, Pestel-Caron M, Barreau M, Caron F, Bouffartigues E, Chevalier S, Etienne M. Understanding Ciprofloxacin Failure in Pseudomonas aeruginosa Biofilm: Persister Cells Survive Matrix Disruption. Front Microbiol 2019; 10:2603. [PMID: 31798554 PMCID: PMC6864029 DOI: 10.3389/fmicb.2019.02603] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 10/28/2019] [Indexed: 12/22/2022] Open
Abstract
Biofilms are commonly recalcitrant to antibiotics, through incompletely elucidated mechanisms such as tolerance and persistence. We aimed at investigating how a Pseudomonas aeruginosa biofilm escapes ciprofloxacin treatment. P. aeruginosa PA14 in vitro mature biofilms were challenged with supra-MIC ciprofloxacin concentrations. Cell viability was quantified by fluorescein diacetate assay. Population dynamics were determined by counts of surviving culturable cells. Biofilms were analyzed using confocal laser scanning microscopy (CLSM), and the expression of genes involved in stringent response, toxin-antitoxin HigB/HigA, and type 3 secretion system (T3SS) was quantified by RT-qPCR in untreated and treated biofilms. Ciprofloxacin exposure resulted in an initial reduction of bacterial counts following a biphasic time-kill curve. After 24 h of treatment, the overall cell activity and the density of culturable cells significantly decreased as compared to untreated biofilm. No resistant mutant was isolated among the <1% surviving cells. Phenotypic adaptation toward persistence appeared to start after only 1 h of antibiotic exposure, by an overexpression of the genes involved in stringent response and in the toxin-antitoxin system, whereas the expression of genes encoding for the T3SS remained unchanged. After 4 h of ciprofloxacin exposure, stringent response genes returned to their basal level of expression. After a prolonged ciprofloxacin exposure, a deep alteration in the matrix structure that became thinner and lost mushroom-like aggregates was observed, in relation with reduced biovolumes of exopolysaccharides and extracellular DNA. These results support that ciprofloxacin might first induce the bacterial killing of most bacterial cells, but simultaneously activate stringent response mechanisms contributing to the switch of a subpopulation toward a persister phenotype. Once the persister phenotype is expressed, and despite an unexpected alteration of the biofilm matrix, ciprofloxacin fails to eradicate biofilm.
Collapse
Affiliation(s)
- Anaïs Soares
- GRAM 2.0, EA 2656, Normandie University, UNIROUEN, Rouen, France.,Microbiology Department, Rouen University Hospital, Rouen, France
| | - Valérie Roussel
- GRAM 2.0, EA 2656, Normandie University, UNIROUEN, Rouen, France
| | - Martine Pestel-Caron
- GRAM 2.0, EA 2656, Normandie University, UNIROUEN, Rouen, France.,Microbiology Department, Rouen University Hospital, Rouen, France
| | - Magalie Barreau
- EA 4312, LMSM, Normandie University, UNIROUEN, Evreux, France
| | - François Caron
- GRAM 2.0, EA 2656, Normandie University, UNIROUEN, Rouen, France.,Infectious Diseases Department, Rouen University Hospital, Rouen, France
| | | | | | - Manuel Etienne
- GRAM 2.0, EA 2656, Normandie University, UNIROUEN, Rouen, France.,Infectious Diseases Department, Rouen University Hospital, Rouen, France
| |
Collapse
|
325
|
Griffith EC, Zhao Y, Singh AP, Conlon BP, Tangallapally R, Shadrick WR, Liu J, Wallace MJ, Yang L, Elmore JM, Li Y, Zheng Z, Miller DJ, Cheramie MN, Lee RB, LaFleur MD, Lewis K, Lee RE. Ureadepsipeptides as ClpP Activators. ACS Infect Dis 2019; 5:1915-1925. [PMID: 31588734 DOI: 10.1021/acsinfecdis.9b00245] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Acyldepsipeptides are a unique class of antibiotics that act via allosterically dysregulated activation of the bacterial caseinolytic protease (ClpP). The ability of ClpP activators to kill nongrowing bacteria represents a new opportunity to combat deep-seated biofilm infections. However, the acyldepsipeptide scaffold is subject to rapid metabolism. Herein, we explore alteration of the potentially metabolically reactive α,β unsaturated acyl chain. Through targeted synthesis, a new class of phenyl urea substituted depsipeptide ClpP activators with improved metabolic stability is described. The ureadepsipeptides are potent activators of Staphylococcus aureus ClpP and show activity against Gram-positive bacteria, including S. aureus biofilms. These studies demonstrate that a phenyl urea motif can successfully mimic the double bond, maintaining potency equivalent to acyldepsipeptides but with decreased metabolic liability. Although removal of the double bond from acyldepsipeptides generally has a significant negative impact on potency, structural studies revealed that the phenyl ureadepsipeptides can retain potency through the formation of a third hydrogen bond between the urea and the key Tyr63 residue in the ClpP activation domain. Ureadepsipeptides represent a new class of ClpP activators with improved drug-like properties, potent antibacterial activity, and the tractability to be further optimized.
Collapse
Affiliation(s)
- Elizabeth C. Griffith
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, United States
| | - Ying Zhao
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, United States
| | - Aman P. Singh
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, United States
| | - Brian P. Conlon
- Antimicrobial Discovery Center, Department of Biology, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Rajendra Tangallapally
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, United States
| | - William R. Shadrick
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, United States
| | - Jiuyu Liu
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, United States
| | - Miranda J. Wallace
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, United States
| | - Lei Yang
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, United States
| | - John M. Elmore
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, United States
| | - Yong Li
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, United States
| | - Zhong Zheng
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, United States
| | - Darcie J. Miller
- Department of Structure Biology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, United States
| | - Martin N. Cheramie
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, United States
| | - Robin B. Lee
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, United States
| | - Michael D. LaFleur
- Arietis Pharma, 650 Albany Street, Suite 114, Boston, Massachusetts 02118, United States
| | - Kim Lewis
- Antimicrobial Discovery Center, Department of Biology, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Richard E. Lee
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, United States
| |
Collapse
|
326
|
Hajhosseini B, Chiou GJ, Dori G, Fukaya E, Chandra V, Meyer S, Gurtner GC. Er:YAG laser vs. sharp debridement in management of chronic wounds: Effects on pain and bacterial load. Wound Repair Regen 2019; 28:118-125. [DOI: 10.1111/wrr.12764] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 09/06/2019] [Accepted: 09/13/2019] [Indexed: 11/30/2022]
Affiliation(s)
- Babak Hajhosseini
- Division of Plastic and Reconstructive SurgeryDepartment of Surgery, Stanford University School of Medicine Stanford California
- Advanced Wound Care CenterStanford University School of Medicine Redwood City California
| | - Grace J. Chiou
- Division of Plastic and Reconstructive SurgeryDepartment of Surgery, Stanford University School of Medicine Stanford California
- Advanced Wound Care CenterStanford University School of Medicine Redwood City California
| | - Gretchen Dori
- Advanced Wound Care CenterStanford University School of Medicine Redwood City California
| | - Eri Fukaya
- Advanced Wound Care CenterStanford University School of Medicine Redwood City California
- Division of Vascular SurgeryDepartment of Surgery, Stanford University School of Medicine Stanford California
| | - Venita Chandra
- Advanced Wound Care CenterStanford University School of Medicine Redwood City California
- Division of Vascular SurgeryDepartment of Surgery, Stanford University School of Medicine Stanford California
| | - Shannon Meyer
- Division of Plastic and Reconstructive SurgeryDepartment of Surgery, Stanford University School of Medicine Stanford California
- Advanced Wound Care CenterStanford University School of Medicine Redwood City California
| | - Geoffrey C. Gurtner
- Division of Plastic and Reconstructive SurgeryDepartment of Surgery, Stanford University School of Medicine Stanford California
- Advanced Wound Care CenterStanford University School of Medicine Redwood City California
| |
Collapse
|
327
|
Robertson SN, Childs PG, Akinbobola A, Henriquez FL, Ramage G, Reid S, Mackay WG, Williams C. Reduction of Pseudomonas aeruginosa biofilm formation through the application of nanoscale vibration. J Biosci Bioeng 2019; 129:379-386. [PMID: 31623950 DOI: 10.1016/j.jbiosc.2019.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 08/13/2019] [Accepted: 09/06/2019] [Indexed: 01/30/2023]
Abstract
Bacterial biofilms pose a significant burden in both healthcare and industrial environments. With the limited effectiveness of current biofilm control strategies, novel or adjunctive methods in biofilm control are being actively pursued. Reported here, is the first evidence of the application of nanovibrational stimulation (nanokicking) to reduce the biofilm formation of Pseudomonas aeruginosa. Nanoscale vertical displacements (approximately 60 nm) were imposed on P. aeruginosa cultures, with a significant reduction in biomass formation observed at frequencies between 200 and 4000 Hz at 24 h. The optimal reduction of biofilm formation was observed at 1 kHz, with changes in the physical morphology of the biofilms. Scanning electron microscope imaging of control and biofilms formed under nanovibrational stimulation gave indication of a reduction in extracellular matrix (ECM). Quantification of the carbohydrate and protein components of the ECM was performed and showed a significant reduction at 24 h at 1 kHz frequency. To model the forces being exerted by nanovibrational stimulation, laser interferometry was performed to measure the amplitudes produced across the Petri dish surfaces. Estimated peak forces on each cell, associated with the nanovibrational stimulation technique, were calculated to be in the order of 10 pN during initial biofilm formation. This represents a potential method of controlling microbial biofilm formation in a number of important settings in industry and medical related processes.
Collapse
Affiliation(s)
- Shaun N Robertson
- Institute of Healthcare, Policy and Practice, School of Health & Life Sciences, University of the West of Scotland, High Street, Paisley PA1 2BE, Scotland, UK; SUPA, Institute of Thin Films, Sensors and Imaging, School of Engineering and Computing, University of the West of Scotland, High Street, Paisley PA1 2BE, Scotland, UK; SUPA, Department of Biomedical Engineering, University of Strathclyde, 40 George Street, Glasgow G1 1QE, Scotland, UK
| | - Peter G Childs
- SUPA, Institute of Thin Films, Sensors and Imaging, School of Engineering and Computing, University of the West of Scotland, High Street, Paisley PA1 2BE, Scotland, UK; Centre for the Cellular Microenvironments (CeMi), School of Engineering, University of Glasgow, G12 8LT, Scotland, UK
| | - Ayorinde Akinbobola
- Institute of Healthcare, Policy and Practice, School of Health & Life Sciences, University of the West of Scotland, High Street, Paisley PA1 2BE, Scotland, UK
| | - Fiona L Henriquez
- Institute of Biomedical and Environmental Health Research, School of Health & Life Sciences, University of the West of Scotland, High Street, Paisley PA1 2BE, Scotland, UK
| | - Gordon Ramage
- School of Medicine, Dentistry and Nursing, MVLS, University of Glasgow, 378 Sauchiehall St, Glasgow G2 3JZ, Scotland, UK
| | - Stuart Reid
- SUPA, Institute of Thin Films, Sensors and Imaging, School of Engineering and Computing, University of the West of Scotland, High Street, Paisley PA1 2BE, Scotland, UK; SUPA, Department of Biomedical Engineering, University of Strathclyde, 40 George Street, Glasgow G1 1QE, Scotland, UK
| | - William G Mackay
- Institute of Healthcare, Policy and Practice, School of Health & Life Sciences, University of the West of Scotland, High Street, Paisley PA1 2BE, Scotland, UK.
| | - Craig Williams
- Institute of Healthcare, Policy and Practice, School of Health & Life Sciences, University of the West of Scotland, High Street, Paisley PA1 2BE, Scotland, UK
| |
Collapse
|
328
|
Viviana Serna González C, Thum M, de Oliveira Ramalho A, Beloto Silva O, Franco Coelho M, Medeiros da Silva Queiroz W, Maria Sebba Tosta de Souza D, Cristina Nogueira P, Lúcia Conceição Gouveia Santos V. Análise da “1a Recomendação Brasileira para o Gerenciamento do Biofilme em Feridas Crônicas e Complexas”. ESTIMA 2019. [DOI: 10.30886/estima.v17.783_pt] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Objetivos: Analisar criticamente a “1ª Recomendação brasileira para o gerenciamento de biofilme em feridas crônicas e complexas”. Método: Realizou-se revisão da literatura atual às informações nele contidas. Resultados: Observou-se que a publicação carece de metodologia compatível com o título, existem lacunas nas recomendações quanto à classificação das evidências e com ausência de fundamentação a partir de importantes consensos internacionais para o tratamento das feridas complexas com suspeita de biofilme, publicados nos últimos três anos. Conclusão: Conclui-se que o manuscrito não deve ser usado como guia de recomendações clínicas, mas como revisão bibliográfica sobre o tema.
Collapse
|
329
|
Viviana Serna González C, Thum M, de Oliveira Ramalho A, Beloto Silva O, Franco Coelho M, Medeiros da Silva Queiroz W, Maria Sebba Tosta de Souza D, Cristina Nogueira P, Lúcia Conceição Gouveia Santos V. Analysis of “1st Brazilian Recommendation for Biofilm Management in Chronic and Complex Wounds”. ESTIMA 2019. [DOI: 10.30886/estima.v17.783_in] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Objectives: Analyze critically the “1st Brazilian Recommendation for Biofilm Management in Chronic and Complex Wounds” (from Portuguese, “1a Recomendação Brasileira para o Gerenciamento de Biofilme em Feridas Crônicas e Complexas”). Method: Reviewing information contained in said document according to current literature. Results: The publication was showed to lack methodology compatible with its title; gaps in the recommendations were perceived regarding evidence classification, as well as an absence of grounding from important international consensus, published in the last three years, about treatment of complex wounds with suspected biofilm. Conclusion: The document was concluded to be inadequate for use as a clinical guideline, being considered only a bibliographic review about the theme.
Collapse
|
330
|
Rupel K, Zupin L, Ottaviani G, Bertani I, Martinelli V, Porrelli D, Vodret S, Vuerich R, Passos da Silva D, Bussani R, Crovella S, Parsek M, Venturi V, Di Lenarda R, Biasotto M, Zacchigna S. Blue laser light inhibits biofilm formation in vitro and in vivo by inducing oxidative stress. NPJ Biofilms Microbiomes 2019; 5:29. [PMID: 31602310 PMCID: PMC6785554 DOI: 10.1038/s41522-019-0102-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 09/02/2019] [Indexed: 11/09/2022] Open
Abstract
Resolution of bacterial infections is often hampered by both resistance to conventional antibiotic therapy and hiding of bacterial cells inside biofilms, warranting the development of innovative therapeutic strategies. Here, we report the efficacy of blue laser light in eradicating Pseudomonas aeruginosa cells, grown in planktonic state, agar plates and mature biofilms, both in vitro and in vivo, with minimal toxicity to mammalian cells and tissues. Results obtained using knock-out mutants point to oxidative stress as a relevant mechanism by which blue laser light exerts its anti-microbial effect. Finally, the therapeutic potential is confirmed in a mouse model of skin wound infection. Collectively, these data set blue laser phototherapy as an innovative approach to inhibit bacterial growth and biofilm formation, and thus as a realistic treatment option for superinfected wounds.
Collapse
Affiliation(s)
- Katia Rupel
- 1Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy
| | - Luisa Zupin
- 1Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy
| | - Giulia Ottaviani
- 1Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy
| | - Iris Bertani
- 2Bacteriology Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149 Trieste, Italy
| | - Valentina Martinelli
- 3Cardiovascular Biology Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149 Trieste, Italy
| | - Davide Porrelli
- 1Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy
| | - Simone Vodret
- 3Cardiovascular Biology Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149 Trieste, Italy
| | - Roman Vuerich
- 3Cardiovascular Biology Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149 Trieste, Italy
| | | | - Rossana Bussani
- 1Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy
| | - Sergio Crovella
- 1Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy.,5Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", 34137 Trieste, Italy
| | - Matthew Parsek
- 4Department of Microbiology, University of Washington, Seattle, WA 98195 USA
| | - Vittorio Venturi
- 2Bacteriology Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149 Trieste, Italy
| | - Roberto Di Lenarda
- 1Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy
| | - Matteo Biasotto
- 1Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy
| | - Serena Zacchigna
- 1Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy.,3Cardiovascular Biology Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149 Trieste, Italy
| |
Collapse
|
331
|
Methods Used for the Eradication of Staphylococcal Biofilms. Antibiotics (Basel) 2019; 8:antibiotics8040174. [PMID: 31590240 PMCID: PMC6963202 DOI: 10.3390/antibiotics8040174] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 09/27/2019] [Accepted: 10/01/2019] [Indexed: 02/07/2023] Open
Abstract
Staphylococcus aureus is considered one of the leading pathogens responsible for community and healthcare-associated infections. Among them, infections caused by methicillin-resistant strains (MRSA) are connected with ineffective or prolonged treatment. The therapy of staphylococcal infections faces many difficulties, not only because of the bacteria's resistance to antibiotics and the multiplicity of virulence factors it produces, but also due to its ability to form a biofilm. The present review focuses on several approaches used for the assessment of staphylococcal biofilm eradication. The methods described here are successfully applied in research on the prevention of biofilm-associated infections, as well as in their management. They include not only the evaluation of the antimicrobial activity of novel compounds, but also the methods for biomaterial functionalization. Moreover, the advantages and limitations of different dyes and techniques used for biofilm characterization are discussed. Therefore, this review may be helpful for those scientists who work on the development of new antistaphylococcal compounds.
Collapse
|
332
|
Engin AB, Engin A. Nanoantibiotics: A Novel Rational Approach to Antibiotic Resistant Infections. Curr Drug Metab 2019; 20:720-741. [DOI: 10.2174/1389200220666190806142835] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 07/19/2019] [Accepted: 07/19/2019] [Indexed: 01/09/2023]
Abstract
Background:The main drawbacks for using conventional antimicrobial agents are the development of multiple drug resistance due to the use of high concentrations of antibiotics for extended periods. This vicious cycle often generates complications of persistent infections, and intolerable antibiotic toxicity. The problem is that while all new discovered antimicrobials are effective and promising, they remain as only short-term solutions to the overall challenge of drug-resistant bacteria.Objective:Recently, nanoantibiotics (nAbts) have been of tremendous interest in overcoming the drug resistance developed by several pathogenic microorganisms against most of the commonly used antibiotics. Compared with free antibiotic at the same concentration, drug delivered via a nanoparticle carrier has a much more prominent inhibitory effect on bacterial growth, and drug toxicity, along with prolonged drug release. Additionally, multiple drugs or antimicrobials can be packaged within the same smart polymer which can be designed with stimuli-responsive linkers. These stimuli-responsive nAbts open up the possibility of creating multipurpose and targeted antimicrobials. Biofilm formation still remains the leading cause of conventional antibiotic treatment failure. In contrast to conventional antibiotics nAbts easily penetrate into the biofilm, and selectively target biofilm matrix constituents through the introduction of bacteria specific ligands. In this context, various nanoparticles can be stabilized and functionalized with conventional antibiotics. These composites have a largely enhanced bactericidal efficiency compared to the free antibiotic.Conclusion:Nanoparticle-based carriers deliver antibiotics with better biofilm penetration and lower toxicity, thus combating bacterial resistance. However, the successful adaptation of nanoformulations to clinical practice involves a detailed assessment of their safety profiles and potential immunotoxicity.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Faculty of Pharmacy, Department of Toxicology, Gazi University, Ankara, Turkey
| | - Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Ankara, Turkey
| |
Collapse
|
333
|
Jørgensen E, Bay L, Skovgaard LT, Bjarnsholt T, Jacobsen S. An Equine Wound Model to Study Effects of Bacterial Aggregates on Wound Healing. Adv Wound Care (New Rochelle) 2019; 8:487-498. [PMID: 31456906 PMCID: PMC6709944 DOI: 10.1089/wound.2018.0901] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 01/10/2019] [Indexed: 01/22/2023] Open
Abstract
Objective: Relevant animal models to study effects of bacterial aggregates on wound healing are lacking. We aimed at establishing an equine wound model with bacterial aggregates to investigate the impact of bacterial inoculation on normal (thorax) and impaired (limb) wound healing. Approach: Wounds were created on three limbs and both thorax sides of six horses. Twelve out of 20 wounds per horse were inoculated with 104 Staphylococcus aureus and 105 Pseudomonas aeruginosa on day 4. Healing was monitored until day 27 by clinical assessment, including wound scoring, surface pH measurements, and digital photography for area determination. Biopsies were used for bacterial culture and for peptide nucleic acid fluorescence in situ hybridization to detect bacterial aggregates. Results: Inoculated limb wounds healed slower than noninoculated limb wounds from day 10 onward (p < 0.0001). Inoculated and noninoculated thorax wounds healed equally well and faster than limb wounds. The odds ratio of detecting bacterial aggregates in inoculated limb wounds was 7.1 (2.4-21.0, p = 0.0086) compared with noninoculated limb wounds and 36.2 (3.8-348, p = 0.0018) compared with thorax wounds. Innovation: This equine wound model with bacterial aggregates might be superior to other animal wound models, as both normal and impaired healing can be studied simultaneously. In this model, many aspects of wound healing, including novel treatments, may be studied. Conclusions: The impaired healing observed in inoculated limb wounds may be related to the persistent bacterial aggregates. Both in capability of clearing inoculated bacteria from the wounds and in healing pattern, thorax wounds were superior to limb wounds.
Collapse
Affiliation(s)
- Elin Jørgensen
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Taastrup, Denmark
| | - Lene Bay
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - Lene T. Skovgaard
- Section of Biostatistics, Department of Public Health, University of Copenhagen, Copenhagen K, Denmark
| | - Thomas Bjarnsholt
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen Ø, Denmark
| | - Stine Jacobsen
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Taastrup, Denmark
| |
Collapse
|
334
|
Abstract
Antibiotic resistance is a growing concern for management of common bacterial infections. Here, we show that antibiotics can be effective at subinhibitory levels when bacteria carry latent phage. Our findings suggest that specific treatment strategies based on the identification of latent viruses in individual bacterial strains may be an effective personalized medicine approach to antibiotic stewardship. Most bacteria and archaea are infected by latent viruses that change their physiology and responses to environmental stress. We use a population model of the bacterium-phage relationship to examine the role that latent phage play in the bacterial population over time in response to antibiotic treatment. We demonstrate that the stress induced by antibiotic administration, even if bacteria are resistant to killing by antibiotics, is sufficient to control the infection under certain conditions. This work expands the breadth of understanding of phage-antibiotic synergy to include both temperate and chronic viruses persisting in their latent form in bacterial populations. IMPORTANCE Antibiotic resistance is a growing concern for management of common bacterial infections. Here, we show that antibiotics can be effective at subinhibitory levels when bacteria carry latent phage. Our findings suggest that specific treatment strategies based on the identification of latent viruses in individual bacterial strains may be an effective personalized medicine approach to antibiotic stewardship.
Collapse
|
335
|
Cooper R, Kirketerp-Møller K. Non-antibiotic antimicrobial interventions and antimicrobial stewardship in wound care. J Wound Care 2019; 27:355-377. [PMID: 29883284 DOI: 10.12968/jowc.2018.27.6.355] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Control of wound infection today relies largely on antibiotics, but the continual emergence of antibiotic-resistant microorganisms threatens a return to the pre-antibiotic era when physicians used antiseptics to prevent and manage infection. Some of those antiseptics are still used today, and others have become available. A diverse variety of non-antibiotic antimicrobial interventions are found on modern formularies. Unlike the mode of action of antibiotics, which affect specific cellular target sites of pathogens, many non-antibiotic antimicrobials affect multiple cellular target sites in a non-specific way. Although this reduces the likelihood of selecting for resistant strains of microorganisms, some have emerged and cross-resistance between antibiotics and antiseptics has been detected. With the prospect of a post-antibiotic era looming, ways to maintain and extend our antimicrobial armamentarium must be found. In this narrative review, current and emerging non-antibiotic antimicrobial strategies will be considered and the need for antimicrobial stewardship in wound care will be explained.
Collapse
Affiliation(s)
- Rose Cooper
- Professor of Microbiology, Department of Biomedical Science, Cardiff School of Health Sciences, Cardiff Metropolitan University, Western Avenue, Cardiff, UK
| | - Klaus Kirketerp-Møller
- Orthopaedic Surgeon, Copenhagen Wound Healing Center, Department of Dermatology and Wounds, Bispebjerg University Hospital, Bispebjerg Bakke 23, DK-2400 Copenhagen NV
| |
Collapse
|
336
|
Preda VG, Săndulescu O. Communication is the key: biofilms, quorum sensing, formation and prevention. Discoveries (Craiova) 2019; 7:e100. [PMID: 32309618 PMCID: PMC7086079 DOI: 10.15190/d.2019.13] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 09/29/2019] [Accepted: 09/29/2019] [Indexed: 12/19/2022] Open
Abstract
Antibiotic resistance is a relevant topic nowadays, representing one of the main causes of infection-related mortality and morbidity at a global level. This phenomenon is worrisome and represents an area of interest for both clinical practice and fundamental research. One important mechanism whereby bacteria acquire resistance to antibiotics and evade the immune system is by forming biofilms. It is estimated that ~80% of the bacteria producing chronic infections can form biofilms. During the process of biofilm formation microorganisms have the ability to communicate with each other through quorum sensing. Quorum sensing regulates the metabolic activity of planktonic cells, and it can induce microbial biofilm formation and increased virulence. In this review we describe the biofilm formation process, quorum sensing, quorum quenching, several key infectious bacteria producing biofilm, methods of prevention and their challenges and limitations. Although progress has been made in the prevention and treatment of biofilm-driven infections, new strategies are required and have to be further developed.
Collapse
Affiliation(s)
- Veronica G. Preda
- Department of Biochemistry and Molecular Biology, University of Bucharest, Faculty of Biology, Bucharest, Romania
- Department of Genetics and Applied Biotechnology, University of Bucharest, Faculty of Biology, Bucharest, Romania
| | - Oana Săndulescu
- Department of Infectious Diseases I, Carol Davila University of Medicine and Pharmacy, National Institute for Infectious Diseases “Prof. Dr. Matei Balș”, Bucharest, Romania
| |
Collapse
|
337
|
Malone M, Schwarzer S, Radzieta M, Jeffries T, Walsh A, Dickson HG, Micali G, Jensen SO. Effect on total microbial load and community composition with two vs six-week topical Cadexomer Iodine for treating chronic biofilm infections in diabetic foot ulcers. Int Wound J 2019; 16:1477-1486. [PMID: 31487117 DOI: 10.1111/iwj.13219] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/18/2019] [Accepted: 08/21/2019] [Indexed: 12/13/2022] Open
Abstract
This study compares two vs six weeks of topical antimicrobial therapy with Cadexomer Iodine in patients with diabetic foot ulcers (DFUs) complicated by chronic biofilm infections. Patients with non-healing DFUs with suspected chronic biofilm infections were eligible for enrolment. Patients were randomised to receive either two or six weeks of treatment with topical Cadexomer Iodine. Tissue biopsies from the ulcers were obtained pre-and-post treatment and underwent DNA sequencing and real-time quantitative polymerase chain reaction (PCR) to determine the total microbial load, community composition, and diversity of bacteria. Scanning electron microscopy confirmed biofilm in all 18 ulcers with suspected chronic biofilm infections. Cadexomer Iodine resulted in 14 of 18 (78%) samples achieving a mean 0.5 log10 reduction in microbial load. Regardless of treatment duration, there was no statistical difference in the reduction of total microbial loads. No difference in the rate of wound healing in the two groups was seen at 6 weeks. Cadexomer Iodine reduces the total microbial load in DFUs with chronic biofilm infections and affects microbial community composition and diversity. All ulcers in both groups showed an initial reduction in wound size with application of Cadexomer Iodine, which might reflect its effect on biofilms.
Collapse
Affiliation(s)
- Matthew Malone
- South West Sydney Limb Preservation and Wound Research, South West Sydney Local Health District, Sydney, New South Wales, Australia.,High Risk Foot Service, Liverpool Hospital, South West Sydney LHD, Sydney, New South Wales, Australia.,Infectious Diseases and Microbiology, School of Medicine, Western Sydney University, Sydney, New South Wales, Australia
| | - Saskia Schwarzer
- South West Sydney Limb Preservation and Wound Research, South West Sydney Local Health District, Sydney, New South Wales, Australia.,High Risk Foot Service, Liverpool Hospital, South West Sydney LHD, Sydney, New South Wales, Australia
| | - Michael Radzieta
- South West Sydney Limb Preservation and Wound Research, South West Sydney Local Health District, Sydney, New South Wales, Australia.,Infectious Diseases and Microbiology, School of Medicine, Western Sydney University, Sydney, New South Wales, Australia
| | - Thomas Jeffries
- Infectious Diseases and Microbiology, School of Medicine, Western Sydney University, Sydney, New South Wales, Australia
| | - Annie Walsh
- South West Sydney Limb Preservation and Wound Research, South West Sydney Local Health District, Sydney, New South Wales, Australia.,High Risk Foot Service, Liverpool Hospital, South West Sydney LHD, Sydney, New South Wales, Australia
| | - Hugh G Dickson
- South West Sydney Limb Preservation and Wound Research, South West Sydney Local Health District, Sydney, New South Wales, Australia
| | - Grace Micali
- Antimicrobial Resistance and Mobile Elements Group, Ingham Institute of Applied Medical Research, Sydney, New South Wales, Australia
| | - Slade O Jensen
- South West Sydney Limb Preservation and Wound Research, South West Sydney Local Health District, Sydney, New South Wales, Australia.,Infectious Diseases and Microbiology, School of Medicine, Western Sydney University, Sydney, New South Wales, Australia.,Antimicrobial Resistance and Mobile Elements Group, Ingham Institute of Applied Medical Research, Sydney, New South Wales, Australia
| |
Collapse
|
338
|
Gabaldón T. Recent trends in molecular diagnostics of yeast infections: from PCR to NGS. FEMS Microbiol Rev 2019; 43:517-547. [PMID: 31158289 PMCID: PMC8038933 DOI: 10.1093/femsre/fuz015] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 05/31/2019] [Indexed: 12/29/2022] Open
Abstract
The incidence of opportunistic yeast infections in humans has been increasing over recent years. These infections are difficult to treat and diagnose, in part due to the large number and broad diversity of species that can underlie the infection. In addition, resistance to one or several antifungal drugs in infecting strains is increasingly being reported, severely limiting therapeutic options and showcasing the need for rapid detection of the infecting agent and its drug susceptibility profile. Current methods for species and resistance identification lack satisfactory sensitivity and specificity, and often require prior culturing of the infecting agent, which delays diagnosis. Recently developed high-throughput technologies such as next generation sequencing or proteomics are opening completely new avenues for more sensitive, accurate and fast diagnosis of yeast pathogens. These approaches are the focus of intensive research, but translation into the clinics requires overcoming important challenges. In this review, we provide an overview of existing and recently emerged approaches that can be used in the identification of yeast pathogens and their drug resistance profiles. Throughout the text we highlight the advantages and disadvantages of each methodology and discuss the most promising developments in their path from bench to bedside.
Collapse
Affiliation(s)
- Toni Gabaldón
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr Aiguader 88, Barcelona 08003, Spain
- Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
- ICREA, Pg Lluís Companys 23, 08010 Barcelona, Spain
| |
Collapse
|
339
|
Kvich L, Fritz B, Crone S, Kragh KN, Kolpen M, Sønderholm M, Andersson M, Koch A, Jensen PØ, Bjarnsholt T. Oxygen Restriction Generates Difficult-to-Culture P. aeruginosa. Front Microbiol 2019; 10:1992. [PMID: 31555231 PMCID: PMC6727857 DOI: 10.3389/fmicb.2019.01992] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 08/13/2019] [Indexed: 11/13/2022] Open
Abstract
Induction of a non-culturable state has been demonstrated for many bacteria, e.g., Escherichia coli and various Vibrio spp. In a clinical perspective, the lack of growth due to these non-culturable bacteria can have major consequences for the treatment of patients. Here, we show how anoxic conditioning (restriction of molecular oxygen, O2) generates difficult-to-culture (DTC) bacteria during biofilm growth. A significant subpopulation of Pseudomonas aeruginosa entered a DTC state after anoxic conditioning, ranging from 5 to 90% of the total culturable population, in both planktonic and biofilm models. Anoxic conditioning also generated DTC subpopulations of Staphylococcus aureus and Staphylococcus epidermidis (89 and 42% of the total culturable population, respectively). Growth of the DTC populations were achieved by substituting O2 with 10 mM NO3– as an alternative electron acceptor for anaerobic respiration or, in the case of P. aeruginosa, by adding sodium pyruvate or catalase as scavengers against reactive oxygen species (ROS) during aerobic respiration. An increase in normoxic plating due to addition of catalase suggests the molecule hydrogen peroxide as a possible mechanism for induction of DTC P. aeruginosa. Anoxic conditioning also generated a true viable but non-culturable (VBNC) population of P. aeruginosa that was not resurrected by substituting O2 with NO3– during anaerobic respiration. These results demonstrate that habituation to an anoxic micro-environment could complicate diagnostic culturing of bacteria, especially in the case of chronic infections where oxygen is restricted due to the host immune response.
Collapse
Affiliation(s)
- Lasse Kvich
- Costerton Biofilm Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Blaine Fritz
- Costerton Biofilm Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Stephanie Crone
- Costerton Biofilm Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kasper N Kragh
- Costerton Biofilm Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mette Kolpen
- Department of Clinical Microbiology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Majken Sønderholm
- Costerton Biofilm Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mikael Andersson
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Anders Koch
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark.,Department of Infectious Diseases, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Peter Ø Jensen
- Costerton Biofilm Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Clinical Microbiology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Thomas Bjarnsholt
- Costerton Biofilm Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Clinical Microbiology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
340
|
|
341
|
Abstract
PURPOSE OF REVIEW Biofilm-associated infections cause difficulties in the management of childhood chronic infections and other diseases, due to the invasive nature of interventions which are often necessary for definitive management. Despite their importance, there are challenges in diagnosing biofilm infections and gaps in clinicians' understanding regarding the significance of biofilms. RECENT FINDINGS Many chronic infections associated with biofilms remain difficult or impossible to eradicate with conventional therapy. Surgical intervention, implant removal or long-term intermittent or suppressive antimicrobial therapy may be required. There are still significant challenges in detecting biofilms which presents a barrier in clinical practice and research. Novel therapies to disrupt biofilms are currently under investigation, which may help reduce the impact of antimicrobial resistance. SUMMARY Biofilm-associated infection should be considered wherever there is clinical concern for an infection affecting prosthetic material, where there is a predisposing condition such as suppurative lung disease; or in the setting of chronic or relapsing infections which may be culture negative. New diagnostic methods for detecting biofilms are a research priority for both clinical diagnosis and the ability to conduct high quality clinical trials of novel antibiofilm interventions.
Collapse
|
342
|
Riangrungroj P, Polizzi KM. BeQuIK (Biosensor Engineered Quorum Induced Killing): designer bacteria for destroying recalcitrant biofilms. Microb Biotechnol 2019; 13:311-314. [PMID: 31328393 PMCID: PMC7017806 DOI: 10.1111/1751-7915.13465] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/05/2019] [Accepted: 07/05/2019] [Indexed: 12/31/2022] Open
Affiliation(s)
- Pinpunya Riangrungroj
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK.,Imperial College Centre for Synthetic Biology, Imperial College London, London, SW7 2AZ, UK
| | - Karen M Polizzi
- Imperial College Centre for Synthetic Biology, Imperial College London, London, SW7 2AZ, UK.,Department of Chemical Engineering, Imperial College London, London, SW7 2AZ, UK
| |
Collapse
|
343
|
Bidossi A, Bottagisio M, De Grandi R, Drago L, De Vecchi E. Chlorquinaldol, a topical agent for skin and wound infections: anti-biofilm activity and biofilm-related antimicrobial cross-resistance. Infect Drug Resist 2019; 12:2177-2189. [PMID: 31410037 PMCID: PMC6650094 DOI: 10.2147/idr.s211007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 06/14/2019] [Indexed: 01/10/2023] Open
Abstract
Purpose Persistence of skin and wound infections is nowadays accepted being linked to bacterial biofilms, which are highly recalcitrant to treatments and contribute to maintain a constant inflammation state and prevent a correct healing. Topical antimicrobials are the most common first-line self-medications; however, treatment failure is not uncommon and emerging resistance to antibiotics is alarming. Chlorquinaldol is an antimicrobial with a wide spectrum of activity and desirable characteristics for topical application. Aim of this study was to evaluate the efficacy of chlorquinaldol to prevent or eradicate S. aureus and P. aeruginosa biofilms, in comparison to classic topical antibiotics like gentamicin and fusidic acid. Methods Minimum inhibitory concentrations (MIC) were assessed for each strain and subinhibitory concentrations (½ and ¼ MIC) were used in the biofilm assay. Antimicrobial assays were performed during biofilm formation or were applied on mature biofilms and were evaluated by means of crystal violet assay and confocal laser scan microscopy. Results Chlorquinaldol and gentamicin were the most effective antimicrobials in both eradicating and preventing pathogens biofilm; however, resistance to methicillin and impermeability to carbapenems impaired chlorquinaldol effect. In addition, similarly to other hydroxyquinolines, aspecific metal chelation is here proposed as chlorquinaldol mode of action. Conclusion Relying on an acceptable antibiofilm and a wide spectrum of activity, an aspecific mode of action and consequent absence of resistance development, chlorquinaldol proved to be a good antimicrobial for topical use.
Collapse
Affiliation(s)
- Alessandro Bidossi
- Laboratory of Clinical Chemistry and Microbiology, IRCCS Orthopedic Institute Galeazzi, Milan, Italy
| | - Marta Bottagisio
- Laboratory of Clinical Chemistry and Microbiology, IRCCS Orthopedic Institute Galeazzi, Milan, Italy
| | - Roberta De Grandi
- Laboratory of Clinical Chemistry and Microbiology, IRCCS Orthopedic Institute Galeazzi, Milan, Italy
| | - Lorenzo Drago
- Laboratory of Clinical Microbiology, Department of Biomedical Science for Health, University of Milan, Milan, Italy
| | - Elena De Vecchi
- Laboratory of Clinical Chemistry and Microbiology, IRCCS Orthopedic Institute Galeazzi, Milan, Italy
| |
Collapse
|
344
|
Swanson T, Wolcott RD, Wallis H, Woodmansey EJ. Understanding biofilm in practice: a global survey of health professionals. J Wound Care 2019; 26:426-440. [PMID: 28795881 DOI: 10.12968/jowc.2017.26.8.426] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
OBJECTIVE The aim of this survey was to examine health professionals' views and practices relating to biofilm in chronic wounds. METHOD A global online survey was conducted to assess the current understanding of biofilm and wound management practices. The survey consisted of 20 questions designed to evaluate health professional knowledge of biofilm, perception and understanding of biofilm behaviour, detection and diagnosis, and treatment. Respondents were classified as 'specialists' if wounds were their primary focus and they developed protocols and determined formularies. Respondents were classified as 'generalists' if wounds were part of multiple indications they treat and they were able to choose wound care products from a restricted list of products. The Pearson's chi-square or Fisher's exact test was used to assess whether the responses were independent of the clinician role, health-care setting and country. RESULTS Overall, 3011 health professionals took part in the survey, of which 397 were excluded or disqualified. Of the remaining 2614 respondents, 1223 (46.8%) completed the entire survey. Although the majority of health professionals were aware of biofilm, knowledge gaps regarding its prevalence in chronic wounds were evident. In general, the majority indicated that they understood that biofilm is detrimental to wound healing. With the exception of wound stalling, there was a lack of consensus on other clinical signs in the detection and diagnosis of biofilm. Knowledge gaps were also evident over the treatment of biofilm and the efficacy of antimicrobial treatments, debridement and wound dressing. CONCLUSION Our results show that though there is a broad recognition of biofilm and its possible role in chronic wounds, there is still a need to educate and increase knowledge on recognition and treatment of biofilm.
Collapse
Affiliation(s)
- T Swanson
- Nurse Practitioner Wound Management, South West Healthcare, Ryot St. Warrnambool. Victoria 3280, Australia
| | - R D Wolcott
- Medical Director, Southwest Regional Wound Care Center, Lubbock, Texas, US
| | - H Wallis
- Senior Global Brand Development Manager, Smith & Nephew Ltd, Hull, UK
| | - E J Woodmansey
- Scientific Communications Manager, Smith & Nephew Ltd, Hull, UK
| |
Collapse
|
345
|
Mihai MM, Dima MB, Dima B, Holban AM. Nanomaterials for Wound Healing and Infection Control. MATERIALS (BASEL, SWITZERLAND) 2019; 12:E2176. [PMID: 31284587 PMCID: PMC6650835 DOI: 10.3390/ma12132176] [Citation(s) in RCA: 233] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 07/02/2019] [Accepted: 07/04/2019] [Indexed: 12/11/2022]
Abstract
Wound healing has been intensely studied in order to develop an "ideal" technique that achieves expeditious recovery and reduces scarring to the minimum, thus ensuring function preservation. The classic approach to wound management is represented by topical treatments, such as antibacterial or colloidal agents, in order to prevent infection and promote a proper wound-healing process. Nanotechnology studies submicroscopic particles (maximum diameter of 100 nm), as well as correlated phenomena. Metal nanoparticles (e.g., silver, gold, zinc) are increasingly being used in dermatology, due to their beneficial effect on accelerating wound healing, as well as treating and preventing bacterial infections. Other benefits include: ease of use, less frequent dressing changes and a constantly moist wound environment. This review highlights recent findings regarding nanoparticle application in wound management.
Collapse
Affiliation(s)
- Mara Madalina Mihai
- Dermavenereology Department, Emergency University Hospital "Elias", 011461 Bucharest, Romania
- Department of Oncologic Dermatology-Emergency University Hospital "Elias", University of Medicine and Pharmacy "Carol Davila", 020021 Bucharest, Romania
| | - Monica Beatrice Dima
- Dermavenereology Department, Emergency University Hospital "Elias", 011461 Bucharest, Romania
| | - Bogdan Dima
- Dermavenereology Department, Emergency University Hospital "Elias", 011461 Bucharest, Romania
| | - Alina Maria Holban
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, 030018 Bucharest, Romania.
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 1-7 Polizu Street, 011061 Bucharest, Romania.
| |
Collapse
|
346
|
Tribler S, Brandt CF, Fuglsang KA, Staun M, Broebech P, Moser CE, Scheike T, Jeppesen PB. Catheter-related bloodstream infections in patients with intestinal failure receiving home parenteral support: risks related to a catheter-salvage strategy. Am J Clin Nutr 2019; 107:743-753. [PMID: 29722835 DOI: 10.1093/ajcn/nqy010] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 01/10/2018] [Indexed: 01/27/2023] Open
Abstract
Background In intestinal failure (IF) patients receiving home parenteral support (HPS), catheter-related bloodstream infections (CRBSIs) frequently result in replacement of their tunneled central venous catheters (CVCs), which may lead to future loss of central venous access. Objective This observational study investigated the consequences of a catheter-salvage strategy related to CRBSIs. Design All CRBSIs from 2002 to 2016 in the Copenhagen IF and microbiological databases were retrospectively analyzed. Catheter salvage was defined by successful antimicrobial therapy with a retained CVC at discharge. Re-occurrences of CRBSIs with the same microbial species and identical antibiogram were defined as a relapse (<30 d) or as a recurrent (30-100 d) infection. Cox regression analyses incorporated a frailty factor to account for recurrent events and overrepresentation by some patients. Cumulative incidence curves are presented with a competing risk model. Results There were 2006 tunneled CVCs inserted in 715 adult HPS patients covering 2014.3 CVC years, with a CRBSI incidence rate of 1.83/1000 (n = 1350) and a mortality rate of 0.007/1000 CVC days (n = 5). The mean ± SD salvage rate was 55.3% ± 5.5%, varying according to infection type [monoinfections (62.9% ± 4.4%) and polyinfections (58.6% ± 17.3%)] and causative microorganism [coagulase-negative Staphylococci (CoNS) (68.1% ± 9.4%), Staphylococcus aureus (42.6% ± 17.5%), and Enterobacteriaceae (54.3% ± 16.7%)]. The overall risk of CRBSI relapse was 7.5%, and the risk of CRBSI recurrence was 7.3%. The HR for a subsequent CRBSI was 14% lower in a replaced than in a retained CVC (95% CI: 0.74, 0.99). The HR for a new CRBSI after catheter salvage was 36% higher after polyinfections than after monoinfections (95% CI: 1.03, 1.79). Enterobacteriaceae entailed an increased risk of CRBSI recurrence compared with CoNS (2.26; 95% CI; 1.08, 4.75) and S. aureus (4.45; 95% CI: 1.28, 15.5). Conclusions High catheter-salvage rates related to CRBSIs were achievable and safe in HPS patients within a broad range of microorganisms but contributed to an increased risk of CRBSI relapse or recurrence.
Collapse
Affiliation(s)
- Siri Tribler
- Departments of Medical Gastroenterology and Clinical Microbiology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Christopher F Brandt
- Departments of Medical Gastroenterology and Clinical Microbiology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Kristian A Fuglsang
- Departments of Medical Gastroenterology and Clinical Microbiology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Michael Staun
- Departments of Medical Gastroenterology and Clinical Microbiology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Per Broebech
- Departments of Medical Gastroenterology and Clinical Microbiology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Claus E Moser
- Clinical Microbiology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Thomas Scheike
- Department of Biostatistics, University of Copenhagen, Copenhagen, Denmark
| | - Palle B Jeppesen
- Departments of Medical Gastroenterology and Clinical Microbiology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
347
|
Mandell JB, Orr S, Koch J, Nourie B, Ma D, Bonar DD, Shah N, Urish KL. Large variations in clinical antibiotic activity against Staphylococcus aureus biofilms of periprosthetic joint infection isolates. J Orthop Res 2019; 37:1604-1609. [PMID: 30919513 PMCID: PMC7141781 DOI: 10.1002/jor.24291] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 03/02/2019] [Indexed: 02/04/2023]
Abstract
Staphylococcus aureus biofilms have a high tolerance to antibiotics, making the treatment of periprosthetic joint infection (PJI) challenging. From a clinical perspective, bacteria from surgical specimens are cultured in a planktonic state to determine antibiotic sensitivity. However, S. aureus exists primarily as established biofilms in PJI. To address this dichotomy, we developed a prospective registry of total knee and hip arthroplasty PJI S. aureus isolates to quantify the activity of clinically important antibiotics against isolates grown as biofilms. S. aureus planktonic minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) were assessed using clinical laboratory standard index assays for 10 antibiotics (cefazolin, clindamycin, vancomycin, rifampin, linezolid, nafcillin, gentamicin, trimethoprim/sulfamethoxazole, doxycycline, and daptomycin). Mature biofilms of each strain were grown in vitro, after which biofilm MIC (MBIC) and biofilm MBC (MBBC) were determined. Overall, isolates grown as biofilms displayed larger variations in antibiotic MICs as compared to planktonic MIC values. Only rifampin, doxycycline, and daptomycin had measurable biofilm MIC values across all S. aureus isolates tested. Biofilm MBC observations complemented biofilm MIC observations; rifampin, doxycycline, and daptomycin were the only antibiotics with measurable biofilm MBC values. 90% of S. aureus biofilms could be killed by rifampin, 50% by doxycycline, and only 15% by daptomycin. Biofilm formation increased bacterial antibiotic tolerance nonspecifically across all antibiotics, in both MSSA and MRSA samples. Rifampin and doxycycline were the most effective antibiotics at killing established S. aureus biofilms. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1604-1609, 2019.
Collapse
Affiliation(s)
- Jonathan B. Mandell
- Arthritis and Arthroplasty Design Group, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania,Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sara Orr
- Arthritis and Arthroplasty Design Group, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - John Koch
- Arthritis and Arthroplasty Design Group, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Blake Nourie
- Arthritis and Arthroplasty Design Group, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Dongzhu Ma
- Arthritis and Arthroplasty Design Group, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Daniel D. Bonar
- Department of Mathematics, Denison University, Granville, Ohio
| | - Neel Shah
- Division of Infectious Disease, Department of Internal Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Kenneth L. Urish
- Arthritis and Arthroplasty Design Group, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania,The Bone and Joint Center, Magee Womens Hospital of the University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania,Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania,Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, Pennsylvania,Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania
| |
Collapse
|
348
|
Crabbé A, Jensen PØ, Bjarnsholt T, Coenye T. Antimicrobial Tolerance and Metabolic Adaptations in Microbial Biofilms. Trends Microbiol 2019; 27:850-863. [PMID: 31178124 DOI: 10.1016/j.tim.2019.05.003] [Citation(s) in RCA: 167] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/25/2019] [Accepted: 05/07/2019] [Indexed: 01/04/2023]
Abstract
Active bacterial metabolism is a prerequisite for optimal activity of many classes of antibiotics. Hence, bacteria have developed strategies to reduce or modulate metabolic pathways to become tolerant. This review describes the tight relationship between metabolism and tolerance in bacterial biofilms, and how physicochemical properties of the microenvironment at the host-pathogen interface (such as oxygen and nutritional content) are key to this relationship. Understanding how metabolic adaptations lead to tolerance brings us to novel approaches to tackle antibiotic-tolerant biofilms. We describe the use of hyperbaric oxygen therapy, metabolism-stimulating metabolites, and alternative strategies to redirect bacterial metabolism towards an antibiotic-susceptible phenotype.
Collapse
Affiliation(s)
- Aurélie Crabbé
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| | - Peter Østrup Jensen
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark; Institute for Inflammation Research, Center for Rheumatology and Spine Diseases, Rigshospitalet, Copenhagen, Denmark
| | - Thomas Bjarnsholt
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
| | - Tom Coenye
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium.
| |
Collapse
|
349
|
Roche ED, Woodmansey EJ, Yang Q, Gibson DJ, Zhang H, Schultz GS. Cadexomer iodine effectively reduces bacterial biofilm in porcine wounds ex vivo and in vivo. Int Wound J 2019; 16:674-683. [PMID: 30868761 PMCID: PMC6850490 DOI: 10.1111/iwj.13080] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 01/04/2019] [Accepted: 01/08/2019] [Indexed: 12/25/2022] Open
Abstract
Biofilms are prevalent in non-healing chronic wounds and implicated in delayed healing. Tolerance to antimicrobial treatments and the host's immune system leave clinicians with limited interventions against biofilm populations. It is therefore essential that effective treatments be rigorously tested and demonstrate an impact on biofilm across multiple experimental models to guide clinical investigations and protocols. Cadexomer iodine has previously been shown to be effective against biofilm in various in vitro models, against methicillin-resistant Staphylococcus aureus biofilm in mouse wounds, and clinically in diabetic foot ulcers complicated by biofilm. Similarities between porcine and human skin make the pig a favoured model for cutaneous wound studies. Two antiseptic dressings and a gauze control were assessed against mature biofilm grown on ex vivo pig skin and in a pig wound model. Significant reductions in biofilm were observed following treatment with cadexomer iodine across both biofilm models. In contrast, silver carboxymethylcellulose dressings had minimal impact on biofilm in the models, with similar results to the control in the ex vivo model. Microscopy and histopathology indicate that the depth of organisms in wound tissue may impact treatment effectiveness. Further work on the promising biofilm efficacy of cadexomer iodine is needed to determine optimal treatment durations against biofilm.
Collapse
Affiliation(s)
- Eric D. Roche
- Advanced Wound Management R&D, Smith & NephewFort WorthTexas
| | - Emma J. Woodmansey
- Clinical, Scientific and Medical Affairs, Smith & NephewKingston upon HullUK
| | - Qingping Yang
- Department of Obstetrics and Gynecology, Institute for Wound ResearchUniversity of FloridaGainesvilleFlorida
| | - Daniel J. Gibson
- Department of Obstetrics and Gynecology, Institute for Wound ResearchUniversity of FloridaGainesvilleFlorida
| | - Hongen Zhang
- Department of Obstetrics and Gynecology, Institute for Wound ResearchUniversity of FloridaGainesvilleFlorida
| | - Gregory S. Schultz
- Department of Obstetrics and Gynecology, Institute for Wound ResearchUniversity of FloridaGainesvilleFlorida
| |
Collapse
|
350
|
Moser C, Lerche CJ, Thomsen K, Hartvig T, Schierbeck J, Jensen PØ, Ciofu O, Høiby N. Antibiotic therapy as personalized medicine - general considerations and complicating factors. APMIS 2019; 127:361-371. [PMID: 30983040 DOI: 10.1111/apm.12951] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 04/07/2019] [Indexed: 12/16/2022]
Abstract
The discovery of antibiotic drugs is considered one of the previous century's most important medical discoveries (Medicine's 10 greatest discoveries. New Haven, CT: Yale University Press, 1998: 263). Appropriate use of antibiotics saves millions of lives each year and prevents infectious complications for numerous people. Still, infections kill unacceptable many people around the world, even in developed countries with easy access to most antibiotic drugs. Optimal use of antibiotics is dependent on the identification of primary and secondary focus, and knowledge on which pathogens to expect in a specific infectious syndrome and information on general patterns of regional antibiotic resistance. Furthermore, sampling for microbiological analysis, knowledge of patient immune status and organ functions, travel history, pharmacokinetics and -dynamics of the different antibiotics and possible biofilm formation are among several factors involved in antibiotic therapy of infectious diseases. The present review aims at describing important considerations when using antibacterial antibiotics and to describe how this is becoming substantially more personalized. The parameters relevant in considering the optimal use of antibiotics to treat infections are shown in Fig. 1 - leading to the most relevant antibiotic therapy for that specific patient. To illustrate this subject, the present review's focus will be on challenges with optimal dosing of antibiotics and risks of underdosing. Especially, in cases highly challenging for achieving the aimed antibiotic effect against bacterial infections - this includes augmented renal clearance (ARC) in sepsis, dosing challenges of antibiotics in pregnancy and against biofilm infections.
Collapse
Affiliation(s)
- Claus Moser
- Department of Clinical Microbiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.,Department of Intensive Care, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Christian Johann Lerche
- Department of Clinical Microbiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Kim Thomsen
- Department of Clinical Microbiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Tom Hartvig
- Department of Intensive Care, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Jens Schierbeck
- Department of Anaesthesiology and Intensive Care, Odense University Hospital, Odense, Denmark
| | - Peter Østrup Jensen
- Department of Clinical Microbiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.,Department of Intensive Care, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Oana Ciofu
- Costerton Biofilm Center, Institute of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Niels Høiby
- Department of Clinical Microbiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.,Costerton Biofilm Center, Institute of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|