1
|
To D, Blanco Massani M, Coraça-Huber DC, Seybold A, Ricci F, Zöller K, Bernkop-Schnürch A. Antibiotic-Polyphosphate Nanocomplexes: A Promising System for Effective Biofilm Eradication. Int J Nanomedicine 2024; 19:9707-9725. [PMID: 39309185 PMCID: PMC11416784 DOI: 10.2147/ijn.s473241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 06/12/2024] [Indexed: 09/25/2024] Open
Abstract
Purpose The eradication of bacterial biofilms poses an enormous challenge owing to the inherently low antibiotic susceptibility of the resident microbiota. The complexation of antibiotics with polyphosphate can substantially improve antimicrobial performance. Methods Nanoparticular complexes of the model drug colistin and polyphosphate (CP-NPs) were developed and characterized in terms of their particle size and morphology, polydispersity index (PDI), zeta potential, and cytotoxicity. Enzyme-triggered monophosphate and colistin release from the CP-NPs was evaluated in the presence of alkaline phosphatase (AP). Subsequently, antimicrobial efficacy was assessed by inhibition experiments on planktonic cultures, as well as time-kill assays on biofilms formed by the model organism Micrococcus luteus. Results The CP-NPs exhibited a spherical morphology with particle sizes <200 nm, PDI <0.25, and negative zeta potential. They showed reduced cytotoxicity toward two human cell lines and significantly decreased hemotoxicity compared with native colistin. Release experiments with AP verified the enzymatic cleavage of polyphosphate and subsequent release of monophosphate and colistin from CP-NPs. Although CP-NPs were ineffective against planktonic M. luteus cultures, they showed major activity against bacterial biofilms, outperforming native colistin treatment. Strongly elevated AP levels in the biofilm state were identified as a potential key factor for the observed findings. Conclusion Accordingly, polyphosphate-based nanocomplexes represent a promising tool to tackle bacterial biofilm.
Collapse
Affiliation(s)
- Dennis To
- Center for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
| | - Mariana Blanco Massani
- Center for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
| | - Débora C Coraça-Huber
- Research Laboratory for Implant Associated Infections (BIOFILM LAB), Experimental Orthopaedics, University Hospital for Orthopaedics and Traumatology, Medical University Innsbruck, Innsbruck, Austria
| | - Anna Seybold
- Department of Zoology, University of Innsbruck, Innsbruck, Austria
| | - Fabrizio Ricci
- Center for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
- Thiomatrix Forschungs- und Beratungs GmbH, Innsbruck, Austria
| | - Katrin Zöller
- Center for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
| | - Andreas Bernkop-Schnürch
- Center for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
2
|
Ferreira L, Pos E, Nogueira DR, Ferreira FP, Sousa R, Abreu MA. Antibiotics with antibiofilm activity - rifampicin and beyond. Front Microbiol 2024; 15:1435720. [PMID: 39268543 PMCID: PMC11391936 DOI: 10.3389/fmicb.2024.1435720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 08/07/2024] [Indexed: 09/15/2024] Open
Abstract
The management of prosthetic joint infections is a complex and multilayered process that is additionally complicated by the formation of bacterial biofilm. Foreign material provides the ideal grounds for the development of an intricate matrix that hinders treatment and creates a difficult environment for antibiotics to act. Surgical intervention is often warranted but requires appropriate adjunctive therapy. Despite available guidelines, several aspects of antibiotic therapy with antibiofilm activity lack clear definition. Given the escalating challenges posed by antimicrobial resistance, extended treatment durations, and tolerance issues, it is essential to ensure that antimicrobials with antibiofilm activity are both potent and diverse. Evidence of biofilm-active drugs is highlighted, and alternatives to classical regimens are further discussed.
Collapse
Affiliation(s)
- Luís Ferreira
- Department of Infectious Diseases, Centro Hospitalar e Universitário de Santo António, Porto, Portugal
| | - Ema Pos
- Department of Infectious Diseases, Centro Hospitalar e Universitário de Santo António, Porto, Portugal
| | | | - Filipa Pinto Ferreira
- Department of Infectious Diseases, Centro Hospitalar e Universitário de Santo António, Porto, Portugal
| | - Ricardo Sousa
- Department of Orthopaedic Surgery, Centro Hospitalar e Universitário de Santo António, Porto, Portugal
- Grupo de Infeção Osteoarticular do Porto, Porto, Portugal
| | - Miguel Araújo Abreu
- Department of Infectious Diseases, Centro Hospitalar e Universitário de Santo António, Porto, Portugal
- Grupo de Infeção Osteoarticular do Porto, Porto, Portugal
| |
Collapse
|
3
|
Hale SJM, Cameron AJ, Lux CA, Biswas K, Kim R, O'Carroll M, Harris PWR, Douglas RG, Wagner Mackenzie B. Polymyxin B and ethylenediaminetetraacetic acid act synergistically against Pseudomonas aeruginosa and Staphylococcus aureus. Microbiol Spectr 2024; 12:e0170923. [PMID: 38168683 PMCID: PMC10845947 DOI: 10.1128/spectrum.01709-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 10/05/2023] [Indexed: 01/05/2024] Open
Abstract
Polymyxin B and ethylenediaminetetraacetic acid are antimicrobials possessing antibiofilm activity. They act by displacement and chelation, respectively, of divalent cations in bacterial membranes and may therefore act synergistically when applied in combination. If so, this combination of agents may be useful for the treatment of diseases like cystic fibrosis (CF), in which biofilms are present on the respiratory epithelium. We used checkerboard assays to investigate the synergy between these agents using reference strains Pseudomonas aeruginosa ATCC 27853 and Staphylococcus aureus ATCC 6538 in planktonic form. We then determined the efficacy of each agent against biofilms of both species grown on 96-pin lids and proceeded to combination testing against the P. aeruginosa reference strain and 10 clinical isolates from patients with CF. Synergism was observed for planktonic forms of both species and for biofilms of P. aeruginosa. The susceptibility of biofilms of P. aeruginosa clinical isolates to these agents was variable compared to the laboratory reference strain. This combination of agents may be useful in the management of biofilm-associated conditions, particularly those amenable to topical therapies. These results provide a basis upon which the antimicrobial and antibiofilm efficacy of preparations containing these agents may be enhanced.IMPORTANCEBacteria living in biofilms produce a protective matrix which makes them difficult to kill. Patients with severe respiratory disease often have biofilms. Polymyxin B is an antibiotic commonly used in topical medications, such as eye drops and nasal sprays. Ethylenediaminetetraacetic acid (EDTA) is used widely as a preservative in medication but also has antimicrobial properties. It has been hypothesized that Polymyxin B and EDTA could have a synergistic relationship: when used in combination their antimicrobial effect is enhanced. Here, we evaluated the levels at which Polymyxin B and EDTA work together to kill common pathogens Pseudomonas aeruginosa and Staphylococcus aureus. We found that Polymyxin B and EDTA were synergistic. This synergy may be useful in the management of planktonic infection with P. aeruginosa and S. aureus, or biofilm infection with P. aeruginosa. This synergy may be beneficial in the treatment of respiratory biofilms, in which P. aeruginosa biofilms are common.
Collapse
Affiliation(s)
- Samuel J M Hale
- Department of Surgery, Faculty of Medical and Health Sciences, The University of Auckland, Grafton, Auckland, New Zealand
| | - Alan J Cameron
- School of Chemical Sciences and School of Biological Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Christian A Lux
- Department of Surgery, Faculty of Medical and Health Sciences, The University of Auckland, Grafton, Auckland, New Zealand
| | - Kristi Biswas
- Department of Surgery, Faculty of Medical and Health Sciences, The University of Auckland, Grafton, Auckland, New Zealand
| | - Raymond Kim
- Department of Surgery, Faculty of Medical and Health Sciences, The University of Auckland, Grafton, Auckland, New Zealand
| | - Mark O'Carroll
- Respiratory Services, Auckland City Hospital, Te Toka Tumai, Te Whatu Ora, Auckland, New Zealand
| | - Paul W R Harris
- School of Chemical Sciences and School of Biological Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Richard G Douglas
- Department of Surgery, Faculty of Medical and Health Sciences, The University of Auckland, Grafton, Auckland, New Zealand
| | - Brett Wagner Mackenzie
- Department of Surgery, Faculty of Medical and Health Sciences, The University of Auckland, Grafton, Auckland, New Zealand
| |
Collapse
|
4
|
Dsouza FP, Dinesh S, Sharma S. Understanding the intricacies of microbial biofilm formation and its endurance in chronic infections: a key to advancing biofilm-targeted therapeutic strategies. Arch Microbiol 2024; 206:85. [PMID: 38300317 DOI: 10.1007/s00203-023-03802-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/04/2023] [Accepted: 12/16/2023] [Indexed: 02/02/2024]
Abstract
Bacterial biofilms can adhere to various surfaces in the environment with human beings being no exception. Enclosed in a self-secreted matrix which contains extracellular polymeric substances, biofilms are intricate communities of bacteria that play a significant role across various sectors and raise concerns for public health, medicine and industries. These complex structures allow free-floating planktonic cells to adopt multicellular mode of growth which leads to persistent infections. This is of great concern as biofilms can withstand external attacks which include antibiotics and immune responses. A more comprehensive and innovative approach to therapy is needed in view of the increasing issue of bacterial resistance brought on by the overuse of conventional antimicrobial medications. Thus, to oppose the challenges posed by biofilm-related infections, innovative therapeutic strategies are being explored which include targeting extracellular polymeric substances, quorum sensing, and persister cells. Biofilm-responsive nanoparticles show promising results by improving drug delivery and reducing the side effects. This review comprehensively examines the factors influencing biofilm formation, host immune defence mechanisms, infections caused by biofilms, diagnostic approaches, and biofilm-targeted therapies.
Collapse
Affiliation(s)
| | - Susha Dinesh
- Department of Bioinformatics, BioNome, Bengaluru, Karnataka, 560043, India.
| | - Sameer Sharma
- Department of Bioinformatics, BioNome, Bengaluru, Karnataka, 560043, India
| |
Collapse
|
5
|
Bayatinejad G, Salehi M, Beigverdi R, Halimi S, Emaneini M, Jabalameli F. In Vitro antibiotic combinations of Colistin, Meropenem, Amikacin, and Amoxicillin/clavulanate against multidrug-resistant Klebsiella pneumonia isolated from patients with ventilator-associated pneumonia. BMC Microbiol 2023; 23:298. [PMID: 37864176 PMCID: PMC10588070 DOI: 10.1186/s12866-023-03039-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 10/03/2023] [Indexed: 10/22/2023] Open
Abstract
BACKGROUND Hospital infections such as ventilator-associated pneumonia (VAP) due to multidrug-resistant Klebsiella pneumoniae (MDR-KP) strains have increased worldwide. In addition, biofilm production by these resistant isolates has confronted clinicians with higher treatment failure and infection recurrence. Given the paucity of new agents and limited data on combination therapy for MDR-KPs, the present study sought to evaluate the in vitro activity of several antibiotic combinations against planktonic and biofilm MDR-KPs isolated from patients with VAP. RESULTS All 10 carbapenem-resistant Klebsiella pneumoniae (CRKP) isolates demonstrated multidrug resistance against the tested antibiotics. At planktonic mode, combinations of colistin-meropenem and amoxicillin/clavulanate in combination with meropenem, colistin, or amikacin showed synergism against 60-70% isolates. On the other hand, in the biofilm state, colistin-based combinations exhibited synergism against 50-70% isolates and the most effective combination was colistin-amikacin with 70% synergy. CONCLUSIONS The results revealed that combinations of amoxicillin/clavulanate with colistin, meropenem, or amikacin in the planktonic mode and colistin with amoxicillin/clavulanate, meropenem, or amikacin in the biofilm mode could effectively inhibit CRKP isolates, and thus could be further explored for the treatment of CRKPs.
Collapse
Affiliation(s)
- Ghazal Bayatinejad
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Salehi
- Department of Infectious Diseases and Tropical Medicine, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Antibiotic Stewardship and Antimicrobial Resistance, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Beigverdi
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahnaz Halimi
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Emaneini
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fereshteh Jabalameli
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Research Center for Antibiotic Stewardship and Antimicrobial Resistance, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Aguilera-Correa JJ, Gisbert-Garzarán M, Mediero A, Fernández-Aceñero MJ, de-Pablo-Velasco D, Lozano D, Esteban J, Vallet-Regí M. Antibiotic delivery from bone-targeted mesoporous silica nanoparticles for the treatment of osteomyelitis caused by methicillin-resistant Staphylococcus aureus. Acta Biomater 2022; 154:608-625. [PMID: 36341887 DOI: 10.1016/j.actbio.2022.10.039] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 10/13/2022] [Accepted: 10/19/2022] [Indexed: 12/14/2022]
Abstract
Osteomyelitis is a hard-to-treat infection of the bone and bone marrow that is mainly caused by Staphylococcus aureus, with an increasing incidence of methicillin-resistant S. aureus (MRSA). Owing to the aggressiveness of these bacteria in colonizing and destroying the bone, systemic antibiotic treatments fail to eradicate the infection. Instead, it normally entails surgery to remove the dead or infected bone. In this work, we report bone-targeted mesoporous silica nanoparticles for the treatment of osteomyelitis. The nanoparticles have been engineered with a functional gelatine/colistin coating able to hamper premature release from the mesopores while effectively disaggregating the bacterial biofilm. Because antibiotic resistance is a global emergency, we have designed two sets of identical nanoparticles, carrying each of them a clinically relevant antibiotic, that have demonstrated to have synergistic effect. The bone-targeted nanoparticles have been thoroughly evaluated in vitro and in vivo, obtaining a notable reduction of the amount of bacteria in the bone in just 24 h after only one dose, and paving the way for localized, nanoparticle-mediated treatment of MRSA-caused osteomyelitis. STATEMENT OF SIGNIFICANCE: In this work, we propose the use of bone-targeted mesoporous silica nanoparticles to address S. aureus-caused osteomyelitis that render synergistic therapeutic effect via multidrug delivery. Because the bacterial biofilm is responsible for an aggressive surgical approach and prolonged antibiotic treatment, the nanoparticles have been functionalized with a functional coating able to both disaggregate the biofilm, hamper premature antibiotic release and protect the intact bone. These engineered nanoparticles are able to effectively target bone tissue both in vitro and in vivo, showing high biocompatibility and elevated antibacterial effect.
Collapse
Affiliation(s)
- J J Aguilera-Correa
- Departamento de Química en Ciencias Farmacéuticas, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain; CIBER de Enfermedades Infecciosas (CIBERINFEC), 28029 Madrid, Spain
| | - M Gisbert-Garzarán
- Departamento de Química en Ciencias Farmacéuticas, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
| | - A Mediero
- Bone and Joint Unit, IIS- Fundación Jimenez Diaz, UAM, Avenida Reyes Católicos, 2 28037 Madrid, Spain
| | | | | | - D Lozano
- Departamento de Química en Ciencias Farmacéuticas, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
| | - J Esteban
- CIBER de Enfermedades Infecciosas (CIBERINFEC), 28029 Madrid, Spain; Clinical Microbiology Department, IIS-Fundación Jiménez Diaz, UAM, Avenida Reyes Católicos, 2 28037 Madrid, Spain.
| | - M Vallet-Regí
- Departamento de Química en Ciencias Farmacéuticas, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain.
| |
Collapse
|
7
|
Noreen A, Masood H, Zaib J, Rafaque Z, Fatima A, Shabbir H, Alam J, Habib A, Noor S, Dil K, Dasti JI. Investigating the Role of Antibiotics on Induction, Inhibition and Eradication of Biofilms of Poultry Associated Escherichia coli Isolated from Retail Chicken Meat. Antibiotics (Basel) 2022; 11:antibiotics11111663. [PMID: 36421307 PMCID: PMC9686770 DOI: 10.3390/antibiotics11111663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/03/2022] [Accepted: 11/07/2022] [Indexed: 11/22/2022] Open
Abstract
Background: Widespread use of antibiotics as growth promoters and prophylactic agents has dramatic consequences for the development of antibiotic resistance. In this study, we investigated effects of selected antibiotics on bacterial biofilms and performed extensive antibiotic and VF profiling of poultry-meat associated E. coli strains. Methods: Antibiotic susceptibility was performed by a disc diffusion method, followed by molecular screening of resistance and virulence determinants. Further biofilm formation assays, MIC-p, MIC-b, MBIC and MBEC, were performed using standard tissue culture plate method. Results: In total, 83 (75%) samples were confirmed as E. coli from poultry sources, 26 different antibiotics were tested, and maximum numbers of the isolates were resistant to lincomycin (100%), while the least resistance was seen against cefotaxime (1%) and polymyxin B (1%). Overall, 48% of the isolates were ESBL producers and 40% showed carbapenemase activity; important virulence genes were detected in following percentages: fimH32 (39%), papC21 (25%), iutA34 (41%), kpsMT-II23 (28%), papEF9 (11%), papGII22 (27%) and fyuA13 (16%). Colistin showed remarkable anti-biofilm activity, while at sub-MIC levels, gentamicin, ceftriaxone and enrofloxin significantly (p < 0.01) inhibited the biofilms. A strong induction of bacterial biofilm, after exposure to sub-minimal levels of colistin clearly indicates risk of bacterial overgrowth in a farm environment, while use of colistin aggravates the risk of emergence of colistin resistant Enterobacteriaceae, a highly undesirable public health scenario.
Collapse
Affiliation(s)
- Aisha Noreen
- Lab of Microbial Genomics and Epidemiology, Department of Microbiology, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Hamid Masood
- Lab of Microbial Genomics and Epidemiology, Department of Microbiology, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Jaweria Zaib
- Lab of Microbial Genomics and Epidemiology, Department of Microbiology, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Zara Rafaque
- Department of Microbiology, Hazara University, Mansehra 21120, Pakistan
| | - Areeta Fatima
- Lab of Microbial Genomics and Epidemiology, Department of Microbiology, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Hira Shabbir
- Lab of Microbial Genomics and Epidemiology, Department of Microbiology, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Javaria Alam
- Lab of Microbial Genomics and Epidemiology, Department of Microbiology, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Aisha Habib
- Lab of Microbial Genomics and Epidemiology, Department of Microbiology, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Saba Noor
- Lab of Microbial Genomics and Epidemiology, Department of Microbiology, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Kinza Dil
- Lab of Microbial Genomics and Epidemiology, Department of Microbiology, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Javid Iqbal Dasti
- Lab of Microbial Genomics and Epidemiology, Department of Microbiology, Quaid-i-Azam University, Islamabad 45320, Pakistan
- Correspondence:
| |
Collapse
|
8
|
Krukiewicz K, Kazek-Kęsik A, Brzychczy-Włoch M, Łos MJ, Ateba CN, Mehrbod P, Ghavami S, Shyntum DY. Recent Advances in the Control of Clinically Important Biofilms. Int J Mol Sci 2022; 23:9526. [PMID: 36076921 PMCID: PMC9455909 DOI: 10.3390/ijms23179526] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 08/17/2022] [Accepted: 08/20/2022] [Indexed: 11/16/2022] Open
Abstract
Biofilms are complex structures formed by bacteria, fungi, or even viruses on biotic and abiotic surfaces, and they can be found in almost any part of the human body. The prevalence of biofilm-associated diseases has increased in recent years, mainly because of the frequent use of indwelling medical devices that create opportunities for clinically important bacteria and fungi to form biofilms either on the device or on the neighboring tissues. As a result of their resistance to antibiotics and host immunity factors, biofilms have been associated with the development or persistence of several clinically important diseases. The inability to completely eradicate biofilms drastically increases the burden of disease on both the patient and the healthcare system. Therefore, it is crucial to develop innovative ways to tackle the growth and development of biofilms. This review focuses on dental- and implant-associated biofilm infections, their prevalence in humans, and potential therapeutic intervention strategies, including the recent advances in pharmacology and biomedical engineering. It lists current strategies used to control the formation of clinically important biofilms, including novel antibiotics and their carriers, antiseptics and disinfectants, small molecule anti-biofilm agents, surface treatment strategies, and nanostructure functionalization, as well as multifunctional coatings particularly suitable for providing antibacterial effects to the surface of implants, to treat either dental- or implant-related bacterial infections.
Collapse
Affiliation(s)
- Katarzyna Krukiewicz
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, M. Strzody 9, 44-100 Gliwice, Poland
- Centre for Organic and Nanohybrid Electronics, Silesian University of Technology, Konarskiego 22B, 44-100 Gliwice, Poland
| | - Alicja Kazek-Kęsik
- Department of Inorganic Chemistry, Analytical Chemistry and Electrochemistry, Silesian University of Technology, 44-100 Gliwice, Poland
- Biotechnology Centre, Silesian University of Technology, Krzywoustego 8 Street, 44-100 Gliwice, Poland
| | - Monika Brzychczy-Włoch
- Department of Molecular Medical Microbiology, Chair of Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Czysta 18 Street, 31-121 Krakow, Poland
| | - Marek J. Łos
- Department of Pathology, Pomeranian Medical University, 71-344 Szczecin, Poland
| | - Collins Njie Ateba
- Food Security and Safety Niche Area, North West University, Private Bag X2046, Mahikeng 2735, South Africa
| | - Parvaneh Mehrbod
- Influenza and Respiratory Viruses Department, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Saeid Ghavami
- Faculty of Medicine in Zabrze, University of Technology in Katowice, Academia of Silesia, 41-800 Zabrze, Poland
- Research Institute of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, MB R3E 3P5, Canada
- Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 3P5, Canada
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 3P5, Canada
| | - Divine Yufetar Shyntum
- Biotechnology Centre, Silesian University of Technology, Krzywoustego 8 Street, 44-100 Gliwice, Poland
| |
Collapse
|
9
|
Hale SJM, Wagner Mackenzie B, Lux CA, Biswas K, Kim R, Douglas RG. Topical Antibiofilm Agents With Potential Utility in the Treatment of Chronic Rhinosinusitis: A Narrative Review. Front Pharmacol 2022; 13:840323. [PMID: 35770097 PMCID: PMC9234399 DOI: 10.3389/fphar.2022.840323] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/24/2022] [Indexed: 11/13/2022] Open
Abstract
The role of bacterial biofilms in chronic and recalcitrant diseases is widely appreciated, and the treatment of biofilm infection is an increasingly important area of research. Chronic rhinosinusitis (CRS) is a complex disease associated with sinonasal dysbiosis and the presence of bacterial biofilms. While most biofilm-related diseases are associated with highly persistent but relatively less severe inflammation, the presence of biofilms in CRS is associated with greater severity of inflammation and recalcitrance despite appropriate treatment. Oral antibiotics are commonly used to treat CRS but they are often ineffective, due to poor penetration of the sinonasal mucosa and the inherently antibiotic resistant nature of bacteria in biofilms. Topical non-antibiotic antibiofilm agents may prove more effective, but few such agents are available for sinonasal application. We review compounds with antibiofilm activity that may be useful for treating biofilm-associated CRS, including halogen-based compounds, quaternary ammonium compounds and derivatives, biguanides, antimicrobial peptides, chelating agents and natural products. These include preparations that are currently available and those still in development. For each compound, antibiofilm efficacy, mechanism of action, and toxicity as it relates to sinonasal application are summarised. We highlight the antibiofilm agents that we believe hold the greatest promise for the treatment of biofilm-associated CRS in order to inform future research on the management of this difficult condition.
Collapse
Affiliation(s)
- Samuel J M Hale
- Department of Surgery, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Brett Wagner Mackenzie
- Department of Surgery, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Christian A Lux
- Department of Surgery, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Kristi Biswas
- Department of Surgery, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Raymond Kim
- Department of Surgery, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Richard G Douglas
- Department of Surgery, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
10
|
Robin B, Nicol M, Le H, Tahrioui A, Schaumann A, Vuillemenot JB, Vergoz D, Lesouhaitier O, Jouenne T, Hardouin J, Potron A, Perrot V, Dé E. MacAB-TolC Contributes to the Development of Acinetobacter baumannii Biofilm at the Solid–Liquid Interface. Front Microbiol 2022; 12:785161. [PMID: 35095797 PMCID: PMC8792954 DOI: 10.3389/fmicb.2021.785161] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 11/22/2021] [Indexed: 12/14/2022] Open
Abstract
Acinetobacter baumannii has emerged as one of the most problematic bacterial pathogens responsible for hospital-acquired and community infections worldwide. Besides its high capacity to acquire antibiotic resistance mechanisms, it also presents high adhesion abilities on inert and living surfaces leading to biofilm development. This lifestyle confers additional protection against various treatments and allows it to persist for long periods in various hospital niches. Due to their remarkable antimicrobial tolerance, A. baumannii biofilms are difficult to control and ultimately eradicate. Further insights into the mechanism of biofilm development will help to overcome this challenge and to develop novel antibiofilm strategies. To unravel critical determinants of this sessile lifestyle, the proteomic profiles of two A. baumannii strains (ATTC17978 and SDF) grown in planktonic stationary phase or in mature solid–liquid (S-L) biofilm were compared using a semiquantitative proteomic study. Of interest, among the 69 common proteins determinants accumulated in the two strains at the S-L interface, we sorted out the MacAB-TolC system. This tripartite efflux pump played a role in A. baumannii biofilm formation as demonstrated by using ΔmacAB-tolC deletion mutant. Complementary approaches allowed us to get an overview of the impact of macAB-tolC deletion in A. baumannii physiology. Indeed, this efflux pump appeared to be involved in the envelope stress response occurring in mature biofilm. It contributes to maintain wild type (WT) membrane rigidity and provides tolerance to high osmolarity conditions. In addition, this system is probably involved in the maintenance of iron and sulfur homeostasis. MacAB-TolC might help this pathogen face and adapt to deleterious conditions occurring in mature biofilms. Increasing our knowledge of A. baumannii biofilm formation will undoubtedly help us develop new therapeutic strategies to tackle this emerging threat to human health.
Collapse
Affiliation(s)
- Brandon Robin
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, Polymers, Biopolymers, Surfaces Laboratory, Rouen, France
| | - Marion Nicol
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, Polymers, Biopolymers, Surfaces Laboratory, Rouen, France
| | - Hung Le
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, Polymers, Biopolymers, Surfaces Laboratory, Rouen, France
| | - Ali Tahrioui
- Normandie Univ, UNIROUEN, LMSM EA4312, Evreux, France
| | - Annick Schaumann
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, Polymers, Biopolymers, Surfaces Laboratory, Rouen, France
- PISSARO Proteomic Facility, IRIB, Mont-Saint-Aignan, France
| | | | - Delphine Vergoz
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, Polymers, Biopolymers, Surfaces Laboratory, Rouen, France
| | | | - Thierry Jouenne
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, Polymers, Biopolymers, Surfaces Laboratory, Rouen, France
- PISSARO Proteomic Facility, IRIB, Mont-Saint-Aignan, France
| | - Julie Hardouin
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, Polymers, Biopolymers, Surfaces Laboratory, Rouen, France
- PISSARO Proteomic Facility, IRIB, Mont-Saint-Aignan, France
| | - Anaïs Potron
- UMR 6249 Chrono-Environnement, CNRS-Université de Bourgogne/Franche-Comté, Besançon, France
| | - Valérie Perrot
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, Polymers, Biopolymers, Surfaces Laboratory, Rouen, France
- *Correspondence: Valérie Perrot,
| | - Emmanuelle Dé
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, Polymers, Biopolymers, Surfaces Laboratory, Rouen, France
- Emmanuelle Dé,
| |
Collapse
|
11
|
Aguilera-Correa J, Gisbert-Garzarán M, Mediero A, Carias-Cálix R, Jiménez-Jiménez C, Esteban J, Vallet-Regí M. Arabic gum plus colistin coated moxifloxacin-loaded nanoparticles for the treatment of bone infection caused by Escherichia coli. Acta Biomater 2022; 137:218-237. [PMID: 34653694 DOI: 10.1016/j.actbio.2021.10.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/20/2021] [Accepted: 10/07/2021] [Indexed: 12/16/2022]
Abstract
Osteomyelitis is an inflammatory process of bone and bone marrow that may even lead to patient death. Even though this disease is mainly caused by Gram-positive organisms, the proportion of bone infections caused by Gram-negative bacteria, such as Escherichia coli, has significantly increased in recent years. In this work, mesoporous silica nanoparticles have been employed as platform to engineer a nanomedicine able to eradicate E. coli- related bone infections. For that purpose, the nanoparticles have been loaded with moxifloxacin and further functionalized with Arabic gum and colistin (AG+CO-coated MX-loaded MSNs). The nanosystem demonstrated high affinity toward E. coli biofilm matrix, thanks to AG coating, and marked antibacterial effect because of the bactericidal effect of moxifloxacin and the disaggregating effect of colistin. AG+CO-coated MX-loaded MSNs were able to eradicate the infection developed on a trabecular bone in vitro and showed pronounced antibacterial efficacy in vivo against an osteomyelitis provoked by E. coli. Furthermore, AG+CO-coated MX-loaded MSNs were shown to be essentially non-cytotoxic with only slight effect on cell proliferation and mild hepatotoxicity, which might be attributed to the nature of both antibiotics. In view of these results, these nanoparticles may be considered as a promising treatment for bone infections caused by enterobacteria, such as E. coli, and introduce a general strategy against bone infections based on the implementation of antibiotics with different but complementary activity into a single nanocarrier. STATEMENT OF SIGNIFICANCE: In this work, we propose a methodology to address E.coli bone infections by using moxifloxacin-loaded mesoporous silica nanoparticles coated with Arabic gum containing colistin (AG+CO-coated MX-loaded MSNs). The in vitro evaluation of this nanosystem demonstrated high affinity toward E. coli biofilm matrix thanks to the Arabic gum coating, a disaggregating and antibacterial effect of colistin, and a remarkable antibiofilm action because of the bactericidal ability of moxifloxacin and colistin. This anti-E. coli capacity of AG+CO-coated MX-loaded MSNs was brought out in an in vivo rabbit model of osteomyelitis where the nanosystem was able to eradicate more than 90% of the bacterial load within the infected bone.
Collapse
|
12
|
Wang L, Lv S, Wang X, Liu B, Wang Z. Ferrate (VI) Oxidation Is an Effective and Safe Way to Degrade Residual Colistin - a Last Resort Antibiotic - in Wastewater. Front Vet Sci 2021; 8:773089. [PMID: 35004924 PMCID: PMC8739907 DOI: 10.3389/fvets.2021.773089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 12/06/2021] [Indexed: 11/13/2022] Open
Abstract
The rise of novel mcr mobile resistance genes seriously threatens the use of colistin as a last resort antibiotic for treatment of multidrug-resistant Gram-negative bacterial infections in humans. Large quantities of colistin are released annually into the environment through animal feces. This leads to environmental toxicity and promotes horizontal transmission of the mcr gene in aqueous environments. We examined colistin degradation catalyzed by the presence of strong oxidant Fe (VI). We found almost complete colistin degradation (>95%) by Fe (VI) at initial colistin levels of 30 μM at a molar ratio of Fe (VI): colistin of 30 using an initial pH 7.0 at 25°C for 60 min. The presence of humic acid did not alter the degradation rate and had no significant impact on the removal of colistin by Fe (VI). Quantitative microbiological assays of Fe (VI)-treated colistin solutions using Escherichia coli, Staphylococcus aureus, and Bacillus subtilis indicated that the residual antibacterial activity was effectively eliminated by Fe (VI) oxidation. Luminescent bacteria toxicity tests using Vibrio fischeri indicated that both colistin and its degradation products in water were of low toxicity and the products showed decreased toxicity compared to the parent drug. Therefore, Fe (VI) oxidation is a highly effective and environment-friendly strategy to degrade colistin in water.
Collapse
Affiliation(s)
- Liqi Wang
- Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, China
| | - Shiming Lv
- Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, China
| | - Xiaoying Wang
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Baosheng Liu
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Zhong Wang
- Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, China
| |
Collapse
|
13
|
Nguyen TK, Peyrusson F, Siala W, Pham NH, Nguyen HA, Tulkens PM, Van Bambeke F. Activity of Moxifloxacin Against Biofilms Formed by Clinical Isolates of Staphylococcus aureus Differing by Their Resistant or Persister Character to Fluoroquinolones. Front Microbiol 2021; 12:785573. [PMID: 34975808 PMCID: PMC8715871 DOI: 10.3389/fmicb.2021.785573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 11/19/2021] [Indexed: 11/13/2022] Open
Abstract
Staphylococcus aureus biofilms are poorly responsive to antibiotics. Underlying reasons include a matrix effect preventing drug access to embedded bacteria, or the presence of dormant bacteria with reduced growth rate. Using 18 clinical isolates previously characterized for their moxifloxacin-resistant and moxifloxacin-persister character in stationary-phase culture, we studied their biofilm production and matrix composition and the anti-biofilm activity of moxifloxacin. Biofilms were grown in microtiter plates and their abundance quantified by crystal violet staining and colony counting; their content in polysaccharides, extracellular DNA and proteins was measured. Moxifloxacin activity was assessed after 24 h of incubation with a broad range of concentrations to establish full concentration-response curves. All clinical isolates produced more biofilm biomass than the reference strain ATCC 25923, the difference being more important for those with high relative persister fractions to moxifloxacin, most of which being also resistant. High biofilm producers expressed icaA to higher levels, enriching the matrix in polysaccharides. Moxifloxacin was less potent against biofilms from clinical isolates than from ATCC 25923, especially against moxifloxacin-resistant isolates with high persister fractions, which was ascribed to a lower concentration of moxifloxacin in these biofilms. Time-kill curves in biofilms revealed the presence of a moxifloxacin-tolerant subpopulation, with low multiplication capacity, whatever the persister character of the isolate. Thus, moxifloxacin activity depends on its local concentration in biofilm, which is reduced in most isolates with high-relative persister fractions due to matrix effects, and insufficient to kill resistant isolates due to their high MIC.
Collapse
Affiliation(s)
- Tiep K. Nguyen
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
- Department of Pharmaceutical Industry, Hanoi University of Pharmacy, Hanoi, Vietnam
| | - Frédéric Peyrusson
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Wafi Siala
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Nhung H. Pham
- Department of Microbiology, Bach Mai Hospital, Hanoi, Vietnam
| | - Hoang A. Nguyen
- The National Center for Drug Information and Adverse Drug Reactions Monitoring, Hanoi University of Pharmacy, Hanoi, Vietnam
| | - Paul M. Tulkens
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Françoise Van Bambeke
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
- *Correspondence: Françoise Van Bambeke,
| |
Collapse
|
14
|
Park NH, Lee SJ, Lee EB, Birhanu BT, Park SC. Colistin Induces Resistance through Biofilm Formation, via Increased phoQ Expression, in Avian Pathogenic Escherichia coli. Pathogens 2021; 10:pathogens10111525. [PMID: 34832681 PMCID: PMC8620993 DOI: 10.3390/pathogens10111525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/12/2021] [Accepted: 11/17/2021] [Indexed: 11/16/2022] Open
Abstract
This study aimed to optimize the colistin-based antibacterial therapy to prevent antimicrobial resistance related to biofilm formation in avian pathogenic Escherichia coli (APEC) in chicken. Of all the bacterial isolates (n = 136), 69 were identified as APEC by polymerase chain reaction (PCR). Through a series of antibiotic susceptibility tests, susceptibility to colistin (<2 μg/mL) was confirmed in all isolates. Hence, a mutant selection window (MSW) was determined to obtain colistin-induced resistant bacteria. The minimum inhibitory concentration (MIC) of colistin against the colistin-induced resistant APEC strains ranged from 8 to 16 μg/mL. To identify the inhibitory activity of colistin against the resistant strains, the mutant prevention concentration (MPC) was investigated for 72 h, and the single and multi-dose colistin activities were determined through the time-kill curve against APEC strains. Bacterial regrowth occurred after 12 h at a double MIC50 concentration (1.00 μg/mL), and regrowth was not inhibited even during multiple exposures. However, upon exposure to 8 μg/mL—a concentration that was close to the MPC—the growth of APEC was inhibited, including in the resistant strains. Additionally, colistin-induced resistant strains showed a slower growth compared with the susceptible ones. Colistin-induced resistant APEC strains did not show colistin resistance gene (mcr-1). However, the expression of higher mgrB and phoQ levels was observed in the resistant strains. Furthermore, these strains showed increased formation of biofilm. Hence, the present study indicated that colistin could induce resistance through the increased formation of biofilm in APEC strains by enhancing the expression of phoQ.
Collapse
Affiliation(s)
- Na-Hye Park
- Laboratory Animal Center, Daegu Gyeongbuk Medical Innovation Foundation, Daegu 41061, Korea;
| | - Seung-Jin Lee
- Reproductive and Developmental Toxicology Research Group, Korea Institute of Toxicology, Daejeon 34114, Korea;
| | - Eon-Bee Lee
- Laboratory of Veterinary Pharmacokinetics and Pharmacodynamics, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Korea;
| | - Biruk Tesfaye Birhanu
- Laboratory of Veterinary Pharmacokinetics and Pharmacodynamics, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Korea;
- Cardiovascular Research Institute, Kyungpook National University, Daegu 41944, Korea
- Correspondence: (B.T.B.); (S.-C.P.); Tel.: +82-53-950-5964 (B.T.B. & S.-C.P.)
| | - Seung-Chun Park
- Laboratory of Veterinary Pharmacokinetics and Pharmacodynamics, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Korea;
- Cardiovascular Research Institute, Kyungpook National University, Daegu 41944, Korea
- Correspondence: (B.T.B.); (S.-C.P.); Tel.: +82-53-950-5964 (B.T.B. & S.-C.P.)
| |
Collapse
|
15
|
Shein AMS, Wannigama DL, Higgins PG, Hurst C, Abe S, Hongsing P, Chantaravisoot N, Saethang T, Luk-In S, Liao T, Nilgate S, Rirerm U, Kueakulpattana N, Laowansiri M, Srisakul S, Muhummudaree N, Techawiwattanaboon T, Gan L, Xu C, Kupwiwat R, Phattharapornjaroen P, Rojanathanes R, Leelahavanichkul A, Chatsuwan T. Novel colistin-EDTA combination for successful eradication of colistin-resistant Klebsiella pneumoniae catheter-related biofilm infections. Sci Rep 2021; 11:21676. [PMID: 34737361 PMCID: PMC8568960 DOI: 10.1038/s41598-021-01052-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 10/20/2021] [Indexed: 12/20/2022] Open
Abstract
Development of an effective therapy to overcome colistin resistance in Klebsiella pneumoniae, a common pathogen causing catheter-related biofilm infections in vascular catheters, has become a serious therapeutic challenge that must be addressed urgently. Although colistin and EDTA have successful roles for eradicating biofilms, no in vitro and in vivo studies have investigated their efficacy in catheter-related biofilm infections of colistin-resistant K. pneumoniae. In this study, colistin resistance was significantly reversed in both planktonic and mature biofilms of colistin-resistant K. pneumoniae by a combination of colistin (0.25-1 µg/ml) with EDTA (12 mg/ml). This novel colistin-EDTA combination was also demonstrated to have potent efficacy in eradicating colistin-resistant K. pneumoniae catheter-related biofilm infections, and eliminating the risk of recurrence in vivo. Furthermore, this study revealed significant therapeutic efficacy of colistin-EDTA combination in reducing bacterial load in internal organs, lowering serum creatinine, and protecting treated mice from mortality. Altered in vivo expression of different virulence genes indicate bacterial adaptive responses to survive in hostile environments under different treatments. According to these data discovered in this study, a novel colistin-EDTA combination provides favorable efficacy and safety for successful eradication of colistin-resistant K. pneumonia catheter-related biofilm infections.
Collapse
Affiliation(s)
- Aye Mya Sithu Shein
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
- Antimicrobial Resistance and Stewardship Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Interdisciplinary Program of Medical Microbiology, Graduate School, Chulalongkorn University, Bangkok, Thailand
| | - Dhammika Leshan Wannigama
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
- Antimicrobial Resistance and Stewardship Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- School of Medicine, Faculty of Health and Medical Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - Paul G Higgins
- Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- German Centre for Infection Research, Partner Site Bonn-Cologne, Cologne, Germany
| | - Cameron Hurst
- Statistics, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Shuichi Abe
- Department of Infectious Diseases and Infection Control, Yamagata Prefectural Central Hospital, Yamagata, Japan
| | - Parichart Hongsing
- Mae Fah Luang University Hospital, Chiang Rai, Thailand
- School of Integrative Medicine, Mae Fah Luang University, Chiang Rai, Thailand
| | - Naphat Chantaravisoot
- Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Systems Biology, Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Thammakorn Saethang
- Department of Computer Science, Faculty of Science, Kasetsart University, Bangkok, Thailand
| | - Sirirat Luk-In
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Tingting Liao
- Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence for Microcirculation, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Sumanee Nilgate
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
- Antimicrobial Resistance and Stewardship Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Ubolrat Rirerm
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
- Antimicrobial Resistance and Stewardship Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Naris Kueakulpattana
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
- Antimicrobial Resistance and Stewardship Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Matchima Laowansiri
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
- Antimicrobial Resistance and Stewardship Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Sukrit Srisakul
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
- Antimicrobial Resistance and Stewardship Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Netchanok Muhummudaree
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
- Antimicrobial Resistance and Stewardship Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Teerasit Techawiwattanaboon
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
- Chula Vaccine Research Center, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Lin Gan
- Department of General Surgery, Fuling Center Hospital of Chongqing City, Chongqing, China
| | - Chenchen Xu
- In-Patient Pharmacy, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Rosalyn Kupwiwat
- Chulabhorn International College of Medicine, Thammasat University, Thammasat University Hospital, Bangkok, Thailand
| | - Phatthranit Phattharapornjaroen
- Department of Emergency Medicine, Center of Excellence, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- Institute of Clinical Sciences, Department of Surgery, Sahlgrenska Academy, Gothenburg University, 40530, Gothenburg, Sweden
| | - Rojrit Rojanathanes
- Center of Excellence in Materials and Bio-Interfaces, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Asada Leelahavanichkul
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
- Translational Research in Inflammation and Immunology Research Unit (TRIRU), Department of Microbiology, Chulalongkorn University, Bangkok, Thailand
| | - Tanittha Chatsuwan
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand.
- Antimicrobial Resistance and Stewardship Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
16
|
Equisetin Restores Colistin Sensitivity against Multi-Drug Resistant Gram-Negative Bacteria. Antibiotics (Basel) 2021; 10:antibiotics10101263. [PMID: 34680843 PMCID: PMC8532683 DOI: 10.3390/antibiotics10101263] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/07/2021] [Accepted: 10/13/2021] [Indexed: 11/17/2022] Open
Abstract
The overuse of antibiotics and the scarcity of new drugs have led to a serious antimicrobial resistance crisis, especially for multi-drug resistant (MDR) Gram-negative bacteria. In the present study, we investigated the antimicrobial activity of a marine antibiotic equisetin in combination with colistin against Gram-negative bacteria and explored the mechanisms of synergistic activity. We tested the synergistic effect of equisetin in combination with colistin on 23 clinical mcr-1 positive isolates and found that 4 µg/mL equisetin combined with 1 µg/mL colistin showed 100% inhibition. Consistently, equisetin restored the sensitivity of 10 species of mcr-1 positive Gram-negative bacteria to colistin. The combination of equisetin and colistin quickly killed 99.9% bacteria in one hour in time-kill assays. We found that colistin promoted intracellular accumulation of equisetin in colistin-resistant E. coli based on LC-MS/MS analysis. Interestingly, equisetin boosted ROS accumulation in E. coli in the presence of colistin. Moreover, we found that equisetin and colistin lost the synergistic effect in two LPS-deficient A. baumannii strains. These findings suggest that colistin destroys the hydrophobic barrier of Gram-negative bacteria, facilitating equisetin to enter the cell and exert its antibacterial effect. Lastly, equisetin restored the activity of colistin in a G. mellonella larvae infection model. Collectively, these results reveal that equisetin can potentiate colistin activity against MDR Gram-negative bacteria including colistin-resistant strains, providing an alternative approach to address Gram-negative pathogens associated with infections in clinics.
Collapse
|
17
|
Powell LC, Abdulkarim M, Stokniene J, Yang QE, Walsh TR, Hill KE, Gumbleton M, Thomas DW. Quantifying the effects of antibiotic treatment on the extracellular polymer network of antimicrobial resistant and sensitive biofilms using multiple particle tracking. NPJ Biofilms Microbiomes 2021; 7:13. [PMID: 33547326 PMCID: PMC7864955 DOI: 10.1038/s41522-020-00172-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 11/24/2020] [Indexed: 01/30/2023] Open
Abstract
Novel therapeutics designed to target the polymeric matrix of biofilms requires innovative techniques to accurately assess their efficacy. Here, multiple particle tracking (MPT) was developed to characterize the physical and mechanical properties of antimicrobial resistant (AMR) bacterial biofilms and to quantify the effects of antibiotic treatment. Studies employed nanoparticles (NPs) of varying charge and size (40-500 nm) in Pseudomonas aeruginosa PAO1 and methicillin-resistant Staphylococcus aureus (MRSA) biofilms and also in polymyxin B (PMB) treated Escherichia coli biofilms of PMB-sensitive (PMBSens) IR57 and PMB-resistant (PMBR) PN47 strains. NP size-dependent and strain-related differences in the diffusion coefficient values of biofilms were evident between PAO1 and MRSA. Dose-dependent treatment effects induced by PMB in PMBSens E. coli biofilms included increases in diffusion and creep compliance (P < 0.05), not evident in PMB treatment of PMBR E. coli biofilms. Our results highlight the ability of MPT to quantify the diffusion and mechanical effects of antibiotic therapies within the AMR biofilm matrix, offering a valuable tool for the pre-clinical screening of anti-biofilm therapies.
Collapse
Affiliation(s)
- Lydia C Powell
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff, UK.
- Centre of Nanohealth, Swansea University Medical School, Swansea University, Swansea, UK.
| | - Muthanna Abdulkarim
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK.
| | - Joana Stokniene
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff, UK
| | - Qiu E Yang
- Medical Microbiology and Infectious Disease, School of Medicine, Cardiff University, Cardiff, UK
| | - Timothy R Walsh
- Medical Microbiology and Infectious Disease, School of Medicine, Cardiff University, Cardiff, UK
| | - Katja E Hill
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff, UK
| | - Mark Gumbleton
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK
| | - David W Thomas
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff, UK
| |
Collapse
|
18
|
Gong H, He L, Zhao Z, Mao X, Zhang C. The specific effect of (R)-(+)-pulegone on growth and biofilm formation in multi-drug resistant Escherichia coli and molecular mechanisms underlying the expression of pgaABCD genes. Biomed Pharmacother 2021; 134:111149. [PMID: 33385683 DOI: 10.1016/j.biopha.2020.111149] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 11/29/2020] [Accepted: 12/14/2020] [Indexed: 01/11/2023] Open
Abstract
E. coli is associated with high rates of infection and resistance to drugs not only in China but also the rest of the world. In addition, the number of E. coli biofilm infections continue to increase with time. Notably, biofilms are attractive targets for the prevention of infections caused by multidrug-resistant bacteria. Moreover, the pgaABCD-encoded Poly-β-1,6-N-acetyl-d-glucosamine (PNAG) plays an important role in biofilm formation. Therefore, this study aimed to explore the specific effect of the (R)-(+)-pulegone (PU) on growth and biofilm formation in multi-drug resistant E. coli. The molecular mechanisms involved were also examined. The results showed that PU had significant antibacterial and antibiofilm formation activity against E. coli K1, with MIC and MBC values of 23.68 and 47.35 mg/mL, respectively. On the other hand, the maximum inhibition rate for biofilm formation in the bacterium was 52.36 % at 94.70 mg/mL of PU. qRT-PCR data showed that PU significantly down-regulated expression of the pgaABCD genes (P < 0.05). PU was also broadly effective against biofilm formation in MG1655 and MG1655/ΔpgaABCD, exhibiting the maximum inhibition rates were 98.23 % and 93.35 %, respectively. In addition, PU destroyed pre-formed mature biofilm in both MG1655 and MG1655/ΔpgaABCD about 95.03 % and 92.4 %, respectively. The study therefore verified that pgaA was a potential and key target for PU in E. coli although it was not the only one. Overall, the findings indicated that PU is a potential and novel inhibitor of drug resistance, This therefore gives insights on new ways of preventing and treating biofilm-associated infections in the food industry as well as in clinical practice.
Collapse
Affiliation(s)
- Haiyan Gong
- The Fifth Affiliated Hospital of Xinjiang Medical University, Xinshi District, No. 118 Henan West Road, Urumqi, Xinjiang, PR China.
| | - Lijuan He
- College of Public Health of Xinjiang Medical University, Shuimogou District, No. 567 Shangde North Road, Urumqi, Xinjiang, PR China.
| | - Zhilong Zhao
- The Fifth Affiliated Hospital of Xinjiang Medical University, Xinshi District, No. 118 Henan West Road, Urumqi, Xinjiang, PR China.
| | - Xinmin Mao
- College of Traditional Chinese Medicine, Key Discipline of Integrated Traditional Chinese and Western Medicine of Autonomous Region from Xinjiang Medical University, Shuimogou District, No. 567 Shangde North Road, Urumqi, Xinjiang, PR China.
| | - Chen Zhang
- Xinjiang Medical University, Shuimogou District, No. 567 Shangde North Road, Urumqi, Xinjiang, PR China.
| |
Collapse
|
19
|
Thermal and non-thermal treatment effects on Staphylococcus aureus biofilms formed at different temperatures and maturation periods. Food Res Int 2020; 137:109432. [PMID: 33233114 DOI: 10.1016/j.foodres.2020.109432] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 05/30/2020] [Accepted: 06/09/2020] [Indexed: 11/20/2022]
Abstract
The objective of this study was to investigate the effect of temperature and maturation period on the resistance of Staphylococcus aureus biofilms to thermal and non-thermal treatments. First, biofilm development was compared at three different temperatures (15, 25, and 37°C) for 5 days. The cell population at 15 and 25°C remained relatively consistent approximately at 6.3 log CFU/cm2, whereas 37°C resulted in the highest cell population on day 1 (7.6 log CFU/cm2) followed by a continual decline. Then, biofilm resistance to steam and sodium hypochlorite (NaOCl) treatments was evaluated. Obtained results highlighted that biofilms had different resistance to both treatments depending on development conditions. Specifically, steam treatment of 10 s eliminated 4.1 log CFU/cm2 of the biofilm formed at 25°C for 5 days. The same treatment inactivated over 5 log population of biofilms developed in other temperature and maturation period conditions. Treatment with NaOCl reduced approximately 1 log CFU/cm2 of biofilm cells developed at 25°C for 5 days. However, inactivation was found to be over 2 log CFU/cm2 under other development conditions. An extracellular polymeric substances (EPS) quantification using 96-well plates and stainless steel coupons was conducted. In the 96-well plate experiment, it was found that the highest amount of polysaccharide was secreted at 25°C (p < 0.05), while total biomass and protein contents were greatest at 37°C (p < 0.05). No significant difference in EPS content was observed for stainless steel, but the results displayed a similar trend to the 96-well plate. In particular, biofilms developed at 25°C tended to secret the highest amount of polysaccharide, which aligned with the current literature. This finding indicated that polysaccharide was the main contribution to the enhanced resistance of S. aureus biofilms. Overall, it was shown that biofilms formed at 25°C for 5 days exhibited the greatest resistance to thermal and nonthermal treatments due to the elevated exopolysaccharide secretion. This study demonstrates that temperature and maturation period significantly affect the resistance of S. aureus biofilms to thermal and non-thermal treatments.
Collapse
|
20
|
Penesyan A, Paulsen IT, Gillings MR, Kjelleberg S, Manefield MJ. Secondary Effects of Antibiotics on Microbial Biofilms. Front Microbiol 2020; 11:2109. [PMID: 32983070 PMCID: PMC7492572 DOI: 10.3389/fmicb.2020.02109] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 08/11/2020] [Indexed: 12/28/2022] Open
Abstract
Biofilms are assemblages of microorganisms attached to each other, or to a surface, and encased in a protective, self-produced matrix. Such associations are now recognized as the predominant microbial growth mode. The physiology of cells in biofilms differs from that of the planktonic cells on which most research has been conducted. Consequently, there are significant gaps in our knowledge of the biofilm lifestyle. Filling this gap is particularly important, given that biofilm cells may respond differently to antibiotics than do planktonic cells of the same species. Understanding the effects of antibiotics on biofilms is of paramount importance for clinical practice due to the increased levels of antibiotic resistance and resistance dissemination in biofilms. From a wider environmental perspective antibiotic exposure can alter the ecology of biofilms in nature, and hence disrupt ecosystems. Biofilm cells display increased resilience toward antibiotics. This resilience is often explained by mechanisms and traits such as decreased antibiotic penetration, metabolically inactive persister cells, and intrinsic resistance by members of the biofilm community. Together, these factors suggest that cells in biofilms are often exposed to subinhibitory concentrations of antimicrobial agents. Here we discuss how cells in biofilms are affected by the presence of antibiotics at subinhibitory concentrations, and the possible ramifications of such secondary effects for healthcare and the environment.
Collapse
Affiliation(s)
- Anahit Penesyan
- School of Chemical Engineering, University of New South Wales, Sydney, NSW, Australia
- Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, Australia
| | - Ian T. Paulsen
- Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, Australia
- ARC Centre of Excellence in Synthetic Biology, Macquarie University, Sydney, NSW, Australia
| | - Michael R. Gillings
- ARC Centre of Excellence in Synthetic Biology, Macquarie University, Sydney, NSW, Australia
- Department of Biological Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, Australia
| | - Staffan Kjelleberg
- Singapore Centre for Environmental Life Sciences Engineering, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
- School of Biological, Earth and Environmental Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Michael J. Manefield
- School of Chemical Engineering, University of New South Wales, Sydney, NSW, Australia
- School of Civil and Environmental Engineering, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
21
|
Hashimoto A, Miyamoto H, Kobatake T, Nakashima T, Shobuike T, Ueno M, Murakami T, Noda I, Sonohata M, Mawatari M. The combination of silver-containing hydroxyapatite coating and vancomycin has a synergistic antibacterial effect on methicillin-resistant Staphylococcus aureus biofilm formation. Bone Joint Res 2020; 9:211-218. [PMID: 32566142 PMCID: PMC7284291 DOI: 10.1302/2046-3758.95.bjr-2019-0326.r1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Aims Biofilm formation is intrinsic to prosthetic joint infection (PJI). In the current study, we evaluated the effects of silver-containing hydroxyapatite (Ag-HA) coating and vancomycin (VCM) on methicillin-resistant Staphylococcus aureus (MRSA) biofilm formation. Methods Pure titanium discs (Ti discs), Ti discs coated with HA (HA discs), and 3% Ag-HA discs developed using a thermal spraying were inoculated with MRSA suspensions containing a mean in vitro 4.3 (SD 0.8) x 106 or 43.0 (SD 8.4) x 105 colony-forming units (CFUs). Immediately after MRSA inoculation, sterile phosphate-buffered saline or VCM (20 µg/ml) was added, and the discs were incubated for 24 hours at 37°C. Viable cell counting, 3D confocal laser scanning microscopy with Airyscan, and scanning electron microscopy were then performed. HA discs and Ag HA discs were implanted subcutaneously in vivo in the dorsum of rats, and MRSA suspensions containing a mean in vivo 7.2 (SD 0.4) x 106 or 72.0 (SD 4.2) x 105 CFUs were inoculated on the discs. VCM was injected subcutaneously daily every 12 hours followed by viable cell counting. Results Biofilms that formed on HA discs were thicker and larger than those on Ti discs, whereas those on Ag-HA discs were thinner and smaller than those on Ti discs. Viable bacterial counts in vivo revealed that Ag-HA combined with VCM was the most effective treatment. Conclusion Ag-HA with VCM has a potential synergistic effect in reducing MRSA biofilm formation and can thus be useful for preventing and treating PJI. Cite this article:Bone Joint Res. 2020;9(5):211–218.
Collapse
Affiliation(s)
- Akira Hashimoto
- Department of Orthopaedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Hiroshi Miyamoto
- Department of Pathology and Microbiology, Faculty of Medicine, Saga University, Saga, Japan
| | - Tomoki Kobatake
- Department of Orthopaedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Takema Nakashima
- Department of Orthopaedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Takeo Shobuike
- Department of Pathology and Microbiology, Faculty of Medicine, Saga University, Saga, Japan
| | - Masaya Ueno
- Department of Orthopaedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Takayuki Murakami
- Department of Pathology and Microbiology, Faculty of Medicine, Saga University, Saga, Japan; Research Section, Medical Division, KYO CERA Corporation, Yasu, Japan
| | - Iwao Noda
- Department of Pathology and Microbiology, Faculty of Medicine, Saga University, Saga, Japan; Research Section, Medical Division, KYO CERA Corporation, Yasu, Japan
| | - Motoki Sonohata
- Department of Orthopaedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Masaaki Mawatari
- Department of Orthopaedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| |
Collapse
|
22
|
Elbourne A, Cheeseman S, Atkin P, Truong NP, Syed N, Zavabeti A, Mohiuddin M, Esrafilzadeh D, Cozzolino D, McConville CF, Dickey MD, Crawford RJ, Kalantar-Zadeh K, Chapman J, Daeneke T, Truong VK. Antibacterial Liquid Metals: Biofilm Treatment via Magnetic Activation. ACS NANO 2020; 14:802-817. [PMID: 31922722 DOI: 10.1021/acsnano.9b07861] [Citation(s) in RCA: 145] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Antibiotic resistance has made the treatment of biofilm-related infections challenging. As such, the quest for next-generation antimicrobial technologies must focus on targeted therapies to which pathogenic bacteria cannot develop resistance. Stimuli-responsive therapies represent an alternative technological focus due to their capability of delivering targeted treatment. This study provides a proof-of-concept investigation into the use of magneto-responsive gallium-based liquid metal (LM) droplets as antibacterial materials, which can physically damage, disintegrate, and kill pathogens within a mature biofilm. Once exposed to a low-intensity rotating magnetic field, the LM droplets become physically actuated and transform their shape, developing sharp edges. When placed in contact with a bacterial biofilm, the movement of the particles resulting from the magnetic field, coupled with the presence of nanosharp edges, physically ruptures the bacterial cells and the dense biofilm matrix is broken down. The antibacterial efficacy of the magnetically activated LM particles was assessed against both Gram-positive and Gram-negative bacterial biofilms. After 90 min over 99% of both bacterial species became nonviable, and the destruction of the biofilms was observed. These results will impact the design of next-generation, LM-based biofilm treatments.
Collapse
Affiliation(s)
- Aaron Elbourne
- School of Science, College of Science, Engineering and Health , RMIT University , Melbourne , Victoria 3001 , Australia
- Nanobiotechnology Laboratory , RMIT University , Melbourne , Victoria 3001 , Australia
| | - Samuel Cheeseman
- School of Science, College of Science, Engineering and Health , RMIT University , Melbourne , Victoria 3001 , Australia
- Nanobiotechnology Laboratory , RMIT University , Melbourne , Victoria 3001 , Australia
| | - Paul Atkin
- School of Engineering, College of Science, Engineering and Health , RMIT University , Melbourne , Victoria 3001 , Australia
- CSIRO Australia , Private Bag 33, Clayton South MDC , Clayton , Victoria 3169 , Australia
| | - Nghia P Truong
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences , Monash University , 399 Royal Parade , Parkville , Victoria 3152 , Australia
| | - Nitu Syed
- School of Engineering, College of Science, Engineering and Health , RMIT University , Melbourne , Victoria 3001 , Australia
| | - Ali Zavabeti
- School of Engineering, College of Science, Engineering and Health , RMIT University , Melbourne , Victoria 3001 , Australia
| | - Md Mohiuddin
- School of Engineering, College of Science, Engineering and Health , RMIT University , Melbourne , Victoria 3001 , Australia
| | - Dorna Esrafilzadeh
- School of Engineering, College of Science, Engineering and Health , RMIT University , Melbourne , Victoria 3001 , Australia
- Graduate School of Biomedical Engineering , University of New South Wales (UNSW) , Kensington , NSW 2052 , Australia
| | - Daniel Cozzolino
- School of Science, College of Science, Engineering and Health , RMIT University , Melbourne , Victoria 3001 , Australia
| | - Chris F McConville
- School of Science, College of Science, Engineering and Health , RMIT University , Melbourne , Victoria 3001 , Australia
| | - Michael D Dickey
- Department of Chemical and Biomolecular Engineering , North Carolina State University , Raleigh , North Carolina 27695 , United States
| | - Russell J Crawford
- School of Science, College of Science, Engineering and Health , RMIT University , Melbourne , Victoria 3001 , Australia
- Nanobiotechnology Laboratory , RMIT University , Melbourne , Victoria 3001 , Australia
| | - Kourosh Kalantar-Zadeh
- School of Chemical Engineering , University of New South Wales (UNSW) , Kensington , NSW 2052 , Australia
| | - James Chapman
- School of Science, College of Science, Engineering and Health , RMIT University , Melbourne , Victoria 3001 , Australia
- Nanobiotechnology Laboratory , RMIT University , Melbourne , Victoria 3001 , Australia
| | - Torben Daeneke
- School of Engineering, College of Science, Engineering and Health , RMIT University , Melbourne , Victoria 3001 , Australia
| | - Vi Khanh Truong
- School of Science, College of Science, Engineering and Health , RMIT University , Melbourne , Victoria 3001 , Australia
- Nanobiotechnology Laboratory , RMIT University , Melbourne , Victoria 3001 , Australia
| |
Collapse
|
23
|
Olivares E, Badel-Berchoux S, Provot C, Prévost G, Bernardi T, Jehl F. Clinical Impact of Antibiotics for the Treatment of Pseudomonas aeruginosa Biofilm Infections. Front Microbiol 2020; 10:2894. [PMID: 31998248 PMCID: PMC6962142 DOI: 10.3389/fmicb.2019.02894] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 12/02/2019] [Indexed: 02/03/2023] Open
Abstract
Bacterial biofilms are highly recalcitrant to antibiotic therapies due to multiple tolerance mechanisms. The involvement of Pseudomonas aeruginosa in a wide range of biofilm-related infections often leads to treatment failures. Indeed, few current antimicrobial molecules are still effective on tolerant sessile cells. In contrast, studies increasingly showed that conventional antibiotics can, at low concentrations, induce a phenotype change in bacteria and consequently, the biofilm formation. Understanding the clinical effects of antimicrobials on biofilm establishment is essential to avoid the use of inappropriate treatments in the case of biofilm infections. This article reviews the current knowledge about bacterial growth within a biofilm and the preventive or inducer impact of standard antimicrobials on its formation by P. aeruginosa. The effect of antibiotics used to treat biofilms of other bacterial species, as Staphylococcus aureus or Escherichia coli, was also briefly mentioned. Finally, it describes two in vitro devices which could potentially be used as antibiotic susceptibility testing for adherent bacteria.
Collapse
Affiliation(s)
- Elodie Olivares
- University of Strasbourg, CHRU Strasbourg, Fédération de Médecine Translationnelle de Strasbourg, EA7290, Institut de Bactériologie, Strasbourg, France.,BioFilm Pharma SAS, Saint-Beauzire, France
| | | | - Christian Provot
- BioFilm Pharma SAS, Saint-Beauzire, France.,BioFilm Control SAS, Saint-Beauzire, France
| | - Gilles Prévost
- University of Strasbourg, CHRU Strasbourg, Fédération de Médecine Translationnelle de Strasbourg, EA7290, Institut de Bactériologie, Strasbourg, France
| | - Thierry Bernardi
- BioFilm Pharma SAS, Saint-Beauzire, France.,BioFilm Control SAS, Saint-Beauzire, France
| | - François Jehl
- University of Strasbourg, CHRU Strasbourg, Fédération de Médecine Translationnelle de Strasbourg, EA7290, Institut de Bactériologie, Strasbourg, France
| |
Collapse
|
24
|
Staphylococcus aureus Pneumonia: Preceding Influenza Infection Paves the Way for Low-Virulent Strains. Toxins (Basel) 2019; 11:toxins11120734. [PMID: 31861176 PMCID: PMC6950557 DOI: 10.3390/toxins11120734] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/05/2019] [Accepted: 12/14/2019] [Indexed: 12/22/2022] Open
Abstract
Staphylococcus aureus is a facultative pathogenic bacterium that colonizes the nasopharyngeal area of healthy individuals, but can also induce severe infection, such as pneumonia. Pneumonia caused by mono- or superinfected S.aureus leads to high mortality rates. To establish an infection, S. aureus disposes of a wide variety of virulence factors, which can vary between clinical isolates. Our study aimed to characterize pneumonia isolates for their virulent capacity. For this, we analyzed isolates from colonization, pneumonia due to S. aureus, and pneumonia due to S. aureus/influenza virus co-infection. A total of 70 strains were analyzed for their virulence genes and the host–pathogen interaction was analyzed through functional assays in cell culture systems. Strains from pneumonia due to S. aureus mono-infection showed enhanced invasion and cytotoxicity against professional phagocytes than colonizing and co-infecting strains. This corresponded to the high presence of cytotoxic components in pneumonia strains. By contrast, strains obtained from co-infection did not exhibit these virulence characteristics and resembled strains from colonization, although they caused the highest mortality rate in patients. Taken together, our results underline the requirement of invasion and toxins to cause pneumonia due to S. aureus mono-infection, whereas in co-infection even low-virulent strains can severely aggravate pneumonia.
Collapse
|
25
|
Frickmann H, Klenk C, Warnke P, Redanz S, Podbielski A. Influence of Probiotic Culture Supernatants on In Vitro Biofilm Formation of Staphylococci. Eur J Microbiol Immunol (Bp) 2018; 8:119-127. [PMID: 30719328 PMCID: PMC6348700 DOI: 10.1556/1886.2018.00022] [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: 08/19/2018] [Accepted: 08/27/2018] [Indexed: 01/15/2023] Open
Abstract
Background The effects of cell-free culture supernatants of probiotic Lactobacillus rhamnosus GG and Streptococcus salivarius K12 on replication and biofilm forming of Staphylococcus aureus and S. epidermidis were assessed in vitro. Methods S. aureus and S. epidermidis strains were exposed to cell-free culture supernatants of L. rhamnosus GG and S. salivarius K12 at different concentrations starting at 0, 4, and 24 h after the onset of incubation. Bacterial amplification was measured on microplate readers, as well as biofilm growth after safranine staining. Scanning electron microscopy was performed for visualization of biofilm status. Results The S. salivarius K12 culture supernatant not only reduced or prevented the formation and maturation of fresh biofilms but even caused a reduction of preformed S. epidermidis biofilms. The L. rhamnosus GG culture supernatant did not show clear inhibitory effects regardless of concentration or time of addition of supernatant, and even concentration-depending promotional effects on the planktonic and biofilm growth of S. aureus and S. epidermidis were observed. Conclusion In particular, the inhibitory effects of the S. salivarius K12 culture supernatant on the formation of staphylococcal biofilms are of potential relevance for biofilm-associated diseases and should be further assessed by in vivo infection models.
Collapse
Affiliation(s)
- Hagen Frickmann
- Institute for Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany.,Department of Microbiology and Hospital Hygiene, Bundeswehr Hospital Hamburg, Hamburg, Germany
| | - Caroline Klenk
- Institute for Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany
| | - Philipp Warnke
- Institute for Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany
| | - Sylvio Redanz
- Institute for Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany.,Kreth Lab, Department of Restorative Dentistry, Oregon Health & Science University, Portland, OR, USA
| | - Andreas Podbielski
- Institute for Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany
| |
Collapse
|
26
|
Cao Q, Wu S, Wang L, Shi X, Li G. Effects of the morphology of sulfobetaine zwitterionic layers grafted onto a silicone surface on improving the hydrophilic stability, anti-bacterial adhesion properties, and biocompatibility. J Appl Polym Sci 2018. [DOI: 10.1002/app.46860] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Qin Cao
- School of Materials Science and Engineering; South China University of Technology; Guangzhou 510640 China
| | - Shuqing Wu
- School of Materials Science and Engineering; South China University of Technology; Guangzhou 510640 China
| | - Liying Wang
- School of Materials Science and Engineering; South China University of Technology; Guangzhou 510640 China
| | - Xuetao Shi
- School of Materials Science and Engineering; South China University of Technology; Guangzhou 510640 China
| | - Guangji Li
- School of Materials Science and Engineering; South China University of Technology; Guangzhou 510640 China
| |
Collapse
|
27
|
MacNair CR, Stokes JM, Carfrae LA, Fiebig-Comyn AA, Coombes BK, Mulvey MR, Brown ED. Overcoming mcr-1 mediated colistin resistance with colistin in combination with other antibiotics. Nat Commun 2018; 9:458. [PMID: 29386620 PMCID: PMC5792607 DOI: 10.1038/s41467-018-02875-z] [Citation(s) in RCA: 177] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 01/04/2018] [Indexed: 12/13/2022] Open
Abstract
Plasmid-borne colistin resistance mediated by mcr-1 may contribute to the dissemination of pan-resistant Gram-negative bacteria. Here, we show that mcr-1 confers resistance to colistin-induced lysis and bacterial cell death, but provides minimal protection from the ability of colistin to disrupt the Gram-negative outer membrane. Indeed, for colistin-resistant strains of Enterobacteriaceae expressing plasmid-borne mcr-1, clinically relevant concentrations of colistin potentiate the action of antibiotics that, by themselves, are not active against Gram-negative bacteria. The result is that several antibiotics, in combination with colistin, display growth-inhibition at levels below their corresponding clinical breakpoints. Furthermore, colistin and clarithromycin combination therapy displays efficacy against mcr-1-positive Klebsiella pneumoniae in murine thigh and bacteremia infection models at clinically relevant doses. Altogether, these data suggest that the use of colistin in combination with antibiotics that are typically active against Gram-positive bacteria poses a viable therapeutic alternative for highly drug-resistant Gram-negative pathogens expressing mcr-1.
Collapse
Affiliation(s)
- Craig R MacNair
- Michael G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, L8N 3ZS, Canada
| | - Jonathan M Stokes
- Michael G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, L8N 3ZS, Canada
| | - Lindsey A Carfrae
- Michael G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, L8N 3ZS, Canada
| | - Aline A Fiebig-Comyn
- Michael G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, L8N 3ZS, Canada
| | - Brian K Coombes
- Michael G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, L8N 3ZS, Canada
| | - Michael R Mulvey
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, R3E 3R2, Canada
| | - Eric D Brown
- Michael G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, L8N 3ZS, Canada.
| |
Collapse
|
28
|
Ribera A, Soldevila L, Rigo-Bonnin R, Tubau F, Padullés A, Gómez-Junyent J, Ariza J, Murillo O. Beta-lactams in continuous infusion for Gram-negative bacilli osteoarticular infections: an easy method for clinical use. Infection 2018; 46:239-244. [PMID: 29363049 DOI: 10.1007/s15010-018-1116-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 01/16/2018] [Indexed: 12/16/2022]
Abstract
Continuous infusion (CI) of beta-lactams could optimize their pharmacokinetic/pharmacodynamic indices, especially in difficult-to-treat infections. PURPOSE To validate an easy-to-use method to guide beta-lactams dosage in CI (formula). METHODS A retrospective analysis was conducted of a prospectively collected cohort (n = 24 patients) with osteoarticular infections caused by Gram-negative bacilli (GNB) managed with beta-lactams in CI. Beta-lactams dose was calculated using a described formula (daily dose = 24 h × beta-lactam clearance × target "steady-state" concentration) to achieve concentrations above the MIC. We correlated the predicted concentration (Cpred = daily dose/24 h × beta-lactam clearance) with the patient's observed concentration (Cobs) measured by UPLC-MS/MS (Spearman's coefficient). RESULTS The most frequent microorganism treated was P. aeruginosa (21 cases; 9 MDR). Beta-lactams in CI were ceftazidime (n = 14), aztreonam (7), and piperacillin/tazobactam (3), mainly used in combination (12 with colistin, 5 with ciprofloxacin) and administered without notable side effects. The plasma Cobs was higher overall than Cpred; the Spearman correlation between both concentrations was rho = 0.6 (IC 95%: 0.2-0.8) for all beta-lactams, and rho = 0.8 (IC 95%: 0.4-1) for those treated with ceftazidime. CONCLUSIONS The formula may be useful in clinical practice for planning the initial dosage of beta-lactams in CI, while we await a systematic therapeutic drug monitoring. The use of beta-lactams in CI was safe.
Collapse
Affiliation(s)
- Alba Ribera
- Infectious Diseases Department, IDIBELL-Hospital Universitari de Bellvitge, Feixa Llarga s/n, Hospitalet de Llobregat, 08907, Barcelona, Spain
| | - Laura Soldevila
- Infectious Diseases Department, IDIBELL-Hospital Universitari de Bellvitge, Feixa Llarga s/n, Hospitalet de Llobregat, 08907, Barcelona, Spain
| | - Raul Rigo-Bonnin
- Clinical Laboratory Department, IDIBELL-Hospital Universitari de Bellvitge, Feixa Llarga s/n, Hospitalet de Llobregat, 08907, Barcelona, Spain
| | - Fe Tubau
- Microbiology Department, IDIBELL-Hospital Universitari de Bellvitge, Feixa Llarga s/n, Hospitalet de Llobregat, 08907, Barcelona, Spain.,Ciber de Enfermedades Respiratorias ISCIII, Madrid, Spain
| | - Ariadna Padullés
- Pharmacy Department, IDIBELL-Hospital Universitari de Bellvitge, Feixa Llarga s/n, Hospitalet de Llobregat, 08907, Barcelona, Spain
| | - Joan Gómez-Junyent
- Infectious Diseases Department, IDIBELL-Hospital Universitari de Bellvitge, Feixa Llarga s/n, Hospitalet de Llobregat, 08907, Barcelona, Spain
| | - Javier Ariza
- Infectious Diseases Department, IDIBELL-Hospital Universitari de Bellvitge, Feixa Llarga s/n, Hospitalet de Llobregat, 08907, Barcelona, Spain
| | - Oscar Murillo
- Infectious Diseases Department, IDIBELL-Hospital Universitari de Bellvitge, Feixa Llarga s/n, Hospitalet de Llobregat, 08907, Barcelona, Spain.
| |
Collapse
|
29
|
Thiol Starvation Induces Redox-Mediated Dysregulation of Escherichia coli Biofilm Components. J Bacteriol 2017; 200:JB.00389-17. [PMID: 29038256 DOI: 10.1128/jb.00389-17] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 10/06/2017] [Indexed: 11/20/2022] Open
Abstract
A hallmark of bacterial biofilms is the production of an extracellular matrix (ECM) that encases and protects the community from environmental stressors. Biofilm formation is an integral portion of the uropathogenic Escherichia coli (UPEC) life cycle. Approximately 2% of UPEC isolates are cysteine auxotrophs. Here, we investigated how cysteine homeostasis impacted UPEC UTI89 strain biofilm formation and, specifically, the production of the ECM components curli and cellulose. Cysteine auxotrophs produced less cellulose and slightly more curli compared to wild-type (WT) strains, and cysteine auxotrophs formed smooth, nonrugose colonies. Cellulose production was restored in cysteine auxotrophs when YfiR was inactivated. YfiR is a redox-sensitive regulator of the diguanylate cyclase, YfiN. The production of curli, a temperature-regulated appendage, was independent of temperature in UTI89 cysteine auxotrophs. In a screen of UPEC isolates, we found that ∼60% of UPEC cysteine auxotrophs produced curli at 37°C, but only ∼2% of cysteine prototrophic UPEC isolates produced curli at 37°C. Interestingly, sublethal concentrations of amdinocillin and trimethoprim-sulfamethoxazole inhibited curli production, whereas strains auxotrophic for cysteine continued to produce curli even in the presence of amdinocillin and trimethoprim-sulfamethoxazole. The dysregulation of ECM components and resistance to amdinocillin in cysteine auxotrophs may be linked to hyperoxidation, since the addition of exogenous cysteine or glutathione restored WT biofilm phenotypes to mutants unable to produce cysteine and glutathione.IMPORTANCE Uropathogenic Escherichia coli (UPEC) bacteria are the predominant causative agent of urinary tract infections (UTIs). UTIs account for billions of dollars of financial burden annually to the health care industry in the United States. Biofilms are an important aspect of the UPEC pathogenesis cascade and for the establishment of chronic infections. Approximately 2% of UPEC isolates from UTIs are cysteine auxotrophs, yet there is relatively little known about the biofilm formation of UPEC cysteine auxotrophs. Here we show that cysteine auxotrophs have dysregulated biofilm components due to a change in the redox state of the periplasm. Additionally, we show the relationship between cysteine auxotrophs, biofilms, and antibiotics frequently used to treat UTIs.
Collapse
|
30
|
Sobke A, Makarewicz O, Baier M, Bär C, Pfister W, Gatermann SG, Pletz MW, Forstner C. Empirical treatment of lower urinary tract infections in the face of spreading multidrug resistance: in vitro study on the effectiveness of nitroxoline. Int J Antimicrob Agents 2017; 51:213-220. [PMID: 29111434 DOI: 10.1016/j.ijantimicag.2017.10.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 10/18/2017] [Accepted: 10/21/2017] [Indexed: 10/18/2022]
Abstract
The spread of antimicrobial resistance challenges the empirical treatment of urinary tract infections (UTIs). Among others, nitrofurantoin is recommended for first-line treatment, but acceptance among clinicians is limited due to chronic nitrofurantoin-induced lung toxicity and insufficient coverage of Enterobacteriaceae other than Escherichia coli. Nitroxoline appears to be an alternative to nitrofurantoin owing to its favourable safety profile, however data on its current in vitro susceptibility are sparse. In this study, susceptibility to nitroxoline was tested against 3012 urinary clinical isolates (including multidrug-resistant bacteria and Candida spp.) by disk diffusion test and/or broth microdilution. At least 91% of all Gram-negatives (n = 2000), Gram-positives (n = 403) and yeasts (n = 132) had inhibition zone diameters for nitroxoline ≥18 mm. Except for Pseudomonas aeruginosa, nitroxoline MIC90 values were ≤16 mg/L and were 2- to >16-fold lower compared with nitrofurantoin. In extended-spectrum β-lactamase (ESBL)-producing Enterobacteriaceae and methicillin-resistant Staphylococcus aureus (MRSA), MIC90 values of nitroxoline were two-fold higher compared with non-ESBL-producing enterobacteria and methicillin-susceptible S. aureus (MSSA). The in vitro efficacies of nitroxoline and nitrofurantoin against ATCC strains of E. coli, Enterococcus faecalis and Proteus mirabilis were compared by time-kill curves in Mueller-Hinton broth and artificial urine. Nitroxoline was non-inferior against E. coli, P. mirabilis and E. faecalis in artificial urine. In conclusion, nitroxoline showed a broad antimicrobial spectrum, with inhibition zone diameters and MICs of nitroxoline well below the EUCAST breakpoint for E. coli for most organisms, and thus may also be a target for therapy of uncomplicated UTIs.
Collapse
Affiliation(s)
- A Sobke
- Institute of Medical Microbiology, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
| | - O Makarewicz
- Center for Infectious Diseases and Infection Control, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
| | - M Baier
- Institute of Medical Microbiology, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
| | - C Bär
- Institute of Medical Microbiology, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
| | - W Pfister
- Institute of Medical Microbiology, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
| | - S G Gatermann
- Department of Medical Microbiology, Ruhr-University Bochum, Universitätsstraße 150, 44801, Bochum, Germany
| | - M W Pletz
- Center for Infectious Diseases and Infection Control, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
| | - C Forstner
- Center for Infectious Diseases and Infection Control, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany; Department of Medicine I, Division of Infectious Diseases and Tropical Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria.
| |
Collapse
|