51
|
Fait ME, Grillo PD, Garrote GL, Prieto ED, Vázquez RF, Saparrat MCN, Morcelle SR. Biocidal and antibiofilm activities of arginine-based surfactants against Candida isolates. Amino Acids 2023; 55:1083-1102. [PMID: 37382761 DOI: 10.1007/s00726-023-03296-z] [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: 03/27/2023] [Accepted: 06/19/2023] [Indexed: 06/30/2023]
Abstract
Amino-acid-based surfactants are a group of compounds that resemble natural amphiphiles and thus are expected to have a low impact on the environment, owing to either the mode of surfactant production or its means of disposal. Within this context, arginine-based tensioactives have gained particular interest, since their cationic nature-in combination with their amphiphilic character-enables them to act as broad-spectrum biocides. This capability is based mainly on their interactive affinity for the microbial envelope that alters the latter's structure and ultimately its function. In the work reported here, we investigated the efficiency of Nα-benzoyl arginine decyl- and dodecylamide against Candida spp. to further our understanding of the antifungal mechanism involved. For the assays, both a Candida albicans and a Candida tropicalis clinical isolates along with a C. albicans-collection strain were used as references. As expected, both arginine-based compounds proved to be effective against the strains tested through inhibiting both the planktonic and the sessile growth. Furthermore, atomic force microscopy techniques and lipid monolayer experiments enabled us to gain insight into the effect of the surfactant on the cellular envelope. The results demonstrated that all the yeasts treated exhibited changes in their exomorphologic structure, with respect to alterations in both roughness and stiffness, relative to the nontreated ones. This finding-in addition to the amphiphiles' proven ability to insert themselves within this model fungal membrane-could explain the changes in the yeast-membrane permeability that could be linked to viability loss and mixed-vesicle release.
Collapse
Affiliation(s)
- M Elisa Fait
- Centro de Investigación de Proteínas Vegetales (CIProVe-UNLP-Centro Asociado CICPBA), Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Centro Asociado CICPBA, Universidad Nacional de La Plata (UNLP), La Plata, Argentina.
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.
| | - Patricia D Grillo
- Centro de Investigación de Proteínas Vegetales (CIProVe-UNLP-Centro Asociado CICPBA), Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Centro Asociado CICPBA, Universidad Nacional de La Plata (UNLP), La Plata, Argentina
- Agencia Nacional de Promoción Científica y Tecnológica (ANPCyT), Buenos Aires, Argentina
| | - Graciela L Garrote
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Centro de Investigación y Desarrollo en Criotecnología de Alimentos (CIDCA, CONICET-UNLP-CICPBA), La Plata, Argentina
| | - Eduardo D Prieto
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA), CONICET, UNLP, CCT-La Plata, La Plata, Argentina
- Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
- Instituto Ciencias de la Salud, Universidad Nacional Arturo Jauretche, Buenos Aires, Argentina
| | - Romina F Vázquez
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), CCT-La Plata, CONICET, UNLP, La Plata, Argentina
| | - Mario C N Saparrat
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Instituto de Fisiología Vegetal (INFIVE-CONICET-UNLP) and Cátedra de Microbiología Agrícola, Facultad de Ciencias Agrarias y Forestales, UNLP, La Plata, Argentina
| | - Susana R Morcelle
- Centro de Investigación de Proteínas Vegetales (CIProVe-UNLP-Centro Asociado CICPBA), Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Centro Asociado CICPBA, Universidad Nacional de La Plata (UNLP), La Plata, Argentina.
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.
| |
Collapse
|
52
|
Enriquez KT, Plummer WD, Neufer PD, Chazin WJ, Dupont WD, Skaar EP. Temporal modelling of the biofilm lifecycle (TMBL) establishes kinetic analysis of plate-based bacterial biofilm dynamics. J Microbiol Methods 2023; 212:106808. [PMID: 37595876 PMCID: PMC10528067 DOI: 10.1016/j.mimet.2023.106808] [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: 06/30/2023] [Revised: 08/12/2023] [Accepted: 08/12/2023] [Indexed: 08/20/2023]
Abstract
Bacterial biofilms are critical to pathogenesis and infection. They are associated with rising rates of antimicrobial resistance. Biofilms are correlated with worse clinical outcomes, making them important to infectious diseases research. There is a gap in knowledge surrounding biofilm kinetics and dynamics which makes biofilm research difficult to translate from bench to bedside. To address this gap, this work employs a well-characterized crystal violet biomass accrual and planktonic cell density assay across a clinically relevant time course and expands statistical analysis to include kinetic information in a protocol termed the TMBL (Temporal Mapping of the Biofilm Lifecycle) assay. TMBL's statistical framework quantitatively compares biofilm communities across time, species, and media conditions in a 96-well format. Measurements from TMBL can reliably be condensed into response features that inform the time-dependent behavior of adherent biomass and planktonic cell populations. Staphylococcus aureus and Pseudomonas aeruginosa biofilms were grown in conditions of metal starvation in nutrient-variable media to demonstrate the rigor and translational potential of this strategy. Significant differences in single-species biofilm formation are seen in metal-deplete conditions as compared to their controls which is consistent with the consensus literature on nutritional immunity that metal availability drives transcriptomic and metabolomic changes in numerous pathogens. Taken together, these results suggest that kinetic analysis of biofilm by TMBL represents a statistically and biologically rigorous approach to studying the biofilm lifecycle as a time-dependent process. In addition to current methods to study the impact of microbe and environmental factors on the biofilm lifecycle, this kinetic assay can inform biological discovery in biofilm formation and maintenance.
Collapse
Affiliation(s)
- Kyle T Enriquez
- Vanderbilt University Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, TN, United States of America; Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN, United States of America; Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - W Dale Plummer
- Department of Biostatistics, Vanderbilt University, Nashville, TN, United States of America
| | - Preston D Neufer
- Department of Chemistry, Vanderbilt University, Nashville, TN, United States of America; Department of Biochemistry, Vanderbilt University, Nashville, TN, United States of America
| | - Walter J Chazin
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, United States of America; Department of Chemistry, Vanderbilt University, Nashville, TN, United States of America; Department of Biochemistry, Vanderbilt University, Nashville, TN, United States of America
| | - William D Dupont
- Department of Biostatistics, Vanderbilt University, Nashville, TN, United States of America
| | - Eric P Skaar
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN, United States of America; Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, United States of America.
| |
Collapse
|
53
|
Brčić J, Tong A, Wender PA, Cegelski L. Conjugation of Vancomycin with a Single Arginine Improves Efficacy against Mycobacteria by More Effective Peptidoglycan Targeting. J Med Chem 2023; 66:10226-10237. [PMID: 37477249 PMCID: PMC10783851 DOI: 10.1021/acs.jmedchem.3c00565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/22/2023]
Abstract
Drug resistant bacterial infections have emerged as one of the greatest threats to public health. The discovery and development of new antimicrobials and anti-infective strategies are urgently needed to address this challenge. Vancomycin is one of the most important antibiotics for the treatment of Gram-positive infections. Here, we introduce the vancomycin-arginine conjugate (V-R) as a highly effective antimicrobial against actively growing mycobacteria and difficult-to-treat mycobacterial biofilm populations. Further improvement in efficacy through combination treatment of V-R to inhibit peptidoglycan synthesis and ethambutol to inhibit arabinogalactan synthesis underscores the ability to identify compound synergies to more effectively target the Achilles heel of the cell-wall assembly. Moreover, we introduce mechanistic activity data and a molecular model derived from a d-Ala-d-Ala-bound vancomycin structure that we hypothesize underlies the molecular basis for the antibacterial improvement attributed to the arginine modification that is specific to peptidoglycan chemistry employed by mycobacteria and distinct from Gram-positive pathogens.
Collapse
Affiliation(s)
- Jasna Brčić
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Alan Tong
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Paul A. Wender
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA
| | - Lynette Cegelski
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
54
|
Kauser A, Parisini E, Suarato G, Castagna R. Light-Based Anti-Biofilm and Antibacterial Strategies. Pharmaceutics 2023; 15:2106. [PMID: 37631320 PMCID: PMC10457815 DOI: 10.3390/pharmaceutics15082106] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/29/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
Biofilm formation and antimicrobial resistance pose significant challenges not only in clinical settings (i.e., implant-associated infections, endocarditis, and urinary tract infections) but also in industrial settings and in the environment, where the spreading of antibiotic-resistant bacteria is on the rise. Indeed, developing effective strategies to prevent biofilm formation and treat infections will be one of the major global challenges in the next few years. As traditional pharmacological treatments are becoming inadequate to curb this problem, a constant commitment to the exploration of novel therapeutic strategies is necessary. Light-triggered therapies have emerged as promising alternatives to traditional approaches due to their non-invasive nature, precise spatial and temporal control, and potential multifunctional properties. Here, we provide a comprehensive overview of the different biofilm formation stages and the molecular mechanism of biofilm disruption, with a major focus on the quorum sensing machinery. Moreover, we highlight the principal guidelines for the development of light-responsive materials and photosensitive compounds. The synergistic effects of combining light-triggered therapies with conventional treatments are also discussed. Through elegant molecular and material design solutions, remarkable results have been achieved in the fight against biofilm formation and antibacterial resistance. However, further research and development in this field are essential to optimize therapeutic strategies and translate them into clinical and industrial applications, ultimately addressing the global challenges posed by biofilm and antimicrobial resistance.
Collapse
Affiliation(s)
- Ambreen Kauser
- Department of Biotechnology, Latvian Institute of Organic Synthesis, Aizkraukles 21, LV-1006 Riga, Latvia; (A.K.); (E.P.)
- Faculty of Materials Science and Applied Chemistry, Riga Technical University, Paula Valdena 3, LV-1048 Riga, Latvia
| | - Emilio Parisini
- Department of Biotechnology, Latvian Institute of Organic Synthesis, Aizkraukles 21, LV-1006 Riga, Latvia; (A.K.); (E.P.)
- Department of Chemistry “G. Ciamician”, University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| | - Giulia Suarato
- Istituto di Elettronica e di Ingegneria dell’Informazione e delle Telecomunicazioni, Consiglio Nazionale delle Ricerche, CNR-IEIIT, Piazza Leonardo da Vinci 32, 20133 Milano, Italy
| | - Rossella Castagna
- Department of Biotechnology, Latvian Institute of Organic Synthesis, Aizkraukles 21, LV-1006 Riga, Latvia; (A.K.); (E.P.)
- Dipartimento di Chimica, Materiali e Ingegneria Chimica “G. Natta”, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133 Milano, Italy
| |
Collapse
|
55
|
Tran NN, Morrisette T, Jorgensen SCJ, Orench-Benvenutti JM, Kebriaei R. Current therapies and challenges for the treatment of Staphylococcus aureus biofilm-related infections. Pharmacotherapy 2023; 43:816-832. [PMID: 37133439 DOI: 10.1002/phar.2806] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 03/10/2023] [Accepted: 03/16/2023] [Indexed: 05/04/2023]
Abstract
Staphylococcus aureus is a major cause of nosocomial and community-acquired infections and contributes to significant increase in morbidity and mortality especially when associated with medical devices and in biofilm form. Biofilm structure provides a pathway for the enrichment of resistant and persistent phenotypes of S. aureus leading to relapse and recurrence of infection. Minimal diffusion of antibiotics inside biofilm structure leads to heterogeneity and distinct physiological activity. Additionally, horizontal gene transfer between cells in proximity adds to the challenges associated with eradication of biofilms. This narrative review focuses on biofilm-associated infections caused by S. aureus, the impact of environmental conditions on biofilm formation, interactions inside biofilm communities, and the clinical challenges that they present. Conclusively, potential solutions, novel treatment strategies, combination therapies, and reported alternatives are discussed.
Collapse
Affiliation(s)
- Nikki N Tran
- Department of Pharmacy, The Ohio State University Wexner Medical Center - The James Cancer Hospital and Solove Research Institute, Columbus, Ohio, USA
| | - Taylor Morrisette
- Department of Clinical Pharmacy and Outcomes Sciences, Medical University of South Carolina College of Pharmacy, Charleston, South Carolina, USA
- Department of Pharmacy Services, Medical University of South Carolina Shawn Jenkins Children's Hospital, Charleston, South Carolina, USA
| | - Sarah C J Jorgensen
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - José M Orench-Benvenutti
- P3 Research Laboratory, Division of Outcomes and Translational Sciences, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Razieh Kebriaei
- P3 Research Laboratory, Division of Outcomes and Translational Sciences, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
56
|
Mahmoud M, Richter P, Lebert M, Burkovski A. Photodynamic Activity of Chlorophyllin and Polyethylenimine on Pseudomonas aeruginosa Planktonic, Biofilm and Persister Cells. Int J Mol Sci 2023; 24:12098. [PMID: 37569471 PMCID: PMC10419130 DOI: 10.3390/ijms241512098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Antimicrobial photodynamic inactivation is considered a promising antimicrobial approach that may not develop resistance in the near future. Here, we investigate the influence of the photosensitizer chlorophyllin (CHL) and the cationic permeabilizer polyethylenimine (PEI), exposed to a red light-emitting diode, on the human pathogen Pseudomonas aeruginosa free-living planktonic cells, the sessile biofilm and persister cells. The broth microdilution checkerboard method was used to test antimicrobial susceptibility. As a substrate for biofilms, the Calgary biofilm device was used, and the quantification of the biofilm biomass was carried out using a crystal violet assay. Serine hydroxamate was used for the induction of persisters. Our findings reveal that PEI ameliorates the antimicrobial activity of CHL against P. aeruginosa planktonic and biofilm states, and the concentration required to eradicate the bacteria in the biofilm is more than fourfold that is required to eradicate planktonic cells. Interestingly, the persister cells are more susceptible to CHL/PEI (31.25/100 µg mL-1) than the growing cells by 1.7 ± 0.12 and 0.4 ± 0.1 log10 reduction, respectively, after 15 min of illumination. These data demonstrate that CHL excited with red light together with PEI is promising for the eradication of P. aeruginosa, and the susceptibility of P. aeruginosa to CHL/PEI is influenced by the concentrations and the exposure time.
Collapse
Affiliation(s)
- Mona Mahmoud
- Department of Biology, Microbiology Division, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany; (M.M.); (A.B.)
- Dairy Department, National Research Centre, Dokki, Giza 12622, Egypt
| | - Peter Richter
- Gravitational Biology Group, Department of Biology, Cell Biology Division, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany;
| | - Michael Lebert
- Gravitational Biology Group, Department of Biology, Cell Biology Division, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany;
- Space Biology Unlimited S.A.S., 33000 Bordeaux, France
| | - Andreas Burkovski
- Department of Biology, Microbiology Division, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany; (M.M.); (A.B.)
| |
Collapse
|
57
|
McLeod D, Wei L, Li Z. A standard 96-well based high throughput microfluidic perfusion biofilm reactor for in situ optical analysis. Biomed Microdevices 2023; 25:26. [PMID: 37493818 DOI: 10.1007/s10544-023-00668-w] [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] [Accepted: 07/18/2023] [Indexed: 07/27/2023]
Abstract
Biofilm infections represent a major public health threat due to their high tolerance to antimicrobials and the lack of specific anti-biofilm drugs. To develop such drugs, it is crucial to have high-throughput biofilm growth systems that can emulate in vivo conditions without the cost and complexity of animal models. However, no current biofilm reactor can provide in vivo-like conditions in a high throughput standard microtiter format. This paper demonstrates a novel high-throughput (HT) microfluidic perfusion biofilm reactor (HT-μPBR) compatible with a standard 96-well microtiter plate for in situ optical analysis. A snap-on liquid-tight cover for standard microtiter plates was designed and fabricated with fluidic channels to provide closed-loop recirculating perfusion. Our system takes steps toward providing in vivo-like conditions with controlled shear stress and nutrient delivery. We describe the system fabrication and usage in optical analysis of biomass and viability of Escherichia coli (E. coli) biofilms. The HT-μPBR was set to perfuse at 1 mL/min corresponding to an average shear rate of approximately [Formula: see text] on the bottom surface of a single well. Biofilms were detected on well plate bottoms and measured using a fluorescence microscope and plate reader to determine biomass and viability. Samples cultured in the HT-μPBR showed increased biomass while maintaining viability after 24 h. The HT-μPBR can further be combined with HT antibiotic susceptibility testing and additional optical techniques such as time-lapse imaging to improve understanding of the drug reaction mechanism as well as the optimization of drug combinations and delivery profiles.
Collapse
Affiliation(s)
- David McLeod
- Department of Biomedical Engineering, The George Washington University, 800 22nd St NW, Washington, DC, USA
| | - Lai Wei
- Department of Biomedical Engineering, The George Washington University, 800 22nd St NW, Washington, DC, USA
| | - Zhenyu Li
- Department of Biomedical Engineering, The George Washington University, 800 22nd St NW, Washington, DC, USA.
| |
Collapse
|
58
|
Kurbatfinski N, Kramer CN, Goodman SD, Bakaletz LO. ESKAPEE pathogens newly released from biofilm residence by a targeted monoclonal are sensitized to killing by traditional antibiotics. Front Microbiol 2023; 14:1202215. [PMID: 37564292 PMCID: PMC10410267 DOI: 10.3389/fmicb.2023.1202215] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 06/30/2023] [Indexed: 08/12/2023] Open
Abstract
Introduction The "silent" antimicrobial resistance (AMR) pandemic is responsible for nearly five million deaths annually, with a group of seven biofilm-forming pathogens, known as the ESKAPEE pathogens, responsible for 70% of these fatalities. Biofilm-resident bacteria, as they exist within the disease site, are canonically highly resistant to antibiotics. One strategy to counter AMR and improve disease resolution involves developing methods to disrupt biofilms. These methods aim to release bacteria from the protective biofilm matrix to facilitate their killing by antibiotics or immune effectors. Several laboratories working on such strategies have demonstrated that bacteria newly released from a biofilm display a transient phenotype of significantly increased susceptibility to antibiotics. Similarly, we developed an antibody-based approach for biofilm disruption directed against the two-membered DNABII family of bacterial DNA-binding proteins, which serve as linchpins to stabilize the biofilm matrix. The incubation of biofilms with α-DNABII antibodies rapidly collapses them to induce a population of newly released bacteria (NRel). Methods In this study, we used a humanized monoclonal antibody (HuTipMab) directed against protective epitopes of a DNABII protein to determine if we could disrupt biofilms formed by the high-priority ESKAPEE pathogens as visualized by confocal laser scanning microscopy (CLSM) and COMSTAT2 analysis. Then, we demonstrated the potentiated killing of the induced NRel by seven diverse classes of traditional antibiotics by comparative plate count. Results To this end, ESKAPEE biofilms were disrupted by 50%-79% using a single tested dose and treatment period with HuTipMab. The NRel of each biofilm were significantly more sensitive to killing than their planktonically grown counterparts (heretofore, considered to be the most sensitive to antibiotic-mediated killing), even when tested at a fraction of the MIC (1/250-1/2 MIC). Moreover, the bacteria that remained within the biofilms of two representative ESKAPEE pathogens after HuTipMab disruption were also significantly more susceptible to killing by antibiotics. Discussion New data presented in this study support our continued development of a combinatorial therapy wherein HuTipMab is delivered to a patient with recalcitrant disease due to an ESKAPEE pathogen to disrupt a pathogenic biofilm, along with a co-delivered dose of an antibiotic whose ability to rapidly kill the induced NRel has been demonstrated. This novel regimen could provide a more successful clinical outcome to those with chronic, recurrent, or recalcitrant diseases, while limiting further contribution to AMR.
Collapse
Affiliation(s)
- Nikola Kurbatfinski
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | - Cameron N. Kramer
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | - Steven D. Goodman
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, United States
| | - Lauren O. Bakaletz
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, United States
| |
Collapse
|
59
|
Kalpana S, Lin WY, Wang YC, Fu Y, Wang HY. Alternate Antimicrobial Therapies and Their Companion Tests. Diagnostics (Basel) 2023; 13:2490. [PMID: 37568853 PMCID: PMC10417861 DOI: 10.3390/diagnostics13152490] [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: 06/30/2023] [Accepted: 07/14/2023] [Indexed: 08/13/2023] Open
Abstract
New antimicrobial approaches are essential to counter antimicrobial resistance. The drug development pipeline is exhausted with the emergence of resistance, resulting in unsuccessful trials. The lack of an effective drug developed from the conventional drug portfolio has mandated the introspection into the list of potentially effective unconventional alternate antimicrobial molecules. Alternate therapies with clinically explicable forms include monoclonal antibodies, antimicrobial peptides, aptamers, and phages. Clinical diagnostics optimize the drug delivery. In the era of diagnostic-based applications, it is logical to draw diagnostic-based treatment for infectious diseases. Selection criteria of alternate therapeutics in infectious diseases include detection, monitoring of response, and resistance mechanism identification. Integrating these diagnostic applications is disruptive to the traditional therapeutic development. The challenges and mitigation methods need to be noted. Applying the goals of clinical pharmacokinetics that include enhancing efficacy and decreasing toxicity of drug therapy, this review analyses the strong correlation of alternate antimicrobial therapeutics in infectious diseases. The relationship between drug concentration and the resulting effect defined by the pharmacodynamic parameters are also analyzed. This review analyzes the perspectives of aligning diagnostic initiatives with the use of alternate therapeutics, with a particular focus on companion diagnostic applications in infectious diseases.
Collapse
Affiliation(s)
- Sriram Kalpana
- Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan 333423, Taiwan;
| | - Wan-Ying Lin
- Department of Medicine, University of California San Diego, San Diego, CA 92093, USA;
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA;
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Yu-Chiang Wang
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA;
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Yiwen Fu
- Department of Medicine, Kaiser Permanente Santa Clara Medical Center, Santa Clara, CA 95051, USA;
| | - Hsin-Yao Wang
- Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan 333423, Taiwan;
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA;
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
| |
Collapse
|
60
|
Kebriaei R, Lehman SM, Shah RM, Stamper KC, Kunz Coyne AJ, Holger D, El Ghali A, Rybak MJ. Optimization of Phage-Antibiotic Combinations against Staphylococcus aureus Biofilms. Microbiol Spectr 2023; 11:e0491822. [PMID: 37199616 PMCID: PMC10269792 DOI: 10.1128/spectrum.04918-22] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 04/21/2023] [Indexed: 05/19/2023] Open
Abstract
Phage therapy has gained attention due to the spread of antibiotic-resistant bacteria and narrow pipeline of novel antibiotics. Phage cocktails are hypothesized to slow the overall development of resistance by challenging the bacteria with more than one phage. Here, we have used a combination of plate-, planktonic-, and biofilm-based screening assays to try to identify phage-antibiotic combinations that will eradicate preformed biofilms of Staphylococcus aureus strains that are otherwise difficult to kill. We have focused on methicillin-resistant S aureus (MRSA) strains and their daptomycin-nonsusceptible vancomycin-intermediate (DNS-VISA) derivatives to understand whether the phage-antibiotic interactions are altered by the changes associated with evolution from MRSA to DNS-VISA (which is known to occur in patients receiving antibiotic therapy). We evaluated the host range and cross-resistance patterns of five obligately lytic S. aureus myophages to select a three-phage cocktail. We screened these phages for their activity against 24-h bead biofilms and found that biofilms of two strains, D712 (DNS-VISA) and 8014 (MRSA), were the most resistant to killing by single phages. Specifically, even initial phage concentrations of 107 PFU per well could not prevent visible regrowth of bacteria from the treated biofilms. However, when we treated biofilms of the same two strains with phage-antibiotic combinations, we prevented bacterial regrowth when using up to 4 orders of magnitude less phage and antibiotic concentrations that were lower than our measured minimum biofilm inhibitory concentration. We did not see a consistent association between phage activity and the evolution of DNS-VISA genotypes in this small number of bacterial strains. IMPORTANCE The extracellular polymeric matrix of biofilms presents an impediment to antibiotic diffusion, facilitating the emergence of multidrug-resistant populations. While most phage cocktails are designed for the planktonic state of bacteria, it is important to take the biofilm mode of growth (the predominant mode of bacterial growth in nature) into consideration, as it is unclear how interactions between any specific phage and its bacterial hosts will depend on the physical properties of the growth environment. In addition, the extent of bacterial sensitivity to any given phage may vary from the planktonic to the biofilm state. Therefore, phage-containing treatments targeting biofilm infections such as catheters and prosthetic joint material may not be merely based on host range characteristics. Our results open avenues to new questions regarding phage-antibiotic treatment efficiency in the eradication of topologically structured biofilm settings and the extent of eradication efficacy relative to the single agents in biofilm populations.
Collapse
Affiliation(s)
- Razieh Kebriaei
- Anti-Infective Research Laboratory, College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Susan M. Lehman
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Rahi M. Shah
- Anti-Infective Research Laboratory, College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Kyle C. Stamper
- Anti-Infective Research Laboratory, College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Ashlan J. Kunz Coyne
- Anti-Infective Research Laboratory, College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Dana Holger
- Anti-Infective Research Laboratory, College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Amer El Ghali
- Anti-Infective Research Laboratory, College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Michael J. Rybak
- Anti-Infective Research Laboratory, College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
- School of Medicine, Wayne State University, Detroit, Michigan, USA
| |
Collapse
|
61
|
Noach N, Lavy E, Reifen R, Friedman M, Kirmayer D, Zelinger E, Ritter A, Yaniv D, Reifen E. Zinc chloride is effective as an antibiotic in biofilm prevention following septoplasty. Sci Rep 2023; 13:8344. [PMID: 37221180 DOI: 10.1038/s41598-023-35069-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 05/11/2023] [Indexed: 05/25/2023] Open
Abstract
Biofilm-state bacterial infections associated with inserted medical devices constitute a massive health and financial problem worldwide. Although bacteria exhibit significantly lower susceptibility to antibiotics in the biofilm state, the most common treatment approach still relies on antibiotics, exacerbating the phenomenon of antibiotic-resistant bacteria. In this study, we aimed to assess whether ZnCl2 coating of intranasal silicone splints (ISSs) can reduce the biofilm infections associated with the insertion of these devices and prevent the overuse of antibiotics while minimizing waste, pollution and costs. We tested the ability of ZnCl2 to prevent biofilm formation on ISS both in vitro and in vivo by using the microtiter dish biofilm formation assay, crystal violet staining, and electron and confocal microscopy. We found a significant decrease in biofilm formation between the treatment group and the growth control when ZnCl2-coated splints were placed in patients' nasal flora. According to these results, infections associated with ISS insertion may be prevented by using ZnCl2 coating, thereby obviating the overuse and abuse of antibiotics.
Collapse
Affiliation(s)
- Noa Noach
- The Institute of Biochemistry, Food Science and Nutrition. The Robert H. Smith Faculty of Agriculture, Food & Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Eran Lavy
- The Koret School of Veterinary Medicine. The Robert H. Smith Faculty of Agriculture, Food & Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Ram Reifen
- The Institute of Biochemistry, Food Science and Nutrition. The Robert H. Smith Faculty of Agriculture, Food & Environment, The Hebrew University of Jerusalem, Rehovot, Israel.
| | - Michael Friedman
- The School of Pharmacy, The Faculty of Medicine, Ein Kerem Campus, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - David Kirmayer
- The School of Pharmacy, The Faculty of Medicine, Ein Kerem Campus, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Einat Zelinger
- FACSI-Faculty of Agriculture Center for Scientific Imaging. The Robert H. Smith Faculty of Agriculture, Food & Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Amit Ritter
- Department of Otolaryngology, Head and Neck Surgery, Rabin Medical Center, Petah Tikva, Israel
| | - Dan Yaniv
- Department of Otolaryngology, Head and Neck Surgery, Rabin Medical Center, Petah Tikva, Israel
| | - Ella Reifen
- Department of Otolaryngology, Head and Neck Surgery, Rabin Medical Center, Petah Tikva, Israel
| |
Collapse
|
62
|
Ge KX, Lung CYK, Lam WYH, Chu CH, Yu OY. A novel glass ionomer cement with silver zeolite for restorative dentistry. J Dent 2023; 133:104524. [PMID: 37080532 DOI: 10.1016/j.jdent.2023.104524] [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: 02/22/2023] [Revised: 04/15/2023] [Accepted: 04/17/2023] [Indexed: 04/22/2023] Open
Abstract
OBJECTIVE To develop an antimicrobial silver zeolite glass ionomer cement (SZ-GIC) and determine its biocompatibility, physical, adhesive and antibacterial properties. METHODS Silver nitrate and sodium zeolite were used to synthesize silver zeolite (SZ). SZ-GICs were prepared by incorporating SZ into GIC at 5% (SZ-GIC5), 2% (SZ-GIC2), or 1% (SZ-GIC1) by weight, respectively. The SZ-GICs were characterized by evaluating surface morphology, topography and elemental composition. SZ-GICs' biocompatibility was assessed by evaluating cell cytotoxicity. Their physical properties were determined by testing setting time, compressive strength, flexural strength, water sorption and solubility. Their adhesive property was assessed by evaluating micro-tensile bond strength. Their antibacterial properties were assessed by evaluating biofilm growth kinetic, metabolic activity, viability and morphology. GIC was used as a control. RESULTS SZ was a three-dimensional crystalline mineral. SZ-GICs (including SZ-GIC 5, 2 and 1) showed similar surface morphology and topography to GIC. SZ-GIC1 and GIC had no difference in cell cytotoxicity (p>0.05). SZ-GICs and GIC showed no difference in setting time (p>0.05). SZ-GICs had higher compressive and flexural strength than GIC (p<0.05). SZ-GIC2 and SZ-GIC1 showed lower water sorption and solubility than GIC (p<0.05). SZ-GICs had higher micro-tensile bond strength than GIC (p<0.05). Biofilms on SZ-GICs' surfaces showed lower colony-forming units, decreased metabolic activities, higher percentages of dead cells and more ruptured bacterial cells compared with those on GIC. CONCLUSION SZ-GIC with silver zeolite at 1% by weight are as biocompatible as conventional GIC. The SZ-GICs have enhanced physical, adhesive and antibacterial properties than GIC. CLINICAL SIGNIFICANCE A silver zeolite glass ionomer cement was developed. The SZ-GICs have great potential for caries prevention and management.
Collapse
Affiliation(s)
- Kelsey Xingyun Ge
- Faculty of Dentistry, The University of Hong Kong, Hong Kong, S.A.R., China
| | | | - Walter Yu-Hang Lam
- Faculty of Dentistry, The University of Hong Kong, Hong Kong, S.A.R., China
| | - Chun-Hung Chu
- Faculty of Dentistry, The University of Hong Kong, Hong Kong, S.A.R., China
| | - Ollie Yiru Yu
- Faculty of Dentistry, The University of Hong Kong, Hong Kong, S.A.R., China.
| |
Collapse
|
63
|
Niu M, Gu X, Yang J, Cui H, Hou X, Ma Y, Wang C, Wei G. Dual-Mechanism Glycolipidpeptide with High Antimicrobial Activity, Immunomodulatory Activity, and Potential Application for Combined Antibacterial Therapy. ACS NANO 2023; 17:6292-6316. [PMID: 36951612 DOI: 10.1021/acsnano.2c10249] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Bacterial drug resistance is becoming increasingly serious, and it is urgent to develop effective antibacterial drugs. Antimicrobial peptides (AMPs), as potential candidates against bacteria, have a broad prospect for development. Herein, a series of AMPs with biological characteristics (net positive charge, amphiphilicity, and α-helix), an AXA motif recognized by membrane bound serine protease type I signal peptidases (SPase I), an FLPII motif to reduce hemolysis, and a monosaccharide motif to improve the stability and activity were designed and synthesized, and among which, the glycolipidpeptide GLP6 (glycosylated LP6 lipopeptide) had excellent antibacterial and immunomodulatory activity, good stability and biocompatibility, and excellent biofilm eradication and membrane penetrating activity. The positively charged spherical aggregates formed by self-assembly of GLP6 could encapsulate tetracycline (TC) to form GLP6@TC with a sustained-release effect, which could enhance the sensitivity of bacteria to the antibiotic and realize combined sterilization. The results of acute peritonitis and bacterial keratitis showed that GLP6@TC had a good combined antibacterial effect and the ability to inhibit interleukin-2 (IL-2), which could significantly reduce the inflammatory response while treating bacterial infection, and it had great potential for application. The results of computer molecular docking showed the AXA motif could effectively bind to SPase I, which was consistent with the results of biological experiments. In general, the study could provide a perspective for the design of AMPs and combined antibacterial therapy.
Collapse
Affiliation(s)
- Mingcong Niu
- Department of Pharmacy Science, Binzhou Medical University, Yantai 264003, China
| | - Xiulian Gu
- Department of Pharmacy Science, Binzhou Medical University, Yantai 264003, China
| | - Jingyi Yang
- Department of Pharmacy Science, Binzhou Medical University, Yantai 264003, China
| | - Haoyu Cui
- Department of Pharmacy Science, Binzhou Medical University, Yantai 264003, China
| | - Xinyi Hou
- Department of Pharmacy Science, Binzhou Medical University, Yantai 264003, China
| | - Yue Ma
- Department of Pharmacy Science, Binzhou Medical University, Yantai 264003, China
| | - Chunhua Wang
- Department of Pharmacy Science, Binzhou Medical University, Yantai 264003, China
| | - Guangcheng Wei
- Department of Pharmacy Science, Binzhou Medical University, Yantai 264003, China
| |
Collapse
|
64
|
Rajapaksha DC, Edirisinghe SL, Nikapitiya C, Whang I, De Zoysa M. The Antimicrobial Peptide Octopromycin Suppresses Biofilm Formation and Quorum Sensing in Acinetobacter baumannii. Antibiotics (Basel) 2023; 12:antibiotics12030623. [PMID: 36978490 PMCID: PMC10044867 DOI: 10.3390/antibiotics12030623] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/03/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Acinetobacter baumannii is an opportunistic bacterial pathogen that causes severe infections in immunocompromised individuals. A. baumannii forms biofilm and produces extracellular matrix, which supports bacteria to survive under harsh conditions and be resistant to antibacterial treatments. In the present study, we investigated the biofilm and quorum-sensing inhibitory effects of antimicrobial peptide, octopromycin in A. baumannii. Field emission-scanning electron microscopy results clearly showed significantly reduced biofilm mass and caused a collapse in biofilm architecture at the minimum inhibitory concentration (50 µg/mL) and minimum bactericidal concentration (200 µg/mL) of octopromycin. Antibiotic-resistant persister cells of A. baumannii were successfully killed by octopromycin treatment, and it inhibited violacein production in Chromobacterium violaceum in a concentration-dependent manner. Octopromycin also inhibited alginate production, surface movements (swarming and swimming), and twitching motility of A. baumannnii, confirming its anti-quorum-sensing activity. Multiple metabolic pathways, two-component regulation systems, quorum-sensing, and antibiotic synthesis-related pathways in A. baumannii biofilms were strongly affected by octopromycin treatment. The collective findings indicate that the antibacterial peptide octopromycin may control A. baumannii biofilms through multi-target interactions. Octopromycin could be a desirable therapeutic option for the prevention and control of A. baumannii infections.
Collapse
Affiliation(s)
- Dinusha Chathurangi Rajapaksha
- College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Shan Lakmal Edirisinghe
- College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Chamilani Nikapitiya
- College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Ilson Whang
- National Marine Biodiversity Institute of Korea (MABIK), 75, Jangsan-ro 101 beon-gil, Janghang-eup, Seochun-gun 33662, Chungchungnam-do, Republic of Korea
| | - Mahanama De Zoysa
- College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| |
Collapse
|
65
|
Antimicrobial and Antibiofilm Potential of Thymus vulgaris and Cymbopogon flexuosus Essential Oils against Pure and Mixed Cultures of Foodborne Bacteria. Antibiotics (Basel) 2023; 12:antibiotics12030565. [PMID: 36978432 PMCID: PMC10044171 DOI: 10.3390/antibiotics12030565] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/07/2023] [Accepted: 03/10/2023] [Indexed: 03/18/2023] Open
Abstract
The spread of pathogenic and food spoilage microorganisms through the food chain still faces major mitigation challenges, despite modern advances. Although multiple cleaning and disinfection procedures are available for microbial load reduction in food-related settings, microbes can still remain on surfaces, equipment, or machinery, especially if they have the ability to form biofilms. The present study assessed the biofilm-forming properties of pure and mixed cultures of foodborne and spoilage bacteria (Listeria monocytogenes, Enterococcus faecalis, Aeromonas hydrophila, Brochothrix thermosphacta), using polystyrene and stainless steel contact surfaces. Subsequently, the antimicrobial and antibiofilm properties of Thymus vulgaris and Cymbopogon flexuosus essential oils—EOs—were evaluated against these bacteria. Moreover, in silico prediction of the absorption and toxicity values of the EOs’ major constituents was also performed, perceiving the putative application in food-related settings. Overall, biofilm formation was observed for all microbes under study, at different temperatures and both contact surfaces. In polystyrene, at 25 °C, when comparing pure with mixed cultures, the combination Listeria–Aeromonas achieved the highest biofilm biomass. Moreover, at 4 °C, increased biofilm formation was detected in stainless steel. Regarding thyme, this EO showed promising antimicrobial features (especially against A. hydrophila, with a MIC of 0.60 µg/µL) and antibiofilm abilities (MBEC of 110.79 µg/µL against L. monocytogenes, a major concern in food settings). As for lemongrass EO, the highest antimicrobial activity, with a MIC of 0.49 µg/µL, was also observed against L. monocytogenes. Overall, despite promising results, the in situ effectiveness of these essential oils, alone or in combination with other antimicrobial compounds, should be further explored.
Collapse
|
66
|
Rottier W, Seidelman J, Wouthuyzen-Bakker M. Antimicrobial treatment of patients with a periprosthetic joint infection: basic principles. ARTHROPLASTY 2023; 5:10. [PMID: 36864531 PMCID: PMC9979546 DOI: 10.1186/s42836-023-00169-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 01/16/2023] [Indexed: 03/04/2023] Open
Abstract
The antibiotic treatment of periprosthetic joint infections (PJI) is complicated by the presence of biofilm produced by bacteria on the abiotic surface of the implant. Bacteria within the deeper layers of the biofilm become metabolically less active, resulting in antibiotic tolerance due to several mechanisms. This review describes the basic principles of antibiotic treatment in PJI in relation to the behavior of bacteria within the biofilm. The concept of biofilm-active antibiotics will be explained from an in vitro as well as in vivo perspective. Evidence from clinical studies on biofilm-active antibiotics in PJI will be highlighted, mainly focusing on the role of rifampicin for Gram-positive microorganisms and fluoroquinolones for Gram-negative microorganisms. The optimal treatment duration will be discussed as the timing of switching to oral antibiotic therapy.
Collapse
Affiliation(s)
- Wouter Rottier
- grid.4830.f0000 0004 0407 1981Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, 9713 GZ the Netherlands
| | - Jessica Seidelman
- grid.26009.3d0000 0004 1936 7961Division of Infectious Diseases, Duke University School of Medicine, Durham, NC 27708 USA
| | - Marjan Wouthuyzen-Bakker
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, 9713 GZ, the Netherlands.
| |
Collapse
|
67
|
Han Y, Jiang N, Xu H, Yuan Z, Xiu J, Mao S, Liu X, Huang J. Extracellular Matrix Rigidities Regulate the Tricarboxylic Acid Cycle and Antibiotic Resistance of Three-Dimensionally Confined Bacterial Microcolonies. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206153. [PMID: 36658695 PMCID: PMC10037996 DOI: 10.1002/advs.202206153] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/22/2022] [Indexed: 06/06/2023]
Abstract
As a major cause of clinical chronic infection, microbial biofilms/microcolonies in host tissues essentially live in 3D-constrained microenvironments, which potentially modulate their spatial self-organization and morphodynamics. However, it still remains unclear whether and how mechanical cues of 3D confined microenvironments, for example, extracellular matrix (ECM) stiffness, exert an impact on antibiotic resistance of bacterial biofilms/microcolonies. With a high-throughput antibiotic sensitivity testing (AST) platform, it is revealed that 3D ECM rigidities greatly modulate their resistance to diverse antibiotics. The microcolonies in 3D ECM with human tissue-specific rigidities varying from 0.5 to 20 kPa show a ≈2-10 000-fold increase in minimum inhibitory concentration, depending on the types of antibiotics. The authors subsequently identified that the increase in 3D ECM rigidities leads to the downregulation of the tricarboxylic acid (TCA) cycle, which is responsible for enhanced antibiotic resistance. Further, it is shown that fumarate, as a potentiator of TCA cycle activity, can alleviate the elevated antibiotic resistance and thus remarkably improve the efficacy of antibiotics against bacterial microcolonies in 3D confined ECM, as confirmed in the chronic infection mice model. These findings suggest fumarate can be employed as an antibiotic adjuvant to effectively treat infections induced by bacterial biofilms/microcolonies in a 3D-confined environment.
Collapse
Affiliation(s)
- Yiming Han
- Department of Mechanics and Engineering Science, and Beijing Innovation Center for Engineering Science and Advanced TechnologyCollege of EngineeringPeking University100871BeijingChina
| | - Nan Jiang
- Department of Mechanics and Engineering Science, and Beijing Innovation Center for Engineering Science and Advanced TechnologyCollege of EngineeringPeking University100871BeijingChina
| | - Hongwei Xu
- Department of Mechanics and Engineering Science, and Beijing Innovation Center for Engineering Science and Advanced TechnologyCollege of EngineeringPeking University100871BeijingChina
| | - Zuoying Yuan
- Department of Mechanics and Engineering Science, and Beijing Innovation Center for Engineering Science and Advanced TechnologyCollege of EngineeringPeking University100871BeijingChina
| | - Jidong Xiu
- Department of Mechanics and Engineering Science, and Beijing Innovation Center for Engineering Science and Advanced TechnologyCollege of EngineeringPeking University100871BeijingChina
| | - Sheng Mao
- Department of Mechanics and Engineering Science, and Beijing Innovation Center for Engineering Science and Advanced TechnologyCollege of EngineeringPeking University100871BeijingChina
| | - Xiaozhi Liu
- Tianjin Key Laboratory of Epigenetics for Organ Development of Premature InfantsFifth Central Hospital of TianjinTianjin300450China
| | - Jianyong Huang
- Department of Mechanics and Engineering Science, and Beijing Innovation Center for Engineering Science and Advanced TechnologyCollege of EngineeringPeking University100871BeijingChina
| |
Collapse
|
68
|
Öztürk FY, Darcan C, Kariptaş E. The Determination, Monitoring, Molecular Mechanisms and Formation of Biofilm in E. coli. Braz J Microbiol 2023; 54:259-277. [PMID: 36577889 PMCID: PMC9943865 DOI: 10.1007/s42770-022-00895-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 12/16/2022] [Indexed: 12/30/2022] Open
Abstract
Biofilms are cell assemblies embedded in an exopolysaccharide matrix formed by microorganisms of a single or many different species. This matrix in which they are embedded protects the bacteria from external influences and antimicrobial effects. The biofilm structure that microorganisms form to protect themselves from harsh environmental conditions and survive is found in nature in many different environments. These environments where biofilm formation occurs have in common that they are in contact with fluids. The gene expression of bacteria in complex biofilm differs from that of bacteria in the planktonic state. The differences in biofilm cell expression are one of the effects of community life. Means of quorum sensing, bacteria can act in coordination with each other. At the same time, while biofilm formation provides many benefits to bacteria, it has positive and negative effects in many different areas. Depending on where they occur, biofilms can cause serious health problems, contamination risks, corrosion, and heat and efficiency losses. However, they can also be used in water treatment plants, bioremediation, and energy production with microbial fuel cells. In this review, the basic steps of biofilm formation and biofilm regulation in the model organism Escherichia coli were discussed. Finally, the methods by which biofilm formation can be detected and monitored were briefly discussed.
Collapse
Affiliation(s)
- Fırat Yavuz Öztürk
- Department of Molecular Biology and Genetic, Faculty of Arts and Science, Bilecik Seyh Edebali University, Bilecik, Turkey.
| | - Cihan Darcan
- Department of Molecular Biology and Genetic, Faculty of Arts and Science, Bilecik Seyh Edebali University, Bilecik, Turkey
| | - Ergin Kariptaş
- Department of Medical Microbiology, Faculty of Medicine, Samsun University, Samsun, Turkey
| |
Collapse
|
69
|
Ghodake V, Dhoble S, Vavilala SL, Patravale V. Anti-biofilm potential against P. aeruginosa biofilm in cystic fibrosis infection by systemically developed garlic extract incorporated liposomal formulation. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
|
70
|
Ali S, Faqir N, Naz F, Jan MI, Khan N, Alotaibi A, Ullah R. A Comprehensive Mechanistic Antibacterial and Antibiofilm Study of Potential Bioactive ((BpA) 2bp)Cu/Zn Complexes via Bactericidal Mechanisms against Escherichia coli. Molecules 2023; 28:molecules28052215. [PMID: 36903459 PMCID: PMC10005605 DOI: 10.3390/molecules28052215] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 02/24/2023] [Accepted: 02/24/2023] [Indexed: 03/04/2023] Open
Abstract
Bacterial resistance to antibiotics and host defense systems is primarily due to bacterial biofilm formation in antibiotic therapy. In the present study, two complexes, bis (biphenyl acetate) bipyridine Cu (II) (1) and bis (biphenyl acetate) bipyridine Zn (II) (2), were tested for their ability to prevent biofilm formation. The minimum inhibitory concentration and minimum bactericidal concentration of complexes 1 and 2 were 46.87 ± 1.822 and 93.75 ± 1.345 and 47.87 ± 1.345 and 94.85 ± 1.466 μg/mL, respectively. The significant activity of both complexes was attributed to the damage caused at the membrane level and was confirmed using an imaging technique. The biofilm inhibitory potential levels of complexes 1 and 2 were 95% and 71%, respectively, while the biofilm eradication potential levels were 95% and 35%, respectively, for both complexes. Both the complexes showed good interactions with the E. coli DNA. Thus, complexes 1 and 2 are good antibiofilm agents that exert their bactericidal actions possibly by disrupting the bacterial membrane and interacting with the bacterial DNA, which can act as a powerful agent to restrain the development of bacterial biofilm on therapeutic implants.
Collapse
Affiliation(s)
- Sajid Ali
- Department of Chemistry, Bacha Khan University, Charsadda 24420, Khyber Pakhtunkhwa, Pakistan
- Correspondence: (S.A.); (R.U.)
| | - Nazma Faqir
- Department of Chemistry, Bacha Khan University, Charsadda 24420, Khyber Pakhtunkhwa, Pakistan
| | - Falak Naz
- Department of Chemistry, Bacha Khan University, Charsadda 24420, Khyber Pakhtunkhwa, Pakistan
| | - Muhammad Ishtiaq Jan
- Department of Chemistry, Kohat University of Science and Technology, Kohat 26000, Khyber Pakhtunkhwa, Pakistan
| | - Naeem Khan
- Department of Chemistry, Kohat University of Science and Technology, Kohat 26000, Khyber Pakhtunkhwa, Pakistan
| | - Amal Alotaibi
- Department of Basic Science, College of Medicine, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Riaz Ullah
- Medicinal Aromatic and Poisonous Plants Research Center, Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
- Correspondence: (S.A.); (R.U.)
| |
Collapse
|
71
|
Foodborne Pathogen Biofilms: Development, Detection, Control, and Antimicrobial Resistance. Pathogens 2023; 12:pathogens12020352. [PMID: 36839624 PMCID: PMC9961813 DOI: 10.3390/pathogens12020352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
Bacteria can grow either as planktonic cells or as communities within biofilms [...].
Collapse
|
72
|
Ortiz-Magdaleno M, Sánchez-Vargas L, Gardea-Contreras D, Campos-Ibarra V, Pozos-Guillén A, Márquez-Preciado R. Antibiofilm properties of silver nanoparticles incorporated into polymethyl methacrylate used for dental applications. Biomed Mater Eng 2023:BME222513. [PMID: 36744329 DOI: 10.3233/bme-222513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Acrylic resins used in dental and biomedical applications do not have antimicrobial properties, their surface is susceptible to colonization of microorganisms. OBJECTIVE The aim of this study was to evaluate the antibiofilm properties of silver nanoparticles (AgNPs) deposited in a polymethyl methacrylate (PMMA) surface against a Staphylococcus aureus biofilm. METHODS The PMMA was impregnated with AgNPs by using the in-situ polymerization method. To determine the solubility of the incorporated silver (Ag+) atomic absorption spectrophotometry was used (AAS) at 24 h, 48 h, 7 days, and 30 days. Thirty specimens of PMMA with AgNPs and without NP (control group) were assembled in the CDC Biofilm Bioreactor system with a cell suspension of S. aureus. The specimens were removed at 6, 12, 24, 48, and 72 h to determine the viability profile and quantify the Arbitrary Fluorescence Units (AFU). RESULTS The AgNPs showed an irregular and quasispherical shape with an average size of 25 nm. AAS analysis demonstrated a low solubility of Ag+. The formation of the S. aureus biofilm increased as the evaluation periods continued up to 72 h. The experimental group showed poor growth, and a decrease in the intensity of the fluorescence demonstrated a statistically significant inhibition of the formation of the biofilm (P < 0.05) in relation to the control group at 6, 12, 24, 48, and 72 h. CONCLUSION AgNPs incorporated into PMMA decreased the growth and maturation of S. aureus biofilm.
Collapse
Affiliation(s)
- Marine Ortiz-Magdaleno
- Basic Science Laboratory, Faculty of Stomatology, San Luis Potosí University, San Luis Potosi, Mexico
| | - Luis Sánchez-Vargas
- Biochemical and Microbiology Laboratory, Faculty of Stomatology, San Luis Potosí University, San Luis Potosi, Mexico
| | - Delia Gardea-Contreras
- Paediatric Dentistry Postgraduate Program, Faculty of Stomatology, San Luis Potosí University, San Luis Potosi, Mexico
| | | | - Amaury Pozos-Guillén
- Basic Science Laboratory, Faculty of Stomatology, San Luis Potosí University, San Luis Potosi, Mexico
| | - Raúl Márquez-Preciado
- Paediatric Dentistry Postgraduate Program, Faculty of Stomatology, San Luis Potosí University, San Luis Potosi, Mexico
| |
Collapse
|
73
|
Curylofo-Zotti FA, Oliveira VDC, Marchesin AR, Borges HS, Tedesco AC, Corona SAM. In vitro antibacterial activity of green tea-loaded chitosan nanoparticles on caries-related microorganisms and dentin after Er:YAG laser caries removal. Lasers Med Sci 2023; 38:50. [PMID: 36689037 DOI: 10.1007/s10103-023-03707-3] [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: 05/30/2022] [Accepted: 01/11/2023] [Indexed: 01/24/2023]
Abstract
This study aimed to determine the inhibitory effects of green tea (Gt), EGCG, and nanoformulations containing chitosan (Nchi) and chitosan+green tea (Nchi+Gt) against Streptococcus mutans and Lactobacillus casei. In addition, the antibacterial effect of nanoformulations was evaluated directly on dentin after the selective removal of carious lesion. At first, the minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) against S. mutans and L. casei isolates were investigated. In parallel, dentin specimens were exposed to S. mutans to induce carious lesions. Soft dentin was selectively removed by Er:YAG laser (n=33) or bur (n=33). Remaining dentin was biomodified with Nchi (n=11) or Gt+Nchi (n=11). Control group (n=11) did not receive any treatment. Dentin scraps were collected at three time points. Microbiological analyses were conducted and evaluated by agar plate counts. Gt at 1:32 dilution inhibited S. mutans growth while 1:16 was efficient against L. casei. EGCG at 1:4 dilution completely inhibited S. mutans and L. casei growth. Independently of the association with Gt, Nchi completely inhibited S. mutans at 1:4 dilution. For L. casei, different concentrations of Nchi (1:32) and Nchi+Gt (1:8) were required to inhibit cell growth. After selective carious removal, viability of S. mutans decreased (p<0.001), without difference between bur and Er:YAG laser (p>0.05). Treatment with Nchi and Nchi+Gt did not influence the microbial load of S. mutans on dentin (p>0.05). Although variations in concentrations were noticed, all compounds showed antibacterial activity against S. mutans and L. casei. Both bur and Er:YAG laser have effectively removed soft dentin and reduced S. mutans counts. Nanoformulations did not promote any additional antibacterial effect in the remaining dentin.
Collapse
Affiliation(s)
- Fabiana Almeida Curylofo-Zotti
- Department of Restorative Dentistry, School of Dentistry of Ribeirao Preto, University of Sao Paulo (USP), Cafe Avenue, s/n 14040-904, Sao Paulo, Ribeirao Preto, Brazil.
| | - Viviane De Cássia Oliveira
- Department of Dental Materials and Prosthodontics, School of Dentistry of Ribeirao Preto, University of Sao Paulo (USP), Ribeirao Preto, Sao Paulo, SP, Brazil
| | - Analu Rodriguez Marchesin
- Department of Restorative Dentistry, School of Dentistry of Ribeirao Preto, University of Sao Paulo (USP), Cafe Avenue, s/n 14040-904, Sao Paulo, Ribeirao Preto, Brazil
| | - Hiago Salge Borges
- Department of Chemistry, Center of Nanotechnology and Tissue Engineering - Photobiology and Photomedicine Research Group, Faculty of Philosophy, Sciences and Letters of Ribeirao Preto, University of Sao Paulo (USP), São Paulo, Brazil
| | - Antonio Claudio Tedesco
- Department of Chemistry, Center of Nanotechnology and Tissue Engineering - Photobiology and Photomedicine Research Group, Faculty of Philosophy, Sciences and Letters of Ribeirao Preto, University of Sao Paulo (USP), São Paulo, Brazil
| | - Silmara Aparecida Milori Corona
- Department of Restorative Dentistry, School of Dentistry of Ribeirao Preto, University of Sao Paulo (USP), Cafe Avenue, s/n 14040-904, Sao Paulo, Ribeirao Preto, Brazil
| |
Collapse
|
74
|
Yang H, Xu Z, Xu Z, Li Y. Mini-Review of Biofilm Interactions with Surface Materials in Industrial Piping System. MEMBRANES 2023; 13:125. [PMID: 36837628 PMCID: PMC9961356 DOI: 10.3390/membranes13020125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/16/2023] [Accepted: 01/17/2023] [Indexed: 06/18/2023]
Abstract
The growth of biofilm, which is caused by microorganism accumulation and growth on wetted surfaces, may damage industrial piping systems, increase maintenance and cleaning costs for the system sterilization, and even divulge the immune system into high risk. This article systematically analyzes the biofilm interactions with piping surface materials from the perspectives of physical convection, and biological and chemical adhesion. The thermodynamics of the flow, bacterial surface sensing, and bio-communication are the most critical factors for biofilm attachment. Furthermore, experimental analysis methods as well as biofilm control and removal approaches, are also included in this study. Finally, the resistance and growth of biofilm, as well as the practical and advanced methodology to control the biofilm and challenges associated with technology, are also discussed. Moreover, this paper may also offer a significant reference for the practice and strategic applications to address the biofilm resistance issues in industrial piping.
Collapse
Affiliation(s)
- Haoyi Yang
- NUS College of Design and Engineering, National University of Singapore, Singapore 118429, Singapore
| | - Zezheng Xu
- UNSW Environment Leadership Program, The University of New South Wales, Kensington, NSW 2052, Australia
| | - Zetong Xu
- Qingdao Huanghai Vocational Institute, Qingdao 266555, China
| | - Yuanzhe Li
- School of Materials Science & Engineering, Nanyang Technological University, Singapore 639798, Singapore
| |
Collapse
|
75
|
Martin LW, Gray AR, Brockway B, Lamont IL. Pseudomonas aeruginosa is oxygen-deprived during infection in cystic fibrosis lungs, reducing the effectiveness of antibiotics. FEMS Microbiol Lett 2023; 370:fnad076. [PMID: 37516450 PMCID: PMC10408701 DOI: 10.1093/femsle/fnad076] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 07/04/2023] [Accepted: 07/20/2023] [Indexed: 07/31/2023] Open
Abstract
Pseudomonas aeruginosa infects the lungs of patients with cystic fibrosis. Sputum expectorated from the lungs of patients contains low levels of oxygen, indicating that P. aeruginosa may be oxygen-deprived during infection. During in vitro growth under oxygen-limiting conditions, a P. aeruginosa reference strain increases expression of a cytochrome oxidase with a high affinity for oxygen, and of nitrate and nitrite reductases that enable it to use nitrate instead of oxygen during respiration. Here, we quantified transcription of the genes encoding these three enzymes in sputum samples from 18 infected patients, and in bacteria isolated from the sputum samples and grown in aerobic and anaerobic culture. In culture, expression of all three genes was increased by averages of 20- to 500-fold in anaerobically grown bacteria compared with those grown aerobically, although expression levels varied greatly between isolates. Expression of the same genes in sputum was similar to that of the corresponding bacteria in anaerobic culture. The isolated bacteria were less susceptible to tobramycin and ciprofloxacin, two widely used anti-pseudomonal antibiotics, when grown anaerobically than when grown aerobically. Our findings show that P. aeruginosa experiences oxygen starvation during infection in cystic fibrosis, reducing the effectiveness of antibiotic treatment.
Collapse
Affiliation(s)
- Lois W Martin
- Department of Biochemistry, University of Otago, Dunedin, 9016, New Zealand
| | - Andrew R Gray
- Biostatistics Centre, University of Otago, Dunedin 9016, New Zealand
| | - Ben Brockway
- Medicine, University of Otago, Dunedin 9016, New Zealand
| | - Iain L Lamont
- Department of Biochemistry, University of Otago, Dunedin, 9016, New Zealand
| |
Collapse
|
76
|
van Dun SCJ, Verheul M, Pijls BGCW, van Prehn J, Scheper H, Galli F, Nibbering PH, de Boer MGJ. Influence of surface characteristics of implant materials on MRSA biofilm formation and effects of antimicrobial treatment. Front Microbiol 2023; 14:1145210. [PMID: 37152752 PMCID: PMC10159048 DOI: 10.3389/fmicb.2023.1145210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 04/03/2023] [Indexed: 05/09/2023] Open
Abstract
Introduction One of the main causes of treatment failure in bacterial prosthetic joint infections (PJI) is biofilm formation. The topography of the biofilm may be associated with susceptibility to antimicrobial treatment. The aims of this study were to assess differences in topography of biofilms on different implant materials and the correlation thereof with susceptibility to antimicrobial treatment. Methods Methicillin-resistant Staphylococcus aureus (MRSA) 7-day mature biofilms were generated on disks made from titanium alloys (Ti-6Al-7Nb and Ti-6Al-4V), synthetic polymer and orthopedic bone cement, commonly used in implant surgery. The surface topography of these implant materials and the biofilms cultured on them was assessed using atomic force microscopy. This provided detailed images, as well as average roughness (Ra) and peak-to-valley roughness (Rt) values in nanometers, of the biofilm and the material surfaces. Bacterial counts within biofilms were assessed microbiologically. Antimicrobial treatment of biofilms was performed by 24-h exposure to the combination of rifampicin and ciprofloxacin in concentrations of 1-, 5- and 10-times the minimal bactericidal concentration (MBC). Finally, treatment-induced differences in bacterial loads and their correlation with biofilm surface parameters were assessed. Results The biofilm surfaces on titanium alloys Ti-6Al-7Nb (Ra = 186 nm) and Ti-6Al-4V (Ra = 270 nm) were less rough than those of biofilms on silicone (Ra = 636 nm). The highest roughness was observed for biofilms on orthopedic bone cement with an Ra of 1,551 nm. Interestingly, the roughness parameters of the titanium alloys themselves were lower than the value for silicone, whereas the surface of the bone cement was the roughest. Treatment with 1- and 5-times the MBC of antibiotics resulted in inter-material differences in colony forming units (CFU) counts, ultimately showing comparable reductions of 2.4-3.0 log CFU/mL at the highest tested concentration. No significant differences in bacterial loads within MRSA biofilms were observed between the various implant materials, upon exposure to increasing concentrations of antibiotics. Discussion The surface parameters of MRSA biofilms were determined by those of the implant materials on which they were formed. The antibiotic susceptibility of MRSA biofilms on the various tested implant materials did not differ, indicating that the efficacy of antibiotics was not affected by the roughness of the biofilm.
Collapse
Affiliation(s)
- Sven C. J. van Dun
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
- *Correspondence: Sven C. J. van Dun,
| | - Mariëlle Verheul
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Bart G. C. W. Pijls
- Department of Orthopedics, Leiden University Medical Center, Leiden, Netherlands
| | - Joffrey van Prehn
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, Netherlands
| | - Henk Scheper
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | | | - Peter H. Nibbering
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Mark G. J. de Boer
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
- Mark G. J. de Boer,
| |
Collapse
|
77
|
Coppola D, Buonocore C, Palisse M, Tedesco P, de Pascale D. Exploring Oceans for Curative Compounds: Potential New Antimicrobial and Anti-Virulence Molecules against Pseudomonas aeruginosa. Mar Drugs 2022; 21:9. [PMID: 36662182 PMCID: PMC9865402 DOI: 10.3390/md21010009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/16/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
Although several antibiotics are already widely used against a large number of pathogens, the discovery of new antimicrobial compounds with new mechanisms of action is critical today in order to overcome the spreading of antimicrobial resistance among pathogen bacteria. In this regard, marine organisms represent a potential source of a wide diversity of unique secondary metabolites produced as an adaptation strategy to survive in competitive and hostile environments. Among the multidrug-resistant Gram-negative bacteria, Pseudomonas aeruginosa is undoubtedly one of the most important species due to its high intrinsic resistance to different classes of antibiotics on the market and its ability to cause serious therapeutic problems. In the present review, we first discuss the general mechanisms involved in the antibiotic resistance of P. aeruginosa. Subsequently, we list the marine molecules identified up until now showing activity against P. aeruginosa, dividing them according to whether they act as antimicrobial or anti-virulence compounds.
Collapse
Affiliation(s)
- Daniela Coppola
- Department of Ecosustainable Marine Biotechnology, Stazione Zoologica Anton Dohrn, Via Ammiraglio Ferdinando Acton 55, 80133 Naples, Italy
| | - Carmine Buonocore
- Department of Ecosustainable Marine Biotechnology, Stazione Zoologica Anton Dohrn, Via Ammiraglio Ferdinando Acton 55, 80133 Naples, Italy
| | - Morgan Palisse
- Département des Sciences de la Vie et de la Terre, Université de Caen Normandie, Boulevard Maréchal Juin CS, CEDEX, 14032 Caen, France
| | - Pietro Tedesco
- Department of Ecosustainable Marine Biotechnology, Stazione Zoologica Anton Dohrn, Via Ammiraglio Ferdinando Acton 55, 80133 Naples, Italy
| | - Donatella de Pascale
- Department of Ecosustainable Marine Biotechnology, Stazione Zoologica Anton Dohrn, Via Ammiraglio Ferdinando Acton 55, 80133 Naples, Italy
| |
Collapse
|
78
|
Neuhaus S, Feßler AT, Dieckmann R, Thieme L, Pletz MW, Schwarz S, Al Dahouk S. Towards a Harmonized Terminology: A Glossary for Biocide Susceptibility Testing. Pathogens 2022; 11:1455. [PMID: 36558789 PMCID: PMC9780826 DOI: 10.3390/pathogens11121455] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/24/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
Abstract
Disinfection is a key strategy to reduce the burden of infections. The contact of bacteria to biocides-the active substances of disinfectants-has been linked to bacterial adaptation and the development of antimicrobial resistance. Currently, there is no scientific consensus on whether the excessive use of biocides contributes to the emergence and spread of multidrug resistant bacteria. The comprehensive analysis of available data remains a challenge because neither uniform test procedures nor standardized interpretive criteria nor harmonized terms are available to describe altered bacterial susceptibility to biocides. In our review, we investigated the variety of criteria and the diversity of terms applied to interpret findings in original studies performing biocide susceptibility testing (BST) of field isolates. An additional analysis of reviews summarizing the knowledge of individual studies on altered biocide susceptibility provided insights into currently available broader concepts for data interpretation. Both approaches pointed out the urgent need for standardization. We, therefore, propose that the well-established and approved concepts for interpretation of antimicrobial susceptibility testing data should serve as a role model to evaluate biocide resistance mechanisms on a single cell level. Furthermore, we emphasize the adaptations necessary to acknowledge the specific needs for the evaluation of BST data. Our approach might help to increase scientific awareness and acceptance.
Collapse
Affiliation(s)
- Szilvia Neuhaus
- German Federal Institute for Risk Assessment, 10589 Berlin, Germany
| | - Andrea T. Feßler
- Centre for Infection Medicine, Department of Veterinary Medicine, Institute of Microbiology and Epizootics, Freie Universität Berlin, 14163 Berlin, Germany
- Veterinary Centre for Resistance Research (TZR), Freie Universität Berlin, 14163 Berlin, Germany
| | - Ralf Dieckmann
- German Federal Institute for Risk Assessment, 10589 Berlin, Germany
| | - Lara Thieme
- Institute of Infectious Diseases and Infection Control, Jena University Hospital, Friedrich-Schiller-University Jena, 07747 Jena, Germany
- Leibniz Center for Photonics in Infection Research, Jena University Hospital, Friedrich Schiller University Jena, 07747 Jena, Germany
| | - Mathias W. Pletz
- Institute of Infectious Diseases and Infection Control, Jena University Hospital, Friedrich-Schiller-University Jena, 07747 Jena, Germany
| | - Stefan Schwarz
- Centre for Infection Medicine, Department of Veterinary Medicine, Institute of Microbiology and Epizootics, Freie Universität Berlin, 14163 Berlin, Germany
- Veterinary Centre for Resistance Research (TZR), Freie Universität Berlin, 14163 Berlin, Germany
| | - Sascha Al Dahouk
- German Federal Institute for Risk Assessment, 10589 Berlin, Germany
- Department of Internal Medicine, RWTH Aachen University Hospital, 52074 Aachen, Germany
| |
Collapse
|
79
|
Cesaria M, Alfinito E, Arima V, Bianco M, Cataldo R. MEED: A novel robust contrast enhancement procedure yielding highly-convergent thresholding of biofilm images. Comput Biol Med 2022; 151:106217. [PMID: 36306585 DOI: 10.1016/j.compbiomed.2022.106217] [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: 07/31/2022] [Revised: 10/04/2022] [Accepted: 10/15/2022] [Indexed: 12/27/2022]
Abstract
Morphological and statistical investigation of biofilm images may be even more critical than the image acquisition itself, in particular in the presence of morphologically complex distributions, due to the unavoidable impact of the measurement technique too. Hence, digital image pre-processing is mandatory for reliable feature extraction and enhancement preliminary to segmentation. Also, pattern recognition in automated deep learning (both supervised and unsupervised) models often requires a preliminary effective contrast-enhancement. However, no universal consensus exists on the optimal contrast enhancement approach. This paper presents and discusses a new general, robust, reproducible, accurate and easy to implement contrast enhancement procedure, briefly named MEED-procedure, able to work on images with different bacterial coverages and biofilm structures, coming from different imaging instrumentations (herein stereomicroscope and transmission microscope). It exploits a proper succession of basic morphological operations (erosion and dilation) and a horizontal line structuring element, to minimize the impact on size and shape of the even finer bacterial features. It systematically enhances the objects of interest, without histogram stretching and/or undesirable artifacts yielded by common automated methods. The quality of the MEED-procedure is ascertained by segmentation tests which demonstrate its robustness regarding the determination of threshold and convergence of the thresholding algorithm. Extensive validation tests over a rich image database, comparison with the literature and comprehensive discussion of the conceptual background support the superiority of the MEED-procedure over the existing methods and demonstrate it is not a routine application of morphological operators.
Collapse
Affiliation(s)
- Maura Cesaria
- University of Salento-Department of Mathematics and Physics "Ennio De Giorgi"- c/o Campus Ecotekne - Lecce, Italy.
| | - Eleonora Alfinito
- University of Salento-Department of Mathematics and Physics "Ennio De Giorgi"- c/o Campus Ecotekne - Lecce, Italy
| | - Valentina Arima
- CNR NANOTEC - Institute of Nanotechnology, c/o Campus Ecotekne, Lecce, Italy
| | - Monica Bianco
- CNR NANOTEC - Institute of Nanotechnology, c/o Campus Ecotekne, Lecce, Italy
| | - Rosella Cataldo
- University of Salento-Department of Mathematics and Physics "Ennio De Giorgi"- c/o Campus Ecotekne - Lecce, Italy.
| |
Collapse
|
80
|
Reiferth VM, Holtmann D, Müller D. Flexible biofilm monitoring device. Eng Life Sci 2022; 22:796-802. [PMID: 36514529 PMCID: PMC9731593 DOI: 10.1002/elsc.202100076] [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: 06/29/2021] [Revised: 05/18/2022] [Accepted: 06/29/2022] [Indexed: 12/16/2022] Open
Abstract
Biofilms and their analysis are increasingly attracting the attention of the scientific community due to the immense importance and impact of biofilms in various natural, technical and medical fields. For these purposes, an optimized and extended antibiofilm assay system based on the Calgary Biofilm Device (MBEC Assay® system) consisting of microtiter plate and PCR tubes was established. Its implementation was used to study the growth characteristics of the sessile phenotype of Pseudomonas fluorescens exposed to antimicrobial peptides. Inhibitory effects of an antimicrobial peptide on P. fluorescens biofilm formation could be determined at a concentration of 250 μg/ml (biofilm prevention concentration (BPC)) using the modified biofilm assay. Similarly, the biofilm bactericidal concentration (BBC) at 125 μg/ml and the minimum biofilm elimination concentration to remove 90% of the total biofilm mass (MBEC90) were measured at a concentration range of 15.625-1.95 μg/ml. In conclusion, this optimized system provides a highly variable, simple, and cost-effective alternative to high-throughput screening based on the Calgary Biofilm Device (CBD).
Collapse
Affiliation(s)
| | - Dirk Holtmann
- Technische Hochschule MittelhessenUniversity of Applied SciencesGiessenGermany
| | - Daniela Müller
- Technische Hochschule MittelhessenUniversity of Applied SciencesGiessenGermany
| |
Collapse
|
81
|
Materazzi A, Bottai D, Campobasso C, Klatt AB, Cesta N, De Masi M, Trampuz A, Tavanti A, Di Luca M. Phage-Based Control of Methicillin Resistant Staphylococcus aureus in a Galleria mellonella Model of Implant-Associated Infection. Int J Mol Sci 2022; 23:ijms232314514. [PMID: 36498843 PMCID: PMC9740198 DOI: 10.3390/ijms232314514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/16/2022] [Accepted: 11/19/2022] [Indexed: 11/23/2022] Open
Abstract
Staphylococcus aureus implant-associated infections are difficult to treat because of the ability of bacteria to form biofilm on medical devices. Here, the efficacy of Sb-1 to control or prevent S. aureus colonization on medical foreign bodies was investigated in a Galleria mellonella larval infection model. For colonization control assays, sterile K-wires were implanted into larva prolegs. After 2 days, larvae were infected with methicillin-resistant S. aureus ATCC 43300 and incubated at 37 °C for a further 2 days, when treatments with either daptomycin (4 mg/kg), Sb-1 (107 PFUs) or a combination of them (3 x/day) were started. For biofilm prevention assays, larvae were pre-treated with either vancomycin (10 mg/kg) or Sb-1 (107 PFUs) before the S. aureus infection. In both experimental settings, K-wires were explanted for colony counting two days after treatment. In comparison to the untreated control, more than a 4 log10 CFU and 1 log10 CFU reduction was observed on K-wires recovered from larvae treated with the Sb-1/daptomycin combination and with their singular administration, respectively. Moreover, pre-infection treatment with Sb-1 was found to prevent K-wire colonization, similarly to vancomycin. Taken together, the obtained results demonstrated the strong potential of the Sb-1 antibiotic combinatory administration or the Sb-1 pretreatment to control or prevent S. aureus-associated implant infections.
Collapse
Affiliation(s)
| | - Daria Bottai
- Department of Biology, University of Pisa, 56127 Pisa, Italy
| | - Claudia Campobasso
- Department of Biology, University of Pisa, 56127 Pisa, Italy
- Department of Biosystems, KU Leuven, 3001 Leuven, Belgium
| | - Ann-Brit Klatt
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Novella Cesta
- PhD Course in Microbiology, Immunology, Infectious Diseases and Transplants (MIMIT), University of Rome Tor Vergata, 00133 Rome, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Margherita De Masi
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Andrej Trampuz
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
- Center for Musculoskeletal Surgery, Charité-Universitätsmedizin Berlin, Berlin Institute of Health, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, 13353 Berlin, Germany
| | - Arianna Tavanti
- Department of Biology, University of Pisa, 56127 Pisa, Italy
| | | |
Collapse
|
82
|
Rega M, Andriani L, Cavallo S, Bonilauri P, Bonardi S, Conter M, Carmosino I, Bacci C. Antimicrobial Resistant E. coli in Pork and Wild Boar Meat: A Risk to Consumers. Foods 2022; 11:foods11223662. [PMID: 36429254 PMCID: PMC9689484 DOI: 10.3390/foods11223662] [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: 10/12/2022] [Revised: 11/04/2022] [Accepted: 11/12/2022] [Indexed: 11/18/2022] Open
Abstract
Antimicrobial-resistant foodborne microorganisms may be transmitted from food producing animals to humans through the consumption of meat products. In this study, meat that was derived from farmed pigs and wild boars was analyzed and compared. Escherichia coli (E. coli) were isolated and tested phenotypically and genotypically for their resistance to quinolones, aminoglycosides and carbapenems. The co-presence of AMR-associated plasmid genes was also evaluated. A quinolone AMR phenotypic analysis showed 41.9% and 36.1% of resistant E. coli derived from pork and wild boars meat, respectively. A resistance to aminoglycosides was detected in the 6.6% of E. coli that was isolated from pork and in 1.8% of the wild boar meat isolates. No resistant profiles were detected for the carbapenems. The quinolone resistance genes were found in 58.3% of the phenotypically resistant pork E. coli and in 17.5% of the wild boar, thus showing low genotypic confirmation rates. The co-presence of the plasmid-related genes was observed only for the quinolones and aminoglycosides, but not for the carbapenems. Wild boar E. coli were the most capable to perform biofilm production when they were compared to pork E. coli. In conclusion, the contamination of pork and wild boar meat by AMR microorganisms could be a threat for consumers, especially if biofilm-producing strains colonize the surfaces and equipment that are used in the food industry.
Collapse
Affiliation(s)
- Martina Rega
- Food Hygiene and Inspection, Veterinary Science Department, University of Parma, Strada del Taglio, 10, 43126 Parma, Italy
| | - Laura Andriani
- Food Hygiene and Inspection, Veterinary Science Department, University of Parma, Strada del Taglio, 10, 43126 Parma, Italy
| | - Silvia Cavallo
- Food Hygiene and Inspection, Veterinary Science Department, University of Parma, Strada del Taglio, 10, 43126 Parma, Italy
| | - Paolo Bonilauri
- Istituto Zooprofilattico Sperimentale della Lombardia ed Emilia Romagna, via Pitagora, 2, 42124 Reggio Emilia, Italy
| | - Silvia Bonardi
- Food Hygiene and Inspection, Veterinary Science Department, University of Parma, Strada del Taglio, 10, 43126 Parma, Italy
| | - Mauro Conter
- Food Hygiene and Inspection, Veterinary Science Department, University of Parma, Strada del Taglio, 10, 43126 Parma, Italy
- Correspondence: ; Tel.: +39-0521-902683
| | - Ilaria Carmosino
- Istituto Zooprofilattico Sperimentale della Lombardia ed Emilia Romagna, via Emilio Diena, 16, 41122 Modena, Italy
| | - Cristina Bacci
- Food Hygiene and Inspection, Veterinary Science Department, University of Parma, Strada del Taglio, 10, 43126 Parma, Italy
| |
Collapse
|
83
|
Metabolomic Profiling of the Responses of Planktonic and Biofilm Vibrio cholerae to Silver Nanoparticles. Antibiotics (Basel) 2022; 11:antibiotics11111534. [DOI: 10.3390/antibiotics11111534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 10/27/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
Abstract
Vibrio cholerae causes cholera and can switch between planktonic and biofilm lifeforms, where biofilm formation enhances transmission, virulence, and antibiotic resistance. Due to antibiotic microbial resistance, new antimicrobials including silver nanoparticles (AgNPs) are being studied. Nevertheless, little is known about the metabolic changes exerted by AgNPs on both microbial lifeforms. Our objective was to evaluate the changes in the metabolomic profile of V. cholerae planktonic and biofilm cells in response to sublethal concentrations of AgNPs using MS2 untargeted metabolomics and chemoinformatics. A total of 690 metabolites were quantified among all groups. More metabolites were significantly modulated in planktonic cells (n = 71) compared to biofilm (n = 37) by the treatment. The chemical class profiles were distinct for both planktonic and biofilm, suggesting a phenotype-dependent metabolic response to the nanoparticles. Chemical enrichment analysis showed altered abundances of oxidized fatty acids (FA), saturated FA, phosphatidic acids, and saturated stearic acid in planktonic cells treated with AgNPs, which hints at a turnover of the membrane. In contrast, no chemical classes were enriched in the biofilm. In conclusion, this study suggests that the response of V. cholerae to silver nanoparticles is phenotype-dependent and that planktonic cells experience a lipid remodeling process, possibly related to an adaptive mechanism involving the cell membrane.
Collapse
|
84
|
Cheng Y, De Bank PA, Bolhuis A. An in vitro and ex vivo wound infection model to test topical and systemic treatment with antibiotics. J Appl Microbiol 2022; 133:2993-3006. [PMID: 35916629 PMCID: PMC9804477 DOI: 10.1111/jam.15756] [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: 04/29/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 01/05/2023]
Abstract
AIMS This study aimed to develop a wound infection model that could be used to test antibiotic-loaded electrospun matrices for the topical treatment of infected skin and compare the effectiveness of this treatment to systemically applied antibiotics. METHODS AND RESULTS 3D-printed flow chambers were made in which Staphylococcus aureus biofilms were grown either on a polycarbonate membrane or explanted porcine skin. The biofilms were then treated either topically, by placing antibiotic-loaded electrospun matrices on top of the biofilms, or systemically by the addition of antibiotics in the growth medium that flowed underneath the membrane or skin. The medium that was used was either a rich medium or an artificial wound fluid. The results showed that microbial viability in the biofilms was reduced to a greater extent with the topical electrospun matrices when compared to systemic treatment. CONCLUSIONS An ex vivo infection model was developed that is flexible and can be used to test both topical and systemic treatment of wound infections. It represents a significant improvement over previous in vitro models that we have used to test electrospun membranes. SIGNIFICANCE AND IMPACT OF THE STUDY The availability of a relatively simple wound infection model in which different delivery methods and dosage regimes can be tested is beneficial for the development of improved treatments for wound infections.
Collapse
Affiliation(s)
- Yanyan Cheng
- Department of Pharmacy and Pharmacology and the Centre for Therapeutic InnovationUniversity of BathBathUK
| | - Paul A. De Bank
- Department of Pharmacy and Pharmacology and the Centre for Therapeutic InnovationUniversity of BathBathUK
| | - Albert Bolhuis
- Department of Pharmacy and Pharmacology and the Centre for Therapeutic InnovationUniversity of BathBathUK
| |
Collapse
|
85
|
Salvador A, Veiga FF, Svidzinski TIE, Negri M. In vitro ability of Fusarium keratoplasticum to form biofilms in venous catheter. Microb Pathog 2022; 173:105868. [DOI: 10.1016/j.micpath.2022.105868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 06/10/2022] [Accepted: 11/02/2022] [Indexed: 11/08/2022]
|
86
|
Wang W, Bao X, Bové M, Rigole P, Meng X, Su J, Coenye T. Antibiofilm Activities of Borneol-Citral-Loaded Pickering Emulsions against Pseudomonas aeruginosa and Staphylococcus aureus in Physiologically Relevant Chronic Infection Models. Microbiol Spectr 2022; 10:e0169622. [PMID: 36194139 PMCID: PMC9602683 DOI: 10.1128/spectrum.01696-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 09/08/2022] [Indexed: 12/31/2022] Open
Abstract
Phytochemicals are promising antibacterials for the development of novel antibiofilm drugs, but their antibiofilm activity in physiologically relevant model systems is poorly characterized. As the host microenvironment can interfere with the activity of the phytochemicals, mimicking the complex environment found in biofilm associated infections is essential to predict the clinical potential of novel phytochemical-based antimicrobials. In the present study, we examined the antibiofilm activity of borneol, citral, and combinations of both as well as their Pickering emulsions against Staphylococcus aureus and Pseudomonas aeruginosa in an in vivo-like synthetic cystic fibrosis medium (SCFM2) model, an in vitro wound model (consisting of an artificial dermis and blood components at physiological levels), and an in vivo Galleria mellonella model. The Pickering emulsions demonstrated an enhanced biofilm inhibitory activity compared to both citral and the borneol/citral combination, reducing the minimum biofilm inhibitory concentration (MBIC) values up to 2 to 4 times against P. aeruginosa PAO1 and 2 to 8 times against S. aureus P8-AE1 in SCMF2. In addition, citral, the combination borneol/citral, and their Pickering emulsions can completely eliminate the established biofilm of S. aureus P8-AE1. The effectiveness of Pickering emulsions was also demonstrated in the wound model with a reduction of up to 4.8 log units in biofilm formation by S. aureus Mu50. Furthermore, citral and Pickering emulsions exhibited a significant degree of protection against S. aureus infection in the G. mellonella model. The present findings reveal the potential of citral- or borneol/citral-based Pickering emulsions as a type of alternative antibiofilm candidate to control pathogenicity in chronic infection. IMPORTANCE There is clearly an urgent need for novel formulations with antimicrobial and antibiofilm activity, but while there are plenty of studies investigating them using simple in vitro systems, there is a lack of studies in which (combinations of) phytochemicals are evaluated in relevant models that closely resemble the in vivo situation. Here, we examined the antibiofilm activity of borneol, citral, and their combination as well as Pickering emulsions (stabilized by solid particles) of these compounds. Activity was tested against Staphylococcus aureus and Pseudomonas aeruginosa in in vitro models mimicking cystic fibrosis sputum and wounds as well as in an in vivo Galleria mellonella model. The Pickering emulsions showed drastically increased antibiofilm activity compared to that of the compounds as such in both in vitro models and protected G. mellonella larvae from S. aureus-induced killing. Our data show that Pickering emulsions from phytochemicals are potentially useful for treating specific biofilm-related chronic infections.
Collapse
Affiliation(s)
- Wen Wang
- School of Food Science and Engineering, South China University of Technology, Guangzhou, Guangdong, China
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
- China-Singapore International Joint Research Institute, Guangzhou, China
| | - Xuerui Bao
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| | - Mona Bové
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| | - Petra Rigole
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| | - Xiaofeng Meng
- School of Food Science and Engineering, South China University of Technology, Guangzhou, Guangdong, China
- China-Singapore International Joint Research Institute, Guangzhou, China
| | - Jianyu Su
- School of Food Science and Engineering, South China University of Technology, Guangzhou, Guangdong, China
- China-Singapore International Joint Research Institute, Guangzhou, China
| | - Tom Coenye
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| |
Collapse
|
87
|
Doolan JA, Williams GT, Hilton KLF, Chaudhari R, Fossey JS, Goult BT, Hiscock JR. Advancements in antimicrobial nanoscale materials and self-assembling systems. Chem Soc Rev 2022; 51:8696-8755. [PMID: 36190355 PMCID: PMC9575517 DOI: 10.1039/d1cs00915j] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Indexed: 11/21/2022]
Abstract
Antimicrobial resistance is directly responsible for more deaths per year than either HIV/AIDS or malaria and is predicted to incur a cumulative societal financial burden of at least $100 trillion between 2014 and 2050. Already heralded as one of the greatest threats to human health, the onset of the coronavirus pandemic has accelerated the prevalence of antimicrobial resistant bacterial infections due to factors including increased global antibiotic/antimicrobial use. Thus an urgent need for novel therapeutics to combat what some have termed the 'silent pandemic' is evident. This review acts as a repository of research and an overview of the novel therapeutic strategies being developed to overcome antimicrobial resistance, with a focus on self-assembling systems and nanoscale materials. The fundamental mechanisms of action, as well as the key advantages and disadvantages of each system are discussed, and attention is drawn to key examples within each field. As a result, this review provides a guide to the further design and development of antimicrobial systems, and outlines the interdisciplinary techniques required to translate this fundamental research towards the clinic.
Collapse
Affiliation(s)
- Jack A Doolan
- School of Chemistry and Forensic Science, University of Kent, Canterbury, Kent CT2 7NH, UK.
- School of Biosciences, University of Kent, Canterbury, Kent CT2 7NJ, UK.
| | - George T Williams
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| | - Kira L F Hilton
- School of Chemistry and Forensic Science, University of Kent, Canterbury, Kent CT2 7NH, UK.
| | - Rajas Chaudhari
- School of Chemistry and Forensic Science, University of Kent, Canterbury, Kent CT2 7NH, UK.
| | - John S Fossey
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| | - Benjamin T Goult
- School of Biosciences, University of Kent, Canterbury, Kent CT2 7NJ, UK.
| | - Jennifer R Hiscock
- School of Chemistry and Forensic Science, University of Kent, Canterbury, Kent CT2 7NH, UK.
| |
Collapse
|
88
|
Drevinek P, Canton R, Johansen HK, Hoffman L, Coenye T, Burgel PR, Davies JC. New concepts in antimicrobial resistance in cystic fibrosis respiratory infections. J Cyst Fibros 2022; 21:937-945. [DOI: 10.1016/j.jcf.2022.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/09/2022] [Accepted: 10/12/2022] [Indexed: 11/22/2022]
|
89
|
Ciofu O, Moser C, Jensen PØ, Høiby N. Tolerance and resistance of microbial biofilms. Nat Rev Microbiol 2022; 20:621-635. [PMID: 35115704 DOI: 10.1038/s41579-022-00682-4] [Citation(s) in RCA: 359] [Impact Index Per Article: 179.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2022] [Indexed: 02/07/2023]
Abstract
Chronic infections caused by microbial biofilms represent an important clinical challenge. The recalcitrance of microbial biofilms to antimicrobials and to the immune system is a major cause of persistence and clinical recurrence of these infections. In this Review, we present the extent of the clinical problem, and the mechanisms underlying the tolerance of biofilms to antibiotics and to host responses. We also explore the role of biofilms in the development of antimicrobial resistance mechanisms.
Collapse
Affiliation(s)
- Oana Ciofu
- Department of Immunology and Microbiology, Costerton Biofilm Center, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Claus Moser
- Department of Immunology and Microbiology, Costerton Biofilm Center, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
| | - Peter Østrup Jensen
- Department of Immunology and Microbiology, Costerton Biofilm Center, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
| | - Niels Høiby
- Department of Immunology and Microbiology, Costerton Biofilm Center, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
90
|
Antimicrobial Peptides Active in In Vitro Models of Endodontic Bacterial Infections Modulate Inflammation in Human Cardiac Fibroblasts. Pharmaceutics 2022; 14:pharmaceutics14102081. [PMID: 36297519 PMCID: PMC9611259 DOI: 10.3390/pharmaceutics14102081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/26/2022] [Accepted: 09/26/2022] [Indexed: 11/30/2022] Open
Abstract
Endodontic and periodontal disease are conditions of infectious origin that can lead to tooth loss or develop into systemic hyperinflammation, which may be associated with a wide variety of diseases, including cardiovascular. Endodontic and periodontal treatment often relies on antibiotics. Since new antimicrobial resistances are a major threat, the use of standard antibiotics is not recommended when the infection is only local. Antimicrobial peptides were recently demonstrated to be valid alternatives for dental treatments. The antimicrobial peptide M33D is a tetrabranched peptide active against Gram-negative and Gram-positive bacteria. It has a long life, unusual for peptides, because its branched form provides resistance to proteases. Here the efficacy of M33D and of its analog M33i/l as antibiotics for local use in dentistry was evaluated. M33D and M33i/l were active against reference strains and multidrug-resistant clinical isolates of Gram-negative and Gram-positive species. Their minimum inhibitory concentration against different strains of dental interest was between 0.4 and 6.0 μM. Both peptides acted rapidly on bacteria, impairing membrane function. They also disrupted biofilm effectively. Disinfection of the root canal is crucial for endodontic treatments. M33D and M33i/l reduced E. faecalis colonies to one-twentieth in a dentin slices model reproducing root canal irrigation. They both captured and neutralized lipopolysaccharide (LPS), a bacterial toxin responsible for inflammation. The release of IL-1β and TNFα by LPS-stimulated murine macrophages was reduced by both peptides. Human cardiac fibroblasts respond to different insults with the release of proinflammatory cytokines, and consequently, they are considered directly involved in atherogenic cardiovascular processes, including those triggered by infections. The presence of M33D and M33i/l at MIC concentration reduced IL6 release from LPS- stimulated human cardiac fibroblasts, hence proving to be promising in preventing bacteria-induced atherogenesis. The two peptides showed low toxicity to mammalian cells, with an EC50 one order of magnitude higher than the average MIC and low hemolytic activity. The development of antimicrobial peptides for dental irrigations and medication is a very promising new field of research that will provide tools to fight dental infections and their severe consequences, while at the same time protecting standard antibiotics from new outbreaks of antimicrobial resistance.
Collapse
|
91
|
Abstract
Pathogenic microorganisms are considered to a major threat to human health, impinging on multiple sectors including hospitals, dentistry, food storage and packaging, and water contamination. Due to the increasing levels of antimicrobial resistance shown by pathogens, often caused by long-term abuse or overuse of traditional antimicrobial drugs, new approaches and solutions are necessary. In this area, antimicrobial polymers are a viable solution to combat a variety of pathogens in a number of contexts. Indeed, polymers with intrinsic antimicrobial activities have long been an intriguing research area, in part, due to their widespread natural abundance in materials such as chitin, chitosan, carrageen, pectin, and the fact that they can be tethered to surfaces without losing their antimicrobial activities. In addition, since the discovery of the strong antimicrobial activity of some synthetic polymers, much work has focused on revealing the most effective structural elements that give rise to optimal antimicrobial properties. This has often been synthesis targeted, with the generation of either new polymers or the modification of natural antimicrobial polymers with the addition of antimicrobial enhancing modalities such as quaternary ammonium or guanidinium groups. In this review, the growing number of polymers showing intrinsic antimicrobial properties from the past decade are highlighted in terms of synthesis; often based on post-synthesis modification and their utilization. This includes as surface coatings, for example on medical devices, such as intravascular catheters, orthopaedic implants and contact lenses, or directly as antibacterial agents (specifically as eye drops). Surface functionalisation with inherently antimicrobial polymers is highlighted and has been achieved via various techniques, including surface-bound initiators allowing RAFT or ATRP surface-based polymerization, or via physical immobilization such as by layer-by-layer techniques. This article also covers the mechanistic modes of action of intrinsic antimicrobial polymers against bacteria, viruses, or fungi.
Collapse
Affiliation(s)
- Meltem Haktaniyan
- EaStCHEM School of Chemistry, University of Edinburgh, David Brewster Road, EH9 3FJ, Edinburgh, UK.
| | - Mark Bradley
- EaStCHEM School of Chemistry, University of Edinburgh, David Brewster Road, EH9 3FJ, Edinburgh, UK.
| |
Collapse
|
92
|
Local Treatment of Driveline Infection with Bacteriophages. Antibiotics (Basel) 2022; 11:antibiotics11101310. [PMID: 36289968 PMCID: PMC9598463 DOI: 10.3390/antibiotics11101310] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/20/2022] [Accepted: 09/20/2022] [Indexed: 11/21/2022] Open
Abstract
Drive line infections (DLI) are common infectious complications after left ventricular assist devices (LVAD) implantation. In case of severe or persistent infections, when conservative management fails, the exchange of the total LVAD may become necessary. We present a case of successful treatment of DL infection with a combination of antibiotics, debridement and local bacteriophage treatment.
Collapse
|
93
|
Synthesized Phosphonium Compounds Demonstrate Resistant Modulatory and Antibiofilm Formation Activities against Some Pathogenic Bacteria. HETEROATOM CHEMISTRY 2022. [DOI: 10.1155/2022/7411957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
A library of six compounds with new hybrids in a single molecule triazole ring attached to the phosphonium salts was synthesized. Click chemistry was, however, used to synthesize the 1-, 2-, and 3-triazole intermediates as a tether for the hybrid phosphonium salts. Their antibacterial activity against Gram-positive bacteria (Staphylococcus aureus and Enterococcus faecalis), Gram-negative bacteria (Escherichia coli and Pseudomonas aeruginosa), and Mycobacterium smegmatis mc2155 was determined using the HT-SPOTi assay. Compound 2 showed the most effective antimicrobial activity as it inhibited the growth of Pseudomonas aeruginosa and Staphylococcus aureus at 0.0125 µg/mL and 31.25 µg/mL, respectively. From the FICI data, compounds 2ET-TOL (2) and RABYL-TOL (4) successfully modulated the activities of amoxicillin against Pseudomonas aeruginosa and Staphylococcus aureus. All the test compounds exhibited a concentration-dependent biofilm formation inhibition against S. aureus, except P-Z (compound 6). Compounds P-MEOXY (1) and 2ET-TOL (2) exhibited mild activity against P. aeruginosa with compound 4 showing antimycobacterial activity at 500 µg/mL.
Collapse
|
94
|
van der Mee-Marquet N, Dos Santos S, Diene SM, Duflot I, Mereghetti L, Valentin AS, François P. Strong Biofilm Formation and Low Cloxacillin Susceptibility in Biofilm-Growing CC398 Staphylococcus aureus Responsible for Bacteremia in French Intensive Care Units, 2021. Microorganisms 2022; 10:microorganisms10091857. [PMID: 36144459 PMCID: PMC9504214 DOI: 10.3390/microorganisms10091857] [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: 08/16/2022] [Revised: 09/07/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
A prospective 3-month study carried out in 267 ICUs revealed an S. aureus nosocomial bacteremia in one admitted patient out of 110 in adult and pediatric sectors, and in one out of 230 newborns; 242 S. aureus bacteremias occurred during the study, including 7.9% MRSA-bacteremias. In one ICU out of ten, the molecular characteristics, antimicrobial susceptibility profiles and biofilm production of the strains responsible for S. aureus bacteremia were studied. Of the 53 strains studied, 9.4% were MRSA and 52.8% were resistant to erythromycin. MLST showed the predominance of CC398 (37.7% of the strains) followed by CC8 (17.0%), CC45 (13.2%) and CC30 (9.4%). The lukF/S genes were absent from our isolates and tst-1 was found in 9.4% of the strains. Under static conditions and without exposure to glucose, biofilm production was rare (9.4% of the strains, without any CC398). The percentage increased to 62.3% for strains grown in broth supplemented with 1% glucose (including 7 out of 9 CC8 and 17 out of the 20 CC398). Further study of the CC398, including whole genome sequencing, revealed (1) highly frequent patient death within seven days after CC398 bacteremia diagnosis (47.4%), (2) 95.0% of the strains producing biofilm when exposed to sub-inhibitory concentrations of cloxacillin, (3) a stronger biofilm production following exposure to cloxacillin than that observed in broth supplemented with glucose only (p < 0.001), (4) a high minimum biofilm eradication concentration of cloxacillin (128 mg/L) indicating a low cloxacillin susceptibility of biofilm-growing CC398, (5) 95.0% of the strains carrying a ϕSa-3 like prophage and its particular evasion cluster (i.e., yielding chp and scin genes), and (6) 30.0% of the strains carrying a ϕMR11-like prophage and yielding a higher ability to produce biofilm. Our results provide evidence that active surveillance is required to avoid spreading of this virulent staphylococcal clone.
Collapse
Affiliation(s)
- Nathalie van der Mee-Marquet
- Centre d’Appui pour la Prévention des Infections Associées aux Soins (CPias) de la Région Centre Val de Loire, Hôpital Bretonneau, Centre Hospitalier Régional Universitaire, 37044 Tours, France
- Correspondence:
| | - Sandra Dos Santos
- Centre d’Appui pour la Prévention des Infections Associées aux Soins (CPias) de la Région Centre Val de Loire, Hôpital Bretonneau, Centre Hospitalier Régional Universitaire, 37044 Tours, France
| | - Seydina M. Diene
- Faculté de Pharmacie, Microbes Evolution Phylogeny and Infections, IHU-Méditerranée Infection, Aix-Marseille Université, 13005 Marseille, France
| | - Isabelle Duflot
- Centre d’Appui pour la Prévention des Infections Associées aux Soins (CPias) de la Région Centre Val de Loire, Hôpital Bretonneau, Centre Hospitalier Régional Universitaire, 37044 Tours, France
| | - Laurent Mereghetti
- Service de Bactériologie, Virologie et Hygiène, Hôpital Trousseau, Centre Hospitalier Régional Universitaire, 37044 Tours, France
| | - Anne-Sophie Valentin
- Centre d’Appui pour la Prévention des Infections Associées aux Soins (CPias) de la Région Centre Val de Loire, Hôpital Bretonneau, Centre Hospitalier Régional Universitaire, 37044 Tours, France
| | - Patrice François
- Department of Medicine, University of Geneva Hospitals, 1205 Geneva, Switzerland
| | | |
Collapse
|
95
|
Guliy OI, Evstigneeva SS, Bunin VD. Microfluidic bioanalytical system for biofilm formation indication. Talanta 2022; 247:123541. [DOI: 10.1016/j.talanta.2022.123541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 05/02/2022] [Accepted: 05/09/2022] [Indexed: 11/27/2022]
|
96
|
Higashihira S, Simpson SJ, Collier CD, Natoli RM, Kittaka M, Greenfield EM. Halicin Is Effective Against Staphylococcus aureus Biofilms In Vitro. Clin Orthop Relat Res 2022; 480:1476-1487. [PMID: 35583504 PMCID: PMC9278916 DOI: 10.1097/corr.0000000000002251] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 04/28/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND Biofilms protect bacteria from the host immune system and many antibiotics, making the treatment of orthopaedic infections difficult. Halicin, a recently discovered antibiotic, has potent activity against nonorthopaedic infections in mice and the planktonic, free-living forms of many bacterial species, including Staphylococcus aureus , a common cause of orthopaedic infections. Importantly, halicin did not induce resistance in vitro and was effective against drug-resistant bacteria and proliferating and quiescent bacteria. Quiescence is an important cause of antibiotic tolerance in biofilms. However, whether halicin acts on biofilms has not been tested. QUESTIONS/PURPOSES (1) Does halicin reduce the viability of S. aureus in less mature and more mature biofilms as it does in planktonic cultures? (2) How do the relative effects of halicin on S. aureus biofilms and planktonic cultures compare with those of conventional antibiotics (tobramycin, cefazolin, vancomycin, or rifampicin) that are commonly used in clinical orthopaedic infections? METHODS To measure minimal biofilm eradication concentrations (MBECs) with less mature 3-day and more mature 7-day biofilms, we used 96-well peg plates that provided high throughput and excellent reproducibility. After S. aureus -Xen36 biofilm formation, planktonic bacteria were removed from the cultures, and the biofilms were exposed to various concentrations of halicin, tobramycin, cefazolin, vancomycin, or rifampicin for 20 hours. Biofilm viability was determined by measuring resazurin reduction or by counting colony-forming units after sonication. To determine effects of halicin and the conventional antibiotics on biofilm viability, we defined MBEC 75 as the lowest concentration that decreased viability by 75% or more. To determine effects on bacterial viability in planktonic cultures, minimum inhibitory concentrations (MICs) were determined with the broth dilution method. Each result was measured in four to 10 independent experiments. RESULTS We found no differences between halicin's effectiveness against planktonic S. aureus and 3-day biofilms (MIC and MBEC 75 for 3-day biofilms was 25 μM [interquartile range 25 to 25 and 25 to 25, respectively]; p > 0.99). Halicin was eightfold less effective against more mature 7-day biofilms (MBEC 75 = 200 μM [100 to 200]; p < 0.001). Similarly, tobramycin was equally effective against planktonic culture and 3-day biofilms (MIC and MBEC 75 for 3-day biofilms was 20 μM [20 to 20 and 10 to 20, respectively]; p > 0.99). Tobramycin's MBEC 75 against more mature 7-day biofilms was 320 μM (320 to 480), which is 16-fold greater than its planktonic MIC (p = 0.03). In contrast, the MBEC 75 for cefazolin, vancomycin, and rifampicin against more mature 7-day biofilms were more than 1000-fold (> 1000; p < 0.001), 500-fold (500 to 875; p < 0.001), and 3125-fold (3125 to 5469; p = 0.004) greater than their planktonic MICs, respectively, consistent with those antibiotics' relative inactivity against biofilms. CONCLUSION Halicin was as effective against S. aureus in less mature 3-day biofilms as those in planktonic cultures, but eightfold higher concentrations were needed for more mature 7-day biofilms. Tobramycin, an antibiotic whose effectiveness depends on biofilm maturity, was also as effective against S. aureus in less mature 3-day biofilms as those in planktonic cultures, but 16-fold higher concentrations were needed for more mature 7-day biofilms. In contrast, cefazolin, vancomycin, and rifampicin were substantially less active against both less and more mature biofilms than against planktonic cultures. CLINICAL RELEVANCE Halicin is a promising antibiotic that may be effective against S. aureus osteomyelitis and infections on orthopaedic implants. Future studies should assess the translational value of halicin by testing its effects in animal models of orthopaedic infections; on the biofilms of other bacterial species, including multidrug-resistant bacteria; and in combination therapy with conventional antibiotics.
Collapse
Affiliation(s)
- Shota Higashihira
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Orthopaedic Surgery, Yokohama City University, Yokohama, Japan
| | - Stefanie Jan Simpson
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Christopher David Collier
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Roman Michael Natoli
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Mizuho Kittaka
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Biomedical Sciences and Comprehensive Care, Indiana University School of Dentistry, Indianapolis, IN, USA
| | - Edward Michael Greenfield
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
97
|
Kim U, Kim JH, Lee SY, Oh SW. Detection of Bacillus cereus and Pseudomonas fluorescens in dual-species biofilm via real-time PCR and eradication using grapefruit seed extract. Lebensm Wiss Technol 2022. [DOI: 10.1016/j.lwt.2022.113679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
98
|
Neutralization of ionic interactions by dextran-based single-chain nanoparticles improves tobramycin diffusion into a mature biofilm. NPJ Biofilms Microbiomes 2022; 8:52. [PMID: 35787627 PMCID: PMC9253323 DOI: 10.1038/s41522-022-00317-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 06/21/2022] [Indexed: 11/18/2022] Open
Abstract
The extracellular matrix protects biofilm cells by reducing diffusion of antimicrobials. Tobramycin is an antibiotic used extensively to treat P. aeruginosa biofilms, but it is sequestered in the biofilm periphery by the extracellular negative charge matrix and loses its efficacy significantly. Dispersal of the biofilm extracellular matrix with enzymes such as DNase I is another promising therapy that enhances antibiotic diffusion into the biofilm. Here, we combine the charge neutralization of tobramycin provided by dextran-based single-chain polymer nanoparticles (SCPNs) together with DNase I to break the biofilm matrix. Our study demonstrates that the SCPNs improve the activity of tobramycin and DNase I by neutralizing the ionic interactions that keep this antibiotic in the biofilm periphery. Moreover, the detailed effects and interactions of nanoformulations with extracellular matrix components were revealed through time-lapse imaging of the P. aeruginosa biofilms by laser scanning confocal microscopy with specific labeling of the different biofilm components.
Collapse
|
99
|
Kaur R, Kanotra M, Sood A, Abdellatif AAH, Bhatia S, Al-Harrasi A, Aleya L, Vargas-De-La-Cruz C, Behl T. Emergence of nutriments as a nascent complementary therapy against antimicrobial resistance. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:49568-49582. [PMID: 35589902 DOI: 10.1007/s11356-022-20775-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 05/08/2022] [Indexed: 06/15/2023]
Abstract
With these growing and evolving years, antimicrobial resistance has become a great subject of interest. The idea of using natural productive ways can be an effective measure against antimicrobial resistance. The growing prevalence of antimicrobial resistance indicates that advanced natural approaches are a topic of concern for fighting the resistance. Many natural products including essential oils, flavonoids, alkaloids and botanicals have been demonstrated as effective bactericidal agents. In this review, we will discuss in detail about the relevance of such natural products to tackle the problem of antimicrobial resistance, antibiotic adjuvants that aim towards non-essential bacterial targets to reduce the prevalence of resistant bacterial infections, latest bioinformatics approach towards antibacterial drug discovery along with an understanding of biogenic nanoparticles in antimicrobial activity.
Collapse
Affiliation(s)
- Rajwinder Kaur
- Chitkara College of Pharmacy, Chitkara University, Patiala, Punjab, India
| | - Muskan Kanotra
- Chitkara College of Pharmacy, Chitkara University, Patiala, Punjab, India
| | - Ankita Sood
- Chitkara College of Pharmacy, Chitkara University, Patiala, Punjab, India
| | - Ahmed A H Abdellatif
- Department of Pharmaceutics, College of Pharmacy, Qassim University, Buraydah, Saudi Arabia
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Al-Azhar University, Assiut, Egypt
| | - Saurabh Bhatia
- Natural & Medical Sciences Research Center, University of Nizwa, Nizwa, Oman
- School of Health Science, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India
| | - Ahmed Al-Harrasi
- Natural & Medical Sciences Research Center, University of Nizwa, Nizwa, Oman
| | - Lotfi Aleya
- Chrono-Environment Laboratory, UMR CNRS 6249, Bourgogne Franche-Comté University, Besancon, France
| | - Celia Vargas-De-La-Cruz
- Department of Pharmacology, Bromatology and Toxicology, Faculty of Pharmacy and Biochemistry, Universidad Nacional Mayor de San Marcos, Lima, Peru
- E-Health Research Center, Universidad de Ciencias Y Humanidades, Lima, Peru
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Patiala, Punjab, India.
| |
Collapse
|
100
|
Okae Y, Nishitani K, Sakamoto A, Kawai T, Tomizawa T, Saito M, Kuroda Y, Matsuda S. Estimation of Minimum Biofilm Eradication Concentration (MBEC) on In Vivo Biofilm on Orthopedic Implants in a Rodent Femoral Infection Model. Front Cell Infect Microbiol 2022; 12:896978. [PMID: 35846761 PMCID: PMC9285014 DOI: 10.3389/fcimb.2022.896978] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 06/01/2022] [Indexed: 12/25/2022] Open
Abstract
The formation of a biofilm on the implant surface is a major cause of intractable implant-associated infection. To investigate the antibiotic concentration needed to eradicate the bacteria inside a biofilm, the minimum biofilm eradication concentration (MBEC) has been used, mostly against in vitro biofilms on plastic surfaces. To produce a more clinically relevant environment, an MBEC assay against biofilms on stainless-steel implants formed in a rat femoral infection model was developed. The rats were implanted with stainless steel screws contaminated by two Staphylococcus aureus strains (UAMS-1, methicillin-sensitive Staphylococcus aureus; USA300LAC, methicillin-resistant Staphylococcus aureus) and euthanized on days 3 and 14. Implants were harvested, washed, and incubated with various concentrations (64–4096 μg/mL) of gentamicin (GM), vancomycin (VA), or cefazolin (CZ) with or without an accompanying systemic treatment dose of VA (20 μg/mL) or rifampicin (RF) (1.5 μg/mL) for 24 h. The implant was vortexed and sonicated, the biofilm was removed, and the implant was re-incubated to determine bacterial recovery. MBEC on the removed biofilm and implant was defined as in vivo MBEC and in vivo implant MBEC, respectively, and the concentrations of 100% and 60% eradication were defined as MBEC100 and MBEC60, respectively. As for in vivo MBEC, MBEC100 of GM was 256–1024 μg/mL, but that of VA and CZ ranged from 2048–4096 μg/mL. Surprisingly, the in vivo implant MBEC was much higher, ranging from 2048 μg/mL to more than 4096 μg/mL. The addition of RF, not VA, as a secondary antibiotic was effective, and MBEC60 on day 3 USA300LAC biofilm was reduced from 1024 μg/mL with GM alone to 128 μg/mL in combination with RF and the MBEC60 on day 14 USA300LAC biofilm was reduced from 2048 μg/mL in GM alone to 256 μg/mL in combination with RF. In conclusion, a novel MBEC assay for in vivo biofilms on orthopedic implants was developed. GM was the most effective against both methicillin-sensitive and methicillin-resistant Staphylococcus aureus, in in vivo biofilms, and the addition of a systemic concentration of RF reduced MBEC of GM. Early initiation of treatment is desired because the required concentration of antibiotics increases with biofilm maturation.
Collapse
|