101
|
Carriot N, Barry-Martinet R, Briand JF, Ortalo-Magné A, Culioli G. Impact of phosphate concentration on the metabolome of biofilms of the marine bacterium Pseudoalteromonas lipolytica. Metabolomics 2022; 18:18. [PMID: 35290545 DOI: 10.1007/s11306-022-01875-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 02/22/2022] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Marine biofilms are the most widely distributed mode of life on Earth and drive biogeochemical cycling processes of most elements. Phosphorus (P) is essential for many biological processes such as energy transfer mechanisms, biological information storage and membrane integrity. OBJECTIVES Our aim was to analyze the effect of a gradient of ecologically relevant phosphate concentrations on the biofilm-forming capacity and the metabolome of the marine bacterium Pseudoalteromonas lipolytica TC8. METHODS In addition to the evaluation of the effect of different phosphate concentration on the biomass, structure and gross biochemical composition of biofilms of P. lipolytica TC8, untargeted metabolomics based on liquid chromatography-mass spectrometry (LC-MS) analysis was used to determine the main metabolites impacted by P-limiting conditions. Annotation of the most discriminating and statistically robust metabolites was performed through the concomitant use of molecular networking and MS/MS fragmentation pattern interpretation. RESULTS At the lowest phosphate concentration, biomass, carbohydrate content and three-dimensional structures of biofilms tended to decrease. Furthermore, untargeted metabolomics allowed for the discrimination of the biofilm samples obtained at the five phosphate concentrations and the highlighting of a panel of metabolites mainly implied in such a discrimination. A large part of the metabolites of the resulting dataset were then putatively annotated. Ornithine lipids were found in increasing quantity when the phosphate concentration decreased, while the opposite trend was observed for oxidized phosphatidylethanolamines (PEs). CONCLUSION This study demonstrated the suitability of LC-MS-based untargeted metabolomics for evaluating the effect of culture conditions on marine bacterial biofilms. More precisely, these results supported the high plasticity of the membrane of P. lipolytica TC8, while the role of the oxidized PEs remains to be clarified.
Collapse
Affiliation(s)
- Nathan Carriot
- Laboratoire MAPIEM, Université de Toulon, EA 4323, La Garde, France
| | | | | | | | - Gérald Culioli
- Laboratoire MAPIEM, Université de Toulon, EA 4323, La Garde, France.
- Institut Méditerranéen de Biodiversité et d'Ecologie Marine et Continentale (IMBE), UMR CNRS-IRD-Avignon, Université-Aix-Marseille Université, Avignon, France.
| |
Collapse
|
102
|
Nielsen JE, Alford MA, Yung DBY, Molchanova N, Fortkort JA, Lin JS, Diamond G, Hancock REW, Jenssen H, Pletzer D, Lund R, Barron AE. Self-Assembly of Antimicrobial Peptoids Impacts Their Biological Effects on ESKAPE Bacterial Pathogens. ACS Infect Dis 2022; 8:533-545. [PMID: 35175731 DOI: 10.1021/acsinfecdis.1c00536] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Antimicrobial peptides (AMPs) are promising pharmaceutical candidates for the prevention and treatment of infections caused by multidrug-resistant ESKAPE pathogens, which are responsible for the majority of hospital-acquired infections. Clinical translation of AMPs has been limited, in part by apparent toxicity on systemic dosing and by instability arising from susceptibility to proteolysis. Peptoids (sequence-specific oligo-N-substituted glycines) resist proteolytic digestion and thus are of value as AMP mimics. Only a few natural AMPs such as LL-37 and polymyxin self-assemble in solution; whether antimicrobial peptoids mimic these properties has been unknown. Here, we examine the antibacterial efficacy and dynamic self-assembly in aqueous media of eight peptoid mimics of cationic AMPs designed to self-assemble and two nonassembling controls. These amphipathic peptoids self-assembled in different ways, as determined by small-angle X-ray scattering; some adopt helical bundles, while others form core-shell ellipsoidal or worm-like micelles. Interestingly, many of these peptoid assemblies show promising antibacterial, antibiofilm activity in vitro in media, under host-mimicking conditions and antiabscess activity in vivo. While self-assembly correlated overall with antibacterial efficacy, this correlation was imperfect. Certain self-assembled morphologies seem better-suited for antibacterial activity. In particular, a peptoid exhibiting a high fraction of long, worm-like micelles showed reduced antibacterial, antibiofilm, and antiabscess activity against ESKAPE pathogens compared with peptoids that form ellipsoidal or bundled assemblies. This is the first report of self-assembling peptoid antibacterials with activity against in vivo biofilm-like infections relevant to clinical medicine.
Collapse
Affiliation(s)
- Josefine Eilsø Nielsen
- Department of Bioengineering, School of Medicine, Stanford University, Stanford, California 94305, United States
- Department of Chemistry, University of Oslo, Oslo 0315, Norway
| | - Morgan Ashley Alford
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Deborah Bow Yue Yung
- Department of Microbiology and Immunology, University of Otago, Dunedin 9054, New Zealand
| | - Natalia Molchanova
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - John A. Fortkort
- Department of Bioengineering, School of Medicine, Stanford University, Stanford, California 94305, United States
| | - Jennifer S. Lin
- Department of Bioengineering, School of Medicine, Stanford University, Stanford, California 94305, United States
| | - Gill Diamond
- Department of Oral Immunology and Infectious Diseases, University of Louisville, School of Dentistry, Louisville, Kentucky 40202, United States
| | - Robert E. W. Hancock
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Håvard Jenssen
- Department of Science and Environment, Roskilde University, Roskilde 4000, Denmark
| | - Daniel Pletzer
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
- Department of Microbiology and Immunology, University of Otago, Dunedin 9054, New Zealand
| | - Reidar Lund
- Department of Chemistry, University of Oslo, Oslo 0315, Norway
| | - Annelise E. Barron
- Department of Bioengineering, School of Medicine, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
103
|
Strain-specific interspecies interactions between co-isolated pairs of Staphylococcus aureus and Pseudomonas aeruginosa from patients with tracheobronchitis or bronchial colonization. Sci Rep 2022; 12:3374. [PMID: 35233050 PMCID: PMC8888623 DOI: 10.1038/s41598-022-07018-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 02/09/2022] [Indexed: 01/20/2023] Open
Abstract
Dual species interactions in co-isolated pairs of Staphylococcus aureus and Pseudomonas aeruginosa from patients with tracheobronchitis or bronchial colonization were examined. The genetic and phenotypic diversity between the isolates was high making the interactions detected strain-specific. Despite this, and the clinical origin of the strains, some interactions were common between some co-isolated pairs. For most pairs, P. aeruginosa exoproducts affected biofilm formation and reduced growth in vitro in its S. aureus counterpart. Conversely, S. aureus did not impair biofilm formation and stimulated swarming motility in P. aeruginosa. Co-culture in a medium that mimics respiratory mucus promoted coexistence and favored mixed microcolony formation within biofilms. Under these conditions, key genes controlled by quorum sensing were differentially regulated in both species in an isolate-dependent manner. Finally, co-infection in the acute infection model in Galleria mellonella larvae showed an additive effect only in the co-isolated pair in which P. aeruginosa affected less S. aureus growth. This work contributes to understanding the complex interspecies interactions between P. aeruginosa and S. aureus by studying strains isolated during acute infection.
Collapse
|
104
|
Sultan AR, Tavakol M, Lemmens-den Toom NA, Croughs PD, Verkaik NJ, Verbon A, van Wamel WJB. Real time monitoring of Staphylococcus aureus biofilm sensitivity towards antibiotics with isothermal microcalorimetry. PLoS One 2022; 17:e0260272. [PMID: 35171906 PMCID: PMC8849495 DOI: 10.1371/journal.pone.0260272] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 11/05/2021] [Indexed: 12/14/2022] Open
Abstract
Biofilm-associated infections with Staphylococcus aureus are difficult to treat even after administration of antibiotics that according to the standard susceptibility assays are effective. Currently, the assays used in the clinical laboratories to determine the sensitivity of S. aureus towards antibiotics are not representing the behaviour of biofilm-associated S. aureus, since these assays are performed on planktonic bacteria. In research settings, microcalorimetry has been used for antibiotic susceptibility studies. Therefore, in this study we investigated if we can use isothermal microcalorimetry to monitor the response of biofilm towards antibiotic treatment in real-time. We developed a reproducible method to generate biofilm in an isothermal microcalorimeter setup. Using this system, the sensitivity of 5 methicillin-sensitive S. aureus (MSSA) and 5 methicillin-resistant S. aureus (MRSA) strains from different genetic lineages were determined towards: flucloxacillin, cefuroxime, cefotaxime, gentamicin, rifampicin, vancomycin, levofloxacin, clindamycin, erythromycin, linezolid, fusidic acid, co-trimoxazole, and doxycycline. In contrast to conventional assays, our calorimetry-based biofilm susceptibility assay showed that S. aureus biofilms, regardless MSSA or MRSA, can survive the exposure to the maximum serum concentration of all tested antibiotics. The only treatment with a single antibiotic showing a significant reduction in biofilm survival was rifampicin, yet in 20% of the strains, emerging antibiotic resistance was observed. Furthermore, the combination of rifampicin with flucloxacillin, vancomycin or levofloxacin was able to prevent S. aureus biofilm from becoming resistant to rifampicin. Isothermal microcalorimetry allows real-time monitoring of the sensitivity of S. aureus biofilms towards antibiotics in a fast and reliable way.
Collapse
Affiliation(s)
- Andi Rofian Sultan
- Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Microbiology, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Mehri Tavakol
- Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Nicole A. Lemmens-den Toom
- Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Peter D. Croughs
- Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Nelianne J. Verkaik
- Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Annelies Verbon
- Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Willem J. B. van Wamel
- Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Center, Rotterdam, The Netherlands
- * E-mail:
| |
Collapse
|
105
|
Antibiofilm and immunomodulatory resorbable nanofibrous filing for dental pulp regenerative procedures. Bioact Mater 2022; 16:173-186. [PMID: 35386316 PMCID: PMC8965695 DOI: 10.1016/j.bioactmat.2022.01.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 01/05/2022] [Accepted: 01/17/2022] [Indexed: 12/22/2022] Open
|
106
|
Baldelli A, Etayash H, Oguzlu H, Mandal R, Jiang F, Hancock RE, Pratap-Singh A. Antimicrobial properties of spray-dried cellulose nanocrystals and metal oxide-based nanoparticles-in-microspheres. CHEMICAL ENGINEERING JOURNAL ADVANCES 2022. [DOI: 10.1016/j.ceja.2022.100273] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
107
|
Castro J, Lima Â, Sousa LGV, Rosca AS, Muzny CA, Cerca N. Crystal Violet Staining Alone Is Not Adequate to Assess Synergism or Antagonism in Multi-Species Biofilms of Bacteria Associated With Bacterial Vaginosis. Front Cell Infect Microbiol 2022; 11:795797. [PMID: 35071046 PMCID: PMC8766793 DOI: 10.3389/fcimb.2021.795797] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 12/15/2021] [Indexed: 12/14/2022] Open
Abstract
Bacterial Vaginosis (BV) involves the presence of a multi-species biofilm adhered to vaginal epithelial cells, but its in-depth study has been limited due to the complexity of the bacterial community, which makes the design of in vitro models challenging. Perhaps the most common experimental technique to quantify biofilms is the crystal violet (CV) staining method. Despite its widespread utilization, the CV method is not without flaws. While biofilm CV quantification within the same strain in different conditions is normally accepted, assessing multi-species biofilms formation by CV staining might provide significant bias. For BV research, determining possible synergism or antagonism between species is a fundamental step for assessing the roles of individual species in BV development. Herein, we provide our perspective on how CV fails to properly quantify an in vitro triple-species biofilm composed of Gardnerella vaginalis, Fannyhessea (Atopobium) vaginae, and Prevotella bivia, three common BV-associated bacteria thought to play key roles in incident BV pathogenesis. We compared the CV method with total colony forming units (CFU) and fluorescence microscopy cell count methods. Not surprisingly, when comparing single-species biofilms, the relationship between biofilm biomass, total number of cells, and total cultivable cells was very different between each tested method, and also varied with the time of incubation. Thus, despite its wide utilization for single-species biofilm quantification, the CV method should not be considered for accurate quantification of multi-species biofilms in BV pathogenesis research.
Collapse
Affiliation(s)
- Joana Castro
- Centre of Biological Engineering (CEB), Laboratory of Research in Biofilms Rosário Oliveira (LIBRO), University of Minho, Braga, Portugal
| | - Ângela Lima
- Centre of Biological Engineering (CEB), Laboratory of Research in Biofilms Rosário Oliveira (LIBRO), University of Minho, Braga, Portugal
| | - Lúcia G V Sousa
- Centre of Biological Engineering (CEB), Laboratory of Research in Biofilms Rosário Oliveira (LIBRO), University of Minho, Braga, Portugal
| | - Aliona S Rosca
- Centre of Biological Engineering (CEB), Laboratory of Research in Biofilms Rosário Oliveira (LIBRO), University of Minho, Braga, Portugal
| | - Christina A Muzny
- Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Nuno Cerca
- Centre of Biological Engineering (CEB), Laboratory of Research in Biofilms Rosário Oliveira (LIBRO), University of Minho, Braga, Portugal
| |
Collapse
|
108
|
Agarwal S, Kallmyer NE, Vang DX, Ramirez AV, Islam MM, Hillier AC, Halverson L, Reuel NF. Single-Walled Carbon Nanotube Probes for the Characterization of Biofilm-Degrading Enzymes Demonstrated against Pseudomonas aeruginosa Extracellular Matrices. Anal Chem 2022; 94:856-865. [PMID: 34939783 PMCID: PMC9150823 DOI: 10.1021/acs.analchem.1c03633] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Hydrolase co-therapies that degrade biofilm extracellular polymeric substances (EPS) allow for a better diffusion of antibiotics and more effective treatment; current methods for quantitatively measuring the enzymatic degradation of EPS are not amendable to high-throughput screening. Herein, we present biofilm EPS-functionalized single-walled carbon nanotube (SWCNT) probes for rapid screening of hydrolytic enzyme selectivity and activity on EPS. The extent of biofilm EPS degradation is quantified by monitoring the quenching of the SWCNT fluorescence. We used this platform to screen 16 hydrolases with varying bond breaking selectivity against a panel of wild-type Pseudomonas aeruginosa and mutants deficient or altered in one or more EPS. Next, we performed concentration-dependent studies of six enzymes on two common strains found in cystic fibrosis (CF) environments and, for each enzyme, extracted three first-order rate constants and their relative contributions by fitting a parallel, multi-site degradation model, with a good model fit (R2 from 0.65 to 0.97). Reaction rates (turnover rates) are dependent on the enzyme concentration and range from 6.67 × 10-11 to 2.80 × 10-3 *s-1 per mg/mL of enzymes. Lastly, we confirmed findings from this new assay using an established crystal-violet staining assay for a subset of hydrolase panels. In summary, our work shows that this modular sensor is amendable to the high-throughput screening of EPS degradation, thereby improving the rate of discovery and development of novel hydrolases.
Collapse
Affiliation(s)
- Sparsh Agarwal
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa, 50011, United States
| | - Nathaniel E Kallmyer
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa, 50011, United States
| | - Dua X Vang
- Department of Plant Pathology and Microbiology, Iowa State University, Ames, Iowa, 50011, United States,Interdepartmental Microbiology Graduate Program, Iowa State University, Ames, Iowa 50011, United States
| | - Alma V Ramirez
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa, 50011, United States
| | - Md Monirul Islam
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa, 50011, United States
| | - Andrew C Hillier
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa, 50011, United States
| | - Larry Halverson
- Department of Plant Pathology and Microbiology, Iowa State University, Ames, Iowa, 50011, United States,Interdepartmental Microbiology Graduate Program, Iowa State University, Ames, Iowa 50011, United States
| | - Nigel F Reuel
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa, 50011, United States,Interdepartmental Microbiology Graduate Program, Iowa State University, Ames, Iowa 50011, United States,Corresponding Author: Prof. Nigel F Reuel, 2114 Sweeney Hall, 618 Bissell Rd, Iowa State University, Ames, IA, 50011, United States, , Ph: 515-294-4592
| |
Collapse
|
109
|
Honey antibacterial activity: A neglected aspect of honey quality assurance as functional food. Trends Food Sci Technol 2021. [DOI: 10.1016/j.tifs.2021.11.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
110
|
Yang S, Zhang J, Yang R, Xu X. Small Molecule Compounds, A Novel Strategy against Streptococcus mutans. Pathogens 2021; 10:pathogens10121540. [PMID: 34959495 PMCID: PMC8708136 DOI: 10.3390/pathogens10121540] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/17/2021] [Accepted: 11/22/2021] [Indexed: 02/05/2023] Open
Abstract
Dental caries, as a common oral infectious disease, is a worldwide public health issue. Oral biofilms are the main cause of dental caries. Streptococcus mutans (S. mutans) is well recognized as the major causative factor of dental caries within oral biofilms. In addition to mechanical removal such as tooth brushing and flossing, the topical application of antimicrobial agents is necessarily adjuvant to the control of caries particularly for high-risk populations. The mainstay antimicrobial agents for caries such as chlorhexidine have limitations including taste confusions, mucosal soreness, tooth discoloration, and disruption of an oral microbial equilibrium. Antimicrobial small molecules are promising in the control of S. mutans due to good antimicrobial activity, good selectivity, and low toxicity. In this paper, we discussed the application of antimicrobial small molecules to the control of S. mutans, with a particular focus on the identification and development of active compounds and their modes of action against the growth and virulence of S. mutans.
Collapse
Affiliation(s)
- Sirui Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu 610041, China; (S.Y.); (J.Z.)
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jin Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu 610041, China; (S.Y.); (J.Z.)
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ran Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu 610041, China; (S.Y.); (J.Z.)
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Correspondence: (R.Y.); (X.X.)
| | - Xin Xu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu 610041, China; (S.Y.); (J.Z.)
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Correspondence: (R.Y.); (X.X.)
| |
Collapse
|
111
|
Etayash H, Alford M, Akhoundsadegh N, Drayton M, Straus SK, Hancock REW. Multifunctional Antibiotic-Host Defense Peptide Conjugate Kills Bacteria, Eradicates Biofilms, and Modulates the Innate Immune Response. J Med Chem 2021; 64:16854-16863. [PMID: 34784220 DOI: 10.1021/acs.jmedchem.1c01712] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Effective anti-infective therapies are required to offset the rise in antibiotic resistance. A novel vancomycin-innate defense regulator conjugate (V-IDR1018) was constructed with multimodal functionality, including bacterial killing, biofilm eradication, and immune modulation. The conjugate killed bacteria within 30 min, exhibited potent activity against persister cells, and showed no susceptibility to antimicrobial resistance in tissue culture assays. Additionally, it stimulated the release of chemokine MCP-1 and anti-inflammatory cytokine IL-10 and suppressed pro-inflammatory IL-1β from lipopolysaccharide-stimulated white blood cells. The conjugate demonstrated ∼90% eradication efficacy when assessed against the MRSA biofilm formed on an organoid human skin equivalent. Similarly, when evaluated in a murine, high-density skin abscess infection model using MRSA or Staphylococcus epidermidis, the conjugate decreased dermonecrosis and reduced bacterial load. The exceptional in vitro and in vivo efficacy of the conjugate, in addition to its safety profile, makes it a valuable candidate to treat complex infectious diseases.
Collapse
Affiliation(s)
- Hashem Etayash
- Centre for Microbial Diseases and Immunity Research, Department of Microbiology and Immunology, University of British Columbia, 2259 Lower Mall Research Station, Vancouver V6T 1Z4, British Columbia, Canada
| | - Morgan Alford
- Centre for Microbial Diseases and Immunity Research, Department of Microbiology and Immunology, University of British Columbia, 2259 Lower Mall Research Station, Vancouver V6T 1Z4, British Columbia, Canada
| | - Noushin Akhoundsadegh
- Centre for Microbial Diseases and Immunity Research, Department of Microbiology and Immunology, University of British Columbia, 2259 Lower Mall Research Station, Vancouver V6T 1Z4, British Columbia, Canada
| | - Matthew Drayton
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver V6T 1Z1, British Columbia, Canada
| | - Suzana K Straus
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver V6T 1Z1, British Columbia, Canada
| | - Robert E W Hancock
- Centre for Microbial Diseases and Immunity Research, Department of Microbiology and Immunology, University of British Columbia, 2259 Lower Mall Research Station, Vancouver V6T 1Z4, British Columbia, Canada
| |
Collapse
|
112
|
Are Uropathogenic Bacteria Living in Multispecies Biofilm Susceptible to Active Plant Ingredient-Asiatic Acid? Biomolecules 2021; 11:biom11121754. [PMID: 34944398 PMCID: PMC8698853 DOI: 10.3390/biom11121754] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/21/2021] [Accepted: 11/22/2021] [Indexed: 02/07/2023] Open
Abstract
Urinary tract infections (UTIs) are a serious health problem in the human population due to their chronic and recurrent nature. Bacteria causing UTIs form multispecies biofilms being resistant to the activity of the conventionally used antibiotics. Therefore, compounds of plant origin are currently being searched for, which could constitute an alternative strategy to antibiotic therapy. Our study aimed to determine the activity of asiatic acid (AA) against biofilms formed by uropathogenic Escherichia coli, Enterobacter cloacae, and Pseudomonas aeruginosa. The influence of AA on the survival, biofilm mass formation by bacteria living in mono-, dual-, and triple-species consortia as well as the metabolic activity and bacterial cell morphology were determined. The spectrophotometric methods were used for biofilm mass synthesis and metabolic activity determination. The survival of bacteria was established using the serial dilution assay. The decrease in survival and a weakening of the ability to create biofilms, both single and multi-species, as well as changes in the morphology of bacterial cells were noticed. As AA works best against young biofilms, the use of AA-containing formulations, especially during the initial stages of infection, seems to be reasonable. However, there is a need for further research concerning AA especially regarding its antibacterial mechanisms of action.
Collapse
|
113
|
Serna N, Carratalá JV, Conchillo-Solé O, Martínez-Torró C, Unzueta U, Mangues R, Ferrer-Miralles N, Daura X, Vázquez E, Villaverde A. Antibacterial Activity of T22, a Specific Peptidic Ligand of the Tumoral Marker CXCR4. Pharmaceutics 2021; 13:1922. [PMID: 34834337 PMCID: PMC8621837 DOI: 10.3390/pharmaceutics13111922] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/21/2021] [Accepted: 11/05/2021] [Indexed: 12/13/2022] Open
Abstract
CXCR4 is a cytokine receptor used by HIV during cell attachment and infection. Overexpressed in the cancer stem cells of more than 20 human neoplasias, CXCR4 is a convenient antitumoral drug target. T22 is a polyphemusin-derived peptide and an effective CXCR4 ligand. Its highly selective CXCR4 binding can be exploited as an agent for the cell-targeted delivery and internalization of associated antitumor drugs. Sharing chemical and structural traits with antimicrobial peptides (AMPs), the capability of T22 as an antibacterial agent remains unexplored. Here, we have detected T22-associated antimicrobial activity and biofilm formation inhibition over Escherichia coli, Staphylococcus aureus and Pseudomonas aeruginosa, in a spectrum broader than the reference AMP GWH1. In contrast to GWH1, T22 shows neither cytotoxicity over mammalian cells nor hemolytic activity and is active when displayed on protein-only nanoparticles through genetic fusion. Under the pushing need for novel antimicrobial agents, the discovery of T22 as an AMP is particularly appealing, not only as its mere addition to the expanding catalogue of antibacterial drugs. The recognized clinical uses of T22 might allow its combined and multivalent application in complex clinical conditions, such as colorectal cancer, that might benefit from the synchronous destruction of cancer stem cells and local bacterial biofilms.
Collapse
Affiliation(s)
- Naroa Serna
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain; (N.S.); (J.V.C.); (O.C.-S.); (C.M.-T.); (N.F.-M.); (E.V.)
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain;
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain;
| | - José Vicente Carratalá
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain; (N.S.); (J.V.C.); (O.C.-S.); (C.M.-T.); (N.F.-M.); (E.V.)
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain;
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain;
| | - Oscar Conchillo-Solé
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain; (N.S.); (J.V.C.); (O.C.-S.); (C.M.-T.); (N.F.-M.); (E.V.)
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain;
| | - Carlos Martínez-Torró
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain; (N.S.); (J.V.C.); (O.C.-S.); (C.M.-T.); (N.F.-M.); (E.V.)
| | - Ugutz Unzueta
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain;
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain;
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain
- Josep Carreras Research Institute, 08916 Barcelona, Spain
| | - Ramón Mangues
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain;
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain
- Josep Carreras Research Institute, 08916 Barcelona, Spain
| | - Neus Ferrer-Miralles
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain; (N.S.); (J.V.C.); (O.C.-S.); (C.M.-T.); (N.F.-M.); (E.V.)
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain;
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain;
| | - Xavier Daura
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain; (N.S.); (J.V.C.); (O.C.-S.); (C.M.-T.); (N.F.-M.); (E.V.)
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain;
- Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain
| | - Esther Vázquez
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain; (N.S.); (J.V.C.); (O.C.-S.); (C.M.-T.); (N.F.-M.); (E.V.)
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain;
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain;
| | - Antonio Villaverde
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain; (N.S.); (J.V.C.); (O.C.-S.); (C.M.-T.); (N.F.-M.); (E.V.)
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain;
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain;
| |
Collapse
|
114
|
Fathollahi A, Coupe SJ. Effect of environmental and nutritional conditions on the formation of single and mixed-species biofilms and their efficiency in cadmium removal. CHEMOSPHERE 2021; 283:131152. [PMID: 34147985 DOI: 10.1016/j.chemosphere.2021.131152] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/29/2021] [Accepted: 06/05/2021] [Indexed: 06/12/2023]
Abstract
Remediation of contaminated water and wastewater using biosorption methods has attracted significant attention in recent decades due to its efficiency, convenience and minimised environmental effects. Bacterial biosorbents are normally deployed as a non-living powder or suspension. Little is known about the mechanisms or rates of bacterial attachment to surfaces and effect of various conditions on the biofilm development, as well as efficiency of living biofilms in the removal of heavy metals. In the present study, the effect of environmental and nutritional conditions such as pH, temperature, concentrations of phosphate, glucose, amino acid, nitrate, calcium and magnesium, on planktonic and biofilm growth of single and mixed bacterial cultures, were measured. Actinomyces meyeri, Bacillus cereus, Escherichia coli, Pseudomonas fluorescens strains were evaluated to determine the optimum biofilm growth conditions. The Cd(II) biosorption efficiencies of the mixed-species biofilm developed in the optimum growth condition, were investigated and modelled using Langmuir, Freundlich and Dubnin Radushkevich models. The biofilm quantification techniques revealed that the optimum concentration of phosphate, glucose, amino acid, nitrate, calcium and magnesium for the biofilm development were 25, 10, 1, 1.5, 5 and 0.5 g L-1, respectively. Further increases in the nutrient concentrations resulted in less biofilm growth. The optimum pH for the biofilm growth was 7 and alkaline or acidic conditions caused significant negative effects on the bacterial attachment and development. The optimum temperatures for the bacterial attachment to the surface were between 25 and 35 °C. The maximum Cd(II) biosorption efficiency (99%) and capacity (18.19 mg g-1) of the mixed-species biofilm, occurred on day 35 (Ci = 0.1 mg L-1) and 1 (Ci = 20 mg L-1) of biofilm growth, respectively. Modelling of the biosorption data revealed that Cd(II) removal by the living biofilm was a physical process by a monolayer of biofilm. The results of present study suggested that environmental and nutritional conditions had a significant effect on bacterial biofilm formation and its efficiency in Cd(II) removal.
Collapse
Affiliation(s)
- Alireza Fathollahi
- Centre for Agroecology Water and Resilience (CAWR), Coventry University, Wolston Lane, Ryton on Dunsmore, CV8 3LG, UK.
| | - Stephen J Coupe
- Centre for Agroecology Water and Resilience (CAWR), Coventry University, Wolston Lane, Ryton on Dunsmore, CV8 3LG, UK
| |
Collapse
|
115
|
Periodically Disturbing the Spatial Structure of Biofilms Can Affect the Production of an Essential Virulence Factor in Pseudomonas aeruginosa. mSystems 2021; 6:e0096121. [PMID: 34581603 PMCID: PMC8547473 DOI: 10.1128/msystems.00961-21] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Understanding the environmental factors that affect the production of virulence factors has major implications in evolution and medicine. While spatial structure is important in virulence factor production, observations of this relationship have occurred in undisturbed or continuously disturbed environments. However, natural environments are subject to periodic fluctuations, including changes in physical forces, which could alter the spatial structure of bacterial populations and impact virulence factor production. Using Pseudomonas aeruginosa PA14, we periodically applied a physical force to biofilms and examined production of pyoverdine. Intermediate frequencies of disturbance reduced the amount of pyoverdine produced compared to undisturbed or frequently disturbed conditions. To explore the generality of this finding, we examined how an intermediate disturbance frequency affected pyoverdine production in 21 different strains of P. aeruginosa. Periodic disturbance increased, decreased, or did not change the amount of pyoverdine produced relative to undisturbed populations. Mathematical modeling predicts that interactions between pyoverdine synthesis rate and biofilm density determine the amount of pyoverdine synthesized. When the pyoverdine synthesis rates are high, depletion of the biofilm due to disturbance reduces the accumulation of pyoverdine. At intermediate synthesis rates, production of pyoverdine increases during disturbance as bacteria dispersed into the planktonic state enjoy increased growth and pyoverdine production rates. At low synthesis rates, disturbance does not alter the amount of pyoverdine produced since disturbance-driven access to nutrients does not augment pyoverdine synthesis. Our results suggest that environmental conditions shape robustness in the production of virulence factors and may lead to novel approaches to treat infections. IMPORTANCE Virulence factors are required to cause infections. Previous work has shown that the spatial organization of a population, such as a biofilm, can increase the production of some virulence factors, including pyoverdine, which is produced by Pseudomonas aeruginosa. Pyoverdine is essential for the infection process, and reducing its production can limit infections. We have discovered that periodically changing the spatial structure of a biofilm of P. aeruginosa strain PA14 using a physical force can reduce the production of pyoverdine. A mathematical model suggests that this is due to the disruption of spatial organization. Using additional strains of P. aeruginosa isolated from patients and the environment, we use experiments and modeling to show that this reduction in pyoverdine is due to interactions between biofilm density and the synthesis rate of pyoverdine. Our results identify conditions where pyoverdine production is reduced and may lead to novel ways to treat infections.
Collapse
|
116
|
Piątkowska E, Paleczny J, Dydak K, Letachowicz K. Antimicrobial activity of hemodialysis catheter lock solutions in relation to other compounds with antiseptic properties. PLoS One 2021; 16:e0258148. [PMID: 34618850 PMCID: PMC8496847 DOI: 10.1371/journal.pone.0258148] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 09/18/2021] [Indexed: 12/15/2022] Open
Abstract
Proper protection of vascular access after haemodialysis is one of the key measures for the prevention of catheter-related infections. Various substances with bactericidal and anticoagulant properties are used to fill catheters, but due to the unsatisfactory clinical effects and occurrence of adverse reactions, the search for new substances is still ongoing. In the present paper, we compared the in vitro antimicrobial activity of solutions used for tunnelled catheter locking (taurolidine, trisodium citrate) and solutions of substances that could potentially be used for this purpose (sodium bicarbonate, polyhexanide-betaine). The studies have been conducted on bacteria that most commonly cause catheter-related infections. The values of both minimum inhibitory concentration and minimum biofilm eradication concentration of the substances were determined. The ability of the tested substances to eradicate biofilm from the dialysis catheter surface was also evaluated. The results showed that polyhexanide-betaine inhibited the growth of all microbes comparably to taurolidine, even after ≥ 32-fold dilution. The activity of trisodium citrate and sodium bicarbonate was significantly lower. Polyhexanide exhibited the highest activity in the eradication of bacterial biofilm on polystyrene plates. The biofilm formed on a polyurethane dialysis catheter was resistant to complete eradication by the test substances. Polyhexanide-betaine and taurolidine showed the highest activity. Inhibition of bacterial growth regardless of species was observed not only at the highest concentration of these compounds but also after dilution 32-128x (taurolidine) and 32-1024x (polyhexanide-betaine). Therefore, it can be assumed that taurolidine application as a locking solution prevents catheter colonization and systemic infection development. Taurolidine displays high antimicrobial efficacy against Gram-positive cocci as well as Gram-negative bacilli. On the contrary, the lowest antibacterial effect displayed product contained sodium bicarbonate. The inhibitions of bacterial growth were not satisfactory to consider it as a substance for colonization prevention. Polyhexanidine-betaine possessed potent inhibitory and biofilm eradication properties comparing to all tested products. PHMB is applied as a wound irrigation solution worldwide. However, based on our results, we assume that the PHMB is a promising substance for catheter locking solutions thanks to its safety and high antimicrobial properties.
Collapse
Affiliation(s)
- Elżbieta Piątkowska
- Department of Pharmaceutical Microbiology and Parasitology, Medical University of Wroclaw, Wroclaw, Poland
| | - Justyna Paleczny
- Department of Pharmaceutical Microbiology and Parasitology, Medical University of Wroclaw, Wroclaw, Poland
| | - Karolina Dydak
- Department of Pharmaceutical Microbiology and Parasitology, Medical University of Wroclaw, Wroclaw, Poland
| | - Krzysztof Letachowicz
- Department of Nephrology and Transplantation Medicine, Medical University of Wroclaw, Wroclaw, Poland
| |
Collapse
|
117
|
Charpentier E, Doudet L, Allart-Simon I, Colin M, Gangloff SC, Gérard S, Reffuveille F. Synergy between Indoloquinolines and Ciprofloxacin: An Antibiofilm Strategy against Pseudomonas aeruginosa. Antibiotics (Basel) 2021; 10:antibiotics10101205. [PMID: 34680786 PMCID: PMC8532862 DOI: 10.3390/antibiotics10101205] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/28/2021] [Accepted: 10/01/2021] [Indexed: 01/01/2023] Open
Abstract
Antibiotic treatments can participate in the formation of bacterial biofilm in case of under dosage. The interest of indoloquinoline scaffold for drug discovery incited us to study the preparation of new indolo [2,3-b]quinoline derivatives by a domino radical process. We tested the effect of two different “indoloquinoline” molecules (Indol-1 and Indol-2) without antimicrobial activity, in addition to ciprofloxacin, on biofilm formation thanks to crystal violet staining and enumeration of adhered bacteria. This association of ciprofloxacin and Indol-1 or Indol-2 attenuated the formation of biofilm up to almost 80% compared to ciprofloxacin alone, or even prevented the presence of adhered bacteria. In conclusion, these data prove that the association of non-antimicrobial molecules with an antibiotic can be a solution to fight against biofilm and antibiotic resistance emergence.
Collapse
Affiliation(s)
- Emilie Charpentier
- EA 4691 Biomatériaux et Inflammation en Site Osseux (BIOS), UFR Pharmacie, Université de Reims Champagne-Ardenne, SFR Cap Santé (FED 4231), 51097 Reims, France; (E.C.); (M.C.); (S.C.G.)
| | - Ludovic Doudet
- Institut de Chimie Moléculaire de Reims (ICMR-UMR CNRS 7312), UFR Pharmacie, Université de Reims Champagne-Ardenne, 51097 Reims, France; (L.D.); (I.A.-S.); (S.G.)
| | - Ingrid Allart-Simon
- Institut de Chimie Moléculaire de Reims (ICMR-UMR CNRS 7312), UFR Pharmacie, Université de Reims Champagne-Ardenne, 51097 Reims, France; (L.D.); (I.A.-S.); (S.G.)
| | - Marius Colin
- EA 4691 Biomatériaux et Inflammation en Site Osseux (BIOS), UFR Pharmacie, Université de Reims Champagne-Ardenne, SFR Cap Santé (FED 4231), 51097 Reims, France; (E.C.); (M.C.); (S.C.G.)
| | - Sophie C. Gangloff
- EA 4691 Biomatériaux et Inflammation en Site Osseux (BIOS), UFR Pharmacie, Université de Reims Champagne-Ardenne, SFR Cap Santé (FED 4231), 51097 Reims, France; (E.C.); (M.C.); (S.C.G.)
| | - Stéphane Gérard
- Institut de Chimie Moléculaire de Reims (ICMR-UMR CNRS 7312), UFR Pharmacie, Université de Reims Champagne-Ardenne, 51097 Reims, France; (L.D.); (I.A.-S.); (S.G.)
| | - Fany Reffuveille
- EA 4691 Biomatériaux et Inflammation en Site Osseux (BIOS), UFR Pharmacie, Université de Reims Champagne-Ardenne, SFR Cap Santé (FED 4231), 51097 Reims, France; (E.C.); (M.C.); (S.C.G.)
- Correspondence:
| |
Collapse
|
118
|
Grigorova EV, Nemchenko UM, Voropaeva NM, Belkova NL, Noskova OA, Savilov ED. Effect of Disinfectants with Different Active Ingredients on Biofilm Formation in Pseudomonas aeruginosa. Bull Exp Biol Med 2021; 171:745-749. [PMID: 34705178 DOI: 10.1007/s10517-021-05308-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Indexed: 11/30/2022]
Abstract
We compared the effects of disinfectants on biofilms of 10 Pseudomonas aeruginosa strains isolated from different loci of patients with purulent-septic infections. Identification was carried out by standard bacteriological methods. To substantiate the prospects of using in hospitals and to assess the effect of disinfectants on P. aeruginosa biofilms, the following disinfectants were used in various concentrations: Sekusept Aktiv, A-DEZ, and Monitor Oxy. All clinical strains of P. aeruginosa showed the ability to form biofilms. Both oxygen-containing and quaternary ammonium compounds effectively inhibited the formation of biofilms. In more than 50% cases, disinfectants with different active ingredients did not destroy pre-formed of P. aeruginosa biofilms.
Collapse
Affiliation(s)
- E V Grigorova
- Research Center for Family Health and Human Reproduction Problems, Irkutsk, Russia.
| | - U M Nemchenko
- Research Center for Family Health and Human Reproduction Problems, Irkutsk, Russia
| | - N M Voropaeva
- Research Center for Family Health and Human Reproduction Problems, Irkutsk, Russia
| | - N L Belkova
- Research Center for Family Health and Human Reproduction Problems, Irkutsk, Russia
| | - O A Noskova
- Research Center for Family Health and Human Reproduction Problems, Irkutsk, Russia
| | - E D Savilov
- Research Center for Family Health and Human Reproduction Problems, Irkutsk, Russia
| |
Collapse
|
119
|
Comparison of Two Cutibacterium acnes Biofilm Models. Microorganisms 2021; 9:microorganisms9102035. [PMID: 34683356 PMCID: PMC8540958 DOI: 10.3390/microorganisms9102035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/13/2021] [Accepted: 09/23/2021] [Indexed: 11/30/2022] Open
Abstract
The study of biofilms in vitro is complex and often limited by technical problems due to simplified models. Here, we compared C. acnes biofilm formation, from species involved in bone and prosthesis infection, in a static model with a dynamic model. Using similar parameters, the percentage of live bacteria within the biofilm was higher in dynamic than in static approach. In both models, bacterial internalization in osteoblast-like cells, playing the role of stress factor, affected this proportion but in opposite ways: increase of live bacteria proportion in the static model (×2.04 ± 0.53) and of dead bacteria proportion (×3.5 ± 1.03) in the dynamic model. This work highlights the huge importance in the selection of a relevant biofilm model in accordance with the environmental or clinical context to effectively improve the understanding of biofilms and the development of better antibiofilm strategies.
Collapse
|
120
|
Lamret F, Varin-Simon J, Velard F, Terryn C, Mongaret C, Colin M, Gangloff SC, Reffuveille F. Staphylococcus aureus Strain-Dependent Biofilm Formation in Bone-Like Environment. Front Microbiol 2021; 12:714994. [PMID: 34557170 PMCID: PMC8453086 DOI: 10.3389/fmicb.2021.714994] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 07/30/2021] [Indexed: 11/23/2022] Open
Abstract
Staphylococcus aureus species is an important threat for hospital healthcare because of frequent colonization of indwelling medical devices such as bone and joint prostheses through biofilm formations, leading to therapeutic failure. Furthermore, bacteria within biofilm are less sensitive to the host immune system responses and to potential antibiotic treatments. We suggested that the periprosthetic bone environment is stressful for bacteria, influencing biofilm development. To provide insights into S. aureus biofilm properties of three strains [including one methicillin-resistant S. aureus (MRSA)] under this specific environment, we assessed several parameters related to bone conditions and expected to affect biofilm characteristics. We reported that the three strains harbored different behaviors in response to the lack of oxygen, casamino acids and glucose starvation, and high concentration of magnesium. Each strain presented different biofilm biomass and live adherent cells proportion, or matrix production and composition. However, the three strains shared common responses in a bone-like environment: a similar production of extracellular DNA and engagement of the SOS response. This study is a step toward a better understanding of periprosthetic joint infections and highlights targets, which could be common among S. aureus strains and for future antibiofilm strategies.
Collapse
Affiliation(s)
- Fabien Lamret
- Université de Reims Champagne-Ardenne, Laboratory BIOS EA 4691, Reims, France
| | | | - Frédéric Velard
- Université de Reims Champagne-Ardenne, Laboratory BIOS EA 4691, Reims, France
| | - Christine Terryn
- Plateforme en Imagerie Cellulaire et Tissulaire, Université de Reims Champagne-Ardenne, Reims, France
| | - Céline Mongaret
- Université de Reims Champagne-Ardenne, Laboratory BIOS EA 4691, Reims, France.,Service Pharmacie, Centre Hospitalier Universitaire de Reims, Reims, France
| | - Marius Colin
- Université de Reims Champagne-Ardenne, Laboratory BIOS EA 4691, Reims, France.,Université de Reims Champagne-Ardenne, UFR de Pharmacie, Reims, France
| | - Sophie C Gangloff
- Université de Reims Champagne-Ardenne, Laboratory BIOS EA 4691, Reims, France.,Université de Reims Champagne-Ardenne, UFR de Pharmacie, Reims, France
| | - Fany Reffuveille
- Université de Reims Champagne-Ardenne, Laboratory BIOS EA 4691, Reims, France.,Université de Reims Champagne-Ardenne, UFR de Pharmacie, Reims, France
| |
Collapse
|
121
|
Shukla SK, Sharma AK, Gupta V, Kalonia A, Shaw P. Challenges with Wound Infection Models in Drug Development. Curr Drug Targets 2021; 21:1301-1312. [PMID: 32116189 DOI: 10.2174/1389450121666200302093312] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/06/2020] [Accepted: 02/06/2020] [Indexed: 01/05/2023]
Abstract
Wound research is an evolving science trying to unfold the complex untold mechanisms behind the wound healing cascade. In particular, interest is growing regarding the role of microorganisms in both acute and chronic wound healing. Microbial burden plays an important role in the persistence of chronic wounds, ultimately resulting in delayed wound healing. It is therefore important for clinicians to understand the evolution of infection science and its various etiologies. Therefore, to understand the role of bacterial biofilm in chronic wound pathogenesis, various in vitro and in vivo models are required to investigate biofilms in wound-like settings. Infection models should be refined comprising an important signet of biofilms. These models are eminent for translational research to obtain data for designing an improved wound care formulation. However, all the existing models possess limitations and do not fit properly in the model frame for developing wound care agents. Among various impediments, one of the major drawbacks of such models is that the wound they possess does not mimic the wound a human develops. Therefore, a novel wound infection model is required which can imitate the human wounds. This review article mainly discusses various in vitro and in vivo models showing microbial colonization, their advantages and challenges. Apart from these models, there are also present ex vivo wound infection models, but this review mainly focused on various in vitro and in vivo models available for studying wound infection in controlled conditions. This information might be useful in designing an ideal wound infection model for developing an effective wound healing formulation.
Collapse
Affiliation(s)
- Sandeep K Shukla
- Institute of Nuclear Medicine & Allied Sciences, Defence Research and Development Organization, SK Mazumdar Marg, Timarpur, Delhi-110054, India
| | - Ajay K Sharma
- Institute of Nuclear Medicine & Allied Sciences, Defence Research and Development Organization, SK Mazumdar Marg, Timarpur, Delhi-110054, India
| | - Vanya Gupta
- Graphic Era deemed to be University, Dehradun, India
| | - Aman Kalonia
- Institute of Nuclear Medicine & Allied Sciences, Defence Research and Development Organization, SK Mazumdar Marg, Timarpur, Delhi-110054, India
| | - Priyanka Shaw
- Institute of Nuclear Medicine & Allied Sciences, Defence Research and Development Organization, SK Mazumdar Marg, Timarpur, Delhi-110054, India
| |
Collapse
|
122
|
Taggar R, Singh S, Bhalla V, Bhattacharyya MS, Sahoo DK. Deciphering the Antibacterial Role of Peptide From Bacillus subtilis subsp. spizizenii Ba49 Against Staphylococcus aureus. Front Microbiol 2021; 12:708712. [PMID: 34489898 PMCID: PMC8417246 DOI: 10.3389/fmicb.2021.708712] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/23/2021] [Indexed: 02/04/2023] Open
Abstract
An increase in antibiotic resistance has led to escalating the need for the development of alternate therapy. Antimicrobial peptides (AMPs) are at the forefront of replacing conventional antibiotics, showing slower development of drug resistance, antibiofilm activity, and the ability to modulate the host immune response. The ESKAPE (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species) pathogens that jeopardize most conventional antibiotics are known to be involved in severe respiratory tract, bloodstream, urinary tract, soft tissue, and skin infections. Among them, S. aureus is an insidious microbe and developed resistance against conventional antibiotics. In the present study, an AMP (named as peptide-Ba49) isolated from Bacillus subtilis subsp. spizizenii strain from Allium cepa (the common onion) exhibited strong antibacterial efficacy against S. aureus ATCC 25923. The mode of action of this peptide-Ba49 on S. aureus was deciphered through various sensitive probes, i.e., DiSC3 (5) and H2DCFDA, suggesting the peptide-Ba49 to be acting upon through change in membrane potential and by triggering the production of reactive oxygen species (ROS). This induced disruption of the cell membrane was further supported by morphological studies using scanning electron microscopy (SEM). Investigations on a possible post-antibiotic effect (PAE) of peptide-Ba49 showed prolonged PAE against S. aureus. Furthermore, the peptide-Ba49 prevented the formation of S. aureus biofilm at low concentration and showed its potential to degrade the mature biofilm of S. aureus. The peptide-Ba49 also exhibited intracellular killing potential against S. aureus ATCC 25923 in the macrophage cells, and moreover, peptide-Ba49 was found to bolster the fibroblast cell migration in the scratch assay at low concentration, exhibiting a wound healing efficacy of this peptide. These studies demonstrated that peptide-Ba49 isolated from the strain B. subtilis subsp. spizizenii could be a therapeutic candidate to combat the pathogenic S. aureus infections.
Collapse
Affiliation(s)
- Ramita Taggar
- CSIR-Institute of Microbial Technology, Sector 39A, Chandigarh, India
| | - Sanpreet Singh
- CSIR-Institute of Microbial Technology, Sector 39A, Chandigarh, India
| | - Vijayender Bhalla
- CSIR-Institute of Microbial Technology, Sector 39A, Chandigarh, India
| | | | - Debendra K Sahoo
- CSIR-Institute of Microbial Technology, Sector 39A, Chandigarh, India.,Academy of Scientific and Innovative Research, New Delhi, India
| |
Collapse
|
123
|
Blanco-Cabra N, López-Martínez MJ, Arévalo-Jaimes BV, Martin-Gómez MT, Samitier J, Torrents E. A new BiofilmChip device for testing biofilm formation and antibiotic susceptibility. NPJ Biofilms Microbiomes 2021; 7:62. [PMID: 34344902 PMCID: PMC8333102 DOI: 10.1038/s41522-021-00236-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 07/15/2021] [Indexed: 12/15/2022] Open
Abstract
Currently, three major circumstances threaten the management of bacterial infections: increasing antimicrobial resistance, expansion of chronic biofilm-associated infections, and lack of an appropriate approach to treat them. To date, the development of accelerated drug susceptibility testing of biofilms and of new antibiofouling systems has not been achieved despite the availability of different methodologies. There is a need for easy-to-use methods of testing the antibiotic susceptibility of bacteria that form biofilms and for screening new possible antibiofilm strategies. Herein, we present a microfluidic platform with an integrated interdigitated sensor (BiofilmChip). This new device allows an irreversible and homogeneous attachment of bacterial cells of clinical origin, even directly from clinical specimens, and the biofilms grown can be monitored by confocal microscopy or electrical impedance spectroscopy. The device proved to be suitable to study polymicrobial communities, as well as to measure the effect of antimicrobials on biofilms without introducing disturbances due to manipulation, thus better mimicking real-life clinical situations. Our results demonstrate that BiofilmChip is a straightforward tool for antimicrobial biofilm susceptibility testing that could be easily implemented in routine clinical laboratories.
Collapse
Affiliation(s)
- Núria Blanco-Cabra
- Bacterial Infections and Antimicrobial Therapies Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Maria José López-Martínez
- Nanobioengineering Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN) Monforte de Lemos 3-5, Madrid, Spain
- Department of Electronics and Biomedical Engineering, University of Barcelona, Barcelona, Spain
| | - Betsy Verónica Arévalo-Jaimes
- Bacterial Infections and Antimicrobial Therapies Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | | | - Josep Samitier
- Nanobioengineering Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN) Monforte de Lemos 3-5, Madrid, Spain
- Department of Electronics and Biomedical Engineering, University of Barcelona, Barcelona, Spain
| | - Eduard Torrents
- Bacterial Infections and Antimicrobial Therapies Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Microbiology Section, Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
124
|
Testing physiologically relevant conditions in minimal inhibitory concentration assays. Nat Protoc 2021; 16:3761-3774. [PMID: 34215865 DOI: 10.1038/s41596-021-00572-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 05/13/2021] [Indexed: 02/06/2023]
Abstract
The minimal inhibitory concentration (MIC) assay uses agar or broth dilution methods to measure, under defined test conditions, the lowest effective concentration of an antimicrobial agent that inhibits visible growth of a bacterium of interest. This assay is used to test the susceptibilities of bacterial isolates and of novel antimicrobial drugs, and is typically done in nutrient-rich laboratory media that have little relevance to in vivo conditions. As an extension to our original protocol on MIC assays (also published in Nature Protocols), here we describe the application of the MIC broth microdilution assay to test antimicrobial susceptibility in conditions that are more physiologically relevant to infections observed in the clinic. Specifically, we describe a platform that can be applied to the preparation of medium that mimics lung and wound exudate or blood conditions for the growth and susceptibility testing of bacteria, including ESKAPE pathogens. This protocol can also be applied to most physiologically relevant liquid medium and aerobic pathogens, and takes 3-4 d to complete.
Collapse
|
125
|
Santana FL, Arenas I, Haney EF, Estrada K, Hancock REW, Corzo G. Identification of a crocodylian β-defensin variant from Alligator mississippiensis with antimicrobial and antibiofilm activity. Peptides 2021; 141:170549. [PMID: 33865931 DOI: 10.1016/j.peptides.2021.170549] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/30/2021] [Accepted: 03/31/2021] [Indexed: 12/19/2022]
Abstract
β-defensin host defense peptides are important components of the innate immune system of vertebrates. Although evidence of their broad antimicrobial, antibiofilm and immunomodulatory activities in mammals have been presented, β-defensins from other vertebrate species, like crocodylians, remain largely unexplored. In this study, five new crocodylian β-defensin variants from Alligator mississippiensis and Crocodylus porosus were selected for synthesis and characterization based on their charge and hydrophobicity values. Linear peptides were synthesized, folded, purified and then evaluated for their antimicrobial and antibiofilm activities against the bacterial pathogens, Salmonella enterica serovar Typhimurium, Staphylococcus aureus, Enterobacter cloacae and Acinetobacter baumannii. The Am23SK variant (SCRFSGGYCIWNWERCRSGHFLVALCPFRKRCCK) from A. mississippiensis displayed promising activity against both planktonic cells and bacterial biofilms, outperforming the human β-defensin 3 under the experimental conditions. Moreover, Am23SK exhibited no cytotoxicity towards mammalian cells and exerted immunomodulatory effects in vitro, moderately suppressing the production of proinflammatory mediators from stimulated human bronchial epithelial cells. Overall, our results have expanded the activity landscape of crocodylian and reptilian β-defensin in general.
Collapse
Affiliation(s)
- Felix L Santana
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, A.P. 510-3, Cuernavaca, Mor., 62250, Mexico; Centre for Microbial Diseases and Immunity Research, University of British Columbia, 2259 Lower Mall Research Station, Vancouver, BC, V6T1Z4, Canada
| | - Iván Arenas
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, A.P. 510-3, Cuernavaca, Mor., 62250, Mexico
| | - Evan F Haney
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, 2259 Lower Mall Research Station, Vancouver, BC, V6T1Z4, Canada
| | - Karel Estrada
- Unidad de Secuenciación Masiva y Bioinformática, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - Robert E W Hancock
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, 2259 Lower Mall Research Station, Vancouver, BC, V6T1Z4, Canada
| | - Gerardo Corzo
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, A.P. 510-3, Cuernavaca, Mor., 62250, Mexico.
| |
Collapse
|
126
|
Crudo F, Aichinger G, Mihajlovic J, Varga E, Dellafiora L, Warth B, Dall'Asta C, Berry D, Marko D. In vitro interactions of Alternaria mycotoxins, an emerging class of food contaminants, with the gut microbiota: a bidirectional relationship. Arch Toxicol 2021; 95:2533-2549. [PMID: 33847775 PMCID: PMC8241668 DOI: 10.1007/s00204-021-03043-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 04/01/2021] [Indexed: 12/11/2022]
Abstract
The human gut microbiota plays an important role in the maintenance of human health. Factors able to modify its composition might predispose the host to the development of pathologies. Among the various xenobiotics introduced through the diet, Alternaria mycotoxins are speculated to represent a threat for human health. However, limited data are currently available about the bidirectional relation between gut microbiota and Alternaria mycotoxins. In the present work, we investigated the in vitro effects of different concentrations of a complex extract of Alternaria mycotoxins (CE; containing eleven mycotoxins; e.g. 0.153 µM alternariol and 2.3 µM altersetin, at the maximum CE concentration tested) on human gut bacterial strains, as well as the ability of the latter to metabolize or adsorb these compounds. Results from the minimum inhibitory concentration assay showed the scarce ability of CE to inhibit the growth of the tested strains. However, the growth kinetics of most of the strains were negatively affected by exposure to the various CE concentrations, mainly at the highest dose (50 µg/mL). The CE was also found to antagonize the formation of biofilms, already at concentrations of 0.5 µg/mL. LC-MS/MS data analysis of the mycotoxin concentrations found in bacterial pellets and supernatants after 24 h incubation showed the ability of bacterial strains to adsorb some Alternaria mycotoxins, especially the key toxins alternariol, alternariol monomethyl ether, and altersetin. The tendency of these mycotoxins to accumulate within bacterial pellets, especially in those of Gram-negative strains, was found to be directly related to their lipophilicity.
Collapse
Affiliation(s)
- Francesco Crudo
- Department of Food Chemistry and Toxicology, University of Vienna, Währinger Str. 38, 1090, Wien, Austria
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124, Parma, Italy
| | - Georg Aichinger
- Department of Food Chemistry and Toxicology, University of Vienna, Währinger Str. 38, 1090, Wien, Austria
| | - Jovana Mihajlovic
- Department of Microbiology and Ecosystem Science, Centre for Microbiology and Environmental Systems Science, University of Vienna, Althanstr. 14, 1090, Vienna, Austria
| | - Elisabeth Varga
- Department of Food Chemistry and Toxicology, University of Vienna, Währinger Str. 38, 1090, Wien, Austria
| | - Luca Dellafiora
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124, Parma, Italy
| | - Benedikt Warth
- Department of Food Chemistry and Toxicology, University of Vienna, Währinger Str. 38, 1090, Wien, Austria
| | - Chiara Dall'Asta
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124, Parma, Italy
| | - David Berry
- Department of Food Chemistry and Toxicology, University of Vienna, Währinger Str. 38, 1090, Wien, Austria
- Department of Microbiology and Ecosystem Science, Centre for Microbiology and Environmental Systems Science, University of Vienna, Althanstr. 14, 1090, Vienna, Austria
- Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Vienna, Austria
| | - Doris Marko
- Department of Food Chemistry and Toxicology, University of Vienna, Währinger Str. 38, 1090, Wien, Austria.
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124, Parma, Italy.
| |
Collapse
|
127
|
Antibiofilm activity of host defence peptides: complexity provides opportunities. Nat Rev Microbiol 2021; 19:786-797. [PMID: 34183822 DOI: 10.1038/s41579-021-00585-w] [Citation(s) in RCA: 145] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2021] [Indexed: 12/21/2022]
Abstract
Host defence peptides (HDPs) are integral components of innate immunity across all living organisms. These peptides can exert direct antibacterial effects, targeting planktonic cells (referred to as antimicrobial peptides), and exhibit antibiofilm (referred to as antibiofilm peptides), antiviral, antifungal and host-directed immunomodulatory activities. In this Review, we discuss how the complex functional attributes of HDPs provide many opportunities for the development of antimicrobial therapeutics, focusing particularly on their emerging antibiofilm properties. The mechanisms of action of antibiofilm peptides are compared and contrasted with those of antimicrobial peptides. Furthermore, obstacles for the practical translation of candidate peptides into therapeutics and the potential solutions are discussed. Critically, HDPs have the value-added assets of complex functional attributes, particularly antibiofilm and anti-inflammatory activities and their synergy with conventional antibiotics.
Collapse
|
128
|
Improving Phage-Biofilm In Vitro Experimentation. Viruses 2021; 13:v13061175. [PMID: 34205417 PMCID: PMC8234374 DOI: 10.3390/v13061175] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 06/14/2021] [Accepted: 06/15/2021] [Indexed: 02/07/2023] Open
Abstract
Bacteriophages or phages, the viruses of bacteria, are abundant components of most ecosystems, including those where bacteria predominantly occupy biofilm niches. Understanding the phage impact on bacterial biofilms therefore can be crucial toward understanding both phage and bacterial ecology. Here, we take a critical look at the study of bacteriophage interactions with bacterial biofilms as carried out in vitro, since these studies serve as bases of our ecological and therapeutic understanding of phage impacts on biofilms. We suggest that phage-biofilm in vitro experiments often may be improved in terms of both design and interpretation. Specific issues discussed include (a) not distinguishing control of new biofilm growth from removal of existing biofilm, (b) inadequate descriptions of phage titers, (c) artificially small overlying fluid volumes, (d) limited explorations of treatment dosing and duration, (e) only end-point rather than kinetic analyses, (f) importance of distinguishing phage enzymatic from phage bacteriolytic anti-biofilm activities, (g) limitations of biofilm biomass determinations, (h) free-phage interference with viable-count determinations, and (i) importance of experimental conditions. Toward bettering understanding of the ecology of bacteriophage-biofilm interactions, and of phage-mediated biofilm disruption, we discuss here these various issues as well as provide tips toward improving experiments and their reporting.
Collapse
|
129
|
Blackman LD, Qu Y, Cass P, Locock KES. Approaches for the inhibition and elimination of microbial biofilms using macromolecular agents. Chem Soc Rev 2021; 50:1587-1616. [PMID: 33403373 DOI: 10.1039/d0cs00986e] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Biofilms are complex three-dimensional structures formed at interfaces by the vast majority of bacteria and fungi. These robust communities have an important detrimental impact on a wide range of industries and other facets of our daily lives, yet their removal is challenging owing to the high tolerance of biofilms towards conventional antimicrobial agents. This key issue has driven an urgent search for new innovative antibiofilm materials. Amongst these emerging approaches are highly promising materials that employ aqueous-soluble macromolecules, including peptides, proteins, synthetic polymers, and nanomaterials thereof, which exhibit a range of functionalities that can inhibit biofilm formation or detach and destroy organisms residing within established biofilms. In this Review, we outline the progress made in inhibiting and removing biofilms using macromolecular approaches, including a spotlight on cutting-edge materials that respond to environmental stimuli for "on-demand" antibiofilm activity, as well as synergistic multi-action antibiofilm materials. We also highlight materials that imitate and harness naturally derived species to achieve new and improved biomimetic and biohybrid antibiofilm materials. Finally, we share some speculative insights into possible future directions for this exciting and highly significant field of research.
Collapse
Affiliation(s)
- Lewis D Blackman
- CSIRO Manufacturing, Research Way, Clayton, VIC 3168, Australia.
| | - Yue Qu
- Infection and Immunity Program, Department of Microbiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia and Department of Infectious Diseases, The Alfred Hospital and Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Peter Cass
- CSIRO Manufacturing, Research Way, Clayton, VIC 3168, Australia.
| | | |
Collapse
|
130
|
Sousa MGDC, Xavier PD, Cantuária APDC, Amorim IA, Almeida JA, Franco OL, Rezende TMB. Antimicrobial and immunomodulatory in vitro profile of double antibiotic paste. Int Endod J 2021; 54:1850-1860. [PMID: 34033685 DOI: 10.1111/iej.13576] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 05/19/2021] [Accepted: 05/21/2021] [Indexed: 12/20/2022]
Abstract
AIM To evaluate the antimicrobial and immunomodulatory activity of double antibiotic paste (DAP) in an in vitro infection model. METHODOLOGY The minimum inhibitory and bactericidal concentrations (MIC and MBC) and the antibiofilm activities (TTC assay) of DAP and its components (ciprofloxacin and metronidazole) were evaluated against Staphylococcus aureus and Enterococcus faecalis compared with triple antibiotic paste (TAP). The cellular viability of RAW 264.7 macrophages (24 and 72 h) and L929 fibroblasts (48 and 72 h) was evaluated by MTT. Furthermore, the production of TNF-α, IL-12, IL-6, IL-1α, IL-10 and NO (on RAW 264.7), besides IL-6, TGF-β and NO (on L929), stimulated with DAP in baseline and associated with heat-killed microbial-antigen conditions was measured by ELISA and Griess reaction. Data were analysed using the one-way ANOVA test with Bonferroni's corrections. RESULTS The MBC of pharmacopoeia DAP was similar to TAP for E. faecalis (0.25 μg. mL-1 ) and lower for S. aureus (DAP 1 μg. mL-1 and TAP 2 μg. mL-1 ; p < .001). Ciprofloxacin was the most effective antibiofilm drug from the pastes (35% of reduction for E. faecalis and S. aureus; p < .0001), and both pastes had a similar antibiofilm eradication against both biofilm species (29% and 35% for S. aureus and 76% and 85% for E. faecalis; p < .0001). DAP was cytotoxic against the tested cells. DAP significantly upregulated IL-1α (p < .001), IL-6 (p < .0001), TNF-α (p < .01) and IL-12 (p < .05; in the absence of antigens) and significantly reduced IL-6 (p < .0001; in the presence of HK-S. aureus) and IL-10 (p < .05; in the presence of both antigens) on macrophages. Furthermore, DAP upregulated IL-6 (p < .001) and NO (p < .05; in the absence of antigens), IL-6 (p < .001; in the presence of HK-S. aureus) and reduced NO (p < .001; in the presence of HK-S. aureus). CONCLUSIONS Double antibiotic paste and TAP had similar antimicrobial activity against S. aureus and E. faecalis. DAP upregulated pro-inflammatory cytokines mainly in the absence of antigens and had pro- and anti-inflammatory activity in RAW 264.7 macrophages and L929 fibroblasts in the presence of antigens involved in pulp infections.
Collapse
Affiliation(s)
- Maurício Gonçalves da Costa Sousa
- Pós-Graduação em Ciências Genômicas e Biotecnologia, Centro de Análises Proteômicas e Bioquímicas, Universidade Católica de Brasília, Brasília, Brazil
| | | | | | - Ingrid Aquino Amorim
- Pós-Graduação em Ciências Genômicas e Biotecnologia, Centro de Análises Proteômicas e Bioquímicas, Universidade Católica de Brasília, Brasília, Brazil.,Curso de Odontologia, Centro Universitário Unieuro, Brasília, Brazil
| | - Jeeser Alves Almeida
- Pós-Graduação em Saúde e Desenvolvimento na Região Centro Oeste, Faculdade de Medicina, Universidade Federal de Mato Grosso do Sul, Campo Grande, Brazil
| | - Octavio Luiz Franco
- Pós-Graduação em Ciências Genômicas e Biotecnologia, Centro de Análises Proteômicas e Bioquímicas, Universidade Católica de Brasília, Brasília, Brazil.,Pós-Graduação em Patologia Molecular, Universidade de Brasília, Brasília, Brazil.,S-Inova Biotech, Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Brazil
| | - Taia Maria Berto Rezende
- Pós-Graduação em Ciências Genômicas e Biotecnologia, Centro de Análises Proteômicas e Bioquímicas, Universidade Católica de Brasília, Brasília, Brazil.,Pós-Graduação em Ciências da Saúde, Faculdade de Ciências da Saúde, Universidade de Brasília, Brasília, Brazil.,Curso de Odontologia, Universidade Católica de Brasília, Brasília, Brazil
| |
Collapse
|
131
|
Biofilm viability checker: An open-source tool for automated biofilm viability analysis from confocal microscopy images. NPJ Biofilms Microbiomes 2021; 7:44. [PMID: 33990612 PMCID: PMC8121819 DOI: 10.1038/s41522-021-00214-7] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 03/12/2021] [Indexed: 12/15/2022] Open
Abstract
Quantifying biofilm formation on surfaces is challenging because traditional microbiological methods, such as total colony-forming units (CFUs), often rely on manual counting. These are laborious, resource intensive techniques, more susceptible to human error. Confocal laser scanning microscopy (CLSM) is a high-resolution technique that allows 3D visualisation of biofilm architecture. In combination with a live/dead stain, it can be used to quantify biofilm viability on both transparent and opaque surfaces. However, there is little consensus on the appropriate methodology to apply in confocal micrograph processing. In this study, we report the development of an image analysis approach to repeatably quantify biofilm viability and surface coverage. We also demonstrate its use for a range of bacterial species and translational applications. This protocol has been created with ease of use and accessibility in mind, to enable researchers who do not specialise in computational techniques to be confident in applying these methods to analyse biofilm micrographs. Furthermore, the simplicity of the method enables the user to adapt it for their bespoke needs. Validation experiments demonstrate the automated analysis is robust and accurate across a range of bacterial species and an improvement on traditional microbiological analysis. Furthermore, application to translational case studies show the automated method is a reliable measurement of biomass and cell viability. This approach will ensure image analysis is an accessible option for those in the microbiology and biomaterials field, improve current detection approaches and ultimately support the development of novel strategies for preventing biofilm formation by ensuring comparability across studies.
Collapse
|
132
|
Curtin AM, Buckley HL. Biofouling detection methods that are widely applicable and useful across disciplines: a mini-review. BIOFOULING 2021; 37:494-505. [PMID: 34193005 DOI: 10.1080/08927014.2021.1926998] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/23/2021] [Accepted: 05/03/2021] [Indexed: 06/13/2023]
Abstract
Biofouling, or the build-up of microorganisms in a biofilm at the solid-water or water-air interface, is an interdisciplinary problem. Biofouling causes various issues including clogging systems, contaminating devices, and creating infections that are extremely difficult to treat, to name but a few. Therefore, engineers, pharmacologists, microbiologists, wastewater treatment operators, chemists, food preservative formulators, home and personal care product formulators, and toxicologists all play a role in studying and have an interest in solving biofouling. High-throughput studies on biofilm prevention and removal can take the form of biofilm antimicrobial microdilution susceptibility (BAMS) tests. Due to vested interests of many disciplines, the results from these tests should be applicable and useful to each discipline. This critical review analyses the focuses, biological implications, and metrics required by each discipline. The possible detection methods that could satisfy each desired metric are then summarized. The detection methods were analysed in order to recommend two methods of biofilm detection, Crystal Violet stain and the LIVE/DEAD BacLight stain, which correspond with three metrics including total biomass, log reduction, and the MIC, BPC, MBIC, MBC, BBC, and/or MBEC values. Determining these three metrics for each BAMS test will allow this type of research to be widely applicable and useful across many disciplines.
Collapse
Affiliation(s)
- Anna M Curtin
- Civil Engineering Department, Green Safe Water Lab, University of Victoria, Victoria, British Columbia, Canada
| | - Heather L Buckley
- Civil Engineering Department, Green Safe Water Lab, University of Victoria, Victoria, British Columbia, Canada
| |
Collapse
|
133
|
Haney EF, Trimble MJ, Hancock REW. Microtiter plate assays to assess antibiofilm activity against bacteria. Nat Protoc 2021; 16:2615-2632. [PMID: 33911258 DOI: 10.1038/s41596-021-00515-3] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 02/03/2021] [Indexed: 02/02/2023]
Abstract
Bacterial biofilms demonstrate high broad-spectrum adaptive antibiotic resistance and cause two thirds of all infections, but there is a lack of approved antibiofilm agents. Unlike the standard minimal inhibitory concentration assay to assess antibacterial activity against planktonic cells, there is no standardized method to evaluate biofilm inhibition and/or eradication capacity of novel antibiofilm compounds. The protocol described here outlines simple and reproducible methods for assessing the biofilm inhibition and eradication capacities of novel antibiofilm agents against adherent bacterial biofilms grown in 96-well microtiter plates. It employs two inexpensive dyes: crystal violet to stain adhered biofilm biomass and 2,3,5-triphenyl tetrazolium chloride to quantify metabolism of the biofilm cells. The procedure is accessible to any laboratory with a plate reader, requires minimal technical expertise or training and takes 4 or 5 d to complete. Recommendations for how biofilm inhibition and eradication results should be interpreted and presented are also described.
Collapse
Affiliation(s)
- Evan F Haney
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michael J Trimble
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Robert E W Hancock
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
134
|
An AY, Choi KYG, Baghela AS, Hancock REW. An Overview of Biological and Computational Methods for Designing Mechanism-Informed Anti-biofilm Agents. Front Microbiol 2021; 12:640787. [PMID: 33927701 PMCID: PMC8076610 DOI: 10.3389/fmicb.2021.640787] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 03/23/2021] [Indexed: 12/29/2022] Open
Abstract
Bacterial biofilms are complex and highly antibiotic-resistant aggregates of microbes that form on surfaces in the environment and body including medical devices. They are key contributors to the growing antibiotic resistance crisis and account for two-thirds of all infections. Thus, there is a critical need to develop anti-biofilm specific therapeutics. Here we discuss mechanisms of biofilm formation, current anti-biofilm agents, and strategies for developing, discovering, and testing new anti-biofilm agents. Biofilm formation involves many factors and is broadly regulated by the stringent response, quorum sensing, and c-di-GMP signaling, processes that have been targeted by anti-biofilm agents. Developing new anti-biofilm agents requires a comprehensive systems-level understanding of these mechanisms, as well as the discovery of new mechanisms. This can be accomplished through omics approaches such as transcriptomics, metabolomics, and proteomics, which can also be integrated to better understand biofilm biology. Guided by mechanistic understanding, in silico techniques such as virtual screening and machine learning can discover small molecules that can inhibit key biofilm regulators. To increase the likelihood that these candidate agents selected from in silico approaches are efficacious in humans, they must be tested in biologically relevant biofilm models. We discuss the benefits and drawbacks of in vitro and in vivo biofilm models and highlight organoids as a new biofilm model. This review offers a comprehensive guide of current and future biological and computational approaches of anti-biofilm therapeutic discovery for investigators to utilize to combat the antibiotic resistance crisis.
Collapse
Affiliation(s)
| | | | | | - Robert E. W. Hancock
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
135
|
Assessing biofilm inhibition and immunomodulatory activity of small amounts of synthetic host defense peptides synthesized using SPOT-array technology. Nat Protoc 2021; 16:1850-1870. [PMID: 33837303 DOI: 10.1038/s41596-021-00500-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 01/06/2021] [Indexed: 12/13/2022]
Abstract
Peptides are promising drug candidates because of their diversity, biocompatibility and spectrum of activities. Here, we describe a protocol for high-throughput screening of SPOT-peptide arrays to assess the antibiofilm, antimicrobial and immunomodulatory activities of synthetic peptides. It is a Protocol Extension of our previous Nature Protocols article, which describes the synthesis of SPOT-peptide arrays and assays for screening antimicrobial activity. This latest protocol allows the simultaneous assessment of hundreds of synthetic host defense peptides to define their overall activity profiles and identify candidate sequences that are suitable for further characterization and development as anti-infectives. When coupled with the SPOT-array technology for peptide synthesis, the described procedures are rapid, inexpensive and straightforward for peptide library screening. The protocols can be implemented in most microbiology or immunology research laboratories without the need for specialists. The time to complete each step ranges between 1 and 4 h with overnight pauses, and datasets related to the antibiofilm and immunomodulatory activities of a large set of peptide sequences can be generated in a few days.
Collapse
|
136
|
The Inhibitory Effects of Ficin on Streptococcus mutans Biofilm Formation. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6692328. [PMID: 33860052 PMCID: PMC8009705 DOI: 10.1155/2021/6692328] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 02/15/2021] [Accepted: 03/16/2021] [Indexed: 02/05/2023]
Abstract
To investigate the effects of ficin on biofilm formation of conditionally cariogenic Streptococcus mutans (S. mutans). Biomass and metabolic activity of biofilm were assessed using crystal violet assay, colony-forming unit (CFU) counting, and MTT assay. Extracellular polysaccharide (EPS) synthesis was displayed by SEM imaging, bacteria/EPS staining, and anthrone method while acid production was revealed by lactic acid assay. Growth curve and live/dead bacterial staining were conducted to monitor bacterial growth state in both planktonic and biofilm form. Total protein and extracellular proteins of S. mutans biofilm were analyzed by protein/bacterial staining and sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE), severally. qRT-PCR was conducted to detect acid production, acid tolerance, and biofilm formation associated genes. Crystal violet assay, CFU counting, and MTT assay showed that the suppression effect of ficin on S. mutans biofilm formation was concentration dependent. 4 mg/mL ficin had significant inhibitory effect on S. mutans biofilm formation including biomass, metabolic activity, EPS synthesis, and lactic acid production (p < 0.05). The growth curves from 0 mg/mL to 4 mg/mL ficin were aligned with each other. There was no significant difference among different ficin groups in terms of live/dead bacterial staining result (p > 0.05). Protein/bacterial staining outcome indicated that ficin inhibit both total protein and biofilm formation during the biofilm development. There were more relatively small molecular weight protein bands in extracellular proteins of 4 mg/mL ficin group when compared with the control. Generally, ficin could inhibit biofilm formation and reduce cariogenic virulence of S. mutans effectively in vitro; thus, it could be a potential anticaries agent.
Collapse
|
137
|
Wang Y, Zhang J, Gao T, Zhang N, He J, Wu F. Covalent immobilization of DJK-5 peptide on porous titanium for enhanced antibacterial effects and restrained inflammatory osteoclastogenesis. Colloids Surf B Biointerfaces 2021; 202:111697. [PMID: 33756295 DOI: 10.1016/j.colsurfb.2021.111697] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 10/21/2022]
Abstract
Currently, implant-related bone infection characterized by aggravated infection-induced inflammatory responses and osteolysis, remains a severe challenge in orthopedic surgery, especially in patients with osteoporosis. Attempts to control such responses using biomaterials with combined immunomodulatory and anti-bacterial properties may provide novel strategies. Herein, DJK-5, a class of host defense peptides (HDPs) with established antimicrobial and immunomodulatory functions, was introduced into porous Ti alloy. Our results indicated that the DJK-5 immobilized surfaces showed intrinsically multifunctional properties, including antibacterial ability, anti-inflammation, biocompatibility and osteolysis-inhibiting properties. The results demonstrated that the antibacterial efficiency of DJK-5 functionalized surfaces was over 90 % for both Gram-positive and Gram-negative bacteria. Specifically, DJK-5 functionalized samples also possessed the excellent anti-bacterial activity against a mixture of bacterial strains, including S. aureus, S. epidermidis and P. aeruginosa, with an antibacterial rate against mixed bacteria reaching 91.36 %, as well as reduced biofilm formation. The remarkable anti-bacterial efficacy was likely based on the direct anti-bacterial effect of DJK-5, which destroyed the integrity of bacteria membranes, leading to the leakage of intracellular materials. Additionally, the immobilized DJK-5 surfaces could indirectly kill bacteria through promoted macrophage capacity to bacteria uptake. Furthermore, DJK-5 functionalized surfaces suppressed inflammatory reaction by decreasing the release of pro-inflammatory factors and increasing the secretions of anti-inflammatory factors, and thereby impeded the activation of NF-κB signal pathway, which resulted in the disruption of the actin rings and decreased Tracp5b expressions. Based on these promising findings, the multi-functional DJK-5 immobilized titanium represents an efficient alternative to realize better osseointegration in sever implant-associated bacterial infections.
Collapse
Affiliation(s)
- Yao Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, PR China
| | - Junwei Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, PR China
| | - Tao Gao
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, PR China
| | - Nihui Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, PR China
| | - Jing He
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, PR China.
| | - Fang Wu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, PR China.
| |
Collapse
|
138
|
Kaźmierczak N, Grygorcewicz B, Piechowicz L. Biofilm Formation and Prevalence of Biofilm-Related Genes Among Clinical Strains of Multidrug-Resistant Staphylococcus aureus. Microb Drug Resist 2021; 27:956-964. [PMID: 33656375 DOI: 10.1089/mdr.2020.0399] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The biofilm-forming Staphylococcus aureus strains are responsible for causing a number of diseases. With the emergence of multidrug resistance they constitute a catastrophic threat to medicine. The ability of 65 clinical strains of multidrug-resistant S. aureus (MDRSA) to form biofilm in vitro was examined in this study and analyzed in relation to SCCmec, spa type, microbial surface components recognizing adhesive matrix molecules (MSCRAMMs), and ica genes. Results obtained from crystal violet and MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] assays showed that all MDRSA strains tested form biofilm but, of 65 strains, only 18 strains (28%) were found to form a biofilm with high metabolic activity and a great amount of biomass. The high proportion of MDRSA isolates in our study made no significant difference for ica and MSCRAMMs genes according to biofilm-forming capacity, except for fib, icaA, and cna gene. In addition, this study demonstrated that strains carrying SCCmec type I showed a significantly decreased biofilm viability compared with the strains harboring SCCmec type II and type IV, but SCCmec type could not serve as a good predictor of biofilm formation. However, we found that significantly weaker metabolic activity was detected in the biofilm of isolates with spa type t011.
Collapse
Affiliation(s)
- Natalia Kaźmierczak
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdansk, Gdańsk, Poland
| | - Bartłomiej Grygorcewicz
- Chair of Microbiology, Immunology and Laboratory Medicine, Department of Laboratory Medicine, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Lidia Piechowicz
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdansk, Gdańsk, Poland
| |
Collapse
|
139
|
Yu K, Alzahrani A, Khoddami S, Ferreira D, Scotland KB, Cheng JTJ, Yazdani‐Ahmadabadi H, Mei Y, Gill A, Takeuchi LE, Yeung E, Grecov D, Hancock REW, Chew BH, Lange D, Kizhakkedathu JN. Self-Limiting Mussel Inspired Thin Antifouling Coating with Broad-Spectrum Resistance to Biofilm Formation to Prevent Catheter-Associated Infection in Mouse and Porcine Models. Adv Healthc Mater 2021; 10:e2001573. [PMID: 33470545 DOI: 10.1002/adhm.202001573] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 12/23/2020] [Indexed: 12/22/2022]
Abstract
Catheter-associated urinary tract infections (CAUTIs) are one of the most commonly occurring hospital-acquired infections. Current coating strategies to prevent catheter-associated biofilm formation are limited by their poor long-term efficiency and limited applicability to diverse materials. Here, the authors report a highly effective non-fouling coating with long-term biofilm prevention activity and is applicable to diverse catheters. The thin coating is lubricous, stable, highly uniform, and shows broad spectrum prevention of biofilm formation of nine different bacterial strains and prevents the migration of bacteria on catheter surface. The coating method is adapted to human-sized catheters (both intraluminal and extraluminal) and demonstrates long-term biofilm prevention activity over 30 days in challenging conditions. The coated catheters are tested in a mouse CAUTI model and demonstrate high efficiency in preventing bacterial colonization of both Gram-positive and Gram-negative bacteria. Furthermore, the coated human-sized Foley catheters are evaluated in a porcine CAUTI model and show consistent efficiency in reducing biofilm formation by Escherichia coli (E. coli) over 95%. The simplicity of the coating method, the ability to apply this coating on diverse materials, and the high efficiency in preventing bacterial adhesion increase the potential of this method for the development of next generation infection resistant medical devices.
Collapse
Affiliation(s)
- Kai Yu
- Centre for Blood Research and Department of Pathology & Laboratory Medicine University of British Columbia Vancouver British Columbia V6T 1Z3 Canada
| | - Amal Alzahrani
- The Stone Centre at VGH Department of Urologic Sciences University of British Columbia Vancouver British Columbia V5Z 1M9 Canada
| | - Sara Khoddami
- The Stone Centre at VGH Department of Urologic Sciences University of British Columbia Vancouver British Columbia V5Z 1M9 Canada
| | - Demian Ferreira
- The Stone Centre at VGH Department of Urologic Sciences University of British Columbia Vancouver British Columbia V5Z 1M9 Canada
| | - Kymora B. Scotland
- The Stone Centre at VGH Department of Urologic Sciences University of British Columbia Vancouver British Columbia V5Z 1M9 Canada
| | - John T. J. Cheng
- Department of Microbiology and Immunology and Centre for Microbial Diseases and Immunity Research University of British Columbia Vancouver British Columbia V6T 1Z4 Canada
| | | | - Yan Mei
- Centre for Blood Research and Department of Pathology & Laboratory Medicine University of British Columbia Vancouver British Columbia V6T 1Z3 Canada
| | - Arshdeep Gill
- Department of Chemistry University of British Columbia Vancouver British Columbia V6T 1Z3 Canada
| | - Lily E. Takeuchi
- Centre for Blood Research and Department of Pathology & Laboratory Medicine University of British Columbia Vancouver British Columbia V6T 1Z3 Canada
| | - Edbert Yeung
- Department of Mechanical Engineering University of British Columbia Vancouver British Columbia V6T 1Z3 Canada
| | - Dana Grecov
- Department of Mechanical Engineering University of British Columbia Vancouver British Columbia V6T 1Z3 Canada
| | - Robert E. W. Hancock
- Department of Microbiology and Immunology and Centre for Microbial Diseases and Immunity Research University of British Columbia Vancouver British Columbia V6T 1Z4 Canada
| | - Ben H. Chew
- The Stone Centre at VGH Department of Urologic Sciences University of British Columbia Vancouver British Columbia V5Z 1M9 Canada
| | - Dirk Lange
- The Stone Centre at VGH Department of Urologic Sciences University of British Columbia Vancouver British Columbia V5Z 1M9 Canada
| | - Jayachandran N. Kizhakkedathu
- Centre for Blood Research and Department of Pathology & Laboratory Medicine University of British Columbia Vancouver British Columbia V6T 1Z3 Canada
- Department of Chemistry University of British Columbia Vancouver British Columbia V6T 1Z3 Canada
- School of Biomedical Engineering University of British Columbia Vancouver British Columbia V6T 1Z3 Canada
| |
Collapse
|
140
|
Saleem H, Mazhar S, Syed Q, Javed MQ, Adnan A. Bio-characterization of food grade pyocyanin bio-pigment extracted from chromogenic Pseudomonas species found in Pakistani native flora. ARAB J CHEM 2021. [DOI: 10.1016/j.arabjc.2021.103005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
141
|
Guedes GMM, Santos-Filho ASP, Regis WFM, Ocadaque CJ, Amando BR, Sidrim JJC, Brilhante RSN, Cordeiro RA, Bandeira SP, Rocha MFG, Castelo-Branco DSCM. Ex situ model of biofilm-associated wounds: providing a host-like environment for the study of Staphylococcus aureus and Pseudomonas aeruginosa biofilms. J Appl Microbiol 2021; 131:1487-1497. [PMID: 33556197 DOI: 10.1111/jam.15026] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 01/17/2021] [Accepted: 02/02/2021] [Indexed: 12/20/2022]
Abstract
AIM This study aimed to assess an ex situ model of biofilm-associated wounds on porcine skin for the study of Staphylococcus aureus and Pseudomonas aeruginosa biofilms in a host-like environment, after 48 to 120 h of incubation. MATERIAL AND RESULTS Ex situ and in vitro biofilms were comparatively analysed. Overall, CFU-counts and matrix quantification yielded significantly (P < 0·05) higher results for ex situ than in vitro biofilms. Confocal microscopy revealed greater (P < 0·05) biomass and thickness at 48-72 h and greater (P < 0·05) robustness at 72 h of growth. S. aureus ex situ biofilms produced less (P < 0·05) siderophore and proteases than in vitro biofilms, while P. aeruginosa ex situ biofilms produced more (P < 0·05) siderophores and less proteases than in vitro biofilms. CONCLUSIONS Biofilms grown ex situ present a greater amount of bacterial cells and polymeric matrix than their in vitro counterparts, reaching maturity at 72 h of growth. Moreover the production of virulence factors differs between ex situ and in vitro biofilms. SIGNIFICANCE AND IMPACT OF THE STUDY These findings emphasize the importance of using ex situ biofilm models, once they mimic in vivo conditions. The use of these models brings perspectives for the pursuit of therapeutic alternatives, as tests may be performed in a host-like environment.
Collapse
Affiliation(s)
- G M M Guedes
- Department of Pathology and Legal Medicine, Postgraduate Program in Medical Microbiology, Group of Applied Medical Microbiology, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - A S P Santos-Filho
- Department of Pathology and Legal Medicine, Postgraduate Program in Medical Microbiology, Group of Applied Medical Microbiology, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - W F M Regis
- Department of Pathology and Legal Medicine, Postgraduate Program in Medical Microbiology, Group of Applied Medical Microbiology, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - C J Ocadaque
- Department of Pathology and Legal Medicine, Postgraduate Program in Medical Microbiology, Group of Applied Medical Microbiology, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - B R Amando
- Department of Pathology and Legal Medicine, Postgraduate Program in Medical Microbiology, Group of Applied Medical Microbiology, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - J J C Sidrim
- Department of Pathology and Legal Medicine, Postgraduate Program in Medical Microbiology, Specialized Medical Mycology Center, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - R S N Brilhante
- Department of Pathology and Legal Medicine, Postgraduate Program in Medical Microbiology, Specialized Medical Mycology Center, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - R A Cordeiro
- Department of Pathology and Legal Medicine, Postgraduate Program in Medical Microbiology, Specialized Medical Mycology Center, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - S P Bandeira
- Department of Pathology and Legal Medicine, Postgraduate Program in Medical Microbiology, Specialized Medical Mycology Center, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - M F G Rocha
- Department of Pathology and Legal Medicine, Postgraduate Program in Medical Microbiology, Specialized Medical Mycology Center, Federal University of Ceará, Fortaleza, Ceará, Brazil.,Postgraduate Program in Veterinary Sciences, College of Veterinary, State University of Ceará, Fortaleza, Ceará, Brazil
| | - D S C M Castelo-Branco
- Department of Pathology and Legal Medicine, Postgraduate Program in Medical Microbiology, Group of Applied Medical Microbiology, Federal University of Ceará, Fortaleza, Ceará, Brazil.,Department of Pathology and Legal Medicine, Postgraduate Program in Medical Microbiology, Specialized Medical Mycology Center, Federal University of Ceará, Fortaleza, Ceará, Brazil
| |
Collapse
|
142
|
Atopobium vaginae and Prevotella bivia Are Able to Incorporate and Influence Gene Expression in a Pre-Formed Gardnerella vaginalis Biofilm. Pathogens 2021; 10:pathogens10020247. [PMID: 33672647 PMCID: PMC7924186 DOI: 10.3390/pathogens10020247] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/18/2021] [Accepted: 02/19/2021] [Indexed: 12/12/2022] Open
Abstract
Bacterial vaginosis (BV) is associated with a highly structured polymicrobial biofilm on the vaginal epithelium where Gardnerella species presumably play a pivotal role. Gardnerella vaginalis, Atopobium vaginae, and Prevotella bivia are vaginal pathogens detected during the early stages of incident BV. Herein, we aimed to analyze the impact of A. vaginae and P. bivia on a pre-established G. vaginalis biofilm using a novel in vitro triple-species biofilm model. Total biofilm biomass was determined by the crystal violet method. We also discriminated the bacterial populations in the biofilm and in its planktonic fraction by using PNA FISH. We further analyzed the influence of A. vaginae and P. bivia on the expression of key virulence genes of G. vaginalis by quantitative PCR. In our tested conditions, A. vaginae and P. bivia were able to incorporate into pre-established G. vaginalis biofilms but did not induce an increase in total biofilm biomass, when compared with 48-h G. vaginalis biofilms. However, they were able to significantly influence the expression of HMPREF0424_0821, a gene suggested to be associated with biofilm maintenance in G. vaginalis. This study suggests that microbial relationships between co-infecting bacteria can deeply affect the G. vaginalis biofilm, a crucial marker of BV.
Collapse
|
143
|
Preda M, Mihai MM, Popa LI, Dițu LM, Holban AM, Manolescu LSC, Popa GL, Muntean AA, Gheorghe I, Chifiriuc CM, Popa MI. Phenotypic and genotypic virulence features of staphylococcal strains isolated from difficult-to-treat skin and soft tissue infections. PLoS One 2021; 16:e0246478. [PMID: 33529240 PMCID: PMC7853507 DOI: 10.1371/journal.pone.0246478] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 01/19/2021] [Indexed: 12/04/2022] Open
Abstract
Chronic infections represent an important burden on the healthcare system and have a significant impact on the patients’ quality of life. While Staphylococcus spp. are commensal bacteria, they can become pathogenic, leading to various types of infections. In this study we aimed to characterize the virulence profiles of staphylococcal strains involved in difficult-to-treat skin and soft tissue infections, from both phenotypic and genotypic points of view. Phenotypic ability of the strains to secrete soluble virulence factors was assessed by a culturing dependent assay and their capacity to develop biofilms on inert substrate was screened by an adapted crystal violet microtiter method. We also tested the presence of several virulence genes by PCR. Most of the studied strains were isolated from purulent secretions of acne lesions and frequently secreted two or three soluble virulence factors. Most frequently secreted soluble virulence factors were caseinase (89%), lipase (71%) and lecithinase (67%). Almost half of the strains produced a well-represented biofilm. The molecular characterization showed the presence of the genes cna, hlg, clfA, and clfB. Staphylococcal strains that produce difficult-to-treat skin and soft tissue infections seem to be characterized by an enhanced ability to produce different soluble virulence factors and to develop biofilms in vitro. Further studies need to be developed in other Staphylococcus spp. infections in order to confirm this hypothesis.
Collapse
Affiliation(s)
- Mădălina Preda
- Department of Microbiology, Parasitology and Virology, Faculty of Midwives and Nursing, ‘Carol Davila’ University of Medicine and Pharmacy, Bucharest, Romania
- ‘Cantacuzino’ National Medico-Military Research and Development Institute, Bucharest, Romania
| | - Mara Mădălina Mihai
- Department of Oncologic Dermatology, ‘Carol Davila’ University of Medicine and Pharmacy, Bucharest, Romania
- Department of Dermatology, ‘Elias’ University Emergency Hospital, Bucharest, Romania
- * E-mail: (MMM); (LIP)
| | - Laura Ioana Popa
- Department of Bioinformatics, The National Institute of Research and Development for Biological Sciences, Bucharest, Romania
- Research Institute of the University of Bucharest (ICUB), Bucharest, Romania
- * E-mail: (MMM); (LIP)
| | - Lia-Mara Dițu
- Research Institute of the University of Bucharest (ICUB), Bucharest, Romania
- Department of Microbiology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Alina Maria Holban
- Research Institute of the University of Bucharest (ICUB), Bucharest, Romania
- Department of Microbiology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Loredana Sabina Cornelia Manolescu
- Department of Microbiology, Parasitology and Virology, Faculty of Midwives and Nursing, ‘Carol Davila’ University of Medicine and Pharmacy, Bucharest, Romania
| | - Gabriela-Loredana Popa
- Department of Microbiology, Faculty of Dental Medicine, ‘Carol Davila’ University of Medicine and Pharmacy, Bucharest, Romania
| | | | - Irina Gheorghe
- Research Institute of the University of Bucharest (ICUB), Bucharest, Romania
- Department of Microbiology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Carmen Mariana Chifiriuc
- Research Institute of the University of Bucharest (ICUB), Bucharest, Romania
- Department of Microbiology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Mircea-Ioan Popa
- ‘Cantacuzino’ National Medico-Military Research and Development Institute, Bucharest, Romania
- Department of Microbiology, Faculty of Medicine, ‘Carol Davila’ University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
144
|
Nassar R, Hachim M, Nassar M, Kaklamanos EG, Jamal M, Williams D, Senok A. Microbial Metabolic Genes Crucial for S. aureus Biofilms: An Insight From Re-analysis of Publicly Available Microarray Datasets. Front Microbiol 2021; 11:607002. [PMID: 33584569 PMCID: PMC7876462 DOI: 10.3389/fmicb.2020.607002] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 12/24/2020] [Indexed: 01/01/2023] Open
Abstract
Bacterial biofilms are microbial lifestyles found in all environments. Up to 80% of human infections and 60–70% of hospital-acquired infections have a biofilm origin, with Staphylococcus aureus one of the leading causes of these infections. Microorganisms in biofilms exhibit significant antimicrobial resistance which poses important treatment challenges, hence the urgent need to identify novel antibiofilm strategies. Microbes form biofilms in response to various factors, and once these 3-dimentional structures form they are highly recalcitrant to removal. The switch from planktonic lifestyle to the biofilm protected mode of growth results in a phenotypic shift in the behavior of the microorganisms in terms of growth rate and gene expression. Given these changes, investigation of microbial gene expression and their modulation at different stages of biofilm maturation is needed to provide vital insight into the behavior of biofilm cells. In this study, we analyzed publicly available transcriptomic dataset of S. aureus biofilms at different stages of maturation to identify consistently upregulated genes irrespective of the biofilm maturation stage. Our reanalysis identified a total of 6 differentially expressed genes upregulated in both 48 and 144-h old S. aureus biofilms. Functional analysis revealed that these genes encode for proteins which play a role in key microbial metabolic pathways. However, these genes, as yet, are unrelated or fully studied in the context of biofilm. Moreover, the findings of this in silico work, suggest that these genes may represent potential novel targets for the development of more effective antibiofilm strategies against S. aureus biofilm-associated infections.
Collapse
Affiliation(s)
- Rania Nassar
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates.,School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff, United Kingdom
| | - Mahmood Hachim
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Mohannad Nassar
- Department of Preventive and Restorative Dentistry, College of Dental Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Eleftherios G Kaklamanos
- Hamdan Bin Mohammed College of Dental Medicine, Mohammed Bin Rashid University of Medicine & Health Sciences, Dubai, United Arab Emirates
| | - Mohamed Jamal
- Hamdan Bin Mohammed College of Dental Medicine, Mohammed Bin Rashid University of Medicine & Health Sciences, Dubai, United Arab Emirates
| | - David Williams
- School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff, United Kingdom
| | - Abiola Senok
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| |
Collapse
|
145
|
Eivazzadeh-Keihan R, Radinekiyan F, Aliabadi HAM, Sukhtezari S, Tahmasebi B, Maleki A, Madanchi H. Chitosan hydrogel/silk fibroin/Mg(OH) 2 nanobiocomposite as a novel scaffold with antimicrobial activity and improved mechanical properties. Sci Rep 2021; 11:650. [PMID: 33436831 PMCID: PMC7804245 DOI: 10.1038/s41598-020-80133-3] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 12/17/2020] [Indexed: 01/29/2023] Open
Abstract
Herein, a novel nanobiocomposite scaffold based on modifying synthesized cross-linked terephthaloyl thiourea-chitosan hydrogel (CTT-CS hydrogel) substrate using the extracted silk fibroin (SF) biopolymer and prepared Mg(OH)2 nanoparticles was designed and synthesized. The biological capacity of this nanobiocomposite scaffold was evaluated by cell viability method, red blood cells hemolytic and anti-biofilm assays. According to the obtained results from 3 and 7 days, the cell viability of CTT-CS/SF/Mg(OH)2 nanobiocomposite scaffold was accompanied by a considerable increment from 62.5 to 89.6% respectively. Furthermore, its low hemolytic effect (4.5%), and as well, the high anti-biofilm activity and prevention of the P. aeruginosa biofilm formation confirmed its promising hemocompatibility and antibacterial activity. Apart from the cell viability, blood biocompatibility, and antibacterial activity of CTT-CS/SF/Mg(OH)2 nanobiocomposite scaffold, its structural features were characterized using spectral and analytical techniques (FT-IR, EDX, FE-SEM and TG). As well as, given the mechanical tests, it was indicated that the addition of SF and Mg(OH)2 nanoparticles to the CTT-CS hydrogel could improve its compressive strength from 65.42 to 649.56 kPa.
Collapse
Affiliation(s)
- Reza Eivazzadeh-Keihan
- Catalysts and Organic Synthesis Research Laboratory, Department of Chemistry, Iran University of Science and Technology, 16846-13114, Tehran, Iran
| | - Fateme Radinekiyan
- Catalysts and Organic Synthesis Research Laboratory, Department of Chemistry, Iran University of Science and Technology, 16846-13114, Tehran, Iran
| | - Hooman Aghamirza Moghim Aliabadi
- Faculty of Chemistry, K.N. Toosi University of Technology, Tehran, Iran
- Protein Chemistry Laboratory, Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Sima Sukhtezari
- Catalysts and Organic Synthesis Research Laboratory, Department of Chemistry, Iran University of Science and Technology, 16846-13114, Tehran, Iran
| | - Behnam Tahmasebi
- School of Chemistry, College of Science, University of Tehran, Tehran, Iran
| | - Ali Maleki
- Catalysts and Organic Synthesis Research Laboratory, Department of Chemistry, Iran University of Science and Technology, 16846-13114, Tehran, Iran.
| | - Hamid Madanchi
- Department of Biotechnology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran.
- Drug Design and Bioinformatics Unit, Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
146
|
Dostert M, Trimble MJ, Hancock REW. Antibiofilm peptides: overcoming biofilm-related treatment failure. RSC Adv 2021; 11:2718-2728. [PMID: 35424252 PMCID: PMC8694000 DOI: 10.1039/d0ra09739j] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 12/18/2020] [Indexed: 12/19/2022] Open
Abstract
Health leaders and scientists worldwide consider antibiotic resistance among the world's most dangerous pathogens as one of the biggest threats to global health. Antibiotic resistance has largely been attributed to genetic changes, but the role and recalcitrance of biofilms, largely due to growth state dependent adaptive resistance, is becoming increasingly appreciated. Biofilms are mono- and multi-species microbial communities embedded in an extracellular, protective matrix. In this growth state, bacteria are transcriptionally primed to survive extracellular stresses. Adaptations, affecting metabolism, regulation, surface charge, immune recognition and clearance, allow bacteria to thrive in the human body and withstand antibiotics and the host immune system. Biofilms resist clearance by multiple antibiotics and have a major role in chronic infections, causing more than 65% of all infections. No specific antibiofilm agents have been developed. Thus, there is a pressing need for alternatives to traditional antibiotics that directly inhibit and/or eradicate biofilms. Host defence peptides (HDPs) are small cationic peptides that are part of the innate immune system to both directly kill microbes but also function to modulate the immune response. Specific HDPs and their derivatives demonstrate broad-spectrum activity against biofilms. In vivo biofilm assays show efficacy in abscess, respiratory, in-dwelling device, contact lens and skin infection models. Further progress has been made through the study of ex vivo organoid and air-liquid interface models to better understand human infections and treatment while relieving the burden and complex nature of animal models. These avenues pave the way for a better understanding and treatment of the underlying cause of chronic infections that challenge the healthcare system.
Collapse
Affiliation(s)
- Melanie Dostert
- Department of Microbiology and Immunology, University of British Columbia Vancouver British Columbia Canada
| | - Michael J Trimble
- Department of Microbiology and Immunology, University of British Columbia Vancouver British Columbia Canada
| | - Robert E W Hancock
- Department of Microbiology and Immunology, University of British Columbia Vancouver British Columbia Canada
| |
Collapse
|
147
|
Candida albicans as an Essential "Keystone" Component within Polymicrobial Oral Biofilm Models? Microorganisms 2020; 9:microorganisms9010059. [PMID: 33379333 PMCID: PMC7823588 DOI: 10.3390/microorganisms9010059] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/23/2020] [Accepted: 12/24/2020] [Indexed: 02/06/2023] Open
Abstract
Background: Existing standardized biofilm assays focus on simple mono-species or bacterial-only models. Incorporating Candida albicans into complex biofilm models can offer a more appropriate and relevant polymicrobial biofilm for the development of oral health products. Aims: This study aimed to assess the importance of interkingdom interactions in polymicrobial oral biofilm systems with or without C. albicans, and test how these models respond to oral therapeutic challenges in vitro. Materials and Methods: Polymicrobial biofilms (two models containing 5 and 10 bacterial species, respectively) were created in parallel in the presence and absence of C. albicans and challenged using clinically relevant antimicrobials. The metabolic profiles and biomasses of these complex biofilms were estimated using resazurin dye and crystal violet stain, respectively. Quantitative PCR was utilized to assess compositional changes in microbial load. Additional assays, for measurements of pH and lactate, were included to monitor fluctuations in virulence "biomarkers." Results: An increased level of metabolic activity and biomass in the presence of C. albicans was observed. Bacterial load was increased by more than a factor of 10 in the presence of C. albicans. Assays showed inclusion of C. albicans impacted the biofilm virulence profiles. C. albicans did not affect the biofilms' responses to the short-term incubations with different treatments. Conclusions: The interkingdom biofilms described herein are structurally robust and exhibit all the hallmarks of a reproducible model. To our knowledge, these data are the first to test the hypothesis that yeasts may act as potential "keystone" components of oral biofilms.
Collapse
|
148
|
Ketone- and Cyano-Selenoesters to Overcome Efflux Pump, Quorum-Sensing, and Biofilm-Mediated Resistance. Antibiotics (Basel) 2020; 9:antibiotics9120896. [PMID: 33322639 PMCID: PMC7763688 DOI: 10.3390/antibiotics9120896] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/02/2020] [Accepted: 12/10/2020] [Indexed: 12/15/2022] Open
Abstract
The emergence of drug-resistant pathogens leads to a gradual decline in the efficacy of many antibacterial agents, which poses a serious problem for proper therapy. Multidrug resistance (MDR) mechanisms allow resistant bacteria to have limited uptake of drugs, modification of their target molecules, drug inactivation, or release of the drug into the extracellular space by efflux pumps (EPs). In previous studies, selenoesters have proved to be promising derivatives with a noteworthy antimicrobial activity. On the basis of these results, two series of novel selenoesters were synthesized to achieve more potent antibacterial activity on Gram-positive and Gram-negative bacteria. Fifteen selenoesters (eight ketone-selenoesters and seven cyano-selenoesters) were investigated with regards to their efflux pump-inhibiting, anti-quorum-sensing (QS), and anti-biofilm effects in vitro. According to the results of the antibacterial activity, the ketone-selenoesters proved to be more potent antibacterial compounds than the cyano-selenoesters. With regard to efflux pump inhibition, one cyano-selenoester on methicillin-resistant S. aureus and one ketone-selenoester on Salmonella Typhimurium were potent inhibitors. The biofilm inhibitory capacity and the ability of the derivatives to disrupt mature biofilms were noteworthy in all the experimental systems applied. Regarding QS inhibition, four ketone-selenoesters and three cyano-selenoesters exerted a noteworthy effect on Vibrio campbellii strains.
Collapse
|
149
|
Screening and characterization of a novel Antibiofilm polypeptide derived from filamentous Fungi. J Proteomics 2020; 233:104075. [PMID: 33309927 DOI: 10.1016/j.jprot.2020.104075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 09/17/2020] [Accepted: 12/06/2020] [Indexed: 12/14/2022]
Abstract
In the present study, 120 fungal isolates were locally isolated from soil and selected according to their ability to antimicrobial activity. Then, selected isolates were tested for their ability to prevent biofilm formation and only one isolate (A01) showed an antibiofilm effect. The isolate A01 identified as Aspergillus tubingensis by sequencing of the 18S ITS region and a segment of β-tubulin gene. Then, 5 fractions were prepared from the culture filtrate of A. tubingensis A01 using the ultrafiltration technique to find active polypeptide fraction. The experiments revealed that one of them had an antibiofilm activity. The MALDI-TOF/MS analyses demonstrated that this polypeptide composed of 92 amino acids and had a molecular mass of 10,087 Da. The sequence alignment showed homology with hypothetical protein (OJI81679.1). The gene coding for this polypeptide consisting of 279 nucleotides, herein we called astucin, was cloned and sequenced from A. tubingensis A01 to confirm results. The MIC of the purified polypeptide was 32 m/L and 128 μg/mL and the MBIC was 2 and 8 μg/mL against Staphylococcus aureus and MRSA, respectively. The results demonstrated that the antimicrobial and antibiofilm activity of astucin, together with its lack of cytotoxicity, makes it an alternative for application in medicine. SIGNIFICANCE: Antibiotic resistance is a global problem and the emergence of antibiotic resistant bacteria reduce the effect the current treatment approaches. In this context, antimicrobial peptides stand out as potentional agents to combat bacterial infection especially, biofilm related infections. Importantly, this study have greatly considered our understanding for fungal derived antibiofilm polypeptides. In this study, traditional selection method combined with crystal violet assay is used to investigate antibiofilm polypeptides. We identified antibiofilm polypeptides purified from A. tubingensis A01. This protein shows antimicrobial and antibiofilm activity against S. aureus.
Collapse
|
150
|
Eivazzadeh-Keihan R, Khalili F, Aliabadi HAM, Maleki A, Madanchi H, Ziabari EZ, Bani MS. Alginate hydrogel-polyvinyl alcohol/silk fibroin/magnesium hydroxide nanorods: A novel scaffold with biological and antibacterial activity and improved mechanical properties. Int J Biol Macromol 2020; 162:1959-1971. [DOI: 10.1016/j.ijbiomac.2020.08.090] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 08/07/2020] [Accepted: 08/08/2020] [Indexed: 12/21/2022]
|